Next Article in Journal
MES SV40 Cells Are Sensitive to Lipopolysaccharide, Peptidoglycan, and Poly I:C Expressing IL-36 Cytokines
Next Article in Special Issue
Understanding In Vitro Pathways to Drug Discovery for TDP-43 Proteinopathies
Previous Article in Journal
Selecting for CRISPR-Edited Knock-In Cells
Previous Article in Special Issue
Towards a Mechanistic Model of Tau-Mediated Pathology in Tauopathies: What Can We Learn from Cell-Based In Vitro Assays?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia

1
Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
2
School of Nursing and Medicine, Emory University, Atlanta, GA 30322, USA
3
Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
4
Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11921; https://doi.org/10.3390/ijms231911921
Submission received: 29 August 2022 / Revised: 27 September 2022 / Accepted: 1 October 2022 / Published: 7 October 2022
(This article belongs to the Special Issue Molecular Mechanisms of Dementia)

Abstract

:
S-equol, a metabolite of soy isoflavone daidzein transformed by the gut microbiome, is the most biologically potent among all soy isoflavones and their metabolites. Soy isoflavones are phytoestrogens and exert their actions through estrogen receptor-β. Epidemiological studies in East Asia, where soy isoflavones are regularly consumed, show that dietary isoflavone intake is inversely associated with cognitive decline and dementia; however, randomized controlled trials of soy isoflavones in Western countries did not generally show their cognitive benefit. The discrepant results may be attributed to S-equol production capability; after consuming soy isoflavones, 40–70% of East Asians produce S-equol, whereas 20–30% of Westerners do. Recent observational and clinical studies in Japan show that S-equol but not soy isoflavones is inversely associated with multiple vascular pathologies, contributing to cognitive impairment and dementia, including arterial stiffness and white matter lesion volume. S-equol has better permeability to the blood–brain barrier than soy isoflavones, although their affinity to estrogen receptor-β is similar. S-equol is also the most potent antioxidant among all known soy isoflavones. Although S-equol is available as a dietary supplement, no long-term trials in humans have examined the effect of S-equol supplementation on arterial stiffness, cerebrovascular disease, cognitive decline, or dementia.

Graphical Abstract

1. Introduction

Nutrition is considered an important modifiable factor in cognitive dysfunction and Alzheimer’s disease and related dementias (ADRD). High adherence to a Mediterranean diet is associated with a reduced risk of global cognitive decline [1]. The MIND (Mediterranean DASH diet Intervention for Neurodegenerative Delay) diet is associated with slower cognitive decline [2]. Evidence suggests that certain nutrients, such as polyphenols and long-chain n-3 fatty acids, can benefit cognitive function due to their antioxidant, anti-inflammatory, and other properties [3,4,5,6,7]. One such class of polyphenolic compounds is soy isoflavones, of which two major constituents are daidzein and genistein. Soy isoflavones are structurally similar to estradiol [8]. While estradiol exerts its biological actions through binding to both estrogen receptor α (ERα) and estrogen receptor β (ERβ), soy isoflavones are ERβ-selective agonists [9]. Soy isoflavones possess antioxidant [10], anti-inflammatory [11], anti-atherosclerotic [12], and vasodilatory [13] properties, with associated health benefits, including those related to cognition [14].
Soy isoflavones are regularly consumed in Japan and other East Asian countries, where their dietary intake is 25–50 mg/day [14,15]. In contrast, consumption is minimal in the US and other Western countries, i.e., <2 mg/day [14,16]. Recent studies in Japan reported that a diet high in soy isoflavones is inversely associated with incident cognitive impairment [17] and dementia [18]. However, a randomized controlled trial (RCT) of soy isoflavones in the US, the Women’s Isoflavone Soy Health (WISH) Trial, showed no significant effect of soy isoflavones on global cognition [19]. This discrepancy between the results in Japan and the US may be due to the difference in S-equol-producing capability. S-equol, a metabolite of a soy isoflavone daidzein, transformed by the gut microbiome, is the most bioactive among all soy isoflavones and their metabolites [20]. After consuming soy isoflavones, 50–70% of Japanese convert daidzein to S-equol. Because dietary intake of soy isoflavones in the US is minuscule [14,16], the rate of S-equol producers is estimated from RCTs of soy isoflavones; 20–30% of individuals assigned to supplementation of soy isoflavones (ranging from 44 to 210 mg/day) produce S-equol [21,22,23,24,25,26]. The difference in the rate of equol producers between Asian and Western countries is believed to be due to the difference in the gut microbiome [27,28,29,30] but not genetics [31,32,33]. A subgroup analysis in WISH showed that S-equol producers had improved cognition compared to the control group [19]. A cross-sectional study in older adults in Japan showed that S-equol production was significantly inversely associated with mild cognitive impairment (MCI) [34]. These results suggest that S-equol may be the key factor for the reported cognitive benefit of soy isoflavones. Here, we provide a narrative review of the bioavailability of S-equol compared to daidzein and genistein, epidemiological and clinical evidence to support that S-equol has cognitive benefits, potential mechanisms of the beneficial effects of S-equol on cognition and neurodegeneration, ERβ and cognition, and neuroprotection by ERβ agonists.

2. Soy Isoflavones and Conversion of Daidzein into S-equol

Soy isoflavones are polyphenols and phytoestrogens, plant-derived compounds structurally similar to 17β-estradiol (Figure 1). Therefore, many biological actions of soy isoflavones are exerted through their binding to ERs. There are two subtypes of ERs: ERα, which is present mainly in the mammary gland, ovary, liver, and bone, and ERβ, which is found in the colon, adipose tissue, and immune system. ERα and ERβ are expressed in the central nervous and cardiovascular systems [8]. Activation of ERα is associated with proliferative responses in the mammary gland, e.g., breast cancer and uterus. While estradiol binds to ERα and ERβ, soy isoflavones preferentially bind to ERβ [9]. Thus, soy isoflavones could potentially have beneficial estrogen-like effects with reduced side effects [8].
Soy isoflavones in soybeans are present as glycosides, bound to a sugar molecule. The sugar molecule is hydrolyzed upon digestion or fermentation, leaving an isoflavone aglycone, e.g., daidzein and genistein. The isoflavone aglycones are more readily absorbed than glucosides in humans [14]. The conversion of daidzein into S-equol (Figure 1) takes place in the intestine by the action of the gut microbiome [35]. Several bacteria that transform daidzein into S-equol have been isolated from the feces of animals and humans. These bacteria are anaerobes, rod-shaped, and Gram positive. In vitro experiments have succeeded in producing S-equol from daidzein-rich soy germ using bacteria [36]. Thus, S-equol has become available as a nutraceutical [37].

3. Bioavailability of S-equol as Compared to Daidzein and Genistein

3.1. Bioavailability in the Circulation

S-equol has the highest bioavailability among all soy isoflavones and their metabolites [20,38]. Preclinical studies show that the affinity of S-equol to ERβ is 18-fold higher than its precursor, daidzein, and S-equol’s affinity is similar to genistein [39]. Unlike daidzein or genistein, S-equol undergoes little biotransformation in the circulation [40,41]. Moreover, the biological activity of S-equol is enhanced by its reduced binding to serum proteins [41]. Almost 50% of S-equol circulates in the free form compared to less than 20% for daidzein and genistein [41,42].

3.2. Permeability to the Gastrointestinal Tract and Blood–Brain Barrier

As described later, in vivo studies report that S-equol and soy isoflavones have beneficial effects on the central nervous and cardiovascular systems. The critical question is whether these compounds can cross the gastrointestinal (GI) tract and the blood–brain barrier (BBB). To answer this question, recently, Johnson et al. used the SwissADME and the high-throughput parallel artificial membrane permeability assay (PAMPA) to determine the permeability properties of S-equol, genistein, daidzein, and other polyphenols through the GI tract and BBB. The SwissADME is a computational predictive software that computes key physicochemical, pharmacokinetic, drug-like, and related parameters for one or multiple molecules [43]. PAMPA is a robust, versatile method for predicting the passive permeability of compounds through the GI tract and BBB [44]. PAMPA uses a combination of phospholipids specific to the tested membrane and a microfiber filter to simulate the biological membrane. SwissADME showed that S-equol, genistein, and daidzein have high gut absorbance and that S-equol and daidzein but not genistein have BBB permeability. PAMPA showed that S-equol has higher GI tract and BBB permeability than daidzein or genistein. Thus, S-equol exhibits high permeability in both the GI tract and BBB.

4. Evidence from Epidemiological and Clinical Studies Evaluating the Cognitive Benefits of S-equol

Recently, two well-designed prospective cohort studies in Japan reported that soy isoflavones and a diet rich in soy conferred cognitive benefits. The National Institute for Longevity Sciences—Longitudinal Study of Aging (NILS-LSA) followed a population-based cohort of 776 men and women (mean age of 68 at baseline) for 8 years [17]. Those with Mini-Mental State Examination (MMSE) scores ≤23 were excluded at baseline. During the mean follow-up of 8 years, MMSE was administered 5 times on average. Incident cognitive impairment was defined as an MMSE score ≤23. Multivariable-adjusted hazard ratio (HR) of incident cognitive impairment associated with one standard deviation (SD) increase in dietary intake of soy isoflavones was 0.55 (95% confidence interval (CI): 0.23, 0.93) in women and 0.89 (95% CI: 0.58, 1.37) in men. The Hisayama Study followed a population-based cohort of 1006 dementia-free men and women (mean age of 68 at baseline) for 16 years. During the follow-up, 271 subjects developed dementia. A diet rich in soy, soy isoflavones, and vegetables was significantly inversely associated with incident dementia (multivariable-adjusted HR in the highest quartile of intake was 0.66 (95% CI: 0.46, 0.95) compared to the lowest quartile (p for trend = 0.02) [18]. However, S-equol was not measured in either study.
A recent cross-sectional study in Japan of 152 subjects (mean age of 69) reported that S-equol producers had significantly higher cognitive scores and lower prevalence of MCI than non-producers and that equol-producing status was a significant determinant of MCI in the multivariable analysis [34]. Other studies in Asians (Singapore [45], Chinese [46], and Asian Americans [46]) reported no significant association of dietary intake of soy isoflavones with cognitive impairment. However, their dietary intake of soy isoflavones (3–9 mg/day) was lower in these studies than in Japan (25–50 mg/day) and their S-equol-producing status was not reported.
Since soy isoflavones are phytoestrogens, most RCTs of soy isoflavones on cognition have been conducted in women, especially post-menopausal women. Our recent systematic review and meta-analysis of 16 RCTs of soy isoflavones on cognition (total number of participants = 1384 (134 male and 1252 female), mean age of 60, dose of soy isoflavones ranging from 60 to 160 mg/day, median duration of intervention of 17 weeks) showed that supplementation of soy isoflavones significantly improved overall cognitive function (standard mean difference 0.19 (95% CI: 0.07–0.32)) and memory (standard mean difference 0.15 (95% CI: 0.03–0.26) in adults [47]. Subgroup analysis showed no statistically significant difference in the duration of the intervention (<6 vs. ≥6 months), dose (<100 vs. ≥100 mg/day), or age (<60 vs. ≥ 60 years). However, the number of participants in most of these RCTs was small (<100), with the duration of intervention less than 1 year. The WISH trial, which had the largest number of participants with the most extended intervention period in these 16 RCTs, did not show significant improvements in overall cognition [19].
The WISH trial, an RCT evaluating the impact of 91 mg/day of soy isoflavones for 2.5 years on cognition among 350 US women (mean age of 61), showed that soy isoflavones did not improve global cognition but improved visual memory [19]. The subgroup analysis by S-equol-producing status did show that only 26% were S-equol producers among women assigned to soy isoflavones and that global cognition marginally significantly improved in S-equol producers compared to the placebo group (mean standardized changes in global cognition (standard error), 0.65 (0.18) vs. 0.31 (0.08) for equol producers and placebo group, respectively, p = 0.08). These results suggest that S-equol rather than soy isoflavones slows cognitive decline.

