Abstract
Compared to other organs, the brain is especially exposed to oxidative stress. In general, brains from young females tend to present lower oxidative damage in comparison to their male counterparts. This has been attributed to higher antioxidant defenses and a better mitochondrial function in females, which has been linked to neuroprotection in this group. However, these differences usually disappear with aging, and the incidence of brain pathologies increases in aged females. Sexual hormones, which suffer a decrease with normal aging, have been proposed as the key factors involved in these gender differences. Here, we provide an overview of redox status and mitochondrial function regulation by sexual hormones and their influence in normal brain aging. Furthermore, we discuss how sexual hormones, as well as phytoestrogens, may play an important role in the development and progression of several brain pathologies, including neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, stroke or brain cancer.
Keywords: Sex hormones, Brain, Redox homeostasis, Mitochondrial function, Aging, Sex differences
Graphical abstract
Highlights
-
•
Sex hormones are reduced with aging, especially in females, affecting redox balance.
-
•
Normal aging is associated to a worse redox homeostasis in the brain.
-
•
Young females show better mitochondrial function and higher antioxidant defenses.
-
•
Development of brain pathologies is influenced by sex hormones and phytoestrogens.
Abbreviations
- 2-ME2
2-methoxyestradiol
- Aβ plaques
amyloid-β plaques
- AD
Alzheimer's disease
- AKT
protein kinase B
- ALS
Amyotrophic lateral sclerosis
- AMPK
AMP-activated protein kinase
- AP-1
activator protein-1
- AR
androgen receptor
- BAT
brown adipose tissue
- CAT
catalase
- CREB
cyclic AMP response element binding protein
- EGFR
epidermal growth factor receptor
- eNOS
endothelial nitric oxide synthase
- ERE
estrogen response elements
- ERK
extracellular signal-regulated kinases
- GABPα
GA-binding protein alpha
- GPER
G protein-coupled estrogen receptor
- GPx
glutathione peroxidase
- GRd
glutathione reductase
- GSH
reduced glutathione
- GSSG
oxidized glutathione
- HD
Huntington's disease
- Htt
huntingtin
- IDH2
isocitrate dehydrogenase 2
- αKGDH
α-Ketoglutarate dehydrogenase
- mtDNA
mitochondrial DNA
- NF-κB
nuclear factor-κB
- Ngb
globin neuroglobin
- NRF2
nuclear factor erythroid 2-related factor
- NRF-1/NRF-2
nuclear respiratory factors 1 and 2
- OXPHOS complexes
oxidative phosphorylation complexes
- PD
Parkinson's disease
- PGC-1α
coactivator peroxisome proliferator-activated receptor gamma coactivator 1 α
- PPARα
peroxisome proliferator-activated receptor alpha
- Prx
peroxiredoxin
- ROS
reactive oxygen species
- SIRTs
sirtuins
- StAR
steroidogenic acute regulatory protein
- SOD
superoxide dismutase
- TFAM
mitochondrial transcription factor A
- Trx
thioredoxin
- TrxR
thioredoxin reductase
- TSPO
translocator protein
- UCPs
uncoupling proteins
- WAT
white adipose tissue
1. Introduction
Oxidative damage and mitochondrial dysfunction play an important role in the development and progression of brain disease [1]. Women seem to be protected at young ages from the development of several brain pathologies, including neurodegenerative diseases, stroke, and cancer. Nevertheless, this protection disappears amongst postmenopausal women and the incidence and/or severity of these diseases significantly increase in this group, which suggests a protective role of estrogens regulating oxidative stress [2].
It is well known that H2O2 can act as a second messenger in the cell targeting several regulatory proteins, such as phosphatases, proteases or transcription factors [3,4]. However, excessive amounts of H2O2 and/or other ROS can become pathologic as they damage cellular structures if not neutralized properly [5,6]. For this reason, ROS production and scavenging are tightly regulated in the cell, through the activity of antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT) and the glutathione peroxidase/reductase (Gpx/GRd) and peroxiredoxin/thioredoxin systems (Prx/Trx). Furthermore, mitochondrial function is also controlled by uncoupling proteins (UCPs), which dissipate the proton gradient generated by the respiratory chain and reduce ROS production [7,8], and some sirtuins (SIRTs), which deacetylate several proteins involved in redox regulation [9].
Most of the proteins implicated in the regulation of redox status and mitochondrial function are under the control of sexual hormones, which could be the reason why young women are more protected against brain injury [10]. Thus, mitochondrial function and antioxidant enzymes, and their differences with gender and aging, have been widely studied to better understand both normal function of the brain and their pathological implications. In this review, we first describe the regulation of antioxidant enzymes and mitochondrial function by sexual hormones. Second, we address the main changes that occur in the brain with aging, and finally, we discuss the influence of sexual hormones on several brain pathologies.
2. Sex hormones and redox regulation
Redox homeostasis and mitochondrial function show sex differences in the brain [11], as well as some pathologies or synaptic transmission related-mechanisms [12,13] that have been described as sex specific processes [14]. Redox homeostasis in the brain is highly important, as an imbalance may contribute to the pathophysiology of many neurologic diseases [15,16].
Sex differences on oxidative stress in the brain, including free radical production, oxidative damage and antioxidant enzymes levels and/or activity, have been studied for a long time [14]. Some investigations show higher oxidative damage in lipids, proteins and DNA in male rats than in females [[17], [18], [19], [20], [21], [22], [23], [24], [25], [26]]. This oxidative damage is due to a higher ROS production in male rats [20,22,[27], [28], [29]] and, moreover, lower antioxidant enzymes levels and/or activity [18,22,23,[30], [31], [32], [33], [34]]. It is worthy to note that, although these studies support a better redox homeostasis in female than in male rats, other reports show no differences [26,33,[35], [36], [37], [38]]. Some authors suggest that age could be responsible for this controversy [31]. In fact, young brains of female rats show higher SOD [18,27,28,31,34] and CAT levels [18,30,32] than male rats of the same age, whereas in older rats these differences are not observed. Moreover, this controversy could be also explained if different parts of the brain are considered. For example, Noschang et al. reported that CAT levels were higher in the striatum of female brain rats, but male rats showed increased CAT levels in the prefrontal cortex. On the other hand, SOD levels were higher in the prefrontal cortex of female rats, and striatum SOD activity was similar in both sexes [34].
Sexual steroid hormones could be the reason of these sex differences observed in the regulation of redox homeostasis in the brain [39]. Sex hormones are steroids derived from cholesterol and are produced by the gonads, the adrenal glands and the placenta. They can reach and cross the blood-brain barrier and enter the central nervous system, where they modulate several physiological functions [40]. In addition, some neurons and glial cells are also capable of synthesize sex hormones de novo independently from peripheral tissues, which are commonly referred to as neurosteroids. These neurosteroids are chemically and biologically identical to circulating steroids [40,41]. Mitochondria are involved in steroidogenesis, as the first and rate-limiting step consists in the transfer of cholesterol inside these organelles. Acute regulatory protein (StAR) and translocator protein (TSPO) have been identified to participate in this transport [42]. Then, cholesterol is converted to pregnenolone, the precursor of steroids, by the cytochrome P450 side-chain cleavage enzyme. Finally, pregnenolone is transported outside the mitochondria and can be converted into the different sex hormones by specific enzymes [10]. Some studies suggest that the enzymes involved in neurosteroidogenesis show a sex-dependent pattern, contributing to the different levels of sexual hormones observed in males and females, although this has been proven in animal models and still needs to be addressed in humans [43].
Female sexual hormones, 17β-estradiol (E2) and progesterone, possess neuroprotective effects in vivo and in vitro at physiological concentrations [[44], [45], [46], [47]]. However, male steroids, androgens and testosterone, usually present neurotoxicity [45,48]. Neuroprotective effects of sexual hormones can occur through genomic and non-genomic mechanisms [14]. Genomic mechanisms are triggered through the interaction with their receptor: the estrogen receptor (ER) α or β [49], the progesterone receptor [50] or the androgen receptor (AR) [51]. Interestingly, it seems that, at physiological concentrations, E2 is involved in neuroprotection through the activation of ERα and not ERβ, as seen by the reduction in the extent of cerebral injury in a mouse model of stroke, while at pharmacological concentrations, E2 activates ER-independent mechanisms [52]. In the classical pathway, the hormone binds to its receptor in the plasmatic membrane or in the nucleus, and this hormone-receptor complex binds directly to the promoter of target genes, through the estrogen response elements (ERE). There are alternative pathways in which the hormone-receptor complex interacts with other transcription factors that bind to DNA, such as activator protein-1 (AP-1) [53], cyclic AMP response element binding protein (CREB) [54], or nuclear factor-κB (NF-κB) [55], being this one especially important since it is known to activate key regulatory genes for maintaining the redox homeostasis of the cell.
Sexual hormones can also activate G-protein-coupled receptor pathways, protein kinases that lead to phosphorylation and activation of transcription factors, resulting in non-genomic effects. One example of this mechanism can be observed with the nuclear factor erythroid 2-related factor (NRF2) estrogenic activation by phosphatidylinositol 3-kinase and glycogen synthase kinase 3β pathways [56]. Finally, whichever the route of activation, the maintenance of redox homeostasis through sex hormones occurs due to the increase in the expression and/or activity of antioxidant enzymes such as SOD, CAT and GPx [14] (Fig. 1).
Fig. 1.
Mechanisms involved in the regulation of redox homeostasis by estradiol. Estradiol can exert its effects by binding to the ERs (ERα or ERβ) or to GPER. Active ERs may bind to the promoter of target genes, which contain an ERE element. Genomic effects include the upregulation of several mitochondrial and metabolic genes, as well as anti-inflammatory and anti-apoptotic genes. Non-genomic effects, such as a reduction of oxidative stress, apoptosis avoidance or anti-inflammatory effects, are mediated through several protein kinases activated by these estrogen receptors.
Moreover, estrogens may also modulate metabolic pathways through an increase of the expression of some electron transport chain proteins, such as cytochrome C and complex IV subunits, as well as citrate synthase enzymatic activity [57]. This control on the respiratory chain through sexual hormones is not only due to the ERE presence in the promoter of these genes, but also to the coordination of mtDNA transcription through nuclear transcription factors that are regulated by sexual hormones [39]. In fact, estrogens stimulate the transcription of nuclear respiratory factors 1 and 2 (NRF-1 and NRF-2), coactivator peroxisome proliferator-activated receptor gamma coactivator 1 α (PGC-1α) and mitochondrial transcription factor A (TFAM), which are involved in mtDNA transcription [58].
Estrogen-related receptor alpha (ERRα), an orphan nuclear receptor, also modulates estrogen signaling [59] and can be coactivated by PGC-1α. ERRα plays an essential role in the regulation of ROS generation [60], and mitochondrial function through the regulation of genes involved in mitogenesis and oxidative phosphorylation, and regulation of mitochondrial replication through TFAM activation [61]. Moreover, PGC-1α induces ERRα and, as PGC-1α mRNA levels increase, ERRα mRNA levels also increase [62].
Finally, phytoestrogens are a huge group of natural compounds which have been found in an extensive number of plants. The common characteristic of these compounds is that they present a chemical structure similar to estrogens and harbor some estrogenic activity [63]. For this reason, phytoestrogens could act through their interaction with the estrogen receptors, although they are able to exert their effects in an ER-independent manner. It is well known that phytoestrogens show antioxidant and anti-inflammatory activities [64], among others, and because of these properties, phytoestrogens could influence the normal function of the brain and modulate the molecular basis of the disease or, at least, palliate some of the symptoms. Phytoestrogens are also able to modulate the activity of ERRα through its direct interaction and activation as agonists [65]. Thus, phytoestrogens modulate mitochondrial biogenesis and function regulating the expression of NRF1, GABPα and PPARα through their interaction with ERRα [66].
3. Sex hormones and uncoupling proteins
UCPs are a family of proteins of the internal mitochondrial membrane whose function is to uncouple the electron transport chain form oxidative phosphorylation, dissipating the proton gradient, causing a decrease in membrane potential [67]. The reduction of the proton gradient contributes to decrease the production of ROS [68].
Five different isoforms of UCPs have been characterized. UCP1 was the first to be described in brown adipose tissue (BAT), tissue in which this protein enables thermogenesis particularly for newborns as well as hibernating animals [69]. UCP2 is expressed in most tissues, but especially in the immune system [67,70,71]. UCP2 has also been extensively investigated in the brain and has been found in different areas. Although there is not much evidence for the thermogenic role of UCP2 in the brain, some studies in rats have shown that it may create temperature variations that could affect the transmission of signals between neurons, increasing the traffic of neurotransmitters [72]. Moreover, it has been described that UCP2 is relevant for adaptive responses in cortical and hippocampal neurons, as well as for perinatal hypoxia-triggered circuit adaptations [73]. Whereas UCP3 is expressed mainly in muscle and BAT [74,75], UCP4 and UCP5 (also known as brain mitochondrial carrier protein 1) are expressed mainly in the brain [67,[76], [77], [78]].
The role of UCP2, UCP4 and UCP5 in oxidative stress modulation of mitochondrial ROS is very clear [72,76,79]. UCP4 has a neuroprotective role in early neuronal development, while UCP5 and UCP2 are important for decreasing ROS production in neurons. All three UCPs have been found decreased in Alzheimer's disease patients, while both UCP4 and UCP5 have been described as protector factors for Parkinson's disease. This reduction of ROS promoted by UCPs in the nervous system could be relevant to avoid or ameliorate neurodegenerative diseases, including Alzheimer's and Parkinson's disease or lateral amyotrophic sclerosis [20,76,78].
