Next Article in Journal
Counteracting Roles of Lipidic Aldehydes and Phenolic Antioxidants on Soy Protein Oxidation Defined by a Chemometric Survey of Solvent and Mechanically Extracted Soybean Meals
Next Article in Special Issue
Natural Acetogenins, Chatenaytrienins-1, -2, -3 and -4, Mitochondrial Potential Uncouplers and Autophagy Inducers—Promising Anticancer Agents
Previous Article in Journal
Use of Winemaking By-Products for the Functionalization of Polylactic Acid for Biomedical Applications
Previous Article in Special Issue
Characterization of 20 Oenological Tannins from Different Botanical Origins for Formulation of Blends with Redox Potential Tuning Ability in Model Wine Solution
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploitation of Quercetin’s Antioxidative Properties in Potential Alternative Therapeutic Options for Neurodegenerative Diseases

by
Viorica Rarinca
1,2,
Mircea Nicusor Nicoara
1,3,*,
Dorel Ureche
4,* and
Alin Ciobica
3,5,6
1
Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
2
Doctoral School of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700506 Iasi, Romania
3
Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
4
Department of Biology, Ecology and Environmental Protection, Faculty of Sciences, University Vasile Alecsandri of Bacau, Calea Marasesti Street, No 157, 600115 Bacau, Romania
5
Center of Biomedical Research, Romanian Academy, No 8, Carol I Avenue, 700506 Iasi, Romania
6
Academy of Romanian Scientists, No 54, Independence Street, Sector 5, 050094 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
Antioxidants 2023, 12(7), 1418; https://doi.org/10.3390/antiox12071418
Submission received: 27 May 2023 / Revised: 5 July 2023 / Accepted: 10 July 2023 / Published: 13 July 2023
(This article belongs to the Collection Advances in Antioxidant Ingredients from Natural Products)

Abstract

:
Oxidative stress (OS) is a condition in which there is an excess of reactive oxygen species (ROS) in the body, which can lead to cell and tissue damage. This occurs when there is an overproduction of ROS or when the body’s antioxidant defense systems are overwhelmed. Quercetin (Que) is part of a group of compounds called flavonoids. It is found in high concentrations in vegetables, fruits, and other foods. Over the past decade, a growing number of studies have highlighted the therapeutic potential of flavonoids to modulate neuronal function and prevent age-related neurodegeneration. Therefore, Que has been shown to have antioxidant, anticancer, and anti-inflammatory properties, both in vitro and in vivo. Due to its antioxidant character, Que alleviates oxidative stress, thus improving cognitive function, reducing the risk of neurodegenerative diseases. On the other hand, Que can also help support the body’s natural antioxidant defense systems, thus being a potentially practical supplement for managing OS. This review focuses on experimental studies supporting the neuroprotective effects of Que in Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and epilepsy.

1. Introduction

Excessive OS is the result of disturbing the balance between oxidation and antioxidant systems, with a tendency to oxidize. OS can cause many reactions, such as protease stimulation, neutrophil infiltration, and the explosion of oxidative intermediates [1]. In addition, OS is thought to play a key role in the progressive degeneration and/or death of nerve cells, especially in neurodegenerative diseases, where it acts as a mediator of the side effects of several neurotoxic substances and as a mechanism of age-related degenerative processes [2]. The ROS scavenger is often used to counteract the effects of OS in neurons [3,4]. Numerous studies have shown that Que, by eliminating oxygen radicals and via metal-chelating operations, attenuates the neuronal damage mediated by OS [5,6].
To survive, aerobic multicellular organisms need molecular oxygen (O2), to the detriment of oxygen, which is susceptible to radical formation due to its electronic structure. Reactive oxygen species are the natural by-products of normal oxygen metabolism and play significant roles in homeostasis and cellular signaling. OS increases in the cellular environment when oxygen homeostasis is not maintained. ROS are oxygen free radicals or small molecules derived from oxygen, such as peroxyl radical (ROO•), hydroxyl radical (•OH), superoxide anion (•O2), and alkoxyl (RO). ROS could also come from non-radicals such as ozone (O3), hypochlorous acid (HOCl), singlet oxygen (1O2), and hydrogen peroxide (H2O2). These non-radicals are oxidizing agents or are easily converted to radicals [7,8].
Neurological diseases are a consequence of genetics, environmental factors, and even age [9]. During periods of environmental stress, such as ultraviolet A (UVA) and ultraviolet B (UVB) radiation, exposure to heat, and ionizing radiation, their levels could increase dramatically. A study by Erden et al. in 2001 [10] showed that exposure to UVA radiation can induce ROS production, leading to damage to cellular elements, but can also induce the benefits that antioxidant Que brings to cells, protecting them from the harmful effects of radiation.
Neurodegeneration is characterized by progressive deterioration of the structure and function of neurons and is accompanied by severe cognitive deficits. Aging is the main risk factor for neurodegenerative disorders such as AD, PD, and HD. Mitochondrial dysfunction and OS also trigger neurodegeneration. Recent studies have supported the mechanisms by which Que supports brain health [11].
The first studies involving flavonoids to reduce OS were performed at the end of the twentieth century [12]. Numerous in vitro and in vivo studies have reported the neuroprotective properties of Que [11,13,14]. Thus, it has been observed to protect neurons from oxidative damage and reduce lipid peroxidation as well (see Figure 1). On the other hand, in addition to its antioxidant properties, Que can inhibit the formation of amyloid-β proteins in the fibrils, counteracting cell lysis and inflammatory cascade [13]. Flavonoids, but also foods containing flavonoids, can have multiple beneficial effects on the treatment of conditions involving OS, such as AD, PD, aging itself, atherosclerosis, and ischemia [12,15,16].
Que, or 2-(3,4-dihydroxy phenyl)-3,5,7- trihydroxychromen-4-one, is commonly found in our diet and is found in abundance as a secondary metabolite in vegetables and fruits. According to the USDA (United States Department of Agriculture) database on flavonoid concentration in foods [18] and Table 1, the highest Que concentrations are in capers, dill weed, oregano, onions, cranberries, cherries, and red fruits; in addition to fruits and vegetables, they are also found in beverages such as red wine and black tea. It has also been isolated and marketed as a dietary supplement, in the form of free aglycone, used in doses of 1000 mg per day, exceeding the usual levels of food intake, which is 200–500 mg per day in individuals who consume large amounts of vegetables and fruits [18,19].
Que is a more potent antioxidant than other antioxidant nutrients, such as vitamin C, vitamin E, and β-carotene [33]. Due to the five hydroxyl groups present in its structure that can bind to ROS, Que has a higher antioxidant potential than many other flavonoids [34,35]. In addition to its antioxidant activity, Que has anti-cancer effects [36,37,38,39,40]; anti-inflammatory [34,41,42,43], antiviral [44,45,46], and antibacterial properties [47,48,49,50,51]; cardioprotective effects [52]; and neuroprotective effects vs. brain ischemia [53,54].
In many studies, Que is also reported to have adverse effects, such as the induction of mutations, chromosomal aberrations, and single-stranded deoxyribonucleic acid (DNA) ruptures in various eukaryotic cell systems in vitro [55].
Que is less toxic than curcumin or gallic acid due to the LD50 value of 484 µg mL−1, while the LD50 value for curcumin is 135 µg mL−1 and that for gallic acid is 304 µg mL−1 [56].
This review focuses on the preventive and therapeutic capacity of Que in neurological and neurodegenerative diseases along with its potential mechanisms of action. Furthermore, we also summarized the biological sources and other pharmacological activities of this antioxidant compound.
Que acts as a protector of neurons against severe OS, but also against free radical attack by easily intercalating its molecules in DNA, thus forming a protective barrier against stronger intercalators and/or ROS attack [57].

2. Methodology

Search Strategy
The current systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines [58], employing several electronic databases (Science Direct, PubMed, and Google Scholar) and using the following keywords: ((quercetin[Title/Abstract]) AND (Alzheimer’s disease[Title/Abstract])) AND (amyloid[Title/Abstract]); (quercetin[Title/Abstract]) AND (Parkinson’s disease[Title/Abstract]); (quercetin[Title/Abstract]) AND (Huntington’s disease[Title/Abstract]); (quercetin[Title/Abstract]) AND (pentylenetetrazol [Title/Abstract]). Inclusion criteria included experimental studies (in vivo and in vitro) until May 2023 in English, which evaluated the antioxidant properties of quercetin in potential alternative therapeutic options for neurodegenerative diseases.
Exclusion Criteria
We applied the following exclusion criteria: (1) conference abstracts, books, book chapters, and unpublished results; (2) non-English papers; (3) reviews, systematic reviews, meta-analyses, and letters.
Data Extraction
Among the initial 868 reports that were collected through electronic search, 554 were omitted due to duplicated results, 47 were ruled out because of the article type, and 190 review articles were omitted and were deemed irrelevant based on abstract and/or title information. Additionally, 1 was excluded because it was not in the English language.
Data Synthesis
Finally, 43 articles were included in this study, as demonstrated in a diagram of the literature search and selection process (see Figure 2). It was thought that the studies would be too heterogeneous to be combined. Therefore, a narrative synthesis was performed. The results are summarized according to the type of neurodegenerative disease (AD, PD, HD, and epilepsy) and the test (in vivo, in vitro).

