Next Article in Journal
Profiling the Physiological Roles in Fish Primary Cell Culture
Next Article in Special Issue
Neuroprotective Action of Humanin and Humanin Analogues: Research Findings and Perspectives
Previous Article in Journal
Heteroplasmy and Individual Mitogene Pools: Characteristics and Potential Roles in Ecological Studies
Previous Article in Special Issue
Adenosine Improves Mitochondrial Function and Biogenesis in Friedreich's Ataxia Fibroblasts Following L-Buthionine Sulfoximine-Induced Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer’s Disease

1
Department of Biology, Lamar University, Beaumont, TX 77705, USA
2
Biological Science, University of Calgary, Calgary, AB T2N 1N4, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biology 2023, 12(11), 1453; https://doi.org/10.3390/biology12111453
Submission received: 7 October 2023 / Revised: 8 November 2023 / Accepted: 14 November 2023 / Published: 20 November 2023

Abstract

:

Simple Summary

Despite the well-documented pathophysiology of Alzheimer’s Disease (AD), treatment options are limited in diversity and efficacy. Thus, the development of new treatments requires an extensive understanding of molecular pathways altered by drugs in development. In this review, we survey the literature regarding common herbal phytochemicals, kaempferol and quercetin, with a specific focus on their multiple mechanisms that alleviate the pathological underpinnings of AD. Here, we utilize the well-documented mechanisms of quercetin to propose a novel multimodal mechanism of kaempferol, and we discuss common herbal sources and the limitations of these potential treatments.

Abstract

Alzheimer’s Disease (AD) is a progressive neurodegenerative disorder impairing cognition and memory in the elderly. This disorder has a complex etiology, including senile plaque and neurofibrillary tangle formation, neuroinflammation, oxidative stress, and damaged neuroplasticity. Current treatment options are limited, so alternative treatments such as herbal medicine could suppress symptoms while slowing cognitive decline. We followed PRISMA guidelines to identify potential herbal treatments, their associated medicinal phytochemicals, and the potential mechanisms of these treatments. Common herbs, including Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum, produced promising pre-clinical results. These herbs are rich in kaempferol and quercetin, flavonoids with a polyphenolic structure that facilitate multiple mechanisms of action. These mechanisms include the inhibition of Aβ plaque formation, a reduction in tau hyperphosphorylation, the suppression of oxidative stress, and the modulation of BDNF and PI3K/AKT pathways. Using pre-clinical findings from quercetin research and the comparatively limited data on kaempferol, we proposed that kaempferol ameliorates the neuroinflammatory state, maintains proper cellular function, and restores pro-neuroplastic signaling. In this review, we discuss the anti-AD mechanisms of quercetin and kaempferol and their limitations, and we suggest a potential alternative treatment for AD. Our findings lead us to conclude that a polyherbal kaempferol- and quercetin-rich cocktail could treat AD-related brain damage.

1. Introduction

Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder characterized by cognitive decline and memory impairment. AD could affect 152 million individuals by 2050 [1]. The progression of AD is influenced by multiple factors, including the accumulation of beta-amyloid plaques (Aβ) and the formation of neurofibrillary tangles (NFTs). The aggregation of Aβ plaques exacerbates the disease by impairing neuronal function and triggering neuroinflammation [2,3,4,5]. Oxidative stress and the presence of neurofibrillary tangles (NFTs) also contribute to the aggregation of Aβ into senile plaques [6,7,8,9,10,11,12,13,14]. NFTs consist of hyperphosphorylated tau proteins that disrupt neuronal transport systems [15,16,17,18,19,20,21]. Neuroinflammation, in turn, exacerbates damage to neuronal integrity [22]. Symptoms of AD include memory loss, impaired learning, emotional changes, cognitive and speech deficits, shortened attention span, and impaired management of daily tasks [23,24,25].
Currently, the treatments available for AD are expensive and have minimal efficacy. Acetylcholinesterase inhibitors (AChEIs), including donepezil, and N-methyl-D-aspartate (NMDA) receptor antagonists, including memantine, are commonly prescribed for AD [26]. AChEIs inhibit the enzymatic degradation of ACh by inhibiting cholinesterase activity [27], while NMDA receptor antagonists limit calcium influx to prevent glutamate-induced cytotoxic cell death [28]. However, these drugs simply suppress symptoms and fail to halt disease progression [26], and only half of the population positively responds to these current treatments [29,30]. Herbal medicine boasts a well-documented history of safe and effective incorporation into traditional Asian diets [31,32]. Preclinical studies have demonstrated that these herbs can enhance cognitive and memory functions [33,34]. These herbs serve as dependable sources of phytochemicals, such as kaempferol and quercetin, that have limited side effects and could combat Alzheimer’s disease [35,36,37]. Specifically, these flavonoids have anti-inflammatory, neuroprotective, and anti-degenerative effects [33,38,39,40,41,42,43,44,45,46].
The objective of this review is to elucidate the anti-AD mechanisms of kaempferol and quercetin. Here, we present a multimodal mechanism of action for kaempferol and quercetin in the treatment of Alzheimer’s disease (AD). First, both flavonoids exert antioxidant effects, which stabilize cellular function and reduce neuroinflammation. Importantly, they also modulate PI3K/AKT signaling to limit Aβ and tau accumulation in toxic aggregates and enhance neuroplasticity by restoring BDNF signaling. These mechanisms ultimately improve memory and cognitive performance in AD patients. To our knowledge, this review represents the first comprehensive exploration of the literature that collectively shows kaempferol’s potential to counteract both tau and Aβ via modulation of the PI3K/AKT/GSK-3β pathway. Additionally, we propose that a polyherbal cocktail, incorporating sources rich in quercetin and kaempferol, could serve as an effective adjunctive or alternative treatment for AD. Finally, we explore the limitations of quercetin and kaempferol and discuss potential strategies for overcoming these challenges.

2. Materials and Methods

We collected data following the PRISMA guidelines for systematic review articles. The articles were sourced from PubMed, ScienceDirect, and Google Scholar, and data collection was conducted up until November 2023. We compiled a relevant list of articles to identify phytochemicals that have been studied for the treatment of Alzheimer’s disease (AD) and their potential to induce therapeutic brain changes related to AD. Our search strategy initially yielded a total of 13,691 papers (13,688 from databases and an additional 3 from other sources). Of these, 2463 studies were screened based on their titles and abstracts, resulting in 378 articles that met the inclusion criteria (Figure 1). We included studies and reviews that explored the anti-AD mechanisms of phytochemicals and those that provided insights into the features of AD. The language was limited to English. Inclusion criteria required that articles discuss topics such as “Alzheimer’s disease”, “herbs”, “kaempferol”, “quercetin”, “inflammation”, “neuroprotection”, “tau”, and “Aβ”. The selected articles encompassed reviews, original research articles, and published clinical trials. Data extraction was carried out independently by a team of three investigators, considering factors such as the year of publication, article types, and the topic of herbs in relation to AD.

3. Hallmarks of Alzheimer’s Disease

Several features of AD, including Aβ plaque accumulation [47,48], tau hyperphosphorylation and neuroinflammation [49], and oxidative stress [50,51,52,53], have been identified as targets for drug development. Moreover, these deficits have been observed in studies with human patients [6,54,55,56,57,58,59,60,61,62,63]. This section will briefly explore the pathophysiology of AD, with a focus on the proposed molecular origins and outcomes of their aberrant activities.
While the origins are still debated, the literature greatly supports the roles of oxidative stress and neuroinflammation as critical drivers of neurodegeneration. Antioxidant deficits facilitate ROS production, driving oxidative stress via lipid peroxidation [57]. Consequently, mitochondrial energy production is impaired and pro-apoptotic signaling follows [57]. Glutamate-induced excitotoxicity could also facilitate oxidative stress [64,65,66,67]. Disrupted ROS clearance establishes the neuroinflammatory microglial and astrocytic hyperactivity [38,68,69,70,71,72] and favors neuronal signaling pathways that impair Aβ clearance [48,73,74]. Finally, proper mitochondrial function is required for Aβ clearance and can, in turn, maintain appropriate tau activity states [75].
Although normal Aβ levels can maintain regular neuronal function [76], failed Aβ clearance from the brain can expedite neurodegeneration by facilitating plaque accumulation and impairing neuronal communication [22,47,48,75,77,78,79]. Moreover, Aβ accumulation further promotes oxidative stress [80,81,82,83]. As Aβ plaques accumulate in the brain due to impaired clearance [84], overzealous astrocytic and microglial responses compound the neuroinflammatory environment by releasing pro-inflammatory factors, promoting neuronal apoptosis [6,49,85,86,87,88,89]. These findings were supported in postmortem tissue [6,54,55,56]. Finally, Aβ signaling significantly impairs LTP [90], facilitating neurodegeneration via low synaptic activity.
AD is one of the most common tauopathies [91]. Aβ plaque accumulation drives tau hyperphosphorylation [47,58,92,93,94,95,96,97,98,99,100], possibly by excess GSK-3β signaling [101]. Likewise, tau hyperphosphorylation also compounds Aβ toxicity [102,103], which has been supported by PET imaging in humans with memory impairment and cognitive decline [60]. These studies demonstrate that Aβ toxicity is necessary for tau hyperphosphorylation [59,60]. Specifically, accumulating Aβ binds to NMDAR, generating excess calcium levels to activate calpain-mediated microtubule-associated protein cleavage [65,104,105]. These events impair mitochondrial function, invoking pro-apoptotic signaling [65,106]. Tau hyperphosphorylation dismantles axonal microtubules to degenerate the axon [15,107,108,109,110], impairing synaptic plasticity [102,103,111,112]. Hyperphosphorylated tau spreads throughout the hippocampus in AD models [113], and uptake may be mediated by clathrin-induced endocytosis [114]. Risk factors such as sleep apnea may potentiate the spread of tau in this manner [115]. Ultimately, these events result in neuronal death and compromise neuroplasticity, thereby driving neuroinflammation and impairing cognitive function.

4. Anti-AD Mechanisms of Quercetin and Kaempferol

Given the limited therapeutics available to AD patients, it is essential to explore alternative treatments, such as plant-derived phytochemicals. Flavonoids, including kaempferol and quercetin, belong to the class of polyphenols commonly found in various herbs. Notably, kaempferol and quercetin possess lipophilic properties [50], which facilitate their easy entry into cells. These phytochemicals are abundant, with an average daily consumption of approximately 23 mg of flavonoids in a typical diet [116,117]. Kaempferol and quercetin produce several beneficial properties, including anti-inflammatory, antioxidant, anti-Aβ, anti-tau, and pro-neuroplastic effects [37,38,39,57,74,118,119,120,121,122,123,124,125,126,127,128]. Moreover, they have demonstrated cognitive and memory-enhancing effects in animal studies [37]. Consequently, this section aims to delve into the commonly studied effects of these phytochemicals.

4.1. Quercetin

Quercetin, the most prevalent flavonoid, is found in several traditional medicinal herbs and is commonly found in fruits and vegetables, including berries, onions, and leeks [118,129,130,131,132,133,134,135,136,137,138,139]. Quercetin intake constitutes approximately 60–75% of total flavonols [140,141], and 25 mg of quercetin is found in the average diet [38]. Quercetin is commonly investigated for its potential anti-neurodegenerative efficacy and is considered safe [51,142]. Quercetin is a 15-carbon flavonoid with two benzene rings connected via a 3-carbon shape (Figure 2) [38,130,143].
Quercetin produces anti-inflammatory effects via multiple signaling pathways, including Nrf2, paraoxonase-2 (PON2), JNK, PKC, and NF-kB [51,118,128,144,145,146,147]. Quercetin dose-dependently protected HT22 hippocampal neurons from glutamate-induced apoptosis by limiting ROS production, impairing the calpain-mediated cleavage of cytoskeletal proteins, and preserving mitochondrial membrane potential [65]. Quercetin also inhibits NO release by inhibiting iNOS activity [33,38,148], which could reduce excess glutamate signaling and minimize the risk of glutamate-induced cytotoxicity in hippocampal neurons in a similar fashion to kaempferol and its derivatives [149]. Moreover, quercetin inhibits COX-2 and TLR4 activity to reduce inflammatory responses [6,39,148]. Interestingly, quercetin may have epigenetic mechanisms by inhibiting lysine acetyltransferase (KAT) activity [150,151] and increasing lysine deacetylase (KDAC) activity [152], suggesting that the flavonoid can bidirectionally regulate autophagy [153], neuroinflammation, and apoptosis [154]. Quercetin also inhibits acetylcholinesterase (AChE) [155], which can enhance alertness and cognitive function in AD patients.
The anti-Aβ effects of quercetin are well studied in AD and related models and have yielded promising therapeutic properties. The hydrophobic groups of quercetin can inhibit the formation of Aβ fibrils [120,121,122,123,156]. Chronic quercetin treatment also slowed Aβ aggregation by potentiating AMPK signaling and inhibiting mitochondrial ROS production, leading to improved memory and object recognition in APPswe/PS1dE9 [80,157]. Quercetin treatment also inhibits the BACE1-mediated cleavage of APP into Aβ by inhibiting NF-kB [74]. Consequently, mitochondrial membrane permeability is restored, and cellular survival is favored over oxidative stress [158]. This anti-neurodegenerative effect could be due to the free radical-quenching structure of the catechol group, reducing neuroinflammation, lipid peroxidation, mitochondrial stress, and DNA damage [38,51]. Elevated SOD, GPx, and Na+-K+ ATPase activity could also be due to quercetin’s anti-Aβ effects [44,78].
In many studies, quercetin and its derivatives reduced tau hyperphosphorylation [23,58,132,159]. In rodent HT22 hippocampal neurons, chronic quercetin treatment inhibited tau phosphorylation at four sites by reducing p-Cdk5 levels, limiting calpain activity, and dramatically reducing Ca2+ influx [58]. In 3xTgAD mice, chronic quercetin inhibited Aβ pathology, reduced NFT levels, and prevented astrocytic and microglial hyperactivity in the amygdala and hippocampus [132,160], showing that the anti-Aβ and anti-tau mechanisms of quercetin depend on its anti-inflammatory effects. Consequently, these mice demonstrated improved learning and memory and decreased anxiety [132], while combined exercise and quercetin treatment robustly improved spatial memory in AD rodents [161]. Studies also found that quercetin enhanced cell viability and morphology by reducing MDA and ROS levels and increasing antioxidant SOD and GSH activity [159,162], limiting NF-κB signaling, restoring mitochondrial membrane potential to baseline, inhibiting tau hyperphosphorylation, and regulating Akt/PI3K/GSK-3β signaling pathway [159,163]. Taken together, these data show that quercetin has a multimodal mechanism of action in treating AD. Of note, the anti-tau and consequent pro-neuroplastic effect of quercetin is further explored in Section 5, but the primary anti-inflammatory, anti-Aβ, ant-tau, and pro-neuroplastic effects of this flavonoid are all dependent on each other.
Figure 2. The chemical structure of quercetin, deduced from PubChem [164].
Figure 2. The chemical structure of quercetin, deduced from PubChem [164].
Biology 12 01453 g002

4.2. Kaempferol

Kaempferol is a common 15-carbon polyphenol (Figure 3) that shares significant structural similarity with quercetin. It is one of the most common flavonoids and is found in a variety of common foods, including fruits and vegetables [129,130,165,166,167,168,169,170]. Multiple preclinical and clinical studies have supported the anti-AD activity of kaempferol [57,149,171,172,173,174]. Kaempferol has pro-neuroplastic, anti-Aβ, anti-tau, anti-inflammatory, and antioxidant properties [29,44,57,171,175,176,177,178,179]. Notably, kaempferol also inhibits AChE like quercetin [180], but this mechanism is beyond the scope of this review.
Like quercetin, kaempferol and its metabolites reduce inflammation and have potent antioxidant properties [181,182,183,184]. Kaempferol also directly modulates neuroinflammation by impairing microglial TLR4 and NF-kB signaling and inhibiting the release of NO, iNOS, PGE2, IL-1β, TNF-α, and IFN-γ [167,185]. Kaempferol also reversed BBB damage [36,186,187]. Kaempferol can also modulate neuroinflammation by regulating epigenetic factors such as SIRT1, a subtype of KDAC [188,189,190]. Kaempferol also prevents cytotoxic damage to PC12 neurons by upregulating SIRT [191]. Other immune factors modulated by kaempferol include COX-2, lipoxygenases, prostacyclin, and leukotrienes [148,187,192,193,194]. Finally, kaempferol may reduce neuroinflammation via Nrf-2 signaling [185].
Like quercetin, kaempferol and its derivatives reverse Aβ-induced damage [29,120,122,124,125,149,195]. Kaempferol-3-O-rhamnoside (K-3-Rh), a kaempferol derivative, limited total Aβ burden and toxicity by disrupting β-sheet formation and impairing Aβ plaque formation in human SH-SY5Y cells [195,196]. However, kaempferol antagonized fibrilization with lower potency compared to quercetin and morin [120,122]. In rodent neuroblastoma cells, kaempferol 3-O-(6″-acetyl)-β-glucopyranoside (KAG) robustly inhibited Aβ-mediated cytotoxic cell death and ROS generation [149]. KAG reversed Aβ-mediated oxidative stress and increased cell survival by regulating caspase-3, Bax, and Bcl-2 signaling [44,64,149,197,198,199,200]. Kaempferol dose-dependently and sex-dependently limited Aβ-induced mitochondrial toxicity in neurons, improving rodent memory in the Y-maze test [57,134,201]. Of note, studies regarding kaempferol’s direct influence on tau are limited; thus, more research is necessary. However, due to its similar phenolic structure to quercetin [165,166], we hypothesize that kaempferol could also reduce tau hyperphosphorylation.
Figure 3. The chemical structure of kaempferol, deduced from PubChem [202].
Figure 3. The chemical structure of kaempferol, deduced from PubChem [202].
Biology 12 01453 g003

5. Kaempferol, Quercetin, and Neuroplasticity

The aberrant brain changes described in Section 3 can impair memory and cognitive function by creating deficits in neuroplasticity. Thus, future AD treatments should also be designed to directly target signaling pathways that can counteract the etiologies of AD. Specifically, we identified the PI3K/AKT signaling pathway as a critical candidate to counteract neurodegeneration. Several studies have suggested that flavonoids can alleviate learning and memory deficits by targeting this signaling pathway [29,203,204,205]. However, other pathways, including the MAPK-ERK1/2 cascade [206], have also been proposed and outlined in a recent review [207]. In this section, we will first explore the impact of Aβ- and tau-mediated neuroinflammation on synaptic plasticity-related neuronal signaling. We will support the necessity of the PI3K/AKT/GSK-3β pathway in AD treatments and investigate the potential roles of kaempferol and quercetin in improving memory and cognition through this pathway.

5.1. Neuroplasticity Deficits in AD

An ideal AD treatment should enhance the expression of plasticity-related genes such as BDNF, a neurotrophic factor that regulates neuronal plasticity and survival [208,209,210,211,212,213,214]. BDNF signaling begins with its binding to the receptor, Trkβ, activating signaling via a variety of pathways like PI3K/AKT [211,215]. Then, AKT or protein kinase B (PKB) [216] can activate the CREB-mediated transcription of BDNF [217,218]. Since Trkβ receptors mediate the pro-neuroplastic effects of BDNF [219], AD drugs must produce a direct or indirect effect on the receptor.
BDNF deficits increase the risk of AD development [220], and BDNF dysfunction due to impaired PI3K and AKT signaling can expedite neurodegeneration [7,41,221,222,223]. The PI3K/AKT signaling pathway has multiple functions, including regulating synaptic plasticity, glucose processing, cell cycle progression, cell proliferation, survival, and apoptosis [167,175,224,225,226]. Moreover, this pathway may protect neurons from Aβ toxicity [224], oxidative stress [227], and neuroinflammation [217]. GSK-3β is downstream of PI3K/AKT, and Aβ can specifically lead to its hyperactivity [7]. However, BDNF and CREB are also vulnerable to Aβ signaling [228] as CREB is regulated by the PI3K/AKT/GSK-3β pathway [211,212,215,229,230,231].
Thus, the Aβ-mediated signaling cascade that degenerates the neuron is as follows (Figure 4A): Aβ binding to NMDAR inhibits PI3K/AKT signaling by activating GSK-3β-mediated tau hyperphosphorylation and CREB downregulation [93,97,210,211,223,229,230,232,233,234,235,236,237]. Consequently, the impaired CREB-mediated transcription of BDNF genes decreases plasticity and facilitates plaque accumulation, as demonstrated in postmortem tissues from humans and human neuronal cells [209,210,229,232]. The absence of protective BDNF and PI3K/AKT activity facilitates the caspase-mediated pro-apoptotic signaling cascade [6,224], degenerating the neuronal circuitry, while tau dissociation from microtubules breaks down the neuronal cytoskeleton [7,233,238,239,240,241,242].
However, future AD treatments could reverse this toxic signaling via the following mechanism: A drug must either directly activate Trkβ or should do so indirectly by enhancing BDNF transcription [210]. The drug can either directly activate PI3K and/or AKT, which would ultimately inhibit GSK-3β via the phosphorylation of its Ser9 residue [224]. In turn, AKT can also inhibit caspase-9 and Bcl-3 to inhibit pro-apoptotic signaling [243,244,245,246]. One study showed that the GSK-3β inhibitor, AR-A014418 (ARA), inhibited BACE1-mediated APP cleavage into Aβ proteins in rodents [48], supporting the necessity of a GSK-3β-inhibiting drug for the treatment of AD. Finally, GSK-3β inhibition also reversed oxidative stress [93,247]. In short, the PI3K/AKT pathway can not only reverse neuroinflammation but can also counteract Aβ-mediated tau hyperphosphorylation by inhibiting GSK-3β.