Different studies have shown that UCPs are differentially expressed between males and females in animal models, although few of them focus on the brain. UCP1 levels are higher in female than male rats in BAT [[81], [82], [83], [84]]. UCP2 levels are also higher in females in BAT [81] and in white adipose tissue (WAT) [82]. In skeletal muscle and WAT, females show increased levels of UCP3 when compared to males [82,85]. In the brain, UCP4 and UCP5 are higher in females [78]. Both proteins are also affected by age, as UCP4 decreases in older males, while no differences are observed in females, and UCP5 levels follows the opposite pattern, increasing with age in females while they are not affected in males [78].
These differences between males and females can be attributed to the effect of sexual hormones [86], as several studies carried out in primary mouse cultures or cancer cell lines have demonstrated that E2, progesterone and testosterone regulate the expression of these proteins [[87], [88], [89]]. In general, female hormones upregulate UCPs, while testosterone downregulates these proteins. Nevertheless, it should be noted that their action is dependent on the tissue and ratio ERα/ERβ. This way, as mentioned before, it seems that E2 exerts its neuroprotective effects through the activation of ERα [52], although some reports suggest that this protective effects, as well as the induction of UCPs, depends on the abundance of ERβ in the tissue [[89], [90], [91], [92]]. This would be consistent with the fact that genistein, which shows higher affinity for ERβ, has a protective effect for mitochondria, enhancing their function and increasing the antioxidant response and expression of UCPs [[92], [93], [94]].
4. Sex hormones and sirtuins
SIRTs belong to a family of histone deacetylases that are dependent on NAD+ for their enzymatic activity, which makes them cellular energy sensors. Some SIRTs also show other enzymatic activities, such as ADP-ribosylation or desuccinylation [95]. Up to seven isoforms (SIRT1-SIRT7) have been identified in mammals, and all of them are expressed in the brain, although they may suffer changes in their expression with aging [96].
The most studied sirtuins are SIRT1 and SIRT3. SIRT1 directly regulates mitochondrial biogenesis through deacetylation of PGC-1α [97], while SIRT3 is critical for reducing oxidative stress and maintaining proper mitochondrial function by regulating both expression and deacetylation levels of several antioxidant proteins [98,99]. It has been reported that SIRT1 levels in mice are modulated by age and sex, and this modulation is area specific in the brain [100]. In humans, SIRT1 also shows a sex-dependent pattern, as its enzymatic activity in serum was found to peak at different ages for men and women [101]. Furthermore, SIRT1 activity in serum and skin was significantly reduced with age, especially for women [101,102], suggesting a role of estrogens in the modulation of SIRTs.
Interestingly, sirtuins have also been described as potent modulators of sexual hormones receptors. SIRT1 has been identified as a coactivator of ERα, but not ERβ, through an independent mechanism not involving deacetylation [[103], [104], [105]]. However, other studies report SIRT1 as a negative regulator of ERα and AR through deacetylation [103] or by inducing AKT-dependent phosphorylation of the receptor [106].
Several reports confirm that sexual hormones regulate the levels of some SIRTs by several mechanisms. Upon activation, ERα is directly involved in the activation of transcription of the SIRT1 gene [104,[107], [108], [109]], which has been associated to protection from cell stressors. For instance, in some rat models, E2 treatment activated SIRT1 in the brain, which resulted in the deacetylation of NF-κB [107] or p53 [110] and the inhibition of proinflammatory and proapoptotic proteins, significantly reducing oxidative stress. Furthermore, E2 induction of SIRT1 seems to be essential for the activation of AMPK and protection from ischemic brain injury [111]. Nevertheless, E2 was reported to reduce the expression of SIRT1 through the activation of Akt/ERK pathway in smooth muscle cells, suggesting that the effects of sexual hormones in SIRT regulation might be tissue dependent [112].
On the other hand, GPER has been also identified to be involved in the upregulation of SIRT1 through the activation of EGFR/ERK/c-fos/AP-1 pathway [113]. Furthermore, activation of AR by testosterone has also been described to regulate SIRT1 expression through the induction of eNOS, which prevented cellular senescence in mice [114].
Fewer studies have linked sexual hormones with SIRT3 modulation. ERβ has been found to recruit some transcription factors, such as Sp1, in the promoter of the SIRT3 gene [115]. ERα might also be involved in the expression of SIRT3, as this receptor induces the expression of Nrf-2, which stimulates an antioxidant response involving also SIRT3 [116]. Notably, it has also been reported that PGC-1α and ERRα can upregulate SIRT3 expression and protein levels [117].
Moreover, 2-methoxyestradiol (2-ME2), a naturally occurring metabolite of E2, regulates both SIRT1 and SIRT3 levels. 2-ME2 has been reported to upregulate SIRT1 protein levels to trigger autophagy [118], while it inhibited SIRT3 activity through physical interaction, resulting in a decrease of mitochondrial mass and activity [119]. Finally, some phytoestrogens have shown the potential to modulate both SIRT1 and SIRT3, presumably through the activation of ER, such as genistein [120], hop-derived phytoestrogens [121] and resveratrol [122].
5. Sex hormones and aging
Brain aging is linked to a decline in mental functions, as well as the development of neuronal dysfunction. This deterioration has been related to a decrease in the levels of sexual hormones with age. Therefore, it has been postulated that the reduction in sex hormone levels could aggravate neuronal dysfunction associated with age [39].
In general, a decline in mitochondrial function and antioxidant capacity in the brain has been described with normal aging [39]. Due to the high metabolic rate and increased sensitivity to oxidative stress, the brain is especially dependent on proper mitochondrial function and an ideal oxidative balance [123]. For this reason, and the relative low levels of antioxidant defense systems, the brain is especially susceptible to oxidative damage [123,124]. Upon oxidative stress in the brain, NRF2 becomes activated to increase the expression of several antioxidant enzymes, and other small antioxidant molecules participate in detoxification of radical species. However, it seems that neurons from some regions of the brain, such as hippocampus, amygdala and prefrontal cortex, are more susceptible to oxidative damage [125].
Among other evidence, a decrease in the activity of α-ketoglutarate dehydrogenase (αKGDH), enzyme of the TCA cycle, has been described during aging. This reduction is related to higher oxidative stress [126] and to an increase in peroxidation of cardiolipin. This lipid, present in the internal mitochondrial membrane, is associated with the inhibition of the complex IV and the worsening of the respiratory chain function in the brain [[127], [128], [129]]. It is necessary to emphasize that there are differences among the different areas of the brain. In this sense, mitochondria of the hippocampus of rats appear to be more susceptible to the effect of age than other areas [130]. Although there are fewer studies in humans, some authors describe a decrease in the activity and protein levels of complex IV in different cerebral sub-regions [131,132], and, more recently, an increase in oxidative damage in mitochondria in the frontal and hippocampus cuts [133]. On the other hand, a deterioration in the capacity of learning and memory, as well as a decrease in the neurogenesis of the hippocampus, have been described with age, and this process could be also modulated by oxidative stress [134].
The brain also suffers a decrease in sexual hormone levels with age, as a result of the decrease in the peripheral synthesis and neurosteroidogenesis, and a more drastic pattern is exhibited in women than in men due to menopause [135]. A decrease in levels of E2, progesterone, testosterone and related metabolites has been described in 24-month-old male mice compared to 7-month-old mice [136], and an age-induced decrease in progesterone levels in mice has also been reported [137]. As mentioned before, mitochondria play an important role in steroid synthesis, so any alterations in mitochondrial function can also affect the levels of sex hormones.
In this regard, previous studies in our laboratory concluded that aged female rat brains had more differentiated mitochondria with greater functional capacity than male brains, and showed a better control of oxidative stress balance, which could be due, in part, to the neuroprotective effect of UCPs [138,139]. The ratio mitochondrial protein/DNA content decreased with aging, shifting towards worse mitochondrial functional capacity and increased mitochondria number [139]. The effect of aging was less marked in females, which accumulated less oxidative damage than males due in part to their greater antioxidant capacity, such as higher GPx activity and higher UCP5 levels. Furthermore, these sex differences gradually increased during aging [140].
In accordance with these studies, other authors have reported that women possess higher antioxidant defenses than men [141,142]. Moreover, in female ovariectomized rats, oxidative stress levels were comparable to those exhibited by males, and a treatment with E2 reverted the effect of the ovariectomy [27,141]. The drop in sex hormones has also been related to a shift to a ketogenic metabolism, with a marked reduction in COX activity and ATP production, and higher oxidative stress [143,144].
Epidemiological data suggest that sex hormones are also involved in the development of several brain pathologies, which will be further developed in the next section of this review. For instance, ischemic strokes occur more frequently among elderly people, and women have a higher risk of suffering this disease and, more concerningly, a poorer functional recovery than men. This trend is repeated in experimental animals, as older females show a more extensive loss of brain tissue than adult females [145].
6. Sex and phytoestrogens influence in the development of brain pathologies
Brain pathologies, especially neurodegenerative disorders, suppose a growing burden as population ages and show a sexual dimorphism in both their incidence and severity. As has been mentioned throughout this review, women seem to be protected at young ages from the development several brain diseases, although the incidence of these pathologies increases after menopause [2]. Moreover, phytoestrogen consumption has been thought to be a key modulator in oxidative stress in the brain and, therefore, to have important effects over brain pathologies development and progression. Twenty years ago, Gélinas and Martinoli demonstrated the protective effects of phytoestrogens on oxidative stress in rat neuronal cells PC12 [146]. More recently published in vivo studies have shown the positive effects of phytoestrogens on cognitive function and the improvement of mitochondrial functionality after phytoestrogen consumption in ovariectomized rats, decreasing the oxidative damage in these animals [147,148]. Moreover, there are clinical trials that relate the consumption of these compounds with a decrease in oxidative stress [149,150] and with a better cognitive ability and a decrease in the risk of suffering from neurodegenerative diseases such as Alzheimer's disease or dementia [[151], [152], [153]]. Table 1 summarizes the effects of the main phytoestrogens on different brain pathologies.
Table 1.
Most studied phytoestrogens, main sources and effects on different brain pathologies.
| Phytoestrogen | Main Sources | Disease | Effects | References |
|---|---|---|---|---|
| Genistein | Soy | Alzheimer disease | Reduces oxidative stress induced by β-amiloid accumulation and activates p38 | [166] |
| Amyotrophic lateral sclerosis | Inhibits the tyrosine kinase activity, avoiding the action of ROS mediators | [196] | ||
| Huntington's disease | Improves the disease symptoms and the inflammatory and oxidative profile and inhibits the cholinesterase activity | [204] | ||
| Stroke | Reduces the cell death of primary cortical neurons subjected to ischemic-like injury in vitro | [213] | ||
| Daidzein | Soy and red clover | Alzheimer disease | Improves the cognitive dysfunction and reduces oxidative stress | [167] |
| Parkinson's disease | Decreases the inflammation | [182] | ||
| Stroke | Reduces the cell death of primary cortical neurons subjected to ischemic-like injury in vitro | [213] | ||
| Brain cancer | Protective role against gliomagenesis | [217] | ||
| Resveratrol | Grapes, mulberries and peanuts | Alzheimer disease | Induces the expression of sirtuin 1 improving the mitochondrial function and reducing the oxidative stress | [168,169] |
| Quercetin | Apples, onions and tomatoes | Alzheimer disease | Restores the cognitive functions reducing the release of β-amiloid | [170] |
| Parkinson's disease | Reduces the expression of proinflammatory cytokines and superoxide anion production, provoking a decrease in the apoptosis ratio of neuronal cells | [[187], [188], [189]] | ||
| Coumestrol | Soy, alfalfa and clovers | Parkinson's disease | Decreases the inflammation | [182] |
| Formononetin | Legumes | Parkinson's disease | Inhibits neuroinflammation and increases ERβ protein expression improving mitochondrial function | [183] |
| Brain cancer | Restores mitochondrial function and inhibits cell signaling and invasion pathways | [233] | ||
| Epigallocatechin-3 gallate | Green tea and chocolate | Parkinson's disease | Reduces ROS production | [185] |
| Biochanin A | Lentils, red clover, alfalfa and cabbage | Stroke | Increases the expression of the glutamate oxaloacetate transaminase, which metabolizes the neurotoxic glutamate in the stroke-affected brain | [212] |
| Brain cancer | Restores mitochondrial function and inhibits cell signaling and invasion pathways | [218] |
6.1. Alzheimer's disease
Alzheimer's disease (AD) is the major neurodegenerative disease amongst the elderly population [154]. AD is characterized by a progressive cognitive loss, significantly affecting short-term memory, speech and reasoning [155]. This is the result of a mitochondrial dysfunction caused by the accumulation of amyloid-β (Aβ) plaques and intracellular fibrillary tangles formed by an abnormal phosphorylation of tau protein [156]. Epidemiological studies suggest a strong correlation between sex and the incidence of AD, since about two thirds of AD patients are women [157]. Furthermore, women manifest a faster cognitive decline and evolution of AD than men, probably due to an increased Aβ accumulation [157,158]. Thus, the drop in estrogens after menopause is recognized as a risk factor for developing AD, since this reduction is associated to lower mitochondrial function and higher oxidative stress [159], as explained before. Some studies also point out that the decrease in androgens could also be related to an increase in AD incidence in men as they age [[159], [160], [161]].
These observations have been also reported in animal models, and some authors suggest that the administration of estradiol may be a strategy to reduce AD incidence [29,162,163]. Noteworthy, SIRT1 and SIRT2 have been found elevated in lymphocytes from AD patients, as well as a reduction in UCP1 [164]. Furthermore, phytoestrogens have been shown to also improve cognitive function in animal and human studies, and they can offer protection against the development of AD. The relationship between the phytoestrogens present in soybeans, such as daidzein and especially genistein, and the improvement of memory and cognitive capabilities has been demonstrated in studies from 20 years ago [153,165]. A recent paper determined that pre-treatment with 0.5 μM of genistein is able to reduce hydrogen peroxide production in neurons treated with β-amiloid, which could suggest that this phytoestrogen would have a protector effect against oxidative stress induced by the β-amiloid accumulation [166]. This protective effects of genistein could be due to the ability of this phytoestrogen to activate p38, a protein involved in the β-amiloid cytotoxicity [166]. Likewise, daidzein also improves the cognitive dysfunction and reduces the oxidative stress in streptozotocin AD-induced rats, restoring antioxidant enzymes and glutathione levels and reducing oxidative damage [167]. Other phytoestrogens like resveratrol are able to induce the expression of sirtuin 1 [168], improving the mitochondrial function and, ultimately, reducing the oxidative stress in cells [169]. Moreover, quercetin has also positive effects restoring the cognitive functions, also reducing the release of β-amiloid in Alzheimer-model transgenic mice [170].
