Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2023 Aug 15;21(10):2097–2109. doi: 10.2174/1570159X21666230203101107

New Perspectives of Taxifolin in Neurodegenerative Diseases

Rong Yang 1, Xinxing Yang 1, Feng Zhang 1,2,*
PMCID: PMC10556370  PMID: 36740800

Abstract

Neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), cerebral amyloid angiopathy (CAA), and Huntington’s disease (HD) are characterized by cognitive and motor dysfunctions and neurodegeneration. These diseases have become more severe over time and cannot be cured currently. Until now, most treatments for these diseases are only used to relieve the symptoms. Taxifolin (TAX), 3,5,7,3,4-pentahydroxy flavanone, also named dihydroquercetin, is a compound derived primarily from Douglas fir and Larix gemelini. TAX has been confirmed to exhibit various pharmacological activities, including anti-inflammation, anti-cancer, anti-virus, and regulation of oxidative stress effects. In the central nervous system, TAX has been demonstrated to inhibit Aβ fibril formation, protect neurons and improve cerebral blood flow, cognitive ability, and dyskinesia. At present, TAX is only applied as a health additive in clinical practice. This review aimed to summarize the application of TAX in neurodegenerative diseases and the underlying neuroprotective mechanisms, such as suppressing inflammation, attenuating oxidative stress, preventing Aβ protein formation, maintaining dopamine levels, and thus reducing neuronal loss.

Keywords: Taxifolin, neurodegenerative disease, neuroinflammation, oxidative stress, neuroprotection, Parkinson’s disease, Alzheimer’s disease

1. INTRODUCTION

Neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), cerebral amyloid angiopathy (CAA), and Huntington’s disease (HD) are characterized by motor symptoms and non-motor symptoms of cognitive dysfunction. The main causes of AD and CAA are amyloid β (Aβ) protein deposition and neuroinflammation. Similar to AD, the main pathological manifestations of PD are α-synuclein aggregation and the formation of Lewy bodies, further resulting in the loss of dopamine (DA) neurons. Studies have confirmed that oxidative stress and neuroinflammation exacerbate DA neuronal damage [1]. Moreover, HD is recognized as motor and cognitive dysfunction irreversibly, and HD has a one-half chance of being passed on to the next generation [2]. Currently, various treatments have been tried to treat neurodegenerative diseases, but they cannot be completely cured due to their complicated pathogenesis [3]. Meanwhile, most drugs used to treat these diseases involveside effects. Thus, opening new alternative avenues for treatment is essential.

Taxifolin (TAX), 3,5,7,3,4-pentahydroxy flavanone, also named dihydroquercetin, is a flavonoid widely found in Pinaceae plants, such as Larch, Douglas fir, and Cedrus deodara [4]. Also, TAX is found in seeds of milk thistle and onion [5, 6]. TAX has been listed as a new food raw material internationally. Meanwhile, TAX has been confirmed to be a health additive in food and drug. Besides, TAX has always been applied clinically for the treatment of cardiovascular and cerebrovascular diseases [7]. In addition, numerous studies have demonstrated the pharmacological activities of TAX, which include anti-cancer [8], anti-inflammatory [9], anti-oxidant [10], anti-viral [11], and anti-bacterial effects [12]. Recently, TAX-mediated neuroprotection has been well studied. For example, TAX inhibited Aβ protein formation and ameliorated cerebral blood flow, further facilitating Aβ clearance in the brain and improving cognitive disorder in the mouse models of CAA and AD, respectively [13]. Moreover, TAX reduced reactive oxygen species (ROS) generation, suppressed inflammation, and protected DA neurons in PD rat model [14, 15]. Otherwise, TAX might confer neuroprotection against HD by its anti-oxidant activity via network pharmacology analysis [16]. In this review, we have mainly summarized the effects of TAX on neurodegenerative diseases, including PD, AD, CAA, and HD, and the underlying mechanisms.

2. OVERVIEW OF TAX

2.1. Physicochemical Property of TAX

TAX is a flavonoid that was first isolated from Douglas fir bark and Siberian larch [17]. The stearic structure of the TAX crystal is C15H12O7, with a molecular weight of 304.25 (Fig. 1). HPLC analysis has revealed that TAX could exist in both trans- and cis-forms, and these trans- and cis-forms crystallize in cells. The melting points of TAX range from 218°C to 253°C [18]. Additionally, TAX is soluble in water-alcohol solutions and polar solvents [19]. Of course, the peculiar structure of TAX provides it with anti-inflammatory and anti-oxidant properties [20].

Fig. (1).

Fig. (1)

Chemical structure of TAX.

2.2. Pharmacological Actions of TAX

2.2.1. Regulation of Oxidative STRESS

Oxidative stress has been confirmed to be closely involved in the pathogenesis of neurodegenerative diseases. Amounts of physiologic and biochemical stimuli, such as expression of misfolded proteins and perturbation in redox status, could disrupt redox homeostasis and subsequently result in the accumulation of unfolded or misfolded proteins in neurodegenerative diseases. However, the brain response to detect and control oxidative stress was accomplished by a complex network of “longevity assurance processes” integrated into the expressions of genes termed vitagenes. For example, heat shock proteins are a highly conserved system participating in the preservation and repair of correct protein conformation [21]. In addition, the hormetic dose responses were mediated by endogenous cellular defense pathways, such as Nrf2- and sirtuin (SIRT)-related signaling pathways that coordinated adaptive stress responses in the treatment of neurodegenerative diseases [22]. Meanwhile, the emerging role of nitric oxide, hydrogen sulfide gases, and carbon monoxide in hormetic-based neuroprotection against neurodegenerative diseases and their relationship with mitochondrial redox signaling should also be paid more attention [23]. As a flavonoid compound, the carbonyl group in the structure of TAX makes it have an obvious anti-oxidant activity [24]. Previously, TAX was usually used for extending the expiration date of food. Recently, TAX was confirmed to activate Nrf2 signaling to restrain inflammation and oxidative stress in lung injury induced by Benzo[a]pyrene [25]. TAX could prevent chlorpyrifos (CPF)-induced neurotoxicity by up-regulating the level of phosphorylated AMP-activated protein kinase (p-AMPK) and activating Nrf2/heme oxygenase (HO-1) pathway [26]. Moreover, TAX could inhibit ROS production and increase calcium concentration to protect GABAergic neurons [27]. Besides, TAX exhibited apparent inhibition for cell proliferation, ROS overproduction, and NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome activation [28]. Also, TAX produced preventive effects on skin cancer, and the underlying mechanism might be associated with activating Nrf2 signaling via an epigenetic pathway. In addition, TAX suppressed cardiac hypertrophy and reduced ventricular fibrosis after pressure overload. These beneficial effects mediated by TAX were at least through the inhibition of ROS production and the activation of extracellular signal-regulated kinase (ERK) 1/2, c-Jun N-terminal kinase (JNK) 1/2, and Smads signaling pathways [29]. On the other hand, TAX improved H2O2-induced oxidative stress damage and inhibited H9C2 cell pyroptosis [30]. Likewise, TAX reduced the expression of caspase 1, thereby inhibiting the recruitment of macrophages and neutrophils. Beyond that, TAX has been shown to have the ability of decreasing ROS-induced malondialdehyde (MDA) levels, superoxide dismutase and glutathione peroxidase expressions [31]. Another evidence also demonstrated that TAX mitigated acute liver injury induced by CCl4 in mice via reducing MDA levels and increasing the expression of anti-oxidant enzyme activities [32]. In cisplatin-induced rat pulmonary damage, TAX conferred protection through its anti-oxidative stress actions [33, 34]. Collectively, these findings suggest that TAX generated anti-oxidative stress properties via the increased anti-oxidant enzyme activities and the decreased MDA levels.

At present, environmental pollution has become increasingly severe, especially heavy metal pollution. TAX was revealed to produce protection against hexavalent chromium-induced oxidative stress injury and monocyte inflammation in vitro [35]. Additionally, TAX was shown to protect retinal pigment epithelium (RPE) cells against oxidative stress-induced apoptosis via the activation of Nrf2 signaling and the phase II anti-oxidant enzyme system [36]. It is worth mentioning that TAX generated protection from Di-2-Ethylhexyl Phthalate (DEHP)-induced apoptosis in chicken cardiomyocytes through the anti-oxidative stress effects and regulation of cytochrome P450 (CYP450) expression [37]. Furthermore, TAX prevented acetaminophen (APAP)-induced hepatotoxicity through increasing anti-oxidant enzyme expressions [38].

2.2.2. Anti-inflammation

Flavonoids possess distinct anti-inflammatory properties with beneficial implications for chronic inflammation-induced diseases [39, 40]. Previous studies have confirmed TAX to present remarkable anti-inflammation effects [9]. For example, TAX inhibited LPS-induced immune response in dendritic cells [41]. Moreover, TAX ameliorated caspase-1-induced inflammatory responses in fatty liver cells [9]. In addition, TAX regulated T helper (Th) cells differentiation by inhibiting Notch1 and Jak2/Stat3 pathways activation to treat psoriasis [42]. In the experimental periodontitis of rats, TAX suppressed inflammatory factors’ production to attenuate periodontitis [43]. Similarly, TAX ameliorated iron overload-induced hepatocellular injury by reducing the production of pro-inflammatory cytokines [44]. In various diabetes animal models, TAX was confirmed to regulate postprandial hyperglycemia through its anti-inflammation and anti-oxidant properties [45]. In high-fat diet/streptozotocin-induced diabetic nephropathy (DN) rats, TAX was discerned to inhibit caveolin-1/nuclear factor-κB (NF-κB) signaling activation and further relieve DN [46]. Also, TAX improved alcoholic liver steatosis by inhibiting the activation of NLRP3 inflammasome [47]. Several studies have indicated TAX to attenuate homeostasis of glucose, inhibit the overactivation of the renin-angiotensin-aldosterone system (RAAS), and reduce inflammatory response via the inactivation of PI3K/AKT signaling pathway in vitro [48]. Additionally, TAX suppressed PI3K/AKT/mTOR and transforming growth factor-β (TGF-β)/Smads pathway activation to exert anti-inflammatory and anti-oxidant effects in order to ameliorate CCl4-induced liver fibrosis [49].

2.2.3. Anti-virus

In 2016, as the most active anti-oxidant compound extracted from Larix gemelinii, TAX was applied for Coxsackievirus B4 (CVB4) prevention, and it produced an anti-virus effect at the early stages of viral reproduction [50]. Furthermore, TAX has been recognized as a possible inhibitor of SARS-CoV-2 replication [51]. Recently, TAX was confirmed to be an effective natural substance for COVID-19 treatment [11].

2.2.4. Anti-bacteria

In addition to anti-virus action, TAX has been reported to exhibit anti-bacterial properties. TAX combined with dapagliflozin was utilized to cure colistin-induced nephrotoxicity in a rat model [52]. Moreover, TAX enhanced the intestinal barrier and modulated gut microbiota by inhibiting NF-κB signaling activation and further generated protection against dextran sulfate sodium-induced colitis [53]. Methicillin-resistant staphylococcus aureus (MRSA) is a common and contagious bacterium with hidden risks. TAX was confirmed to generate protection from MRSA-induced mouse pneumonia and prolong the mouse’s life span [54]. Helicobacter pylori (HP), a potent strain of bacteria, could worsen stomach ulcers. TAX attenuated HP-induced gastric ulcers [55].