5.2. Quercetin and Kaempferol Resolve AD-Related Plasticity Deficits

The multimodal mechanisms of kaempferol and quercetin collectively slow neurodegeneration by combating the impairments that are illustrated in Figure 4A and are described in Table 1. Specifically, the restoration of proper PI3K/AKT signaling will greatly improve synaptic plasticity deficits in AD [7]. While quercetin’s interaction with each component of this signaling pathway has already been documented [7], kaempferol’s mechanisms are still unclear. However, since kaempferol’s structure is similar to that of quercetin [165], we propose that kaempferol has a nearly identical mechanism with respect to the signaling pathway in this subsection. Finally, we will propose the potential outcomes of these molecular interactions.
Molecular docking studies suggested that quercetin can bind PI3K, AKT, and GSK3β [213,250,255,256,257,260,264,265]. Specifically, quercetin can bind to PI3K [256], consequently activating AKT signaling [265], or quercetin can directly bind to AKT [257]. In preclinical studies, quercetin reduced GSK-3β activity, which decreased tau hyperphosphorylation and reduced pro-apoptotic signaling [7,38,159]. Quercetin treatment in rodents also increased BDNF, Trkβ, PI3K, and AKT expression [243,266]. Consequently, quercetin enhanced neurite outgrowth in hippocampal neurons [36] and ameliorated the stress-induced downregulation of CREB and BDNF [40], suggesting that quercetin could potently replenish neuroplasticity in the AD brain. Moreover, quercetin inhibited Aβ by restoring Trkβ signaling and CREB-mediated BDNF transcription, increasing the viability of SH-SY5Y cells [252]. Finally, quercetin’s dual pro-neuroplastic and anti-inflammatory effects may also be related to the quercetin-mediated downregulation of BACE1 expression via the inhibition of NF-kB [253,254,264,267]. Taken together, these data suggest that quercetin antagonizes Aβ-induced GSK-3β signaling relative to tau by activating the PI3K/AKT pathway and directly inhibiting GSK-3β [7,225,241,255,256,260]. Consequently, proper BDNF levels can be restored to replenish neuronal plasticity in the AD brain. Similar chemicals, such as epigallocatechin-3-gallate (EGCG), attenuated tau hyperphosphorylation in a similar mechanism [23,268,269,270]. Thus, quercetin clearly has dual neuroprotective and pro-neuroplastic mechanisms in cells [33,65,252], and the clinical outcomes of quercetin’s pro-neuroplastic mechanisms were supported by its memory and cognition-boosting effects in rodent models of AD and Parkinson’s disease [23,38,44,271,272,273,274,275,276]. Select molecular targets of quercetin are described in Table 1.
Kaempferol may have similar pro-neuroplastic mechanisms to quercetin, and some of its molecular targets are outlined in Table 1. First, kaempferol improved hippocampal plasticity following traumatic brain injury in young rodents [277] and improved memory in rodents [29,57] and Drosophila [173]. Moreover, kaempferol dose-dependently maintained cell viability following Aβ treatment in multiple studies [29,149,195,248]. This could be due to kaempferol’s inhibition of BACE1-mediated Aβ synthesis [253,254] or the activation of the PI3K/AKT signaling pathway, enhancing CREB-mediated BDNF transcription [175,211,258]. Although one molecular docking study suggested that kaempferol may have minimal affinity for GSK-3β [250], kaempferol likely inhibits GSK-3β indirectly by first binding and activating PI3K [256] or AKT [175,185,257]. Via this mechanism, kaempferol prevents tau hyperphosphorylation, protecting neuronal morphology and function [47,278,279,280,281]. Then, AKT can activate CREB-mediated BDNF transcription [217]. Supporting this pro-neuroplastic mechanism, kaempferol and its metabolite, kaempferide, produced similar effects that resulted in Trkβ signaling [171,210] and enhanced BDNF expression in Aβ-treated mice [243]. Taken together, these data suggest that kaempferol enhances neuroplasticity to reverse Aβ damage by activating the PI3K/AKT cascade, which potentiates CREB-mediated BDNF transcription. However, kaempferol produces the opposite effect on this signaling pathway in microglial cells [167] and cancer cells [282]. Thus, kaempferol’s effects on the PI3K/AKT signaling cascade are dynamic and depend on cell lineage.
Despite the lack of literature demonstrating a direct modulation of tau by kaempferol, there is plenty of evidence to support the possibility that kaempferol inhibits tau hyperphosphorylation via the PI3K/AKT pathway and by antagonizing Aβ-mediated GSK-3β signaling [29,149,195]. This mechanism prevents neuronal degeneration and a loss of synaptic plasticity. Thus, the pro-neuroplastic effect of kaempferol requires the inhibition of GSK-3β and CREB phosphorylation. Remarkably, a recent molecular docking study suggested that kaempferol could bind to NMDAR [259]. However, in vivo studies are still required to confirm this effect.
These data suggest a clear anti-AD mechanism of quercetin and kaempferol, as outlined in Figure 4B. First, quercetin and kaempferol could enter the cell cytoplasm due to their lipophilic polyphenolic structure. Quercetin and kaempferol scavenge ROS and activate PI3K/AKT signaling to inhibit GSK-3β. Specifically, they can bind directly to PI3K or AKT to activate protective signaling, inhibiting GSK-3β and preventing tau hyperphosphorylation. This signaling cascade reduces the formation of NFTs in the AD brain. GSK-3β inhibition can also antagonize Aβ-NMDAR interactions. Thus, downstream pro-apoptotic signaling mediators are also inhibited by quercetin and kaempferol treatment. Due to reduced NFT and amyloid plaque formation, microglial hyperactivity decreases in the absence of the burden of clearance. Thus, progressive neuroinflammatory signaling is slowed, allowing surrounding neuronal synapses to survive. After chronic quercetin treatment, progressive elevations in BDNF release rebuild damaged synapses by favoring neurotrophic signaling over cytotoxic Aβ signaling, improving memory and cognition. Of note, molecular docking studies have not supported the possibility that kaempferol and quercetin can directly bind to tau protein, supporting their indirect inhibitory mechanism via GSK-3β inhibition. Taken together, kaempferol and quercetin share multiple mechanisms that slow AD progression by first limiting ROS activity, NFT aggregation, and Aβ-mediated toxic signaling, slowing neurodegeneration.
Figure 4. (A) Neuroplasticity deficits accelerate AD progression and must be treated. Impaired PI3K-AKT signaling facilitates GSK3β-mediated phosphorylation of tau. Aβ may potentiate tau hyperphosphorylation via GSK3β. (B) Kaempferol and quercetin (K/Q) invoke the PI3K/AKT pathway to antagonize Aβ and reduce tau hyperphosphorylation in neurons. As a result, neuroplasticity is increased in the AD brain [283].
Figure 4. (A) Neuroplasticity deficits accelerate AD progression and must be treated. Impaired PI3K-AKT signaling facilitates GSK3β-mediated phosphorylation of tau. Aβ may potentiate tau hyperphosphorylation via GSK3β. (B) Kaempferol and quercetin (K/Q) invoke the PI3K/AKT pathway to antagonize Aβ and reduce tau hyperphosphorylation in neurons. As a result, neuroplasticity is increased in the AD brain [283].
Biology 12 01453 g004

6. Quercetin and Kaempferol in Common Herbs

Although data on the co-treatment of quercetin and kaempferol are still somewhat limited, the abundance of both compounds in several common herbs requires the investigation of the synergistic effects of both flavonoids, in addition to their interactions with other herbal phytochemicals. Flavonoid-rich herbs are commonly employed in traditional Chinese medicine (TCM), in which an emphasis is placed on the utility of natural treatments. Moreover, these herbs are generally safe for consumption [224]. Kaempferol is the second most common flavonoid in traditional medicinal herbs, following quercetin [225,284]. Other reviews have assessed the efficacy and safety of natural medicine in the treatment of neurodegenerative diseases [7,224], highlighting the potential medicinal properties of herbs in treating AD. Flavonoids are commonly found in herbs such as Schima wallichii Korth, Maesa membranacea, Ginkgo biloba, and many more [175,225,278]. These phytochemicals could work synergistically with each other and with other herbal components to invoke anti-AD effects. Thus, we explore common herbal sources of kaempferol and quercetin, describe the anti-AD mechanisms of herbs, and propose a design for a future AD treatment based on the current evidence of these effects.
Ginkgo biloba is a quercetin- and kaempferol-rich herb proposed to treat AD [285]. G. biloba improves memory and cognition by inhibiting ROS, facilitating hippocampal neuron proliferation, halting Aβ plaque accumulation, and reducing tau hyperphosphorylation [47,286,287,288]. Moreover, this effect is associated with reduced GSK-3β activity and the increased expression of PSD-95 and synapsin-1 [47]. As seen with kaempferol and quercetin alone, G. biloba potentiates PI3K/AKT relative to CREB signaling to promote neuroplasticity [287,289,290,291,292,293]. Hippophae rhamnoides extracts are also rich in quercetin and kaempferol, and they enhanced neuronal differentiation and neurite outgrowth via PI3K/AKT and ERK signaling [294,295]. However, clinical trials have revealed the inconsistent efficacy of G. biloba on cognition and other AD-related parameters [296]. Camellia sinensis is another kaempferol- and quercetin-rich herb commonly grown to produce black and green tea [297,298]. C. sinensis extracts improved spatial memory and reduced hippocampal Aβ fibrillization in AD rodents and had greater antioxidant effects compared to other herbs [298,299]. Kaempferol and its derivatives are found in the leaves of Maesa membranacea, Schima wallichii Korth, Carthamus tinctorius, Panax ginseng, and several other herbs [175,188,225,278]. S. wallichii was neuroprotective due to the promotion of hippocampal and cortical AKT signaling [175], and M. membranacea could protect H202-treated SH-SY5Y cells [225] and hippocampal tissue [300] via the same pathway due to their kaempferol abundance. C. tinctorus is rich in kaempferol, produces a similar effect, and invokes protective AMPK signaling [188]. Finally, recent studies also suggested that other herbs such as Morenga oleifera, Cuscuta chinensis, Allium cepa, Litchi chinensis, Prakia roxburghii, Radix astragali, Acoritatan Fagopyrum tataricum, Carthami flos, Punica granatum, and Cyperi rhizoma [251,257,264,301,302,303,304,305] may also be great sources of kaempferol and/or quercetin and produce anti-AD effects. Their medicinal properties and expression of kaempferol and quercetin are outlined in Table 2.
Polyherbal cocktails, such as Chaihu shugan san (CSS) and Huangqi Sijunzi (HQSJDZ), could treat AD and its risk factors. CSS is abundant in kaempferol and quercetin and contains herbs such as Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum [256]. Specifically, the antidepressant effect of CSS is mediated by increased PI3K/AKT/BDNF signaling and decreased GSK-3β and IL-2 activity [256], suggesting that polyherbal cocktails may be protected from AD development. HQSJDZ, rich in kaempferol and quercetin, had cholinergic, anti-inflammatory, and anti-GSK-3β effects [278,306]. Moreover, a cocktail of C. sinensis, Hypericum perforatum, and Bacopa monnieri produced robust antioxidant effects compared to single-herb treatment [298]. These data suggest that polyherbal treatment may be superior to single-herb therapy.
Due to the well-documented effects of quercetin and kaempferol on Aβ, GSK-3β, PI3K/AKT, and multiple pro-inflammatory molecules, it is possible that both phytochemicals, given their abundance, contribute vastly to the anti-AD effects of several herbs. Such herbs include Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum. The herbal sources outlined in Table 2 may also be great additions to the treatment protocol that can enhance the dietary intake of kaempferol and quercetin. According to the practice of TCM, it is possible that a multi-herb cocktail containing varying amounts of these herbs could alleviate AD symptoms, as seen with current medications, but it may also halt progression relative to a unique multi-modal mechanism. Multiple studies have suggested that the synergistic effects of polyherbal treatments produce greater anti-AD efficacy compared to single-herb treatment [256,278,298]. Thus, the research and development of future AD drugs should consider the applications of these common herbs in future drug cocktails. On the other hand, since clinical trials featuring Ginkgo biloba extracts have demonstrated controversial results on the progression of AD [296], single-herb treatments may be insufficient to treat AD.
Table 2. Plant sources of kaempferol and quercetin and/or their metabolites and a description of reported herbal health effects.
Table 2. Plant sources of kaempferol and quercetin and/or their metabolites and a description of reported herbal health effects.
Species NameKaempferolQuercetinExample Health EffectsReference
Ginkgo biloba++Memory and cognition improvement[285,296,307,308]
Camellia sinensis++Improved memory and antioxidant effects[297,298,299]
Maesa membranacea++Neuroprotective[175,188,225,278]
Schima wallichii Korth+Neuroprotective[175,187,225,278]
Carthamus tinctorius++Neuroprotective[175,187,225,278,309]
Panax ginseng++Neuroprotective[175,187,225,278,310]
Morenga oleifera++Memory improvement[300,301,302]
Cuscuta chinensis++Memory improving,
Neuroprotective, Hepatoprotective,
Immunomodulatory
[311]
Allium cepa++Anti-inflammatory[312,313]
Hippophae rhamnoides L.++Anti-inflammatory[294,295]
Litchi chinensis++Neuroprotective[303,314]
Prakia roxburghii-+Neuroprotective[304]
Radix astragali++Neuroprotective[213]
Fagopyrum tataricum (L.)++Decrease neurotoxicity[251]
Carthami flos++Anti-ischemic[213]
Punica granatum++Anti-inflammatory[264,315]
Cyperi rhizoma++Antidepressant[257]

7. Limitations of Kaempferol and Quercetin Treatment

7.1. Bioavailability

Despite the promising effects of these herbs and flavonoids in AD treatment, low bioavailability and blood-brain barrier (BBB) permeability are common obstacles interfering with drug delivery to the brain [316,317,318]. Thus, structural manipulations are commonly required to improve the bioavailability of flavonoids. Moreover, the varying dietary intake of macromolecules like fats and carbohydrates also impacts BBB permeability relative to polyphenols [44]. Other factors, such as aging or diagnosis with AD, may increase BBB permeability to peripheral chemicals [39,319,320,321]. However, tau hyperphosphorylation and astrocytic hyperactivity invoke neuroinflammatory signaling that damages BBB integrity and increases its permeability [112,322,323,324]. The limited BBB permeability may also explain the lack of clinical trials in humans [68].
Despite its lipophilicity and easy oral administration in common foods, quercetin treatment for AD may be challenged by its limited bioavailability relative to the brain [3,258]. Since quercetin absorption is predominantly mediated by the small intestine, it is vulnerable to extensive first-pass metabolism [133,258,325]. While its distribution was evidenced in the plasma, liver, heart, spleen, kidneys, and lungs, quercetin levels were non-detectable in the rat brain [326,327]. Hence, it has around 65% BBB permeability [321,328] and is absorbed in the stomach with 24–53% bioavailability [329]. The P-glycoprotein transporter, which is a BBB efflux transporter, has a high affinity for free, unaltered quercetin and greatly reduces its bioavailability by pumping quercetin away from the brain [51,330]. While in vitro studies showed the promising antioxidant effects of quercetin, most studies in animal models have demonstrated limited efficacy [3,331]. These data show that quercetin’s limited bioavailability could debilitate anti-AD effects [258].
Chemical modifications are necessary to ensure quercetin distribution to the brain, as some metabolites may also have higher efficacy than quercetin alone. For instance, quercetin–glucoside conjugation enhanced its bioavailability [129]. Quercetin glycosides are commonly available in fruits and vegetables, improving its delivery to the CNS [51,332]. Glucuronidation in the liver also increased the distribution of quercetin to the brain in oxidative stress models [23]. Moreover, in vivo studies showed that lipid nanoparticle-loaded quercetin enhances its entry into the brain [39,44,146,158,163,333,334]. Moreover, quercetin loading into selenium nanoparticles improved brain distribution and anti-Aβ mechanisms [335]. However, excess selenium levels in the body can produce oxidative stress [336,337], potentially limiting the clinical efficacy of this approach.
Like quercetin, free kaempferol generally has low oral bioavailability due to metabolic degradation [324,338,339]. Kaempferol is generally slowly absorbed in the GI tract and can be distributed to several tissues [326,340], suggesting that the primary limitation of kaempferol treatment is limited bioavailability. However, several modifications to improve its BBB permeability have been proposed. First, nanoparticle loading also improves kaempferol bioavailability [194,334,341,342,343,344], and kaempferol–sugar conjugates also demonstrate superior protective efficacy [36]. For instance, nanoparticle-loaded kaempferol has more robust anti-inflammatory effects than kaempferol alone [68]. Clinical trials revealed that quercetin had superior memory-modulating activity in AD patients compared to healthy elderly controls [345,346,347], suggesting that the increased BBB permeability in AD may, in turn, improve flavonoid bioavailability and efficacy in neurodegenerative brains. Several other forms of delivery have been proposed for both flavonoids, including gold-infused nanoparticles [348,349], multi-targeted drugs [350], extracellular vesicles [351], and intranasal administration [352]. Finally, other proposed nanoformulation delivery systems include nanomatrixes, nanoemulsions, nanostructured lipid carriers, and nanocomplexes [343,344,353].

7.2. Adverse Health Effects and Other Limitations

Most studies show promising medical benefits for kaempferol and quercetin and suggest that they are safe in a variety of doses. For example, quercetin is included in the Food and Drug Administration’s Generally Recognized as Safe (GRAS) list for supplemental use of up to 500 mg per serving in foods and beverages [129,354]. However, flavonoids’ clinical efficacy may also be limited by adverse effects [329]. While the Ames test suggested that quercetin could have carcinogenic properties, most studies have opposed this finding and suggested that quercetin is safe [355]. One study suggested that high-dose quercetin treatment reduced neuronal survival, induced oxidative stress, and inhibited AKT [356]. Thus, physicians should carefully manage the abundance of quercetin in the AD patient’s diet to maintain its proper anti-degenerative effects. Moreover, the efficacy of quercetin may be limited in AD patients who are also diagnosed with leukemia, as quercetin inhibits the PI3K/AKT signaling pathway in HG3 cells [282]. It is possible that, since most dietary quercetin is distributed to peripheral sites, lower concentrations in the brain may decrease its efficacy in AD.
Although kaempferol is most likely safe to consume [339] and most studies showed low toxicity in mice [357,358,359], some studies have reported concerns about potential mutagenic effects in people with iron and folic acid deficiencies [338,339,360]. Since the excess inhibition of GSK-3β may produce toxic effects in cells [233], kaempferol’s low-affinity GSK-3β interactions may underlie its generally low toxicity. In a 4-week randomized, double-blind clinical trial, participants were divided into a group that received 50 mg of kaempferol daily and a placebo group; kaempferol was reported as mostly safe, but the small sample size of 24 in each group limits this study [359]. Overall, the majority of work on the herb suggests it to be safe, even in high doses, but more clinical trials are highly recommended.

8. Discussion

Since AD still lacks a true cure, and currently available medications are insufficient to halt disease progression, the field has sought out multimodal treatments for AD. However, little progress in drug development has been made in recent decades, necessitating new alternative treatments. Thus, the objective of this review was to deduce the anti-AD mechanisms of kaempferol and quercetin. These phytochemicals were selected for multiple reasons, including their abundance [38,116] and their multimodal mechanisms (Figure 5) that include antioxidant, anti-inflammatory, pro-neuroplastic, and neuroprotective effects. Thus, quercetin and kaempferol may treat Alzheimer’s disease, and we aimed to explore their anti-amyloidogenic, antioxidant, anti-inflammatory, anti-tau, and pro-neuroplastic mechanisms [6,29,38,39,51,127,128,149,159,167,361]. In turn, phytochemicals may not only reduce AD symptoms [29,33,132] but also delay the progression of the disorder. Of note, the efficacy of these flavonoids to produce the effects outlined in this review depends on any chemical modifications that may occur throughout the absorption and distribution of phytochemicals to the brain.
Perhaps the most significant contribution of this review is the complex anti-degenerative mechanism of kaempferol. We utilized the available literature to show that kaempferol’s dual anti-tau and anti-Aβ mechanisms are due to its modulation of the PI3K/AKT/GSK-3β signaling pathway. Both phytochemicals resolve oxidative stress by increasing antioxidant levels and inhibiting ROS signaling [119]. Meanwhile, they halt inflammatory signaling [29,38] to commence a neuroprotective effect. Then, resolved microglial and astrocytic activity facilitates proper Aβ clearance from the brain [6] and reduces continued neuronal damage due to the neuroinflammatory environment [122,188,195]. The modulation of PI3K/AKT/GSK-3β and Trkβ/BDNF signaling potentiates neuroplasticity and protects neurons from insults like Aβ [10,240], decreasing tau hyperphosphorylation and preserving the neuronal cytoskeletal structure. These phytochemicals, in turn, protect neuronal networks [33,40], improving memory and cognitive function in AD patients. Other flavonoids with heterocyclic structures [362], including morin [363,364,365,366], rutin [367,368], and luteolin [369,370,371], share many similar anti-AD properties relative to kaempferol and quercetin. However, rutin [368] failed to increase BDNF levels, like kaempferol and quercetin.
Due to the superior efficacy of polyherbal treatments, such as HQSJDZ and CSS [256,278,298], we proposed that polyherbal treatment, containing quercetin- and kaempferol-rich herbs like Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum may produce superior anti-AD efficacy compared to single-herb supplements. Recent studies also suggested that herbs such as Morenga oleifera, Cuscuta chinensis, Allium cepa, Hippophae rhamnoides, Litchi chinensis, Prakia roxburghii, Radix astragali, Fagopyrum tataricum, and Carthami flos [251,294,301,302,303,304] may also be candidates for polyherbal treatment. However, a recent review noted that kaempferol and quercetin are widely available in hundreds of herbs, and it is possible that they may not be as abundant as other phytochemicals in some species [372], supporting the necessity of polyherbal treatment to obtain biologically effective concentrations.
As previously mentioned, clinical trials suggest that kaempferol and quercetin could treat AD in humans [135,346,347,373,374], but single-herb treatment was unsuccessful in clinical trials [296]. Future trials should assess bioavailability-enhancing delivery methods for quercetin and kaempferol. However, recent studies also suggested that both quercetin and kaempferol have the ability to maintain and protect BBB integrity [375,376,377,378,379]. This could possibly be due to their anti-inflammatory properties that could be invoked if they reach the brain. Of course, clinical trials should continue to assess the efficacy of herbal sources in AD-related symptoms. However, the misuse of herbal treatments may produce side effects, including gastrointestinal discomfort, insomnia, and tachycardia [298]. Thus, studies assessing these side effects are limited and require further investigation [36,37]. Nonetheless, these natural herbs are generally considered safe, and toxic effects are uncommon [51,116,142]. Finally, an investigation of interactions between these polyphenols and other drugs commonly prescribed to AD patients is required.
Although the data presented in this review showcase the great potential of these herbs in AD treatment, a few limitations have impacted this review. Specifically, studies investigating the tau hyperphosphorylation-inhibiting mechanisms of these herbs may be limited due to the rapid dephosphorylation of the protein in postmortem AD tissues [15,279]. Moreover, the abundantly described bioavailability limitations of both herbs critically limit the efficiency of human studies. This could be one reason underlying the lack of kaempferol and quercetin’s clinical efficacy to date. Clinical trials investigating compounds that increase the bioavailability of these phytochemicals are still needed. Since quercetin and kaempferol are naturally abundant in the average diet, future clinical trials can be easily conducted. Finally, while molecular docking studies show the potential pharmacodynamic interactions between kaempferol/quercetin and the outlined pro-neuroplastic targets, these approaches are merely estimates of binding affinity based on the crystal structures of the target protein and the molecular structures of the ligand, and they could be vulnerable to mispredictions [380]. Thus, future studies must either employ competition assays or ligand inhibitor/antagonist studies to confidently elucidate the true affinity of kaempferol and quercetin for the targets of interest. Nonetheless, recent data support the exciting potential of kaempferol and quercetin to slow the progression of AD and alleviate the symptoms.