6.2. Parkinson's disease
Parkinson's disease (PD) is characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra, and the presence of Lewy bodies. This results in the consequent depletion of dopamine [171] and the development of motor symptoms that include tremor, bradykinesia and rigidity, and other non-motor manifestations such as cognitive decline [172]. One of the possible causes of the appearance of this disease is neuroinflammation, a process characterized by the activated microglia and the up-regulation of proinflammatory gene expression [173]. In the case of PD, men present two-fold higher incidence rates than women, which has been observed in human patients and animal models [172,174,175]. Furthermore, women tend to develop PD at a later age than men, with milder symptoms and slower disease progression [176,177]. In addition, a more marked mitochondrial dysfunction has been described in men with PD with respect to women [178], suggesting a neuroprotective role for sex hormones. In fact, testosterone and estradiol levels were found reduced in male PD patients compared to healthy individuals, and higher levels of these hormones could be associated to better cognitive ability in PD patients [179]. Interestingly, UCP4 and UCP2 have been associated to a better mitochondrial function and protection from some motor symptoms of PD in mice [180,181].
A study carried out by Jantaratnotai and collaborators in 2013 demonstrated that soy phytoestrogens, like genistein, daidzein and coumestrol, were able to decrease both the expression of genes related to the inflammation and the activation of signaling pathways associated to the inflammatory process [182], which could delay PD progression. Formononetin, a non-steroidal isoflavone, inhibits neuroinflammation and increases ERβ protein expression in BV2 mouse microglia cells [183]. In addition to neuroinflammation, a prolonged situation of oxidative stress may stimulate the development and progression of PD [184]. Catechins are a group of polyphenols which can be found in vegetal products like tea and chocolate. Among them, epigallocatechin-3 gallate stands out, since it has the potential to reduce ROS production, increasing antioxidant enzymes expression [185]. Quercetin has also shown anti-inflammatory and antioxidant effects [186]. This phytoestrogen is able to reduce the expression of proinflammatory cytokines and superoxide anion production in microglial N9 cells [187], suggesting that quercetin could modulate the expression and activity of the electron transport chain complexes, provoking a decrease in the apoptosis ratio of neuronal cells [188,189].
6.3. Amyotrophic lateral sclerosis
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that affects motor neurons of the cortex, brain stem and spinal cord, resulting in neuromuscular dysfunction and paralysis [190]. The causes of sporadic ALS are still unknown, but it is believed that a mix between environmental and genetic factors may be responsible. A redox disbalance seems to be involved in the pathogeny, since a reduction in SOD1 or TrxR1 activities is found in most patients with ALS [191,192]. This disease is more frequent in men than women, although the difference is lost at older ages, and men usually show different clinical manifestations than women [193].
In a mice model for ALS, females also show a slower progression of this disease, although ovariectomy abrogates this difference and E2 treatment reverses its effects [194]. Furthermore, a better mitochondrial function and lower oxidative stress were reported in female mice [195], which are related to the presence of E2. Finally, some phytoestrogens such as genistein are able to act like a neuroprotector agent against this disease, mainly for its estrogenic activity and also for its ability to inhibit the tyrosine kinase activity, avoiding the action of ROS mediators [196].
6.4. Huntington's disease
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expansion mutation in the huntingtin (Htt) gene. HD patients suffer a wide range of motor, cognitive and psychiatric symptoms [197]. In the case of HD, women show a poorer prognostic, since they usually present a faster progression and more severe symptoms than men [198,199]. Interestingly, E2 treatment, through the activation of ERα, induces the expression of Htt in a dose- and time-dependent manner [200], as well as globin neuroglobin (Ngb) expression [201]. The interaction between Htt and Ngb confers neurons protection from oxidative stress and apoptosis. However, in striatal neurons with mutated Htt, the effect of E2 is abolished, and Htt and Ngb interaction is lost, resulting in increased apoptosis [202]. Furthermore, some SIRTs, especially SIRT1, seem to have a role in the development of this disease, as their expression is increased in some areas of the brain of HD patients and are associated to metabolic changes [203].
On the other hand, genistein can revert the effects of the 3-Nitropropionic acid, which is a mitochondrial toxin able to mimic the symptoms of HD in rodents [204]. In this study, carried out by Menze and collaborators in 2015, several effects of genistein have been checked in ovariectomized rats, reverting the main symptoms of the disease, such as the locomotive improvement and the increased retention latencies in the passive avoidance task. Genistein treatment improved the inflammatory and oxidative profile of the animals and, moreover, inhibited the cholinesterase activity [204].
6.5. Stroke
Stroke is caused by a reduction in the cerebral blood flow, which results in cell death due to the lack of oxygen and nutrients. Stroke is the leading cause of death in developed countries, and the incidence is higher in men than women, although this difference is lost in postmenopausal women. In fact, in the 10 years following menopause, the risk of stroke doubles in women and the outcome tends to be poorer [205,206]. E2 has been reported to have a neuroprotective role in some animals, including humans, by improving learning, memory, secondary symptoms such as depression, and by reducing the recovery time after a stroke [[207], [208], [209]]. Some genetic variants of UCP and SIRT genes have been associated to the development of stroke [210]. Interestingly, Guo et al. [111] recently showed that E2 treatment stimulates both expression and activity of SIRT1, which stimulates AMPK activation and results in neuroprotection from stroke. Furthermore, the activation of ERs may improve the severity and the outcome in females after stroke [211].
In the pathophysiologic setting of cerebral ischemia, excitotoxic levels of glutamate contribute to neuronal cell death. The isoflavone biochanin A has the capacity to increase the expression of the glutamate oxaloacetate transaminase, which metabolizes the neurotoxic glutamate in the stroke-affected brain [212]. In 2010, Schreihofer and Redmond discovered that daidzein, its metabolite equol, and genistein, were able to reduce the cell death of primary cortical neurons subjected to ischemic-like injury in vitro, exerting this effect in an ER-dependent manner, more concretely the ER-kinase pathway [213].
6.6. Brain cancer
Brain cancer is a relatively rare disease compared to other cancer types, although it is the second more common cancer in children. Approximately 23700 new cases per year are reported, accounting for the 1.4% of all new cancer cases [214]. Epidemiologic studies support a sex difference for brain cancer incidence, as men are twice as likely to develop medulloblastoma, ependymoma, and gliomas [215]. Furthermore, a recent study reported a better outcome and survival for women than men, as they responded better to the standard therapy, and identified specific transcriptome signatures for glioblastomas [216].
Oxidative stress and inflammation are involved in brain cancer development and progression, and since phytoestrogens have antioxidant and anti-inflammatory activities, they could be potential candidates for brain cancer prevention. An epidemiologic study carried out in San Francisco Bay Area in 2006 demonstrated that the consumption of foods containing phytoestrogens, especially daidzein, seems to have a protective role against gliomagenesis [217]. Moreover, recent studies showed that the combination of the phytoestrogens formononetin or biochanin A and the cytotoxic treatment temozolomide enhanced the anticancer effect in glioblastoma multiforme cells, with greater inhibition of cell signaling and invasion pathways, and restoring the mitochondrial function [218,219].
6.7. Other brain diseases
Other brain diseases such as autism spectrum disorders [220], depression [221], multiple sclerosis [222], or traumatic brain injury [223,224], also show gender differences in incidence and/or outcome, suggesting the influence of sex hormones. Some phytoestrogens, especially isoflavones like genistein or daidzein, have been used for the treatment of psychiatric disorders, taking advantage of their estrogenic activity [225]. For instance, the consumption of soy phytoestrogens can affect the immune system activation through an ERα-dependent pathway [226], which could modulate seizure propensity in epileptic patients [227]. An isoflavone-rich diet, with high amounts of genistein, daidzein and equol, can reduce the anxious behavior in mice [228]. Moreover, an epidemiologic study revealed that a soy-supplemented diet decreased the depression and anxiety levels in postmenopausal women [229]. In the same way, it has been proved that genistein administration decreased the dopaminergic activity in schizophrenic rat models [230]. Despite the numerous described beneficial effects of phytoestrogens, especially related to their antioxidant and anti-inflammatory properties, there are studies which link phytoestrogen consumption in the childhood with the increment of autistic behaviors [231]. In addition, large doses of genistein administered chronically may induce cytotoxicity and apoptosis in the rat brain [232]. This highlights the need to further study both sexual hormones and phytoestrogens and understand the molecular mechanisms involved in their effects.
7. Concluding remarks
Sexual hormones play a crucial role in the brain regulating mitochondrial function and the antioxidant response. This way, sexual hormones have a strong influence on oxidative stress, as they can modulate key proteins involved in these processes, such as antioxidant enzymes, UCPs and SIRTs. Even though sexual hormones could explain, at least in part, the sexual dimorphism observed for some brain pathologies, and studies in animal models support the idea of E2 as a potential treatment, the data from clinical trials involving patients remains questionable. Differences between both sexes need to be addressed both in preclinical and clinical studies, as well as age-related changes, to elucidate whether E2 supplementation could be a potential treatment, and to establish dose, time and form of administration. Nevertheless, a deeper understanding of the effects of sexual hormones in different brain pathologies could yield potential gender- and age-specific therapeutic interventions. Furthermore, molecules mimicking the effect of sexual hormones, such as phytoestrogens, could hold promise as new neuroprotective strategies, as they could stimulate antioxidant defenses and improve mitochondrial function, reducing oxidative damage and its influence in the development of brain pathologies.
Funding source
The authors received no specific funding for this work.
Declaration of competing interest
None.