2.2.5. Anti-apoptosis

TAX was found to reduce cadmium (Cd)-induced cytotoxicity and apoptosis [56]. In addition, TAX promoted the osteogenesis of human bone marrow mesenchymal stem cells (HBMSCs) and further inhibited NF-κB signaling pathway activation [57]. Besides, TAX improved bone formation in alveolar bone successfully in the experimental periodontitis model and decreased bone cell apoptosis [43]. In the myocardial ischemia or reperfusion injury rat model, TAX conferred protection through regulating the mitochondrial apoptosis pathway [58].

2.2.6. Anti-DNA Injury

Surprisingly, TAX was revealed to generate protection against DNA damage [59]. Although TAX did not exhibit DNA toxicity in vitro, more clinical experiments are still needed to prove it in vivo [60]. These findings suggest that TAX combined with other flavonoids might be a noteworthy anti-cancer medicine potentially [61, 62].

2.2.7. Anti-cancer

An interesting finding reported that TAX could recover various biochemical changes caused by cancer, such as body weight, tumor growth, and various metabolic enzyme activities [63]. Also, TAX had been shown to have the capacity of inhibiting proliferation, migration and invasion of highly aggressive breast cancer cells [8]. In addition, TAX suppressed two enzyme activities of 3β-hydroxysteroid dehydrogenase and 17α-hydroxylase/17 in human with IC50 value of about 100 μM. Based on this finding, TAX might be serviceable for treating prostate cancer [64]. Furthermore, TAX reduced the expression of ATP-binding cassette subfamily B member 1 (ABCB1) and the function of P-glycoprotein (P-gp) via uncompetitive inhibition of rhodamine 123 and doxorubicin efflux, suggesting that TAX might be useful for multiple drug resistance (MDR) to chemotherapeutic agents in cancer [65]. Similarly, TAX suppressed liver cells' abnormal growth and promoted the apoptosis of liver cancer cells, thus producing anti-cancer effects [66]. For lung cancer, TAX attenuated the stemness of lung cancer cells possibly through the inactivation of PI3K and OCT4 signaling [67]. Moreover, TAX improved dimethylbenzanthracene (DMBA)-induced mammary carcinoma via down-regulating the aryl hydrocarbon receptor (AhR) signaling pathway [68]. Besides, in the solar UV-induced skin carcinogenesis mouse model, TAX inhibited epidermal growth factor (EGF)-induced cell transformation to produce protection against UV-induced skin carcinogenesis [69]. In addition, TAX was found to block epithelial-mesenchymal transition (EMT) to prevent migration and invasion of gastric cancer cells and thus inhibit tumor growth. The potential mechanism was associated with suppressing the activation of the AhR/CYP450 1A1 (CYP1A1) signaling pathway [70]. In vitro studies, TAX exhibited its preventive potential for colon cancer by enhancing the apoptotic signaling in HCT116 and HT29 colon cancer cells [71]. Together, TAX might be a prodrug or a potential adjuvant for cancer treatment [67].

2.2.8. Anti-hyperglycemia

In the diabetic retinopathy (DR) rat model, TAX reduced retinal ganglion blood vessel damage and decreased oxidative stress and inflammation reactions [72]. Pre-administration with TAX improved the postprandial hyperglycemia in rats and also decreased triglyceride absorption by suppressing pancreatic lipase [73]. Recent studies indicate that TAX derivatives are capable of inhibiting the activity of carbohydrate-hydrolyzing enzymes, decreasing carbohydrate absorption in diabetes [74].

2.2.9. Anti-glycation

Advanced glycation end-products (AGEs) formation is associated with type 2 diabetes. TAX could inhibit AGEs production and show the effect of anti-glycation [75]. Additionally, TAX more efficiently inhibited glycation in the collagen-fructose reaction and the elastin-glyceraldehyde reaction than quercetin and luteolin [76].

2.2.10. Other Biological Activities

TAX inhibited the xanthine oxidase activity in the liver to achieve a strong urine-lowering effect, suggesting that TAX might be a potential substance for treating hyperuricemia [77]. Moreover, TAX uncompetitively suppressed kidney 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) activity against steroid substrates in human and various animal models [78]. Likewise, TAX inhibited osteoclastogenesis via restraining NF-κB signaling activation and decreased bone loss in the mouse model of calvarial osteolysis, which suggested that TAX might be one of the potential treatment means for osteoporosis and rheumatoid arthritis [79]. In addition, TAX suppressed obesity by reducing excessive inflammation [80]. In the high-fat feeding mouse model, TAX presented anti-obesity and gut microbiota-modulating effects [81]. In recent years, TAX has been confirmed to alleviate psoriatic dermatitis in the mouse model and inhibit imiquimod-induced cell over-proliferation [82, 83]. Additionally, TAX suppressed mast cell activation by inhibition of AKT/NF-κB and mitogen-activated protein kinases (MAPKs)/cytoplasmic phospholipase A2 (cPLA2) signaling pathway, suggesting that TAX could alleviate mast cell-mediated passive cutaneous anaphylaxis reaction [84]. Increasingly, TAX was able to improve skin viscoelasticity, which was used for the skincare component [85]. Moreover, it was well confirmed that flavonoid-conjugated compounds could enhance the regeneration process and repair hair follicles and sebaceous glands after chemical burns. Further, TAX-conjugated acetaldehyde-based agents were found to be more effective than the wound healing agent Olasol. Therefore, TAX conjugated with carbonyl compounds could be used for burn healing [86]. In addition, in chronic obstructive pulmonary disease (COPD) mouse model and CSE-induced human bronchial epithelial (HBE) cells, TAX conferred beneficial effects via attenuating oxidative stress and reducing expressions of iron death-related proteins, such as glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11) [87]. Another evidence showed the interaction of TAX to exist with bovine hemoglobin at physiological pH, revealing TAX to be located in the hemoglobin moiety [88]. Furthermore, TAX could interact with amino acids located in the active site, in which TAX was recognized as the most principal lead molecule and its minimal inhibition concentration was ≤ 12.5 μg/mL [89].

Besides, TAX could attenuate oxidative damage caused by UV injury [69]. Also, TAX showed an anti-aging effect [90], indicating the application of TAX to not only be used for the treatment of certain diseases but also applied as a skin care product. Moreover, TAX could be used in combination with other drugs to enhance the therapeutic effects in clinical practice. For example, glucosamine alendronate (GA) combined with TAX appeared to be more effective than GA treatment alone in maintaining the strength of the femur and bone mineral density (BMD) in the osteoporosis rat model [91]. On the other hand, the combination of TAX and andrographolide enhanced the anti-proliferation and anti-cancer effects of andrographolide [92]. In addition, daily application of TAX (1.5 g/kg) improved the anti-oxidant status and promoted the growth of broiler chicken [93]. Also, TAX was confirmed as a food supplement to improve the immune status of gilthead seabream (Sparus aurata L.) and activate immune cells in vitro [94]. On the other hand, the purified TAX was proved to confer beneficial effects against biofouling bacteria, algal spore germination, and mollusk foot adhesion, respectively. TAX isolated from Streptomyces sampsonii PM33 derived from a mangrove forest was found to be a promising candidate for the development of eco-friendly anti-fouling agents [95]. Collectively, TAX exhibited a variety of pharmacological activities, suggesting that more possibilities of TAX would be applied in future clinical practice.

3. TAX AND NEURODEGENERATIVE DISEASES

In CNS, TAX was verified to provide neuroprotection against neurodegeneration via reducing inflammatory responses [13] and ROS production [96]. However, the underlying neuroprotective mechanisms warrant further exploration.

3.1. TAX and PD

PD is one of the most common neurodegenerative diseases, mainly characterized by DA neuronal loss in the substantia nigra [97]. PD patients present motor symptoms, such as static tremor, bradykinesia, and muscle rigidity, and non-motor symptoms, such as depression, constipation, and hypoxia [98]. PD is more common in the older population, affecting up to 1% of the population over 60 years old, increasing with aging and getting worse [99]. Until now, most drugs have been utilized for PD treatment. However, these drugs have limited efficacy and unignorable side effects [100].

Although the pathogenesis of PD is currently poorly defined, age, neuroinflammation, and oxidative stress are the most important factors in PD progression [101, 102]. The anti-Parkinson potential of silymarin has been proven. Silymarin is a mixture of flavonolignans (silybin, isosilybin, silychristin, and siliandrin) and a flavonoid (TAX), suggesting that TAX might be useful for PD [14]. Moreover, TAX was found to maintain DA levels in the brain and attenuate proteasome inhibitor-induced neurotoxicity [103, 104]. In human neuroblastoma SH-SY5Y cells, TAX decreased ROS production and further attenuated cell apoptosis [105]. In addition, TAX was discerned to reduce the production of peroxide in glial cells via inhibiting the deamination of monoamine oxidase (MAO) or scavenging free radicals [106]. Recent studies have indicated TAX to ameliorate oxidative stress and inhibit the expression of pro-inflammatory genes, such as IL-1β and TNF-α, via the inactivation of NF-κB signaling, and it ultimately improved rotenone-induced PD rat model [15]. Thus, these findings indicate that TAX-mediated dopamine neuroprotection might be related to suppressing pro-inflammatory factors release and reducing oxidative stress-induced neuronal damage [14, 107].

On the other hand, type 2 diabetes is associated with the risk of PD, which might be due to the production of hyperuricemia [108]. Luckily, TAX has been proven to potentially inhibit hyperuricemia [77]. Besides, another increasing risk factor related to PD is cancer. A clinical case report indicated melanoma patients to have a higher risk of PD [109]. TAX was confirmed to attenuate cellular melanogenesis via inhibiting tyrosinase enzymatic activity despite increasing tyrosinase protein levels [110]. Since PD not only includes motor symptoms but also presents non-motor symptoms, such as depression and constipation, TAX was proven to reduce the level of 5-hydroxytryptamine and further produce anti-depression effects [111]. In conclusion, TAX generated neuroprotection against PD directly or indirectly. The underlying neuroprotective mechanisms of TAX on PD are shown in Fig. (2).

Fig. (2).

Fig. (2)

Potential mechanisms underlying TAX-mediated neuroprotection against PD. TAX conferred dopamine neuroprotection via decreasing the production of ROS and further attenuating apoptosis, suppressing glial cells-mediated neuroinflammation and inhibiting NF-κB signaling pathway activation.