3. Oxidative Stress

OS is a condition in which there is an imbalance between the production of ROS and the body’s ability to neutralize them, causing damage to lipids, proteins, and DNA. ROS are naturally produced by the body as a by-product of normal metabolism but can also be produced by environmental factors such as exposure to pollution, radiation, and certain chemicals. In the context of neurological disease, OS has been shown to play a role in the development and progression of several conditions.
Recent studies have shown that Que can protect against OS-induced cell death by inhibiting the activity of caspase-3 [59] and increasing the body’s antioxidant capacity by regulating glutathione (GSH) levels [60]. Que has also been shown to reduce inflammation and OS and improve wound healing in animal models of Alzheimer’s disease [43] and reduce inflammatory pain by inhibiting OS pathways [61] (see Figure 2). These findings suggest that Que may be beneficial in reducing OS and inflammation in humans [62].
In AD, OS has been shown to accelerate the formation of amyloid plaques in the brain, which are a hallmark of the disease. Also, in PD, OS can cause the death of dopaminergic neurons, leading to loss of motor function. In HD, stress can increase levels of the glutamate neurotransmitter in the brain, which can lead to neuron death and worsening symptoms. Thus, in Figure 3, we highlight the enzymes that have the role of protecting the body from OS and have neuroprotective effects in the case of neurodegenerative diseases.

3.1. Alzheimer’s Disease

Alzheimer’s disease is the most common neurodegenerative disease, accounting for about two-thirds (60–80%) of all cases of dementia, and it affects mainly the elderly (aged 65 or older) [63]. The pathogenesis of AD is commonly associated with the extracellular accumulation of amyloid-β (1–40, 1–42) aggregates and the hyperphosphorylation of tau proteins, leading to neurofibrillary tangles (NFT) and synaptic dysfunction [13,64,65,66]. An estimated 44 million people worldwide are affected by AD or a related form of dementia, with a prevalence rate of 4.6 million new cases each year. The prevalence rate of AD increases with age: the rate doubles every 5 years from the age of 60 [67,68].
OS plays an important role in AD, which, through ROS generation, can amplify or initiate the disease (Table 2). The reduction reaction of hydrogen peroxide results in the production of reactive oxygen species, thus damaging brain tissue and disrupting brain-cell repair [69,70,71]. Thus, the administration (ad) of Que before the treatment decreases the damage to the cell membrane induced by the OS caused by H2O2 [70].
AD is characterized by neuronal loss, which is preceded by the extracellular accumulation of Aβ1–40, and Aβ1–42. Antioxidants such as Que increase the resistance of neurons to OS by modulating cell-death mechanisms. Thus, Que protects the mouse hippocampal cell line HT-22 from glutamate-induced oxidative toxicity and lipid peroxidation by blocking the production of free radicals [69]. Also, pretreatment of primary hippocampal cultures with Que significantly attenuated Aβ1–42-induced cytotoxicity, protein oxidation (protein carbonyl, 3-nitrotyrosine), lipid peroxidation (protein-bound 4-hydroxy-2-nonenal), and apoptosis. There were also observed protective effects against Aβ1–42 toxicity by modulating OS at lower concentrations (5 and 10 μM), while in the cases of higher concentrations (20 and 40 μM), the effects were not only non-neuroprotective, but toxic [13].
However, recent research shows that the more hydroxyl groups the structure of the molecule contains, the stronger the anti-amyloidogenic activity. Therefore, one of the potential benefits of Que is its ability to act as an anti-amyloidogenic agent due to its five hydroxyl groups (see Figure 1), which means it can prevent the formation of amyloid plaques in the brain [72,73].
Numerous studies demonstrate anticancer and apoptosis-inducing effects in vitro on a variety of cancer cell lines, including murine neuroblastoma HT-22 cells or human-brain microvascular endothelial cells [69,74,75,76]. Que was found to be non-cytotoxic and strongly protected HT-22 cells from fibril formation [76]. In addition, Ishige et al. in 2001 [69], using the HT-22 mouse hippocampal cell line, found three distinct mechanisms of flavonoid protection in cell death, including increased intracellular GSH, a direct decrease in ROS levels, and the prevention of Ca2+ influx.
In a study on the stable cell line of the Swedish mutant of amyloid precursor protein (APP695-transfected SH-SY5Y), no effects of Que were observed in the middle-late stage of AD; instead, effects were observed in the mid-early stage, when the reduction in β-amyloid-converting enzyme 1 (BACE1) activity was recorded [73].
The optimal concentration of Que required for the effective destabilization of Aβ fibrils has been found to be in the range of 0.1–1 μM [74,77]. Thus, in a study using neurons of the rat hippocampal region, optimal doses of Que administration were beneficial for protecting against Aβ25–35-induced amnesic injury by reducing lipid peroxidase, ROS, and GPx [77]. In another study on pheochromocytoma (PC12) cells, Que was found to increase the survival rate of H2O2-damaged cells, decrease lipid peroxidation and GSH level, and provide mitochondrial protection mechanisms [70]. On the other hand, Yu et al. in 2020 [78] showed that Que has beneficial effects, so it can increase the PC12 cell-survival damaged by Aβ25–35, antagonize the toxicity of Aβ, promote cell proliferation, and provide some neuroprotective effects.
Also, in a study involving homozygotic transgenic mouse line B6.129S7-Sod2tm1Leb/J, where Que was administered orally at a dose of 50 mg kg−1 body weight (b.w.) twice a week for four weeks, the results showed that Que had a protective effect against hydrogen peroxide- and paraquat-induced OS in the mice [71]. In addition, Que has the effect of reducing anion superoxide levels that increased with H2O2 and Aβ treatment in hippocampal neurons or human-brain microvascular endothelial cells (hBMECs) [71,75].
Quercetin-3-glucuronide (Q3G), a glucuronide conjugate of Que, has been identified as a potential intervention for AD due to its ability to target the brain. Thus, several studies have shown that Q3G may be able to alleviate neuroinflammation and reduce OS in nerve cells, both of which are associated with AD [79,80]. Additionally, Q3G has been found to specifically localize in human brain tissue, suggesting that it may be able to cross the blood–brain barrier (BBB) and reach areas of the brain affected by AD [79]. The mechanism by which Que can cross the BBB is through passive diffusion; this is a process that allows small molecules to move across the BBB from an area of high concentration to an area of low concentration [81]. Thus, Ho et al. in 2013 [82] observed that Q3G significantly reduced the generation of β-amyloid peptides using cultures of primary neurons generated by the mouse model Tg2576 AD.
A high concentration of Que was found in Ginkgo biloba, thus showing that Ginkgo biloba extract (EGb761) and its constituents, Que and ginkgolide B, have protective effects against the cytotoxic action of Aβ1–42 via intraperitoneal administration (i.p.), thereby ameliorating the oxidative phosphorylation deficits, and mitochondrial dysfunction in AD [83]. Another plant used in pharmacotherapy and in which Que is found is Acanthopanax henryi, which can potentiate cholinergic activity by inhibiting acetylcholinesterase (AchE) [84].
Table 2. Protective effects against OS, neuroinflammation, and Aβ accumulation induced by Que in vitro.
Table 2. Protective effects against OS, neuroinflammation, and Aβ accumulation induced by Que in vitro.
Types of QueConcentration of QueModelExposureEffectsRef.
QueDosage: 2.2 μM;
Duration: 24 h;
HT-22 mouse hippocampal cellH2O2 lipid peroxidation, ↑ intracellular GSH, ROS[69]
Dosage: 10–100 µmol L−1;
Duration: 10 min;
PC12 cellsH2O2↓ lipid peroxidation, ↓ GSH, mitochondrial protection mechanisms[70]
Dosage: 50 mg; kg−1 b.w.;
Duration: 2 times a week for 4 weeks;
homozygotic transgenic mouse line B6.129S7-Sod2tm1Leb/J H2O2 and Aβ↓ ROS levels, improved the typical morphology of mitochondria, prevented mitochondrial dysfunction[71]
Dosage: 10 μM;APP695-transfected SH-SY5Y cells25–35↓ ROS, ↓ BACE, ↓ Aβ, ↓ GSH, ↓ lipid peroxidation [73]
Dosage: 10 and 50 μM;
Duration: 7 days;
hek cells1–42 or Aβ1–40↓ Aβ peptides, ↓ the performed mature fibrils[74]
Dosage: 5 or 10 mg kg−1 b.w.;
Ad: p.o.;
Duration: once daily;
hBMECsfAβ1–40↓ SOD, ↓ LDH[75]
Dosage: 2.4 µg mL−1;HT-22 murine neuroblastoma cells25–35↓ amyloidogenic Aβ peptides,
inhibited Aβ fibril formation.
[76]
Dosage: 10, 20, 40, and 80 μmol L−1;
Duration: 24 h, 48 h, and 72 h;
PC12 cells25–35↑ the survival rate of PC12 injured by Aβ25-35, promoted cell proliferation, and antagonized the toxicity of Aβ, ↓ ROS[78]
Q3GDosage: 25 μmol L−1;Tg2576 AD primary neuron cultures1–40,1–42↑ neuronal survival, ↑ c-Jun N-terminal kinases, ↓ stress-induced impairments[82]
Que/Ginkgo bilobaDosage: 1.5–6 μg mL−1;SHSY5Y human neuroblastoma cells1–42↓ Akt signaling pathways, ↓ Aβ toxicity, ↓ platelet-activating factor[83]
Que/
Acanthopanax henryi
Dosage: 2.5, 5, 10, 20, and 40 μg mL−1;cell-free system ↓ AchE activity, ↑ antioxidant activity [84]
Abbreviations: ↑, increase; ↓, decrease; Aβ, amyloid beta-peptide; AchE, acetylcholinesterase; BACE, β-amyloid-converting enzyme 1; b.w., body weight; CAT, catalase; GPx, superoxide dismutase; GSH, glutathione; hBMECs, human-brain microvascular endothelial cells; hek, human embryonic kidney; LDH, lactate dehydrogenase; OS, oxidative stress; PC12, pheochromocytoma; Q3G, quercetin-3-glucuronide; Que, quercetin; ROS, reactive oxygen species; SOD, superoxide dismutase.
In mitochondria, the first free radical to form is the superoxide radical, which is catalyzed by superoxide dismutase (SOD) and can cause irreversible damage to nucleic acids, proteins, phospholipids, and/or signaling pathways, thus contributing to apoptosis and intoxication [85].
In vivo studies (Table 3) in triple transgenic mice models of AD (3xTg-AD) have shown that Que can disaggregate amyloid fibrils, such as extracellular amyloid β-peptide, tauopathy astrogliosis, and microgliosis in the hippocampus and amygdala, and improves their spatial memory and learning [86,87]. Additionally, the results showed that Que tended to improve active behaviors of 3xTG-AD mice and decreased neurodegeneration markers in mice [87]. Additionally, in the case of APPswe/PS1dE9 transgenic mice, it was observed that long-term Que consumption prevents memory loss, Aβ-induced neurotoxicity, and mitochondrial dysfunctions [88].
Furthermore, Hayakawa et al. in 2015 [89] showed that Que has memory-enhancing effects in older mice and delays the deterioration of memory in the early stages of Alzheimer’s, since it reduces eIF2a and ATF4 expression by inducing GADD34 in the brain. Also, Que can partially block the effect of other genes that play an important role in Alzheimer’s disease, such as tumor necrosis factor-alfa (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6) [90].
On the other hand, oral (p.o.) treatment with 500 mg kg−1 b.w. Que for 10 days can significantly increase brain apoE levels and reduce insoluble Aβ levels in the cortex of 5xFAD amyloid-model mice [91].
Interestingly, this memory impairment was markedly ameliorated by oral treatment with Que nanoencapsulated in zein nanoparticles (25 mg kg−1 every 48 h for 2 months), while the administration of free Que was not able to reverse the faulty behavior, despite a higher administration frequency [92]. Also, pretreatment with Que decreases the effects induced by Aβ1–42 in adult male Sprague Dawley rats [93].
Scopolamine administration causes short-term and long-term memory loss because it blocks muscarinic cholinergic receptors in the brain and interferes with learning and memory [94,95]. There are studies that have found that Que alleviates scopolamine-induced memory deficits by protecting against neuroinflammation and neurodegeneration by inhibiting OS and acetylcholinesterase activity, reverses synaptic loss in the cortex and hippocampus of the brain of adult mice, and suppresses memory impairment [94,95].
Aluminum is a toxic metal that has neurological effects, including Alzheimer’s disease, by generating ROS [96]. Increased production of reactive oxygen species leads to the disruption of cellular antioxidant defense systems and to the release of cytochrome c from the mitochondria into the cytosol, resulting in apoptotic cell death [96,97]. Thus, the administration of 10 mg kg−1 b.w Que reduces the effects induced by aluminum, thus reducing OS. In addition, it prevents cytochrome c translocation [96].
In addition, Hou et al. in 2010 [98] showed that flavonols can antagonize the toxicity of Aβ and improve the expression of brain-derived neurotrophic factor (BDNF) in the hippocampus of double transgenic mice.
Table 3. Protective effects against oxidative stress, neuroinflammation, and Aβ accumulation induced by Que in vivo.
Table 3. Protective effects against oxidative stress, neuroinflammation, and Aβ accumulation induced by Que in vivo.
Types of QueConcentration ModelExposureEffectsRef.
QueDosage: 25 mg kg−1 b.w.;
Ad: i.p.;
Duration: every 2 days for 3 months;
3xTg-AD mice ↓ tauopathy, ↓ β-amyloidosis, ↑ memory, ↑ learning, ↓ microgliosis, ↓ astrogliosis[86]
Dosage: 100 mg kg−1 b.w.;
Ad: gavage;
Duration: every 48 h for 12 months;
3xTg-AD mice ↓ neurodegeneration, ↓ β-amyloidosis[87]
Dosage: 20 and 40 mg kg−1 b.w.;
Ad: p.o.;
Duration: 16 weeks;
adult male C57BL mice ↑ MMP, ↑ ATP levels, ↓ ROS[88]
Dosage: 20 mg;
Ad: p.o.;
Duration: 5 weeks;
APP23 AD mice model ↓ eIF2α, ↓ ATF4, ↓ GADD34, ↑ memory in aged mice, ↓ memory deterioration in the early stage of AD, ↓ memory dysfunction, ↓ OS[89]
Dosage: 1% in mouse chow;
Ad: p.o.;
Duration: from 3 to 13 months;
double transgenic female mice ↓ neuroinflammation,
↓ neurodegeneration, ↓ IL-1β
[90]
Dosage: 25 mg kg−1;
Ad: p.o.;
Duration: 2 times a week for 2 months;
SAMP8 mice ↑ the cognition and memory impairments, ↓ astrogliosis[92]
Dosage: 100 mg kg−1 b.w.;
Ad: p.o.;
Duration: 22 days;
adult male Sprague Dawley rats1–42↑ expression of Nrf2/HO-1 in rat brains, ↓ Aβ1-42 level, ↓ antioxidant activity[93]
Dosage: 12.5 and 25 mg kg−1;miceScopolamine↓ OS, ↓ AchE activity[94]
Dosage: 30 mg kg−1 b.w.;
Ad: i.p.;
Duration: every day for 8 days;
male albino Wistar ratsScopolamine abridged transfer latency, ↓ avoidance response, ↓ 3,4-methylenedioxyamphetamine, acetylcholinesterase levels, ↑ CAT, ↑ GSH levels[95]
Dosage: 10 mg kg −1 b.w.;
Ad: p.o.;
Duration: every day for 12 weeks;
male albino Wistar ratsaluminum↓ ROS production,
↑ mitochondrial superoxide dismutase activity
[96]
Que/ginkgo flavonolsDosage: 4.8% in extract, all based on weight;double transgenic (TgAPP/PS1) mice-reversed the spatial learning deficit[98]
Abbreviations: ↑, increase; ↓, decrease; Aβ, amyloid beta-peptide; AchE, acetylcholinesterase; ATP, adenosine triphosphate; b.w., body weight; CAT, catalase; GPx, glutathione peroxidase; GSH, glutathione; hBMECs, human-brain microvascular endothelial cells; hek, human embryonic kidney; IL-1β, interleukin 1 beta; i.p., intraperitoneal; LDH, lactate dehydrogenase; MMP, matrix metalloproteinases; OS, oxidative stress; PC12, pheochromocytoma; Q3G, quercetin-3-glucuronide; Que, quercetin; ROS, reactive oxygen species; SOD, superoxide dismutase.