References
- 1.Cardoso S., Correia S., Carvalho C., Candeias E., Plácido A.I., Duarte A.I., Seiça R.M., Moreira P.I. Perspectives on mitochondrial uncoupling proteins-mediated neuroprotection. J. Bioenerg. Biomembr. 2015;47(2015):119–131. doi: 10.1007/s10863-014-9580-x. [DOI] [PubMed] [Google Scholar]
- 2.Ventura-Clapier R., Moulin M., Piquereau J., Lemaire C., Mericskay M., Veksler V., Garnier A. Mitochondria: a central target for sex differences in pathologies. Clin. Sci. 2017 doi: 10.1042/CS20160485. [DOI] [PubMed] [Google Scholar]
- 3.Pani G., Galeotti T., Chiarugi P. Metastasis: cancer cell's escape from oxidative stress. Canc. Metastasis Rev. 2010;29:351–378. doi: 10.1007/s10555-010-9225-4. [DOI] [PubMed] [Google Scholar]
- 4.Janssen-Heininger Y.M.W., Mossman B.T., Heintz N.H., Forman H.J., Kalyanaraman B., Finkel T., Stamler J.S., Rhee S.G., van der Vliet A. Redox-based regulation of signal transduction: principles, pitfalls, and promises. Free Radic. Biol. Med. 2008;45:1–17. doi: 10.1016/j.freeradbiomed.2008.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Laurent A., Nicco C., Chéreau C., Goulvestre C., Alexandre J., Alves A., Lévy E., Goldwasser F., Panis Y., Soubrane O., Weill B., Batteux F. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Canc. Res. 2005;65:948–956. [PubMed] [Google Scholar]
- 6.Galadari S., Rahman A., Pallichankandy S., Thayyullathil F. Reactive oxygen species and cancer paradox: to promote or to suppress? Free Radic. Biol. Med. 2017;104:144–164. doi: 10.1016/j.freeradbiomed.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 7.Dalla Pozza E., Fiorini C., Dando I., Menegazzi M., Sgarbossa A., Costanzo C., Palmieri M., Donadelli M. Role of mitochondrial uncoupling protein 2 in cancer cell resistance to gemcitabine. Biochim. Biophys. Acta Mol. Cell Res. 2012;1823:1856–1863. doi: 10.1016/j.bbamcr.2012.06.007. [DOI] [PubMed] [Google Scholar]
- 8.Baffy G., Derdak Z., Robson S.C. Mitochondrial recoupling: a novel therapeutic strategy for cancer? Br. J. Canc. 2011;105:469–474. doi: 10.1038/bjc.2011.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Verdin E., Hirschey M.D., Finley L.W.S., Haigis M.C. Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem. Sci. 2010;35:669–675. doi: 10.1016/j.tibs.2010.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lejri I., Grimm A., Eckert A. Mitochondria, estrogen and female brain aging. Front. Aging Neurosci. 2018;10:1–12. doi: 10.3389/fnagi.2018.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Demarest T.G., McCarthy M.M. Sex differences in mitochondrial (dys)function: implications for neuroprotection. J. Bioenerg. Biomembr. 2015;47:173–188. doi: 10.1007/s10863-014-9583-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Godar S.C., Bortolato M. Gene-sex interactions in schizophrenia: focus on dopamine neurotransmission. Front. Behav. Neurosci. 2014;8:71. doi: 10.3389/fnbeh.2014.00071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wickens M.M., Bangasser D.A., Briand L.A. Sex differences in psychiatric disease: a focus on the glutamate system. Front. Mol. Neurosci. 2018;11:197. doi: 10.3389/fnmol.2018.00197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ruszkiewicz J.A., Miranda-Vizuete A., Tinkov A.A., Skalnaya M.G., Skalny A.V., Tsatsakis A., Aschner M. Sex-specific differences in redox homeostasis in brain norm and disease. J. Mol. Neurosci. 2019;67:312–342. doi: 10.1007/s12031-018-1241-9. [DOI] [PubMed] [Google Scholar]
- 15.Ren X., Zou L., Zhang X., Branco V., Wang J., Carvalho C., Holmgren A., Lu J. Redox signaling mediated by thioredoxin and glutathione systems in the central nervous system. Antioxidants Redox Signal. 2017;27:989–1010. doi: 10.1089/ars.2016.6925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Salim S. Oxidative stress and the central nervous system. J. Pharmacol. Exp. Therapeut. 2016;360:201–205. doi: 10.1124/jpet.116.237503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Candeias E., Duarte A.I., Sebastião I., Fernandes M.A., Plácido A.I., Carvalho C., Correia S., Santos R.X., Seiça R., Santos M.S., Oliveira C.R., Moreira P.I. Middle-aged diabetic females and males present distinct susceptibility to alzheimer disease-like pathology. Mol. Neurobiol. 2017;54:6471–6489. doi: 10.1007/s12035-016-0155-1. [DOI] [PubMed] [Google Scholar]
- 18.Chakraborti A., Gulati K., Banerjee B.D., Ray A. Possible involvement of free radicals in the differential neurobehavioral responses to stress in male and female rats. Behav. Brain Res. 2007;179:321–325. doi: 10.1016/J.BBR.2007.02.018. [DOI] [PubMed] [Google Scholar]
- 19.Cole T.B., Coburn J., Dao K., Roqué P., Chang Y.-C., Kalia V., Guilarte T.R., Dziedzic J., Costa L.G. Sex and genetic differences in the effects of acute diesel exhaust exposure on inflammation and oxidative stress in mouse brain. Toxicology. 2016;374:1–9. doi: 10.1016/J.TOX.2016.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Guevara R., Santandreu F.M., Valle A., Gianotti M., Oliver J., Roca P. Sex-dependent differences in aged rat brain mitochondrial function and oxidative stress. Free Radic. Biol. Med. 2009;46:169–175. doi: 10.1016/j.freeradbiomed.2008.09.035. [DOI] [PubMed] [Google Scholar]
- 21.Katalinic V., Modun D., Music I., Boban M. Gender differences in antioxidant capacity of rat tissues determined by 2,2′-azinobis (3-ethylbenzothiazoline 6-sulfonate; ABTS) and ferric reducing antioxidant power (FRAP) assays. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2005;140:47–52. doi: 10.1016/J.CCA.2005.01.005. [DOI] [PubMed] [Google Scholar]
- 22.Silva T.L.A., Braz G.R.F., Silva S.C. de A., Pedroza A.A. da S., Freitas C. de M., Ferreira D.J.S., da Silva A.I., Lagranha C.J. Serotonin transporter inhibition during neonatal period induces sex-dependent effects on mitochondrial bioenergetics in the rat brainstem. Eur. J. Neurosci. 2018;48:1620–1634. doi: 10.1111/ejn.13971. [DOI] [PubMed] [Google Scholar]
- 23.Sobočanec S., Balog T., Kušić B., Šverko V., Šarić A., Marotti T. Differential response to lipid peroxidation in male and female mice with age: correlation of antioxidant enzymes matters. Biogerontology. 2008;9:335–343. doi: 10.1007/s10522-008-9145-7. [DOI] [PubMed] [Google Scholar]
- 24.Guevara R., Gianotti M., Roca P., Oliver J. Age and sex-related changes in rat brain mitochondrial function. Cell. Physiol. Biochem. 2011;27:201–206. doi: 10.1016/j.exger.2011.08.003. [DOI] [PubMed] [Google Scholar]
- 25.Jung M.E., Metzger D.B. A sex difference in oxidative stress and behavioral suppression induced by ethanol withdrawal in rats. Behav. Brain Res. 2016;314:199–214. doi: 10.1016/J.BBR.2016.07.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Uzun H., Kayali R., Çakatay U. The chance of gender dependency of oxidation of brain proteins in aged rats. Arch. Gerontol. Geriatr. 2010;50:16–19. doi: 10.1016/J.ARCHGER.2009.01.002. [DOI] [PubMed] [Google Scholar]
- 27.Borrás C., Sastre J., García-Sala D., Lloret A., Pallardó F.V., Viña J. Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males. Free Radic. Biol. Med. 2003;34:546–552. doi: 10.1016/s0891-5849(02)01356-4. [DOI] [PubMed] [Google Scholar]
- 28.Khalifa A.R.M., Abdel-Rahman E.A., Mahmoud A.M., Ali M.H., Noureldin M., Saber S.H., Mohsen M., Ali S.S. Sex-specific differences in mitochondria biogenesis, morphology, respiratory function, and ROS homeostasis in young mouse heart and brain. Phys. Rep. 2017;5:e13125. doi: 10.14814/phy2.13125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lloret A., Badía M.-C., Mora N.J., Ortega A., Pallardó F.V., Alonso M.-D., Atamna H., Viña J. Gender and age-dependent differences in the mitochondrial apoptogenic pathway in Alzheimer's disease. Free Radic. Biol. Med. 2008;44:2019–2025. doi: 10.1016/J.FREERADBIOMED.2008.02.017. [DOI] [PubMed] [Google Scholar]
- 30.Dkhil M.A., Al-Shaebi E.M., Lubbad M.Y., Al-Quraishy S. Impact of sex differences in brain response to infection with Plasmodium berghei. Parasitol. Res. 2016;115:415–422. doi: 10.1007/s00436-015-4803-6. [DOI] [PubMed] [Google Scholar]
- 31.Ehrenbrink G., Hakenhaar F.S., Salomon T.B., Petrucci A.P., Sandri M.R., Benfato M.S. Antioxidant enzymes activities and protein damage in rat brain of both sexes. Exp. Gerontol. 2006;41:368–371. doi: 10.1016/J.EXGER.2006.02.007. [DOI] [PubMed] [Google Scholar]
- 32.Krolow R., Noschang C.G., Arcego D., Andreazza A.C., Peres W., Gonçalves C.A., Dalmaz C. Consumption of a palatable diet by chronically stressed rats prevents effects on anxiety-like behavior but increases oxidative stress in a sex-specific manner. Appetite. 2010;55:108–116. doi: 10.1016/J.APPET.2010.03.013. [DOI] [PubMed] [Google Scholar]
- 33.Mármol F., Rodríguez C.A., Sánchez J., Chamizo V.D. Anti-oxidative effects produced by environmental enrichment in the hippocampus and cerebral cortex of male and female rats. Brain Res. 2015;1613:120–129. doi: 10.1016/J.BRAINRES.2015.04.007. [DOI] [PubMed] [Google Scholar]
- 34.Noschang C., Krolow R., Arcego D.M., Toniazzo A.P., Huffell A.P., Dalmaz C. Neonatal handling affects learning, reversal learning and antioxidant enzymes activities in a sex-specific manner in rats. Int. J. Dev. Neurosci. 2012;30:285–291. doi: 10.1016/J.IJDEVNEU.2012.01.010. [DOI] [PubMed] [Google Scholar]
- 35.Brocardo P.S., Boehme F., Patten A., Cox A., Gil-Mohapel J., Christie B.R. Anxiety- and depression-like behaviors are accompanied by an increase in oxidative stress in a rat model of fetal alcohol spectrum disorders: protective effects of voluntary physical exercise. Neuropharmacology. 2012;62:1607–1618. doi: 10.1016/J.NEUROPHARM.2011.10.006. [DOI] [PubMed] [Google Scholar]
- 36.Charradi K., Mahmoudi M., Bedhiafi T., Kadri S., Elkahoui S., Limam F., Aouani E. Dietary supplementation of grape seed and skin flour mitigates brain oxidative damage induced by a high-fat diet in rat: gender dependency. Biomed. Pharmacother. 2017;87:519–526. doi: 10.1016/J.BIOPHA.2017.01.015. [DOI] [PubMed] [Google Scholar]
- 37.Giménez-Llort L., García Y., Buccieri K., Revilla S., Suñol C., Cristofol R., Sanfeliu C. Gender-specific neuroimmunoendocrine response to treadmill exercise in 3xTg-AD mice. Int. J. Alzheimer's Dis. 2010;2010:128354. doi: 10.4061/2010/128354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Harish G., Venkateshappa C., Mahadevan A., Pruthi N., Srinivas Bharath M.M., Shankar S.K. Effect of premortem and postmortem factors on the distribution and preservation of antioxidant activities in the cytosol and synaptosomes of human brains. Biopreserv. Biobanking. 2012;10:253–265. doi: 10.1089/bio.2012.0001. [DOI] [PubMed] [Google Scholar]
- 39.Gaignard P., Liere P., Thérond P., Schumacher M., Slama A., Guennoun R. Role of sex hormones on brain mitochondrial function, with special reference to aging and neurodegenerative diseases. Front. Aging Neurosci. 2017;9:406. doi: 10.3389/fnagi.2017.00406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Reddy D.S., Bakshi K. Horm. Signal. Biol. Med. Elsevier Inc.; 2020. Neurosteroids: biosynthesis, molecular mechanisms, and neurophysiological functions in the human brain; pp. 69–82. [DOI] [Google Scholar]
- 41.Yilmaz C., Karali K., Fodelianaki G., Gravanis A., Chavakis T., Charalampopoulos I., Alexaki V.I. Neurosteroids as regulators of neuroinflammation. Front. Neuroendocrinol. 2019;55 doi: 10.1016/j.yfrne.2019.100788. [DOI] [PubMed] [Google Scholar]
- 42.Papadopoulos V., Miller W.L. Role of mitochondria in steroidogenesis. Best Pract. Res. Clin. Endocrinol. Metabol. 2012;26:771–790. doi: 10.1016/j.beem.2012.05.002. [DOI] [PubMed] [Google Scholar]
- 43.Mendell A.L., MacLusky N.J. Neurosteroid metabolites of gonadal steroid hormones in neuroprotection: implications for sex differences in neurodegenerative disease. Front. Mol. Neurosci. 2018;11:1–18. doi: 10.3389/fnmol.2018.00359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Engler-Chiurazzi E.B., Brown C.M., Povroznik J.M., Simpkins J.W. Estrogens as neuroprotectants: estrogenic actions in the context of cognitive aging and brain injury. Prog. Neurobiol. 2017;157:188–211. doi: 10.1016/J.PNEUROBIO.2015.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Liu M., Kelley M.H., Herson P.S., Hurn P.D. Neuroprotection of sex steroids. Minerva Endocrinol. 2010:127–143. [PMC free article] [PubMed] [Google Scholar]