9. Conclusions

Kaempferol and quercetin clearly exhibit multimodal mechanisms that halt AD progression and alleviate symptoms. Given the multifaceted nature of AD pathogenesis, future treatments need to adopt a multimodal approach that targets the Aβ-tau signaling pathway via the modulation of the PI3K/AKT/GSK3β signaling cascade, leading to a pro-neuroplastic effect via enhanced BDNF signaling. To our knowledge, our review demonstrates how kaempferol and quercetin address various aspects of AD, including neuroinflammation, oxidative stress, reduced plasticity, and Aβ and tau signaling. Notably, our review is the first to propose that kaempferol can mitigate both tau hyperphosphorylation and Aβ toxicity by directly targeting the PI3K/AKT/GSK3β pathway. Additionally, we suggest that polyherbal cocktails rich in kaempferol and quercetin may yield robust anti-AD effects, and we identified potential herbal sources of kaempferol and quercetin. Finally, we discuss the limitations that currently impede the efficacy of kaempferol/quercetin treatment, and suggest potential adjustments to circumvent these challenges. Together, these changes can improve the anti-AD efficacy of natural flavonoids and could be ideal adjunctive or alternative treatments to currently available drugs.

Author Contributions

Conception, design of this review, and final editing of the manuscript, M.V.; Collecting recent reports and writing the original draft, C.A. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Department of Biology at Lamar University for their support.

Conflicts of Interest

The authors declare no conflict of interest.

Herbs Manuscript Abbreviations

amyloid beta
AChEIsacetylcholinesterase inhibitors
AChEacetylcholinesterase
AChacetylcholine
ADAlzheimer’s disease
AKTprotein Kinase B
AMPKAMP-activated protein kinase
APPamyloid precursor protein
BACE1beta-site APP cleaving enzyme 1
Baxbcl-2-like protein 4
BBBblood-brain barrier
BDNFbrain-derived neurotrophic factor
Cdk5cyclin-dependent kinase 5
p-Cdk5phosphorylated forms of Cdk5
CNScentral nervous system
COX-1cyclooxygenase-1
COX-2cyclooxygenase-2
CREBcAMP response element-binding protein
CSSChaihu shugan san
EGCGepigallocatechin-3-gallate
EPMelevated plus-maze test
ERK1/2extracellular receptor signal-regulated kinase 1&2
GLUT4glucose transporter type 4
GSHglutathione
GSK3βglycogen synthase kinase-3 beta
GPxglutathione peroxidase
HT22immortalized mouse hippocampal cell line
HO-1heme oxygenase-1
HQSJDZ Huangqi Sijunzi
H2O2hydrogen peroxide
IDEinsulin-degrading enzyme
IFN-γinterferon gamma
IL-1βinterleukin-1β
IL-2interleukin-2
iNOSinducible nitric oxide synthase
ICRstrain of Swiss mice produced at the Institute of Cancer Research
IRinsulin resistance
I/Rcerebral ischemia/reperfusion
IRS1insulin response substrate-1
JNKc-Jun N-terminal kinase
KAGkaempferol 3-O-(6″-acetyl)-β-glucopyranoside
KATlysine acetylase
KDAClysine deacetylase
K-3-Rhkaempferol-3-O-rhamnoside
K/Qkaempferol and quercetin co-treatment
LPSlipopolysaccharide
PGE2prostaglandin E2
PI3Kphosphoinositide 3-kinases
PI3K/AKT/GSK-3βphosphoinositide 3-kinase/protein kinase B/glycogen synthase kinase-3 beta signaling pathway
PKCprotein kinase C
PP2Aprotein phosphatase 2
PSD-95postsynaptic density protein 95
MAPmicrotubule-associated protein
MAPKmitogen-activated protein kinase
MDAmalondialdehyde
MLK2mixed lineage kinase 2
NF-kBnuclear factor kappa B
NFTsneurofibrillary tangles
NMDARsN-methyl-d-aspartate receptors
NOnitric oxide
Nrf2nuclear factor erythroid 2-related factor 2
NR2BN-methyl d-aspartate receptor subtype 2B
PETpositron emission tomography
PON2paroxonase 2
ROSreactive oxygen species
SIRT1Sirtuin 1
Serserine
Ser9serine 9
SODsuperoxide dismutase
STZstreptozotocin
TCMtraditional Chinese medicine
Thrthreonine
TLRstoll-like receptors
TLR2toll-like receptor 2
TLR4toll-like receptor 4
TLR9toll-like receptor 9
TNF-αtumor necrosis factor-α
Trkβtropomycin-related kinase β
3 × Tg AD micetriple transgenic Alzheimer’s disease mice