3.2. TAX and AD

AD is characterized by the commonest cause of dementia with cognitive impairment. Most cases occur in the elderly more than 60 years old [112]. The significant pathological feature of AD is the accumulation of Aβ protein misfolding. Of course, the pathogenesis of AD is not just the accumulation of Aβ, but is also related to the immune mechanism in the brain. The misfolding and aggregation of Aβ proteins were found to be linked to pattern recognition receptors on microglia and astroglia, and further produced a congenital immune response. This process was accompanied by a release of various inflammatory mediators, and these mediators were closely associated with the progression and severity of AD [113-115]. In addition, Aβ aggregation, neurofibrillary tangles, and neurodegeneration have been verified to be the predominant inflammatory inducers in AD brain, while activated microglia and astroglia have been reported to exacerbate AD progression [116]. However, neuroinflammation could further accelerate neuronal death in the brain. Previous studies have indicated Aβ42 activated cPLA2 and prostaglandin E2 (PGE2) to damage synapses [117, 118]. In Aβ42-induced AD mouse model, TAX promoted the clearance of Aβ42 and inhibited the expressions of cPLA2 and pro-inflammatory mediators [119]. Moreover, the computational analysis demonstrated that the covalent admixture formed between the oxidized form of flavonoid (+) -TAX and Aβ could chemically react with Aβ-specific recognition motifs and thus exhibit anti-Aβ aggregation properties [120]. Since Hen egg white lysozyme (HEWL) protein production was recognized to be closely associated with Aβ formation, TAX inhibited HEWL fibril formation and then reduced mature Aβ aggregation in a dose-dependent manner [121,122]. Moreover, microcirculation plays an important role in the brain, and the decreased brain capillary density might be another risk factor for cognitive impairment in AD [123]. In a clinical trial, TAX combined with ascorbic acid for the treatment of atherosclerosis improved cognitive dysfunction and microcirculation [124, 125]. Furthermore, TAX was shown to improve cognitive disorders and protect hippocampal neurons in the mouse model of AD [126]. Also, TAX inhibited Aβ formation and promoted its clearance in the brain, and thus ameliorated brain’s cognitive function [127, 128]. Since β-site amyloid precursor protein cleaving enzyme 1 (BACE1) is known to be a rate-limiting enzyme producing Aβ, TAX has been confirmed to be an effective BACE1 inhibitor [129]. In addition, TAX inhibited pro-inflammatory factors production and then attenuated LPS-induced cognitive dysfunctions. The underlying mechanisms were associated with the promotion of SIRT1 activation and the inhibition of phosphorylated-JAK2/phosphorylated-STAT3-coupled NF-κB-linked BACE1 expression [130]. Moreover, TAX expressed the inhibition of intracerebral Aβ generation by suppressing apolipoprotein E (APOE)-ERK1/ 2-Aβ precursor protein axis, which was responsible for the secretion of Aβ [131, 132]. Besides, TAX generated neuroprotection against cognitive dysfunctions via its anti-oxidant and suppression of glutathione (GSH) depletion actions [105]. Likewise, TAX was found to be capable of inhibiting the activity of the neurotransmitter acetylcholinesterase and butyrylcholinesterase from blocking the transmission of these two enzymes in the synaptic cleft and ultimately attenuate neuronal damage [133, 134].

Additionally, CAA is related to the development of lobar intracerebral hemorrhages (ICHs). CAA is pathologically characterized by cerebrovascular Aβ accumulation (deposition of Aβ in the tunica media). Further, studies have confirmed Aβ to exist not only in cortical, leptomeningeal arteries, arterioles and capillaries of most CAA patients but also in the venules of brain in rare cases of CAA [135, 136]. Recently, AD and aging have been confirmed to be other risk factors of CAA [137]. Interestingly, the shared role of Aβ deposition in AD and CAA was arguably the clearest instance of crosstalk between neurodegenerative and cerebrovascular disorders. The pathogenic pathways of AD and CAA intersected at the levels of Aβ generation, its circulation within the interstitial fluid, and perivascular drainage pathways and brain clearance. However, their mechanisms of brain injury and disease presentation diverged [138]. Thus, the treatment for CAA would be effective for AD. TAX was confirmed to promote the breakdown of Aβ and inhibit Aβ assembly and oligomer formation. After TAX treatment, the higher blood Aβ level was discerned, suggesting TAX could promote the clearance of Aβ from the brain to the circulation. Therefore, TAX restored cerebral blood flow (CBF) and cerebrovascular reactivity (CVR), and reduced the accumulation of Aβ and cognitive disorders in the CAA mouse model [139]. In addition, TAX was found to elevate blood Aβ level to maintain cerebrovascular integrity in the CAA mouse model [140]. Furthermore, TAX was verified to facilitate disassembly, block oligomer formation, and increase the clearance of Aβ, and then improve memory impairment in a mouse model of CAA [141]. On the other hand, oxidative stress was reported to aggravate CAA. Inhibition of ROS production might be promising and beneficial for CAA treatment [142]. Recently, microglial activation was observed in CAA-related inflammation. It has been well studied that triggering receptor expressed on myeloid cell 2 (TREM2) is expressed only on microglia in the brain, and TREM2 overexpression would aggravate the neuroinflammation in the brain [143]. TAX could suppress oxidative stress and neuroinflammation, and reduce the overexpression of TREM2 to further confer neuroprotection in the CAA mouse model [131]. Collectively, TAX might open new alternative avenues for AD treatment. The potential mechanism of TAX in AD is shown in Fig. (3).

Fig. (3).

Fig. (3)

Potential mechanisms underlying TAX-mediated neuroprotection against AD or CAA. In AD, TAX conferred neuroprotection via decreasing the level of cPLA2 to reduce the synthesis of PGE2, inhibiting neuroinflammation, suppressing the activation of APOE and ERK1/2 signaling to decrease Aβ production, up-regulating SIRT1 expression to inhibit Aβ-induced JAK-2/STAT3 pathway activation, and then decreasing BACE1 expression to ultimately inhibit Aβ aggregation in neurons. In CAA, TAX promoted the breakdown of Aβ, inhibited Aβ oligomer formation, cleared Aβ from the brain to the circulation, and restored the cerebral blood flow (CBF) and cerebrovascular reactivity (CVR).

3.3. TAX and HD

HD is one of the neurodegenerative diseases with unique clinical presentation, including chorea and dystonia, dyscoordination, cognitive decline, and behavioral disorder. Further, studies have demonstrated mutant huntingtin (mHTT) to aggregate mitochondrial and metabolism dysfunctions and inhibit the expression of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) [144, 145]. Also, mHTT could inhibit autophagy to clear misfolded Aβ proteins and further damage neurons. Therefore, at present, the use of genetic techniques to reduce transcription of mHTT mRNA is the main therapeutic approach. Additionally, in HD patients and HD mouse models, glial cells’ activation and up-regulation of the mRNA levels of various pro-inflammatory factors were discerned [146]. Thus, inhibition of glial cells’ activation and NF-κB signaling pathway has been considered to be a potential avenue to treat HD [147, 148]. Recent studies have confirmed TAX to be a potentially promising alternative for HD treatment due to its anti-oxidant activity [16]. However, the mechanism underlying TAX-mediated neuroprotection against HD has not been reported till yet.

3.4. TAX and Other Neurological Diseases

It is well known that cerebral ischemia reperfusion (I/R) injury is related to the production of free radicals, intracellular calcium overload, excitatory amino acid toxicity, high leukocyte aggregation, inflammatory reactions, and lack of high-energy phosphate compounds. TAX could suppress the expression of inflammatory mediators, such as cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), and inhibit the activation of NF-κB signaling and leukocyte infiltration [149, 150]. Besides, two key counter-receptors, macrophage antigen 1 (Mac-1) and intercellular adhesion molecule 1 (ICAM-1), were found to be closely associated with the limited leukocyte infiltration. TAX was capable of inhibiting these two receptor expressions and then improving cerebral I/R injury [151].

4. BIOAVAILABILITY OF TAX

Previous studies have found that TAX bound to glyoxalic acid condensation could become a new compound consisting of two TAX unit dimers, having more effective anti-oxidant effects than TAX [152]. In addition, TAX-chitosan compositions were found to present more apparent anti-hypoxia actions in the process of preventive oral therapy under experimental hypoxia conditions than TAX alone [153]. On the other hand, in order to improve the bioavailability of TAX, γ-cyclodextrin (γ-CD) was employed as an inclusion complex. Further, TAX-γ-CD inclusion complex by emulsion solvent evaporation and freeze drying combination method had demonstrated better bioavailability and anti-oxidant capacity than TAX [154]. Moreover, to achieve better water-soluble biological properties, TAX was contained in the loop of β-cyclodextrin (β-CD), and TAX-β-CD could release TAX slowly and support a low concentration of the free form of TAX for long term, thus making TAX penetrate into the bloodstream [155].

CONCLUSION

Flavonoids have always been one of the important anti-inflammatory and anti-oxidant compounds. In clinical practice, flavonoids have presented promising beneficial effects in various diseases. As a typical flavonoid compound, TAX not only has significant anti-oxidant and anti-inflammatory activity, but also generates anti-apoptosis, anti-bacterial, and anti-cancer capacity. Besides, TAX was confirmed to exert protective effects against cardiovascular and liver diseases. Recently, studies on TAX in CNS have attracted wide attention. Current studies have indicated TAX to improve cognitive and motor impairment in neurodegenerative diseases, including PD, AD, and HD. The underlying mechanisms have been found to be related to clearing Aβ protein, reducing neuroinflammatory response, increasing DA level, and regulating oxidative stress (Table 1). Additionally, TAX has been confirmed to present a few side effects in various clinical studies. Importantly, TAX and its metabolites could be detected in rat brain [156]. Thus, TAX might have a good safety profile to be an ideal candidate for neurodegenerative disease treatment. However, current studies are limited to clinical and basic experiments. Collectively, TAX extends our understanding of the neuroprotective properties of TAX in neurodegenerative diseases. Since the etiology and pathogenesis of neurodegenerative diseases are complex, more clinical trials of TAX warrant future investigation.

Table 1.

Mechanisms underlying TAX-mediated neuroprotection against neurodegenerative diseases.

Neurodegenerative Disease Biological Target Results References
Parkinson’s disease (PD) Rotenone-induced rat model Inhibiting NF-κB signaling pathway activation and the
expressions of inflammatory factors
Akinmoladun et al. [15]
MPTP mouse model Maintaining DA level Pérez et al. [104]
SH-SY5Y cell Decreasing ROS production and further attenuating cell
apoptosis
Kim et al. [105]
Alzheimer’s disease (AD) and cerebral amyloid angiopathy (CAA) 42-induced SH-SY5Y cell Reducing cPLA2 and PGE2 levels Wang et al. [119]
42-induced mouse model Inhibiting Aβ42 fibril formation Sato et al. [128]
Neuroblastoma N2a Swe cell line Downregulating BACE1 expression to inhibit Aβ formation Park et al. [130]
Mouse primary hippocampal
neurons
LPS-induced mouse model
Decreasing activation of glial cells and suppressing TREM2 expression
Inhibiting the oxidative injury and inflammatory response
Inoue et al. [131]
Tg-SwDI mouse Reducing Aβ1-40 fibril formation and improving cerebrovascular disorders Saito et al. [139]
Huntington’s disease (HD) Network pharmacology analysis Anti-oxidant activity De Oliveira et al.
[16]

ACKNOWLEDGEMENTS

The authors have acknowledged the Servier Medical Art image bank that was used to create schematic Figure 2, Figure 3, and Graphical Abstract.