5. Potential Protective Mechanisms of S-equol for Cognitive Decline and ADRD

While amyloid-β deposition and phosphorylated tau are major therapeutic targets being tested in recent RCTs [48] or developed [49] for Alzheimer’s disease (AD), vascular, inflammatory, immune, metabolic, and other factors are also involved in pathophysiology of AD/ADRD. Among these, vascular contributions to cognitive impairment and dementia (VCID) [50] are an emerging and promising approach to dementia prevention. This is further evidenced by the Systolic Blood Pressure Intervention Trial (SPRINT) MIND RCT, which reported that intensive blood pressure treatment lowered the risk for MCI and slowed the progression of white matter lesions (WML) [51,52]. Evidence shows that S-equol affects various factors related to ADRD, including arterial stiffness and WML.

5.1. Potential Effect of S-equol on Arterial Stiffness

Arterial stiffness is emerging as a high priority as a therapeutic target for AD/ADRD. Arterial stiffness significantly predicts cognitive decline [53,54,55,56,57,58] and incident dementia [59]. Arterial stiffness is also associated with multiple forms of dementia-related pathology, including WML volume, cerebral microbleeds, amyloid-β deposition in the brain, phosphorylated tau, and neuroinflammation in the cerebral spinal fluid [60,61,62,63]. We recently conducted a systematic review and meta-analysis of eight RCTs of soy isoflavones on arterial stiffness [64]. Arterial stiffness was assessed with carotid-femoral pulse wave velocity (PWV), brachial-ankle PWV, and Cardio Ankle Vascular Index (CAVI), all of which are currently widely used in clinical practice and research [58] and with other methods. Supplementation of soy isoflavones significantly improved arterial stiffness compared to placebo (standardized mean difference −0.33, 95% CI: −0.47, −0.19). Because of the short intervention period (24 h to 12 weeks) and the small number of participants in each study (n = 17 to 213), trials with longer intervention duration and a larger number of participants are imperative to determine whether supplementation of soy isoflavones improves arterial stiffness.
Three trials directly tested whether supplementation of S-equol improved arterial stiffness. Usui et al. conducted a crossover RCT of 10 mg/day of S-equol supplementation for 12 weeks among 54 middle-aged overweight or obese men and women with metabolic syndrome and on arterial stiffness by CAVI [32]. Supplementation of S-equol significantly improved arterial stiffness (change in CAVI: 0.1 ± 0.1 for placebo and −0.2 ± 0.1 for equol, p < 0.01). Yoshikata et al. conducted a single-arm study of 10 mg/day of S-equol supplementation for 12 months among 74 middle-aged women [65]. They assessed arterial stiffness by brachial-ankle PWV. They observed a significant reduction in arterial stiffness (1402 cm/s at baseline and 1376 after 12 months, p < 0.01). Yoshitaka et al. more recently conducted an RCT to study the effect of 10 mg/day of S-equol supplementation on arterial stiffness by brachial-ankle PWV for 3 months among 57 postmenopausal women [66]. They did not observe a significant difference in the change in brachial-ankle PWV between the intervention and control groups, likely due to the small sample size. However, after the 3-month intervention, they observed that significantly more women in the control group were found to have worse brachial-ankle PWV than those in the intervention group (26.7% vs. 7.3%, p < 0.04). Since arterial stiffness increases monotonically with age [67], these results suggest that supplementation of S-equol inhibits the progression of arterial stiffness. RCTs of S-equol and arterial stiffness with longer duration and a larger number of participants are warranted.
Major underlying mechanisms of arterial stiffness include pathological collagen production and elastin degradation in the arterial wall, endothelial dysfunction, oxidative stress, and inflammation [58]. Preclinical studies show that S-equol inhibits abnormal collagen synthesis and elastase, improves endothelial function, and has antioxidant and anti-inflammatory properties (Table 1). Dubey et al. reported that in human aortic smooth muscle cells, phytoestrogens, including S-equol, dose-dependently inhibited collagen synthesis, proliferation, and migration of smooth muscle cells [68]. This effect is partly mediated through estrogen receptors (ERs) and possibly the direct inhibition of mitogen-activated protein kinase (MAPK). Gopaul et al. reported that treatment with equol decreased the expression of the elastase enzyme by 35% in 8-week organotypic cell cultures of human dermal fibroblasts [69]. This effect is through ERβ. We recently conducted a systematic review on the potential cardioprotective effects of S-equol and showed that S-equol improves endothelial function and possesses antioxidant and anti-inflammatory properties in preclinical studies [70].

5.2. Inverse Association of S-equol with White Matter Lesions (WMLs)

WMLs are a significant risk factor for age-related cognitive decline and ADRD [78,79]. The SPRINT-MIND MRI sub-study reported that progression of WMLs was significantly slower in individuals treated with more aggressive blood pressure (BP) control, although the effect size is relatively small, i.e., a mean between-group difference for change in WML volume of −0.54 cm3 (95% CI: −0.87 to −0.20 cm3) [51,52]. We recently reported a longitudinal association of S-equol-producing status with WML% (WML volume divided by total brain volume) [80]. This study was nested within a prospective cohort study in Japan [81]. Normal cognition was defined as ≤1 abnormal test result over all domains of the neuropsychological test battery and 91 cognitively normal older adults (45 men and 46 women, mean age of 82) underwent structural brain MRI. Blood levels of S-equol and soy isoflavones were measured using serum samples collected and stored 6–9 years before the imaging study. Soy isoflavones were detected in the blood of all 91 participants and 45 participants (49%) were non-producers. We divided equol producers into a high or low category using the median. Compared to high equol producers, non-producers had >100% higher age- and sex-adjusted WML% (1.19% (95% CI: 0.97, 1.49), 0.89% (95% CI: 0.67, 1.17), and 0.58% (95% CI: 0.44, 0.72) for non-producers, low and high producers, respectively, p for trend <0.01). The significant inverse association remained even after additionally adjusting for hypertension, diabetes, dyslipidemia, apolipoprotein E4 (ApoE4), and years of education. Sex-stratified analysis showed that S-equol-producing status was significantly inversely associated with WML% in both sexes. Of note, serum levels of soy isoflavones were not significantly associated with WML%, suggesting that S-equol but not soy isoflavones may be a strong determinant of the progression of WML volume.
Studies consistently associate [62,82] higher arterial stiffness with WMLs in older adults, independent of BP and other cardiovascular risk factors, suggesting that the S-equol effect on arterial stiffness may be one potential mechanism. In addition, a recent meta-analysis in human studies reported a significant inverse association of WMLs with cerebral blood flow [83].
A preclinical study reported that equol increased cerebral blood flow and had vasorelaxant effects on the cerebral artery. Yu et al. investigated the impact of equol on cerebral blood flow and the underlying molecular mechanisms [84]. Single-dose intraperitoneal injection of equol (both S-equol and R-equol) in adult male Sprague-Dawley rats resulted in a significant dose-dependent increase in regional cerebral blood flow in the parietal lobe without significant changes in BP. They also showed that vasorelaxation induced by equol was endothelium independent through activation of large-conductance Ca2+ activated K+ channels in the vascular smooth muscle cells. Jackman et al. reported that using adult normotensive and hypertensive Sprague-Dawley rats, equol displayed vasorelaxant activity both in normotensive and hypertensive rats [85]. They also showed that vasorelaxant responses of equol were independent of the endothelium, nitric oxide synthase, or K+ channels. S-equol is reported to improve endothelial function in human endothelial cells of the umbilical vein [74,75,76,77], aorta [77], and pulmonary artery [86]. Still, no study has examined the effect of S-equol on brain microvascular endothelial cells [87].

5.3. Other Neuroprotective Properties of S-equol

5.3.1. Antioxidant Properties

Oxidative stress is generally caused by an imbalance between the production and accumulation of reactive oxygen species (ROS) in cells and tissues [88]. Mitochondria mainly produce ROS during both physiological and pathological conditions. ROS accumulation exceeds antioxidant defense and is referred to as oxidative stress [89]. The brain is vulnerable to oxidative stress due to high oxygen consumption, polyunsaturated fat-rich content, and high iron content. Oxidative stress is associated with various pathways leading to neurodegeneration and AD [90,91,92], including amyloid-β accumulation [93,94], mitochondrial dysfunction and apoptosis [93,94,95], cell membrane damage, protein damage [96], DNA damage [96], and neuroinflammation [95].
S-equol has the highest antioxidant properties among all soy isoflavones and their metabolites [20,38]. Antioxidant properties of S-equol are greater than vitamins C and E in in vitro studies [38,97]. Lie et al. showed in deoxycorticosterone acetate-salt-induced hypertensive rats treated with low- (10 mg/kg/BW) and high- (20 mg/kg/BW) S-equol for 4 weeks that S-equol improved both long- and short-term memory and that S-equol increased antioxidant activity in the brain by elevating the activities of superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx) levels and decreasing malondialdehyde (MDA) content and acetylcholinesterase (AChE) activity [98]. Ma et al. showed in male and ovariectomized female Sprague-Dawley rats fed with S-equol (250 ppm), genistein (500 ppm), or isoflavone-reduced diet for two weeks before 90 min transient middle cerebral artery occlusion followed by reperfusion that both S-equol and genistein reduced infarct size and that S-equol and genistein reduced oxidase activity of nicotinamide adenine dinucleotide phosphate and superoxide levels in the rat brain [99]. Furthermore, S-equol reduced plasma thiobarbituric acid reactive substances, a by-product of lipid peroxidation and a biomarker of oxidative stress. Yu et al. examined whether S-equol confers neuroprotection against hypoxia injury by inhibiting the generation of ROS [100]. Using a rat pheochromocytoma cell line exposed to hypoxia injury, they showed that pretreatment with S-equol dose-dependently restored the cell viability and decreased MDA content and lactate dehydrogenase (LDH) activity.

5.3.2. Anti-Inflammatory Properties

Microglia and astrocytes are key regulators of the central nervous system inflammatory responses [101]. Microglial cells are principally responsible for immune defense in the central nervous system. Under pathological conditions, microglial cells are over-activated and produce a variety of proinflammatory cytokines, which can cause neuroinflammation and, subsequently, neurodegeneration. Astrocytes are active players in neuroinflammation and their response may be beneficial or detrimental depending on the stimuli offered by the inflamed milieu [102].
Subedi et al., using microglia and astrocyte cell lines, studied the anti-neuroinflammatory and neuroprotective effects of equol [103]. Equol (both S- and R-equol) inhibited the lipopolysaccharide (LPS)-induced activation of toll-like receptor 4 (TLR4), MAPK, nuclear factor kappa B (NF-κB)-mediated transcription of inflammatory mediators, production of nitric oxide (NO), secretion of tumor necrosis factor-α (TNFα), and interleukin 6 (IL-6). In addition, equol protects neurons from neuroinflammatory injury through the downregulation of neuronal apoptosis. Equol increased the production of nerve growth factors in astrocytes. Moriyama et al., using astrocytes stimulated by LPS, reported that S-equol attenuated LPS-induced increase in NO with a concomitant decrease in expression of inducible NO synthase [104]. This effect was partially mediated via the G protein-coupled receptor 30 on the cell surface. Lu et al. reported in an LPS-induced depression model in mice that S-equol significantly decreased the levels of pro-inflammatory cytokines (TNFα, IL-6, IL-1β) in the hippocampi [105]. In addition, S-equol significantly upregulated the expression of synaptic plasticity-related proteins and downregulated TLR4 and NF-κB signaling pathway in the hippocampi of LPS-treated mice.