3.2. Parkinson’s Disease

Parkinson’s disease is the second most common neurodegenerative disorder worldwide, affecting 1% of the global population aged 65 years and older; it has significant morbidity and mortality [99]. An increasing percentage of research indicates the association of PD with microglial activation, resulting in an increase in various inflammatory mediators and neuroinflammation [100,101]. 1-Methyl-4-phenylpyridinium (MPP+) is the ultimate toxic agent formed by the metabolism of MPTP and can activate glial cells to induce neuroinflammation [102]. Research has shown that MPP+ induces microglial activation and the degeneration of dopaminergic neurons (Table 4), as well as the generation of ROS in dopaminergic neurons [103]. On the other hand, Que administration protects microglia cells against MPP+-induced increases in the mRNA and protein levels of IL-1, IL-6, and TNF-α, due to its antioxidant action [102]. In addition to the loss of dopaminergic neurons in the substantia nigra pars compacta, PD is also characterized by the abnormal accumulation and aggregation of α-synuclein (α-Syn) in the form of Lewy bodies [104,105]. Thus, the formation of α-Syn fibrils can be inhibited by Que and oxidized by Que through their 1:1 covalent binding [105].
Another neurotoxic synthetic organic compound used by researchers to selectively destroy dopaminergic and noradrenergic neurons is 6-hydroxydopamine (6-OHDA) [106,107]. It is a hydroxylated analogue of dopamine and is a benzenetriol with hydroxyl groups on the phenyl ring at positions 2, 4, and 5. Isoquercetin, a flavonol derived from Que, has also been found to have protective effects against 6-OHDA-induced oxidative damage in a rat model of PD. They observed that antioxidant enzymes, catalase (CAT), SOD, GPX, and GSH levels, which were previously attenuated by 6-OHDA, increased significantly [106,107].
In vitro studies have shown that Que can improve mitochondrial quality control, reduce OS, and increase levels of antioxidant enzymes (Table 5) [102,105,106,107]. Instead, in vivo studies in mice and 6-OHDA-induced PD rat models demonstrated that Que can improve locomotor and muscle activity, increase striatal dopamine levels, and protect neurons from mitochondrial dysfunction [108,109,110,111,112].
Studies have shown that Que has neuroprotective effects against MPTP-induced neurotoxicity in Wistar rats and adult male C57BL/6 mice [108,109,110]. Que was found to reduce OS and neuroinflammatory cytokines in rats [108,109], as well as restore motor and non-motor symptoms (depression and cognitive impairment) of PD in rats injected with rotenone [113,114]. Additionally, Que supplementation was found to improve striatal cholinergic function and reduce rotenone-induced OS in rats [114].
On the other hand, the administration of fish oil can attenuate rotenone-induced oxidative impairments and mitochondrial dysfunctions in rat brains [115]. Combined oral supplementation with fish oil and Que has been found to enhance neuroprotection in a chronic rotenone rat model, suggesting potential relevance for PD [115].