References

  1. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef] [PubMed]
  2. Tamagno, E.; Guglielmotto, M.; Vasciaveo, V.; Tabaton, M. Oxidative Stress and Beta Amyloid in Alzheimer’s Disease. Which Comes First: The Chicken or the Egg? Antioxidants 2021, 10, 1479. [Google Scholar] [CrossRef] [PubMed]
  3. Riche, K.; Lenard, N.R. Quercetin’s Effects on Glutamate Cytotoxicity. Molecules 2022, 27, 7620. [Google Scholar] [CrossRef]
  4. Yu, H.; Wu, J. Amyloid-β: A double agent in Alzheimer’s disease? Biomed. Pharmacother. 2021, 139, 111575. [Google Scholar] [CrossRef] [PubMed]
  5. Ozben, T.; Ozben, S. Neuro-inflammation and anti-inflammatory treatment options for Alzheimer’s disease. Clin. Biochem. 2019, 72, 87–89. [Google Scholar] [CrossRef] [PubMed]
  6. Minter, M.R.; Taylor, J.M.; Crack, P.J. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J. Neurochem. 2016, 136, 457–474. [Google Scholar] [CrossRef]
  7. Kitagishi, Y.; Nakanishi, A.; Ogura, Y.; Matsuda, S. Dietary regulation of PI3K/AKT/GSK-3β pathway in Alzheimer’s disease. Alzheimer's Res. Ther. 2014, 6, 35. [Google Scholar] [CrossRef]
  8. Gabbouj, S.; Ryhänen, S.; Marttinen, M.; Wittrahm, R.; Takalo, M.; Kemppainen, S.; Martiskainen, H.; Tanila, H.; Haapasalo, A.; Hiltunen, M.; et al. Altered Insulin Signaling in Alzheimer’s Disease Brain—Special Emphasis on PI3K-Akt Pathway. Front. Neurosci. 2019, 13, 629. [Google Scholar] [CrossRef]
  9. Bhaskar, K.; Miller, M.; Chludzinski, A.; Herrup, K.; Zagorski, M.; Lamb, B.T. The PI3K-Akt-mTOR pathway regulates Abeta oligomer induced neuronal cell cycle events. Mol. Neurodegener. 2009, 4, 14. [Google Scholar] [CrossRef]
  10. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef]
  11. Moldogazieva, N.T.; Mokhosoev, I.M.; Mel'nikova, T.I.; Porozov, Y.B.; Terentiev, A.A. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxid. Med. Cell. Longev. 2019, 2019, 3085756. [Google Scholar] [CrossRef]
  12. Uddin, M.S.; Kabir, M.T. Oxidative Stress in Alzheimer’s Disease: Molecular Hallmarks of Underlying Vulnerability. In Biological, Diagnostic and Therapeutic Advances in Alzheimer’s Disease; Ashraf, G., Alexiou, A., Eds.; Springer: Singapore, 2019. [Google Scholar] [CrossRef]
  13. Youssef, P.; Chami, B.; Lim, J.; Middleton, T.; Sutherland, G.T.; Witting, P.K. Evidence supporting oxidative stress in a moderately affected area of the brain in Alzheimer’s disease. Sci. Rep. 2018, 8, 11553. [Google Scholar] [CrossRef]
  14. Gao, W.; Wang, W.; Peng, Y.; Deng, Z. Antidepressive effects of kaempferol mediated by reduction of oxidative stress, proinflammatory cytokines and up-regulation of AKT/β-catenin cascade. Metab. Brain Dis. 2019, 34, 485–494. [Google Scholar] [CrossRef] [PubMed]
  15. Alquezar, C.; Arya, S.; Kao, A.W. Tau Post-translational Modifications: Dynamic Transformers of Tau Function, Degradation, and Aggregation. Front. Neurol. 2021, 11, 595532. [Google Scholar] [CrossRef] [PubMed]
  16. Merino-Serrais, P.; Benavides-Piccione, R.; Blazquez-Llorca, L.; Kastanauskaite, A.; Rábano, A.; Avila, J.; DeFelipe, J. The influence of phospho-tau on dendritic spines of cortical pyramidal neurons in patients with Alzheimer’s disease. Brain 2013, 136, 1913–1928. [Google Scholar] [CrossRef] [PubMed]
  17. Spittaels, K.; Haute, C.V.D.; Van Dorpe, J.; Geerts, H.; Mercken, M.; Bruynseels, K.; Lasrado, R.; Vandezande, K.; Laenen, I.; Boon, T.; et al. Glycogen synthase kinase-3β phosphorylates protein tau and rescues the axonopathy in the central nervous system of human four-repeat tau transgenic mice. J. Biol. Chem. 2000, 275, 41340–41349. [Google Scholar] [CrossRef]
  18. Tatebayashi, Y.; Haque, N.; Tung, Y.-C.; Iqbal, K.; Grundke-Iqbal, I. Role of tau phosphorylation by glycogen synthase kinase-3β in the regulation of organelle transport. J. Cell Sci. 2004, 117, 1653–1663. [Google Scholar] [CrossRef]
  19. Jaworski, T.; Kügler, S.; van Leuven, F. Modeling of tau-mediated synaptic and neuronal degeneration in Alzheimer’s disease. Int. J. Alzheimer's Dis. 2010, 2010, 573138. [Google Scholar] [CrossRef]
  20. Hoover, B.R.; Reed, M.N.; Su, J.; Penrod, R.D.; Kotilinek, L.A.; Grant, M.K.; Pitstick, R.; Carlson, G.A.; Lanier, L.M.; Yuan, L.-L.; et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 2010, 68, 1067–1081. [Google Scholar] [CrossRef]
  21. Thies, E.; Mandelkow, E.-M. Missorting of tau in neurons causes degeneration of synapses that can be rescued by the kinase MARK2/Par-1. J. Neurosci. 2007, 27, 2896–2907. [Google Scholar] [CrossRef]
  22. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2013, 79, 1–12. [Google Scholar] [CrossRef] [PubMed]
  23. Sabogal-Guáqueta, A.M.; Muñoz-Manco, J.I.; Ramírez-Pineda, J.R.; Lamprea-Rodriguez, M.; Osorio, E.; Cardona-Gómez, G.P. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015, 93, 134–145. [Google Scholar] [CrossRef]
  24. Reitz, C.; Brayne, C.; Mayeux, R. Epidemiology of Alzheimer disease. Nat. Rev. Neurol. 2011, 7, 137–152. [Google Scholar] [CrossRef] [PubMed]
  25. Joe, E.; Ringman, J.M. Cognitive symptoms of Alzheimer’s disease: Clinical management and prevention. BMJ 2019, 367, l6217. [Google Scholar] [CrossRef] [PubMed]
  26. Casey, D.A.; Antimisiaris, D.; O’Brien, J. Drugs for Alzheimer’s disease: Are they effective? Pharm. Ther. 2010, 35, 208–211. [Google Scholar]
  27. Francis, P.T. The interplay of neurotransmitters in Alzheimer’s disease. CNS Spectrums 2005, 10 (Suppl. S18), 6–9. [Google Scholar] [CrossRef]
  28. Wang, R.; Reddy, P.H. Role of Glutamate and NMDA Receptors in Alzheimer’s Disease. J. Alzheimer's Dis. 2017, 57, 1041–1048. [Google Scholar] [CrossRef]
  29. Kouhestani, S.; Jafari, A.; Babaei, P. Kaempferol attenuates cognitive deficit via regulating oxidative stress and neuroinflammation in an ovariectomized rat model of sporadic dementia. Neural Regen. Res. 2018, 13, 1827–1832. [Google Scholar] [CrossRef]
  30. Farlow, M.R.; Miller, M.L.; Pejovic, V. Treatment options in Alzheimer’s disease: Maximizing benefit, managing expectations. Dement. Geriatr. Cogn. Disord. 2008, 25, 408–422. [Google Scholar] [CrossRef]
  31. Tian, J.; Shi, J.; Zhang, X.; Wang, Y. Herbal therapy: A new pathway for the treatment of Alzheimer’s disease. Alzheimer's Res. Ther. 2010, 2, 30. [Google Scholar] [CrossRef]
  32. Lee, J.; Jin, C.; Cho, S.Y.; Park, S.U.; Jung, W.S.; Moon, S.K.; Park, J.M.; Ko, C.N.; Cho, K.H.; Kwon, S. Herbal medicine treatment for Alzheimer disease: A protocol for a systematic review and meta-analysis. Medicine 2020, 99, e21745. [Google Scholar] [CrossRef]
  33. Chen, M.M.; Yin, Z.Q.; Zhang, L.Y.; Liao, H. Quercetin promotes neurite growth through enhancing intracellular cAMP level and GAP-43 expression. Chin. J. Nat. Med. 2015, 13, 667–672. [Google Scholar] [CrossRef]
  34. Zhang, X.W.; Chen, J.Y.; Ouyang, D.; Lu, J.H. Quercetin in Animal Models of Alzheimer’s Disease: A Systematic Review of Preclinical Studies. Int. J. Mol. Sci. 2020, 21, 493. [Google Scholar] [CrossRef]
  35. Scarmeas, N.; Stern, Y.; Tang, M.X.; Mayeux, R.; Luchsinger, J.A. Mediterranean diet and risk for Alzheimer’s disease. Ann. Neurol. 2006, 59, 912–921. [Google Scholar] [CrossRef]
  36. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef]
  37. Sreenivasmurthy, S.G.; Liu, J.Y.; Song, J.X.; Yang, C.B.; Malampati, S.; Wang, Z.Y.; Huang, Y.Y.; Li, M. Neurogenic traditional Chinese medicine as a promising strategy for the treatment of Alzheimer’s disease. Int. J. Mol. Sci. 2017, 18, 272. [Google Scholar] [CrossRef]
  38. Khan, A.; Ali, T.; Rehman, S.U.; Khan, M.S.; Alam, S.I.; Ikram, M.; Muhammad, T.; Saeed, K.; Badshah, H.; Kim, M.O. Neuroprotective Effect of Quercetin Against the Detrimental Effects of LPS in the Adult Mouse Brain. Front. Pharmacol. 2018, 9, 1383. [Google Scholar] [CrossRef]
  39. Testa, G.; Gamba, P.; Badilli, U.; Gargiulo, S.; Maina, M.; Guina, T.; Calfapietra, S.; Biasi, F.; Cavalli, R.; Poli, G.; et al. Loading into nanoparticles improves quercetin’s efficacy in preventing neuroinflammation induced by oxysterols. PLoS ONE 2014, 9, e96795. [Google Scholar] [CrossRef]
  40. Ma, Z.X.; Zhang, R.Y.; Rui, W.J.; Wang, Z.Q.; Feng, X. Quercetin alleviates chronic unpredictable mild stress-induced depressive-like behaviors by promoting adult hippocampal neurogenesis via FoxG1/CREB/ BDNF signaling pathway. Behav. Brain Res. 2021, 406, 113245. [Google Scholar] [CrossRef]
  41. Das, D.; Biswal, S.; Barhwal, K.K.; Chaurasia, O.P.; Hota, S.K. Kaempferol Inhibits Extra-synaptic NMDAR-Mediated Downregulation of TRkβ in Rat Hippocampus During Hypoxia. Neuroscience 2018, 392, 77–91. [Google Scholar] [CrossRef]
  42. Hussein, R.M.; Mohamed, W.R.; Omar, H.A. A neuroprotective role of kaempferol against chlorpyrifos-induced oxidative stress and memory deficits in rats via GSK3β-Nrf2 signaling pathway. Pestic. Biochem. Physiol. 2018, 152, 29–37. [Google Scholar] [CrossRef] [PubMed]
  43. Yu, L.; Chen, C.; Wang, L.F.; Kuang, X.; Liu, K.; Zhang, H.; Du, J.R. Neuroprotective effect of kaempferol glycosides against brain injury and neuroinflammation by inhibiting the activation of NF-κB and STAT3 in transient focal stroke. PLoS ONE 2013, 8, e55839. [Google Scholar] [CrossRef] [PubMed]
  44. Azam, S.; Jakaria, M.; Kim, I.S.; Kim, J.; Haque, M.E.; Choi, D.K. Regulation of Toll-Like Receptor (TLR) Signaling Pathway by Polyphenols in the Treatment of Age-Linked Neurodegenerative Diseases: Focus on TLR4 Signaling. Front. Immunol. 2019, 10, 1000. [Google Scholar] [CrossRef] [PubMed]
  45. Hou, Y.; Aboukhatwa, M.A.; Lei, D.L.; Manaye, K.; Khan, I.; Luo, Y. Anti-depressant natural flavonols modulate BDNF and beta amyloid in neurons and hippocampus of double TgAD mice. Neuropharmacology 2010, 58, 911–920. [Google Scholar] [CrossRef]
  46. Kim, J.H.; Kim, H.Y.; Cho, E.J. Protective effects of kaempferol, quercetin, and its glycosides on amyloid beta-induced neurotoxicity in C6 glial cell. J. Appl. Biol. Chem. 2019, 62, 327–332. [Google Scholar] [CrossRef]
  47. Zeng, K.; Li, M.; Hu, J.; Mahaman, Y.A.R.; Bao, J.; Huang, F.; Xia, Y.; Liu, X.; Wang, Q.; Wang, J.Z.; et al. Ginkgo biloba Extract EGb761 Attenuates Hyperhomocysteinemia-induced AD Like Tau Hyperphosphorylation and Cognitive Impairment in Rats. Curr. Alzheimer Res. 2018, 15, 89–99. [Google Scholar] [CrossRef]
  48. Ly, P.T.; Wu, Y.; Zou, H.; Wang, R.; Zhou, W.; Kinoshita, A.; Zhang, M.; Yang, Y.; Cai, F.; Woodgett, J.; et al. Inhibition of GSK3β-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J. Clin. Investig. 2013, 123, 224–235. [Google Scholar] [CrossRef]
  49. Latta, C.H.; Brothers, H.M.; Wilcock, D.M. Neuroinflammation in Alzheimer’s disease; A source of heterogeneity and target for personalized therapy. Neuroscience 2015, 302, 103–111. [Google Scholar] [CrossRef]
  50. Karuppagounder, S.S.; Madathil, S.K.; Pandey, M.; Haobam, R.; Rajamma, U.; Mohanakumar, K.P. Quercetin up-regulates mitochondrial complex-I activity to protect against programmed cell death in rotenone model of Parkinson’s disease in rats. Neuroscience 2013, 236, 136–148. [Google Scholar] [CrossRef]
  51. Zaplatic, E.; Bule, M.; Shah, S.Z.A.; Uddin, M.S.; Niaz, K. Molecular mechanisms underlying protective role of quercetin in attenuating Alzheimer’s disease. Life Sci. 2019, 224, 109–119. [Google Scholar] [CrossRef]
  52. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol. 2009, 7, 65–74. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.G.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta 2014, 1842, 1240–1247. [Google Scholar] [CrossRef] [PubMed]
  54. Beach, T.G.; Walker, R.; McGeer, E.G. Patterns of gliosis in Alzheimer’s disease and aging cerebrum. Glia 1989, 2, 420–436. [Google Scholar] [CrossRef]
  55. Delacourte, A. General and dramatic glial reaction in Alzheimer brains. Neurology 1990, 40, 33. [Google Scholar] [CrossRef] [PubMed]
  56. Arends, Y.M.; Duyckaerts, C.; Rozemuller, J.M.; Eikelenboom, P.; Hauw, J.J. Microglia, amyloid and dementia in alzheimer disease. A correlative study. Neurobiol. Aging 2000, 21, 39–47. [Google Scholar] [CrossRef]
  57. Kim, J.K.; Choi, S.J.; Cho, H.Y.; Hwang, H.J.; Kim, Y.J.; Lim, S.T.; Kim, C.J.; Kim, H.K.; Peterson, S.; Shin, D.H. Protective effects of kaempferol (3,4’,5,7-tetrahydroxyflavone) against amyloid beta peptide (Abeta)-induced neurotoxicity in ICR mice. Biosci. Biotechnol. Biochem. 2010, 74, 397–401. [Google Scholar] [CrossRef]
  58. Shen, X.Y.; Luo, T.; Li, S.; Ting, O.Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin inhibits okadaic acid-induced tau protein hyperphosphorylation through the Ca2+-calpain-p25-CDK5 pathway in HT22 cells. Int. J. Mol. Med. 2018, 41, 1138–1146. [Google Scholar] [CrossRef]
  59. Busche, M.A.; Hyman, B.T. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat. Neurosci. 2020, 23, 1183–1193. [Google Scholar] [CrossRef]
  60. Sperling, R.A.; Mormino, E.C.; Schultz, A.P.; Betensky, R.A.; Papp, K.V.; Amariglio, R.E.; Hanseeuw, B.J.; Buckley, R.; Chhatwal, J.; Hedden, T.; et al. The impact of amyloid-beta and tau on prospective cognitive decline in older individuals. Ann. Neurol. 2019, 85, 181–193. [Google Scholar] [CrossRef]
  61. Pievani, M.; de Haan, W.; Wu, T.; Seeley, W.W.; Frisoni, G.B. Functional network disruption in the degenerative dementias. Lancet Neurol. 2011, 10, 829–843. [Google Scholar] [CrossRef]
  62. Sakakibara, R.; Kawai, T. Cerebrospinal fluid oxidative stress markers in Alzheimer’s disease. Neurol. Clin. Neurosci. 2020, 8, 232–240. [Google Scholar] [CrossRef]
  63. Willette, A.A.; Li, T.; Willette, S.A.; Larsen, B.A.; Pollpeter, A.; Klinedinst, B.S.; Moody, S.; Barnett, N.; Parvin, M.; Pappas, C.; et al. Oxidative stress biomarkers and longitudinal changes in human brain imaging across the Alzheimer’s disease continuum. Alzheimer’s Dement. 2022, 18, e068364. [Google Scholar] [CrossRef]
  64. Kim, H.G.; Ju, M.S.; Shim, J.S.; Kim, M.C.; Lee, S.H.; Huh, Y.; Kim, S.Y.; Oh, M.S. Mulberry fruit protects dopaminergic neurons in toxin-induced Parkinson’s disease models. Br. J. Nutr. 2010, 104, 8–16. [Google Scholar] [CrossRef] [PubMed]
  65. Song, K.S.; Yang, E.J.; Kim, G.S.; Kim, J.A. Protective effects of onion-derived quercetin on glutamate-mediated hippocampal neuronal cell death. Pharmacogn. Mag. 2013, 9, 302–308. [Google Scholar] [CrossRef] [PubMed]
  66. Greenwood, S.M.; Connolly, C.N. Dendritic and mitochondrial changes during glutamate excitotoxicity. Neuropharmacology 2007, 53, 891–898. [Google Scholar] [CrossRef]
  67. Mattson, M.P. Apoptosis in neurodegenerative disorders. Nat. Rev. Mol. Cell Biol. 2000, 1, 120–130. [Google Scholar] [CrossRef]
  68. Simpson, D.S.; Oliver, P.L. ROS generation in microglia: Understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  69. Di Filippo, M.; Sarchielli, P.; Picconi, B.; Calabresi, P. Neuroinflammation and synaptic plasticity: Theoretical basis for a novel, immune-centred, therapeutic approach to neurological disorders. Trends Pharmacol. Sci. 2008, 29, 402–412. [Google Scholar] [CrossRef]
  70. Chen, W.W.; Zhang, X.; Huang, W.J. Role of neuroinflammation in neurodegenerative diseases. Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef]
  71. Kempuraj, D.; Thangavel, R.; Selvakumar, G.P.; Zaheer, S.; Ahmed, M.E.; Raikwar, S.P.; Zahoor, H.; Saeed, D.; Natteru, P.A.; Iyer, S.; et al. Brain and peripheral atypical inflammatory mediators potentiate neuroinflammation and neurodegeneration. Front. Cell. Neurosci. 2017, 11, 216. [Google Scholar] [CrossRef]
  72. Kempuraj, D.; Thangavel, R.; Natteru, P.A.; Selvakumar, G.P.; Saeed, D.; Zahoor, H.; Zaheer, S.; Iyer, S.S.; Zaheer, A. Neuroinflammation induces neurodegeneration. J. Neurol. Neurosurg. Spine 2016, 1, 1003. [Google Scholar] [PubMed]
  73. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef] [PubMed]
  74. Paris, D.; Mathura, V.; Ait-Ghezala, G.; Beaulieu-Abdelahad, D.; Patel, N.; Bachmeier, C.; Mullan, M. Flavonoids lower Alzheimer’s Aβ production via an NFκB dependent mechanism. Bioinformation 2011, 6, 229–236. [Google Scholar] [CrossRef] [PubMed]
  75. Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef]
  76. Huang, W.J.; Zhang, X.; Chen, W.W. Role of oxidative stress in Alzheimer’s disease. Biomed. Rep. 2016, 4, 519–522. [Google Scholar] [CrossRef]
  77. Cai, Z.; Zhao, B.; Ratka, A. Oxidative stress and β-amyloid protein in Alzheimer’s disease. NeuroMolecular Med. 2011, 13, 223–250. [Google Scholar] [CrossRef]
  78. Kim, H.R.; Lee, P.; Seo, S.W.; Roh, J.H.; Oh, M.; Oh, J.S.; Oh, S.J.; Kim, J.S.; Jeong, Y. Comparison of Amyloid β and Tau Spread Models in Alzheimer’s Disease. Cereb. Cortex 2019, 29, 4291–4302. [Google Scholar] [CrossRef]
  79. Ismail, R.; Parbo, P.; Madsen, L.S.; Hansen, A.K.; Hansen, K.V.; Schaldemose, J.L.; Kjeldsen, P.L.; Stokholm, M.G.; Gottrup, H.; Eskildsen, S.F.; et al. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: A longitudinal PET study. J. Neuroinflammation 2020, 17, 151. [Google Scholar] [CrossRef]
  80. Wang, D.M.; Li, S.Q.; Wu, W.L.; Zhu, X.Y.; Wang, Y.; Yuan, H.Y. Effects of long-term treatment with quercetin on cognition and mitochondrial function in a mouse model of Alzheimer’s disease. Neurochem. Res. 2014, 39, 1533–1543. [Google Scholar] [CrossRef]
  81. Cha, M.Y.; Han, S.H.; Son, S.M.; Hong, H.S.; Choi, Y.J.; Byun, J.; Mook Jung, I. Mitochondria-specific accumulation of amyloid beta induces mitochondrial dysfunction leading to apoptotic cell death. PLoS ONE 2012, 7, e34929. [Google Scholar] [CrossRef]
  82. Moreira, P.I.; Santos, M.S.; Moreno, A.; Rego, A.C.; Oliveira, C. Effect of amyloid beta-peptide on permeability transition pore: A comparative study. J. Neurosci. Res. 2002, 69, 257–267. [Google Scholar] [CrossRef] [PubMed]
  83. Beal, M.F. Mitochondria take centre stage in aging and neurodegeneration. Ann. Neurol. 2005, 58, 495–505. [Google Scholar] [CrossRef] [PubMed]
  84. Li, Y.; Rusinek, H.; Butler, T.; Glodzik, L.; Pirraglia, E.; Babich, J.; Mozley, P.D.; Nehmeh, S.; Pahlajani, S.; Wang, X.; et al. Decreased CSF clearance and increased brain amyloid in Alzheimer’s disease. Fluids Barriers CNS 2022, 19, 21. [Google Scholar] [CrossRef]
  85. Heneka, M.T.; Kummer, M.P.; Latz, E. Innate immune activation in neurodegenerative disease. Nat. Rev. Immunol. 2014, 14, 463–477. [Google Scholar] [CrossRef]
  86. Heneka, M.T.; Carson, M.J.; Khoury, J.E.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [PubMed]
  87. Arai, H.; Suzuki, H.; Yoshiyama, T.; Lobello, K.; Peng, Y.; Liu, E.; Ketter, N.; Margolin, R.; Jackson, N.; Fujimoto, Y. Safety, tolerability and immunogenicity of an immunotherapeutic vaccine (vanutide cridificar [ACC-001]) and the QS-21 adjuvant in Japanese individuals with mild-to-moderate Alzheimer’s disease: A phase IIa, multicenter, randomized, adjuvant and placebo clinical trial. Alzheimer’s Dement. 2013, 9, 282. [Google Scholar]
  88. Doody, R.S.; Raman, R.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; He, F.; Sun, X.; Thomas, R.G.; et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N. Engl. J. Med. 2013, 369, 341–350. [Google Scholar] [CrossRef]
  89. Doody, R.S.; Farlow, M.; Aisen, P.S. Alzheimer’s Disease Cooperative Study Data Analysis and Publication Committee. Phase 3 trials of solanezumab and bapineuzumab for Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 1460. [Google Scholar]
  90. Li, S.; Selkoe, D.J. A mechanistic hypothesis for the impairment of synaptic plasticity by soluble Aβ oligomers from Alzheimer’s brain. J. Neurochem. 2020, 154, 583–597. [Google Scholar] [CrossRef]