5.3.3. Reduction in Amyloid-β Induced Neurotoxicity and Tau Phosphorylation

Major neuropathological hallmarks of AD involve protein aggregation as amyloid-β in extracellular plaques and phosphorylated tau in intracellular neurofibrillary tangles [106]. Several studies reported neuroprotective effects of genistein on amyloid-β-induced neurotoxicity and tau phosphorylation [107,108,109,110,111,112]. Compared to genistein, a few studies have investigated the effect of S-equol on amyloid-β-induced neurotoxicity and tau-phosphorylation. Using SH-SY5Y neuroblastoma cells exposed to amyloid-β, Tsai et al. examined the effect of S-equol and 17β-estradiol [113]. Exposure to amyloid-β reduced cell survival and caused downregulation of steroid receptor coactivator-1 (SRC-1) and extracellular signal-regulated kinase (ERK)1/2. Pretreatment with S-equol or 17β-estradiol reversed these effects of amyloid-β. ER agonists blocked the effects of both S-equol and 17β-estradiol. Using cultured hippocampal neurons, Zhao et al. investigated whether phytoestrogens, including equol and genistein, exert neuroprotective or neurotrophic effects [114]. They showed that following exposure to neurotoxic agents, including amyloid β25-35, phytoestrogens significantly reduced a marker of membrane damage but were ineffective in preventing a decline in neuronal mitochondrial viability. Hirohata et al. investigated the anti-fibrillization, anti-oligomerization, and fibril-destabilizing effects of genistein, glycitein, and equol on Aβ1-40 and Aβ1-42 levels in vitro using nucleation-dependent polymerization monitored by thioflavin T fluorescence, atomic force microscopy, electron microscopy, and photo-induced cross-linking [115]. They showed that glycitein interacted with Aβ monomers, oligomers, and fibrils, and the effect of equol was much weaker than glycitein. Using SH-SY5Y neuroblastoma cells cultured with amyloid-β and 1-methyl-4-phenyl-pyridine, Sorensen et al. investigated the effect of equol and genistein on the neurotoxicant-induced elevation of intracellular calcium [116]. The neurotoxicants caused an increase in Ca2+ and both equol and genistein attenuated the increase in Ca2+.
Since genistein and S-equol are not only structurally similar but also possess similar affinity to ERβ, effects and mechanisms of genistein on amyloid-β and tau phosphorylation may, to some extent, apply to those of S-equol. Okamura et al., using the human cell lines Daudi, Jurkat, U937, and K562, showed that genistein decreased the generation of amyloid-β by downregulation of the transmembrane protein presenilin 1, which is involved in the amyloid precursor protein (APP) cleavage [107]. Li et al., using female ob/ob and control mice fed with a standard diet or diet with genistein for 4 weeks, showed that genistein reduced amyloid-β deposition and the level of hyper-phosphorylated tau in the brain and that genistein increased the expression levels of the neurotrophic factors, nerve growth factor, and brain-derived neurotrophic factors [112]. Shentu et al., using HEK293-T cells co-infected with a cancerous inhibitor of protein phosphatase 2A (CIP2A) and APP plasmids or CIP2A and tau plasmids, investigated the inhibitory effects of genistein on APP/tau phosphorylation [111]. They showed that genistein significantly reduced APP/tau phosphorylation and amyloid-β production. Pierzynowska et al., using a streptozotocin-induced rat model of the sporadic form of AD, investigated the effect of genistein and showed that genistein degraded amyloid-β and phosphorylated tau protein in the brain [109]. Using retinoic-acid differentiated SH-SY5Y cells treated with amyloid-β25-35, Petry et al. examined the effect of genistein against amyloid-β-induced cell death and underlying mechanisms [108]. Genistein partially inhibited amyloid-β-induced cell death, primarily apoptosis, and the protective effect of genistein was associated with inhibition of amyloid-β-induced Akt inactivation and tau hyperphosphorylation. In another study using male Wistar rats with a bilateral intracerebroventricular infusion of amyloid-β1-42, Petry et al. investigated the effect of genistein on memory impairment and its underlying mechanisms [110]. Genistein improved amyloid-β-induced cognitive impairment by attenuating synaptotoxicity, hyperphosphorylation of tau, and inactivating ERK.

5.3.4. Other Reported Mechanisms

Ariyani et al. studied the effect of equol on cell differentiation and proliferation, using a mouse primary cerebellar culture, Neuro-2A clonal cells, and an astrocyte-enriched culture [117]. S-equol increased the dendrite arborization of Purkinje cells induced by triiodothyronine and the neurite growth of Neuro-2A cell differentiation. This effect was mediated through both ER and GPR30. On the other hand, in astrocytes, S-equol induced cell proliferation and migration, which was mediated through GPR30.
Using a non-contact co-culture model with LPS-BV2 conditioned media and human neuroblastoma SH-SY5Y cells, Johnson et al. demonstrated that S-equol had cytoprotective effects by decreasing LPS-BV2-conditioned media-induced cytotoxicity in SH-SY5Y cells and that S-equol had neuroprotective effects by reducing Parkinson’s disease-related neurotoxin (6-hydroxydopamine and 1-methyl-4-phenylpyridimium)-induced cytotoxicity in SH-SY5Y cells. [118]

6. ERβ and Cognition

ERβ is part of the steroid-activated nuclear receptor family of transcription factors and is present in several tissues throughout the body [119,120]. While ERβ density decreases with age, there is no change in estradiol affinity with aging. In contrast, ERα decreases in several receptors and is responsive with age [121]. ERβ is the predominant ER expressed in the brain. It is highly expressed in the hippocampus, cerebellum, and frontal cortex, with an essential role in learning and memory and promoting synaptic plasticity [122,123,124]. While ERα and ERβ mediate the effects of estradiol on normal brain function, ERβ has a more substantial influence on areas related to cognition [122,125].
Single nucleotide polymorphisms (SNPs) in the intragenic region of ESR2, the gene that encodes ERβ, are related to cognitive performance. Some minor alleles are associated with decreased memory and poor executive function, whereas major alleles have cognitive-domain-specific protection [126]. A study in women aged 54–55 found that ESR2 SNPs were associated with decreased semantic memory, as measured by the Boston Naming Test. The addition of the ApoE4 allele to the analyses demonstrated that the effects of ESR2 SNPs on executive function were dependent on the ApoE4 allele, where ESR2 SNP-related impairments in semantic memory were ApoE4 allele independent [126]. Further, ESR2 polymorphisms influence the incidence of AD [127]. Thus, through variations in ESR2 SNPs, ERβ plays a crucial role in neurocognitive health and increasing AD susceptibility.
ERβ activation improves performance in hippocampus-dependent memory tasks and increases recognition memory through activation of the pre-frontal cortex and hippocampal signaling pathways [123,128]. ERβ is implicated in transcriptional regulation of genes involved in the maintenance of hippocampal function with aging and expression of phosphorylated MAPK/ERK, which is implicated in AD development [129,130]. ERβ stimulation induces protein kinase B (Akt) and tropomyosin receptor kinase B (TrKB) expression, which are involved in hippocampal-dependent behaviors, such as memory and cognition [131]. Further, studies with ERβ knockout mice found impaired spatial learning and severely disrupted memory behaviors [132]. ERβ was found to play a role in cAMP response element-binding protein (CREB) expression and phosphorylation, a cellular transcription factor involved in learning and long-term memory [128,133]. In the cerebellum, ERβ plays a crucial role in motor learning by potentiating neuronal plasticity and synaptogenesis [134]. As ERβ promotes estrogen-mediated neuronal plasticity and memory function, selective targeting of ERβ may help maintain cognitive function and decrease the incidence of AD.

7. Neuroprotection by ERβ Agonists

ERs are intracellular transcription factors. Both ERα and ERβ are expressed in the brain and cardiovascular system [8]. ERα is predominantly present in the mammary gland, uterus, bone, and adipose tissue, while ERβ is mainly expressed in lung, prostate, immune cells, and adipose tissues.
ERβ has a minor role in the proliferation of the mammary gland or endometrium. Thus, selectively targeting ERβ is an attractive way to harness the beneficial effects of ER while reducing unwanted adverse effects. For this reason, pharmaceutical companies and academic institutes have developed ERβ agonists as potential treatment agents for cancer, cardiovascular disease (CVD), and neurological disorders, including ADRDs [135]. Table 2 summarizes ERβ agonists that have been developed for neuroprotection.
Diarylproprionitrile (DPN) was the first ERβ agonist to be discovered. DPN has a 70-fold ERβ selectivity over ERα in the binding assay. In preclinical studies, DPN improved maintenance of cognitive function, suppressed anxiety-like behaviors, and reduced neuroinflammation, pro-inflammatory cytokines, and neural cell death [136,137,138,139,140,141].
PhytoSERM is a combination of genistein, daidzein, and equol and has an 83-fold ERβ selectivity over ERα [145]. Preclinical studies show that PhytoSERM potentiated mitochondrial bioenergetics and increased the expression of mitochondrial antiapoptotic proteins and amyloid-β degrading enzymes [144,145]. In addition, in a female triple-transgenic mouse model of AD, 9-month treatment with PhytoSERM promoted physical health, improved spatial recognition memory, and attenuated amyloid-β deposition in the brain compared to controls [142]. Among ERβ agonists listed in Table 2, PhytoSERM is the only one tested in humans. An RCT of 50 mg/day, 100 mg/day, or placebo for 12 weeks among 71 women aged 45–60 years was conducted to assess the safety and tolerability of PhytoSERM [142]. A retrospective responder analysis of the trial showed that 50 mg/day of PhytoSERM significantly preserved cognitive function in areas of verbal learning and executive function [143]. An RCT of PhytoSERM among pre- and peri-menopausal women will soon start (R01 AG075122). This RCT aims to determine the efficacy of PhytoSERM to sustain glucose metabolism (because the menopausal transition is accompanied by reduction in cerebral metabolic rate of glucose, which correlates with progression of AD biomarkers later in life), on cognitive function and other outcomes.
Silibinin is an active milk thistle extract and is a selective ERβ agonist. Silibinin is used to treat toxic liver damage and is an adjunctive therapy for chronic hepatitis and cirrhosis [154]. Preclinical studies show that silibinin protects against amyloid-β-induced anxiety/depression-like behavior [147] and memory deficit [148]. EGX358 or ISP358-2 has a 750-fold selectivity for ERβ over ERα in a cell-based assay. Preclinical studies showed that EGX358 improved memory [150,151]. 8β-VE2 has a 183-fold ERβ selectivity in the transactivation assay [155]. Injection of 8β-VE2 in aromatase knockout mice prevented dopaminergic cell death in the medial preoptic area [152]. SERM-beta1 and SERM-beta2 are derivatives of tetrahydorfluorenose and have a 129-fold ERβ-selective ligand. Treatment with these agents for 4 days in ovariectomized rats showed an increase in the total number of cells in the dentate gyrus of the hippocampus [153].

8. Conclusions

It has long been hypothesized that S-equol producers, compared to non-producers, have more health benefits from consuming soy isoflavones [20], including alleviating postmenopausal symptoms, bone health, breast and prostate cancers, CVD, and cognitive decline. Some but not all observational studies have supported this hypothesis [30,35]. Inconsistent results in these observational studies may partly be due to changes in S-equol-producing status over time [156,157]. In contrast, although evidence from preclinical studies supports health benefits of S-equol, as reviewed above, the possibility cannot be ruled out that some phenotypes associated with S-equol producers bring these health benefits.
S-equol is available as a dietary supplement and seven RCTs of S-equol have been conducted and reported [32,66,158,159,160,161,162]. Almost all these trials investigated the effect of S-equol on menopausal-related symptoms with short durations of intervention (8 to 12 weeks). An RCT of S-equol supplementation for 24 months among 400 older adults without dementia will soon start (R01 AG074971). This trial will answer whether a 24-month supplementation of S-equol will significantly improve arterial stiffness, slow the progression of WML volume and cognitive decline, and whether S-equol itself or phenotypes associated with equol producers result in such benefits.