3.3. Huntington’s Disease

Along with AD and PD, HD is a major health problem worldwide, with a major financial impact [116]. HD is an autosomal dominant inherited disorder, the treatment of which is clinically available but provides only symptomatic relief. These drugs are available by prescription and have side effects such as anxiety and depression.
For an experimental model of HD, 3-nitropropionic acid (3-NPA) was administered, which altered the mitochondrial metabolism, decreased cellular ATP level, and included nerve-cell death by increasing OS.
In a study by Sandhir and Mehrotra [117] in which female Wistar rats were used as model organisms and in which Que was orally administered at a dose of 25 mg kg−1 for 21 days, for 17 of these 21 days concomitantly with 3-NPA, an attenuation of motor deficits was observed which were assessed using the narrow-beam walking test and fingerprint analysis. Furthermore, molecular changes induced by 3-NPA acid were observed, which were reversed, thus increasing the level of OS and lowering the ATP concentration [117].
On the other hand, a study by Chakraborty [118] failed to confirm the beneficial effect of Que on the 3-NP-induced striatal neuronal lesion (Table 6). However, the conditions of the two studies varied little. Chakraborty used male rats as model organisms, the duration of administration of 3-NP and Que was 4 days, and the concentration was higher (25–50 mg kg−1) than that which was administered by Sandhir and Mehrotra [117], where the dose was administered subchronically (25 mg kg−1). Although Que had no effects on 3-NP-induced striatal neuronal injury, it significantly attenuated neurotoxin-induced anxiety, decreased microglial proliferation, and increased the number of astrocytes in the lesion core [118].
In addition, Que in combination with other antioxidants, such as lycopene, decreases anxiety and depression [119]. Furthermore, the use of dietary antioxidants as adjuvants with n-3 fatty acids is increasingly being used, as they offer a higher degree of protection. Thus, the efficacy of Que in combination with fish oil was observed in a rat model previously treated with 3-NPA, where it decreased OS and improved motor function [120].
Quinolinic acid (QA) is also a paradigm of HD, and co-administration of antioxidants such as Que with sesamol minimizes neurochemical, behavioral, and biochemical alterations in rat brains [121]. However, these data appear inconsistent and unequivocal conclusions cannot be drawn.

3.4. Epilepsy

Epilepsy is a neurological disorder characterized by recurrent spontaneous seizures (Table 7), being caused by an imbalance in excitatory and inhibitory neurotransmission [122]. Glutamate and γ-amino butyric acid (GABA) are the major excitatory and inhibitory neurotransmitters in the CNS [123]. A GABA receptor antagonist is pentylenetetrazol (PTZ), which is used to create a chemically induced seizure model in animals [123]. The frequency and severity of these recurrent seizures can vary. Thus, a low dose of Que (25 mg kg −1) can reduce the number and duration of spike-wave discharges in WAG/Rij rats [124]. In addition, a reduction in the levels of TNF-alpha, IL-6, and NO was observed compared with the control group.
In a study using PTZ-induced seizure model rats, Que administration at 10 mg kg−1 intraperitoneally 30 min before PTZ injection significantly prolonged the onset and reduced the severity of the seizure, but, at an increased concentration of 40 mg kg−1, Que failed to prevent the effects of PTZ [123]. Also, Nassiri-Asl et al. [125] showed that the administration of 35 mg kg−1 PTZ after 50 mg kg−1 Que reduces seizure severity during kindling and improves performance in a passive avoidance task in kindled rats. Also, supplementation of levetiracetam with quercetin improved depression that is associated with epilepsy, led to decreased immobility time and reduced seizure severity. [126]. In addition, Choudhary et al. in 2011 [127] isolated and evaluated the antiepileptic potential of both the acute and chronic flavonoid fractions of Anisomeles malabarica leaves. Toxic effects were observed In the acute treatment (25 and 50 mg kg−1, i.p.) while for the chronic treatment for one week (6.25 and 12.5 mg kg−1, i.p.) a significant antiepileptic effect was observed, without causing neurotoxic side effects [127].

4. Conclusions

Research on Que’s neuroprotective effects suggests that it can be used to protect against various neurodegenerative diseases. Que has been shown to reduce OS and inflammation, which are both associated with neurodegeneration. It has also been found to reduce hippocampal tau phosphorylation, which is a marker of AD.
In addition, Que has been found to act through numerous mechanistic targets to provide neuroprotection, including the modulation of receptor pathways. It has also been found to have protective effects when combined with vitamin C, lycopene, fish oil, or sesamol.
Overall, research suggests that Que may be an effective agent for the prevention of progressive age-related neurodegenerative diseases such as AD, PD, HD, and epilepsy, respectively. However, more research is needed to draw more concrete conclusions about the efficacy of Que in these disorders.