- 46.Siddiqui A.N., Siddiqui N., Khan R.A., Kalam A., Jabir N.R., Kamal M.A., Firoz C.K., Tabrez S. Neuroprotective role of steroidal sex hormones: an overview. CNS Neurosci. Ther. 2016;22:342–350. doi: 10.1111/cns.12538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Spychala M.S., Honarpisheh P., McCullough L.D. Sex differences in neuroinflammation and neuroprotection in ischemic stroke. J. Neurosci. Res. 2017;95:462–471. doi: 10.1002/jnr.23962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Quillinan N., Deng G., Grewal H., Herson P.S. Androgens and stroke: good, bad or indifferent? Exp. Neurol. 2014;259:10–15. doi: 10.1016/J.EXPNEUROL.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Marino M., Galluzzo P., Ascenzi P. Estrogen signaling multiple pathways to impact gene transcription. Curr. Genom. 2006:497–508. doi: 10.2174/138920206779315737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Grimm S.L., Hartig S.M., Edwards D.P. Progesterone receptor signaling mechanisms. J. Mol. Biol. 2016;428:3831–3849. doi: 10.1016/J.JMB.2016.06.020. [DOI] [PubMed] [Google Scholar]
- 51.Mhaouty-Kodja S. Role of the androgen receptor in the central nervous system. Mol. Cell. Endocrinol. 2018;465:103–112. doi: 10.1016/J.MCE.2017.08.001. [DOI] [PubMed] [Google Scholar]
- 52.Dubal D.B., Zhu H., Yu J., Rau S.W., Shughrue P.J., Merchenthaler I., Kindy M.S., Wise P.M. Estrogen receptor alpha, not beta, is a critical link in estradiol-mediated protection against brain injury. Proc. Natl. Acad. Sci. Unit. States Am. 2001;98:1952–1957. doi: 10.1073/pnas.041483198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kushner P.J., Agard D.A., Greene G.L., Scanlan T.S., Shiau A.K., Uht R.M., Webb P. Estrogen receptor pathways to AP-1. J. Steroid Biochem. Mol. Biol. 2000:311–317. doi: 10.1016/s0960-0760(00)00108-4. [DOI] [PubMed] [Google Scholar]
- 54.Carlstrom L., Ke Z., Unnerstall J.R., Cohen R.S., Pandey S.C. Estrogen modulation of the cyclic AMP response element-binding protein pathway. Neuroendocrinology. 2001;74:227–243. doi: 10.1159/000054690. [DOI] [PubMed] [Google Scholar]
- 55.Kalaitzidis D., Gilmore T.D. Transcription factor cross-talk: the estrogen receptor and NF-κB. Trends Endocrinol. Metabol. 2005;16:46–52. doi: 10.1016/J.TEM.2005.01.004. [DOI] [PubMed] [Google Scholar]
- 56.Wu J., Williams D., Walter G.A., Thompson W.E., Sidell N. Estrogen increases Nrf2 activity through activation of the PI3K pathway in MCF-7 breast cancer cells. Exp. Cell Res. 2014;328:351–360. doi: 10.1016/J.YEXCR.2014.08.030. [DOI] [PubMed] [Google Scholar]
- 57.Razmara A., Duckles S.P., Krause D.N., Procaccio V. Estrogen suppresses brain mitochondrial oxidative stress in female and male rats. Brain Res. 2007;1176:71–81. doi: 10.1016/J.BRAINRES.2007.08.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen J.-Q., Cammarata P.R., Baines C.P., Yager J.D. Regulation of mitochondrial respiratory chain biogenesis by estrogens/estrogen receptors and physiological, pathological and pharmacological implications. Biochim. Biophys. Acta Mol. Cell Res. 2009;1793:1540–1570. doi: 10.1016/J.BBAMCR.2009.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Giguère V. Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocr. Rev. 2008;29:677–696. doi: 10.1210/er.2008-0017. [DOI] [PubMed] [Google Scholar]
- 60.St-Pierre J., Drori S., Uldry M., Silvaggi J.M., Rhee J., Jäger S., Handschin C., Zheng K., Lin J., Yang W., Simon D.K., Bachoo R., Spiegelman B.M. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell. 2006;127:397–408. doi: 10.1016/j.cell.2006.09.024. [DOI] [PubMed] [Google Scholar]
- 61.Kelly D.P., Scarpulla R.C. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 2004;18:357–368. doi: 10.1101/gad.1177604. [DOI] [PubMed] [Google Scholar]
- 62.Schreiber S.N., Knutti D., Brogli K., Uhlmann T., Kralli A. The transcriptional coactivator PGC-1 regulates the expression and activity of the orphan nuclear receptor estrogen-related receptor α (ERRα) J. Biol. Chem. 2003;278:9013–9018. doi: 10.1074/jbc.M212923200. [DOI] [PubMed] [Google Scholar]
- 63.Kurzer M.S., Xu X. Dietary phytoestrogens. Annu. Rev. Nutr. 1997;17:353–381. doi: 10.1146/annurev.nutr.17.1.353. [DOI] [PubMed] [Google Scholar]
- 64.Middleton E., Kandaswami C., Theoharides T.C. The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol. Rev. 2000 [PubMed] [Google Scholar]
- 65.May F.E.B. Novel drugs that target the estrogen-related receptor alpha: their therapeutic potential in breast cancer. Canc. Manag. Res. 2014;6:225–252. doi: 10.2147/CMAR.S35024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mootha V.K., Handschin C., Arlow D., Xie X., Pierre J. St, Sihag S., Yang W., Altshuler D., Puigserver P., Patterson N., Willy P.J., Schulman I.G., Heyman R.A., Lander E.S., Spiegelman B.M. Errα and Gabpa/b specify PGC-1α-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc. Natl. Acad. Sci. U. S. A. 2004;101:6570–6575. doi: 10.1073/pnas.0401401101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Echtay K.S., Bienengraeber M., Mayinger P., Heimpel S., Winkler E., Druhmann D., Frischmuth K., Kamp F., Huang S. Uncoupling proteins : martin Klingenberg ’ s contributions for 40 years. Arch. Biochem. Biophys. 2018;657:41–55. doi: 10.1016/j.abb.2018.09.006. [DOI] [PubMed] [Google Scholar]
- 68.Valle A., Oliver J., Roca P. Role of uncoupling proteins in cancer. Cancers. 2010;2 doi: 10.3390/cancers2020567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Klingenberg M. Mechanism and evolution of the uncoupling protein of brown adipose tissue Trends Biochem. Sci. 15 (3) (1990) 108–112. Trends Biochem. Sci. 1990;15:108–112. doi: 10.1016/0968-0004(90)90194-g. [DOI] [PubMed] [Google Scholar]
- 70.Gimeno R.E., Dembski M., Weng X., Deng N., Shyjan A.W., Gimeno C.J., Iris F., Ellis S.J., Woolf E.A., Tartaglia L.A. Cloning and characterization of an uncoupling protein homolog: a potential molecular mediator of human thermogenesis. Diabetes. 1997;46:900–906. doi: 10.2337/diab.46.5.900. [DOI] [PubMed] [Google Scholar]
- 71.Fleury C., Neverova M., Collins S., Raimbault S., Champigny O., Levi-Meyrueis C., Bouillaud F., Seldin M.F., Surwit R.S., Ricquier D., Warden C.H. Uncoupling protein-2: a novel gene linked to obesity and hyperinsulinemia. Nat. Genet. 1997;15:269–272. doi: 10.1038/ng0397-269. [DOI] [PubMed] [Google Scholar]
- 72.Gaudry M.J., Jastroch M. Neuroscience Letters Molecular evolution of uncoupling proteins and implications for brain function. Neurosci. Lett. 2019;696:140–145. doi: 10.1016/j.neulet.2018.12.027. [DOI] [PubMed] [Google Scholar]
- 73.Varela L., Schwartz M.L., Horvath T.L. Mitochondria controlled by UCP2 determine hypoxia-induced synaptic remodeling in the cortex and hippocampus. Neurobiol. Dis. 2016;90:68–74. doi: 10.1016/j.nbd.2016.01.004. [DOI] [PubMed] [Google Scholar]
- 74.Vidal-puig A., Solanes G., Grujic D., Flier J.S., Lowell B.B. vol. 82. 1997. pp. 79–82. (UCP3 : an Uncoupling Protein Homologue Expressed Preferentially and Abundantly in Skeletal Muscle and Brown Adipose Tissue). [DOI] [PubMed] [Google Scholar]
- 75.Boss O., Samec S., Paoloni-giacobino A., Rossier C., Dulloo A., Seydoux J., Muzzin P., Giacobino J. Uncoupling protein-3 : a new member of the mitochondrial carrier family with tissue-specific expression. FEBS Lett. 1997;408:39–42. doi: 10.1016/S0014-5793(97)00384-0. [DOI] [PubMed] [Google Scholar]
- 76.Zhao R.Z., Jiang S., Zhang L., Yu Z.B. Mitochondrial electron transport chain, ROS generation and uncoupling (Review) Int. J. Mol. Med. 2019;44:3–15. doi: 10.3892/ijmm.2019.4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Valle A., Guevara R., Garcia-Palmer F.J., Roca P., Oliver J. Sexual dimorphism in liver mitochondrial oxidative capacity is conserved under caloric restriction conditions. Am. J. Physiol. Cell Physiol. 2007;293:C1302–C1308. doi: 10.1152/ajpcell.00203.2007. 00203.2007[pii] [DOI] [PubMed] [Google Scholar]
- 78.Guevara R., Gianotti M., Oliver J., Roca P. Age and sex-related changes in rat brain mitochondrial oxidative status. Exp. Gerontol. 2011;46:923–928. doi: 10.1016/j.exger.2011.08.003. S0531-5565(11)00202-6[pii] [DOI] [PubMed] [Google Scholar]
- 79.Andrews Z.B., Liu Z.-W., Walllingford N., Erion D.M., Borok E., Friedman J.M., Tschöp M.H., Shanabrough M., Cline G., Shulman G.I., Coppola A., Gao X.-B., Horvath T.L., Diano S. UCP2 mediates ghrelin's action on NPY/AgRP neurons by lowering free radicals. Nature. 2008;454:846–851. doi: 10.1038/nature07181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Justo R., Frontera M., Pujol E., Rodriguez-Cuenca S., Llado I., Garcia-Palmer F.J., Roca P., Gianotti M. Gender-related differences in morphology and thermogenic capacity of brown adipose tissue mitochondrial subpopulations. Life Sci. 2005;76:1147–1158. doi: 10.1016/j.lfs.2004.08.019. S0024-3205(04)00945-2[pii] [DOI] [PubMed] [Google Scholar]
- 82.Rodriguez A.M., Quevedo-Coli S., Roca P., Palou A. Sex-dependent dietary obesity, induction of UCPs, and leptin expression in rat adipose tissues. Obes. Res. 2001;9:579–588. doi: 10.1038/oby.2001.75. [DOI] [PubMed] [Google Scholar]
- 83.Rodriguez-Cuenca S., Pujol E., Justo R., Frontera M., Oliver J., Gianotti M., Roca P. Sex-dependent thermogenesis, differences in mitochondrial morphology and function, and adrenergic response in brown adipose tissue. J. Biol. Chem. 2002;277:42958–42963. doi: 10.1074/jbc.M207229200M207229200. [pii] [DOI] [PubMed] [Google Scholar]
- 84.Frontera M., Pujol E., Rodriguez-Cuenca S., Catala-Niell A., Roca P., Garcia-Palmer F.J., Gianotti M. Rat brown adipose tissue thermogenic features are altered during mid-pregnancy. Cell. Physiol. Biochem. 2005;15:203–210. doi: 10.1159/000086407. CPB2005015005203[pii] [DOI] [PubMed] [Google Scholar]
- 85.Rodriguez A.M., Roca P., Bonet M.L., Pico C., Oliver P., Palou A. Positive correlation of skeletal muscle UCP3 mRNA levels with overweight in male, but not in female, rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003;285:R880–R888. doi: 10.1152/ajpregu.00698.200200698.2002. [pii] [DOI] [PubMed] [Google Scholar]
- 86.Valle A., Santandreu F.M., Garcia-Palmer F.J., Roca P., Oliver J. The serum levels of 17beta-estradiol, progesterone and triiodothyronine correlate with brown adipose tissue thermogenic parameters during aging. Cell. Physiol. Biochem. 2008;22:337–346. doi: 10.1159/000149812. 000149812[pii] [DOI] [PubMed] [Google Scholar]
- 87.Sastre-Serra J., Valle A., Company M.M., Garau I., Oliver J., Roca P. Estrogen down-regulates uncoupling proteins and increases oxidative stress in breast cancer. Free Radic. Biol. Med. 2010;48:506–512. doi: 10.1016/j.freeradbiomed.2009.11.025. S0891-5849(09)00738-2[pii] [DOI] [PubMed] [Google Scholar]
- 88.Rodriguez A.M., Monjo M., Roca P., Palou A. Opposite actions of testosterone and progesterone on UCP1 mRNA expression in cultured brown adipocytes. Cell. Mol. Life Sci. 2002:1714–1723. doi: 10.1007/PL00012499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Miro A.M., Sastre-Serra J., Pons D.G., Valle A., Roca P., Oliver J. 17beta-Estradiol regulates oxidative stress in prostate cancer cell lines according to ERalpha/ERbeta ratio. J. Steroid Biochem. Mol. Biol. 2011;123:133–139. doi: 10.1016/j.jsbmb.2010.12.004. [DOI] [PubMed] [Google Scholar]
- 90.Sastre-Serra J., Nadal-Serrano M., Pons D.G., Valle A., Garau I., Garcia-Bonafe M., Oliver J., Roca P. The oxidative stress in breast tumors of postmenopausal women is ERalpha/ERbeta ratio dependent. Free Radic. Biol. Med. 2013;61C:11–17. doi: 10.1016/j.freeradbiomed.2013.03.005. [DOI] [PubMed] [Google Scholar]
- 91.Nadal-Serrano M., Sastre-Serra J., Pons D.G., Miro A.M., Oliver J., Roca P. The ERalpha/ERbeta ratio determines oxidative stress in breast cancer cell lines in response to 17beta-estradiol. J. Cell. Biochem. 2012;113:3178–3185. doi: 10.1002/jcb.24192. [DOI] [PubMed] [Google Scholar]
- 92.Nadal-Serrano M., Pons D.G., Sastre-Serra J., Blanquer-Rossello Mdel M., Roca P., Oliver J. Genistein modulates oxidative stress in breast cancer cell lines according to ERalpha/ERbeta ratio: effects on mitochondrial functionality, sirtuins, uncoupling protein 2 and antioxidant enzymes. Int. J. Biochem. Cell Biol. 2013;45:2045–2051. doi: 10.1016/j.biocel.2013.07.002. [DOI] [PubMed] [Google Scholar]
- 93.Roca P., Sastre-Serra J., Nadal-Serrano M., Pons D.G., Blanquer-Rossello M.D.M., Oliver J. Phytoestrogens and mitochondrial biogenesis in breast cancer. Influence of estrogen receptors ratio. Curr. Pharmaceut. Des. 2014:5594–5618. doi: 10.2174/1381612820666140306100709. [DOI] [PubMed] [Google Scholar]