Author Contributions

A.S., W.W., B.B. and C.V.V. drafted the initial version of this review paper. J.G., J.L., S.G., A.F., M.L. and T.M.H., together with A.S., W.W., B.B. and C.V.V. revised and finalized the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This study is supported by the National Institute on Aging of the National Institutes of Health (R01 AG074971).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Coelho-Júnior, H.J.; Trichopoulou, A.; Panza, F. Cross-sectional and longitudinal associations between adherence to Mediterranean diet with physical performance and cognitive function in older adults: A systematic review and meta-analysis. Ageing Res. Rev. 2021, 70, 101395. [Google Scholar] [CrossRef]
  2. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Barnes, L.L.; Bennett, D.A.; Aggarwal, N.T. MIND diet slows cognitive decline with aging. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2015, 11, 1015–1022. [Google Scholar] [CrossRef] [Green Version]
  3. Scarmeas, N.; Anastasiou, C.A.; Yannakoulia, M. Nutrition and prevention of cognitive impairment. Lancet Neurol. 2018, 17, 1006–1015. [Google Scholar] [CrossRef]
  4. Chen, X.; Maguire, B.; Brodaty, H.; O’Leary, F. Dietary Patterns and Cognitive Health in Older Adults: A Systematic Review. J. Alzheimers Dis. 2019, 67, 583–619. [Google Scholar] [CrossRef] [PubMed]
  5. van den Brink, A.C.; Brouwer-Brolsma, E.M.; Berendsen, A.A.; van de Rest, O. The Mediterranean, Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Diets Are Associated with Less Cognitive Decline and a Lower Risk of Alzheimer’s Disease-A Review. Adv. Nutr. 2019, 10, 1040–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Solfrizzi, V.; . Custodero, C.; Lozupone, M.; Imbimbo, B.P.; Valiani, V.; Agosti, P.; Schilardi, A.; D’Introno, A.; La Montagna, M.; Calvani, M.; et al. Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer’s Disease and Late-Life Cognitive Disorders: A Systematic Review. J. Alzheimers Dis. 2017, 59, 815–849. [Google Scholar] [CrossRef] [Green Version]
  7. Morris, M.C. Nutrition and risk of dementia: Overview and methodological issues. Ann. N. Y. Acad. Sci. 2016, 1367, 31–37. [Google Scholar] [CrossRef] [Green Version]
  8. Paterni, I.; Granchi, C.; Katzenellenbogen, J.A.; Minutolo, F. Estrogen receptors alpha (ERα) and beta (ERβ): Subtype-selective ligands and clinical potential. Steroids 2014, 90, 13–29. [Google Scholar] [CrossRef] [Green Version]
  9. An, J.; Tzagarakis-Foster, C.; Scharschmidt, T.C.; Lomri, N.; Leitman, D.C. Estrogen receptor beta-selective transcriptional activity and recruitment of coregulators by phytoestrogens. J. Biol. Chem. 2001, 276, 17808–17814. [Google Scholar] [CrossRef] [Green Version]
  10. Ruiz-Larrea, M.B.; Mohan, A.R.; Paganga, G.; Miller, N.J.; Bolwell, G.P.; Rice-Evans, C.A. Antioxidant activity of phytoestrogenic isoflavones. Free Radic. Res. 1997, 26, 63–70. [Google Scholar] [CrossRef]
  11. Chacko, B.K.; Chandler, R.T.; Mundhekar, A.; Khoo, N.; Pruitt, H.M.; Kucik, D.F.; Parks, D.A.; Kevil, C.G.; Barnes, S.; Patel, R.P. Revealing anti-inflammatory mechanisms of soy isoflavones by flow: Modulation of leukocyte-endothelial cell interactions. Am. J. Physiol. —Heart Circ. Physiol. 2005, 289, H908–H915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Anthony, M.S.; Clarkson, T.B.; Bullock, B.C.; Wagner, J.D. Soy Protein Versus Soy Phytoestrogens in the Prevention of Diet-Induced Coronary Artery Atherosclerosis of Male Cynomolgus Monkeys. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 2524–2531. [Google Scholar] [CrossRef] [PubMed]
  13. Yamagata, K. Soy Isoflavones Inhibit Endothelial Cell Dysfunction and Prevent Cardiovascular Disease. J. Cardiovasc. Pharm. 2019, 74, 201–209. [Google Scholar] [CrossRef] [PubMed]
  14. Messina, M. Soy and Health Update: Evaluation of the Clinical and Epidemiologic Literature. Nutrients 2016, 8, 754. [Google Scholar] [CrossRef] [Green Version]
  15. Messina, M.; Nagata, C.; Wu, A.H. Estimated Asian adult soy protein and isoflavone intakes. Nutr. Cancer 2006, 55, 1–12. [Google Scholar] [CrossRef]
  16. Bai, W.; Wang, C.; Ren, C. Intakes of total and individual flavonoids by US adults. Int. J. Food Sci. Nutr. 2014, 65, 9–20. [Google Scholar] [CrossRef]
  17. Nakamoto, M.; Otsuka, R.; Nishita, Y.; Tange, C.; Tomida, M.; Kato, Y.; Imai, T.; Sakai, T.; Ando, F.; Shimokata, H. Soy food and isoflavone intake reduces the risk of cognitive impairment in elderly Japanese women. Eur. J. Clin. Nutr. 2018, 72, 1458–1462. [Google Scholar] [CrossRef]
  18. Ozawa, M.; Ninomiya, T.; Ohara, T.; Doi, Y.; Uchida, K.; Shirota, T.; Yonemoto, K.; Kitazono, T.; Kiyohara, Y. Dietary patterns and risk of dementia in an elderly Japanese population: The Hisayama Study. Am. J. Clin. Nutr. 2013, 97, 1076–1082. [Google Scholar] [CrossRef] [Green Version]
  19. Henderson, V.W.; St John, J.A.; Hodis, H.N.; Kono, N.; McCleary, C.A.; Franke, A.A.; Mack, W.J. Long-term soy isoflavone supplementation and cognition in women: A randomized, controlled trial. Neurology 2012, 78, 1841–1848. [Google Scholar] [CrossRef] [Green Version]
  20. Setchell, K.D.; Brown, N.M.; Lydeking-Olsen, E. The clinical importance of the metabolite sequel-a clue to the effectiveness of soy and its isoflavones. J. Nutr. 2002, 132, 3577–3584. [Google Scholar] [CrossRef]
  21. Hodis, H.N.; Mack, W.J.; Kono, N.; Azen, S.P.; Shoupe, D.; Hwang-Levine, J.; Petitti, D.; Whitfield-Maxwell, L.; Yan, M.; Franke, A.A.; et al. Isoflavone Soy Protein Supplementation and Atherosclerosis Progression in Healthy Postmenopausal Women. Stroke 2011, 42, 3168–3175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. A Greany, K.; A Nettleton, J.; E Wangen, K.; Thomas, W.; Kurzer, M.S. Consumption of isoflavone-rich soy protein does not alter homocysteine or markers of inflammation in postmenopausal women. Eur. J. Clin. Nutr. 2008, 62, 1419–1425. [Google Scholar] [CrossRef] [Green Version]
  23. Ma, Y.; Chiriboga, D.; Olendzki, B.C.; Nicolosi, R.; Merriam, P.A.; Ockene, I.S. Effect of soy protein containing isoflavones on blood lipids in moderately hypercholesterolemic adults: A randomized controlled trial. J. Am. Coll. Nutr. 2005, 24, 275–285. [Google Scholar] [CrossRef]
  24. West, S.G.; Hilpert, K.F.; Juturu, V.; Bordi, P.L.; Lampe, J.W.; Mousa, S.A.; Kris-Etherton, P.M. Effects of including soy protein in a blood cholesterol-lowering diet on markers of cardiac risk in men and in postmenopausal women with and without hormone replacement therapy. J. Womens Health 2005, 14, 253–262. [Google Scholar] [CrossRef] [PubMed]
  25. Pop, E.A.; Fischer, L.M.; Coan, A.D.; Gitzinger, M.; Nakamura, J.; Zeisel, S.H. Effects of a high daily dose of soy isoflavones on DNA damage, apoptosis, and estrogenic outcomes in healthy postmenopausal women: A phase I clinical trial. Menopause 2008, 15 Pt 1, 684–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gardner, C.D.; Messina, M.; Kiazand, A.; Morris, J.L.; Franke, A.A. Effect of two types of soy milk and dairy milk on plasma lipids in hypercholesterolemic adults: A randomized trial. J. Am. Coll. Nutr. 2007, 26, 669–677. [Google Scholar] [CrossRef]
  27. Setchell, K.D.; Clerici, C. Equol: History, chemistry, and formation. J. Nutr. 2010, 140, 1355S–1362S. [Google Scholar] [CrossRef] [Green Version]
  28. Setchell, K.D.R.; Cassidy, A. Dietary Isoflavones: Biological Effects and Relevance to Human Health. J. Nutr. 1999, 129, 758. [Google Scholar] [CrossRef] [Green Version]
  29. Jackson, R.L.; Greiwe, J.S.; Schwen, R.J. Emerging evidence of the health benefits of S-equol, an estrogen receptor beta agonist. Nutr. Rev. 2011, 69, 432–448. [Google Scholar] [CrossRef]
  30. Sekikawa, A.; Ihara, M.; Lopez, O.; Kakuta, C.; Lopresti, B.; Higashiyama, A.; Aizenstein, H.; Chang, Y.-F.; Mathis, C.; Miyamoto, Y.; et al. Effect of S-equol and Soy Isoflavones on Heart and Brain. Curr. Cardiol. Rev. 2019, 15, 114–135. [Google Scholar] [CrossRef] [PubMed]
  31. Nagata, C.; Ueno, T.; Uchiyama, S.; Nagao, Y.; Yamamoto, S.; Shibuya, C.; Kashiki, Y.; Shimizu, H. Dietary and lifestyle correlates of urinary excretion status of equol in Japanese women. Nutr. Cancer 2008, 60, 49–54. [Google Scholar] [CrossRef]
  32. Usui, T.; Tochiya, M.; Sasaki, Y.; Muranaka, K.; Yamakage, H.; Himeno, A.; Shimatsu, A.; Inaguma, A.; Ueno, T.; Uchiyama, S.; et al. Effects of natural S-equol supplements on overweight or obesity and metabolic syndrome in the Japanese, based on sex and equol status. Clin. Endocrinol. 2013, 78, 365–372. [Google Scholar] [CrossRef]
  33. Maskarinec, G.; Yamakawa, R.; Hebshi, S.; A Franke, A. Urinary isoflavonoid excretion and soy consumption in three generations of Japanese women in Hawaii. Eur. J. Clin. Nutr. 2007, 61, 255–261. [Google Scholar] [CrossRef] [Green Version]
  34. Igase, M.; Igase, K.; Tabara, Y.; Ohyagi, Y.; Kohara, K. Cross-sectional study of equol producer status and cognitive impairment in older adults. Geriatr. Gerontol. Int. 2017, 17, 2103–2108. [Google Scholar] [CrossRef] [PubMed]
  35. Mayo, B.; Vázquez, L.; Flórez, A.B. Equol: A Bacterial Metabolite from The Daidzein Isoflavone and Its Presumed Beneficial Health Effects. Nutrients 2019, 11, 2231. [Google Scholar] [CrossRef] [Green Version]
  36. Yuan, J.-P.; Wang, J.-H.; Liu, X. Metabolism of dietary soy isoflavones to equol by human intestinal microflora--implications for health. Mol. Nutr. Food Res. 2007, 51, 765–781. [Google Scholar] [CrossRef]
  37. Newton, K.M.; Reed, S.D.; Uchiyama, S.; Qu, C.; Ueno, T.; Iwashita, S.; Gunderson, G.; Fuller, S.; Lampe, J.W. A cross-sectional study of equol producer status and self-reported vasomotor symptoms. Menopause 2015, 22, 489–495. [Google Scholar] [CrossRef] [PubMed]
  38. Mitchell, J.H.; Gardner, P.T.; McPhail, D.B.; Morrice, P.C.; Collins, A.R.; Duthie, G.G. Antioxidant efficacy of phytoestrogens in chemical and biological model systems. Arch. Biochem. Biophys. 1998, 360, 142–148. [Google Scholar] [CrossRef]
  39. Muthyala, R.S.; Ju, Y.H.; Sheng, S.; Williams, L.D.; Doerge, D.R.; Katzenellenbogen, B.S.; Helferich, W.G.; A Katzenellenbogen, J. Equol, a natural estrogenic metabolite from soy isoflavones: Convenient preparation and resolution of R- and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorg. Med. Chem. 2004, 12, 1559–1567. [Google Scholar] [CrossRef]
  40. Setchell, K.D.R.; Zhao, X.; Shoaf, S.E.; Ragland, K. The pharmacokinetics of S-(-)equol administered as SE5-OH tablets to healthy postmenopausal women. J. Nutr. 2009, 139, 2037–2043. [Google Scholar] [CrossRef]
  41. Setchell, K.D.R.; Clerici, C. Equol: Pharmacokinetics and biological actions. J. Nutr. 2010, 140, 1363S–1368S. [Google Scholar] [PubMed] [Green Version]
  42. Yang, Z.; Kulkarni, K.; Zhu, W.; Hu, M. Bioavailability and pharmacokinetics of genistein: Mechanistic studies on its ADME. Anti-Cancer Agents Med. Chem. 2012, 12, 1264–1280. [Google Scholar] [CrossRef]
  43. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [Green Version]
  44. Bicker, J.; Alves, G.; Fortuna, A.; Soares-Da-Silva, P.; Falcão, A. A new PAMPA model using an in-house brain lipid extract for screening the blood-brain barrier permeability of drug candidates. Int. J. Pharm. 2016, 501, 102–111. [Google Scholar] [CrossRef]