  91. Barbier, P.; Zejneli, O.; Martinho, M.; Lasorsa, A.; Belle, V.; Smet-Nocca, C.; Tsvetkov, P.O.; Devred, F.; Landrieu, I. Role of Tau as a Microtubule-Associated Protein: Structural and Functional Aspects. Front. Aging Neurosci. 2019, 11, 204. [Google Scholar] [CrossRef]
  92. Stancu, I.C.; Vasconcelos, B.; Terwel, D.; Dewachter, I. Models of β-amyloid induced Tau-pathology: The long and “folded” road to understand the mechanism. Mol. Neurodegener. 2014, 9, 51. [Google Scholar] [CrossRef] [PubMed]
  93. Takashima, A.; Honda, T.; Yasutake, K.; Michel, G.; Murayama, O.; Murayama, M.; Ishiguro, K.; Yamaguchi, H. Activation of tau protein kinase I/glycogen synthase kinase-3beta by amyloid beta peptide (25–35) enhances phosphorylation of tau in hippocampal neurons. Neurosci. Res. 1998, 31, 317–323. [Google Scholar] [CrossRef]
  94. Ferreira, A.; Lu, Q.; Orecchio, L.; Kosik, K.S. Selective phosphorylation of adult tau isoforms in mature hippocampal neurons exposed to fibrillar A beta. Mol. Cell. Neurosci. 1997, 9, 220–234. [Google Scholar] [CrossRef]
  95. Zheng, W.H.; Bastianetto, S.; Mennicken, F.; Ma, W.; Kar, S. Amyloid beta peptide induces tau phosphorylation and loss of cholinergic neurons in rat primary septal cultures. Neuroscience 2002, 115, 201–211. [Google Scholar] [CrossRef]
  96. Ma, Q.L.; Lim, G.P.; Harris-White, M.E.; Yang, F.; Ambegaokar, S.S.; Ubeda, O.J.; Glabe, C.G.; Teter, B.; Frautschy, S.A.; Cole, G.M. Antibodies against beta-amyloid reduce Abeta oligomers, glycogen synthase kinase-3beta activation and tau phosphorylation in vivo and in vitro. J. Neurosci. Res. 2006, 83, 374–384. [Google Scholar] [CrossRef]
  97. Tackenberg, C.; Grinschgl, S.; Trutzel, A.; Santuccione, A.C.; Frey, M.C.; Konietzko, U.; Grimm, J.; Brandt, R.; Nitsch, R.M. NMDA receptor subunit composition determines beta-amyloid-induced neurodegeneration and synaptic loss. Cell Death Dis. 2013, 4, e608. [Google Scholar] [CrossRef]
  98. Wang, W.Y.; Tan, M.S.; Yu, J.T.; Tan, L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015, 3, 136. [Google Scholar] [CrossRef] [PubMed]
  99. Vogel, J.W.; Iturria-Medina, Y.; Strandberg, O.T.; Smith, R.; Levitis, E.; Evans, A.C.; Hansson, O.; Alzheimer’s Disease Neuroimaging Initiative; Swedish BioFinder Study. Spread of pathological tau proteins through communicating neurons in human Alzheimer’s disease. Nat. Commun. 2020, 11, 2612, Erratum in Nat. Commun. 2021, 12, 4862. [Google Scholar] [CrossRef] [PubMed]
  100. Giacobini, E.; Gold, G. Alzheimer disease therapy—Moving from amyloid-β to tau. Nat. Rev. Neurol. 2013, 9, 677–686. [Google Scholar] [CrossRef]
  101. Amaral, A.C.; Perez-Nievas, B.G.; Chong, M.S.T.; Gonzalez-Martinez, A.; Argente-Escrig, H.; Rubio-Guerra, S.; Commins, C.; Muftu, S.; Eftekharzadeh, B.; Hudry, E.; et al. Isoform-selective decrease of glycogen synthase kinase-3-beta (GSK-3β) reduces synaptic tau phosphorylation, transcellular spreading, and aggregation. Iscience 2021, 24, 102058. [Google Scholar] [CrossRef]
  102. Mandelkow, E.-M.; Mandelkow, E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb. Perspect. Med. 2012, 2, a006247. [Google Scholar] [CrossRef] [PubMed]
  103. Roberson, E.D.; Scearce-Levie, K.; Palop, J.J.; Yan, F.; Cheng, I.H.; Wu, T.; Gerstein, H.; Yu, G.Q.; Mucke, L. Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer’s disease mouse model. Science 2007, 316, 750–754. [Google Scholar] [CrossRef] [PubMed]
  104. Duchen, M.R. Mitochondria and calcium: From cell signaling to cell death. J. Physiol. 2000, 529, 57–68. [Google Scholar] [CrossRef] [PubMed]
  105. Squìer, M.K.; Miller, A.C.; Malkinson, A.M.; Cohen, J.J. Calpain activation in apoptosis. J. Cell. Physiol. 1994, 159, 229–237. [Google Scholar] [CrossRef]
  106. Maher, P.; Schubert, D. Signaling by reactive oxygen species in the nervous system. Cell. Mol. Life Sci. 2000, 57, 1287–1305. [Google Scholar] [CrossRef] [PubMed]
  107. Darling, A.L.; Uversky, V.N. Intrinsic disorder and posttranslational modifications: The darker side of the biological dark matter. Front. Genet. 2018, 9, 158. [Google Scholar] [CrossRef]
  108. Barber, K.W.; Rinehart, J. The ABCs of PTMs. Nat. Chem. Biol. 2018, 14, 188–192. [Google Scholar] [CrossRef]
  109. Buee, L.; Bussiere, T.; Buee-Scherrer, V.; Delacourte, A.; Hof, P.R. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Rev. 2000, 33, 95–130. [Google Scholar] [CrossRef]
  110. Xia, C.; Makaretz, S.J.; Caso, C.; McGinnis, S.; Gomperts, S.N.; Sepulcre, J.; Gomez-Isla, T.; Hyman, B.T.; Schultz, A.; Vasdev, N.; et al. Association of in vivo [18F]AV-1451 tau PET imaging results with cortical atrophy and symptoms in typical and atypical Alzheimer disease. JAMA Neurol. 2017, 74, 427–436. [Google Scholar] [CrossRef]
  111. Bejanin, A.; Schonhaut, D.R.; La Joie, R.; Kramer, J.H.; Baker, S.L.; Sosa, N.; Ayakta, N.; Cantwell, A.; Janabi, M.; Lauriola, M.; et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimeras disease. Brain 2017, 140, 3286–3300. [Google Scholar] [CrossRef]
  112. Fleeman, R.M.; Proctor, E.A. Astrocytic Propagation of Tau in the Context of Alzheimer’s Disease. FFront. Cell. Neurosci. 2021, 15, 645233. [Google Scholar] [CrossRef] [PubMed]
  113. Wegmann, S.; Bennett, R.E.; Delorme, L.; Robbins, A.B.; Hu, M.; MacKenzie, D.; Kirk, M.J.; Schiantarelli, J.; Tunio, N.; Amaral, A.C.; et al. Experimental evidence for the age dependence of tau protein spread in the brain. Sci. Adv. 2019, 5, eaaw6404. [Google Scholar] [CrossRef] [PubMed]
  114. Wei, Y.; Liu, M.; Wang, D. The propagation mechanisms of extracellular tau in Alzheimer’s disease. J. Neurol. 2022, 269, 1164–1181. [Google Scholar] [CrossRef]
  115. Kazim, S.F.; Sharma, A.; Saroja, S.R.; Seo, J.H.; Larson, C.S.; Ramakrishnan, A.; Wang, M.; Blitzer, R.D.; Shen, L.; Peña, C.J.; et al. Chronic Intermittent Hypoxia Enhances Pathological Tau Seeding, Propagation, and Accumulation and Exacerbates Alzheimer-like Memory and Synaptic Plasticity Deficits and Molecular Signatures. Biol. Psychiatry 2022, 91, 346–358. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, R.H. Health-promoting components of fruits and vegetables in the diet. Adv. Nutr. Int. Rev. J. 2013, 4, 384S–392S. [Google Scholar] [CrossRef]
  117. Hertog, M.G.L.; Feskens, E.J.M.; Hollman, P.C.H.; Katan, M.B.; Kromhout, D. Dietary antioxidant flavonoids and risk of coronary heart disease: The Zutphen Elderly Study. Lancet 1993, 342, 1007–1011. [Google Scholar] [CrossRef]
  118. Anhê, G.F.; Okamoto, M.M.; Kinote, A.; Sollon, C.; Lellis-Santos, C.; Anhê, F.F.; Lima, G.A.; Hirabara, S.M.; Velloso, L.A.; Bordin, S.; et al. Quercetin decreases inflammatory response and increases insulin action in skeletal muscle of ob/ob mice and in L6 myotubes. Eur. J. Pharmacol. 2012, 689, 285–293. [Google Scholar] [CrossRef]
  119. Simunkova, M.; Alwasel, S.H.; Alhazza, I.M.; Jomova, K.; Kollar, V.; Rusko, M.; Valko, M. Management of oxidative stress and other pathologies in Alzheimer’s disease. Arch. Toxicol. 2019, 93, 2491–2513. [Google Scholar] [CrossRef]
  120. Hanaki, M.; Murakami, K.; Akagi, K.; Irie, K. Structural insights into mechanisms for inhibiting amyloid β42 aggregation by non-catechol-type flavonoids. Bioorganic Med. Chem. 2016, 24, 304–313. [Google Scholar] [CrossRef]
  121. Porat, Y.; Abramowitz, A.; Gazit, E. Inhibition of amyloid fibril formation by polyphenols: Structural similarity and aromatic interactions as a common inhibition mechanism. Chem. Biol. Drug Des. 2006, 67, 27–37. [Google Scholar] [CrossRef]
  122. Ono, K.; Yoshiike, Y.; Takashima, A.; Hasegawa, K.; Naiki, H.; Yamada, M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: Implications for the prevention and therapeutics of Alzheimer’s disease. J. Neurochem. 2003, 87, 172–181. [Google Scholar] [CrossRef]
  123. Jiménez-Aliaga, K.; Bermejo-Bescós, P.; Benedí, J.; Martín-Aragón, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
  124. Sato, M.; Murakami, K.; Uno, M.; Nakagawa, Y.; Katayama, S.; Akagi, K.; Masuda, Y.; Takegoshi, K.; Irie, K. Site-specific inhibitory mechanism for amyloid β42 aggregation by catechol-type flavonoids targeting the Lys residues. J. Biol. Chem. 2013, 288, 23212–23224. [Google Scholar] [CrossRef]
  125. Yu, X.; Li, Y.; Mu, X. Effect of Quercetin on PC12 Alzheimer’s Disease Cell Model Induced by Aβ25-35 and Its Mechanism Based on Sirtuin1/Nrf2/HO-1 Pathway. BioMed Res. Int. 2020, 2020, 8210578. [Google Scholar] [CrossRef] [PubMed]
  126. Kumar, S.; Krishnakumar, V.G.; Morya, V.; Gupta, S.; Datta, B. Nanobiocatalyst facilitated aglycosidic quercetin as a potent inhibitor of tau protein aggregation. Int. J. Biol. Macromol. 2019, 138, 168–180. [Google Scholar] [CrossRef] [PubMed]
  127. Luo, C.; Yang, H.; Tang, C.; Yao, G.; Kong, L.; He, H.; Zhou, Y. Kaempferol alleviates insulin resistance via hepatic IKK/NF-κB signal in type 2 diabetic rats. Int. Immunopharmacol. 2015, 28, 744–750. [Google Scholar] [CrossRef]
  128. Peng, J.; Li, Q.; Li, K.; Zhu, L.; Lin, X.; Lin, X.; Shen, Q.; Li, G.; Xie, X. Quercetin Improves Glucose and Lipid Metabolism of Diabetic Rats: Involvement of Akt Signaling and SIRT1. J. Diabetes Res. 2017, 2017, 3417306. [Google Scholar] [CrossRef]
  129. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [PubMed]
  130. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
  131. Crozier, A.; Lean, M.E.J.; McDonald, M.S.; Black, C. Quantitative analysis of the flavonoid content of commercial tomatoes, onions, lettuce, and celery. J. Agric. Food Chem. 1997, 45, 590–595. [Google Scholar] [CrossRef]
  132. Xu, M.; Huang, H.; Mo, X.; Zhu, Y.; Chen, X.; Li, X.; Peng, X.; Xu, Z.; Chen, L.; Rong, S.; et al. Quercetin-3-O-Glucuronide Alleviates Cognitive Deficit and Toxicity in Aβ1-42 -Induced AD-Like Mice and SH-SY5Y Cells. Mol. Nutr. Food Res. 2021, 65, e2000660. [Google Scholar] [CrossRef]
  133. Shen, P.; Lin, W.; Deng, X.; Ba, X.; Han, L.; Chen, Z.; Qin, K.; Huang, Y.; Tu, S. Potential Implications of Quercetin in Autoimmune Diseases. Front. Immunol. 2021, 12, 689044. [Google Scholar] [CrossRef]
  134. Babaei, F.; Mirzababaei, M.; Nassiri-Asl, M. Quercetin in Food: Possible Mechanisms of Its Effect on Memory. J. Food Sci. 2018, 83, 2280–2287. [Google Scholar] [CrossRef] [PubMed]
  135. Yang, D.; Wang, T.; Long, M.; Li, P. Quercetin: Its Main Pharmacological Activity and Potential Application in Clinical Medicine. Oxidative Med. Cell. Longev. 2020, 2020, 8825387. [Google Scholar] [CrossRef] [PubMed]
  136. Nishihira, J.; Nishimura, M.; Kurimoto, M.; Kagami-Katsuyama, H.; Hattori, H.; Nakagawa, T.; Muro, T.; Kobori, M. The effect of 24-week continuous intake of quercetin-rich onion on age-related cognitive decline in healthy elderly people: A randomized, double-blind, placebo-controlled, parallel-group comparative clinical trial. J. Clin. Biochem. Nutr. 2021, 69, 203–215. [Google Scholar] [CrossRef]
  137. Bayazid, A.B.; Lim, B.O. Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1. Nutrients 2022, 14, 5132. [Google Scholar] [CrossRef]
  138. Islam, M.S.; Quispe, C.; Hossain, R.; Islam, M.T.; Al-Harrasi, A.; Al-Rawahi, A.; Martorell, M.; Mamurova, A.; Seilkhan, A.; Altybaeva, N.; et al. Neuropharmacological Effects of Quercetin: A Literature-Based Review. Neuropharmacology 2022, 12, 665031. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, Q.; Naeem, A.; Zou, J.; Yu, C.; Wang, Y.; Chen, J.; Ping, Y. Isolation of Phenolic Compounds from Raspberry Based on Molecular Imprinting Techniques and Investigation of Their Anti-Alzheimer’s Disease Properties. Molecules 2022, 27, 6893. [Google Scholar] [CrossRef]
  140. Ulusoy, H.G.; Sanlier, N. A minireview of quercetin: From its metabolism to possible mechanisms of its biological activities. Crit. Rev. Food Sci. Nutr. 2020, 60, 3290–3303. [Google Scholar] [CrossRef]
  141. Xiao, L.; Luo, G.; Tang, Y.; Yao, P. Quercetin and iron metabolism: What we know and what we need to know. Food Chem. Toxicol. 2018, 114, 190–203. [Google Scholar] [CrossRef]
  142. Lesjak, M.; Beara, I.; Simin, N.; Pintać, D.; Majkić, T.; Bekvalac, K.; Orčić, D.; Mimica-Dukić, N. Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J. Funct. Foods 2018, 40, 68–75. [Google Scholar] [CrossRef]
  143. Chahar, M.K.; Sharma, N.; Dobhal, M.P.; Joshi, Y.C. Flavonoids: A versatile source of anticancer drugs. Pharmacogn. Rev. 2011, 5, 1. [Google Scholar]
  144. Nakagawa, T.; Ohta, K. Quercetin Regulates the Integrated Stress Response to Improve Memory. Int. J. Mol. Sci. 2019, 20, 2761. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, Y.W.; Liu, X.L.; Kong, L.; Zhang, M.Y.; Chen, Y.J.; Zhu, X.; Hao, Y.C. Neuroprotection of quercetin on central neurons against chronic high glucose through enhancement of Nrf2/ARE/glyoxalase-1 pathway mediated by phosphorylation regulation. Biomed. Pharmacother. 2019, 109, 2145–2154. [Google Scholar] [CrossRef]
  146. Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxidative Med. Cell. Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef]
  147. Wei, C.; Li, S.; Zhu, Y.; Chen, W.; Li, C.; Xu, R. Network pharmacology identify intersection genes of quercetin and Alzheimer’s disease as potential therapeutic targets. Front. Aging Neurosci. 2022, 14, 902092. [Google Scholar] [CrossRef] [PubMed]
  148. García-Mediavilla, M.V.; Crespo, I.; Collado, P.S.; Esteller, A.; Sánchez-Campos, S.; Tuñón, M.J.; González-Gallego, J. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway in Chang Liver cells. Eur. J. Pharmacol. 2007, 557, 221–229. [Google Scholar] [CrossRef] [PubMed]
  149. Song, K.S.; Jeong, W.S.; Jun, M. Inhibition of β-amyloid peptide-induced neurotoxicity by kaempferol 3-O-(6″-acetyl)-β-glucopyranoside from butterbur (Petasites japonicus) leaves in B103 cells. Food Sci. Biotechnol. 2012, 21, 845–851. [Google Scholar] [CrossRef]
  150. Kaypee, S.; Singh, S.; Swarnkar, S.; Kundu, T.K. Emerging epigenetic therapies—Lysine acetyltransferase inhibitors. In Epigenetic Cancer Therapy; Academic Press: Cambridge, MA, USA, 2023; pp. 459–505. [Google Scholar]
  151. Xiao, X.; Shi, D.; Liu, L.; Wang, J.; Xie, X.; Kang, T.; Deng, W. Quercetin suppresses cyclooxygenase-2 expression and angiogenesis through inactivation of P300 signaling. PLoS ONE 2011, 6, e22934. [Google Scholar] [CrossRef]
  152. Pei, Y.; Parks, J.S.; Kang, H.W. Quercetin alleviates high-fat diet-induced inflammation in brown adipose tissue. J. Funct. Foods 2021, 85, 104614. [Google Scholar] [CrossRef]
  153. Son, S.M.; Park, S.J.; Fernandez-Estevez, M.; Rubinsztein, D.C. Autophagy regulation by acetylation-implications for neurodegenerative diseases. Exp. Mol. Med. 2021, 53, 30–41. [Google Scholar] [CrossRef] [PubMed]
  154. Fiorentino, F.; Mai, A.; Rotili, D. Lysine Acetyltransferase Inhibitors From Natural Sources. Front. Pharmacol. 2020, 11, 1243. [Google Scholar] [CrossRef] [PubMed]
  155. Liao, Y.; Mai, X.; Wu, X.; Hu, X.; Luo, X.; Zhang, G. Exploring the Inhibition of Quercetin on Acetylcholinesterase by Multispectroscopic and In Silico Approaches and Evaluation of Its Neuroprotective Effects on PC12 Cells. Molecules 2022, 27, 7971. [Google Scholar] [CrossRef] [PubMed]
  156. Alghamdi, A.; Birch, D.J.; Vyshemirsky, V.; Rolinski, O.J. Impact of the Flavonoid Quercetin on β-Amyloid Aggregation Revealed by Intrinsic Fluorescence. J. Phys. Chem. B 2022, 126, 7229–7237. [Google Scholar] [CrossRef]
  157. Ho, C.L.; Kao, N.J.; Lin, C.I.; Cross, T.L.; Lin, S.H. Quercetin Increases Mitochondrial Biogenesis and Reduces Free Radicals in Neuronal SH-SY5Y Cells. Nutrients 2022, 14, 3310. [Google Scholar] [CrossRef] [PubMed]
  158. Bao, D.; Wang, J.; Pang, X.; Liu, H. Protective Effect of Quercetin against Oxidative Stress-Induced Cytotoxicity in Rat Pheochromocytoma (PC-12) Cells. Molecules 2017, 22, 1122. [Google Scholar] [CrossRef]
  159. Jiang, W.; Luo, T.; Li, S.; Zhou, Y.; Shen, X.-Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin Protects against Okadaic Acid-Induced Injury via MAPK and PI3K/Akt/GSK3β Signaling Pathways in HT22 Hippocampal Neurons. PLoS ONE 2016, 11, e0152371. [Google Scholar] [CrossRef]
  160. Paula, P.C.; Maria, S.G.; Luis, C.H.; Patricia, C.G. Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 2019, 24, 2287. [Google Scholar] [CrossRef]
  161. Molaei, A.; Hatami, H.; Dehghan, G.; Sadeghian, R.; Khajehnasiri, N. Synergistic effects of quercetin and regular exercise on the recovery of spatial memory and reduction of parameters of oxidative stress in animal model of Alzheimer’s disease. EXCLI J. 2020, 19, 596–612. [Google Scholar] [CrossRef]
  162. Dhawan, S.; Kapil, R.; Singh, B. Formulation development and systematic optimization of solid lipid nanoparticles of quercetin for improved brain delivery. J. Pharm. Pharmacol. 2011, 63, 342–351. [Google Scholar] [CrossRef]
  163. Chen, J.; Deng, X.; Liu, N.; Li, M.; Liu, B.; Fu, Q.; Qu, R.; Ma, S. Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of ER stress in a manner dependent on AMPK pathway. J. Funct. Foods 2016, 22, 463–476. [Google Scholar] [CrossRef]
  164. National Center for Biotechnology Information. PubChem Compound Summary for CID 5280343, Quercetin. Retrieved 9 November 2023. 2023. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Quercetin (accessed on 6 October 2023).
  165. Dimitrić Marković, J.M.; Milenković, D.; Amić, D.; Popović-Bijelić, A.; Mojović, M.; Pašti, I.A.; Marković, Z.S. Energy requirements of the reactions of kaempferol and selected radical species in different media: Towards the prediction of the possible radical scavenging mechanisms. Struct. Chem. 2014, 25, 1795–1804. [Google Scholar] [CrossRef]
  166. Wang, L.; Tu, Y.C.; Lian, T.W.; Hung, J.T.; Yen, J.H.; Wu, M.J. Distinctive antioxidant and antiinflammatory effects of flavonols. J. Agric. Food Chem. 2006, 54, 9798–9804. [Google Scholar] [CrossRef] [PubMed]
  167. Park, S.E.; Sapkota, K.; Kim, H.; Kim, S.J. Kaempferol acts through mitogen-activated protein kinases and protein kinase B/AKT to elicit protection in a model of neuroinflammation in BV2 microglial cells. Br. J. Pharmacol. 2011, 164, 1008–1025. [Google Scholar] [CrossRef]
  168. Olszewska, M. Separation of quercetin, sexangularetin, kaempferol and isorhamnetin for simultaneous HPLC determination of flavonoid aglycones in inflorescences, leaves and fruits of three Sorbus species. J. Pharm. Biomed. Anal. 2008, 48, 629–635. [Google Scholar] [CrossRef]
  169. Kiziltaş, H. Comprehensive evaluation of Reseda lutea L. (Wild Mignonette) and 7 isolated flavonol glycosides: Determination of antioxidant activity, anti-Alzheimer, antidiabetic and cytotoxic effects with in vitro and in silico methods. Turk. J. Chem. 2022, 46, 1185–1198. [Google Scholar] [CrossRef]
  170. Sulfahri; Wardhani, R.; Makatita, F.A.; Iskandar, I.W. Utilization of Nypa fruit in Alzheimer’s Disease: An In Silico Approach. J. Phys. Conf. Ser. 2019, 1341, 022003. [Google Scholar] [CrossRef]
  171. Yuan, Y.; Zhai, Y.; Chen, J.; Xu, X.; Wang, H. Kaempferol Ameliorates Oxygen-Glucose Deprivation/Reoxygenation-Induced Neuronal Ferroptosis by Activating Nrf2/SLC7A11/GPX4 Axis. Biomolecules 2021, 11, 923. [Google Scholar] [CrossRef]
  172. Uysal, M.; Celikten, M.; Beker, M.; Polat, N.; Huseyinbas, O.; Terzioglu-Usak, S.; Elibol, B. Kaempferol treatment ameliorates memory impairments in STZ-induced neurodegeneration by acting on reelin signaling. Acta Neurobiol. Exp. (Wars) 2023, 83, 236–245. [Google Scholar] [CrossRef]
  173. Beg, T.; Jyoti, S.; Naz, F.; Rahul, X.; Ali, F.; Ali, S.K.; Reyad, A.M.; Siddique, Y.H. Protective Effect of Kaempferol on the Transgenic Drosophila Model of Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2018, 17, 421–429. [Google Scholar] [CrossRef]
