OS is a condition in which there is an imbalance between 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, oxidative stress has been shown to play a role in the development and progression of several conditions.
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) [
48]. The pathogenesis of AD is commonly associated with extracellular accumulation of amyloid-β (1-40, 1-42) aggregates and hyperphosphorylation of tau proteins, leading to neurofibrillary tangles (NFT) and synaptic dysfunction [
13,
49,
50,
51]. 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 [
52,
53].
Oxidative stress plays an important role in AD, which, through ROS generation, can amplify or initiate the disease. The reduction reaction of hydrogen peroxide results in the production of reactive oxygen species, thus damaging brain tissue and disrupting brain cell repair [
54,
55]. Thus, the administration (ad) of quercetin before the treatment decreases the damage to the cell membrane induced by the oxidative stress caused by H
2O
2 [
55].
AD is characterized by neuronal loss, which is preceded by extracellular accumulation of Aβ
1–40, and Aβ
1–42. Antioxidants such as quercetin increase the resistance of neurons to oxidative stress by modulating cell death mechanisms. Thus, quercetin protects the mouse hippocampal cell line HT-22 from glutamate-induced oxidative toxicity and lipid peroxidation, by blocking the production of free radicals [
54]. Also, pretreatment of primary hippocampal cultures with quercetin significantly attenuated Aβ
1–42 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 Aβ
1–42 toxicity by modulating oxidative stress at lower concentrations (5 and 10 μM), while in the cases of higher concentrations (20 and 40 μM), 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 quercetin is its ability to act as an anti-amyloidogenic agent due to its five hydroxyl groups (see
Figure 2), which means it can prevent the formation of amyloid plaques in the brain [
56,
57].
Numerous studies demonstrate anticancer and apoptosis-inducing effects in vitro on a variety of cancer cell lines, including murine neuroblastoma HT-22 cells [
54,
58,
59]. Quercetin was found to be non-cytotoxic and strongly protected HT-22 cells from fibril formation [
59]. In addition, Ishige et al., 2001, [
54] using the HT-22 mouse hippocampal cell line found three distinct mechanisms of flavonoid protection in cell death, including increased intracellular GSH, direct decrease in ROS levels, and prevention of Ca
2+ influx.
In a study on the stable cell line of the Swedish mutant of amyloid precursor protein (APP695 transfected SH-SY5Y) no effects of quercetin were observed in the middle-late stage of AD; instead, effects were observed in the mid-early stage, when the reduction of β-amyloid converting enzyme 1 (BACE1) activity was recorded [
57].
The optimal concentration of quercetin required for effective destabilization of Aβ fibrils has been found to be in the range of 0.1–1 μM [
58,
60]. Thus, in a study using neurons of the rat hippocampal region, optimal doses of quercetin administration were beneficial for protecting against Aβ
25-35-induced amnesic injury by reducing lipid peroxidase, ROS, and GPx [
60]. In another study on pheochromocytoma (PC12) cells, quercetin was found to increase the survival rate of H
2O
2-damaged cells, decrease lipid peroxidation and GSH level, and provide mitochondrial protection mechanisms [
55]. On the other hand, Yu et al., 2020, [
61] have shown that quercetin has beneficial effects, so it can increase PC12 cell survival damaged by Aβ
25-35, antagonize toxicity Aβ, promote cell proliferation, and provide some neuroprotective effects.
Quercetin-3-glucuronide (Q3G), a glucuronide conjugate of quercetin, 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 oxidative stress in nerve cells, both of which being associated with AD [
62,
63]. Additionally, Q3G has been found to specifically localize in human brain tissue, suggesting that it may be able to cross the blood-brain barrier and reach areas of the brain affected by AD [
62]. Thus, Ho et al., 2013, [
64] observed that quercetin-3-O-glucuronide significantly reduced the generation of β-amyloid peptides by cultures of primary neurons generated by the mouse model Tg2576 AD.