  45. Talaei, M.; Feng, L.; Yuan, J.-M.; Pan, A.; Koh, W.-P. Dairy, soy, and calcium consumption and risk of cognitive impairment: The Singapore Chinese Health Study. Eur. J. Nutr. 2020, 59, 1541–1552. [Google Scholar] [CrossRef]
  46. Greendale, G.A.; Huang, M.H.; Leung, K.; Crawford, S.L.; Gold, E.B.; Wight, R.; Waetjen, E.; Karlamangla, A.S. Dietary phytoestrogen intakes and cognitive function during the menopausal transition: Results from the Study of Women’s Health Across the Nation Phytoestrogen Study. Menopause 2012, 19, 894–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Cui, C.; Birru, R.L.; E Snitz, B.; Ihara, M.; Kakuta, C.; Lopresti, B.J.; Aizenstein, H.J.; Lopez, O.L.; A Mathis, C.; Miyamoto, Y.; et al. Effects of soy isoflavones on cognitive function: A systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 2020, 78, 134–144. [Google Scholar] [CrossRef]
  48. Avgerinos, K.I.; Ferrucci, L.; Kapogiannis, D. Effects of monoclonal antibodies against amyloid-β on clinical and biomarker outcomes and adverse event risks: A systematic review and meta-analysis of phase III RCTs in Alzheimer’s disease. Ageing Res. Rev. 2021, 68, 101339. [Google Scholar] [CrossRef]
  49. Cummings, J.; Lee, G.; Zhong, K.; Fonseca, J.; Taghva, K. Alzheimer’s disease drug development pipeline: 2021. Alzheimers Dement. 2021, 7, e12179. [Google Scholar] [CrossRef] [PubMed]
  50. Zlokovic, B.V.; Gottesman, R.F.; Bernstein, K.E.; Seshadri, S.; McKee, A.; Snyder, H.; Greenberg, S.M.; Yaffe, K.; Schaffer, C.B.; Yuan, C.; et al. Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement. 2020, 16, 1714–1733. [Google Scholar] [CrossRef]
  51. The SPRINT MIND Investigators for the SPRINT Research Group; Nasrallah, I.M.; Pajewski, N.M.; Auchus, A.P.; Chelune, G.; Cheung, A.K.; Cleveland, M.L.; Coker, L.H.; Crowe, M.G.; Cushman, W.C.; et al. Association of Intensive vs Standard Blood Pressure Control With Cerebral White Matter Lesions. JAMA 2019, 322, 524–534. [Google Scholar] [CrossRef] [PubMed]
  52. Williamson, J.D.; Pajewski, N.M.; Auchus, A.P.; Bryan, R.N.; Chelune, G.; Cheung, A.K.; Cleveland, M.L.; Coker, L.H.; Crowe, M.G.; Cushman, W.C.; et al. Effect of Intensive vs Standard Blood Pressure Control on Probable Dementia: A Randomized Clinical Trial. JAMA 2019, 321, 553–561. [Google Scholar] [PubMed] [Green Version]
  53. Zeki Al Hazzouri, A.; Newman, A.B.; Simonsick, E.; Sink, K.M.; Sutton Tyrrell, K.; Watson, N.; Satterfield, S.; Harris, T.; Yaffe, K. Pulse wave velocity and cognitive decline in elders: The Health, Aging, and Body Composition study. Stroke 2013, 44, 388–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hajjar, I.; Goldstein, F.C.; Martin, G.S.; Quyyumi, A.A. Roles of Arterial Stiffness and Blood Pressure in Hypertension-Associated Cognitive Decline in Healthy Adults. Hypertension 2016, 67, 171–175. [Google Scholar] [CrossRef] [Green Version]
  55. Waldstein, S.R.; Rice, S.C.; Thayer, J.F.; Najjar, S.S.; Scuteri, A.; Zonderman, A.B. Pulse pressure and pulse wave velocity are related to cognitive decline in the Baltimore Longitudinal Study of Aging. Hypertension 2008, 51, 99–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Araghi, M.; Shipley, M.J.; Wilkinson, I.B.; McEniery, C.M.; Valencia-Hernández, C.A.; Kivimaki, M.; Sabia, S.; Singh-Manoux, A.; Brunner, E.J. Association of aortic stiffness with cognitive decline: Whitehall II longitudinal cohort study. Eur. J. Epidemiol. 2019, 35, 861–869. [Google Scholar] [CrossRef] [Green Version]
  57. Alvarez-Bueno, C.; Cunha, P.G.; Martinez-Vizcaino, V.; Pozuelo-Carrascosa, D.P.; Visier-Alfonso, M.E.; Jimenez-Lopez, E.; Cavero-Redondo, I. Arterial Stiffness and Cognition Among Adults: A Systematic Review and Meta-Analysis of Observational and Longitudinal Studies. J. Am. Heart Assoc. 2020, 9, e014621. [Google Scholar] [CrossRef] [PubMed]
  58. Chirinos, J.A.; Segers, P.; Hughes, T.; Townsend, R. Large-Artery Stiffness in Health and Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 74, 1237–1263. [Google Scholar] [CrossRef] [PubMed]
  59. Cui, C.; Sekikawa, A.; Kuller, L.H.; Lopez, O.L.; Newman, A.B.; Kuipers, A.L.; Mackey, R.H. Aortic Stiffness is Associated with Increased Risk of Incident Dementia in Older Adults. J. Alzheimers Dis. 2018, 66, 297–306. [Google Scholar] [CrossRef]
  60. Hughes, T.M.; Kuller, L.H.; Barinas-Mitchell, E.J.; Mackey, R.H.; McDade, E.M.; Klunk, W.E.; Aizenstein, H.J.; Cohen, A.D.; Snitz, B.E.; Mathis, C.A.; et al. Pulse wave velocity is associated with beta-amyloid deposition in the brains of very elderly adults. Neurology 2013, 81, 1711–1718. [Google Scholar] [CrossRef] [PubMed]
  61. Hughes, T.M.; Kuller, L.H.; Barinas-Mitchell, E.J.; McDade, E.M.; Klunk, W.E.; Cohen, A.D.; Mathis, C.A.; Dekosky, S.T.; Price, J.C.; Lopez, O.L. Arterial Stiffness and beta-Amyloid Progression in Nondemented Elderly Adults. JAMA Neurol. 2014, 71, 562–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hughes, T.M.; Wagenknecht, L.E.; Craft, S.; Mintz, A.; Heiss, G.; Palta, P.; Wong, D.; Zhou, Y.; Knopman, D.; Mosley, T.H.; et al. Arterial stiffness and dementia pathology: Atherosclerosis Risk in Communities (ARIC)-PET Study. Neurology 2018, 90, e1248–e1256. [Google Scholar] [CrossRef]
  63. Moore, E.E.; Liu, D.; Li, J.; Schimmel, S.J.; Cambronero, F.E.; Terry, J.G.; Nair, S.; Pechman, K.R.; Moore, M.E.; Bell, S.P.; et al. Association of Aortic Stiffness With Biomarkers of Neuroinflammation, Synaptic Dysfunction, and Neurodegeneration. Neurology 2021, 97, e329. [Google Scholar] [CrossRef]
  64. Man, B.; Cui, C.; Zhang, X.; Sugiyama, D.; Barinas-Mitchell, E.; Sekikawa, A. The effect of soy isoflavones on arterial stiffness: A systematic review and meta-analysis of randomized controlled trials. Eur. J. Nutr. 2020, 60, 603–614. [Google Scholar] [CrossRef] [PubMed]
  65. Yoshikata, R.; Myint, K.Z.Y.; Ohta, H. Effects of Equol Supplement on Bone and Cardiovascular Parameters in Middle-Aged Japanese Women: A Prospective Observational Study. J. Altern. Complement Med. 2018, 24, 701–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Yoshikata, R.; Myint, K.Z.Y.; Ohta, H.; Ishigaki, Y. Effects of an equol-containing supplement on advanced glycation end products, visceral fat and climacteric symptoms in postmenopausal women: A randomized controlled trial. PLoS ONE 2021, 16, e0257332. [Google Scholar] [CrossRef]
  67. AlGhatrif, M.; Strait, J.B.; Morrell, C.H.; Canepa, M.; Wright, J.; Elango, P.; Scuteri, A.; Najjar, S.S.; Ferrucci, L.; Lakatta, E.G. Longitudinal trajectories of arterial stiffness and the role of blood pressure: The Baltimore Longitudinal Study of Aging. Hypertension 2013, 62, 934–941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Dubey, R.K.; Gillespie, D.G.; Imthurn, B.; Rosselli, M.; Jackson, E.K.; Keller, P.J. Phytoestrogens inhibit growth and MAP kinase activity in human aortic smooth muscle cells. Hypertension 1999, 33 Pt 2, 177–182. [Google Scholar] [CrossRef] [Green Version]
  69. Gopaul, R.; Knaggs, H.E.; Lephart, E.D. Biochemical investigation and gene analysis of equol: A plant and soy-derived isoflavonoid with antiaging and antioxidant properties with potential human skin applications. Biofactors 2012, 38, 44–52. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, X.; Veliky, C.V.; Birru, R.L.; Barinas-Mitchell, E.; Magnani, J.W.; Sekikawa, A. Potential Protective Effects of Equol (Soy Isoflavone Metabolite) on Coronary Heart Diseases-From Molecular Mechanisms to Studies in Humans. Nutrients 2021, 13, 3739. [Google Scholar] [CrossRef]
  71. Iulita, M.F.; Noriega de la Colina, A.; Girouard, H. Arterial stiffness, cognitive impairment and dementia: Confounding factor or real risk? J. Neurochem. 2018, 144, 527–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Pase, M.P.; Beiser, A.; Himali, J.J.; Tsao, C.; Satizabal, C.L.; Vasan, R.S.; Seshadri, S.; Mitchell, G.F. Aortic Stiffness and the Risk of Incident Mild Cognitive Impairment and Dementia. Stroke 2016, 47, 2256–2261. [Google Scholar] [CrossRef] [Green Version]
  73. Poels, M.M.; van Oijen, M.; Mattace-Raso, F.U.; Hofman, A.; Koudstaal, P.J.; Witteman, J.C.; Breteler, M.M. Arterial stiffness, cognitive decline, and risk of dementia: The Rotterdam study. Stroke 2007, 38, 888–892. [Google Scholar] [CrossRef] [Green Version]
  74. Joy, S.; Siow, R.C.; Rowlands, D.J.; Becker, M.; Wyatt, A.W.; Aaronson, P.I.; Coen, C.W.; Kallo, I.; Jacob, R.; Mann, G.E. The isoflavone Equol mediates rapid vascular relaxation: Ca2+-independent activation of endothelial nitric-oxide synthase/Hsp90 involving ERK1/2 and Akt phosphorylation in human endothelial cells. J. Biol. Chem. 2006, 281, 27335–27345. [Google Scholar] [CrossRef] [Green Version]
  75. Rowlands, D.J.; Chapple, S.; Siow, R.C.; Mann, G.E. Equol-stimulated mitochondrial reactive oxygen species activate endothelial nitric oxide synthase and redox signaling in endothelial cells: Roles for F-actin and GPR30. Hypertension 2011, 57, 833–840. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, T.; Liang, X.; Shi, L.; Wang, L.; Chen, J.; Kang, C.; Zhu, J.; Mi, M. Estrogen receptor and PI3K/Akt signaling pathway involvement in S-(-)equol-induced activation of Nrf2/ARE in endothelial cells. PLoS ONE 2013, 8, e79075. [Google Scholar] [CrossRef] [Green Version]
  77. Zhang, T.; Hu, Q.; Shi, L.; Qin, L.; Zhang, Q.; Mi, M. Equol Attenuates Atherosclerosis in Apolipoprotein E-Deficient Mice by Inhibiting Endoplasmic Reticulum Stress via Activation of Nrf2 in Endothelial Cells. PLoS ONE 2016, 11, e0167020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Lopez, O.L.; Becker, J.T.; Chang, Y.; Klunk, W.E.; Mathis, C.; Price, J.; Aizenstein, H.J.; Snitz, B.; Cohen, A.D.; DeKosky, S.T.; et al. Amyloid deposition and brain structure as long-term predictors of MCI, dementia, and mortality. Neurology 2018, 90, e1920–e1928. [Google Scholar] [CrossRef]
  79. Alber, J.; Alladi, S.; Bae, H.; Barton, D.A.; Beckett, L.A.; Bell, J.M.; Berman, S.E.; Biessels, G.J.; Black, S.E.; Bos, I.; et al. White matter hyperintensities in vascular contributions to cognitive impairment and dementia (VCID): Knowledge gaps and opportunities. Alzheimers Dement. 2019, 5, 107–117. [Google Scholar] [CrossRef] [PubMed]
  80. Sekikawa, A.; Higashiyama, A.; Lopresti, B.J.; Ihara, M.; Aizenstein, H.; Watanabe, M.; Chang, Y.; Kakuta, C.; Yu, Z.; Mathis, C.; et al. Associations of equol-producing status with white matter lesion and amyloid-β deposition in cognitively normal elderly Japanese. Alzheimers Dement. 2020, 6, e12089. [Google Scholar] [CrossRef]
  81. Okamura, T.; Kokubo, Y.; Watanabe, M.; Higashiyama, A.; Miyamoto, Y.; Yoshimasa, Y.; Okayama, A. Low-density lipoprotein cholesterol and non-high-density lipoprotein cholesterol and the incidence of cardiovascular disease in an urban Japanese cohort study: The Suita study. Atherosclerosis 2009, 203, 587–592. [Google Scholar] [CrossRef]
  82. Zhai, F.-F.; Ye, Y.-C.; Chen, S.-Y.; Ding, F.-M.; Han, F.; Yang, X.-L.; Wang, Q.; Zhou, L.-X.; Ni, J.; Yao, M.; et al. Arterial Stiffness and Cerebral Small Vessel Disease. Front. Neurol. 2018, 9, 723. [Google Scholar] [CrossRef] [PubMed]