  174. Zhang, N.; Xu, H.; Wang, Y.; Yao, Y.; Liu, G.; Lei, X.; Sun, H.; Wu, X.; Li, J. Protective mechanism of kaempferol against Aβ25-35-mediated apoptosis of pheochromocytoma (PC-12) cells through the ER/ERK/MAPK signalling pathway. Arch. Med Sci. 2020, 17, 406–416. [Google Scholar] [CrossRef]
  175. Sun, J.; Wang, J.; Hu, L.; Yan, J. K-3-Rh Protects Against Cerebral Ischemia/Reperfusion Injury by Anti-Apoptotic Effect Through PI3K-Akt Signaling Pathway in Rat. Neuropsychiatr. Dis. Treat. 2020, 16, 1217–1227. [Google Scholar] [CrossRef]
  176. Al-Brakati, A.; Albarakati, A.J.A.; Lokman, M.S.; Theyab, A.; Algahtani, M.; Menshawi, S.; AlAmri, O.D.; Al Omairi, N.E.; Essawy, E.A.; Kassab, R.B.; et al. Possible Role of Kaempferol in Reversing Oxidative Damage, Inflammation, and Apoptosis-Mediated Cortical Injury Following Cadmium Exposure. Neurotox. Res. 2021, 39, 198–209. [Google Scholar] [CrossRef]
  177. Ai, R.; Zhuang, X.X.; Anisimov, A.; Lu, J.H.; Fang, E.F. A synergized machine learning plus cross-species wet-lab validation approach identifies neuronal mitophagy inducers inhibiting Alzheimer disease. Autophagy 2022, 18, 939–941. [Google Scholar] [CrossRef]
  178. Zarei, M.; Mohammadi, S.; Komaki, A.; Golipour Choshali, Z. Antidepressant-like Effects of Intra-cerebroventricular Microinjection of Kaempferol in Male Rats: Involvement of 5-HT2 Receptors. Avicenna J. Neuro Psycho Physiol. 2022, 9, 23–30. [Google Scholar]
  179. Rita, L.; Neumann, N.R.; Laponogov, I.; Gonzalez, G.; Veselkov, D.; Pratico, D.; Aalizadeh, R.; Thomaidis, N.S.; Thompson, D.C.; Vasiliou, V.; et al. Alzheimer’s disease: Using gene/protein network machine learning for molecule discovery in olive oil. Hum. Genom. 2023, 17, 57. [Google Scholar] [CrossRef]
  180. Karunakaran, K.B.; Thiyagaraj, A.; Santhakumar, K. Novel insights on acetylcholinesterase inhibition by Convolvulus pluricaulis, scopolamine and their combination in zebrafish. Nat. Prod. Bioprospecting 2022, 12, 6. [Google Scholar] [CrossRef] [PubMed]
  181. Simunkova, M.; Barbierikova, Z.; Jomova, K.; Hudecova, L.; Lauro, P.; Alwasel, S.H.; Alhazza, I.; Rhodes, C.J.; Valko, M. Antioxidant vs. Prooxidant Properties of the Flavonoid, Kaempferol, in the Presence of Cu(II) Ions: A ROS-Scavenging Activity, Fenton Reaction and DNA Damage Study. Int. J. Mol. Sci. 2021, 22, 1619. [Google Scholar] [CrossRef] [PubMed]
  182. Ajiboye, B.O.; Ojo, O.A.; Okesola, M.A.; Akinyemi, A.J.; Talabi, J.Y.; Idowu, O.T.; Fadaka, A.O.; Boligon, A.A.; de Campos, M.M.A. In vitro antioxidant activities and inhibitory effects of phenolic extract of Senecio biafrae (Oliv and Hiern) against key enzymes linked with type II diabetes mellitus and Alzheimer’s disease. Food Sci. Nutr. 2018, 6, 1803–1810. [Google Scholar] [CrossRef] [PubMed]
  183. Shabir, I.; Pandey, V.K.; Shams, R.; Dar, A.H.; Dash, K.K.; Khan, S.A.; Bashir, I.; Jeevarathinam, G.; Rusu, A.V.; Esatbeyoglu, T.; et al. Promising bioactive properties of quercetin for potential food applications and health benefits: A review. Front. Nutr. 2022, 9, 999752. [Google Scholar] [CrossRef]
  184. Álvarez-Berbel, I.; Espargaró, A.; Viayna, A.; Caballero, A.B.; Busquets, M.A.; Gámez, P.; Luque, F.J.; Sabaté, R. Three to Tango: Inhibitory Effect of Quercetin and Apigenin on Acetylcholinesterase, Amyloid-β Aggregation and Acetylcholinesterase-Amyloid Interaction. Pharmaceutics 2022, 14, 2342. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, J.; Mao, J.; Wang, R.; Li, S.; Wu, B.; Yuan, Y. Kaempferol Protects Against Cerebral Ischemia Reperfusion Injury Through Intervening Oxidative and Inflammatory Stress Induced Apoptosis. Front. Pharmacol. 2020, 11, 424. [Google Scholar] [CrossRef] [PubMed]
  186. Dong, X.; Zhou, S.; Nao, J. Kaempferol as a therapeutic agent in Alzheimer’s disease: Evidence from preclinical studies. Ageing Res. Rev. 2023, 87, 101910. [Google Scholar] [CrossRef] [PubMed]
  187. Li, W.H.; Cheng, X.; Yang, Y.L.; Liu, M.; Zhang, S.S.; Wang, Y.H.; Du, G.H. Kaempferol attenuates neuroinflammation and blood brain barrier dysfunction to improve neurological deficits in cerebral ischemia/reperfusion rats. Brain Res. 2019, 1722, 146361. [Google Scholar] [CrossRef]
  188. El-Kott, A.F.; Abd-Lateif, A.-E.M.; Khalifa, H.S.; Morsy, K.; Ibrahim, E.H.; Bin-Jumah, M.; Abdel-Daim, M.M.; Aleya, L. Kaempferol protects against cadmium chloride-induced hippocampal damage and memory deficits by activation of silent information regulator 1 and inhibition of poly (ADP-Ribose) polymerase-1. Sci. Total. Environ. 2020, 728, 138832. [Google Scholar] [CrossRef]
  189. Lin, H.; Wang, X.; Zhao, J.; Lin, Z. Protective effect of kaempferol against cognitive and neurological disturbances induced by d-galactose and aluminum chloride in mice. J. Funct. Foods 2023, 100, 105385. [Google Scholar] [CrossRef]
  190. Selvi, R.B.; Swaminathan, A.; Chatterjee, S.; Shanmugam, M.K.; Li, F.; Ramakrishnan, G.B.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Inhibition of p300 lysine acetyltransferase activity by luteolin reduces tumor growth in head and neck squamous cell carcinoma (HNSCC) xenograft mouse model. Oncotarget 2015, 6, 43806–43818. [Google Scholar] [CrossRef]
  191. Zhou, Y.P.; Li, G.C. Kaempferol protects cell damage in in vitro ischemia reperfusion model in rat neuronal PC12 cells. BioMed Res. Int. 2020, 2020, 2461079. [Google Scholar] [CrossRef]
  192. Kadioglu, O.; Nass, J.; Saeed, M.E.; Schuler, B.; Efferth, T. Kaempferol Is an Anti-Inflammatory Compound with Activity towards NF-κB Pathway Proteins. Anticancer Res. 2015, 35, 2645–2650. [Google Scholar]
  193. Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef]
  194. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef] [PubMed]
  195. Sharoar, G.; Thapa, A.; Shahnawaz, M.; Ramasamy, V.S.; Woo, E.-R.; Shin, S.Y.; Park, I.-S. Keampferol-3-O-rhamnoside abrogates amyloid beta toxicity by modulating monomers and remodeling oligomers and fibrils to non-toxic aggregates. J. Biomed. Sci. 2012, 19, 104. [Google Scholar] [CrossRef] [PubMed]
  196. Chowdhury, M.A.; Ko, H.J.; Lee, H.; Aminul Haque, M.; Park, I.S.; Lee, D.S.; Woo, E.R. Oleanane triterpenoids from Akebiae Caulis exhibit inhibitory effects on Aβ42 induced fibrillogenesis. Arch. Pharm. Res. 2017, 40, 318–327. [Google Scholar] [CrossRef] [PubMed]
  197. Guo, Q.; Sebastian, L.; Sopher, B.L. Increased vulnerability of hippocampal neurons from presenilin-1 mutant knock-in mice to amyloid-β peptide toxicity. J. Neurochem. 1999, 72, 1019–1029. [Google Scholar] [CrossRef] [PubMed]
  198. Ishige, K.; Schubert, D.; Sagara, Y. Flavonoids protect neuronal cells from oxidative stress by three distinct mechanisms. Free. Radic. Biol. Med. 2001, 30, 433–446. [Google Scholar] [CrossRef]
  199. Miranda, S.; Opazo, C.; Larrondo, L.F.; Munoz, F.J. The role of oxidative stress in the toxicity induced by amyloid β-peptide in Alzheimer’s disease. Prog. Neurobiol. 2000, 62, 633–648. [Google Scholar] [CrossRef]
  200. Jafari, A.; Babaei, P.; Rohampour, K.; Rashtiani, S. The Effect of Kaempferol on Autophagy and Nrf-2 Signaling in a Rat Model of Aβ1-42-induced Alzheimer’s Disease. Casp. J. Neurol. Sci. 2022, 8, 7–16. [Google Scholar] [CrossRef]
  201. Xie, C.; Zhuang, X.X.; Niu, Z.; Ai, R.; Lautrup, S.; Zheng, S.; Jiang, Y.; Han, R.; Gupta, T.S.; Cao, S.; et al. Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow. Nat. Biomed. Eng. 2022, 6, 76–93. [Google Scholar] [CrossRef]
  202. Kaempferol: National Center for Biotechnology Information. PubChem Compound Summary for CID 5280863, Kaempferol. Retrieved 9 November 2023. 2023. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Kaempferol (accessed on 6 October 2023).
  203. Kouhestani, S.; Zare, S.; Babaei, P. Effects of pure flavonoid of medlar leaves on passive avoidance learning and memory in Alzheimer model of ovariectomized rats. J. Guilan Univ. Med. Sci. 2017, 26, 62–71. [Google Scholar]
  204. Krishnaveni, M. Flavonoid in enhancing memory function. J. Pharm. Res. 2012, 5, 3870–3874. [Google Scholar]
  205. Spencer, J.P. The impact of fruit flavonoids on memory and cognition. Br. J. Nutr. 2010, 104, 40–47. [Google Scholar] [CrossRef] [PubMed]
  206. Liu, L.; Liu, Y.; Zhen, Y.; Guo, T.; Wang, C.; Shen, L.; Li, W. Quercetin inhibits cytotoxicity of PC12 cells induced by amyloid-beta 25–35 via stimulating estrogen receptor α, activating ERK1/2, and inhibiting apoptosis. Open Life Sci. 2022, 17, 230–242. [Google Scholar] [CrossRef]
  207. Jin, S.; Zhang, L.; Wang, L. Kaempferol, a potential neuroprotective agent in neurodegenerative diseases: From chemistry to medicine. Biomed. Pharmacother. 2023, 165, 115215. [Google Scholar] [CrossRef]
  208. Damirchi, A.; Hosseini, F.; Babaei, P. Mental Training Enhances Cognitive Function and BDNF More Than Either Physical or Combined Training in Elderly Women With MCI: A Small-Scale Study. Am. J. Alzheimers Dis. Other Demen. 2018, 33, 20–29. [Google Scholar] [CrossRef] [PubMed]
  209. Lee, J.; Fukumoto, H.; Orne, J.; Klucken, J.; Raju, S.; Vanderburg, C.R.; Irizarry, M.C.; Hyman, B.T.; Ingelsson, M. Decreased levels of BDNF protein in Alzheimer temporal cortex are independent of BDNF polymorphisms. Exp. Neurol. 2005, 194, 91–96. [Google Scholar] [CrossRef] [PubMed]
  210. Yan, T.; He, B.; Xu, M.; Wu, B.; Xiao, F.; Bi, K.; Jia, Y. Kaempferide prevents cognitive decline via attenuation of oxidative stress and enhancement of brain-derived neurotrophic factor/tropomyosin receptor kinase B/cAMP response element-binding signaling pathway. Phytotherapy Res. 2019, 33, 1065–1073. [Google Scholar] [CrossRef]
  211. Amidfar, M.; de Oliveira, J.; Kucharska, E.; Budni, J.; Kim, Y.K. The role of CREB and BDNF in neurobiology and treatment of Alzheimer’s disease. Life Sci. 2020, 257, 118020. [Google Scholar] [CrossRef]
  212. Walton, M.R.; Dragunow, M. Is CREB a key to neuronal survival? Trends Neurosci. 2000, 23, 48–53. [Google Scholar] [CrossRef]
  213. Gao, Q.; Tian, D.; Han, Z.; Lin, J.; Chang, Z.; Zhang, D.; Ma, D. Network pharmacology and molecular docking analysis on molecular targets and mechanisms of buyang huanwu decoction in the treatment of ischemic stroke. Evid. -Based Complement. Altern. Med. 2021, 2021, 1–15. [Google Scholar] [CrossRef]
  214. Wang, Z.-H.; Xiang, J.; Liu, X.; Yu, S.P.; Manfredsson, F.P.; Sandoval, I.M.; Wu, S.; Wang, J.Z.; Ye, K. Deficiency in BDNF/TrkB neurotrophic activity stimulates δ-secretase by upregulating C/EBPβ in Alzheimer’s disease. Cell Rep. 2019, 28, 655–669. [Google Scholar] [CrossRef]
  215. Connor, B.; Young, D.; Yan, Q.; Faull, R.L.M.; Synek, B.; Dragunow, M. Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Mol. Brain Res. 1997, 49, 71–81. [Google Scholar] [CrossRef] [PubMed]
  216. Levenga, J.; Wong, H.; Milstead, R.; LaPlante, L.; Hoeffer, C.A. Immunohistological Examination of AKT Isoforms in the Brain: Cell-Type Specificity That May Underlie AKT’s Role in Complex Brain Disorders and Neurological Disease. Cereb. Cortex Commun. 2021, 2, tgab036. [Google Scholar] [CrossRef] [PubMed]
  217. Zarneshan, S.N.; Fakhri, S.; Khan, H. Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: A mechanistic approach. Pharmacol. Res. 2022, 177, 106099. [Google Scholar] [CrossRef] [PubMed]
  218. Pak, M.E.; Yang, H.J.; Li, W.; Kim, J.K.; Go, Y. Yuk-Gunja-Tang attenuates neuronal death and memory impairment via ERK/CREB/BDNF signaling in the hippocampi of experimental Alzheimer’s disease model. Front. Pharmacol. 2022, 13, 1014840. [Google Scholar] [CrossRef] [PubMed]
  219. Jain, V.; Baitharu, I.; Prasad, D.; Ilavazhagan, G. Enriched environment prevents hypobaric hypoxia induced memory impairment and neurodegeneration: Role of BDNF/PI3K/GSK3β pathway coupled with CREB activation. PLoS ONE 2013, 8, e62235. [Google Scholar] [CrossRef]
  220. Gao, L.; Zhang, Y.; Sterling, K.; Song, W. Brain-derived neurotrophic factor in Alzheimer’s disease and its pharmaceutical potential. Transl. Neurodegener. 2022, 11, 4. [Google Scholar] [CrossRef]
  221. Rada, P.; Rojo, A.I.; Chowdhry, S.; McMahon, M.; Hayes, J.D.; Cuadrado, A. SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol. Cell. Biol. 2011, 31, 1121–1133. [Google Scholar] [CrossRef]
  222. Kume, T.; Kouchiyama, H.; Kaneko, S.; Maeda, T.; Kaneko, S.; Akaike, A.; Shimohama, S.; Kihara, T.; Kimura, J.; Wada, K.; et al. BDNF prevents NO mediated glutamate cytotoxicity in cultured cortical neurons. Brain Res. 1997, 756, 200–204. [Google Scholar] [CrossRef]
  223. Mercado-Gómez, O.; Hernández-Fonseca, K.; Villavicencio-Queijeiro, A.; Massieu, L.; Chimal-Monroy, J.; Arias, C. Inhibition of Wnt and PI3K signaling modulates GSK-3beta activity and induces morphological changes in cortical neurons: Role of tau phosphorylation. Neurochem. Res. 2008, 33, 1599–1609. [Google Scholar] [CrossRef]
  224. Long, H.Z.; Cheng, Y.; Zhou, Z.W.; Luo, H.Y.; Wen, D.D.; Gao, L.C. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer’s Disease and Parkinson’s Disease. Front. Pharmacol. 2021, 12, 648636. [Google Scholar] [CrossRef]
  225. Jantas, D.; Malarz, J.; Le, T.N.; Stojakowska, A. Neuroprotective Properties of Kempferol Derivatives from Maesa membranacea against Oxidative Stress-Induced Cell Damage: An Association with Cathepsin D Inhibition and PI3K/Akt Activation. Int. J. Mol. Sci. 2021, 22, 10363. [Google Scholar] [CrossRef] [PubMed]
  226. Wu, J.; Liu, H.; Chu, T.; Jiang, P.; Li, S.T. Neuregulin-1β attenuates sepsis-induced diaphragm atrophy by activating the PI3K/Akt signaling pathway. J. Muscle Res. Cell Motil. 2019, 40, 43–51. [Google Scholar] [CrossRef] [PubMed]
  227. Kandezi, N.; Mohammadi, M.; Ghaffari, M.; Gholami, M.; Motaghinejad, M.; Safari, S. Novel Insight to Neuroprotective Potential of Curcumin: A Mechanistic Review of Possible Involvement of Mitochondrial Biogenesis and PI3/Akt/GSK3 or PI3/Akt/CREB/BDNF Signaling Pathways. Int. J. Mol. Cell. Med. 2020, 9, 1–32. [Google Scholar] [CrossRef] [PubMed]
  228. Tanqueiro, S.R.; Ramalho, R.M.; Rodrigues, T.M.; Lopes, L.V.; Sebastião, A.M.; Diógenes, M.J. Inhibition of NMDA Receptors Prevents the Loss of BDNF Function Induced by Amyloid β. Front. Pharmacol. 2018, 9, 237. [Google Scholar] [CrossRef]
  229. Garzon, D.J.; Fahnestock, M. Oligomeric amyloid decreases basal levels of brain-derived neurotrophic factor (BDNF) mRNA via specific downregulation of BDNF transcripts IV and V in differentiated human neuroblastoma cells. J. Neurosci. 2007, 27, 2628–2635. [Google Scholar] [CrossRef]
  230. Tong, L.; Thornton, P.L.; Balazs, R.; Cotman, C.W. β-amyloid-(1–42) impairs activity-dependent cAMP-response element-binding protein signaling in neurons at concentrations in which cell survival is not compromised. J. Biol. Chem. 2001, 276, 17301–17306. [Google Scholar] [CrossRef]
  231. Cowansage, K.K.; LeDoux, J.E.; Monfils, M.H. Brain-derived neurotrophic factor: A dynamic gatekeeper of neural plasticity. Curr. Mol. Pharmacol. 2010, 3, 12–29. [Google Scholar] [CrossRef]
  232. Rosa, E.; Fahnestock, M. CREB expression mediates amyloid β-induced basal BDNF downregulation. Neurobiol. Aging 2015, 36, 2406–2413. [Google Scholar] [CrossRef]
  233. DaRocha-Souto, B.; Coma, M.; Perez-Nievas, B.; Scotton, T.; Siao, M.; Sánchez-Ferrer, P.; Hashimoto, T.; Fan, Z.; Hudry, E.; Barroeta, I. Activation of glycogen synthase kinase-3 beta mediates β-amyloid induced neuritic damage in Alzheimer’s disease. Neurobiol. Dis. 2012, 45, 425–437. [Google Scholar] [CrossRef]
  234. Barco, A.; Pittenger, C.; Kandel, E.R. CREB, memory enhancement and the treatment of memory disorders: Promises, pitfalls and prospects. Expert Opin. Ther. Targets 2003, 7, 101–114. [Google Scholar] [CrossRef]
  235. Christensen, R.; Marcussen, A.B.; Wörtwein, G.; Knudsen, G.; Aznar, S. Aβ (1–42) injection causes memory impairment, lowered cortical and serum BDNF levels, and decreased hippocampal 5-HT2A levels. Exp. Neurol. 2008, 210, 164–171. [Google Scholar] [CrossRef] [PubMed]
  236. Ciaramella, A.; Salani, F.; Bizzoni, F.; Orfei, M.D.; Langella, R.; Angelucci, F.; Spalletta, G.; Taddei, A.R.; Caltagirone, C.; Bossù, P. The stimulation of dendritic cells by amyloid beta 1–42 reduces BDNF production in Alzheimer’s disease patients. Brain Behav. Immun. 2013, 32, 29–32. [Google Scholar] [CrossRef] [PubMed]
  237. Zussy, C.; Brureau, A.; Keller, E.; Marchal, S.; Blayo, C.; Delair, B.; Ixart, G.; Maurice, T.; Givalois, L. Alzheimer’s disease related markers, cellular toxicity and behavioral deficits induced six weeks after oligomeric amyloid-β peptide injection in rats. PLoS ONE 2013, 8, e53117. [Google Scholar] [CrossRef] [PubMed]
  238. Wu, H.; Lu, D.; Jiang, H.; Xiong, Y.; Qu, C.; Li, B.; Mahmood, A.; Zhou, D.; Chopp, M. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/AKT pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J. Neurotrauma 2008, 25, 130–139. [Google Scholar] [CrossRef]
  239. Hu, Y.S.; Long, N.; Pigino, G.; Brady, S.T.; Lazarov, O. Molecular mechanisms of environmental enrichment: Impairments in AKT/GSK3β, neurotrophin-3 and CREB signaling. PLoS ONE. 2013, 8, e64460. [Google Scholar] [CrossRef] [PubMed]
  240. Nordberg, A. PET imaging of amyloid in Alzheimer’s disease. Lancet Neurol. 2004, 3, 519–527. [Google Scholar] [CrossRef]
  241. Caricasole, A.; Copani, A.; Caruso, A.; Caraci, F.; Iacovelli, L.; Sortino, M.A.; Terstappen, G.C.; Nicoletti, F. The Wnt pathway, cell-cycle activation and beta-amyloid: Novel therapeutic strategies in Alzheimer’s disease? Trends Pharmacol. Sci. 2003, 24, 233–238. [Google Scholar] [CrossRef]
  242. Lee, C.W.; Lau, K.F.; Miller, C.C.; Shaw, P.C. Glycogen synthase kinase-3 beta-mediated tau phosphorylation in cultured cell lines. Neuroreport 2003, 14, 257–260. [Google Scholar] [CrossRef]
  243. Yao, R.Q.; Qi, D.S.; Yu, H.L.; Liu, J.; Yang, L.H.; Wu, X.X. Quercetin attenuates cell apoptosis in focal cerebral ischemia rat brain via activation of BDNF-TrkB-PI3K/Akt signaling pathway. Neurochem. Res. 2012, 37, 2777–2786. [Google Scholar] [CrossRef]
  244. Datta, S.R.; Dudek, H.; Tao, X.; Masters, S.; Fu, H.; Gotoh, Y.; Greenberg, M.E. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 1997, 91, 231–241. [Google Scholar] [CrossRef]
  245. Cardone, M.H.; Roy, N.; Stennicke, H.R.; Salvesen, G.S.; Franke, T.F.; Stanbridge, E.; Frisch, S.; Reed, J.C. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998, 282, 1318–1321. [Google Scholar] [CrossRef] [PubMed]
  246. Cheng, B.; Martinez, A.A.; Morado, J.; Scofield, V.; Roberts, J.L.; Maffi, S.K. Retinoic acid protects against proteasome inhibition associated cell death in SH-SY5Y cells via the AKT pathway. Neurochem. Int. 2013, 62, 31–42. [Google Scholar] [CrossRef] [PubMed]
  247. Lee, K.Y.; Koh, S.H.; Noh, M.Y.; Park, K.W.; Lee, Y.J.; Kim, S.H. Glycogen synthase kinase-3beta activity plays very important roles in determining the fate of oxidative stress-inflicted neuronal cells. Brain Res. 2007, 1129, 89–99. [Google Scholar] [CrossRef] [PubMed]
  248. Gu, Y.; Zhang, X.; Xu, A.; Chen, W.; Liu, K.; Wu, L.; Mo, S.; Hu, Y.; Liu, M.; Luo, Q. Protein-ligand binding affinity prediction with edge awareness and supervised attention. iScience 2022, 26, 105892. [Google Scholar] [CrossRef]
  249. Jain, A.N. Surflex: Fully automatic flexible molecular docking using a molecular similarity-based search engine. J. Med. Chem. 2003, 46, 499–511. [Google Scholar] [CrossRef]