Author Contributions

Conceptualization, methodology, analysis, writing—original draft preparation, V.R. and A.C.; writing—review and editing, supervision, validation, M.N.N. and D.U. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are thankful to the Romanian Ministry of Research, Innovation and Digitization, within Program 1—Development of the national RD system, Subprogram 1.2—Institutional Performance—RDI excellence funding projects, Contract no. 11PFE/30.12.2021, for financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and analyzed during the current study are available from the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhang, Y.M.; Zhang, Z.Y.; Wang, R.X. Protective Mechanisms of Quercetin Against Myocardial Ischemia Reperfusion Injury. Front. Physiol. 2020, 11, 956. [Google Scholar] [CrossRef] [PubMed]
  2. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shukla, V.; Mishra, S.K.; Pant, H.C. Oxidative Stress in Neurodegeneration. Adv. Pharmacol. Sci. 2011, 2011, 572634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gkekas, I.; Gioran, A.; Boziki, M.K.; Grigoriadis, N.; Chondrogianni, N.; Petrakis, S. Oxidative Stress and Neurodegeneration: Interconnected Processes in PolyQ Diseases. Antioxidants 2021, 10, 1450. [Google Scholar] [CrossRef]
  5. Bandiwadekar, A.; Jose, J.; Khayatkashani, M.; Habtemariam, S.; Kashani, H.R.K.; Nabavi, S.M. Emerging Novel Approaches for the Enhanced Delivery of Natural Products for the Management of Neurodegenerative Diseases. J. Mol. Neurosci. 2021, 72, 653–676. [Google Scholar] [CrossRef] [PubMed]
  6. Chapman, J.M.; Muday, G.K. Flavonols modulate lateral root emergence by scavenging reactive oxygen species in Arabidopsis thaliana. J. Biol. Chem. 2021, 296, 100222. [Google Scholar] [CrossRef]
  7. Ozcan, A.; Ogun, M. Biochemistry of Reactive Oxygen and Nitrogen Species. In Basic Principles and Clinical Significance of Oxidative Stress; Intechopen: London, UK, 2015. [Google Scholar]
  8. Klebanoff, S.J. Oxygen Metabolism and the Toxic Properties of Phagocytes. Ann. Intern. Med. 1980, 93, 391–398. [Google Scholar] [CrossRef]
  9. Sheikh, S.; Safia, U.; Haque, E.; Mir, S.S. Neurodegenerative Diseases: Multifactorial Conformational Diseases and Their Therapeutic Interventions. J. Neurodegener. Dis. 2013, 2013, 1–8. [Google Scholar] [CrossRef] [Green Version]
  10. Erden Inal, M.; Kahraman, A.; Köken, T. Beneficial effects of quercetin on oxidative stress induced by ultraviolet A. Clin. Exp. Dermatol. 2001, 26, 536–539. [Google Scholar] [CrossRef]
  11. Elumalai, P.; Lakshmi, S. Role of Quercetin Benefits in Neurodegeneration. Benefits Nat. Prod. Neurodegener. Dis. 2016, 12, 229–245. [Google Scholar] [CrossRef]
  12. Halliwell, B. Oxidants and human disease: Some new concepts 1. FASEB J. 1987, 1, 358–364. [Google Scholar] [CrossRef]
  13. Ansari, M.A.; Abdul, H.M.; Joshi, G.; Opii, W.O.; Butterfield, D.A. Protective effect of quercetin in primary neurons against Aβ(1–42): Relevance to Alzheimer’s disease. J. Nutr. Biochem. 2009, 20, 269–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Islam, 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. Front. Pharmacol. 2021, 12, 665031. [Google Scholar] [CrossRef] [PubMed]
  15. Ames, B.N.; Shigenaga, M.K.; Hagen, T.M. Oxidants, antioxidants, and the degenerative diseases of aging. Proc. Natl. Acad. Sci. USA 1993, 90, 7915–7922. [Google Scholar] [CrossRef] [PubMed]
  16. Zalpoor, H.; Nabi-Afjadi, M.; Forghaniesfidvajani, R.; Tavakol, C.; Farahighasreaboonasr, F.; Pakizeh, F.; Dana, V.G.; Seif, F. Quercetin as a JAK–STAT inhibitor: A potential role in solid tumors and neurodegenerative diseases. Cell. Mol. Biol. Lett. 2022, 27, 60. [Google Scholar] [CrossRef]
  17. 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, 1991. [Google Scholar] [CrossRef]
  18. Haytowitz, D.; Wu, X.; Bhagwat, S. USDA Database for the Flavonoid Content of Selected Foods, Release 3.3; USDA Agricultureal Research Service: Washington, DC, USA, 2018.
  19. Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm, G.W.; Williams, G.M.; Lines, T.C. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem. Toxicol. 2007, 45, 2179–2205. [Google Scholar] [CrossRef]
  20. Hoffmann-Ribani, R.; Huber, L.S.; Rodriguez-Amaya, D.B. Flavonols in fresh and processed Brazilian fruits. J. Food Compos. Anal. 2009, 22, 263–268. [Google Scholar] [CrossRef]
  21. Tsao, R.; Yang, R.; Young, J.C.; Zhu, H. Polyphenolic Profiles in Eight Apple Cultivars Using High-Performance Liquid Chromatography (HPLC). J. Agric. Food Chem. 2003, 51, 6347–6353. [Google Scholar] [CrossRef]
  22. Sakakibara, H.; Honda, Y.; Nakagawa, S.; Ashida, H.; Kanazawa, K. Simultaneous Determination of All Polyphenols in Vegetables, Fruits, and Teas. J. Agric. Food Chem. 2003, 51, 571–581. [Google Scholar] [CrossRef]
  23. Dragovic-Uzelac, V.; Levaj, B.; Mrkic, V.; Bursac, D.; Boras, M. The content of polyphenols and carotenoids in three apricot cultivars depending on stage of maturity and geographical region. Food Chem. 2007, 102, 966–975. [Google Scholar] [CrossRef]
  24. Bilyk, A.; Sapers, G.M. Varietal differences in the quercetin, kaempferol, and myricetin contents of highbush blueberry, cranberry, and thornless blackberry fruits. J. Agric. Food Chem. 1986, 34, 585–588. [Google Scholar] [CrossRef]
  25. Kirakosyan, A.; Seymour, E.M.; Urcuyo Llanes, D.E.; Kaufman, P.B.; Bolling, S.F. Chemical profile and antioxidant capacities of tart cherry products. Food Chem. 2009, 115, 20–25. [Google Scholar] [CrossRef]
  26. Justesen, U.; Knuthsen, P.; Leth, T. Quantitative analysis of flavonols, flavones, and flavanones in fruits, vegetables and beverages by high-performance liquid chromatography with photo-diode array and mass spectrometric detection. J. Chromatogr. A 1998, 799, 101–110. [Google Scholar] [CrossRef]
  27. Giuffrida, D.; Salvo, F.; Ziino, M.; Toscano, G.; Dugo, G. Initial Investigation on Some Chemical Constituents of Capers (Capparis Spinosa L.) from the Island of Salina. Ital. J. Food Sci. 2002, 14, 25. [Google Scholar]
  28. Lugast, A.; Hóvári, J. Flavonoid aglycons in foods of plant origin I. vegetables. Acta Aliment. 2000, 29, 345–352. [Google Scholar] [CrossRef]
  29. Lin, L.-Z.; Mukhopadhyay, S.; Robbins, R.J.; Harnly, J.M. Identification and quantification of flavonoids of Mexican oregano (Lippia graveolens) by LC-DAD-ESI/MS analysis. J. Food Compos. Anal. 2007, 20, 361–369. [Google Scholar] [CrossRef] [Green Version]
  30. Arabbi, P.R.; Genovese, M.I.; Lajolo, F.M. Flavonoids in Vegetable Foods Commonly Consumed in Brazil and Estimated Ingestion by the Brazilian Population. J. Agric. Food Chem. 2004, 52, 1124–1131. [Google Scholar] [CrossRef]
  31. Innocenti, M.; Gallori, S.; Giaccherini, C.; Ieri, F.; Vincieri, F.F.; Mulinacci, N. Evaluation of the Phenolic Content in the Aerial Parts of Different Varieties of Cichorium intybus L. J. Agric. Food Chem. 2005, 53, 6497–6502. [Google Scholar] [CrossRef]
  32. Price, K.R.; Rhodes, M.J.C.; Barnes, K.A. Flavonol Glycoside Content and Composition of Tea Infusions Made from Commercially Available Teas and Tea Products. J. Agric. Food Chem. 1998, 46, 2517–2522. [Google Scholar] [CrossRef]
  33. Mishra, D.; Flora, S.J.S. Quercetin Administration During Chelation Therapy Protects Arsenic-Induced Oxidative Stress in Mice. Biol. Trace Element Res. 2008, 122, 137–147. [Google Scholar] [CrossRef] [PubMed]
  34. 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]
  35. Dhaouadi, Z.; Nsangou, M.; Garrab, N.; Anouar, E.; Marakchi, K.; Lahmar, S. DFT study of the reaction of quercetin with O2 and OH radicals. J. Mol. Struct. THEOCHEM 2009, 904, 35–42. [Google Scholar] [CrossRef]
  36. Reyes-Farias, M.; Carrasco-Pozo, C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. Int. J. Mol. Sci. 2019, 20, 3177. [Google Scholar] [CrossRef] [Green Version]
  37. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Fallahi, F.; Taghavipour, M.; Ghasemi, Y.; Akbari, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; et al. Quercetin and cancer: New insights into its therapeutic effects on ovarian cancer cells. Cell Biosci. 2020, 10, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Shafabakhsh, R.; Asemi, Z. Quercetin: A natural compound for ovarian cancer treatment. J. Ovarian Res. 2019, 12, 55. [Google Scholar] [CrossRef] [Green Version]
  39. Tang, S.-M.; Deng, X.-T.; Zhou, J.; Li, Q.-P.; Ge, X.-X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed. Pharmacother. 2020, 121, 109604. [Google Scholar] [CrossRef] [PubMed]
  40. Ezzati, M.; Yousefi, B.; Velaei, K.; Safa, A. A review on anti-cancer properties of Quercetin in breast cancer. Life Sci. 2020, 248, 117463. [Google Scholar] [CrossRef]
  41. Hou, D.-D.; Zhang, W.; Gao, Y.-L.; Sun, Y.-Z.; Wang, H.-X.; Qi, R.-Q.; Chen, H.-D.; Gao, X.-H. Anti-inflammatory effects of quercetin in a mouse model of MC903-induced atopic dermatitis. Int. Immunopharmacol. 2019, 74, 105676. [Google Scholar] [CrossRef]
  42. Kawabata, K.; Baba, N.; Sakano, T.; Hamano, Y.; Taira, S.; Tamura, A.; Baba, S.; Natsume, M.; Ishii, T.; Murakami, S.; et al. Functional properties of anti-inflammatory substances from quercetin-treated Bifidobacterium adolescentis. Biosci. Biotechnol. Biochem. 2018, 82, 689–697. [Google Scholar] [CrossRef] [Green Version]