  83. Stewart, C.R.; Stringer, M.S.; Shi, Y.; Thrippleton, M.J.; Wardlaw, J.M. Associations Between White Matter Hyperintensity Burden, Cerebral Blood Flow and Transit Time in Small Vessel Disease: An Updated Meta-Analysis. Front. Neurol. 2021, 12, 647848. [Google Scholar] [CrossRef]
  84. Yu, W.; Wang, Y.; Song, Z.; Zhao, L.-M.; Li, G.-R.; Deng, X.-L. Equol increases cerebral blood flow in rats via activation of large-conductance Ca(2+)-activated K(+) channels in vascular smooth muscle cells. Pharmacol. Res. 2016, 107, 186–194. [Google Scholar] [CrossRef]
  85. Jackman, K.A.; Woodman, O.L.; Chrissobolis, S.; Sobey, C.G. Vasorelaxant and antioxidant activity of the isoflavone metabolite equol in carotid and cerebral arteries. Brain Res. 2007, 1141, 99–107. [Google Scholar] [CrossRef] [PubMed]
  86. Cheng, C.; Wang, X.; Weakley, S.M.; Kougias, P.; Lin, P.H.; Yao, Q.; Chen, C. The soybean isoflavonoid equol blocks ritonavir-induced endothelial dysfunction in porcine pulmonary arteries and human pulmonary artery endothelial cells. J. Nutr. 2010, 140, 12–17. [Google Scholar] [CrossRef] [Green Version]
  87. Stone, N.L.; England, T.J.; O’Sullivan, S.E. A Novel Transwell Blood Brain Barrier Model Using Primary Human Cells. Front. Cell. Neurosci. 2019, 13, 230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S. Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [Green Version]
  90. Scuderi, S.A.; Ardizzone, A.; Paterniti, I.; Esposito, E.; Campolo, M. Antioxidant and Anti-inflammatory Effect of Nrf2 Inducer Dimethyl Fumarate in Neurodegenerative Diseases. Antioxidants 2020, 9, 630. [Google Scholar] [CrossRef]
  91. Carvalho, C.; Moreira, P.I. Oxidative Stress: A Major Player in Cerebrovascular Alterations Associated to Neurodegenerative Events. Front. Physiol. 2018, 9, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Herman, F.; Westfall, S.; Brathwaite, J.; Pasinetti, G.M. Suppression of Presymptomatic Oxidative Stress and Inflammation in Neurodegeneration by Grape-Derived Polyphenols. Front. Pharmacol. 2018, 9, 867. [Google Scholar] [CrossRef]
  93. Guo, C.; Sun, L.; Chen, X.; Zhang, D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen. Res. 2013, 8, 2003–2014. [Google Scholar] [PubMed]
  94. Grimm, A.; Friedland, K.; Eckert, A. Mitochondrial dysfunction: The missing link between aging and sporadic Alzheimer’s disease. Biogerontology 2016, 17, 281–296. [Google Scholar] [CrossRef]
  95. Lim, J.-H.; Gerhart-Hines, Z.; Dominy, J.E.; Lee, Y.; Kim, S.; Tabata, M.; Xiang, Y.K.; Puigserver, P. Oleic acid stimulates complete oxidation of fatty acids through protein kinase A-dependent activation of SIRT1-PGC1α complex. J. Biol. Chem. 2013, 288, 7117–7126. [Google Scholar] [CrossRef] [Green Version]
  96. Tadokoro, K.; Ohta, Y.; Inufusa, H.; Loon, A.F.N.; Abe, K. Prevention of Cognitive Decline in Alzheimer’s Disease by Novel Antioxidative Supplements. Int. J. Mol. Sci. 2020, 21, 1974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Arora, A.; Nair, M.G.; Strasburg, G.M. Antioxidant activities of isoflavones and their biological metabolites in a liposomal system. Arch Biochem. Biophys. 1998, 356, 133–141. [Google Scholar] [CrossRef]
  98. Liu, T.-H.; Tsai, T.-Y. Effects of equol on deoxycorticosterone acetate salt-induced hypertension and associated vascular dementia in rats. Food Funct. 2016, 7, 3444–3457. [Google Scholar] [CrossRef]
  99. Ma, Y.; Sullivan, J.C.; Schreihofer, D.A. Dietary genistein and equol (4’, 7 isoflavandiol) reduce oxidative stress and protect rats against focal cerebral ischemia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 299, R871–R877. [Google Scholar] [CrossRef]
  100. Yu, W.; Deng, X.; Ma, Z.; Wang, Y. Equol protects PC12 neuronal cells against hypoxia/reoxygenation injury in vitro by reducing reactive oxygen species production. Nan Fang Yi Ke Da Xue Xue Bao 2016, 36, 1–7. [Google Scholar]
  101. Vainchtein, I.D.; Molofsky, A.V. Astrocytes and Microglia: In Sickness and in Health. Trends Neurosci. 2020, 43, 144–154. [Google Scholar] [CrossRef] [PubMed]
  102. Colombo, E.; Farina, C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol. 2016, 37, 608–620. [Google Scholar] [CrossRef] [PubMed]
  103. Subedi, L.; Ji, E.; Shin, D.; Jin, J.; Yeo, J.H.; Kim, S.Y. Equol, a Dietary Daidzein Gut Metabolite Attenuates Microglial Activation and Potentiates Neuroprotection In Vitro. Nutrients 2017, 9, 207. [Google Scholar] [CrossRef] [Green Version]
  104. Moriyama, M.; Hashimoto, A.; Satoh, H.; Kawabe, K.; Ogawa, M.; Takano, K.; Nakamura, Y. S-Equol, a Major Isoflavone from Soybean, Inhibits Nitric Oxide Production in Lipopolysaccharide-Stimulated Rat Astrocytes Partially via the GPR30-Mediated Pathway. Int. J. Inflam. 2018, 2018, 8496973. [Google Scholar] [CrossRef] [Green Version]
  105. Lu, C.; Gao, R.; Zhang, Y.; Jiang, N.; Chen, Y.; Sun, J.; Wang, Q.; Fan, B.; Liu, X.-M.; Wang, Z.F. S-equol, a metabolite of dietary soy isoflavones, alleviates lipopolysaccharide-induced depressive-like behavior in mice by inhibiting neuroinflammation and enhancing synaptic plasticity. Food Funct. 2021, 12, 5770–5778. [Google Scholar] [CrossRef] [PubMed]
  106. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
  107. Matsuda, S.; Okumura, N.; Yoshida, H.; Nishimura, Y.; Murakami, M.; Kitagishi, Y. Genistein downregulates presenilin 1 and ubiquilin 1 expression. Mol. Med. Rep. 2012, 5, 559–561. [Google Scholar] [CrossRef]
  108. Petry, F.D.S.; Coelho, B.P.; Gaelzer, M.M.; Kreutz, F.; Guma, F.T.C.R.; Salbego, C.G.; Trindade, V.M.T. Genistein protects against amyloid-beta-induced toxicity in SH-SY5Y cells by regulation of Akt and Tau phosphorylation. Phytother. Res. 2020, 34, 796–807. [Google Scholar] [CrossRef]
  109. Pierzynowska, K.; Podlacha, M.; Gaffke, L.; Majkutewicz, I.; Mantej, J.; Węgrzyn, A.; Osiadły, M.; Myślińska, D. Węgrzyn G.; Autophagy-dependent mechanism of genistein-mediated elimination of behavioral and biochemical defects in the rat model of sporadic Alzheimer’s disease. Neuropharmacology 2019, 148, 332–346. [Google Scholar] [CrossRef]
  110. Petry, F.D.S.; Hoppe, J.B.; Klein, C.P.; dos Santos, B.G.; Hözer, R.M.; Bifi, F.; Matté, C.; Salbego, C.G.; Trindade, V.M.T. Genistein attenuates amyloid-beta-induced cognitive impairment in rats by modulation of hippocampal synaptotoxicity and hyperphosphorylation of Tau. J. Nutr. Biochem. 2021, 87, 108525. [Google Scholar] [CrossRef]
  111. Shentu, Y.-P.; Hu, W.-T.; Liang, J.-W.; Liuyang, Z.-Y.; Wei, H.; Qun, W.; Wang, X.-C.; Wang, J.-Z.; Westermarck, J.; Liu, R. Genistein Decreases APP/tau Phosphorylation and Ameliorates Aβ Overproduction Through Inhibiting CIP2A. Curr. Alzheimer Res. 2019, 16, 732–740. [Google Scholar] [CrossRef] [PubMed]
  112. Li, R.-Z.; Ding, X.-W.; Geetha, T.; Al-Nakkash, L.; Broderick, T.L.; Babu, J.R. Beneficial Effect of Genistein on Diabetes-Induced Brain Damage in the ob/ob Mouse Model. Drug Des. Devel. Ther. 2020, 14, 3325–3336. [Google Scholar] [CrossRef] [PubMed]
  113. Tsai, M.-C.; Lin, S.-H.; Hidayah, K.; Lin, C.-I. Equol Pretreatment Protection of SH-SY5Y Cells against Aβ (25-35)-Induced Cytotoxicity and Cell-Cycle Reentry via Sustaining Estrogen Receptor Alpha Expression. Nutrients 2019, 11, 2356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhao, L.; Chen, Q.; Brinton, R.D. Neuroprotective and neurotrophic efficacy of phytoestrogens in cultured hippocampal neurons. Exp. Biol. Med. 2002, 227, 509–519. [Google Scholar] [CrossRef]
  115. Hirohata, M.; Ono, K.; Takasaki, J.-I.; Takahashi, R.; Ikeda, T.; Morinaga, A.; Yamada, M. Anti-amyloidogenic effects of soybean isoflavones in vitro: Fluorescence spectroscopy demonstrating direct binding to Aβ monomers, oligomers and fibrils. Biochim. Et Biophys. Acta 2012, 1822, 1316–1324. [Google Scholar] [CrossRef] [Green Version]
  116. Sørensen, I.F.; Purup, S.; Ehrich, M. Modulation of neurotoxicant-induced increases in intracellular calcium by phytoestrogens differ for amyloid beta peptide (Abeta) and 1-methyl-4-phenyl-pyridine (MPP(+)). J. Appl. Toxicol. 2009, 29, 84–89. [Google Scholar] [CrossRef]
  117. Ariyani, W.; Miyazaki, W.; Koibuchi, N. A Novel Mechanism of S-equol Action in Neurons and Astrocytes: The Possible Involvement of GPR30/GPER1. Int. J. Mol. Sci. 2019, 20, 5178. [Google Scholar] [CrossRef] [Green Version]
  118. Johnson, S.L.; Park, H.Y.; Vattem, D.A.; Grammas, P.; Ma, H.; Seeram, N.P. Equol, a Blood-Brain Barrier Permeable Gut Microbial Metabolite of Dietary Isoflavone Daidzein, Exhibits Neuroprotective Effects against Neurotoxins Induced Toxicity in Human Neuroblastoma SH-SY5Y Cells and Caenorhabditis elegans. Plant Foods Hum. Nutr. 2020, 75, 512–517. [Google Scholar] [CrossRef]
  119. Heldring, N.; Pike, A.; Andersson, S.; Matthews, J.; Cheng, G.; Hartman, J.; Tujague, M.; Ström, A.; Treuter, E.; Warner, M.; et al. Estrogen receptors: How do they signal and what are their targets. Physiol. Rev. 2007, 87, 905–931. [Google Scholar] [CrossRef] [Green Version]
  120. Sugiyama, N.; Barros, R.P.; Warner, M.; Gustafsson, J.A. ERbeta: Recent understanding of estrogen signaling. Trends Endocrinol. Metab. 2010, 21, 545–552. [Google Scholar] [CrossRef]
  121. Kelly, J.F.; Bienias, J.L.; Shah, A.; Meeke, K.A.; Schneider, J.A.; Soriano, E.; Bennett, D.A. Levels of estrogen receptors alpha and beta in frontal cortex of patients with Alzheimer’s disease: Relationship to Mini-Mental State Examination scores. Curr. Alzheimer Res. 2008, 5, 45–51. [Google Scholar] [CrossRef] [PubMed]
  122. Almey, A.; Milner, T.A.; Brake, W.G. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm. Behav. 2015, 74, 125–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Liu, F.; Day, M.; Muñiz, L.C.; Bitran, D.; Arias, R.; Revilla-Sanchez, R.; Grauer, S.; Zhang, G.; Kelley, C.; Pulito, V.; et al. Activation of estrogen receptor-beta regulates hippocampal synaptic plasticity and improves memory. Nat. Neurosci. 2008, 11, 334–343. [Google Scholar] [CrossRef] [PubMed]
  124. Hwang, W.J.; Lee, T.Y.; Kim, N.S.; Kwon, J.S. The Role of Estrogen Receptors and Their Signaling across Psychiatric Disorders. Int. J. Mol. Sci. 2020, 22, 373. [Google Scholar] [CrossRef]
  125. Kim, H.J.; Casadesus, G. Estrogen-mediated effects on cognition and synaptic plasticity: What do estrogen receptor knockout models tell us? Biochim. Biophys. Acta 2010, 1800, 1090–1093. [Google Scholar] [CrossRef] [Green Version]
  126. Fehsel, K.; Schikowski, T.; Jänner, M.; Hüls, A.; Voussoughi, M.; Schulte, T.; Vierkötter, A.; Teichert, T.; Herder, C.; Sugiri, D.; et al. Estrogen receptor beta polymorphisms and cognitive performance in women: Associations and modifications by genetic and environmental influences. J. Neural Transm. 2016, 123, 1369–1379. [Google Scholar] [CrossRef]
  127. Janicki, S.C.; Park, N.; Cheng, R.; Lee, J.H.; Schupf, N.; Clark, L.N. Estrogen receptor β variants modify risk for Alzheimer’s disease in a multiethnic female cohort. J. Alzheimers Dis. 2014, 40, 83–93. [Google Scholar] [CrossRef] [Green Version]
  128. Vargas, K.G.; Milic, J.; Zaciragic, A.; Wen, K.-X.; Jaspers, L.; Nano, J.; Dhana, K.; Bramer, W.M.; Kraja, B.; van Beeck, E.; et al. The functions of estrogen receptor beta in the female brain: A systematic review. Maturitas 2016, 93, 41–57. [Google Scholar] [CrossRef]