- 94.Pons D.G., Nadal-Serrano M., Blanquer-Rossello M.M., Sastre-Serra J., Oliver J., Roca P. Genistein modulates proliferation and mitochondrial functionality in breast cancer cells depending on ERalpha/ERbeta ratio. J. Cell. Biochem. 2014;115:949–958. doi: 10.1002/jcb.24737. [DOI] [PubMed] [Google Scholar]
- 95.Satoh A., Imai S.I. Systemic regulation of mammalian ageing and longevity by brain sirtuins. Nat. Commun. 2014;5:1–11. doi: 10.1038/ncomms5211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Braidy N., Poljak A., Grant R., Jayasena T., Mansour H., Chan-Ling T., Smythe G., Sachdev P., Guillemin G.J. Differential expression of sirtuins in the aging rat brain. Front. Cell. Neurosci. 2015;9:1–16. doi: 10.3389/fncel.2015.00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Brenmoehl J., Hoeflich A. Dual control of mitochondrial biogenesis by sirtuin 1 and sirtuin 3. Mitochondrion. 2013;13:755–761. doi: 10.1016/J.MITO.2013.04.002. [DOI] [PubMed] [Google Scholar]
- 98.Torrens-Mas M., Oliver J., Roca P., Sastre-Serra J. SIRT3: oncogene and tumor suppressor in cancer. Cancers. 2017;9 doi: 10.3390/cancers9070090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Torrens-Mas M., Hernández-López R., Pons D.G., Roca P., Oliver J., Sastre-Serra J. Sirtuin 3 silencing impairs mitochondrial biogenesis and metabolism in colon cancer cells. Am. J. Physiol. Cell Physiol. 2019 doi: 10.1152/ajpcell.00112.2019. [DOI] [PubMed] [Google Scholar]
- 100.Lafontaine-Lacasse M., Richard D., Picard F. Effects of age and gender on Sirt 1 mRNA expressions in the hypothalamus of the mouse. Neurosci. Lett. 2010;480:1–3. doi: 10.1016/j.neulet.2010.01.008. [DOI] [PubMed] [Google Scholar]
- 101.Lee H.J., Yang S.J. Aging-related correlation between serum sirtuin 1 activities and basal metabolic rate in women, but not in men. Clin. Nutr. Res. 2017;6:18. doi: 10.7762/cnr.2017.6.1.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Massudi H., Grant R., Braidy N., Guest J., Farnsworth B., Guillemin G.J. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PloS One. 2012 doi: 10.1371/journal.pone.0042357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Moore R.L., Dai Y., Faller D.V. Sirtuin 1 (SIRT1) and steroid hormone receptor activity in cancer. J. Endocrinol. 2012;213:37–48. doi: 10.1530/JOE-11-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Elangovan S., Ramachandran S., Venkatesan N., Ananth S., Gnana-Prakasam J.P., Martin P.M., Browning D.D., Schoenlein P.V., Prasad P.D., Ganapathy V., Thangaraju M. SIRT1 is essential for oncogenic signaling by estrogen/estrogen receptor α in breast cancer. Canc. Res. 2011;71:6654–6664. doi: 10.1158/0008-5472.CAN-11-1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bayele H.K. A conserved mechanism of sirtuin signalling through steroid hormone receptors. Biosci. Rep. 2019;39:1–18. doi: 10.1042/BSR20193535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Moore R.L., Faller D.V. SIRT1 represses estrogen-signaling, ligand-independent ERα-mediated transcription, and cell proliferation in estrogen-responsive breast cells. J. Endocrinol. 2013;216:273–285. doi: 10.1530/JOE-12-0102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zheng Y., Hu Q., Manaenko A., Zhang Y., Peng Y., Xu L., Tang J., Tang J., Zhang J. 17β-Estradiol attenuates hematoma expansion through ERα/Sirt1/NF-κB pathway in hyperglycemic intracerebral hemorrhage mice. Stroke. 2015;46:485–491. doi: 10.2217/FON.09.6.Dendritic. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bendale D.S., Karpe P.A., Chhabra R., Shete S.P., Shah H., Tikoo K. 17-β Oestradiol prevents cardiovascular dysfunction in post-menopausal metabolic syndrome by affecting SIRT1/AMPK/H3 acetylation. Br. J. Pharmacol. 2013;170:779–795. doi: 10.1111/bph.12290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Liarte S., Alonso-Romero J.L., Nicolás F.J. SIRT1 and estrogen signaling cooperation for breast cancer onset and progression. Front. Endocrinol. 2018;9:1–9. doi: 10.3389/fendo.2018.00552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Khan M., Shah S.A., Kim M.O. 17β-Estradiol via SIRT1/acetyl-p53/NF-kB signaling pathway rescued postnatal rat brain against acute ethanol intoxication. Mol. Neurobiol. 2018;55:3067–3078. doi: 10.1007/s12035-017-0520-8. [DOI] [PubMed] [Google Scholar]
- 111.Guo J.M., Shu H., Wang L., Xu J.J., Niu X.C., Zhang L. SIRT1-dependent AMPK pathway in the protection of estrogen against ischemic brain injury. CNS Neurosci. Ther. 2017;23:360–369. doi: 10.1111/cns.12686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lee C.-H., Su S.-C., Chiang C.-F., Chien C.-Y., Hsu C.-C., Yu T.-Y., Huang S.-M., Shieh Y.-S., Kao H.-W., Tsai C.-S., Hung Y.-J., Lin C.-Y. Estrogen modulates vascular smooth muscle cell function through downregulation of SIRT1. Oncotarget. 2017;8:110039–110051. doi: 10.18632/oncotarget.22546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Santolla M.F., Avino S., Pellegrino M., De Francesco E.M., De Marco P., Lappano R., Vivacqua A., Cirillo F., Rigiracciolo D.C., Scarpelli A., Abonante S., Maggiolini M. SIRT1 is involved in oncogenic signaling mediated by GPER in breast cancer. Cell Death Dis. 2015;6 doi: 10.1038/cddis.2015.201. e1834-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ota H., Akishita M., Akiyoshi T., Kahyo T., Setou M., Ogawa S., Iijima K., Eto M., Ouchi Y. Testosterone deficiency accelerates neuronal and vascular aging of samp8 mice: protective role of enos and sirt1. PloS One. 2012;7:1–10. doi: 10.1371/journal.pone.0029598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Panza S., Santoro M., De Amicis F., Morelli C., Passarelli V., D'Aquila P., Giordano F., Cione E., Passarino G., Bellizzi D., Aquila S. Estradiol via estrogen receptor beta influences ROS levels through the transcriptional regulation of SIRT3 in human seminoma TCam-2 cells. Tumor Biol. 2017;39 doi: 10.1177/1010428317701642. 101042831770164. [DOI] [PubMed] [Google Scholar]
- 116.Oh J.Y., Choi G.E., Lee H.J., Jung Y.H., Chae C.W., Kim J.S., Lee C.K., Han H.J. 17β-Estradiol protects mesenchymal stem cells against high glucose-induced mitochondrial oxidants production via Nrf2/Sirt3/MnSOD signaling. Free Radic. Biol. Med. 2019;130:328–342. doi: 10.1016/j.freeradbiomed.2018.11.003. [DOI] [PubMed] [Google Scholar]
- 117.Kong X., Wang R., Xue Y., Liu X., Zhang H., Chen Y., Fang F., Chang Y. Sirtuin 3, a new target of PGC-1α, plays an important role in the suppression of ROS and mitochondrial biogenesis. PloS One. 2010;5 doi: 10.1371/journal.pone.0011707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Cho H.I., Seo M.J., Lee S.M. 2-Methoxyestradiol protects against ischemia/reperfusion injury in alcoholic fatty liver by enhancing sirtuin 1-mediated autophagy. Biochem. Pharmacol. 2017;131:40–51. doi: 10.1016/j.bcp.2017.02.008. [DOI] [PubMed] [Google Scholar]
- 119.Gorska-Ponikowska M., Kuban-Jankowska A., Eisler S.A., Perricone U., Lo Bosco G., Barone G., Nussberger S. 2-Methoxyestradiol affects mitochondrial biogenesis pathway and succinate dehydrogenase complex flavoprotein subunit a in osteosarcoma cancer cells. CANCER GENOMICS PROTEOMICS. 2018;15:73–89. doi: 10.21873/cgp.20067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Nadal-Serrano M., Pons D.G., Sastre-Serra J., Blanquer-Rosselló M. del M., Roca P., Oliver J. Genistein modulates oxidative stress in breast cancer cell lines according to ERα/ERβ ratio: effects on mitochondrial functionality, sirtuins, uncoupling protein 2 and antioxidant enzymes. Int. J. Biochem. Cell Biol. 2013;45:2045–2051. doi: 10.1016/j.biocel.2013.07.002. [DOI] [PubMed] [Google Scholar]
- 121.Blanquer-Rosselló M.M., Oliver J., Valle A., Roca P. Effect of xanthohumol and 8-prenylnaringenin on MCF-7 breast cancer cells oxidative stress and mitochondrial complexes expression. J. Cell. Biochem. 2013;114:2785–2794. doi: 10.1002/jcb.24627. [DOI] [PubMed] [Google Scholar]
- 122.Blanquer-Rosselló M. del M., Hernández-López R., Roca P., Oliver J., Valle A. Resveratrol induces mitochondrial respiration and apoptosis in SW620 colon cancer cells. Biochim. Biophys. Acta Gen. Subj. 2017;1891:431–440. doi: 10.1016/j.bbagen.2016.10.009. [DOI] [PubMed] [Google Scholar]
- 123.Kann O., Kovács R. Mitochondria and neuronal activity. Am. J. Physiol. Cell Physiol. 2007;292:C641–C657. doi: 10.1152/ajpcell.00222.2006. [DOI] [PubMed] [Google Scholar]
- 124.Poon H.F., Vaishnav R.A., V Getchell T., Getchell M.L., Butterfield D.A. Quantitative proteomics analysis of differential protein expression and oxidative modification of specific proteins in the brains of old mice. Neurobiol. Aging. 2006;27:1010–1019. doi: 10.1016/j.neurobiolaging.2005.05.006. [DOI] [PubMed] [Google Scholar]
- 125.Salim S. Oxidative stress and the central nervous system. J. Pharmacol. Exp. Therapeut. 2017;360:201–205. doi: 10.1124/jpet.116.237503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tretter L., Adam-Vizi V. Alpha-ketoglutarate dehydrogenase: a target and generator of oxidative stress. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2005;360:2335–2345. doi: 10.1098/rstb.2005.1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Petrosillo G., De Benedictis V., Ruggiero F.M., Paradies G. Decline in cytochrome c oxidase activity in rat-brain mitochondria with aging. Role of peroxidized cardiolipin and beneficial effect of melatonin. J. Bioenerg. Biomembr. 2013;45:431–440. doi: 10.1007/s10863-013-9505-0. [DOI] [PubMed] [Google Scholar]
- 128.Petrosillo G., Matera M., Casanova G., Ruggiero F.M., Paradies G. Mitochondrial dysfunction in rat brain with aging Involvement of complex I, reactive oxygen species and cardiolipin. Neurochem. Int. 2008;53:126–131. doi: 10.1016/j.neuint.2008.07.001. [DOI] [PubMed] [Google Scholar]
- 129.Lenaz G., Genova M.L. Structure and organization of mitochondrial respiratory complexes: a new understanding of an old subject. Antioxidants Redox Signal. 2010;12:961–1008. doi: 10.1089/ars.2009.2704. [DOI] [PubMed] [Google Scholar]
- 130.Pandya J.D., Royland J.E., MacPhail R.C., Sullivan P.G., Kodavanti P.R.S. Age- and brain region-specific differences in mitochondrial bioenergetics in Brown Norway rats. Neurobiol. Aging. 2016;42:25–34. doi: 10.1016/j.neurobiolaging.2016.02.027. [DOI] [PubMed] [Google Scholar]
- 131.Ojaimi J., Masters C.L., Opeskin K., McKelvie P., Byrne E. Mitochondrial respiratory chain activity in the human brain as a function of age. Mech. Ageing Dev. 1999;111:39–47. doi: 10.1016/s0047-6374(99)00071-8. [DOI] [PubMed] [Google Scholar]
- 132.D.A. Cottrell, E.L. Blakely, M.A. Johnson, P.G. Ince, G.M. Borthwick, D.M. Turnbull, Cytochrome c oxidase deficient cells accumulate in the hippocampus and choroid plexus with age., Neurobiol. Aging 22 (n.d.) 265–272. [DOI] [PubMed]
- 133.Venkateshappa C., Harish G., Mahadevan A., Srinivas Bharath M.M., Shankar S.K. Elevated oxidative stress and decreased antioxidant function in the human hippocampus and frontal cortex with increasing age: implications for neurodegeneration in Alzheimer's disease. Neurochem. Res. 2012;37:1601–1614. doi: 10.1007/s11064-012-0755-8. [DOI] [PubMed] [Google Scholar]
- 134.Berry A., Amrein I., Nötzli S., Lazic S.E., Bellisario V., Giorgio M., Pelicci P.G., Alleva E., Lipp H.-P., Cirulli F. Sustained hippocampal neurogenesis in females is amplified in P66(Shc-/-) mice: an animal model of healthy aging. Hippocampus. 2012;22:2249–2259. doi: 10.1002/hipo.22042. [DOI] [PubMed] [Google Scholar]
- 135.Feldman H.A., Longcope C., Derby C.A., Johannes C.B., Araujo A.B., Coviello A.D., Bremner W.J., McKinlay J.B. Age trends in the level of serum testosterone and other hormones in middle-aged men: longitudinal results from the Massachusetts Male Aging Study. J. Clin. Endocrinol. Metab. 2002 doi: 10.1210/jcem.87.2.8201. [DOI] [PubMed] [Google Scholar]
- 136.Caruso D., Barron A.M., Brown M.A., Abbiati F., Carrero P., Pike C.J., Garcia-Segura L.M., Melcangi R.C. Age-related changes in neuroactive steroid levels in 3xTg-AD mice. Neurobiol. Aging. 2013;34:1080–1089. doi: 10.1016/j.neurobiolaging.2012.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Gaignard P., Savouroux S., Liere P., Pianos A., Thérond P., Schumacher M., Slama A., Guennoun R. Effect of sex differences on brain mitochondrial function and its suppression by ovariectomy and in aged mice. Endocrinology. 2015;156:2893–2904. doi: 10.1210/en.2014-1913. [DOI] [PubMed] [Google Scholar]