  43. Saeedi-Boroujeni, A.; Mahmoudian-Sani, M.-R. Anti-inflammatory potential of Quercetin in COVID-19 treatment. J. Inflamm. 2021, 18, 1–9. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, M.; Yu, Q.; Xiao, H.; Li, M.; Huang, Y.; Zhang, Q.; Li, P. The Inhibitory Activities and Antiviral Mechanism of Medicinal Plant Ingredient Quercetin Against Grouper Iridovirus Infection. Front. Microbiol. 2020, 11, 586331. [Google Scholar] [CrossRef] [PubMed]
  45. Di Petrillo, A.; Orrù, G.; Fais, A.; Fantini, M.C. Quercetin and its derivates as antiviral potentials: A comprehensive review. Phytotherapy Res. 2022, 36, 266–278. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, C.H.; Kim, J.E.; Song, Y.J. Antiviral Activities of Quercetin and Isoquercitrin Against Human Herpesviruses. Molecules 2020, 25, 2379. [Google Scholar] [CrossRef]
  47. Wang, S.; Yao, J.; Zhou, B.; Yang, J.; Chaudry, M.T.; Wang, M.; Xiao, F.; Li, Y.; Yin, W. Bacteriostatic Effect of Quercetin as an Antibiotic Alternative In Vivo and Its Antibacterial Mechanism In Vitro. J. Food Prot. 2018, 81, 69–78. [Google Scholar] [CrossRef] [PubMed]
  48. Olewnik-Kruszkowska, E.; Gierszewska, M.; Richert, A.; Grabska-Zielińska, S.; Rudawska, A.; Bouaziz, M. Antibacterial Films Based on Polylactide with the Addition of Quercetin and Poly(Ethylene Glycol). Materials 2021, 14, 1643. [Google Scholar] [CrossRef]
  49. Júnior, S.D.D.C.; Santos, J.V.D.O.; Campos, L.A.D.A.; Pereira, M.A.; Santos-Magalhaes, N.; Cavalcanti, I.M.F. Antibacterial and antibiofilm activities of quercetin against clinical isolates of Staphyloccocus aureus and Staphylococcus saprophyticus with resistance profile. Int. J. Environ. Agric. Biotechnol. 2018, 3, 1948–1958. [Google Scholar] [CrossRef] [Green Version]
  50. Jaisinghani, R.N. Antibacterial properties of quercetin. Microbiol. Res. 2017, 8, 1. [Google Scholar] [CrossRef] [Green Version]
  51. Aljadaan, S.A.N.; Elias, R.S.; Al-Anssari, R.A. Investigation of the Antioxidant and Antibacterial Activity of Novel Quercetin Derivatives. Biointerface Res. Appl. Chem. 2020, 10, 7329–7336. [Google Scholar] [CrossRef]
  52. Patel, R.V.; Mistry, B.M.; Shinde, S.K.; Syed, R.; Singh, V.; Shin, H.-S. Therapeutic potential of quercetin as a cardiovascular agent. Eur. J. Med. Chem. 2018, 115, 889–904. [Google Scholar] [CrossRef]
  53. Zhang, L.; Ma, J.; Yang, F.; Li, S.; Ma, W.; Chang, X.; Yang, L. Neuroprotective Effects of Quercetin on Ischemic Stroke: A Literature Review. Front. Pharmacol. 2022, 13, 854249. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, R.; Shen, Y.J.; Chen, M.; Zhao, J.Y.; Chen, S.H.; Zhang, W.; Song, J.K.; Li, L.; Du, G.H. Quercetin attenuates ischemia reperfusion injury by protecting the blood-brain barrier through Sirt1 in MCAO rats. J. Asian Nat. Prod. Res. 2022, 24, 278–289. [Google Scholar] [CrossRef]
  55. Andres, S.; Pevny, S.; Ziegenhagen, R.; Bakhiya, N.; Schäfer, B.; Hirsch-Ernst, K.I.; Lampen, A. Safety Aspects of the Use of Quercetin as a Dietary Supplement. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef]
  56. Harishkumar, R.; Reddy, L.P.K.; Karadkar, S.H.; Al Murad, M.; Karthik, S.S.; Manigandan, S.; Selvaraj, C.I.; Christopher, J.G. Toxicity and Selective Biochemical Assessment of Quercetin, Gallic Acid, and Curcumin in Zebrafish. Biol. Pharm. Bull. 2019, 42, 1969–1976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. 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] [PubMed] [Green Version]
  58. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  59. Kook, D.; Wolf, A.H.; Yu, A.L.; Neubauer, A.S.; Priglinger, S.G.; Kampik, A.; Welge-Lu¨ssen, U.C. The Protective Effect of Quercetin against Oxidative Stress in the Human RPE In Vitro. Investig. Opthalmology Vis. Sci. 2008, 49, 1712–1720. [Google Scholar] [CrossRef] [Green Version]
  60. 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] [Green Version]
  61. Valério, D.A.; Georgetti, S.R.; Magro, D.A.; Casagrande, R.; Cunha, T.M.; Vicentini, F.T.M.C.; Vieira, S.M.; Fonseca, M.J.V.; Ferreira, S.H.; Cunha, F.Q.; et al. Quercetin Reduces Inflammatory Pain: Inhibition of Oxidative Stress and Cytokine Production. J. Nat. Prod. 2009, 72, 1975–1979. [Google Scholar] [CrossRef]
  62. Grewal, A.K.; Singh, T.G.; Sharma, D.; Sharma, V.; Singh, M.; Rahman, 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]
  63. De Ture, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kommaddi, R.P.; Das, D.; Karunakaran, S.; Nanguneri, S.; Bapat, D.; Ray, A.; Shaw, E.; Bennett, D.A.; Nair, D.; Ravindranath, V. Aβ mediates F-actin disassembly in dendritic spines leading to cognitive deficits in Alzheimer’s disease. J. Neurosci. 2018, 38, 1085–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Parent, M.J.; Zimmer, E.R.; Shin, M.; Kang, M.S.; Fonov, V.S.; Mathieu, A.; Aliaga, A.; Kostikov, A.; Do Carmo, S.; Dea, D.; et al. Multimodal Imaging in Rat Model Recapitulates Alzheimer’s Disease Biomarkers Abnormalities. J. Neurosci. 2017, 37, 12263–12271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wallace, R.A.; Dalton, A.J. What can we learn from study of Alzheimer’s disease in patients with Down syndrome for early-onset Alzheimer’s disease in the general population? Alzheimer’s Res. Ther. 2011, 3, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hollingworth, P.; Harold, D.; Jones, L.; Owen, M.J.; Williams, J. Alzheimer’s disease genetics: Current knowledge and future challenges. Int. J. Geriatr. Psychiatry 2011, 26, 793–802. [Google Scholar] [CrossRef]
  68. Mayeux, R.; Stern, Y. Epidemiology of Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006239. [Google Scholar] [CrossRef] [Green Version]
  69. 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]
  70. Heo, H.J.; Lee, C.Y. Protective Effects of Quercetin and Vitamin C against Oxidative Stress-Induced Neurodegeneration. J. Agric. Food Chem. 2004, 52, 7514–7517. [Google Scholar] [CrossRef]
  71. Godoy, J.A.; Lindsay, C.B.; Quintanilla, R.A.; Carvajal, F.J.; Cerpa, W.; Inestrosa, N.C. Quercetin Exerts Differential Neuroprotective Effects Against H2O2 and Aβ Aggregates in Hippocampal Neurons: The Role of Mitochondria. Mol. Neurobiol. 2017, 54, 7116–7128. [Google Scholar] [CrossRef]
  72. 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]
  73. 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] [PubMed]
  74. 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] [PubMed]
  75. Li, Y.; Zhou, S.; Li, J.; Sun, Y.; Hasimu, H.; Liu, R.; Zhang, T. Quercetin protects human brain microvascular endothelial cells from fibrillar β-amyloid1–40-induced toxicity. Acta Pharm. Sin. B 2015, 5, 47–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kim, H.; Park, B.-S.; Lee, K.-G.; Choi, C.Y.; Jang, S.S.; Kim, Y.-H.; Lee, S.-E. Effects of Naturally Occurring Compounds on Fibril Formation and Oxidative Stress of β-Amyloid. J. Agric. Food Chem. 2005, 53, 8537–8541. [Google Scholar] [CrossRef]
  77. Rattanajarasroj, S.; Unchern, S. Comparable Attenuation of Aβ25–35-Induced Neurotoxicity by Quercitrin and 17β-Estradiol in Cultured Rat Hippocampal Neurons. Neurochem. Res. 2010, 35, 1196–1205. [Google Scholar] [CrossRef]
  78. 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]
  79. Ishisaka, A.; Mukai, R.; Terao, J.; Shibata, N.; Kawai, Y. Specific localization of quercetin-3-O-glucuronide in human brain. Arch. Biochem. Biophys. 2014, 557, 11–17. [Google Scholar] [CrossRef]
  80. Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective Effects of Quercetin in Alzheimer’s Disease. Biomolecules 2020, 10, 59. [Google Scholar] [CrossRef] [Green Version]
  81. Ren, S.-C.; Suo, Q.-F.; Du, W.-T.; Pan, H.; Yang, M.-M.; Wang, R.-H.; Liu, J. Quercetin permeability across blood-brain barrier and its effect on the viability of U251 cells. Sichuan da xue xue bao. Yi xue ban = J. Sichuan Univ. Med. Sci. Ed. 2010, 41, 751–754. [Google Scholar]
  82. Ho, L.; Ferruzzi, M.G.; Janle, E.M.; Wang, J.; Gong, B.; Lobo, J.; Cooper, B.; Wu, Q.L.; Talcott, S.T.; Percival, S.S.; et al. Identification of brain-targeted bioactive dietary quercetin-3-O-glucuronide as a novel intervention for Alzheimer’s disease. FASEB J. 2013, 27, 769–781. [Google Scholar] [CrossRef] [Green Version]
  83. Shi, C.; Zhao, L.; Zhu, B.; Li, Q.; Yew, D.T.; Yao, Z.; Xu, J. Protective effects of Ginkgo biloba extract (EGb761) and its constituents quercetin and ginkgolide B against β-amyloid peptide-induced toxicity in SH-SY5Y cells. Chem. Interact. 2009, 181, 115–123. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, X.D.; Liu, X.Q.; Kim, Y.H.; Whang, W.K. Chemical constituents and their acetyl cholinesterase inhibitory and antioxidant activities from leaves of Acanthopanax henryi: Potential complementary source against Alzheimer’s disease. Arch. Pharmacal Res. 2014, 37, 606–616. [Google Scholar] [CrossRef]
  85. Miriyala, S.; Holley, A.K.; Clair, D.K.S. Mitochondrial superoxide dismutase—Signals of distinction. Anti-Cancer Agents Med. Chem. 2012, 11, 181–190. [Google Scholar] [CrossRef] [Green Version]