  129. Kim, E.K.; Choi, E.-J. Pathological roles of MAPK signaling pathways in human diseases. Biochim. Biophys. Acta 2010, 1802, 396–405. [Google Scholar] [CrossRef] [Green Version]
  130. Han, X.; Aenlle, K.K.; Bean, L.A.; Rani, A.; Semple-Rowland, S.L.; Kumar, A.; Foster, T.C. Role of estrogen receptor α and β in preserving hippocampal function during aging. J. Neurosci. 2013, 33, 2671–2683. [Google Scholar] [CrossRef] [Green Version]
  131. Spencer-Segal, J.; Tsuda, M.; Mattei, L.; Waters, E.; Romeo, R.; Milner, T.; McEwen, B.; Ogawa, S. Estradiol acts via estrogen receptors alpha and beta on pathways important for synaptic plasticity in the mouse hippocampal formation. Neuroscience 2012, 202, 131–146. [Google Scholar] [CrossRef]
  132. Rissman, E.F.; Heck, A.L.; Leonard, J.E.; Shupnik, M.A.; Gustafsson, J.A. Disruption of estrogen receptor beta gene impairs spatial learning in female mice. Proc. Natl. Acad. Sci. USA 2002, 99, 3996–4001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bourtchuladze, R.; Frenguelli, B.; Blendy, J.; Cioffi, D.; Schutz, G.; Silva, A.J. Deficient long-term memory in mice with a targeted mutation of the cAMP-responsive element-binding protein. Cell 1994, 79, 59–68. [Google Scholar] [CrossRef]
  134. Khan, M.M.; Wakade, C.; de Sevilla, L.; Brann, D.W. Selective estrogen receptor modulators (SERMs) enhance neurogenesis and spine density following focal cerebral ischemia. J. Steroid Biochem. Mol. Biol. 2015, 146, 38–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Nilsson, S.; Koehler, K.F.; Gustafsson, J. Development of subtype-selective oestrogen receptor-based therapeutics. Nat. Rev. Drug Discov. 2011, 10, 778–792. [Google Scholar] [CrossRef] [PubMed]
  136. Hernández-Fonseca, K.; Massieu, L.; de la Cadena, S.G.; Guzmán, C.; Camacho-Arroyo, I. Neuroprotective role of estradiol against neuronal death induced by glucose deprivation in cultured rat hippocampal neurons. Neuroendocrinology 2012, 96, 41–50. [Google Scholar] [CrossRef]
  137. Crawford, D.; Mangiardi, M.; Song, B.; Patel, R.; Du, S.; Sofroniew, M.V.; Voskuhl, R.R.; Tiwari-Woodruff, S.K. Oestrogen receptor β ligand: A novel treatment to enhance endogenous functional remyelination. Brain 2010, 133, 2999–3016. [Google Scholar] [CrossRef]
  138. Suwanna, N.; Thangnipon, W.; Soi-Ampornkul, R. Neuroprotective effects of diarylpropionitrile against β-amyloid peptide-induced neurotoxicity in rat cultured cortical neurons. Neurosci. Lett. 2014, 578, 44–49. [Google Scholar] [CrossRef]
  139. Suwanna, N.; Thangnipon, W.; Kumar, S.; De Vellis, J. Neuroprotection by diarylpropionitrile in mice with spinal cord injury. Excli J. 2014, 13, 1097–1103. [Google Scholar]
  140. Sárvári, M.; Kalló, I.; Hrabovszky, E.; Solymosi, N.; Liposits, Z. Ovariectomy and subsequent treatment with estrogen receptor agonists tune the innate immune system of the hippocampus in middle-aged female rats. PLoS ONE 2014, 9, e88540. [Google Scholar]
  141. Sárvári, M.; Kalló, I.; Hrabovszky, E.; Solymosi, N.; Rodolosse, A.; Liposits, Z. Long-Term Estrogen Receptor Beta Agonist Treatment Modifies the Hippocampal Transcriptome in Middle-Aged Ovariectomized Rats. Front. Cell Neurosci. 2016, 10, 149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Schneider, L.S.; Hernandez, G.; Zhao, L.; Franke, A.A.; Chen, Y.L.; Pawluczyk, S.; Mack, W.J.; Brinton, R.D. Safety and feasibility of estrogen receptor-β targeted phytoSERM formulation for menopausal symptoms: Phase 1b/2a randomized clinical trial. Menopause 2019, 26, 874–884. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, Y.; Hernandez, G.; Mack, W.J.; Schneider, L.S.; Yin, F.; Brinton, R.D. Retrospective analysis of phytoSERM for management of menopause-associated vasomotor symptoms and cognitive decline: A pilot study on pharmacogenomic effects of mitochondrial haplogroup and APOE genotype on therapeutic efficacy. Menopause 2020, 27, 57–65. [Google Scholar] [CrossRef]
  144. Yao, J.; Zhao, L.; Mao, Z.; Chen, S.; Wong, K.C.; To, J.; Brinton, R.D. Potentiation of brain mitochondrial function by S-equol and R/S-equol estrogen receptor beta-selective phytoSERM treatments. Brain Res. 2013, 1514, 128–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhao, L.; Mao, Z.; Brinton, R.D. A select combination of clinically relevant phytoestrogens enhances estrogen receptor beta-binding selectivity and neuroprotective activities in vitro and in vivo. Endocrinology 2009, 150, 770–783. [Google Scholar] [CrossRef] [Green Version]
  146. Zhao, L.; Mao, Z.; Chen, S.; Schneider, L.S.; Brinton, R.D. Early intervention with an estrogen receptor β-selective phytoestrogenic formulation prolongs survival, improves spatial recognition memory, and slows progression of amyloid pathology in a female mouse model of Alzheimer’s disease. J. Alzheimers Dis. 2013, 37, 403–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Song, X.; Liu, B.; Cui, L.; Zhou, B.; Liu, W.; Xu, F.; Hayashi, T.; Hattori, S.; Ushiki-Kaku, Y.; Tashiro, S.-I.; et al. Silibinin ameliorates anxiety/depression-like behaviors in amyloid β-treated rats by upregulating BDNF/TrkB pathway and attenuating autophagy in hippocampus. Physiol. Behav. 2017, 179, 487–493. [Google Scholar] [CrossRef] [PubMed]
  148. Song, X.; Zhou, B.; Cui, L.; Lei, D.; Zhang, P.; Yao, G.; Xia, M.; Hayashi, T.; Hattori, S.; Ushiki-Kaku, Y.; et al. Silibinin ameliorates Aβ(25-35)-induced memory deficits in rats by modulating autophagy and attenuating neuroinflammation as well as oxidative stress. Neurochem Res. 2017, 42, 1073–1083. [Google Scholar] [CrossRef] [PubMed]
  149. Fernandes, V.; Sharma, D.; Kalia, K.; Tiwari, V. Neuroprotective effects of silibinin: An in silico and in vitro study. Int. J. Neurosci. 2018, 128, 935–945. [Google Scholar] [CrossRef] [PubMed]
  150. Hanson, A.M.; Perera, K.L.I.S.; Kim, J.; Pandey, R.K.; Sweeney, N.; Lu, X.; Imhoff, A.; Mackinnon, A.; Wargolet, A.J.; Van Hart, R.M.; et al. A-C Estrogens as Potent and Selective Estrogen Receptor-Beta Agonists (SERBAs) to Enhance Memory Consolidation under Low-Estrogen Conditions. J. Med. Chem. 2018, 61, 4720–4738. [Google Scholar] [CrossRef] [PubMed]
  151. Fleischer, A.W.; Schalk, J.C.; Wetzel, E.A.; Hanson, A.M.; Sem, D.S.; Donaldson, W.A.; Frick, K.M. Long-term oral administration of a novel estrogen receptor beta agonist enhances memory and alleviates drug-induced vasodilation in young ovariectomized mice. Horm. Behav. 2021, 130, 104948. [Google Scholar] [CrossRef] [PubMed]
  152. Hill, R.A.; Chow, J.; Fritzemeier, K.; Simpson, E.R.; Boon, W.C. Fas/FasL-mediated apoptosis in the arcuate nucleus and medial preoptic area of male ArKO mice is ameliorated by selective estrogen receptor alpha and estrogen receptor beta agonist treatment, respectively. Mol. Cell Neurosci. 2007, 36, 146–157. [Google Scholar] [CrossRef]
  153. Clark, J.; Alves, S.; Gundlah, C.; Rocha, B.; Birzin, E.; Cai, S.-J.; Flick, R.; Hayes, E.; Ho, K.; Warrier, S.; et al. Selective estrogen receptor-beta (SERM-beta) compounds modulate raphe nuclei tryptophan hydroxylase-1 (TPH-1) mRNA expression and cause antidepressant-like effects in the forced swim test. Neuropharmacology 2012, 63, 1051–1063. [Google Scholar] [CrossRef] [PubMed]
  154. Federico, A.; Dallio, M.; Loguercio, C. Silymarin/Silybin and Chronic Liver Disease: A Marriage of Many Years. Molecules 2017, 22, 191. [Google Scholar] [CrossRef] [Green Version]
  155. Hillisch, A.; Peters, O.; Kosemund, D.; Müller, G.; Walter, A.; Schneider, B.; Reddersen, G.; Elger, W.; Fritzemeier, K.H. Dissecting physiological roles of estrogen receptor alpha and beta with potent selective ligands from structure-based design. Mol. Endocrinol. 2004, 18, 1599–1609. [Google Scholar] [CrossRef] [Green Version]
  156. Franke, A.A.; Lai, J.F.; Pagano, I.; Morimoto, Y.; Maskarinec, G. Equol production changes over time in pre-menopausal women. Br. J. Nutr. 2012, 107, 1201–1206. [Google Scholar] [CrossRef]
  157. Franke, A.A.; Lai, J.F.; Halm, B.M.; Pagano, I.; Kono, N.; Mack, W.J.; Hodis, H.N. Equol production changes over time in postmenopausal women. J. Nutr. Biochem. 2012, 23, 573–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Ishiwata, N.; Melby, M.; Mizuno, S.; Watanabe, S. New equol supplement for relieving menopausal symptoms: Randomized, placebo-controlled trial of Japanese women. Menopause 2009, 16, 141–148. [Google Scholar] [CrossRef] [PubMed]
  159. Tousen, Y.; Ezaki, J.; Fujii, Y.; Ueno, T.; Nishimuta, M.; Ishimi, Y. Natural S-equol decreases bone resorption in postmenopausal, non-equol-producing Japanese women: A pilot randomized, placebo-controlled trial. Menopause 2011, 18, 563–574. [Google Scholar] [CrossRef]
  160. Aso, T.; Uchiyama, S.; Matsumura, Y.; Taguchi, M.; Nozaki, M.; Takamatsu, K.; Ishizuka, B.; Kubota, T.; Mizunuma, H.; Ohta, H. A natural S-equol supplement alleviates hot flushes and other menopausal symptoms in equol nonproducing postmenopausal Japanese women. J. Womens Health 2012, 21, 92–100. [Google Scholar] [CrossRef] [Green Version]
  161. Jenks, B.H.; Iwashita, S.; Nakagawa, Y.; Ragland, K.; Lee, J.; Carson, W.H.; Ueno, T.; Uchiyama, S. A pilot study on the effects of S-equol compared to soy isoflavones on menopausal hot flash frequency. J. Womens Health 2012, 21, 674–682. [Google Scholar] [CrossRef]
  162. Oyama, A.; Ueno, T.; Uchiyama, S.; Aihara, T.; Miyake, A.; Kondo, S.; Matsunaga, K. The effects of natural S-equol supplementation on skin aging in postmenopausal women: A pilot randomized placebo-controlled trial. Menopause 2012, 19, 202–210. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of daidzein, genistein (two major soy isoflavones), equol, and estradiol. Daidzein and genistein are two major soy isoflavones and comprise >95% of their dietary source. Soy isoflavones and S-equol are structurally similar to estradiol. S-equol is a metabolite of daidzein, transformed by the gut microbiome. Daidzein is reduced to S-equol through the intermediate dihydrodaidzein and then converted by deoxygenation to yield S-equol. [27] Daidzein, genistein and equol are structurally similar to estradiol (images of chemical structures are from Wikimedia Commons and Pixabay).
Figure 1. Chemical structures of daidzein, genistein (two major soy isoflavones), equol, and estradiol. Daidzein and genistein are two major soy isoflavones and comprise >95% of their dietary source. Soy isoflavones and S-equol are structurally similar to estradiol. S-equol is a metabolite of daidzein, transformed by the gut microbiome. Daidzein is reduced to S-equol through the intermediate dihydrodaidzein and then converted by deoxygenation to yield S-equol. [27] Daidzein, genistein and equol are structurally similar to estradiol (images of chemical structures are from Wikimedia Commons and Pixabay).
Ijms 23 11921 g001
Table 1. Potential mechanisms of S-equol to slow the progression of arterial stiffness (image of the white matter lesion is from Wikimedia Commons; images of amyloid β plaques and a cross-sectional view of the aorta were created with BioRender).
Table 1. Potential mechanisms of S-equol to slow the progression of arterial stiffness (image of the white matter lesion is from Wikimedia Commons; images of amyloid β plaques and a cross-sectional view of the aorta were created with BioRender).
 