- 138.Guevara R., Santandreu F.M., Valle A., Gianotti M., Oliver J., Roca P. Sex-dependent differences in aged rat brain mitochondrial function and oxidative stress. Free Radic. Biol. Med. 2009;46:169–175. doi: 10.1016/J.FREERADBIOMED.2008.09.035. [DOI] [PubMed] [Google Scholar]
- 139.Guevara R., Gianotti M., Roca P., Oliver J. Age and sex-related changes in rat brain mitochondrial function. Cell. Physiol. Biochem. 2011;27:201–206. doi: 10.1159/000327945. [DOI] [PubMed] [Google Scholar]
- 140.Guevara R., Gianotti M., Oliver J., Roca P. Age and sex-related changes in rat brain mitochondrial oxidative status. Exp. Gerontol. 2011;46:923–928. doi: 10.1016/J.EXGER.2011.08.003. [DOI] [PubMed] [Google Scholar]
- 141.Viña J., Borrás C. Women live longer than men: understanding molecular mechanisms offers opportunities to intervene by using estrogenic compounds. Antioxidants Redox Signal. 2010 doi: 10.1089/ars.2009.2952. [DOI] [PubMed] [Google Scholar]
- 142.Mandal P.K., Tripathi M., Sugunan S. Brain oxidative stress: detection and mapping of anti-oxidant marker “Glutathione” in different brain regions of healthy male/female, MCI and Alzheimer patients using non-invasive magnetic resonance spectroscopy. Biochem. Biophys. Res. Commun. 2012 doi: 10.1016/j.bbrc.2011.11.047. [DOI] [PubMed] [Google Scholar]
- 143.Yao J., Hamilton R.T., Cadenas E., Brinton R.D. Decline in mitochondrial bioenergetics and shift to ketogenic profile in brain during reproductive senescence. Biochim. Biophys. Acta Gen. Subj. 2010 doi: 10.1016/j.bbagen.2010.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Yin F., Yao J., Sancheti H., Feng T., Melcangi R.C., Morgan T.E., Finch C.E., Pike C.J., Mack W.J., Cadenas E., Brinton R.D. The perimenopausal aging transition in the female rat brain: decline in bioenergetic systems and synaptic plasticity. Neurobiol. Aging. 2015 doi: 10.1016/j.neurobiolaging.2015.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Chisholm N.C., Sohrabji F. Astrocytic response to cerebral ischemia is influenced by sex differences and impaired by aging. Neurobiol. Dis. 2016;85:245–253. doi: 10.1016/j.nbd.2015.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Gélinas S., Martinoli M.G. Neuroprotective effect of estradiol and phytoestrogens on MPP+-induced cytotoxicity in neuronal PC12 cells. J. Neurosci. Res. 2002 doi: 10.1002/jnr.10315. [DOI] [PubMed] [Google Scholar]
- 147.Dong Y., Wang Y., Liu Y., Yang N., Zuo P. Phytoestrogen α-Zearalanol ameliorates memory impairment and neuronal DNA oxidation in ovariectomized mice. Clinics. 2013;68:1255–1262. doi: 10.6061/clinics/2013(09)13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Moreira A.C., Silva A.M., Branco A.F., Baldeiras I., Pereira G.C., Seiça R., Santos M.S., Sardão V.A. Phytoestrogen coumestrol improves mitochondrial activity and decreases oxidative stress in the brain of ovariectomized Wistar-Han rats. J. Funct. Foods. 2017;34:329–339. doi: 10.1016/j.jff.2017.05.002. [DOI] [Google Scholar]
- 149.Celec P., Hodosy J., Pálffy R., Gardlík R., Halčák L., Ostatníková D. The short-term effects of soybean intake on oxidative and carbonyl stress in men and women. Molecules. 2013;18:5190–5200. doi: 10.3390/molecules18055190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Pusparini Yenny, Hidayat A. Effect of soy isoflavone supplementation on endothelial dysfunction and oxidative stress in equol-producing postmenopausal women. Endocr. Metab. Immune Disord. - Drug Targets. 2015;15:71–79. doi: 10.2174/1871530314666141202123309. [DOI] [PubMed] [Google Scholar]
- 151.Evans H.M., Howe P.R.C., Wong R.H.X. Clinical evaluation of effects of chronic resveratrol supplementation on cerebrovascular function, cognition, mood, physical function and general well-being in postmenopausal women—rationale and study design. Nutrients. 2016;8 doi: 10.3390/nu8030150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.File S.E., Hartley D.E., Elsabagh S., Duffy R., Wiseman H. Cognitive improvement after 6 weeks of soy supplements in postmenopausal women is limited to frontal lobe function. Menopause. 2005;12:193–201. doi: 10.1097/00042192-200512020-00014. [DOI] [PubMed] [Google Scholar]
- 153.Veldman C.M., Cantorna M.T., DeLuca H.F. Eating soya improves human memory. Psychopharmacology (Berlin) 2001 doi: 10.1007/s002130100845. [DOI] [PubMed] [Google Scholar]
- 154.Prince M., Wimo A., Guerchet M., Ali G., Wu Y., Prina M. 2015. The Global Impact of Dementia - an Analysis of Prevalence, Incidence, Cost and Trends. [Google Scholar]
- 155.Selkoe D.J. Alzheimer's disease: genes, proteins, and therapy. Physiol. Rev. 2001 doi: 10.1152/physrev.2001.81.2.741. [DOI] [PubMed] [Google Scholar]
- 156.Schmitt K., Grimm A., Kazmierczak A., Strosznajder J.B., Götz J., Eckert A. Insights into mitochondrial dysfunction: aging, amyloid-β, and tau–A deleterious trio. Antioxidants Redox Signal. 2011 doi: 10.1089/ars.2011.4400. [DOI] [PubMed] [Google Scholar]
- 157.Mielke M.M., Vemuri P., Rocca W.A. Clinical epidemiology of Alzheimer's disease: assessing sex and gender differences. Clin. Epidemiol. 2014 doi: 10.2147/CLEP.S37929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Corder E.H., Ghebremedhin E., Taylor M.G., Thal D.R., Ohm T.G., Braak H. The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism. Ann. N. Y. Acad. Sci. 2004 doi: 10.1196/annals.1297.005. [DOI] [PubMed] [Google Scholar]
- 159.Vest R.S., Pike C.J. Gender, sex steroid hormones, and Alzheimer's disease. Horm. Behav. 2013 doi: 10.1016/j.yhbeh.2012.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Pike C.J. Sex and the development of Alzheimer's disease. J. Neurosci. Res. 2017 doi: 10.1002/jnr.23827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Rosario E.R., Carroll J., Pike C.J. Testosterone regulation of Alzheimer-like neuropathology in male 3xTg-AD mice involves both estrogen and androgen pathways. Brain Res. 2010 doi: 10.1016/j.brainres.2010.08.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Amtul Z., Wang L., Westaway D., Rozmahel R.F. Neuroprotective mechanism conferred by 17beta-estradiol on the biochemical basis of Alzheimer's disease. Neuroscience. 2010 doi: 10.1016/j.neuroscience.2010.05.031. [DOI] [PubMed] [Google Scholar]
- 163.Yao J., Irwin R., Chen S., Hamilton R., Cadenas E., Brinton R.D. Ovarian hormone loss induces bioenergetic deficits and mitochondrial β-amyloid. Neurobiol. Aging. 2012 doi: 10.1016/j.neurobiolaging.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Cornelius C., Trovato Salinaro A., Scuto M., Fronte V., Cambria M.T., Pennisi M., Bella R., Milone P., Graziano A., Crupi R., Cuzzocrea S., Pennisi G., Calabrese V. Cellular stress response, sirtuins and UCP proteins in Alzheimer disease: role of vitagenes. Immun. Ageing. 2013;10 doi: 10.1186/1742-4933-10-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Kim H., Xu J., Su Y., Xia H., Li L., Peterson P., Murphy-Ullrich J., Barnes S. Actions of the soy phytoestrogen genistein in models of human chronic disease: potential involvement of transforming growth factor β. Biochem. Soc. Trans. 2005 doi: 10.1042/bst0290216. [DOI] [PubMed] [Google Scholar]
- 166.Viña J., Lloret A., Vallés S.L., Borrás C., Badía M.C., Pallardó F.V., Sastre J., Alonso M.D. Mitochondrial oxidant signalling in Alzheimer's disease. J. Alzheim. Dis. 2007 doi: 10.3233/JAD-2007-11205. [DOI] [PubMed] [Google Scholar]
- 167.Wei J., Yang F., Gong C., Shi X., Wang G. Protective effect of daidzein against streptozotocin-induced Alzheimer's disease via improving cognitive dysfunction and oxidative stress in rat model. J. Biochem. Mol. Toxicol. 2019 doi: 10.1002/jbt.22319. [DOI] [PubMed] [Google Scholar]
- 168.Baur J.A., Sinclair D.A. Therapeutic potential of resveratrol: the in vivo evidence. Nat. Rev. Drug Discov. 2006 doi: 10.1038/nrd2060. [DOI] [PubMed] [Google Scholar]
- 169.Stacchiotti A., Favero G., Rezzani R. Resveratrol - Adding Life to Years, Not Adding Years to Life. 2019. Resveratrol and SIRT1 activators for the treatment of aging and age-related diseases. [DOI] [Google Scholar]
- 170.Sabogal-Guáqueta A.M., Muñoz-Manco J.I., Ramírez-Pineda J.R., Lamprea-Rodriguez M., Osorio E., Cardona-Gómez G.P. The flavonoid quercetin ameliorates Alzheimer's disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer's disease model mice. Neuropharmacology. 2015 doi: 10.1016/j.neuropharm.2015.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Skaper S.D. The brain as a target for inflammatory processes and neuroprotective strategies. Ann. N. Y. Acad. Sci. 2007 doi: 10.1196/annals.1403.002. [DOI] [PubMed] [Google Scholar]
- 172.Georgiev D., Hamberg K., Hariz M., Forsgren L., Hariz G.M. Gender differences in Parkinson's disease: a clinical perspective. Acta Neurol. Scand. 2017 doi: 10.1111/ane.12796. [DOI] [PubMed] [Google Scholar]
- 173.Hunot S., Hirsch E.C. Neuroinflammatory processes in Parkinson's disease. Ann. Neurol. 2003 doi: 10.1002/ana.10481. [DOI] [PubMed] [Google Scholar]
- 174.Rodríguez-Navarro J.A., Solano R.M., Casarejos M.J., Gomez A., Perucho J., García De Yébenes J., Mena M.A. Gender differences and estrogen effects in parkin null mice. J. Neurochem. 2008 doi: 10.1111/j.1471-4159.2008.05569.x. [DOI] [PubMed] [Google Scholar]
- 175.Gillies G.E., Murray H.E., Dexter D., McArthur S. Sex dimorphisms in the neuroprotective effects of estrogen in an animal model of Parkinson's disease. Pharmacol. Biochem. Behav. 2004 doi: 10.1016/j.pbb.2004.04.022. [DOI] [PubMed] [Google Scholar]
- 176.Haaxma C.A., Bloem B.R., Borm G.F., Oyen W.J.G., Leenders K.L., Eshuis S., Booij J., Dluzen D.E., Horstink M.W.I.M. Gender differences in Parkinson's disease. J. Neurol. Neurosurg. Psychiatry. 2007 doi: 10.1136/jnnp.2006.103788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Miller I.N., Cronin-Golomb A. Gender differences in Parkinson's disease: clinical characteristics and cognition. Mov. Disord. 2010 doi: 10.1002/mds.23388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Weiduschat N., Mao X., Beal M.F., Nirenberg M.J., Shungu D.C., Henchcliffe C. Sex differences in cerebral energy metabolism in Parkinson's disease: a phosphorus magnetic resonance spectroscopic imaging study. Park. Relat. Disord. 2014;20:545–548. doi: 10.1016/j.parkreldis.2014.02.003. [DOI] [PubMed] [Google Scholar]
- 179.Nitkowska M., Tomasiuk R., Czyzyk M., Friedman A. Prolactin and sex hormones levels in males with Parkinson's disease. Acta Neurol. Scand. 2015;131:411–416. doi: 10.1111/ane.12334. [DOI] [PubMed] [Google Scholar]
- 180.Wu K., Liu J., Zhuang N., Wang T. UCP4A protects against mitochondrial dysfunction and degeneration in pink1/parkin models of Parkinson's disease. Faseb. J. 2014;28:5111–5121. doi: 10.1096/fj.14-255802. [DOI] [PubMed] [Google Scholar]
- 181.Conti B., Sugama S., Lucero J., Winsky-Sommerer R., Wirz S.A., Maher P., Andrews Z., Barr A.M., Morale M.C., Paneda C., Pemberton J., Gaidarova S., Behrens M.M., Beal F., Sanna P.P., Horvath T., Bartfai T. Uncoupling protein 2 protects dopaminergic neurons from acute 1,2,3,6-methyl-phenyl-tetrahydropyridine toxicity. J. Neurochem. 2005;93:493–501. doi: 10.1111/j.1471-4159.2005.03052.x. [DOI] [PubMed] [Google Scholar]
- 182.Jantaratnotai N., Utaisincharoen P., Sanvarinda P., Thampithak A., Sanvarinda Y. Phytoestrogens mediated anti-inflammatory effect through suppression of IRF-1 and pSTAT1 expressions in lipopolysaccharide-activated microglia. Int. Immunopharm. 2013 doi: 10.1016/j.intimp.2013.07.013. [DOI] [PubMed] [Google Scholar]
- 183.El-Bakoush A., Olajide O.A. Formononetin inhibits neuroinflammation and increases estrogen receptor beta (ERβ) protein expression in BV2 microglia. Int. Immunopharm. 2018 doi: 10.1016/j.intimp.2018.06.016. [DOI] [PubMed] [Google Scholar]
- 184.Blesa J., Trigo-Damas I., Quiroga-Varela A., Del Rey N.L.G. Parkinson's disease-associated mutations affect mitochondrial function. Mitochondrial Mech. Degener. Repair Park. Dis. 2016 doi: 10.1007/978-3-319-42139-1_7. [DOI] [Google Scholar]
- 185.Ye Q., Ye L., Xu X., Huang B., Zhang X., Zhu Y., Chen X. Epigallocatechin-3-gallate suppresses 1-methyl-4-phenyl-pyridine-induced oxidative stress in PC12 cells via the SIRT1/PGC-1α signaling pathway. BMC Compl. Alternative Med. 2012 doi: 10.1186/1472-6882-12-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.de Oliveira M.R., Nabavi S.M., Braidy N., Setzer W.N., Ahmed T., Nabavi S.F. Quercetin and the mitochondria: a mechanistic view. Biotechnol. Adv. 2016 doi: 10.1016/j.biotechadv.2015.12.014. [DOI] [PubMed] [Google Scholar]