A high concentration of quercetin was found in Ginkgo biloba, thus showing that
Ginkgo biloba extract (EGb761) and its constituents, quercetin and ginkgolide B, which by intraperitoneal administration (i.p.) have protective effects against the cytotoxic action of Aβ
1–42, thereby ameliorating oxidative phosphorylation deficits, and mitochondrial dysfunction in AD [
65]. Another plant used in pharmacotherapy and in which quercetin is found is
Acanthopanax henryi, that it can potentiate cholinergic activity by inhibiting acetylcholinesterase (AchE) [
66].
Table 2.
Protective effects against oxidative stress, neuroinflammation and Aβ accumulation, induced by quercetin in vitro.
Table 2.
Protective effects against oxidative stress, neuroinflammation and Aβ accumulation, induced by quercetin in vitro.
Types of quercetin |
Concentration of quercetin |
Model |
Exposure |
Effects |
Ref. |
Quercetin |
Dosage: 2.2 μM Duration: 24 h. |
HT-22 mouse hippocampal cell |
H2O2
|
↓lipid peroxidation, ↑intracellular GSH, ↓ROS |
[54] |
Dosage: 10-100 µmol L−1 Duration: 10 min |
PC12 cells |
H2O2
|
↓lipid peroxidation, ↓GSH, mitochondrial protection mechanisms. |
[55] |
Dosage: 10 μM |
APP695-transfected SH-SY5Y cells |
Aβ25–35
|
↓ROS, ↓BACE, ↓Aβ, ↓GSH, ↓lipid peroxidation |
[57] |
Dosage: 10 and 50 μM Duration: 7 days |
HT-22 mouse hippocampal cells |
Aβ1–42 or Aβ1–40
|
↓Aβ peptides, ↓the performed mature fibrils |
[58] |
Dosage: 2.4 µg mL−1 |
HT-22 murine neuroblastoma cells |
Aβ25–35
|
↓amyloidogenic Aβ peptides, inhibited Aβ fibril formation. |
[59] |
Quercetin-3′-glucoside |
Dosage: 10, 20, 40, and 80 μmol L−1 Duration: 24 h, 48 h, and 72 h |
PC12 cells |
Aβ25–35
|
↑ the survival rate of PC12 injured by Aβ25-35, promote cell proliferation, and antagonize the toxicity of Aβ, ↑ CREB/BDNF signaling pathway, ↓ROS |
[61] |
Q3G |
Dosage: 50 mg kg−1 Ad: gavage Duration: 24 h, 48 h, and 72 h |
APP695-transfected SH-SY5Y cells |
Aβ1–42
|
↓Aβ peptides, ↑CREB signaling, ↓Aβ aggregation, ↑mitogen-activated protein |
[64] |
Dosage: 50 mg kg −1 Ad: gavage Duration: 4 months |
Tg2576 AD primary neuron cultures |
|
↑neuronal survival, ↑c-Jun N-terminal kinases, ↓stress-induced impairments |
|
|
|
|
|
|
Quercetin/Ginkgo biloba
|
Dosage: 1.5-6 μg mL−1 Ad: i.p.
|
SHSY5Y human neuroblastoma cells |
Aβ1–42
|
↓ Akt signaling pathways, ↓Aβ toxicity, ↓platelet-activating factor |
[65] |
Quercetin/ Acanthopanax henryi
|
Dosage: 2.5, 5, 10, 20, and 40 μg mL−1 |
Cell free system |
|
↓ AchE activity, ↑antioxidant activity
|
[66] |
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 [
67].
Thus, it has been observed that the administration of quercetin can attenuate the levels of the SOD enzyme, reduce the amount of lactate dehydrogenase (LDH) release and protect human brain microvascular endothelial cells (hBMECs) from fibrillar β-amyloid1–40 (fAβ
1–40)-induced toxicity [
68]. In vivo studies in triple transgenic mice model of AD (3xTg-AD) have shown that quercetin 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 [
69,
70]. Additionally, the results showed that quercetin tended to improve active behaviors of 3xTG-AD mice and decreased neurodegeneration markers in mice [
70]. Additionally, in the case of APPswe/PS1dE9 transgenic mice it was observed that long-term quercetin consumption prevents memory loss, Aβ-induced neurotoxicity, and mitochondrial dysfunctions [
71].