  250. Simayi, J.; Bayinsang; Nuermaimaiti, M.; Hailati, S.; Han, M.; Reheman, Z.; Wumaier, A.; Zhou, W. A Network Pharmacology-Based Study on the Mechanism of Dibutyl Phthalate of Ocimum basilicum L. against Alzheimer’s Disease through the AKT/GSK-3β Pathway. BioMed Res. Int. 2022, 2022, 9494548. [Google Scholar] [CrossRef]
  251. Rahmatkar, S.N.; Rana, A.K.; Kumar, R.; Singh, D. Fagopyrum tataricum (L.) Gaertn interacts with Gsk-3β/Nrf-2 signalling to protect neurotoxicity in a zebrafish model. J. Ethnopharmacol. 2023, 319, 117187. [Google Scholar] [CrossRef]
  252. Chiu, Y.J.; Teng, Y.S.; Chen, C.M.; Sun, Y.C.; Hsieh-Li, H.M.; Chang, K.H.; Lee-Chen, G.J. A Neuroprotective Action of Quercetin and Apigenin through Inhibiting Aggregation of Aβ and Activation of TRKB Signaling in a Cellular Experiment. Biomol. Ther. 2023, 31, 285–297. [Google Scholar] [CrossRef]
  253. Das, S.; Sengupta, S.; Chakraborty, S. Scope of β-secretase (bace1)-targeted therapy in alzheimer’s disease: Emphasizing the flavonoid based natural scaffold for bace1 inhibition. CS Chem. Neurosci. 2020, 11, 3510–3522. [Google Scholar] [CrossRef]
  254. Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and flavones as BACE-1 inhibitors: Structure–activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Et Biophys. Acta (BBA)-Gen. Subj. 2008, 1780, 819–825. [Google Scholar] [CrossRef]
  255. Li, E.; Yan, K.; Zhang, R.; Zou, P.; Li, S.; Ma, Q.; Liao, B. Kaempferol Protects Against Apoptosis in PC12 Cells Exposed to Hydrogen Peroxide by Activating Akt1. Nat. Prod. Commun. 2023, 18, 1934578X231170448. [Google Scholar] [CrossRef]
  256. Zhang, S.; Lu, Y.; Chen, W.; Shi, W.; Zhao, Q.; Zhao, J.; Li, L. Network Pharmacology and Experimental Evidence: PI3K/AKT Signaling Pathway is Involved in the Antidepressive Roles of Chaihu Shugan San. Drug Des. Dev. Ther. 2021, 15, 3425–3441. [Google Scholar] [CrossRef]
  257. Shi, Y.; Chen, M.; Zhao, Z.; Pan, J.; Huang, S. Network pharmacology and molecular docking analyses of mechanisms underlying effects of the cyperi rhizoma-chuanxiong rhizoma herb pair on depression. Evid. -Based Complement. Altern. Med. 2021, 2021, 5704578. [Google Scholar] [CrossRef] [PubMed]
  258. Zhang, S.S.; Liu, M.; Liu, D.N.; Shang, Y.F.; Du, G.H.; Wang, Y.H. Network pharmacology analysis and experimental validation of kaempferol in the treatment of ischemic stroke by inhibiting apoptosis and regulating neuroinflammation involving neutrophils. Int. J. Mol. Sci. 2022, 23, 12694. [Google Scholar] [CrossRef] [PubMed]
  259. Touati, I.; Abdalla, M.; Ali, N.H.; AlRuwaili, R.; Alruwaili, M.; Britel, M.R.; Maurady, A. Constituents of Stachys plants as potential dual inhibitors of AChE and NMDAR for the treatment of Alzheimer’s disease: A molecular docking and dynamic simulation study. J. Biomol. Struct. Dyn. 2023, 1–17. [Google Scholar] [CrossRef]
  260. Rasouli, H.; Hosseini Ghazvini, S.M.B.; Yarani, R.; Altıntaş, A.; Jooneghani, S.G.N.; Ramalho, T.C. Deciphering inhibitory activity of flavonoids against tau protein kinases: A coupled molecular docking and quantum chemical study. J. Biomol. Struct. Dyn. 2022, 40, 411–424. [Google Scholar] [CrossRef]
  261. Omar, S.H.; Scott, C.J.; Hamlin, A.S.; Obied, H.K. Biophenols: Enzymes (β-secretase, Cholinesterases, histone deacetylase and tyrosinase) inhibitors from olive (Olea europaea L.). Fitoterapia 2018, 128, 118–129. [Google Scholar] [CrossRef]
  262. Chukwuma, I.F.; Ezeorba, T.P.C.; Nworah, F.N.; Apeh, V.O.; Khalid, M.; Sweilam, S.H. Bioassay-guided identification of potential Alzheimer’s disease therapeutic agents from Kaempferol-Enriched fraction of Aframomum melegueta seeds using in vitro and chemoinformatics approaches. Arab. J. Chem. 2023, 16, 105089. [Google Scholar] [CrossRef]
  263. Pandey, D.; Pal, T.; Sharma, A. Phytochemicals as Potential Anti-Alzheimer’s Agents- An In-Silico Evidence. J. Dis. Markers 2022, 7, 1047. [Google Scholar]
  264. Grewal, A.K.; Singh, T.G.; Sharma, D.; Sharma, V.; Singh, M.; Rahman, M.H.; Najda, A.; Walasek-Janusz, M.; Kamel, M.; Albadrani, G.M.; et al. Mechanistic insights and perspectives involved in neuroprotective action of quercetin. Biomed. Pharmacother. 2021, 140, 111729. [Google Scholar] [CrossRef]
  265. Gong, P.; Wang, D.; Cui, D.; Yang, Q.; Wang, P.; Yang, W.; Chen, F. Anti-aging function and molecular mechanism of Radix Astragali and Radix Astragali preparata via network pharmacology and PI3K/Akt signaling pathway. Phytomedicine 2021, 84, 153509. [Google Scholar] [CrossRef]
  266. Sadighparvar, S.; Darband, S.G.; Yousefi, B.; Kaviani, M.; Ghaderi-Pakdel, F.; Mihanfar, A.; Mobaraki, K.; Majidinia, M. Combination of quercetin and exercise training attenuates depression in rats with 1,2-dimethylhydrazine-induced colorectal cancer: Possible involvement of inflammation and BDNF signalling. Exp. Physiol. 2020, 105, 1598–1609. [Google Scholar] [CrossRef]
  267. Khan, H.; Singh, A.; Thapa, K.; Garg, N.; Grewal, A.K.; Singh, T.G. Therapeutic modulation of the phosphatidylinositol 3-kinases (PI3K) pathway in cerebral ischemic injury. Brain Res. 2021, 1761, 147399. [Google Scholar] [CrossRef] [PubMed]
  268. Rezai-Zadeh, K.; Arendash, G.W.; Hou, H.; Fernandez, F.; Jensen, M.; Runfeldt, M.; Shytle, R.D.; Tan, J. Green tea epigallocatechin-3-gallate (EGCG) reduces beta-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Res. 2008, 1214, 177–187. [Google Scholar] [CrossRef] [PubMed]
  269. Gong, E.J.; Park, H.R.; Kim, M.E.; Piao, S.; Lee, E.; Jo, D.G.; Chung, H.Y.; Ha, N.C.; Mattson, M.P.; Lee, J. Morin attenuates tau hyperphosphorylation by inhibiting GSK3beta. Neurobiol. Dis. 2011, 44, 223–230. [Google Scholar] [CrossRef] [PubMed]
  270. Miyai, S.; Yamaguchi, A.; Iwasaki, T.; Shamsa, F.; Ohtsuki, K. Biochemical characterization of epigallocatechin-3-gallate as an effective stimulator for the phosphorylation of its binding proteins by glycogen synthase kinase-3beta in vitro. Biol. Pharm. Bull. 2010, 33, 1932–1937. [Google Scholar] [CrossRef]
  271. Ashrafpour, M.; Parsaei, S.; Sepehri, H. Quercetin improved spatial memory dysfunctions in rat model of intracerebroventricular streptozotocin-induced Alzheimer’s disease. Natl. J. Physiol. Pharm. Pharmacol. 2015, 5, 411. [Google Scholar] [CrossRef]
  272. Tong-un, T.; Muchimapura, S.; Phachonpai, W.; Wattanathorn, J. Effects of quercetin encapsulated liposomes via nasal administration: A novel cognitive enhancer. Am. J. Appl. Sci. 2010, 7, 906–913. [Google Scholar] [CrossRef]
  273. Sriraksa, N.; Wattanathorn, J.; Muchimapura, S.; Tiamkao, S.; Brown, K.; Chaisiwamongkol, K. Cognitive-enhancing effect of quercetin in a rat model of Parkinson’s disease induced by 6-hyrodoxydopamine. J. Evid. Based Complement. Alternat. Med. 2011, 2012, 823206. [Google Scholar] [CrossRef]
  274. Elreedy, H.A.; Elfiky, A.M.; Ahmed Mahmoud, A.; Ibrahim, K.S.; Ghazy, M.A. Effect Of Quercetin As Therapeutic And Protective Agent In Aluminum Chloride-Induced Alzheimer’s Disease Rats. Egypt. J. Chem. 2022, 65, 633–641. [Google Scholar] [CrossRef]
  275. Elfiky, A.M.; Mahmoud, A.A.; Elreedy, H.A.; Ibrahim, K.S.; Ghazy, M.A. Quercetin stimulates the non-amyloidogenic pathway via activation of ADAM10 and ADAM17 gene expression in aluminum chloride-induced Alzheimer’s disease rat model. Life Sci. 2021, 285, 119964. [Google Scholar] [CrossRef] [PubMed]
  276. Singh, N.K.; Garabadu, D. Quercetin Exhibits α7nAChR/Nrf2/HO-1-Mediated Neuroprotection Against STZ-Induced Mitochondrial Toxicity and Cognitive Impairments in Experimental Rodents. Neurotox. Res. 2021, 39, 1859–1879. [Google Scholar] [CrossRef]
  277. Parent, M.; Chitturi, J.; Santhakumar, V.; Hyder, F.; Sanganahalli, B.G.; Kannurpatti, S.S. Kaempferol Treatment after Traumatic Brain Injury during Early Development Mitigates Brain Parenchymal Microstructure and Neural Functional Connectivity Deterioration at Adolescence. J. Neurotrauma 2020, 37, 966–974. [Google Scholar] [CrossRef] [PubMed]
  278. Zhang, W.; Lv, M.; Shi, Y.; Mu, Y.; Yao, Z.; Yang, Z. Network Pharmacology-Based Study of the Underlying Mechanisms of Huangqi Sijunzi Decoction for Alzheimer’s Disease. Evid. Based Complement. Altern. Med. 2021, 2021, 6480381. [Google Scholar] [CrossRef] [PubMed]
  279. Matsuo, E.S.; Shin, R.W.; Billingsley, M.L.; van de Voorde, A.; O’Connor, M.; Trojanowski, J.Q.; Lee, V.M. Biopsy-derived adult human brain tau is phosphorylated at many of the same sites as Alzheimer’s disease paired helical filament tau. Neuron 1994, 13, 989–1002. [Google Scholar] [CrossRef]
  280. Iqbal, K.; Liu, F.; Gong, C.X. Tau and neurodegenerative disease: The story so far. Nat. Rev. Neurol. 2016, 12, 15–27. [Google Scholar] [CrossRef]
  281. Xia, Y.; Liu, R.; Chen, R.; Tian, Q.; Zeng, K.; Hu, J.; Liu, X.; Wang, Q.; Wang, P.; Wang, X.C.; et al. Novel multipotent AChEI-CCB attenuates hyperhomocysteinemia-induced memory deficits and Neuropathologies in rats. J. Alzheimer's Dis. 2014, 42, 1029–1039. [Google Scholar] [CrossRef]
  282. Russo, M.; Milito, A.; Spagnuolo, C.; Carbone, V.; Rosén, A.; Minasi, P.; Lauria, F.; Russo, G.L. CK2 and PI3K are direct molecular targets of quercetin in chronic lymphocytic leukaemia. Oncotarget 2017, 8, 42571–42587. [Google Scholar] [CrossRef]
  283. Images Created. Available online: BioRender.com (accessed on 6 October 2023).
  284. Fang, J.; Wang, L.; Wu, T.; Yang, C.; Gao, L.; Cai, H.; Liu, J.; Fang, S.; Chen, Y.; Tan, W.; et al. Network pharmacology-based study on the mechanism of action for herbal medicines in Alzheimer treatment. J. Ethnopharmacol. 2017, 196, 281–292. [Google Scholar] [CrossRef]
  285. Luo, Y.; Smith, J.V.; Paramasivam, V.; Burdick, A.; Curry, K.J.; Buford, J.P.; Khan, I.; Netzer, W.J.; Xu, H.; Butko, P. Inhibition of amyloid-β aggregation and caspase-3 activation by the Ginkgo biloba extract EGb761. Proc. Natl. Acad. Sci. USA 2002, 99, 12197–12202. [Google Scholar] [CrossRef]
  286. Longpré, F.; Garneau, P.; Christen, Y.; Ramassamy, C. Protection by EGb 761 against beta-amyloid-induced neurotoxicity: Involvement of NF-kappaB, SIRT1, and MAPKs pathways and inhibition of amyloid fibril formation. Free. Radic. Biol. Med. 2006, 41, 1781–1794. [Google Scholar] [CrossRef]
  287. Tchantchou, F.; Xu, Y.; Wu, Y.; Christen, Y.; Luo, Y. EGb 761 enhances adult hippocampal neurogenesis and phosphorylation of CREB in transgenic mouse model of Alzheimer’s disease. FASEB J. 2007, 21, 2400–2408. [Google Scholar] [CrossRef] [PubMed]
  288. Kwon, K.J.; Lee, E.J.; Cho, K.S.; Cho, D.H.; Shin, C.Y.; Han, S.H. Ginkgo biloba extract (Egb761) attenuates zinc-induced tau phosphorylation at Ser262 by regulating GSK3β activity in rat primary cortical neurons. Food Funct. 2015, 6, 2058–2067. [Google Scholar] [CrossRef] [PubMed]
  289. Lejri, I.; Grimm, A.; Eckert, A. Ginkgo biloba extract increases neurite outgrowth and activates the Akt/mTOR pathway. PLoS ONE. 2019, 14, e0225761. [Google Scholar] [CrossRef] [PubMed]
  290. Rhein, V.; Song, X.; Wiesner, A.; Ittner, L.M.; Baysang, G.; Meier, F.; Ozmen, L.; Bluethmann, H.; Dröse, S.; Brandt, U.; et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimer’s disease mice. Proc. Natl. Acad. Sci. USA 2009, 106, 20057–20062. [Google Scholar] [CrossRef]
  291. Muller, W.E.; Heiser, J.; Leuner, K. Effects of the standardized Ginkgo biloba extract EGb 761(R) on neuroplasticity. Int. Psychogeriatrics 2012, 24 (Suppl. S1), S21–S24. [Google Scholar] [CrossRef]
  292. Xu, Y.; Cui, C.; Pang, C.; Christen, Y.; Luo, Y. Restoration of impaired phosphorylation of cyclic AMP response element-binding protein (CREB) by EGb 761 and its constituents in Abeta-expressing neuroblastoma cells. Eur. J. Neurosci. 2007, 26, 2931–2939. [Google Scholar] [CrossRef]
  293. Tchantchou, F.; Lacor, P.N.; Cao, Z.; Lao, L.; Hou, Y.; Cui, C.; Klein, W.L.; Luo, Y. Stimulation of neurogenesis and synaptogenesis by bilobalide and quercetin via common final pathway in hippocampal neurons. J. Alzheimer's Dis. 2009, 18, 787–798. [Google Scholar] [CrossRef]
  294. Tian, R.; Wang, H.; Xiao, Y.; Hu, P.; Du, R.; Shi, X.; Wang, Z.; Xie, Y. Fabrication of nanosuspensions to improve the oral bioavailability of total flavones from Hippophae rhamnoides L. and their comparison with an inclusion complex. AAPS Pharmscitech 2020, 21, 249. [Google Scholar] [CrossRef]
  295. Xia, C.X.; Gao, A.X.; Zhu, Y.; Dong, T.T.; Tsim, K.W. Flavonoids from Seabuckthorn (Hippophae rhamnoides L.) restore CUMS-induced depressive disorder and regulate the gut microbiota in mice. Food Funct. 2023, 14, 7426–7438. [Google Scholar] [CrossRef]
  296. Liu, H.; Ye, M.; Guo, H. An Updated Review of Randomized Clinical Trials Testing the Improvement of Cognitive Function of Ginkgo biloba Extract in Healthy People and Alzheimer’s Patients. Front. Pharmacol. 2020, 10, 1688. [Google Scholar] [CrossRef] [PubMed]
  297. Jeganathan, B.; Punyasiri, P.A.; Kottawa-Arachchi, J.D.; Ranatunga, M.A.; Abeysinghe, I.S.; Gunasekare, M.T.; Bandara, B.M. Genetic Variation of Flavonols Quercetin, Myricetin, and Kaempferol in the Sri Lankan Tea (Camellia sinensis L.) and Their Health-Promoting Aspects. Int. J. Food Sci. 2016, 2016, 6057434. [Google Scholar] [CrossRef] [PubMed]
  298. Ishan, T.; Lalit, S.; Girdhari, G.; Gupta, G.L. Synergistic antioxidant activity of three medicinal plants Hypericum perforatum, Bacopa monnieri, and Camellia sinensis. Indo Am. J. Pharm. Res. 2014, 4, 2563–2568. [Google Scholar]
  299. Mahmoodzadeh, T.; Kashani, M.H.K.; Ramshini, H.; Moslem, A.; Mohammad-Zadeh, M. Effect of Camellia sinensis on Spatial Memory in a Rat Model of Alzheimer’s Disease. J. Biomed. 2016, 1, e5340. [Google Scholar] [CrossRef]
  300. Onasanwo, S.A.; Adamaigbo, V.O.; Adebayo, O.G.; Eleazer, S.E. Moringa oleifera-supplemented diet protect against cortico-hippocampal neuronal degeneration in scopolamine-induced spatial memory deficit in mice: Role of oxido-inflammatory and cholinergic neurotransmission pathway. Metab. Brain Dis. 2021, 36, 2445–2460. [Google Scholar] [CrossRef]
  301. Sermkaew, N.; Plyduang, T. Self-microemulsifying drug delivery systems of Moringa oleifera extract for enhanced dissolution of kaempferol and quercetin. Acta Pharm. 2020, 70, 77–88. [Google Scholar] [CrossRef]
  302. Liu, W.L.; Wu, B.F.; Shang, J.H.; Wang, X.F.; Zhao, Y.L.; Huang, A.X. Moringa oleifera seed ethanol extract and its active component kaempferol potentiate pentobarbital-induced sleeping behaviours in mice via a GABAergic mechanism. Pharm. Biol. 2022, 60, 810–824. [Google Scholar] [CrossRef]
  303. Sun, Y.; Wu, A.; Li, X.; Qin, D.; Jin, B.; Liu, J.; Tang, Y.; Wu, J.; Yu, C. The seed of Litchi chinensis fraction ameliorates hippocampal neuronal injury in an Aβ25-35-induced Alzheimer’s disease rat model via the AKT/GSK-3β pathway. Pharm. Biol. 2020, 58, 35–43. [Google Scholar] [CrossRef]
  304. El Gizawy, H.A.; Abo-Salem, H.M.; Ali, A.A.; Hussein, M.A. Phenolic Profiling and Therapeutic Potential of Certain Isolated Compounds from Parkia roxburghii against AChE Activity as well as GABAA α5, GSK-3β, and p38α MAP-Kinase Genes. ACS Omega 2021, 6, 20492–20511. [Google Scholar] [CrossRef]
  305. Singh, S.; Singh, T.G.; Mahajan, K.; Dhiman, S. Medicinal plants used against various inflammatory biomarkers for the management of rheumatoid arthritis. J. Pharm. Pharmacol. 2020, 72, 1306–1327. [Google Scholar] [CrossRef]
  306. Liu, Y.; Xue, Q.; Li, A.; Li, K.; Qin, X. Mechanisms exploration of herbal pair of HuangQi-DanShen on cerebral ischemia based on metabonomics and network pharmacology. J. Ethnopharmacol. 2020, 253, 112688. [Google Scholar] [CrossRef] [PubMed]
  307. Moriwaki, M.; Tominaga, E.; Kito, K.; Nakagawa, R.; Kapoor, M.P.; Matsumiya, Y.; Fukuhara, T.; Kobayashi, J.; Satomoto, K.; Yamagata, H.; et al. Bioavailability of Flavonoids in Ginkgo biloba Extract-γ-Cyclodextrin Complex. Nat. Prod. Commun. 2023, 18, 1934578X231170221. [Google Scholar] [CrossRef]
  308. Zubčić, K.; Radovanović, V.; Vlainić, J.; Hof, P.R.; Oršolić, N.; Šimić, G.; Jazvinšćak Jembrek, M. PI3K/Akt and ERK1/2 Signalling Are Involved in Quercetin-Mediated Neuroprotection against Copper-Induced Injury. Oxidative Med. Cell. Longev. 2020, 2020, 9834742. [Google Scholar] [CrossRef] [PubMed]
  309. Permana, A.D.; Maddeppungeng, N.M.; Asma, N.; Rahim, A.; Nainu, F.; Bahar, M.A.; Yulianty, R. Validation of HPLC-UV method for simultaneous determination of quercetin and luteolin from chartamus tinctorius L in solid lipid nanoparticles incorporated in floating gel in situ formulation. Microchem. J. 2023, 194, 109373. [Google Scholar] [CrossRef]
  310. Cho, K.M.; Lee, H.Y.; Cho, D.Y.; Jung, J.G.; Kim, M.J.; Bin Jeong, J.; Jang, S.N.; Lee, G.O.; Sim, H.-S.; Kang, M.J.; et al. Comprehensive Comparison of Chemical Composition and Antioxidant Activity of Panax ginseng Sprouts by Different Cultivation Systems in a Plant Factory. Plants 2022, 11, 1818. [Google Scholar] [CrossRef]
  311. Lin, M.K.; Lee, M.S.; Huang, H.C.; Cheng, T.J.; Cheng, Y.D.; Wu, C.R. Cuscuta chinensis and C. campestris attenuate scopolamine-induced memory deficit and oxidative damage in mice. Molecules 2018, 23, 3060. [Google Scholar] [CrossRef]
  312. Marefati, N.; Ghorani, V.; Shakeri, F.; Boskabady, M.; Kianian, F.; Rezaee, R.; Boskabady, M.H. A review of anti-inflammatory, antioxidant, and immunomodulatory effects of Allium cepa and its main constituents. Pharm. Biol. 2021, 59, 285–300. [Google Scholar] [CrossRef]
  313. Sagar, N.A.; Pareek, S.; Benkeblia, N.; Xiao, J. Onion (Allium cepa L.) bioactives: Chemistry, pharmacotherapeutic functions, and industrial applications. Food Front. 2022, 3, 380–412. [Google Scholar] [CrossRef]
  314. Tan, S.; Tang, J.; Shi, W.; Wang, Z.; Xiang, Y.; Deng, T.; Gao, X.; Li, W.; Shi, S. Effects of three drying methods on polyphenol composition and antioxidant activities of Litchi chinensis Sonn. Food Sci. Biotechnol. 2019, 29, 351–358. [Google Scholar] [CrossRef]
  315. Singh, B.; Singh, J.P.; Kaur, A.; Singh, N. Phenolic compounds as beneficial phytochemicals in pomegranate (Punica granatum L.) peel: A review. Food Chem. 2018, 261, 75–86. [Google Scholar] [CrossRef]
  316. Chen, Z.; Liu, L.; Gao, C.; Chen, W.; Vong, C.T.; Yao, P.; Yang, Y.; Li, X.; Tang, X.; Wang, S.; et al. Astragali Radix (Huangqi): A promising edible immunomodulatory herbal medicine. J. Ethnopharmacol. 2020, 258, 112895. [Google Scholar] [CrossRef]
  317. Pool, H.; Mendoza, S.; Xiao, H.; McClements, D.J. Encapsulation and release of hydrophobic bioactive components in nanoemulsion-based delivery systems: Impact of physical form on quercetin bioaccessibility. Food Funct. 2013, 4, 162–174. [Google Scholar] [CrossRef]
  318. Bangar, S.P.; Chaudhary, V.; Sharma, N.; Bansal, V.; Ozogul, F.; Lorenzo, J.M. Kaempferol: A flavonoid with wider biological activities and its applications. Crit. Rev. Food Sci. Nutr. 2022, 1–25. [Google Scholar] [CrossRef]
  319. Elahy, M.; Jackaman, C.; Mamo, J.C.; Lam, V.; Dhaliwal, S.S.; Giles, C.; Nelson, D.; Takechi, R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun. Ageing 2015, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  320. Popescu, B.O.; Toescu, E.C.; Popescu, L.M.; Bajenaru, O.; Muresanu, D.F.; Schultzberg, M.; Bogdanovic, N. Blood-brain barrier alterations in ageing and dementia. J. Neurol. Sci. 2009, 283, 99–106. [Google Scholar] [CrossRef] [PubMed]
  321. Leoni, V.; Masterman, T.; Patel, P.; Meaney, S.; Diczfalusy, U.; Björkhem, I. Side chain oxidized oxysterols in cerebrospinal fluid and the integrity of blood-brain and blood-cerebrospinal fluid barriers. J. Lipid Res. 2003, 44, 793–799. [Google Scholar] [CrossRef] [PubMed]