LIST OF ABBREVIATIONS

AD

Alzheimer’s Disease

Amyloid β

BMD

Bone Mineral Density

CAA

Cerebral Amyloid Angiopathy

COPD

Chronic Obstructive Pulmonary Disease

DA

Dopamine

DN

Diabetic Nephropathy

GA

Glucosamine Alendronate

HBE

Human Bronchial Epithelial

HD

Huntington’s Disease

iNOS

Inducible Nitric Oxide Synthase

MDR

Multiple Drug Resistance

MRSA

Methicillin-resistant Staphylococcus aureus

PD

Parkinson’s Disease

RAAS

Renin-angiotensin-aldosterone System

ROS

Reactive Oxygen Species

AUTHORS’ CONTRIBUTIONS

FZ, XXY and RY contributed to the writing of the manuscript and the design of the figures.

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

This study was supported by the National Natural Science Foundation of China (No. 82160690), the Science and Technology Foundation of Guizhou Province (No. ZK[2021]-014) and the Collaborative Innovation Center of the Chinese Ministry of Education (No. 2020-39).

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

REFERENCES

  • 1.Dugger B.N., Dickson D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017;9(7):a028035. doi: 10.1101/cshperspect.a028035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ghosh R., Tabrizi S.J. Clinical features of huntington’s disease. Adv. Exp. Med. Biol. 2018;1049:1–28. doi: 10.1007/978-3-319-71779-1_1. [DOI] [PubMed] [Google Scholar]
  • 3.Heemels M.T. Neurodegenerative diseases. Nature. 2016;539(7628):179. doi: 10.1038/539179a. [DOI] [PubMed] [Google Scholar]
  • 4.Sunil C., Xu B. An insight into the health-promoting effects of taxifolin (dihydroquercetin). Phytochemistry. 2019;166:112066. doi: 10.1016/j.phytochem.2019.112066. [DOI] [PubMed] [Google Scholar]
  • 5.Kim N.C., Graf T.N., Sparacino C.M., Wani M.C., Wall M.E. Complete isolation and characterization of silybins and isosilybins from milk thistle (Silybum marianum) Electronic supplementary information (ESI) available: HPLC chromatograms of isolates and extracts. See http://www.rsc.org/suppdata/ob/b3/b300099k/. Org. Biomol. Chem. 2003;1(10):1684–1689. doi: 10.1039/b300099k. [DOI] [PubMed] [Google Scholar]
  • 6.Weidmann A.E. Dihydroquercetin: More than just an impurity? Eur. J. Pharmacol. 2012;684(1-3):19–26. doi: 10.1016/j.ejphar.2012.03.035. [DOI] [PubMed] [Google Scholar]
  • 7.Guo H., Zhang X., Cui Y., Zhou H., Xu D., Shan T., Zhang F., Guo Y., Chen Y., Wu D. Taxifolin protects against cardiac hypertrophy and fibrosis during biomechanical stress of pressure overload. Toxicol. Appl. Pharmacol. 2015;287(2):168–177. doi: 10.1016/j.taap.2015.06.002. [DOI] [PubMed] [Google Scholar]
  • 8.Li J., Hu L., Zhou T., Gong X., Jiang R., Li H., Kuang G., Wan J., Li H. Taxifolin inhibits breast cancer cells proliferation, migration and invasion by promoting mesenchymal to epithelial transition via β-catenin signaling. Life Sci. 2019;232:116617. doi: 10.1016/j.lfs.2019.116617. [DOI] [PubMed] [Google Scholar]
  • 9.Zhan Z.Y., Wu M., Shang Y., Jiang M., Liu J., Qiao C.Y., Ye H., Lin Y.C., Piao M.H., Sun R.H., Zhang Z.H., Jiao J.Y., Wu Y.L., Nan J.X., Lian L.H. Taxifolin ameliorate high-fat-diet feeding plus acute ethanol binge-induced steatohepatitis through inhibiting inflammatory caspase-1-dependent pyroptosis. Food Funct. 2021;12(1):362–372. doi: 10.1039/D0FO02653K. [DOI] [PubMed] [Google Scholar]
  • 10.Ahiskali I., Pinar C.L., Kiki M., Cankaya M., Kunak C.S., Altuner D. Effect of taxifolin on methanol-induced oxidative and inflammatory optic nerve damage in rats. Cutan. Ocul. Toxicol. 2019;38(4):384–389. doi: 10.1080/15569527.2019.1637348. [DOI] [PubMed] [Google Scholar]
  • 11.Bernatova I., Liskova S. Mechanisms modified by (−)-epicatechin and taxifolin relevant for the treatment of hypertension and viral infection: Knowledge from preclinical studies. Antioxidants. 2021;10(3):467. doi: 10.3390/antiox10030467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Artem’eva O.A., Pereselkova D.A., Fomichev Y.P. Dihydroquercetin, the bioactive substance, to be used against pathogenic microorganisms as an alternative to antibiotics. Sh. Biol. 2015;50(4):513–519. doi: 10.15389/agrobiology.2015.4.513eng. [DOI] [Google Scholar]
  • 13.Tanaka M., Saito S., Inoue T., Satoh-Asahara N., Ihara M. Novel therapeutic potentials of taxifolin for amyloid-β-associated neurodegenerative diseases and other diseases: Recent advances and future perspectives. Int. J. Mol. Sci. 2019;20(9):2139. doi: 10.3390/ijms20092139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ullah H., Khan H. Anti-parkinson potential of silymarin: Mechanistic insight and therapeutic standing. Front. Pharmacol. 2018;9:422. doi: 10.3389/fphar.2018.00422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Akinmoladun A.C., Famusiwa C.D., Josiah S.S., Lawal A.O., Olaleye M.T., Akindahunsi A.A. Dihydroquercetin improves rotenone‐induced parkinsonism by regulating NF‐κB‐mediated inflammation pathway in rats. J. Biochem. Mol. Toxicol. 2022;36(5):e23022. doi: 10.1002/jbt.23022. [DOI] [PubMed] [Google Scholar]
  • 16.de Oliveira N.K.S., Almeida M.R.S., Pontes F.M.M., Barcelos M.P., Silva G.M., de Paula da Silva C.H.T., Cruz R.A.S., da Silva Hage-Melim L.I. Molecular docking, physicochemical properties, pharmacokinetics and toxicity of flavonoids present in euterpe oleracea martius. Curr. Computeraided Drug Des. 2021;17(4):589–617. doi: 10.2174/1573409916666200619122803. [DOI] [PubMed] [Google Scholar]
  • 17.Pew J.C. A flavonone from Douglas-fir heartwood. J. Am. Chem. Soc. 1948;70(9):3031–3034. doi: 10.1021/ja01189a059. [DOI] [PubMed] [Google Scholar]
  • 18.Kiehlmann E., Li E.P.M. Isomerization of dihydroquercetin. J. Nat. Prod. 1995;58(3):450–455. doi: 10.1021/np50117a018. [DOI] [Google Scholar]
  • 19.Rogozhin V.V., Peretolchin D.V. Kinetic regulations of dihydroquercetin oxidation with horseradish peroxide. Russ. J. Bioorganic Chem. 2009;35(5):576–580. doi: 10.1134/S1068162009050069. [DOI] [PubMed] [Google Scholar]
  • 20.Topal F., Nar M., Gocer H., Kalin P., Kocyigit U.M., Gülçin İ., Alwasel S.H. Antioxidant activity of taxifolin: An activity–structure relationship. J. Enzyme Inhib. Med. Chem. 2016;31(4):674–683. doi: 10.3109/14756366.2015.1057723. [DOI] [PubMed] [Google Scholar]
  • 21.Calabrese V., Guagliano E., Sapienza M., Mancuso C., Butterfield D.A., Stella A.M. Redox regulation of cellular stress response in neurodegenerative disorders. Ital. J. Biochem. 2006;55(3-4):263–282. [PubMed] [Google Scholar]
  • 22.Calabrese V., Cornelius C., Dinkova-Kostova A.T., Calabrese E.J., Mattson M.P. Cellular stress responses, the hormesis paradigm, and vitagenes: Novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid. Redox Signal. 2010;13(11):1763–1811. doi: 10.1089/ars.2009.3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Calabrese V., Mancuso C., Calvani M., Rizzarelli E., Butterfield D.A., Giuffrida S.A.M. Nitric oxide in the central nervous system: Neuroprotection versus neurotoxicity. Nat. Rev. Neurosci. 2007;8(10):766–775. doi: 10.1038/nrn2214. [DOI] [PubMed] [Google Scholar]
  • 24.Akinmoladun A.C., Olaniyan O.O., Famusiwa C.D., Josiah S.S., Olaleye M.T. Ameliorative effect of quercetin, catechin, and taxifolin on rotenone-induced testicular and splenic weight gain and oxidative stress in rats. J. Basic Clin. Physiol. Pharmacol. 2020 doi: 10.1515/jbcpp-2018-0230. [DOI] [PubMed] [Google Scholar]
  • 25.Islam J., Shree A., Vafa A., Afzal S.M., Sultana S. Taxifolin ameliorates Benzo[a]pyrene-induced lung injury possibly via stimulating the Nrf2 signalling pathway. Int. Immunopharmacol. 2021;96:107566. doi: 10.1016/j.intimp.2021.107566. [DOI] [PubMed] [Google Scholar]
  • 26.Zhang C., Zhan J., Zhao M., Dai H., Deng Y., Zhou W., Zhao L. Protective mechanism of Taxifolin for chlorpyrifos neurotoxicity in BV2 cells. Neurotoxicology. 2019;74:74–80. doi: 10.1016/j.neuro.2019.05.010. [DOI] [PubMed] [Google Scholar]
  • 27.Turovskaya M.V., Gaidin S.G., Mal’tseva V.N., Zinchenko V.P., Turovsky E.A. Taxifolin protects neurons against ischemic injury in vitro via the activation of antioxidant systems and signal transduction pathways of GABAergic neurons. Mol. Cell. Neurosci. 2019;96:10–24. doi: 10.1016/j.mcn.2019.01.005. [DOI] [PubMed] [Google Scholar]
  • 28.Ding T., Wang S., Zhang X., Zai W., Fan J., Chen W., Bian Q., Luan J., Shen Y., Zhang Y., Ju D., Mei X. Kidney protection effects of dihydroquercetin on diabetic nephropathy through suppressing ROS and NLRP3 inflammasome. Phytomedicine. 2018;41:45–53. doi: 10.1016/j.phymed.2018.01.026. [DOI] [PubMed] [Google Scholar]
  • 29.Kuang H., Tang Z., Zhang C., Wang Z., Li W., Yang C., Wang Q., Yang B., Kong A.N. Taxifolin activates the Nrf2 anti-oxidative stress pathway in mouse skin epidermal JB6 P+ cells through epigenetic modifications. Int. J. Mol. Sci. 2017;18(7):1546. doi: 10.3390/ijms18071546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ye Y., Wang X., Cai Q., Zhuang J., Tan X., He W. Zhao. M. Protective effect of taxifolin on H2O2-induced H9C2 cell pyroptosis. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2017;42(12):1367–1374. doi: 10.11817/j.issn.1672-7347.2017.12.003. [DOI] [PubMed] [Google Scholar]