  86. 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] [PubMed] [Green Version]
  87. Paula, P.-C.; Maria, S.-G.A.; Luis, C.-H.; Patricia, C.-G.G. Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 2019, 24, 2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. 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]
  89. Hayakawa, M.; Itoh, M.; Ohta, K.; Li, S.; Ueda, M.; Wang, M.-X.; Nishida, E.; Islam, S.; Suzuki, C.; Ohzawa, K.; et al. Quercetin reduces eIF2α phosphorylation by GADD34 induction. Neurobiol. Aging 2015, 36, 2509–2518. [Google Scholar] [CrossRef]
  90. Jung, S.H.; Murphy, E.A.; McClellan, J.L.; Carmichael, M.D.; Davis, J.M. The dietary flavonoid quercetin decreases neuroinflammation in a mouse model of Alzheimer’s disease. FASEB J. 2010, 24, 604–617. [Google Scholar] [CrossRef]
  91. Zhang, X.; Hu, J.; Zhong, L.; Wang, N.; Yang, L.; Liu, C.-C.; Li, H.; Wang, X.; Zhou, Y.; Zhang, Y.; et al. Quercetin stabilizes apolipoprotein E and reduces brain Aβ levels in amyloid model mice. Neuropharmacology 2016, 108, 179–192. [Google Scholar] [CrossRef]
  92. Moreno, L.C.G.E.I.; Puerta, E.; Suárez-Santiago, J.E.; Santos-Magalhães, N.S.; Ramirez, M.J.; Irache, J.M. Effect of the oral administration of nanoencapsulated quercetin on a mouse model of Alzheimer’s disease. Int. J. Pharm. 2017, 14, 927–937. [Google Scholar] [CrossRef]
  93. Li, Y.; Tian, Q.; Li, Z.; Dang, M.; Lin, Y.; Hou, X. Activation of Nrf2 signaling by sitagliptin and quercetin combination against β-amyloid induced Alzheimer’s disease in rats. Drug Dev. Res. 2019, 80, 837–845. [Google Scholar] [CrossRef] [PubMed]
  94. Olayinka, J.N.; Eduviere, A.; Adeoluwa, O.; Akinluyi, E.; Obisesan, A.; Akawa, O.; Adebanjo, A. Quercetin mitigates scopolamine-induced memory dysfunction: Impact on oxidative stress and cholinergic mechanisms. Metab. Brain Dis. 2022, 37, 265–277. [Google Scholar] [CrossRef] [PubMed]
  95. Palle, S.; Neerati, P. Quercetin nanoparticles attenuates scopolamine induced spatial memory deficits and pathological damages in rats. Bull. Fac. Pharmacy, Cairo Univ. 2017, 55, 101–106. [Google Scholar] [CrossRef] [Green Version]
  96. Sharma, D.; Wani, W.; Sunkaria, A.; Kandimalla, R.; Sharma, R.; Verma, D.; Bal, A.; Gill, K. Quercetin attenuates neuronal death against aluminum-induced neurodegeneration in the rat hippocampus. Neuroscience 2016, 324, 163–176. [Google Scholar] [CrossRef]
  97. Wani, W.Y.; Gudup, S.; Sunkaria, A.; Bal, A.; Singh, P.P.; Kandimalla, R.J.; Sharma, D.R.; Gill, K.D. Protective efficacy of mitochondrial targeted antioxidant MitoQ against dichlorvos induced oxidative stress and cell death in rat brain. Neuropharmacology 2011, 61, 1193–1201. [Google Scholar] [CrossRef] [PubMed]
  98. 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] [PubMed] [Green Version]
  99. Halli-Tierney, A.D.; Luker, J.; Carroll, D.G. Parkinson Disease. Am. Fam. Physician 2020, 102, 679–691. [Google Scholar] [CrossRef] [PubMed]
  100. Vargas-Restrepo, F.; Guaqueta, A.M.S.; Cardona-Gómez, G.P. Quercetin ameliorates inflammation in CA1 hippocampal region in aged triple transgenic Alzheimer’s disease mice model. Biomedica 2018, 38, 69–76. [Google Scholar] [CrossRef] [PubMed]
  101. 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] [PubMed]
  102. Bournival, J.; Plouffe, M.; Renaud, J.; Provencher, C.; Martinoli, M.-G. Quercetin and Sesamin Protect Dopaminergic Cells from MPP+-Induced Neuroinflammation in a Microglial (N9)-Neuronal (PC12) Coculture System. Oxidative Med. Cell. Longev. 2012, 2012, 1–11. [Google Scholar] [CrossRef] [Green Version]
  103. Bureau, G.; Longpré, F.; Martinoli, M.-G. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J. Neurosci. Res. 2008, 86, 403–410. [Google Scholar] [CrossRef]
  104. Amanzadeh, E.; Esmaeili, A.; Rahgozar, S.; Nourbakhshnia, M. Application of quercetin in neurological disorders: From nutrition to nanomedicine. Rev. Neurosci. 2019, 30, 555–572. [Google Scholar] [CrossRef]
  105. Zhu, M.; Han, S.; Fink, A.L. Oxidized quercetin inhibits α-synuclein fibrillization. Biochim. et Biophys. Acta (BBA)—Gen. Subj. 2013, 1830, 2872–2881. [Google Scholar] [CrossRef] [PubMed]
  106. Magalingam, K.B.; Radhakrishnan, A.; Haleagrahara, N. Protective effects of flavonol isoquercitrin, against 6-hydroxy dopamine (6-OHDA)—Induced toxicity in PC12 cells. BMC Res. Notes 2014, 7, 49. [Google Scholar] [CrossRef] [Green Version]
  107. Magalingam, K.B.; Radhakrishnan, A.; Haleagrahara, N. Protective effects of quercetin glycosides, rutin, and isoquercetrin against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in rat pheochromocytoma (PC-12) cells. Int. J. Immunopathol. Pharmacol. 2016, 29, 30–39. [Google Scholar] [CrossRef] [Green Version]
  108. Pany, S.; Pal, A.; Sahu, P.K. Neuroprotective Effect of Quercetin in Neurotoxicity Induced Rats: Role of Neuroinflammation in Neurodegeneration. Asian J. Pharm. Clin. Res. 2014, 7, 152–156. [Google Scholar]
  109. Kumar, P.; Singh, S.; Jamwal, S. Neuroprotective potential of quercetin in combination with piperine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. Neural Regen. Res. 2017, 12, 1137–1144. [Google Scholar] [CrossRef]
  110. Lv, C.; Hong, T.; Yang, Z.; Zhang, Y.; Wang, L.; Dong, M.; Zhao, J.; Mu, J.; Meng, Y. Effect of Quercetin in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-Induced Mouse Model of Parkinson’s Disease. Evid.-Based Complement. Altern. Med. 2012, 2012, 928643. [Google Scholar] [CrossRef] [Green Version]
  111. 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-Hydroxydopamine. Evid.-Based Complement. Altern. Med. 2011, 2012, 823206. [Google Scholar] [CrossRef] [Green Version]
  112. Mehdizadeh, M.; Joghataei, M.T.; Nobakht, M.; Aryanpour, R. Neuroprotective Effect of Quercetin in a Model of Parkinson’s Disease in Rat: A Histochemical Analysis. Basic Clin. Neurosci. 2009, 1, 3–6. [Google Scholar]
  113. Karuppagounder, S.; Madathil, S.; Pandey, M.; Haobam, R.; Rajamma, U.; Mohanakumar, K. 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] [PubMed]
  114. Madiha, S.; Batool, Z.; Tabassum, S.; Liaquat, L.; Sadir, S.; Shahzad, S.; Naqvi, F.; Saleem, S.; Yousuf, S.; Nawaz, A.; et al. Quercetin exhibits potent antioxidant activity, restores motor and non-motor deficits induced by rotenone toxicity. PLoS ONE 2021, 16, e0258928. [Google Scholar] [CrossRef] [PubMed]
  115. Denny Joseph, K.M.; Muralidhara. Combined Oral Supplementation of Fish Oil and Quercetin Enhances Neuroprotection in a Chronic Rotenone Rat Model: Relevance to Parkinson’s Disease. Neurochem. Res. 2015, 40, 894–905. [Google Scholar] [CrossRef]
  116. Medina, A.; Mahjoub, Y.; Shaver, L.; Pringsheim, T. Prevalence and Incidence of Huntington’s Disease: An Updated Systematic Review and Meta-Analysis. Mov. Disord. 2022, 37, 2327–2335. [Google Scholar] [CrossRef] [PubMed]
  117. Sandhir, R.; Mehrotra, A. Quercetin supplementation is effective in improving mitochondrial dysfunctions induced by 3-nitropropionic acid: Implications in Huntington’s disease. Biochim. et Biophys. Acta (BBA)—Mol. Basis Dis. 2013, 1832, 421–430. [Google Scholar] [CrossRef] [Green Version]
  118. Chakraborty, J.; Singh, R.; Dutta, D.; Naskar, A.; Rajamma, U.; Mohanakumar, K.P. Quercetin Improves Behavioral Deficiencies, Restores Astrocytes and Microglia, and Reduces Serotonin Metabolism in 3-Nitropropionic Acid-Induced Rat Model of Huntington’s Disease. CNS Neurosci. Ther. 2014, 20, 10–19. [Google Scholar] [CrossRef]
  119. Jain, D.; Gangshettiwar, A. Combination of lycopene, quercetin and poloxamer188 alleviates anxiety and depression in 3-nitropropionic acid-induced Huntingtons disease in rats. J. Intercult. Ethnopharmacol. 2014, 3, 186–191. [Google Scholar] [CrossRef]
  120. Joseph, K.D. Muralidhara Enhanced neuroprotective effect of fish oil in combination with quercetin against 3-nitropropionic acid induced oxidative stress in rat brain. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2013, 40, 83–92. [Google Scholar] [CrossRef]
  121. Kuhad, A.; Singla, S.; Arora, V.; Chopra, K. Neuroprotective effect of sesamol and quercetin against QA induced neurotoxicity: An experimental paradigm of Huntington’s disease. J. Neurol. Sci. 2013, 333, e149–e150. [Google Scholar] [CrossRef]
  122. Patel, D.C.; Tewari, B.P.; Chaunsali, L.; Sontheimer, H. Neuron–glia interactions in the pathophysiology of epilepsy. Nat. Rev. Neurosci. 2019, 20, 282–297. [Google Scholar] [CrossRef]
  123. Sefil, F.; Kahraman, I.; Dokuyucu, R.; Gokce, H.; Ozturk, A.; Tutuk, O.; Aydin, M.; Ozkan, U.; Pinar, N. Ameliorating effect of quercetin on acute pentylenetetrazole induced seizures in rats. Int. J. Clin. Exp. Med. 2014, 7, 2471–2477. [Google Scholar] [PubMed]
  124. Kızılaslan, N.; Aydın, D.; Sumbul, O.; Koroglu, R.; Aygun, H. The effect of quercetin on absence epilepsy in WAG/Rij rats. Neurol. Res. 2023, 1–7. [Google Scholar] [CrossRef] [PubMed]
  125. Nassiri-Asl, M.; Moghbelinejad, S.; Abbasi, E.; Yonesi, F.; Haghighi, M.-R.; Lotfizadeh, M.; Bazahang, P. Effects of quercetin on oxidative stress and memory retrieval in kindled rats. Epilepsy Behav. 2013, 28, 151–155. [Google Scholar] [CrossRef] [PubMed]