- 187.Bournival J., Plouffe M., Renaud J., Provencher C., Martinoli M.-G. Quercetin and sesamin protect dopaminergic cells from MPP + -induced neuroinflammation in a microglial (N9)-neuronal (PC12) coculture system. Oxid. Med. Cell. Longev. 2012 doi: 10.1155/2012/921941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Karuppagounder S.S., Madathil S.K., Pandey M., Haobam R., Rajamma U., Mohanakumar K.P. Quercetin up-regulates mitochondrial complex-I activity to protect against programmed cell death in rotenone model of Parkinson's disease in rats. Neuroscience. 2013 doi: 10.1016/j.neuroscience.2013.01.032. [DOI] [PubMed] [Google Scholar]
- 189.Park S.E., Sapkota K., Choi J.H., Kim M.K., Kim Y.H., Kim K.M., Kim K.J., Oh H.N., Kim S.J., Kim S. Rutin from dendropanax morbifera leveille protects human dopaminergic cells against rotenone induced cell injury through inhibiting JNK and p38 MAPK signaling. Neurochem. Res. 2014 doi: 10.1007/s11064-014-1259-5. [DOI] [PubMed] [Google Scholar]
- 190.Hirano A. Neuropathology of ALS: an overview. Neurology. 2012 doi: 10.1212/wnl.47.4_suppl_2.63s. [DOI] [PubMed] [Google Scholar]
- 191.Robberecht W., Sapp P., Viaene M.K., Rosen D., McKenna‐Yasek D., Haines J., Horvitz R., Theys P., Brown R. Rapid communication: Cu/Zn superoxide dismutase activity in familial and sporadic amyotrophic lateral sclerosis. J. Neurochem. 1994 doi: 10.1046/j.1471-4159.1994.62010384.x. [DOI] [PubMed] [Google Scholar]
- 192.Mitchell J., Morris A., de Belleroche J. Thioredoxin reductase 1 haplotypes modify familial amyotrophic lateral sclerosis onset. Free Radic. Biol. Med. 2009 doi: 10.1016/j.freeradbiomed.2008.09.041. [DOI] [PubMed] [Google Scholar]
- 193.McCombe P.A., Henderson R.D. Effects of gender in amyotrophic lateral sclerosis. Gend. Med. 2010 doi: 10.1016/j.genm.2010.11.010. [DOI] [PubMed] [Google Scholar]
- 194.Il Choi C., Lee Y.D., Gwag B.J., Cho S.I., Kim S.S., Suh-Kim H. Effects of estrogen on lifespan and motor functions in female hSOD1 G93A transgenic mice. J. Neurol. Sci. 2008 doi: 10.1016/j.jns.2007.10.024. [DOI] [PubMed] [Google Scholar]
- 195.Cacabelos D., Ramírez-Núñez O., Granado-Serrano A.B., Torres P., Ayala V., Moiseeva V., Povedano M., Ferrer I., Pamplona R., Portero-Otin M., Boada J. Early and gender-specific differences in spinal cord mitochondrial function and oxidative stress markers in a mouse model of ALS. Acta Neuropathol. Commun. 2016 doi: 10.1186/s40478-015-0271-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Trieu V.N., Uckun F.M. Genistein is neuroprotective in murine models of familial amyotrophic lateral sclerosis and stroke. Biochem. Biophys. Res. Commun. 1999 doi: 10.1006/bbrc.1999.0577. [DOI] [PubMed] [Google Scholar]
- 197.McColgan P., Tabrizi S.J. Huntington's disease: a clinical review. Eur. J. Neurol. 2018 doi: 10.1111/ene.13413. [DOI] [PubMed] [Google Scholar]
- 198.Zielonka D., Marinus J., Roos R.A.C., De Michele G., Di Donato S., Putter H., Marcinkowski J., Squitieri F., Bentivoglio A.R., Landwehrmeyer G.B. The influence of gender on phenotype and disease progression in patients with Huntington's disease. Park. Relat. Disord. 2013 doi: 10.1016/j.parkreldis.2012.09.012. [DOI] [PubMed] [Google Scholar]
- 199.Dorner J.L., Miller B.R., Barton S.J., Brock T.J., Rebec G.V. Sex differences in behavior and striatal ascorbate release in the 140 CAG knock-in mouse model of Huntington's disease. Behav. Brain Res. 2007 doi: 10.1016/j.bbr.2006.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Nuzzo M.T., Fiocchetti M., Servadio M., Trezza V., Ascenzi P., Marino M. 17β-Estradiol modulates huntingtin levels in rat tissues and in human neuroblastoma cell line. Neurosci. Res. 2016;103:59–63. doi: 10.1016/j.neures.2015.07.013. [DOI] [PubMed] [Google Scholar]
- 201.Fiocchetti M., Ascenzi P., Marino M. Neuroprotective effects of 17β-estradiol rely on estrogen receptor membrane initiated signals. Front. Physiol. 2012 doi: 10.3389/fphys.2012.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nuzzo M., Marino M. Estrogen/Huntingtin: a novel pathway involved in neuroprotection. Neural Regen. Res. 2016 doi: 10.4103/1673-5374.179045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Baldo B., Gabery S., Soylu-Kucharz R., Cheong R.Y., Henningsen J.B., Englund E., McLean C., Kirik D., Halliday G. Petersén, SIRT1 is increased in affected brain regions and hypothalamic metabolic pathways are altered in Huntington disease. Neuropathol. Appl. Neurobiol. 2018 doi: 10.1111/nan.12514. 0–3. [DOI] [PubMed] [Google Scholar]
- 204.Menze E.T., Esmat A., Tadros M.G., Abdel-Naim A.B., Khalifa A.E. Genistein improves 3-NPA-induced memory impairment in ovariectomized rats: impact of its antioxidant, anti-inflammatory and acetylcholinesterase modulatory properties. PloS One. 2015 doi: 10.1371/journal.pone.0117223. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 205.Lisabeth L., Bushnell C. Stroke risk in women: the role of menopause and hormone therapy. Lancet Neurol. 2012 doi: 10.1016/S1474-4422(11)70269-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Sohrabji F., Okoreeh A., Panta A. Sex hormones and stroke: beyond estrogens. Horm. Behav. 2019;111:87–95. doi: 10.1016/j.yhbeh.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Carpenter R.S., Iwuchukwu I., Hinkson C.L., Reitz S., Lee W., Kukino A., Zhang A., Pike M.M., Ardelt A.A. High-dose estrogen treatment at reperfusion reduces lesion volume and accelerates recovery of sensorimotor function after experimental ischemic stroke. Brain Res. 2016 doi: 10.1016/j.brainres.2016.01.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Morán J., Perez-Basterrechea M., Garrido P., Díaz E., Alonso A., Otero J., Colado E., González C. Effects of estrogen and phytoestrogen treatment on an in vitro model of recurrent stroke on HT22 neuronal cell line. Cell. Mol. Neurobiol. 2017;37:405–416. doi: 10.1007/s10571-016-0372-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Su Q., Cheng Y., Jin K., Cheng J., Lin Y., Lin Z., Wang L., Shao B. Estrogen therapy increases BDNF expression and improves post-stroke depression in ovariectomy-treated rats. Exp. Ther. Med. 2016;12:1843–1848. doi: 10.3892/etm.2016.3531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Dong C., Della-Morte D., Wang L., Cabral D., Beecham A. Association of the sirtuin and mitochondrial uncoupling protein genes with carotid plaque. PloS One. 2011;6:27157. doi: 10.1371/journal.pone.0027157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Selvaraj U.M., Zuurbier K.R., Whoolery C.W., Plautz E.J., Chambliss K.L., Kong X., Zhang S., Kim S.H., Katzenellenbogen B.S., Katzenellenbogen J.A., Mineo C., Shaul P.W., Stowe A.M. Selective nonnuclear estrogen receptor activation decreases stroke severity and promotes functional recovery in female mice. Endocrinology. 2018;159:3848–3859. doi: 10.1210/en.2018-00600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Khanna S., Stewart R., Gnyawali S., Harris H., Balch M., Spieldenner J., Sen C.K., Rink C. Phytoestrogen isoflavone intervention to engage the neuroprotective effect of glutamate oxaloacetate transaminase against stroke. Faseb. J. 2017 doi: 10.1096/fj.201700353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Schreihofer D.A., Redmond L. Soy phytoestrogens are neuroprotective against stroke-like injury in vitro. Neuroscience. 2009 doi: 10.1016/j.neuroscience.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Torre L.A., Siegel R.L., Ward E.M., Jemal A. Global cancer incidence and mortality rates and trends--an update. Cancer Epidemiol. Biomark. Prev. 2016;25:16–27. doi: 10.1158/1055-9965.EPI-15-0578. [DOI] [PubMed] [Google Scholar]
- 215.Sun T., Plutynski A., Ward S., Rubin J.B. An integrative view on sex differences in brain tumors. Cell. Mol. Life Sci. 2015;72:3323–3342. doi: 10.1007/s00018-015-1930-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Yang W., Warrington N.M., Taylor S.J., Whitmire P., Carrasco E., Singleton K.W., Wu N., Lathia J.D., Berens M.E., Kim A.H., Barnholtz-sloan J.S., Swanson K.R., Luo J., Rubin J.B. Sex differences in GBM revealed by analysis of patient imaging, transcriptome, and survival data. Sci. Transl. Med. 2019;11 doi: 10.1126/scitranslmed.aao5253.Sex. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Tedeschi-Blok N., Lee M., Sison J.D., Miike R., Wrensch M. Inverse association of antioxidant and phytoestrogen nutrient intake with adult glioma in the San Francisco Bay Area: a case-control study. BMC Canc. 2006 doi: 10.1186/1471-2407-6-148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Desai V., Jain A., Shaghaghi H., Summer R., Lai J.C.K., Bhushan A. Combination of biochanin A and temozolomide impairs tumor growth by modulating cell metabolism in glioblastoma multiforme. Anticancer Res. 2019;39:57–66. doi: 10.21873/anticanres.13079. [DOI] [PubMed] [Google Scholar]
- 219.Zhang X., Ni Q., Wang Y., Fan H., Li Y. Synergistic anticancer effects of formononetin and temozolomide on glioma C6 cells. Biol. Pharm. Bull. 2018 doi: 10.1248/bpb.b18-00002. [DOI] [PubMed] [Google Scholar]
- 220.Rivet T.T., Matson J.L. Review of gender differences in core symptomatology in autism spectrum disorders. Res. Autism Spectr. Disord. 2011 doi: 10.1016/j.rasd.2010.12.003. [DOI] [Google Scholar]
- 221.Albert P.R. Why is depression more prevalent in women? J. Psychiatry Neurosci. 2015 doi: 10.1503/jpn.150205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Harbo H.F., Gold R., Tintora M. Sex and gender issues in multiple sclerosis. Ther. Adv. Neurol. Disord. 2013 doi: 10.1177/1756285613488434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Ng I., Lee K.K., Lim J.H.G., Wong H.B., Yan X.Y. Investigating gender differences in outcome following severe traumatic brain injury in a predominantly Asian population. Br. J. Neurosurg. 2006 doi: 10.1080/02688690600682259. [DOI] [PubMed] [Google Scholar]
- 224.Ley E.J., Short S.S., Liou D.Z., Singer M.B., Mirocha J., Melo N., Bukur M., Salim A. Gender impacts mortality after traumatic brain injury in teenagers. J. Trauma Acute Care Surg. 2013 doi: 10.1097/TA.0b013e31829d024f. [DOI] [PubMed] [Google Scholar]
- 225.Estrada-Camarena E., López-Rubalcava C., Valdés-Sustaita B., Azpilcueta-Morales G.S., González-Trujano E.M. A Multidiscip. Look Menopause. 2017. Use of phytoestrogens for the treatment of psychiatric symptoms associated with menopause transition. [DOI] [Google Scholar]
- 226.Curran E.M., Judy B.M., Newton L.G., Lubahn D.B., Rottinghaus G.E., Macdonald R.S., Franklin C., Estes D.M. Dietary soy phytoestrogens and ERα signalling modulate interferon gamma production in response to bacterial infection. Clin. Exp. Immunol. 2004 doi: 10.1111/j.1365-2249.2003.02368.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Ravizza T., Kostoula C., Vezzani A. Immunity activation in brain cells in epilepsy: mechanistic insights and pathological consequences. Neuropediatrics. 2013 doi: 10.1055/s-0033-1358601. [DOI] [PubMed] [Google Scholar]
- 228.Lephart E.D., Adlercreutz H., Lund T.D. Dietary soy phytoestrogen effects on brain structure and aromatase in Long-Evans rats. Neuroreport. 2001 doi: 10.1097/00001756-200111160-00015. [DOI] [PubMed] [Google Scholar]
- 229.Balk J.L., Whiteside D.A., Naus G., DeFerrari E., Roberts J.M. A pilot study of the effects of phytoestrogen supplementation on postmenopausal endometrium. J. Soc. Gynecol. Invest. 2002 doi: 10.1016/S1071-5576(02)00152-1. [DOI] [PubMed] [Google Scholar]
- 230.Suresh P., Raju A.B. Antidopaminergic effects of leucine and genistein on shizophrenic rat models. Neurosciences. 2013 [PubMed] [Google Scholar]
- 231.Westmark C.J. Soy infant formula and seizures in children with autism: a retrospective study. PloS One. 2014 doi: 10.1371/journal.pone.0080488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Choi E.J., Lee B.H. Evidence for genistein mediated cytotoxicity and apoptosis in rat brain. Life Sci. 2004 doi: 10.1016/j.lfs.2004.01.010. [DOI] [PubMed] [Google Scholar]
- 233.Wang G., Zhang D., Yang S., Wang Y., Tang Z., Fu X. Co-Administration of genistein with doxorubicin-loaded polypeptide nanoparticles weakens the metastasis of malignant prostate cancer by amplifying oxidative damage. Biomater. Sci. 2018;6:827–835. doi: 10.1039/c7bm01201b. [DOI] [PubMed] [Google Scholar]