  • 31.Li Z., Yu Y., Li Y., Ma F., Fang Y., Ni C., Wu K., Pan P., Ge R.S. Taxifolin attenuates the developmental testicular toxicity induced by di-n-butyl phthalate in fetal male rats. Food Chem. Toxicol. 2020;142:111482. doi: 10.1016/j.fct.2020.111482. [DOI] [PubMed] [Google Scholar]
  • 32.Yang C.L., Lin Y.S., Liu K.F., Peng W.H., Hsu C.M. Hepatoprotective mechanisms of taxifolin on carbon tetrachloride-induced acute liver injury in mice. Nutrients. 2019;11(11):2655. doi: 10.3390/nu11112655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Unver E., Tosun M., Olmez H., Kuzucu M., Cimen F.K., Suleyman Z. The effect of taxifolin on cisplatin-induced pulmonary damage in rats: A biochemical and histopathological evaluation. Mediators Inflamm. 2019;2019:1–6. doi: 10.1155/2019/3740867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kurt N., Gunes O., Suleyman B., Bakan N. The effect of taxifolin on high-dose-cisplatin-induced oxidative liver injury in rats. Adv. Clin. Exp. Med. 2021;30(10):1025–1030. doi: 10.17219/acem/138318. [DOI] [PubMed] [Google Scholar]
  • 35.Cao X., Bi R., Hao J., Wang S., Huo Y., Demoz R.M., Banda R., Tian S., Xin C., Fu M., Pi J., Liu J. A study on the protective effects of taxifolin on human umbilical vein endothelial cells and THP-1 cells damaged by hexavalent chromium: A probable mechanism for preventing cardiovascular disease induced by heavy metals. Food Funct. 2020;11(5):3851–3859. doi: 10.1039/D0FO00567C. [DOI] [PubMed] [Google Scholar]
  • 36.Xie X., Feng J., Kang Z., Zhang S., Zhang L., Zhang Y., Li X., Tang Y. Taxifolin protects RPE cells against oxidative stressinduced apoptosis. Mol. Vis. 2017;23:520–528. [PMC free article] [PubMed] [Google Scholar]
  • 37.Cai J., Shi G., Zhang Y., Zheng Y., Yang J., Liu Q., Gong Y., Yu D., Zhang Z. Taxifolin ameliorates DEHP-induced cardiomyocyte hypertrophy via attenuating mitochondrial dysfunction and glycometabolism disorder in chicken. Environ. Pollut. 2019;255(Pt 1):113155. doi: 10.1016/j.envpol.2019.113155. [DOI] [PubMed] [Google Scholar]
  • 38.Hu C., Ye J., Zhao L., Li X., Wang Y., Liu X., Pan L., You L., Chen L., Jia Y., Zhang J. 5,7,3′4′-flavan-on-ol (taxifolin) protects against acetaminophen-induced liver injury by regulating the glutathione pathway. Life Sci. 2019;236:116939. doi: 10.1016/j.lfs.2019.116939. [DOI] [PubMed] [Google Scholar]
  • 39.Hämäläinen M., Nieminen R., Asmawi M., Vuorela P., Vapaatalo H., Moilanen E. Effects of flavonoids on prostaglandin E2 production and on COX-2 and mPGES-1 expressions in activated macrophages. Planta Med. 2011;77(13):1504–1511. doi: 10.1055/s-0030-1270762. [DOI] [PubMed] [Google Scholar]
  • 40.Kwon J.H., Kim S.B., Park K.H., Lee M.W. Antioxidative and anti-inflammatory effects of phenolic compounds from the roots of Ulmus macrocarpa. Arch. Pharm. Res. 2011;34(9):1459–1466. doi: 10.1007/s12272-011-0907-4. [DOI] [PubMed] [Google Scholar]
  • 41.Kim Y.J., Choi S.E., Lee M.W., Lee C.S. Taxifolin glycoside inhibits dendritic cell responses stimulated by lipopolysaccharide and lipoteichoic acid. J. Pharm. Pharmacol. 2010;60(11):1465–1472. doi: 10.1211/jpp.60.11.0007. [DOI] [PubMed] [Google Scholar]
  • 42.Yuan X., Li N., Zhang M., Lu C., Du Z., Zhu W., Wu D. Taxifolin attenuates IMQ-induced murine psoriasis-like dermatitis by regulating T helper cell responses via Notch1 and JAK2/STAT3 signal pathways. Biomed. Pharmacother. 2020;123:109747. doi: 10.1016/j.biopha.2019.109747. [DOI] [PubMed] [Google Scholar]
  • 43.Lektemur A.A., Kızıldağ A., Özdede M., Karakan N.C., Özmen Ö. The effects of taxifolin on alveolar bone in experimental periodontitis in rats. Arch. Oral Biol. 2020;117:104823. doi: 10.1016/j.archoralbio.2020.104823. [DOI] [PubMed] [Google Scholar]
  • 44.Salama S.A., Kabel A.M. Taxifolin ameliorates iron overload-induced hepatocellular injury: Modulating PI3K/AKT and p38 MAPK signaling, inflammatory response, and hepatocellular regeneration. Chem. Biol. Interact. 2020;330:109230. doi: 10.1016/j.cbi.2020.109230. [DOI] [PubMed] [Google Scholar]
  • 45.Rehman K., Chohan T.A., Waheed I., Gilani Z., Akash M.S.H. Taxifolin prevents postprandial hyperglycemia by regulating the activity of α‐amylase: Evidence from an in vivo and in silico studies. J. Cell. Biochem. 2019;120(1):425–438. doi: 10.1002/jcb.27398. [DOI] [PubMed] [Google Scholar]
  • 46.Zhao Y., Huang W., Wang J., Chen Y., Huang W., Zhu Y. Taxifolin attenuates diabetic nephropathy in streptozotocin-induced diabetic rats. Am. J. Transl. Res. 2018;10(4):1205–1210. [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang Y., Jin Q., Li X., Jiang M., Cui B.W., Xia K.L., Wu Y.L., Lian L.H., Nan J.X. Amelioration of alcoholic liver steatosis by dihydroquercetin through the modulation of AMPK-dependent lipogenesis mediated by P2X7R-NLRP3-inflammasome activation. J. Agric. Food Chem. 2018;66(19):4862–4871. doi: 10.1021/acs.jafc.8b00944. [DOI] [PubMed] [Google Scholar]
  • 48.Gao L., Yuan P., Zhang Q., Fu Y., Hou Y., Wei Y., Zheng X., Feng W. Taxifolin improves disorders of glucose metabolism and water-salt metabolism in kidney via PI3K/AKT signaling pathway in metabolic syndrome rats. Life Sci. 2020;263:118713. doi: 10.1016/j.lfs.2020.118713. [DOI] [PubMed] [Google Scholar]
  • 49.Liu X., Liu W., Ding C., Zhao Y., Chen X., Ling D., Zheng Y., Cheng Z. Taxifolin, extracted from waste larix olgensis roots, attenuates CCl4-induced liver fibrosis by regulating the PI3K/AKT/] mTOR and TGF-β1/smads signaling pathways. Drug Des. Devel. Ther. 2021;15:871–887. doi: 10.2147/DDDT.S281369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Galochkina A.V., Zarubaev V.V., Kiselev O.I., Babkin V.A., Ostroukhova L.A. Antiviral activity of the dihydroquercetin during the Coxsackievirus В4 replication in vitro. Probl. Virol. 2016;61(1):27–31. doi: 10.18821/0507-4088-2016-61-1-27-31. [DOI] [PubMed] [Google Scholar]
  • 51.Al-Karmalawy A.A., Farid M.M., Mostafa A., Ragheb A.Y., H Mahmoud S., Shehata M., Shama N.M.A., GabAllah M., Mostafa-Hedeab G., Marzouk M.M. Naturally available flavonoid aglycones as potential antiviral drug candidates against SARS-CoV-2. Molecules. 2021;26(21):6559. doi: 10.3390/molecules26216559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kabel A.M., Salama S.A. Effect of taxifolin/dapagliflozin combination on colistin-induced nephrotoxicity in rats. Hum. Exp. Toxicol. 2021;40(10):1767–1780. doi: 10.1177/09603271211010906. [DOI] [PubMed] [Google Scholar]
  • 53.Hou J., Hu M., Zhang L., Gao Y., Ma L., Xu Q. Dietary taxifolin protects against dextran sulfate sodium-induced colitis via NF-κB signaling, enhancing intestinal barrier and modulating gut microbiota. Front. Immunol. 2021;11:631809. doi: 10.3389/fimmu.2020.631809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang L., Wang G., Qu H., Wang K., Jing S., Guan S., Su L., Li Q., Wang D. Taxifolin, an inhibitor of sortase a, interferes with the adhesion of methicillin-resistant Staphylococcal aureus. Front. Microbiol. 2021;12:686864. doi: 10.3389/fmicb.2021.686864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Stenger M.F.C., Cechinel-Filho V., Greco F.A., Venzon L., Meurer M.C., França T.C.S., Longo B., Somensi L.B., Mariano L.N.B., Cruz A.B., Macchiarulo A., Schoubben A., Ricci M., Belle B.T.M., da Silva L.M. Taxifolin and gastro-adhesive microparticles containing taxifolin promotes gastric healing in vivo, inhibits Helicobacter pylori in vitro and proton pump reversibly in silico. Chem. Biol. Interact. 2021;339:109445. doi: 10.1016/j.cbi.2021.109445. [DOI] [PubMed] [Google Scholar]
  • 56.Moon S.H., Lee C.M., Nam M.J. Cytoprotective effects of taxifolin against cadmium-induced apoptosis in human keratinocytes. Hum. Exp. Toxicol. 2019;38(8):992–1003. doi: 10.1177/0960327119846941. [DOI] [PubMed] [Google Scholar]
  • 57.Wang Y.J., Zhang H.Q., Han H.L., Zou Y.Y., Gao Q.L., Yang G.T. Taxifolin enhances osteogenic differentiation of human bone marrow mesenchymal stem cells partially via NF-κB pathway. Biochem. Biophys. Res. Commun. 2017;490(1):36–43. doi: 10.1016/j.bbrc.2017.06.002. [DOI] [PubMed] [Google Scholar]
  • 58.Tang Z., Yang C., Zuo B., Zhang Y., Wu G., Wang Y., Wang Z. Taxifolin protects rat against myocardial ischemia/reperfusion injury by modulating the mitochondrial apoptosis pathway. PeerJ. 2019;7:e6383. doi: 10.7717/peerj.6383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Živković L., Bajić V., Topalović D., Bruić M., Spremo-Potparević B. Antigenotoxic effects of biochaga and dihydroquercetin (Taxifolin) on H2O2-induced dna damage in human whole blood cells. Oxid. Med. Cell. Longev. 2019;2019:1–8. doi: 10.1155/2019/5039372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rajnochová Svobodová A., Ryšavá A., Psotová M., Kosina P., Zálešák B., Ulrichová J., Vostálová J. The phototoxic potential of the flavonoids, taxifolin and quercetin. Photochem. Photobiol. 2017;93(5):1240–1247. doi: 10.1111/php.12755. [DOI] [PubMed] [Google Scholar]
  • 61.Amalinei R.L., Trifan A., Cioanca O., Miron S.D., Mihai C.T., Rotinberg P., Miron A. Polyphenol-rich extract from Pinus sylvestris L. bark-chemical and antitumor studies. Rev. Med. Chir. Soc. Med. Nat. Iasi. 2014;118(2):551–557. [PubMed] [Google Scholar]