Furthermore, Hayakawa et al., 2015, [
72] have shown that quercetin 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, quercetin 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), IL-1β, IL-6 [
73].
Quercetin has the effect of reducing anion superoxide levels that increased with H
2O
2 and Aβ treatment in hippocampal neurons [
74,
75]. On the other hand, oral (p.o.) treatment with 500 mg kg
−1 b.w. quercetin for 10 days can significantly increase brain apoE levels and reduce insoluble Aβ levels in the cortex of 5xFAD amyloid model mice [
76].
Interestingly, this memory impairment was markedly ameliorated by oral treatment with quercetin nanoencapsulated in zein nanoparticles (25 mg kg
−1 every 48 h for 2 months), while the administration of free quercetin was not able to reverse the faulty behavior, despite a higher administration frequency [
77].
In a study involving homozygotic transgenic mouse line B6.129S7-Sod2tm1Leb/J, where quercetin 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 quercetin had a protective effect against hydrogen peroxide- and paraquat-induced oxidative stress in the mice [
74].
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 [
78,
79]. There are studies that have found that quercetin alleviates scopolamine-induced memory deficits by protecting against neuroinflammation and neurodegeneration by inhibiting oxidative stress and acetylcholinesterase activity, reverses synaptic loss in the cortex and hippocampus of the brain of adult mice, and suppresses memory impairment [
78,
79].
Aluminum is a toxic metal that has neurological effects, including Alzheimer’s disease, by generating ROS [
80]. Increased production of reactive oxygen species leads to the disruption of cellular antioxidant defense systems and to the release of cytochrome c from mitochondria into the cytosol, resulting in apoptotic cell death [
80,
81]. Thus, the administration of 10 mg kg
−1 b.w quercetin reduces the effects induced by aluminum, thus reducing oxidative stress, in addition, it prevents cytochrome c translocation [
80].
In addition, Hou et al., 2010 [
82] have shown that flavonols can antagonize A toxicity 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 quercetin in vivo.
Table 3.
Protective effects against oxidative stress, neuroinflammation, and Aβ accumulation, induced by quercetin in vivo.
Types of quercetin |
Concentration |
Model |
Exposure |
Effects |
Ref. |
Quercetin |
Dosage: 5 or 10 mg kg−1 b.w.; Ad: p.o.; Duration: once daily; |
hBMECs |
fAβ1–40
|
↓SOD, ↓LDH |
[68] |
Dosage: 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 |
[69] |
Dosage: 100 mg kg−1 b.w.; Ad: gavage; Duration: every 48 h for 12 months; |
3xTg-AD mice |
|
↓neurodegeneration, ↓β-amyloidosis |
[70] |
Dosage: 20 and 40 mg kg−1 b.w.; Ad: p.o.; Duration: 16 weeks; |
adult male C57BL mice |
|
↑ MMP, ↑ATP levels, ↓ROS |
[71] |
Dosage: 20 mg; Ad: p.o.; Duration: 5 weeks; |
APP23 AD mice model |
Aβ |
↓eIF2α, ↓ATF4, ↓GADD34, ↑memory in aged mice, ↓memory deterioration in the early stage of AD, ↓memory dysfunction, ↓OS |
[72] |
Dosage: 1% in mouse chow; Ad: p.o.; Duration: from 3 to 13 months; |
double transgenic female mice |
|
↓neuroinflammation, ↓neurodegeneration, ↓ IL-1β |
[73] |
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 |
[74] |
Dosage: 100 mg kg−1 b.w.; Ad: p.o.; Duration: 22 days; |
adult male Sprague-Dawley rats |
Aβ1–42 |
↑expression of Nrf2/HO-1 in rat brain, ↓Aβ1-42 level, ↓antioxidant activity |