  126. Singh, T.; Kaur, T.; Goel, R.K. Adjuvant quercetin therapy for combined treatment of epilepsy and comorbid depression. Neurochem. Int. 2017, 104, 27–33. [Google Scholar] [CrossRef]
  127. Choudhary, N.; Bijjem, K.R.V.; Kalia, A.N. Antiepileptic potential of flavonoids fraction from the leaves of Anisomeles malabarica. J. Ethnopharmacol. 2011, 135, 238–242. [Google Scholar] [CrossRef]
Figure 1. Main effects of Que [14,16,17].
Figure 1. Main effects of Que [14,16,17].
Antioxidants 12 01418 g001
Figure 2. The PRISMA flow chart of the selection process for the included studies.
Figure 2. The PRISMA flow chart of the selection process for the included studies.
Antioxidants 12 01418 g002
Figure 3. Diagram with possible neuroprotective effects of quercetin. Abbreviations: ↑, increase; ↓, decrease; Aβ, amyloid beta-peptide; CAT, catalase; GPx, superoxide dismutase; GSH, glutathione; OS, oxidative stress; ROS, reactive oxygen species; SOD, superoxide dismutase [14,16,47].
Figure 3. Diagram with possible neuroprotective effects of quercetin. Abbreviations: ↑, increase; ↓, decrease; Aβ, amyloid beta-peptide; CAT, catalase; GPx, superoxide dismutase; GSH, glutathione; OS, oxidative stress; ROS, reactive oxygen species; SOD, superoxide dismutase [14,16,47].
Antioxidants 12 01418 g003
Table 1. Que content (mg 100 g−1 or mg 100 mL−1) in selected foods and beverages [18].
Table 1. Que content (mg 100 g−1 or mg 100 mL−1) in selected foods and beverages [18].
Source QueReferences
FoodCommon NameScientific NameActive Portionsmg 100 g−1 Weight
FruitsAcerolaMalpighia emarginataFruits4.74[20]
AppleMalus domesticaFruits19.36[21]
Cranberry Vaccinium oxycoccusFruits25.0[22]
ApricotsPrunus armeniacaFruits1.63[23]
BlackberriesRubus spp.Fruits3.58[24]
Blueberries Vaccinium spp.Fruits7.67[24]
Cherries Prunus aviumFruits17.44[25]
CranberriesVaccinium macrocarponFruits14.84[24]
GrapefruitCitrus paradisiFruits0.50[26]
GrapesVitis viniferaFruits3.7[26]
VegetablesCapers, raw Capparis spinosaFlower buds233.84[27]
Onions, raw Allium cepaBulbs34.8[26]
Dill weed, freshAnethum graveolensLeaves74.5[28]
Oregano Origanum vulgareLeaves42.00[29]
Tarragon, fresh Artemisia dracunculusLeaves10.00[26]
ChicoryCichorium intybusLeaves25.2[30]
Beverages mg 100 mL−1
Black tea 2.50[31]
Red wine 3.16[32]
Source: Phenol Explorer and USDA Database for the Flavonoid Content of Selected Foods.
Table 4. Protective effects against oxidative stress and neuroinflammation induced by Que in vitro in the case of Parkinson’s disease.
Table 4. Protective effects against oxidative stress and neuroinflammation induced by Que in vitro in the case of Parkinson’s disease.
Types of QueConcentration ModelExposureEffectsRef.
QueDosage: 0.1 μMMicroglial (N9)-neuronal (PC12) cellsMPP↓ iNOS gene expression, ↓ ROS, ↓ cellular death, ↓ DNA fragmentation, ↓apoptosis, ↓ nuclear translocation of apoptosis-inducing factor, ↓ caspase-3 activation [102]
Dosage: 10 mMPC12 cellsα-Synuclein↓ Aβ fibrillation[105]
IsoquercetinDosage: 10, 50, and 100 μMPC12 cells6-OHDA↓ ROS, ↑ SOD, ↑ GSH, ↑ CAT, ↑ GPx[106]
Quercetin glycosideDosage: 10, 50, and 100 μMPC12 cells6-OHDA↑ antioxidant activity, ↑ GSH, ↑ GPx [107]
Abbreviations: ↑, increase; ↓, decrease; 6-OHDA, 6-hydroxydopamine; Aβ, amyloid beta-peptide; CAT, catalase; DNA, 6-hydroxydopamine; GPx, glutathione peroxidase; GSH, glutathione; iNOS, inducible nitric oxide synthase; MPP, 1-methyl-4-phenylpyridinium; LDH, lactate dehydrogenase; MMP, matrix metalloproteinases; OS, oxidative stress; Q3G, quercetin-3-glucuronide; Que, quercetin; ROS, reactive oxygen species; SOD, superoxide dismutase.
Table 5. Protective effects against oxidative stress and neuroinflammation of Que in vivo in the case of Parkinson’s disease.
Table 5. Protective effects against oxidative stress and neuroinflammation of Que in vivo in the case of Parkinson’s disease.
Types of QueConcentration ModelExposureEffectsRef.
QueDosage: 25 mg kg−1
Ad: p.o.
Wistar ratsHaloperidol MPTP↓ cataleptic score, ↑ actophotometer activity score, ↑ GSH, ↓ lipid peroxidation, ↓ ROS[108]
Dosage: 25 and 50 mg kg−1
Ad: intragastrically
Duration: 14 days
Wistar ratsMPTP↓ TNF-α, ↓ IL-1β and ↓ IL-6, ↓ glutamate level,[109]
Dosage: 50, 100, and 200 mg kg−1
Ad: p.o.
Duration: 14 days
adult male C57BL/6 miceMPTP↓ striatal dopamine depletion, ↓ level of acetylcholine, ↑ AchE activity, ↑ motor deficits, ↑ GPx, ↑ SOD[110]
Dosage: 100, 200, and 300 mg kg −1
Duration: 14 days
Wistar rats6-OHDA↑ spatial memory, ↓ OS, ↓ AchE activity, ↑ antioxidant activity, ↓ neuronal damage[111]
Dosage: 20 mg kg−1
Ad: i.p.
Duration: 1 month
Wistar rats6-OHDA↓ neuroplastic changes in neural circuits, ↓ excitability in neurons involved in epilepsy, ↓ NMDA receptor functionality[112]
Dosage: 25–75 mg kg−1
Duration: 12 h intervals for 4 days
Wistar ratsRotenone↓ nigral GSH depletion, ↓ ROS, ↓ striatal DA loss, ↑ mitochondrial complex, ↓ neuronal death[113]
Dosage: 50 mg kg−1
Ad: p.o.
Duration: 14 days
Wistar ratsRotenone↑ AchE activity, ↑ SOD, ↓ GPx, ↓ CAT[114]
Que + fish oilDosage: 25 mg kg−1
Ad: p.o.
Duration: 28 days
Wistar ratsRotenone↑ mitochondrial functions, ↑ GSH, ↑ antioxidant defenses[115]
Abbreviations: ↑, increase; ↓, decrease; 6-OHDA, 6-hydroxydopamine; AchE, acetylcholinesterase; CAT, catalase; DNA, deoxyribonucleic acid; GPx, glutathione peroxidase; GSH, glutathione; iNOS, inducible nitric oxide synthase; i.p., intraperitoneal; MPP, 1-methyl-4-phenylpyridinium; MMP, matrix metalloproteinases; NMDA, N-methyl-D-aspartate; OS, oxidative stress; p.o., oral; Q3G, quercetin-3-glucuronide; Que, quercetin; ROS, reactive oxygen species; SOD, superoxide dismutase.
Table 6. Protective effects against oxidative stress and neuroinflammation induced by Que in vivo in the case of Huntington’s disease.
Table 6. Protective effects against oxidative stress and neuroinflammation induced by Que in vivo in the case of Huntington’s disease.
Types of QueConcentration ModelExposureEffectsRef.
QueDosage: 25 mg kg−1
Ad: p.o.
Duration: 21 days
Wistar rats3-NPA↑ ATP, ↑ activity of complex II and V enzyme of respiratory chain complex, ↓ ROS, ↑ SOD, ↑ CAT, ↓ lipid peroxidation,[117]
Dosage: 25–50 mg kg−1
Ap: i.p.
Duration: 4 days
Sprague Dawley rats3-NPA↓ gait despair, ↓ microglial proliferation, ↓ anxiety, ↑ astrocyte numbers in the lesion core, ↓ motor coordination deficits, ↓ serotonin metabolism [118]
Que + lycopeneDosage: 50 mg kg−1
Duration: 14 days
Wistar rats3-NPA↓ anxiety, ↓ depression [119]
Que + fish oilDosage: 25 mg kg−1Wistar rats3-NPA↓ OS, ↑ motor function [120]
Que + sesamolDosage: 25, 50, and 100 mg kg−1
Ad: i.p.
Duration: 14 days before and 14 days after QA administration
Wistar ratsQA↓ behavioral, biochemical, and neurochemical alterations in the rat brain, ↑ antioxidant effects, ↑ anti-inflammatory activity [121]
Abbreviations: ↑, increase; ↓, decrease; 3-NPA, 3-nitropropionic acid; CAT, catalase; GSH, glutathione; i.p., intraperitoneal; MPP, 1-methyl-4-phenylpyridinium; OS, oxidative stress; p.o., oral; QA, quinolinic acid; Que, quercetin; ROS, reactive oxygen species; SOD, superoxide dismutase.
Table 7. Protective effects against oxidative stress and neuroinflammation induced by Que in the case of epilepsy.
Table 7. Protective effects against oxidative stress and neuroinflammation induced by Que in the case of epilepsy.
Types of QueConcentrationModelType of TestExposureEffectsRef.
QueDosage: 5, 10, 20, and 40 mg kg−1Albino ratsin vivoPTZ↑ antiseizure effect, ↑ anticonvulsant effect[123]
Dosage: 25, 50, and 100 mg kg−1
Ad: i.p.
Wistar ratsin vivoPTZ↑ anticonvulsant effects, ↓ seizure severity, ↓ lipid peroxidation, ↑ antioxidant effect, ↑ memory retrieval in the passive avoidance task[125]
Dosage: 10, 20, and 40 mg kg−1
Ad.: p.o.
Duration: 15 days
Swiss albino micein vivoPTZ↑ immobility time, ↓ seizure severity[126]
Que/
Anisomelesma labarica
Dosage: 25 and 50 mg kg−1
Ad: i.p.
Wistar ratsin vivoPTZ↓ locomotor activity and motor
activity performance
[127]
Dosage: 6.25 and 12.5 mg kg−1
Ad: i.p.
Duration: 1 week
Wistar ratsin vivoPTZpotentiating the GABAergic system, inhibition of the NMDA receptor and Na+ channels.
Abbreviations: ↑, increase; ↓, decrease; GABA, glutamate and γ-amino butyric acid; i.p., intraperitoneal; NMDA, N-methyl-D-aspartate; p.o., oral; PTZ, pentylenetetrazol; Que, quercetin.
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

Rarinca, V.; Nicoara, M.N.; Ureche, D.; Ciobica, A. Exploitation of Quercetin’s Antioxidative Properties in Potential Alternative Therapeutic Options for Neurodegenerative Diseases. Antioxidants 2023, 12, 1418. https://doi.org/10.3390/antiox12071418

AMA Style

Rarinca V, Nicoara MN, Ureche D, Ciobica A. Exploitation of Quercetin’s Antioxidative Properties in Potential Alternative Therapeutic Options for Neurodegenerative Diseases. Antioxidants. 2023; 12(7):1418. https://doi.org/10.3390/antiox12071418

Chicago/Turabian Style

Rarinca, Viorica, Mircea Nicusor Nicoara, Dorel Ureche, and Alin Ciobica. 2023. "Exploitation of Quercetin’s Antioxidative Properties in Potential Alternative Therapeutic Options for Neurodegenerative Diseases" Antioxidants 12, no. 7: 1418. https://doi.org/10.3390/antiox12071418

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