  • 62.Jomová K., Hudecova L., Lauro P., Simunkova M., Alwasel S.H., Alhazza I.M., Valko M. A switch between antioxidant and prooxidant properties of the phenolic compounds myricetin, morin, 3′,4′-dihydroxyflavone, taxifolin and 4-hydroxy-coumarin in the presence of copper(II) ions: A spectroscopic, absorption titration and dna damage study. Molecules. 2019;24(23):4335. doi: 10.3390/molecules24234335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Haque M.W., Bose P., Siddique M.U.M., Sunita P., Lapenna A., Pattanayak S.P. Taxifolin binds with LXR (α & β) to attenuate DMBA-induced mammary carcinogenesis through mTOR/Maf-1/] PTEN pathway. Biomed. Pharmacother. 2018;105:27–36. doi: 10.1016/j.biopha.2018.05.114. [DOI] [PubMed] [Google Scholar]
  • 64.Ge F., Tian E., Wang L., Li X., Zhu Q., Wang Y., Zhong Y., Ge R.S. Taxifolin suppresses rat and human testicular androgen biosynthetic enzymes. Fitoterapia. 2018;125:258–265. doi: 10.1016/j.fitote.2018.01.017. [DOI] [PubMed] [Google Scholar]
  • 65.Chen H.J., Chung Y.L., Li C.Y., Chang Y.T., Wang C., Lee H.Y., Lin H.Y., Hung C.C. Taxifolin resensitizes multidrug resistance cancer cells via uncompetitive inhibition of p-glycoprotein function. Molecules. 2018;23(12):3055. doi: 10.3390/molecules23123055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Butt S.S., Khan K., Badshah Y., Rafiq M., Shabbir M. Evaluation of pro-apoptotic potential of taxifolin against liver cancer. PeerJ. 2021;9:e11276. doi: 10.7717/peerj.11276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wang R., Zhu X., Wang Q., Li X., Wang E., Zhao Q., Wang Q., Cao H. The anti-tumor effect of taxifolin on lung cancer via suppressing stemness and epithelial-mesenchymal transition in vitro and oncogenesis in nude mice. Ann. Transl. Med. 2020;8(9):590. doi: 10.21037/atm-20-3329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Haque M.W., Pattanayak S.P. Taxifolin inhibits 7,12-dimethylbenz(a)anthracene-induced breast carcinogenesis by regulating AhR/CYP1A1 signaling pathway. Pharmacogn. Mag. 2018;13:S749–S755. doi: 10.4103/pm.pm_315_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Oi N., Chen H., Ok Kim M., Lubet R.A., Bode A.M., Dong Z. Taxifolin suppresses UV-induced skin carcinogenesis by targeting EGFR and PI3K. Cancer Prev. Res. 2012;5(9):1103–1114. doi: 10.1158/1940-6207.CAPR-11-0397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Xie J., Pang Y., Wu X. Taxifolin suppresses the malignant progression of gastric cancer by regulating the AhR/CYP1A1 signaling pathway. Int. J. Mol. Med. 2021;48(5):197. doi: 10.3892/ijmm.2021.5030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Razak S., Afsar T., Ullah A., Almajwal A., Alkholief M., Alshamsan A., Jahan S. Taxifolin, a natural flavonoid interacts with cell cycle regulators causes cell cycle arrest and causes tumor regression by activating Wnt/β -catenin signaling pathway. BMC Cancer. 2018;18(1):1043. doi: 10.1186/s12885-018-4959-4. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 72.Ahiskali I., Pinar C.L., Kiki M., Mammadov R., Ozbek Bilgin A., Hacimuftuoglu A., Cankaya M., Keskin Cimen F., Altuner D. Effect of taxifolin on development of retinopathy in alloxan-induced diabetic rats. Cutan. Ocul. Toxicol. 2019;38(3):227–232. doi: 10.1080/15569527.2019.1588289. [DOI] [PubMed] [Google Scholar]
  • 73.Su H., Ruan Y.T., Li Y., Chen J.G., Yin Z.P., Zhang Q.F. in vitro and in vivo inhibitory activity of taxifolin on three digestive enzymes. Int. J. Biol. Macromol. 2020;150:31–37. doi: 10.1016/j.ijbiomac.2020.02.027. [DOI] [PubMed] [Google Scholar]
  • 74.Yoon K.D., Lee J.Y., Kim T.Y., Kang H., Ha K.S., Ham T.H., Ryu S.N., Kang M.Y., Kim Y.H., Kwon Y.I. In vitro and in vivo anti-hyperglycemic activities of taxifolin and its derivatives isolated from pigmented rice (Oryzae sativa L. cv. Superhongmi). J. Agric. Food Chem. 2020;68(3):742–750. doi: 10.1021/acs.jafc.9b04962. [DOI] [PubMed] [Google Scholar]
  • 75.Harris C., Beaulieu L.P., Fraser M.H., McIntyre K., Owen P., Martineau L., Cuerrier A., Johns T., Haddad P., Bennett S., Arnason J. Inhibition of advanced glycation end product formation by medicinal plant extracts correlates with phenolic metabolites and antioxidant activity. Planta Med. 2011;77(2):196–204. doi: 10.1055/s-0030-1250161. [DOI] [PubMed] [Google Scholar]
  • 76.Muramatsu D., Uchiyama H., Kida H., Iwai A. Cell cytotoxity and anti-glycation activity of taxifolin-rich extract from Japanese larch, Larix kaempferi. Heliyon. 2019;5(7):e02047. doi: 10.1016/j.heliyon.2019.e02047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Adachi S., Nihei K., Ishihara Y., Yoshizawa F., Yagasaki K. Anti-hyperuricemic effect of taxifolin in cultured hepatocytes and model mice. Cytotechnology. 2017;69(2):329–336. doi: 10.1007/s10616-016-0061-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wu C., Cao S., Hong T., Dong Y., Li C., Wang Q., Sun J., Ge R.S. Taxifolin inhibits rat and human 11β-hydroxysteroid dehydrogenase 2. Fitoterapia. 2017;121:112–117. doi: 10.1016/j.fitote.2017.07.004. [DOI] [PubMed] [Google Scholar]
  • 79.Zhang H.Q., Wang Y.J., Yang G.T., Gao Q.L., Tang M.X. Taxifolin inhibits receptor activator of NF-κB Ligand-induced osteoclastogenesis of human bone marrow-derived macrophages in vitro and prevents lipopolysaccharide-induced bone loss in vivo. Pharmacology. 2019;103(1-2):101–109. doi: 10.1159/000495254. [DOI] [PubMed] [Google Scholar]
  • 80.Muramatsu D., Uchiyama H., Kida H., Iwai A. in vitro anti-inflammatory and anti-lipid accumulation properties of taxifolinrich extract from the Japanese larch, Larix kaempferi. Heliyon. 2020;6(12):e05505. doi: 10.1016/j.heliyon.2020.e05505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Su H., Wang W.J., Zheng G.D., Yin Z.P., Li J.E., Chen L.L., Zhang Q.F. The anti‐obesity and gut microbiota modulating effects of taxifolin in C57BL / 6J mice fed with a high‐fat diet. J. Sci. Food Agric. 2022;102(4):1598–1608. doi: 10.1002/jsfa.11496. [DOI] [PubMed] [Google Scholar]
  • 82.Bucak M.N., Keskin N., Ili P., Bodu M., Akalın P.P., Öztürk A.E., Özkan H., Topraggaleh T.R., Sari F., Başpınar N., Dursun Ş. Decreasing glycerol content by co-supplementation of trehalose and taxifolin hydrate in ram semen extender: Microscopic, oxidative stress, and gene expression analyses. Cryobiology. 2020;96:19–29. doi: 10.1016/j.cryobiol.2020.09.001. [DOI] [PubMed] [Google Scholar]
  • 83.Di T., Zhai C., Zhao J., Wang Y., Chen Z., Li P. Taxifolin inhibits keratinocyte proliferation and ameliorates imiquimod-induced psoriasis-like mouse model via regulating cytoplasmic phospholipase A2 and PPAR-γ pathway. Int. Immunopharmacol. 2021;99:107900. doi: 10.1016/j.intimp.2021.107900. [DOI] [PubMed] [Google Scholar]
  • 84.Pan S., Zhao X., Ji N., Shao C., Fu B., Zhang Z., Wang R., Qiu Y., Jin M., Kong D. Inhibitory effect of taxifolin on mast cell activation and mast cell-mediated allergic inflammatory response. Int. Immunopharmacol. 2019;71:205–214. doi: 10.1016/j.intimp.2019.03.038. [DOI] [PubMed] [Google Scholar]
  • 85.Micek I., Nawrot J., Seraszek-Jaros A., Jenerowicz D., Schroeder G., Spiżewski T., Suchan A., Pawlaczyk M., Gornowicz-Porowska J. Taxifolin as a promising ingredient of cosmetics for adult skin. Antioxidants. 2021;10(10):1625. doi: 10.3390/antiox10101625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shubina V.S., Shatalin Y.V. Skin regeneration after chemical burn under the effect of taxifolin-based preparations. Bull. Exp. Biol. Med. 2012;154(1):152–157. doi: 10.1007/s10517-012-1897-z. [DOI] [PubMed] [Google Scholar]
  • 87.Liu X., Ma Y., Luo L., Zong D., Li H., Zeng Z., Cui Y., Meng W., Chen Y. Dihydroquercetin suppresses cigarette smoke induced ferroptosis in the pathogenesis of chronic obstructive pulmonary disease by activating Nrf2-mediated pathway. Phytomedicine. 2022;96:153894. doi: 10.1016/j.phymed.2021.153894. [DOI] [PubMed] [Google Scholar]
  • 88.Chowdhury S., Bhuiya S., Haque L., Das S. In-depth investigation of the binding of flavonoid taxifolin with bovine hemoglobin at physiological pH: Spectroscopic and molecular docking studies. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2020;225:117513. doi: 10.1016/j.saa.2019.117513. [DOI] [PubMed] [Google Scholar]
  • 89.Kozhikkadan Davis C., Nasla K., Anjana A.K., Rajanikant G.K. Taxifolin as dual inhibitor of Mtb DNA gyrase and isoleucyl-tRNA synthetase: in silico molecular docking, dynamics simulation and in vitro assays. In Silico Pharmacol. 2018;6(1):8. doi: 10.1007/s40203-018-0045-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lee C.W., Park N.H., Kim J.W., Um B.H., Shpatov A.V., Shults E.E., Sorokina I.V., Popov S.A. Study of skin anti-ageing and anti-inflammatory effects of dihydroquercetin, natural triterpenoinds, and their synthetic derivatives. Russ. J. Bioorganic Chem. 2012;38(3):328–334. doi: 10.1134/S1068162012030028. [DOI] [PubMed] [Google Scholar]