 
Arterial stiffness is an emerging target for preventing cognitive decline and dementia
 
• Emerging as a potential therapeutic target for preventing vascular contribution of cognitive impairment and dementia (VCID) and Alzheimer’s disease and related dementias (ADRD) [58,71].
• Significantly associated with cognitive decline [53,54,55,56,57,58].
• Signifincatly associated with multiple forms of dementia-related pathology, including white matter lesion volume and Amyloid-β deposition in the brain [53,54,55,56,57,58].
• Significantly associated with incident dementia in some [59] but not all [72,73] prospective cohort studies.
Ijms 23 11921 i001
 
 
Pathophysiology of arterial stiffness
 
Major underlying mechanisms of arterial stiffness include pathological collagen production and elastin degradation in the arterial wall, endothelial dysfunction, oxidative stress, and inflammation [58].
Ijms 23 11921 i002
 
 
Potential mechanisms of S-equol that slow the progression of arterial stiffness
 
• S-equol inhibits abnormal collagen synthesis, proliferation, and migration of smooth muscle cells in the human aorta [68].
• S-equol may inhibit the degradation of elastin in the arterial wall by decreasing the expression of the elastase enzyme [69].
• S-equol improves endothelial function in the human aorta and umbilical vein endothelial cells [74,75,76,77].
• S-equol possesses anti-inflammatory and antioxidant properties [70].
Table 2. Estrogen receptor β agonists are being investigated for neuroprotection.
Table 2. Estrogen receptor β agonists are being investigated for neuroprotection.
NameFindings
Diarylproprionitrile (DPN)Treatment with DPN-protected rat cultured hippocampal neurons from glucose deprivation-induced cell death [136].
Treatment with DPN of chronic experimental autoimmune encephalomyelitis mice (transgenic proteolipid protein-enhanced green fluorescent protein transgenic mice in the C57BL/6J background) prevented histopathological (fewer demyelinated, damaged axons and more myelinated axons) and increased mature oligodendrocyte numbers [137].
Pretreatment with DPN decreased neuronal cell death by reducing reactive oxygen species in Aβ1-42 induced oxidative stress and inflammation in primary rat cortical cell culture [138]. In addition, DPN pretreatment decreased pro-inflammatory cytokines (IL-1β and IL-6)
In C57/B1/6 female mice performed with spinal cord injury, the administration of DPN reduced neural apoptosis and inflammation compared to untreated groups [139].In middle-aged female Harlan-Wistar rats with ovariectomy followed by no replacement or replacement with estradiol, DPN, or LE2 (ERα agonist), gene expression related to the innate immune system, specifically macrophage-associated and complement genes, in the hippocampus was investigated. Ovariectomy caused the upregulation of these gene expressions, and estradiol, DPN, and LE2 attenuated the increase in these gene expressions [140].
In middle-aged female Harlan-Wistar rats implanted subcutaneously in the neck with DPN or vehicle only for 29 days, the effect of DPN on hippocampal transcriptome was investigated. DPN contributed to regulating transcription, translation, neurogenesis, neuromodulation, and neuroprotection in the hippocampal formation [141].
PhytoSERMA first human study of PhytoSERM-Phase 1b/2a randomized, double-blind placebo-controlled study of 50 or 100 mg/day PhytoSERM for 12 weeks, recruiting 71 peri-menopausal women aged 45–60, showed that both 50 and 100 mg/day were well tolerated but based on safety outcomes, 50 mg/day was considered preferable [142].
A retrospective responder analysis of the trial mentioned above showed that 50 mg/day of PhytoSERM significantly reduced hot flashes and preserved cognitive function in verbal learning and executive function [143].
In mitochondrial markers in rat hippocampal neuronal cultures and a female mouse ovariectomy model, both S-equol phytoSERM and R/S-equol phytoSERM treatments potentiated mitochondrial bioenergetics [144].
In rat primary hippocampal neuronal cultures challenged with neurotoxic glutamate or amyloid-β1–42, phytoSERM showed a greater effect on neuronal survival than genistein, daidzein, equol, or IBSO03569. In ovariectomized adult female rats, treatment with phytoSERV significantly enhanced brain mitochondrial bioenergetics. Furthermore, in western blot analyses of hippocampal protein, phytoSERM increased expression of brain mitochondrial anti-apoptotic protein Bcl-2 and Bcl-xL as well as insulin-degrading enzyme and neprilysin, which are amyloid-β degrading enzymes [145].
A 9-month supplementation of phytoSERM in a female triple transgenic mouse model of Alzheimer’s disease promoted physical health, prolonged survival, improved spatial recognition memory, and attenuated amyloid-β deposition and plaque formation in the brain [146].
SilibininOral gavage of silibinin protected amyloid-β1–42 induced anxiety/depression-like behaviors in male Sprague-Dawley rats. Silibinin significantly suppressed neuronal damage induced by amyloid-β1–42 in hippocampus regions, increased brain-derived neurotrophic factor, and attenuated the increase of autophagy levels in the hippocampus [147].
Silibinin-protected amyloid-β25–35 induced memory deficits in male Sprague-Dawley rats. Silibinin increased the expression of anti-inflammatory cytokine (interleukin 4) and decreased the expression of pro-inflammatory cytokines (interleukin 1β) in the hippocampus. Silibinin also increased glutathione levels and decreased malondialdehyde levels, indicating antioxidant properties of silibinin [148].
Treatment with silibinin of astrocyte induced with lipopolysaccharide decreased the reactive form of astrocyte, attenuated nitrite release, increased glutathione levels, reduced malonaldehyde, and reduced glial fibrillary activated protein [149].
EGX358 or ISP358-2Intrahippocampal infusion or intraperitoneal injection of ISP358-2 enhanced memory consolidation in C57BL/6 ovariectomized female mice [150].
EGX358 reduced drug-induced vasodilation and enhanced spatial and object recognition memory without adversely affecting anxiety- or depression-like behaviors in C57BL/6 ovariectomized female mice gavaged with EGX358 for 9 weeks [151].
8β-VE2Injecting 8β-VE2 for 6 weeks in aromatase knockout male mice prevented dopaminergic cell death in the medial preoptic area but not in the rostral and medial arcuate nucleus [152].
SERM-beta1 and SERM-beta2Treatment with SERM-beta 1 or SERM-beta 2 for 4 days in ovariectomized Sprague-Dawley rats stimulated an increase in the total number of cells in the dentate gyrus of the hippocampus [153].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sekikawa, A.; Wharton, W.; Butts, B.; Veliky, C.V.; Garfein, J.; Li, J.; Goon, S.; Fort, A.; Li, M.; Hughes, T.M. Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia. Int. J. Mol. Sci. 2022, 23, 11921. https://doi.org/10.3390/ijms231911921

AMA Style

Sekikawa A, Wharton W, Butts B, Veliky CV, Garfein J, Li J, Goon S, Fort A, Li M, Hughes TM. Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia. International Journal of Molecular Sciences. 2022; 23(19):11921. https://doi.org/10.3390/ijms231911921

Chicago/Turabian Style

Sekikawa, Akira, Whitney Wharton, Brittany Butts, Cole V. Veliky, Joshua Garfein, Jiatong Li, Shatabdi Goon, Annamaria Fort, Mengyi Li, and Timothy M. Hughes. 2022. "Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia" International Journal of Molecular Sciences 23, no. 19: 11921. https://doi.org/10.3390/ijms231911921

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

Article Metrics

Back to TopTop