[75] |
|
|
|
|
|
|
Dosage: 25 mg kg−1; Ad: p.o.; Duration: 2 times a week for 2 months; |
SAMP8 mice |
|
↑ the cognition and memory impairments, ↓ astrogliosis |
[77] |
|
Dosage: 12.5 and 25 mg kg−1; |
mice |
Scopolamine |
↓OS, ↓AchE activity |
[78] |
|
Dosage: 30 mg kg−1 b.w.; Ad: i.p.; Duration: every day for 8 days; |
male albino Wistar rats |
Scopolamine |
abridged transfer latency, ↓avoidance response, ↓3,4-methylenedioxyamphetamine, acetylcholinesterase levels, ↑CAT, ↑ GSH levels |
[79] |
|
Dosage: 10 mg kg −1 b.w.; Ad: p.o.; Duration: every day for 12 weeks; |
male albino Wistar rats |
aluminum |
↓ROS production, ↑mitochondrial superoxide dismutase activity |
[80] |
Quercetin/ ginkgo flavonols |
Dosage: 4.8% in extract, all based on weight;
|
Double Transgenic (TgAPP/PS1) mice |
- |
Reversed the spatial learning deficit |
[82] |
Several studies have shown that type 2 diabetes is a risk factor for the onset of AD [
83,
84]. Moreover, changes in glucose regulation that accompany type 2 diabetes appear to affect regions of the hippocampus involved in age-related memory loss [
84].
The link between AD and type 2 diabetes is not fully understood. However, uncontrolled blood sugar can increase the risk of AD [
85]. Some researchers have referred to AD as “diabetes of the brain” or “type 3 diabetes” [
86,
87,
88]. Environmental factors and comorbidities, as well as other diseases that a certain person suffers from, can make them more susceptible to the onset of AD, but all studies so far point to the fact that the accumulation of amyloid is the fundamental cause of dementia.
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 [
89]. An increasing percentage of research indicates the association of Parkinson’s disease with microglial activation, resulting in an increase in various inflammatory mediators and neuroinflammation [
90,
91]. 1-Methyl-4-phenylpyridinium (MPP+) is the ultimate toxic agent formed by metabolism of MPTP and can activate glial cells to induce neuroinflammation [
92]. Research has shown that MPP+ induces microglial activation and degeneration of dopaminergic neurons, as well as generation of ROS in dopaminergic neurons [
93]. On the other hand, quercetin administration protects microglia cells against MPP+- induced increases in mRNA protein level of IL-1, IL-6 and TNF-α, due to its antioxidant action [
92]. 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 [
94,
95]. Thus, the formation of α-Syn fibrillations can be inhibited by quercetin and oxidized quercetin through their 1:1 covalent binding [
95].
Another neurotoxic synthetic organic compound used by researchers to selectively destroy dopaminergic and noradrenergic neurons is 6-hydroxydopamine (6-OHDA) [
96,
97]. It is a hydroxylated analogue of dopamine and is a benzenetriol with hydrogens on the phenyl ring at positions 2, 4 and 5. Isoquercetin, a flavonol derived from quercetin, has also been found to have protective effects against 6-OHDA-induced oxidative damage in a rat model of Parkinson’s disease, they observed that antioxidant enzymes, catalase (CAT), SOD, GPX and GSH levels, which were previously attenuated by 6-OHDA, increased significantly [
96,
97].
Table 4.
Protective effects against oxidative stress and neuroinflammation, induced by quercetin in vitro, in the case of Parkinson’s disease.
Table 4.
Protective effects against oxidative stress and neuroinflammation, induced by quercetin in vitro, in the case of Parkinson’s disease.
Types of quercetin |
Concentration |
Model |
Exposure |
Effects |
Ref. |
Quercetin |
Dosage: 0.1 μM
|
Microglial (N9)-neuronal (PC12) cells |
MPP |
↓iNOS gene expression, ↓ROS, ↓ cellular death, ↓DNA fragmentation, ↑apoptosis, ↓nuclear translocation of apoptosis-inducing factor, ↓caspase-3activation |
[92] |
Dosage:10 mM
|
PC12 cells |
α-Synuclein |
↓Aβ fibrillation |
[95] |
Isoquercetin |
Dosage: 10, 50, and 100 μM
|
PC12 cells |
6-OHDA |
↓ROS, ↑SOD, ↑GSH, ↑CAT, ↑GPx |
[96] |
Quercetin glycoside |
Dosage: 10, 50, and 100 μM |
PC12 cells |
6-OHDA |
↑antioxidant activity, ↑GSH, ↑GPx |
[97] |
In vitro studies have shown that quercetin can improve mitochondrial quality control, reduce oxidative stress, and increase levels of antioxidant enzymes [
92,
95,
96,
97]. Instead, in vivo studies in mice and 6-OHDA-induced PD rat models demonstrated that quercetin can improve locomotor and muscle activity, increase striatal dopamine levels, and protect neurons from mitochondrial dysfunction [
98,
99,
100,
101,
102].
Studies have shown that quercetin has neuroprotective effects against MPTP-induced neurotoxicity in Wistar rats and adult male C57BL/6 mice [
98,
99,
100]. Quercetin was found to reduce oxidative stress and neuroinflammatory cytokines in rats [
98,
99], as well as restores motor and non-motor symptoms (depression and cognitive impairment) of PD in rats injected with rotenone [
103,
104]. Additionally, quercetin supplementation was found to improve striatal cholinergic function and reduce rotenone-induced oxidative stress in rats [
104].
On the other hand, administration of fish oil can attenuate rotenone-induced oxidative impairments and mitochondrial dysfunctions in rat brain [
105]. Combined oral supplementation with fish oil and quercetin has been found to enhance neuroprotection in a chronic rotenone rat model, suggesting potential relevance for Parkinson’s disease [
105].
Table 5.
Protective effects against oxidative stress and neuroinflammation of quercetin in vivo in the case of Parkinson’s disease.
Table 5.
Protective effects against oxidative stress and neuroinflammation of quercetin in vivo in the case of Parkinson’s disease.
Types of quercetin |
Concentration |
Model |
Exposure |
Effects |
Ref. |
Quercetin |
Dosage: 25 mg kg−1 Ad: p.o.
|
Wistar rats |
Haloperidol MPTP |
↓cataleptic score, ↑actophotometer activity score, ↑GSH, ↓lipid peroxidation, ↓ROS |
[98] |
Dosage: 25 and 50 mg kg−1, Ad: intragastrically Duration: 14 days |
Wistar rats |
MPTP |
↓TNF-α, ↓IL-1β and ↓IL-6, ↓glutamate level, |
[99] |
Dosage: 50, 100 and 200 mg kg−1 Ad: p.o. Duration: 14 days. |
adult male C57BL/6 mice |
MPTP |
↓striatal dopamine depletion, ↓level of acetylcholine, ↑AchE activity, ↑motor deficits, ↑GPx, ↑SOD |
[100] |
Dosage: 100, 200 and 300 mg kg −1 Duration: 14 days |
Wistar rats |
6-OHDA |
↑spatial memory, ↓OS, ↓AchE activity, ↑antioxidant activity, ↓neuronal damage |
[101] |
Dosage: 20 mg kg−1 Ad: i.p. Duration: 1 month. |
Wistar rats |
6-OHDA |
↓neuroplastic changes in neural circuits, ↓excitability in neurons involved in epilepsy, ↓NMDA receptor functionality |
[102] |
Dosage: 25-75 mg kg−1 Duration: 12 h intervals for 4 days |
Wistar rats |
Rotenone |
↓nigral GSH depletion, ↓ROS, ↓striatal DA loss, ↑mitochondrial complex, ↓neuronal death |
[103] |
Dosage: 50 mg kg−1, Ad: p.o. Duration: 14 days |
Wistar rats |
Rotenone |
↑AchE activity, ↑SOD, ↓GPx, ↓CAT |
[104] |
Quercetin + fish oil |
Dosage: 25 mg kg−1 Ad: p.o. Duration: 28 days |
Wistar rats |
Rotenone |
↑mitochondrial functions, ↑GSH, ↑antioxidant defenses |
[105] |
3.3. Huntington’s disease
Along with Alzheimer’s and Parkinson’s, Huntington’s disease (HD) is a major health problem worldwide, with a major financial impact [
106]. Huntington’s disease is an autosomal dominant, inherited disorder, 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 Huntington’s disease, 3-nitropropionic acid (3-NPA) is administered, which alters the mitochondrial metabolism, decreases cellular ATP level, and includes the nerve cell death, by increasing OS.