  • 91.Muraleva N.A., Ofitserov E.N., Tikhonov V.P., Kolosova N.G. Efficacy of glucosamine alendronate alone & in combination with dihydroquercetin for treatment of osteoporosis in animal model. Indian J. Med. Res. 2012;135(2):221–227. [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhang Z.R., Al Zaharna M., Wong M.M.K., Chiu S.K., Cheung H.Y. Taxifolin enhances andrographolide-induced mitotic arrest and apoptosis in human prostate cancer cells via spindle assembly checkpoint activation. PLoS One. 2013;8(1):e54577. doi: 10.1371/journal.pone.0054577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pirgozliev V., Mansbridge S.C., Whiting I.M., Arthur C., Rose S.P., Atanasov A.G. Antioxidant status and growth performance of broiler chickens fed diets containing graded levels of supplementary dihydroquercetin. Res. Vet. Sci. 2021;141:63–65. doi: 10.1016/j.rvsc.2021.10.001. [DOI] [PubMed] [Google Scholar]
  • 94.Awad E., Awaad A.S., Esteban M.A. Effects of dihydroquercetin obtained from deodar (Cedrus deodara) on immune status of gilthead seabream (Sparus aurata L.). Fish Shellfish Immunol. 2015;43(1):43–50. doi: 10.1016/j.fsi.2014.12.009. [DOI] [PubMed] [Google Scholar]
  • 95.Gopikrishnan V., Radhakrishnan M., Shanmugasundaram T., Ramakodi M.P., Balagurunathan R. Isolation, characterization and identification of antibiofouling metabolite from mangrove derived Streptomyces sampsonii PM33. Sci. Rep. 2019;9(1):12975. doi: 10.1038/s41598-019-49478-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Okkay U., Ferah Okkay I., Cicek B., Aydin I.C., Ozkaraca M. Hepatoprotective and neuroprotective effect of taxifolin on hepatic encephalopathy in rats. Metab. Brain Dis. 2022;37(5):1541–1556. doi: 10.1007/s11011-022-00952-3. [DOI] [PubMed] [Google Scholar]
  • 97.Kalia L.V., Lang A.E. Parkinson’s disease. Lancet. 2015;386(9996):896–912. doi: 10.1016/S0140-6736(14)61393-3. [DOI] [PubMed] [Google Scholar]
  • 98.Cabreira V., Massano J. Parkinson’s disease: Clinical review and update. Acta Med. Port. 2019;32(10):661–670. doi: 10.20344/amp.11978. [DOI] [PubMed] [Google Scholar]
  • 99.Tysnes O.B., Storstein A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017;124(8):901–905. doi: 10.1007/s00702-017-1686-y. [DOI] [PubMed] [Google Scholar]
  • 100.Nakaoka S., Ishizaki T., Urushihara H., Satoh T., Ikeda S., Yamamoto M., Nakayama T. Prescribing pattern of anti-Parkinson drugs in Japan: A trend analysis from 2005 to 2010. PLoS One. 2014;9(6):e99021. doi: 10.1371/journal.pone.0099021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pajares M., I Rojo A., Manda G., Boscá L., Cuadrado A. Inflammation in parkinson’s disease: Mechanisms and therapeutic implications. Cells. 2020;9(7):1687. doi: 10.3390/cells9071687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yasuhara T. Neurobiology research in parkinson’s disease. Int. J. Mol. Sci. 2020;21(3):793. doi: 10.3390/ijms21030793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Nam Y.J., Lee D.H., Shin Y.K., Sohn D.S., Lee C.S. Flavanonol taxifolin attenuates proteasome inhibition-induced apoptosis in differentiated PC12 cells by suppressing cell death process. Neurochem. Res. 2015;40(3):480–491. doi: 10.1007/s11064-014-1493-x. [DOI] [PubMed] [Google Scholar]
  • 104.Pérez-H J., Carrillo-S C., García E., Ruiz-Mar G., Pérez-Tamayo R., Chavarría A. Neuroprotective effect of silymarin in a MPTP mouse model of Parkinson's disease. Toxicology. 2014;319:38–43. doi: 10.1016/j.tox.2014.02.009. [DOI] [PubMed] [Google Scholar]
  • 105.Kim A., Nam Y.J., Lee C.S. Taxifolin reduces the cholesterol oxidation product-induced neuronal apoptosis by suppressing the Akt and NF-κB activation-mediated cell death. Brain Res. Bull. 2017;134:63–71. doi: 10.1016/j.brainresbull.2017.07.008. [DOI] [PubMed] [Google Scholar]
  • 106.Abushouk A.I., Negida A., Ahmed H., Abdel-Daim M.M. Neuroprotective mechanisms of plant extracts against MPTP induced neurotoxicity: Future applications in Parkinson’s disease. Biomed. Pharmacother. 2017;85:635–645. doi: 10.1016/j.biopha.2016.11.074. [DOI] [PubMed] [Google Scholar]
  • 107.Dok-Go H., Lee K.H., Kim H.J., Lee E.H., Lee J., Song Y.S., Lee Y.H., Jin C., Lee Y.S., Cho J. Neuroprotective effects of antioxidative flavonoids, quercetin, (+)-dihydroquercetin and quercetin 3-methyl ether, isolated from Opuntia ficus-indica var. saboten. Brain Res. 2003;965(1-2):130–136. doi: 10.1016/S0006-8993(02)04150-1. [DOI] [PubMed] [Google Scholar]
  • 108.Xu Q., Park Y., Huang X., Hollenbeck A., Blair A., Schatzkin A., Chen H. Diabetes and risk of Parkinson’s disease. Diabetes Care. 2011;34(4):910–915. doi: 10.2337/dc10-1922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wirdefeldt K., Weibull C.E., Chen H., Kamel F., Lundholm C., Fang F., Ye W. Parkinson’s disease and cancer: A register-based family study. Am. J. Epidemiol. 2014;179(1):85–94. doi: 10.1093/aje/kwt232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.An S.M., Kim H.J., Kim J.E., Boo Y.C. Flavonoids, taxifolin and luteolin attenuate cellular melanogenesis despite increasing tyrosinase protein levels. Phytother. Res. 2008;22(9):1200–1207. doi: 10.1002/ptr.2435. [DOI] [PubMed] [Google Scholar]
  • 111.Liu T., Zhou N., Cao Y., Xu R., Liu Z., Zheng X., Feng W. Investigating the basis for the antidepressant effects of Gleditsiae spina using an integrated metabolomic strategy. Iran. J. Basic Med. Sci. 2021;24(4):524–530. doi: 10.22038/ijbms.2021.51975.11781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ballard C., Gauthier S., Corbett A., Brayne C., Aarsland D., Jones E. Alzheimer’s disease. Lancet. 2011;377(9770):1019–1031. doi: 10.1016/S0140-6736(10)61349-9. [DOI] [PubMed] [Google Scholar]
  • 113.Selkoe D.J. Alzheimer’s disease. Cold Spring Harb. Perspect. Biol. 2011;3(7):a004457. doi: 10.1101/cshperspect.a004457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Heneka M.T., Carson M.J., Khoury J.E., Landreth G.E., Brosseron F., Feinstein D.L., Jacobs A.H., Wyss-Coray T., Vitorica J., Ransohoff R.M., Herrup K., Frautschy S.A., Finsen B., Brown G.C., Verkhratsky A., Yamanaka K., Koistinaho J., Latz E., Halle A., Petzold G.C., Town T., Morgan D., Shinohara M.L., Perry V.H., Holmes C., Bazan N.G., Brooks D.J., Hunot S., Joseph B., Deigendesch N., Garaschuk O., Boddeke E., Dinarello C.A., Breitner J.C., Cole G.M., Golenbock D.T., Kummer M.P. Neuroinflammation in alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405. doi: 10.1016/S1474-4422(15)70016-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mintun M.A., Lo A.C., Duggan E.C., Wessels A.M., Ardayfio P.A., Andersen S.W., Shcherbinin S., Sparks J., Sims J.R., Brys M., Apostolova L.G., Salloway S.P., Skovronsky D.M. Donanemab in early Alzheimer’s disease. N. Engl. J. Med. 2021;384(18):1691–1704. doi: 10.1056/NEJMoa2100708. [DOI] [PubMed] [Google Scholar]
  • 116.Akiyama H., Barger S., Barnum S., Bradt B., Bauer J., Cole G.M., Cooper N.R., Eikelenboom P., Emmerling M., Fiebich B.L., Finch C.E., Frautschy S., Griffin W.S., Hampel H., Hull M., Landreth G., Lue L., Mrak R., Mackenzie I.R., McGeer P.L., O’Banion M.K., Pachter J., Pasinetti G., Plata-Salaman C., Rogers J., Rydel R., Shen Y., Streit W., Strohmeyer R., Tooyoma I., Van Muiswinkel F.L., Veerhuis R., Walker D., Webster S., Wegrzyniak B., Wenk G., Wyss-Coray T. Inflammation and alzheimer’s disease. Neurobiol. Aging. 2000;21(3):383–421. doi: 10.1016/S0197-4580(00)00124-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Bate C., Williams A. cAMP-inhibits cytoplasmic phospholipase A2 and protects neurons against amyloid-β-induced synapse damage. Biology. 2015;4(3):591–606. doi: 10.3390/biology4030591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Bate C., Williams A. α-Synuclein-induced synapse damage in cultured neurons is mediated by cholesterol-sensitive activation of cytoplasmic phospholipase A2. Biomolecules. 2015;5(1):178–193. doi: 10.3390/biom5010178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Wang Y., Wang Q., Bao X., Ding Y., Shentu J., Cui W., Chen X., Wei X., Xu S. Taxifolin prevents β-amyloid-induced impairments of synaptic formation and deficits of memory via the inhibition of cytosolic phospholipase A2/prostaglandin E2 content. Metab. Brain Dis. 2018;33(4):1069–1079. doi: 10.1007/s11011-018-0207-5. [DOI] [PubMed] [Google Scholar]
  • 120.Ginex T., Trius M., Luque F.J. Computational study of the azamichael addition of the flavonoid (+)-taxifolin in the inhibition of β-amyloid fibril aggregation. Chemistry. 2018;24(22):5813–5824. doi: 10.1002/chem.201706072. [DOI] [PubMed] [Google Scholar]
  • 121.Mahdavimehr M., Meratan A.A., Ghobeh M., Ghasemi A., Saboury A.A., Nemat-Gorgani M. Inhibition of HEWL fibril formation by taxifolin: Mechanism of action. PLoS One. 2017;12(11):e0187841. doi: 10.1371/journal.pone.0187841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Mahdavimehr M., Katebi B., Meratan A.A. Effect of fibrillation conditions on the anti-amyloidogenic properties of polyphenols and their involved mechanisms. Int. J. Biol. Macromol. 2018;118(Pt A):552–560. doi: 10.1016/j.ijbiomac.2018.06.109. [DOI] [PubMed] [Google Scholar]
  • 123.Ambrose C.T. Neuroangiogenesis: A vascular basis for Alzheimer’s disease and cognitive decline during aging. J. Alzheimers Dis. 2012;32(3):773–788. doi: 10.3233/JAD-2012-120067. [DOI] [PubMed] [Google Scholar]
  • 124.Plotnikov M.B., Plotnikov D.M., Aliev O.I., Maslov M.Y., Vasiliev A.S., Alifirova V.M., Tyukavkina N.A. Hemorheological and antioxidant effects of Ascovertin in patients with sclerosis of cerebral arteries. Clin. Hemorheol. Microcirc. 2004;30(3-4):449–452. [PubMed] [Google Scholar]