  322. Blair, L.J.; Frauen, H.D.; Zhang, B.; Nordhues, B.A.; Bijan, S.; Lin, Y.-C.; Zamudio, F.; Hernandez, L.D.; Sabbagh, J.J.; Selenica, M.-L.B.; et al. Tau depletion prevents progressive blood-brain barrier damage in a mouse model of tauopathy. Acta Neuropathol. Commun. 2015, 3, 8. [Google Scholar] [CrossRef]
  323. Majerova, P.; Michalicova, A.; Cente, M.; Hanes, J.; Vegh, J.; Kittel, A.; Kosikova, N.; Cigankova, V.; Mihaljevic, S.; Jadhav, S.; et al. Trafficking of immune cells across the blood-brain barrier is modulated by neurofibrillary pathology in tauopathies. PLoS ONE 2019, 14, e0217216. [Google Scholar] [CrossRef]
  324. Moradi-Afrapoli, F.; Oufir, M.; Walter, F.R.; Deli, M.A.; Smiesko, M.; Zabela, V.; Butterweck, V.; Hamburger, M. Validation of UHPLC-MS/MS methods for the determination of kaempferol and its metabolite 4-hydroxyphenyl acetic acid, and application to in vitro blood-brain barrier and intestinal drug permeability studies. J. Pharm. Biomed. Anal. 2016, 128, 264–274. [Google Scholar] [CrossRef]
  325. Gupta, A.; Kaur, C.D.; Saraf, S.; Saraf, S. Formulation, characterization, and evaluation of ligand-conjugated biodegradable quercetin nanoparticles for active targeting. Artif. Cells Nanomed. Biotechnol. 2016, 44, 960–970. [Google Scholar] [CrossRef]
  326. Dong, Y.; Tao, B.; Xue, X.; Feng, C.; Ren, Y.; Ma, H.; Zhang, J.; Si, Y.; Zhang, S.; Liu, S.; et al. Molecular mechanism of Epicedium treatment for depression based on network pharmacology and molecular docking technology. BMC Complement. Med. Ther. 2021, 21, 222. [Google Scholar] [CrossRef] [PubMed]
  327. Wu, D.; Zhang, L.; Zhang, S. Distribution of quercetin in plasma and tissues in rats. Chin. J. Hosp. Pharm. 2008, 28, 1822–1824. [Google Scholar]
  328. Orhan, I.E. Cholinesterase Inhibitory Potential of Quercetin towards Alzheimer’s Disease—A Promising Natural Molecule or Fashion of the Day?—A Narrowed Review. Curr. Neuropharmacol. 2021, 19, 2205–2213. [Google Scholar] [CrossRef] [PubMed]
  329. Imran, M.; Thabet, H.K.; Alaqel, S.I.; Alzahrani, A.R.; Abida, A.; Alshammari, M.K.; Kamal, M.; Diwan, A.; Asdaq, S.M.B.; Alshehri, S. The Therapeutic and Prophylactic Potential of Quercetin against COVID-19: An Outlook on the Clinical Studies, Inventive Compositions, and Patent Literature. Antioxidants 2022, 11, 876. [Google Scholar] [CrossRef] [PubMed]
  330. Huebbe, P.; Wagner, A.E.; Boesch-Saadatmandi, C.; Sellmer, F.; Wolffram, S.; Rimbach, G. Effect of dietary quercetin on brain quercetin levels and the expression of antioxidant and Alzheimer’s disease relevant genes in mice. Pharmacol. Res. 2010, 61, 242–246. [Google Scholar] [CrossRef]
  331. Crozier, A.; Del Rio, D.; Clifford, M.N. Bioavailability of Dietary Flavonoids and Phenolic Compounds. Mol. Asp. Med. 2010, 31, 446–467. [Google Scholar] [CrossRef]
  332. Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem. 2015, 26, 201–210. [Google Scholar] [CrossRef]
  333. Ebrahimpour, S.; Zakeri, M.; Esmaeili, A. Crosstalk between obesity, diabetes, and alzheimer’s disease: Introducing quercetin as an effective triple herbal medicine. Ageing Res. Rev. 2020, 62, 101095. [Google Scholar] [CrossRef]
  334. Pinheiro, R.; Granja, A.; Loureiro, J.; Pereira, M.; Pinheiro, M.; Neves, A.; Reis, S. RVG29-Functionalized Lipid Nanoparticles for Quercetin Brain Delivery and Alzheimer’s Disease. Pharm. Res. 2020, 37, 139. [Google Scholar] [CrossRef]
  335. Qi, Y.; Yi, P.; He, T.; Song, X.; Liu, Y.; Li, Q.; Zheng, J.; Song, R.; Liu, C.; Zhang, Z.; et al. Quercetin-loaded selenium nanoparticles inhibit amyloid-β aggregation and exhibit antioxidant activity. Colloids Surf. A Physicochem. Eng. Asp. 2020, 602, 125058. [Google Scholar] [CrossRef]
  336. Chen, G.; Yang, F.; Fan, S.; Jin, H.; Liao, K.; Li, X.; Liu, G.-B.; Liang, J.; Zhang, J.; Xu, J.F.; et al. Immunomodulatory roles of selenium nanoparticles: Novel arts for potential immunotherapy strategy development. Front. Immunol. 2022, 13, 956181. [Google Scholar] [CrossRef] [PubMed]
  337. Kieliszek, M.; Błażejak, S.; Bzducha-Wróbel, A.; Kot, A.M. Effect of selenium on growth and antioxidative system of yeast cells. Mol. Biol. Rep. 2019, 46, 1797–1808. [Google Scholar] [CrossRef] [PubMed]
  338. Colombo, M.; de Lima Melchiades, G.; Michels, L.R.; Figueiró, F.; Bassani, V.L.; Teixeira, H.F.; Koester, L.S. Solid Dispersion of Kaempferol: Formulation Development, Characterization, and Oral Bioavailability Assessment. AAPS PharmSciTech 2019, 20, 106. [Google Scholar] [CrossRef] [PubMed]
  339. Alkandahri, M.Y.; Pamungkas, B.T.; Oktoba, Z.; Shafirany, M.Z.; Sulastri, L.; Arfania, M.; Anggraeny, E.N.; Pratiwi, A.; Astuti, F.D.; Indriyani, D.S.Y.; et al. Hepatoprotective Effect of Kaempferol: A Review of the Dietary Sources, Bioavailability, Mechanisms of Action, and Safety. Adv. Pharmacol. Pharm. Sci. 2023, 2023, 1387665. [Google Scholar] [CrossRef] [PubMed]
  340. Zhang, M.; Kong, L.; Luo, C.; Li, X.; Zhou, Y. Pharmacokinetic Study of Keampferol in Rabbits. China Pharm. 2014, 25, 4040–4042. [Google Scholar]
  341. Salehi, B.; Machin, L.; Monzote, L.; Sharifi-Rad, J.; Ezzat, S.M.; Salem, M.A.; Merghany, R.M.; El Mahdy, N.M.; Kılıç, C.S.; Sytar, O.; et al. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health. ACS Omega 2020, 5, 11849–11872. [Google Scholar] [CrossRef]
  342. Liu, Y.; Gong, Y.; Xie, W.; Huang, A.; Yuan, X.; Zhou, H.; Zhu, X.; Chen, X.; Liu, J.; Liu, J.; et al. Microbubbles in combination with focused ultrasound for the delivery of quercetin-modified sulfur nanoparticles through the blood brain barrier into the brain parenchyma and relief of endoplasmic reticulum stress to treat Alzheimer’s disease. Nanoscale 2020, 12, 6498–6511. [Google Scholar]
  343. Chandekar, L.; Katgeri, R.; Takke, A. The Potential Clinical Uses and Nanoformulation Strategies of Kaempferol, a Dietary Flavonoid. Rev. Bras. de Farm. 2022, 32, 693–707. [Google Scholar]
  344. Benameur, T.; Soleti, R.; Porro, C. The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases. Nutrients 2021, 13, 1318. [Google Scholar] [CrossRef]
  345. Nishimura, M.; Ohkawara, T.; Nakagawa, T.; Muro, T.; Sato, Y.; Satoh, H.; Kobori, M.; Nishihira, J. A randomized, double-blind, placebo-controlled study evaluating the effects of quercetin-rich onion on cognitive function in elderly subjects. Funct. Foods Heal. Dis. 2017, 7, 353. [Google Scholar] [CrossRef]
  346. Broman-Fulks, J.J.; Canu, W.H.; Trout, K.L.; Nieman, D.C. The effects of quercetin supplementation on cognitive functioning in a community sample: A randomized, placebo-controlled trial. Ther. Adv. Psychopharmacol. 2012, 2, 131–138. [Google Scholar] [CrossRef]
  347. Nakagawa, T.; Itoh, M.; Ohta, K.; Hayashi, Y.; Hayakawa, M.; Yamada, Y.; Akanabe, H.; Chikaishi, T.; Nakagawa, K.; Itoh, Y.; et al. Improvement of memory recall by quercetin in rodent contextual fear conditioning and human early-stage Alzheimer’s disease patients. Neuroreport 2016, 27, 671–676. [Google Scholar] [CrossRef] [PubMed]
  348. Liu, Y.; Zhou, H.; Yin, T.; Gong, Y.; Yuan, G.; Chen, L.; Liu, J. Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid Interface Sci. 2019, 552, 388–400. [Google Scholar] [CrossRef] [PubMed]
  349. Rananaware, P.; Pandit, P.; Naik, S.; Mishra, M.; Keria, R.S.; Brahmkhatri, V.P. Anti-amyloidogenic property of gold nanoparticle decorated querc. RSC Adv. 2022, 12, 23661–23674. [Google Scholar] [CrossRef] [PubMed]
  350. Zhu, C.; Yang, Y.; Li, X.; Chen, X.; Lin, X.; Wu, X. Develop potential multi-target drugs by self-assembly of quercetin with amino acids and metal ion to achieve significant efficacy in anti-Alzheimer’s disease. Nano Res. 2022, 15, 5173–5182. [Google Scholar]
  351. Cui, Z.; Zhao, X.; Amevor, F.K.; Du, X.; Wang, Y.; Li, D.; Shu, G.; Tian, Y.; Zhao, X. Therapeutic application of quercetin in aging-related diseases: SIRT1 as a potential mechanism. Front. Immunol. 2022, 13, 943321. [Google Scholar] [CrossRef] [PubMed]
  352. Karthika, C.; Appu, A.P.; Akter, R.; Rahman, H.; Tagde, P.; Ashraf, G.M.; Abdel-Daim, M.M.; Ul Hassan, S.S.; Abid, A.; Bungau, S. Potential innovation against Alzheimer’s disorder: A tricomponent combination of natural antioxidants (vitamin E, quercetin, and basil oil) and the development of its intranasal delivery. Environ. Sci. Pollut. Res. 2022, 29, 10950–10965. [Google Scholar]
  353. Alaqeel, N.K.; AlSheikh, M.H.; Al-Hariri, M.T. Quercetin Nanoemulsion Ameliorates Neuronal Dysfunction in Experimental Alzheimer’s Disease Model. Antioxidants 2022, 11, 19866. [Google Scholar] [CrossRef]
  354. Center for Food Safety and Applied Nutrition. (n.d.). Gras Notice Inventory—Agency Response Letter Gras Notice no. GRN 000341. Archive. Available online: https://www.cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set=GRASNotices&id=341 (accessed on 6 October 2023).
  355. Batiha, G.E.; Beshbishy, A.M.; Ikram, M.; Mulla, Z.S.; El-Hack, M.E.A.; Taha, A.E.; Algammal, A.M.; Elewa, Y.H.A. The Pharmacological Activity, Biochemical Properties, and Pharmacokinetics of the Major Natural Polyphenolic Flavonoid: Quercetin. Foods 2020, 9, 374. [Google Scholar] [CrossRef]
  356. Spencer, J.P.; Rice-Evans, C.; Williams, R.J. Modulation of pro-survival Akt/protein kinase B and ERK1/2 signaling cascades by quercetin and its in vivo metabolites underlie their action on neuronal viability. J. Biol. Chem. 2003, 278, 34783–34793. [Google Scholar] [CrossRef]
  357. Kimoto, H.; Fujiwara, S.; Koyama, N.; Uesugi, T. Genotoxicity and subchronic toxicity of a kaempferol aglycone-rich product produced from horseradish leaves. Fundam. Toxicol. Sci. 2022, 9, 71–83, 2189-115X. [Google Scholar] [CrossRef]
  358. Yangzom, P.; Amruthanand, S.; Sharma, M.; Mahajan, S.; Lingaraju, M.C.; Parida, S.; Sahoo, M.; Kumar, D.; Singh, T.U. Subacute 28 days oral toxicity study of kaempferol and biochanin-A in the mouse model. J. Biochem. Mol. Toxicol. 2022, 36, e23090. [Google Scholar] [CrossRef] [PubMed]
  359. Akiyama, M.; Mizokami, T.; Ito, H.; Ikeda, Y. A randomized, placebo-controlled trial evaluating the safety of excessive administration of kaempferol aglycone. Food Sci. Nutr. 2023, 11, 5427–5437. [Google Scholar] [CrossRef] [PubMed]
  360. Hart, J.J.; Tako, E.; Wiesinger, J.; Glahn, R.P. Polyphenolic Profiles of Yellow Bean Seed Coats and Their Relationship with Iron Bioavailability. J. Agric. Food Chem. 2020, 68, 769–778. [Google Scholar] [CrossRef]
  361. Vaez, S.; Parivr, K.; Amidi, F.; Rudbari, N.H.; Moini, A.; Amini, N. Quercetin and polycystic ovary syndrome; inflammation, hormonal parameters and pregnancy outcome: A randomized clinical trial. Am. J. Reprod. Immunol. 2023, 89, e13644. [Google Scholar] [CrossRef]
  362. Thakur, K.; Zhu, Y.Y.; Feng, J.Y.; Zhang, J.G.; Hu, F.; Prasad, C.; Wei, Z.J. Morin as an imminent functional food ingredient: An update on its enhanced efficacy in the treatment and prevention of metabolic syndromes. Food Funct. 2020, 11, 8424–8443. [Google Scholar] [CrossRef]
  363. Carmona, V.; Martín-Aragón, S.; Goldberg, J.; Schubert, D.; Bermejo-Bescós, P. Several targets involved in Alzheimer’s disease amyloidogenesis are affected by morin and isoquercitrin. Nutr. Neurosci. 2020, 23, 575–590. [Google Scholar] [CrossRef]
  364. Gur, C.; Kandemir, F.M.; Darendelioglu, E.; Caglayan, C.; Kucukler, S.; Kandemir, O.; Ileriturk, M. Morin protects against acrylamide-induced neurotoxicity in rats: An investigation into different signal pathways. Environ. Sci. Pollut. Res. 2021, 28, 49808–49819. [Google Scholar] [CrossRef]
  365. Mohammadi, N.; Asle-Rousta, M.; Rahnema, M.; Amini, R. Morin attenuates memory deficits in a rat model of Alzheimer’s disease by ameliorating oxidative stress and neuroinflammation. Eur. J. Pharmacol. 2021, 910, 174506. [Google Scholar] [CrossRef]
  366. Soubh, A.A.; El-Gazar, A.A.; Mohamed, E.A.; Awad, A.S.; El-Abhar, H.S. Further insights for the role of Morin in mRTBI: Implication of non-canonical Wnt/PKC-α and JAK-2/STAT-3 signaling pathways. Int. Immunopharmacol. 2021, 100, 108123. [Google Scholar] [CrossRef]
  367. Thabet, N.M.; Moustafa, E.M. Protective effect of rutin against brain injury induced by acrylamide or gamma radiation: Role of PI3K/AKT/GSK-3β/NRF-2 signalling pathway. Arch. Physiol. Biochem. 2018, 124, 185–193. [Google Scholar] [CrossRef]
  368. Çelik, H.; Kandemir, F.M.; Caglayan, C.; Özdemir, S.; Çomaklı, S.; Kucukler, S.; Yardım, A. Neuroprotective effect of rutin against colistin-induced oxidative stress, inflammation and apoptosis in rat brain associated with the CREB/BDNF expressions. Mol. Biol. Rep. 2020, 47, 2023–2034. [Google Scholar] [CrossRef]
  369. Ahmad, S.; Jo, M.H.; Ikram, M.; Khan, A.; Kim, M.O. Deciphering the potential neuroprotective effects of luteolin against Aβ1–42-Induced alzheimer’s disease. Int. J. Mol. Sci. 2021, 22, 9583. [Google Scholar] [CrossRef] [PubMed]
  370. He, H.; Chen, X. Luteolin Attenuates Cognitive Dysfunction Induced By Chronic Cerebral Hypoperfusion Through the Modulation of The PI3K/Akt Pathway in Rats. J. Vet. Res. 2021, 65, 341–349. [Google Scholar] [CrossRef] [PubMed]
  371. Sawmiller, D.; Li, S.; Shahaduzzaman; Smith, A.J.; Obregon, D.; Giunta, B.; Borlongan, C.V.; Sanberg, P.R.; Tan, J. Luteolin reduces Alzheimer’s disease pathologies induced by traumatic brain injury. Int. J. Mol. Sci. 2014, 15, 895–904. [Google Scholar] [CrossRef]
  372. Zeng, P.; Su, H.F.; Ye, C.Y.; Qiu, S.W.; Shi, A.; Wang, J.Z.; Zhou, X.W.; Tian, Q. A Tau Pathogenesis-Based Network Pharmacology Approach for Exploring the Protections of Chuanxiong Rhizoma in Alzheimer’s Disease. Front. Pharmacol. 2022, 13, 877806. [Google Scholar] [CrossRef]
  373. Yi, H.; Peng, H.; Wu, X.; Xu, X.; Kuang, T.; Zhang, J.; Du, L.; Fan, G. The Therapeutic Effects and Mechanisms of Quercetin on Metabolic Diseases: Pharmacological Data and Clinical Evidence. Oxidative Med. Cell. Longev. 2021, 2021, 6678662. [Google Scholar] [CrossRef] [PubMed]
  374. Michala, A.S.; Pritsa, A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022, 10, 37. [Google Scholar] [CrossRef]
  375. Cheng, X.; Yang, Y.L.; Yang, H.; Wang, Y.H.; Du, G.H. Kaempferol alleviates LPS-induced neuroinflammation and BBB dysfunction in mice via inhibiting HMGB1 release and down-regulating TLR4/MyD88 pathway. Int. Immunopharmacol. 2018, 56, 29–35. [Google Scholar] [CrossRef]
  376. Yang, Y.L.; Cheng, X.; Li, W.H.; Liu, M.; Wang, Y.H.; Du, G.H. Kaempferol Attenuates LPS-Induced Striatum Injury in Mice Involving Anti-Neuroinflammation, Maintaining BBB Integrity, and Down-Regulating the HMGB1/TLR4 Pathway. Int. J. Mol. Sci. 2019, 20, 491. [Google Scholar] [CrossRef]
  377. Silva Dos Santos, J.; Gonçalves Cirino, J.P.; de Oliveira Carvalho, P.; Ortega, M.M. The Pharmacological Action of Kaempferol in Central Nervous System Diseases: A Review. Front. Pharmacol. 2021, 11, 565700. [Google Scholar] [CrossRef] [PubMed]
  378. Rifaai, R.A.; Mokhemer, S.A.; Saber, E.A.; El-Aleem, S.A.; El-Tahawy, N.F. Neuroprotective effect of quercetin nanoparticles: A possible prophylactic and therapeutic role in Alzheimer’s disease. J. Chem. Neuroanat. 2020, 107, 101795. [Google Scholar] [CrossRef]
  379. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [PubMed]
  380. Pantsar, T.; Poso, A. Binding Affinity via Docking: Fact and Fiction. Molecules 2018, 23, 1899. [Google Scholar] [CrossRef]
Figure 1. Flowchart depicting the article screening and selection process according to PRISMA guidelines.
Figure 1. Flowchart depicting the article screening and selection process according to PRISMA guidelines.
Biology 12 01453 g001
Figure 5. A graphical summary of the underlying mechanisms behind AD progression (pathogenesis), the proposed mechanisms of kaempferol and quercetin (K/Q), where K/Q represents kaempferol and quercetin, and the impact of these molecular changes on behavior and disease progression (outcomes). Each category is presented in a top-to-bottom chronological order.
Figure 5. A graphical summary of the underlying mechanisms behind AD progression (pathogenesis), the proposed mechanisms of kaempferol and quercetin (K/Q), where K/Q represents kaempferol and quercetin, and the impact of these molecular changes on behavior and disease progression (outcomes). Each category is presented in a top-to-bottom chronological order.
Biology 12 01453 g005
Table 1. Kaempferol and Quercetin and molecular interactions with select molecules relevant to neuroplasticity in AD. These affinity or potency values are deduced from molecular docking studies (affinity) and competition assays (IC50; potency) or were indirect interactions evidenced in the literature. Docking scores (DS) of 5 or higher indicate the high affinity of a compound for the protein of interest [248,249]. Or, affinity from docking studies may be expressed as binding energies (BE) in -kcal/mol. The more negative the value, the higher the binding affinity. If studies have not supported direct binding to a certain target, the affinity column is noted as “Indirect”.
Table 1. Kaempferol and Quercetin and molecular interactions with select molecules relevant to neuroplasticity in AD. These affinity or potency values are deduced from molecular docking studies (affinity) and competition assays (IC50; potency) or were indirect interactions evidenced in the literature. Docking scores (DS) of 5 or higher indicate the high affinity of a compound for the protein of interest [248,249]. Or, affinity from docking studies may be expressed as binding energies (BE) in -kcal/mol. The more negative the value, the higher the binding affinity. If studies have not supported direct binding to a certain target, the affinity column is noted as “Indirect”.
Molecular TargetPhytochemicalMechanismAffinity (DS, BE, or IC50)References
GSK-3βKaempferolInhibit4.6 (DS, mice);
−7.9 kcal/mol (human brain docking)
−9.2 kcal/mol (zebrafish)
[243,250,251]
QuercetinInhibit5.64 (DS);
−8.8 kcal/mol (human brain docking)
−9.0 kcal/mol (zebrafish)
[243,250,251]
KaempferolInhibitIndirect[171]
QuercetinInhibitIndirect[252]
BACE1KaempferolInhibitIC50 = 14.7 µM[253,254]
QuercetinInhibitIC50 = 5.4 µM[253,254]
TauKaempferolInhibit hyperactivationIndirect[47]
QuercetinInhibit hyperactivationIndirect[255]
PI3KKaempferolActivate5.19 (DS, neurons)[256]
QuercetinActivate7.04 (MD, neurons)[256]
AKT1KaempferolActivate5.13 (MD, neurons);
−9.3 kcal/mol
[256,257]
QuercetinActivate5.03 (MD, neurons),
−9.4 kcal/mol;
−7.96 kcal/mol
[213,256,257]
BDNFKaempferolUpregulateIndirect[258]
QuercetinUpregulateIndirect[252]
CREBKaempferolActivateIndirect[211]
QuercetinActivateIndirect[252]
NMDARKaempferolReverse Aβ binding−10.84 kcal/mol[259]
QuercetinReverse Aβ bindingIndirect[255,260]
HDACKaempferolActivateNot Found[188,189]
QuercetinActivateIC50 = 105.1 µM[261]
AChEKaempferolInhibit−10.26 kcal/mol;
between −8.6 and −9.22 kcal/mol
[259,262,263]
QuercetinInhibit−7.9 kcal/mol;
IC50 = 4.59 ± 0.27 µM
[155,263]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alexander, C.; Parsaee, A.; Vasefi, M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer’s Disease. Biology 2023, 12, 1453. https://doi.org/10.3390/biology12111453

AMA Style

Alexander C, Parsaee A, Vasefi M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer’s Disease. Biology. 2023; 12(11):1453. https://doi.org/10.3390/biology12111453

Chicago/Turabian Style

Alexander, Claire, Ali Parsaee, and Maryam Vasefi. 2023. "Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer’s Disease" Biology 12, no. 11: 1453. https://doi.org/10.3390/biology12111453

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