In a study by Sandhir and Mehrotra [
107], in which female Wistar rats were used as model organisms, in which quercetin was orally administered at a dose of 25 mg kg
−1 for 21 days, for 17 of these 21 days concomitantly with 3-NPA, was observed an attenuation of motor deficits 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 oxidative stress, and lowering the ATP concentration [
107].
On the other hand, a study by Chakraborty [
108] failed to confirm the beneficial effect of quercetin on the 3-NP-induced striatal neuronal lesion. However, the conditions of the two studies varied little, Chakraborty used male rats as model organisms, and the duration of administration of 3-NP and quercetin was 4 days, and the concentration was higher (25-50 mg kg
−1), than that which was administered by Sandhir and Mehrotra, [
107] where the dose was administered subchronically (25 mg kg
−1). Although quercetin 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 [
108].
In addition, quercetin in combination with other antioxidants, such as lycopene, decreases anxiety and depression [
109]. 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 quercetin in combination with fish oil was observed in a rat model previously treated with 3-NPA, where it decreased OS, and improved motor function [
110].
Quinolinic acid (QA) is also a paradigm of Huntington’s disease, and co-administration of antioxidants such as quercetin with sesamol minimizes neurochemical, behavioral, and biochemical alterations in rat brains [
111]. However, these data appear inconsistent and unequivocal conclusions cannot be drawn.
Table 6.
Protective effects against oxidative stress and neuroinflammation, induced by quercetin in vitro in the case of Huntington’s disease.
Table 6.
Protective effects against oxidative stress and neuroinflammation, induced by quercetin in vitro in the case of Huntington’s disease.
Types of quercetin |
Concentration |
Model |
Exposure |
Effects |
Ref. |
Quercetin |
Dosage: 25 mg kg−1 Ad: p.o. Duration: 21 days; |
Wistar rats |
3-NPA |
↑ATP, ↑activity of complex II and V enzyme of respiratory chain complex, ↓ROS, ↑SOD, ↑CAT, ↓lipid peroxidation |
[107] |
|
Dosage: 25–50 mg kg−1 Ap: i.p.; Duration: 4 days |
Sprague Dawley rats
|
3-NPA |
↓gait despair, ↓microglial proliferation, ↓anxiety, ↑astrocyte numbers in the lesion core, ↓motor coordination deficits, ↓serotonin metabolism |
[108] |
Quercetin + lycopene |
Dosage: 50 mg kg−1, Duration: 14 days |
Wistar rats |
3-NPA
|
↓anxiety, ↓depression |
[109] |
Quercetin + fish oil |
Dosage: 25 mg kg−1
|
Wistar rats |
3-NPA |
↓OS, ↑motor function |
[110] |
Quercetin + sesamol |
Dosage: 25, 50, and 100 mg kg−1, Ad: i.p. Duration: 14 days before and 14 days after quinolinic acid administration |
Wistar rats |
QA |
↓behavioral, biochemical, and neurochemical alterations in the rat brain, ↑antioxidant effects, ↑anti-inflammatory activity |
[111] |