  • 125.Plotnikov M.B., Plotnikov D.M., Alifirova V.M., Aliev O.I., Maslov M.I., Vasil’ev A.S., Tiukavkina N.A. Clinical efficacy of a novel hemorheological drug ascovertin in patients with vascular encephalopathy. Zh. Nevrol. Psikhiatr. Im. S. S. Korsakova. 2004;104(12):33–37. [PubMed] [Google Scholar]
  • 126.Gunesch S., Hoffmann M., Kiermeier C., Fischer W., Pinto A.F.M., Maurice T., Maher P., Decker M. 7-O-Esters of taxifolin with pronounced and overadditive effects in neuroprotection, anti-neuroinflammation, and amelioration of short-term memory impairment in vivo. Redox Biol. 2020;29:101378. doi: 10.1016/j.redox.2019.101378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Sato M., Murakami K., Uno M., Ikubo H., Nakagawa Y., Katayama S., Akagi K., Irie K. Structure-activity relationship for (+)-taxifolin isolated from silymarin as an inhibitor of amyloid β aggregation. Biosci. Biotechnol. Biochem. 2013;77(5):1100–1103. doi: 10.1271/bbb.120925. [DOI] [PubMed] [Google Scholar]
  • 128.Sato M., Murakami K., Uno M., Nakagawa Y., Katayama S., Akagi K., Masuda Y., Takegoshi K., Irie K. Site-specific inhibitory mechanism for amyloid β42 aggregation by catechol-type flavonoids targeting the Lys residues. J. Biol. Chem. 2013;288(32):23212–23224. doi: 10.1074/jbc.M113.464222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Das S., Majumder T., Sarkar A., Mukherjee P., Basu S. Flavonoids as BACE1 inhibitors: QSAR modelling, screening and in vitro evaluation. Int. J. Biol. Macromol. 2020;165(Pt A):1323–117. doi: 10.1016/j.ijbiomac.2020.09.232. [DOI] [PubMed] [Google Scholar]
  • 130.Park S.Y., Kim H.Y., Park H.J., Shin H.K., Hong K.W., Kim C.D. Concurrent treatment with taxifolin and cilostazol on the lowering of β-amyloid accumulation and neurotoxicity via the suppression of P-JAK2/P-STAT3/NF-κB/BACE1 signaling pathways. PLoS One. 2016;11(12):e0168286. doi: 10.1371/journal.pone.0168286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Inoue T., Saito S., Tanaka M., Yamakage H., Kusakabe T., Shimatsu A., Ihara M., Satoh-Asahara N. Pleiotropic neuroprotective effects of taxifolin in cerebral amyloid angiopathy. Proc. Natl. Acad. Sci. USA. 2019;116(20):10031–10038. doi: 10.1073/pnas.1901659116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Huang Y.W.A., Zhou B., Wernig M., Südhof T.C. ApoE2, ApoE3, and ApoE4 differentially stimulate APP transcription and Aβ secretion. Cell. 2017;168(3):427–441.e21. doi: 10.1016/j.cell.2016.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Gocer H., Topal F., Topal M., Küçük M., Teke D., Gülçin İ., Alwasel S.H., Supuran C.T. Acetylcholinesterase and carbonic anhydrase isoenzymes I and II inhibition profiles of taxifolin. J. Enzyme Inhib. Med. Chem. 2015;31(3):1–7. doi: 10.3109/14756366.2015.1036051. [DOI] [PubMed] [Google Scholar]
  • 134.Kim J.W., Im S., Jeong H.R., Jung Y.S., Lee I., Kim K.J., Park S.K., Kim D.O. Neuroprotective effects of korean red pine (Pinus densiflora) bark extract and its phenolics. J. Microbiol. Biotechnol. 2018;28(5):679–687. doi: 10.4014/jmb.1801.01053. [DOI] [PubMed] [Google Scholar]
  • 135.Weber S.A., Patel R.K., Lutsep H.L. Cerebral amyloid angiopathy: Diagnosis and potential therapies. Expert Rev. Neurother. 2018;18(6):503–513. doi: 10.1080/14737175.2018.1480938. [DOI] [PubMed] [Google Scholar]
  • 136.Kozberg M.G., Perosa V., Gurol M.E., van Veluw S.J. A practical approach to the management of cerebral amyloid angiopathy. Int. J. Stroke. 2021;16(4):356–369. doi: 10.1177/1747493020974464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Jäkel L., Van Nostrand W.E., Nicoll J.A.R., Werring D.J., Verbeek M.M. Animal models of cerebral amyloid angiopathy. Clin. Sci. 2017;131(19):2469–2488. doi: 10.1042/CS20170033. [DOI] [PubMed] [Google Scholar]
  • 138.Greenberg S.M., Bacskai B.J., Hernandez-Guillamon M., Pruzin J., Sperling R., van Veluw S.J. Cerebral amyloid angiopathy and Alzheimer disease — one peptide, two pathways. Nat. Rev. Neurol. 2020;16(1):30–42. doi: 10.1038/s41582-019-0281-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Saito S., Yamamoto Y., Maki T., Hattori Y., Ito H., Mizuno K., Harada-Shiba M., Kalaria R.N., Fukushima M., Takahashi R., Ihara M. Taxifolin inhibits amyloid-β oligomer formation and fully restores vascular integrity and memory in cerebral amyloid angiopathy. Acta Neuropathol. Commun. 2017;5(1):26. doi: 10.1186/s40478-017-0429-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Saito S., Ihara M. New therapeutic approaches for Alzheimer’s disease and cerebral amyloid angiopathy. Front. Aging Neurosci. 2014;6:290. doi: 10.3389/fnagi.2014.00290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Saito S., Tanaka M., Satoh-Asahara N., Carare R.O., Ihara M. Taxifolin: A potential therapeutic agent for cerebral amyloid angiopathy. Front. Pharmacol. 2021;12:643357. doi: 10.3389/fphar.2021.643357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Park L., Koizumi K., El Jamal S., Zhou P., Previti M.L., Van Nostrand W.E., Carlson G., Iadecola C. Age-dependent neurovascular dysfunction and damage in a mouse model of cerebral amyloid angiopathy. Stroke. 2014;45(6):1815–1821. doi: 10.1161/STROKEAHA.114.005179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Fassler M., Rappaport M.S., Cuño C.B., George J. Engagement of TREM2 by a novel monoclonal antibody induces activation of microglia and improves cognitive function in Alzheimer’s disease models. J. Neuroinflammation. 2021;18(1):19. doi: 10.1186/s12974-020-01980-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Walker F.O. Huntington’s disease. Lancet. 2007;369(9557):218–228. doi: 10.1016/S0140-6736(07)60111-1. [DOI] [PubMed] [Google Scholar]
  • 145.McColgan P., Tabrizi S.J. Huntington’s disease: A clinical review. Eur. J. Neurol. 2018;25(1):24–34. doi: 10.1111/ene.13413. [DOI] [PubMed] [Google Scholar]
  • 146.Björkqvist M., Wild E.J., Thiele J., Silvestroni A., Andre R., Lahiri N., Raibon E., Lee R.V., Benn C.L., Soulet D., Magnusson A., Woodman B., Landles C., Pouladi M.A., Hayden M.R., Khalili-Shirazi A., Lowdell M.W., Brundin P., Bates G.P., Leavitt B.R., Möller T., Tabrizi S.J. A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. J. Exp. Med. 2008;205(8):1869–1877. doi: 10.1084/jem.20080178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Qi L., Zhang X.D., Wu J.C., Lin F., Wang J., DiFiglia M., Qin Z.H. The role of chaperone-mediated autophagy in huntingtin degradation. PLoS One. 2012;7(10):e46834. doi: 10.1371/journal.pone.0046834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wu Z., Parry M., Hou X.Y., Liu M.H., Wang H., Cain R., Pei Z.F., Chen Y.C., Guo Z.Y., Abhijeet S., Chen G. Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington’s disease. Nat. Commun. 2020;11(1):1105. doi: 10.1038/s41467-020-14855-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Maksimovich N.Y., Dremza I.K., Troyan E.I., Maksimovich Y.N., Borodinskiĭ A.N. The correcting effects of dihydroquercetin in cerebral ischemia-reperfusion injury. Biomed. Khim. 2014;60(6):643–650. doi: 10.18097/PBMC20146006643. [DOI] [PubMed] [Google Scholar]
  • 150.Stegner D., Klaus V., Nieswandt B. Platelets as modulators of cerebral ischemia/reperfusion injury. Front. Immunol. 2019;10:2505. doi: 10.3389/fimmu.2019.02505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Wang Y.H., Wang W.Y., Chang C.C., Liou K.T., Sung Y.J., Liao J.F., Chen C.F., Chang S., Hou Y.C., Chou Y.C., Shen Y.C. Taxifolin ameliorates cerebral ischemia-reperfusion injury in rats through its anti-oxidative effect and modulation of NF-kappa B activation. J. Biomed. Sci. 2006;13(1):127–141. doi: 10.1007/s11373-005-9031-0. [DOI] [PubMed] [Google Scholar]
  • 152.Shubina V.S., Shatalin Y.V. Antioxidant and iron-chelating properties of taxifolin and its condensation product with glyoxylic acid. J. Food Sci. Technol. 2017;54(6):1467–1475. doi: 10.1007/s13197-017-2573-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Murach E.I., Baranov I.A., Erlykina E.I., Koryagin A.S., Mochalova A.E., Smirnova L.A. Adaptogenic effects of dihydroquercetin-chitosan composition during modeling of acute hypoxia. Bull. Exp. Biol. Med. 2014;156(3):306–309. doi: 10.1007/s10517-014-2336-0. [DOI] [PubMed] [Google Scholar]
  • 154.Zu Y., Wu W., Zhao X., Li Y., Zhong C., Zhang Y. The high water solubility of inclusion complex of taxifolin-γ-CD prepared and characterized by the emulsion solvent evaporation and the freeze drying combination method. Int. J. Pharm. 2014;477(1-2):148–158. doi: 10.1016/j.ijpharm.2014.10.027. [DOI] [PubMed] [Google Scholar]
  • 155.Zinchenko V.P., Kim IuA., Tarakhovskiĭ IuS., Bronnikov G.E. Biological activity of water-soluble nanostructures of dihydroquercetin with cyclodextrins. Biofizika. 2011;56(3):433–438. [PubMed] [Google Scholar]
  • 156.Yang P., Xu F., Li H.F., Wang Y., Li F.C., Shang M.Y., Liu G.X., Wang X., Cai S.Q. Detection of 191 taxifolin metabolites and their distribution in rats using HPLC-ESI-IT-TOF-MSn. Molecules. 2016;21(9):1209. doi: 10.3390/molecules21091209. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES