Next Article in Journal
High-Performance One-Dimensional Sub-5 nm Transistors Based on Poly(p-phenylene ethynylene) Molecular Wires
Next Article in Special Issue
Upgrading the Bioactive Potential of Hazelnut Oil Cake by Aspergillus oryzae under Solid-State Fermentation
Previous Article in Journal
Enhancing Sustainable Analytical Chemistry in Liquid Chromatography: Guideline for Transferring Classical High-Performance Liquid Chromatography and Ultra-High-Pressure Liquid Chromatography Methods into Greener, Bluer, and Whiter Methods
Previous Article in Special Issue
Analysis of Polyphenol Extract from Hazel Leaf and Ameliorative Efficacy and Mechanism against Hyperuricemia Zebrafish Model via Network Pharmacology and Molecular Docking
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Quercetin as a Plant-Derived Bioactive Agent in Preventive Medicine and Treatment in Skin Disorders

1
Department of Food Industry Technology and Engineering, Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland
2
Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
*
Authors to whom correspondence should be addressed.
Molecules 2024, 29(13), 3206; https://doi.org/10.3390/molecules29133206
Submission received: 31 May 2024 / Revised: 30 June 2024 / Accepted: 4 July 2024 / Published: 5 July 2024
(This article belongs to the Special Issue Research and Application of Food By-Products, 2nd Edition)

Abstract

:
Quercetin, a bioactive plant flavonoid, is an antioxidant, and as such it exhibits numerous beneficial properties including anti-inflammatory, antiallergic, antibacterial and antiviral activity. It occurs naturally in fruit and vegetables such as apples, blueberries, cranberries, lettuce, and is present in plant waste such as onion peel or grape pomace which constitute good sources of quercetin for technological or pharmaceutical purposes. The presented study focuses on the role of quercetin in prevention and treatment of dermatological diseases analyzing its effect at a molecular level, its signal transduction and metabolism. Presented aspects of quercetin potential for skin treatment include protection against aging and UV radiation, stimulation of wound healing, reduction in melanogenesis, and prevention of skin oxidation. The article discusses quercetin sources (plant waste products included), methods of its medical administration, and perspectives for its further use in dermatology and diet therapy.

1. Introduction

The directions of development in the food industry are strictly connected to the dominating trends in the food sector. Currently, worldwide tendencies point to sustainable development, with a focus on lab-grown food. It involves transferring production from the natural world to the laboratories in the face of growing ecological challenges like environmental degradation, climate change, and shrinking natural resources. The trend reflects the increased awareness and expectations of customers regarding sustainable development, and is a potential solution to the issue of raw materials recovery in the situation of the global raw material shortage and the growing problems with waste management [1]. The food industry fits perfectly into the latest developments with its new technologies for utilizing waste materials or enriching food with compounds obtained from waste [2,3,4]. That, in turn, results in further advancements like the health-promoting qualities of food, or the reduction and/or management of byproducts, particularly those from plant production, which are used as the main source of health-benefitting bioactive compounds like polyphenols [5].
Quercetin (Que), 3,3′,4′,5,7-pentahydroxyflavone, is an organic compound in the group of flavonols which are a class of flavonoids. In nature, quercetin occurs as the aglycone of flavonoid glycosides forming isoquercetin with glucose, rutin with rutinose, hyperoside with galactose, and quercetrin with rhamnose (Figure 1).
Quercetin-3-O-glucoside, naturally occurring as a yellow plant pigment, is the most common Que glycoside [6]. The compound is sparingly soluble in water, and soluble in alcohol [7], lipids [8] and organic solvents [9]. Quercetin is not synthesized in the human body [10].
Figure 1. Structure of quercetin and its derivatives [11].
Figure 1. Structure of quercetin and its derivatives [11].
Molecules 29 03206 g001
Quercetin is widely distributed in many plant-derived products; foods high in quercetin include, e.g., onion, capers, grapes or berries. Depending on the dietary habits, some of them can be the main sources of quercetin. However, the concentration of bioactive compounds, including quercetin, depends on numerous factors: plant maturity, harvest time, and farming techniques [12,13,14,15]. It should be noted that byproducts contain up to several times more quercetin than edible parts of plants and therefore, waste may be a good source of quercetin and other bioactive compounds used in food, chemical, beauty, and pharmaceutical industries. Table 1 presents the concentration of quercetin in foods and food industry byproducts.
Quercetin is highly valued for the health benefits it offers (Figure 2). Its most important properties include the ability to neutralize free radicals, and its anticancer, anti-diabetic, anti-aging, antibacterial and anti-inflammatory effects observed even in cases of inflammation associated with chronic diseases [23,24,25,26,27,28,29,30,31,32]. Additionally, a positive impact on human skin has been confirmed in research [33,34,35,36]. Due to its beneficial health effects and the fact that it is easily available from plants and food industry byproducts, quercetin has the potential to be used in medicine, especially in disease prevention, treatment of skin diseases and injuries. Thus, the therapeutic potential of quercetin requires a summary providing an insight into the research conducted to date, especially into its impact on biochemical mechanisms and expression of enzymes and proteins, which is the main objective of this paper.
Figure 2. The biological activities of quercetin and its mechanism [37] (list of abbreviations used in Figure 2 and in the paper, with their descriptions, is in Table 2).
Figure 2. The biological activities of quercetin and its mechanism [37] (list of abbreviations used in Figure 2 and in the paper, with their descriptions, is in Table 2).
Molecules 29 03206 g002

2. Anti-Aging Effects and UV Protection

Quercetin prevents degradation of collagen due to UV radiation in human skin and inhibits MMP-1 and COX-2 expression. Quercetin inhibits UV-induced AP-1 activity and NF-κB (Figure 2). Additionally, quercetin may reduce phosphorylation of ERK, JNK, and AKT, and STAT3Kinase assays using purified protein demonstrated quercetin’s ability to directly inhibit the activity of PKCδ and JAK2. This suggests its possible direct interaction with PKCδ and JAK2 in the skin, counteracting UV-induced aging. [35]. Research by Vicentini et al. [38] confirmed reduction in skin irritation due to UV radiation by inhibition of NF-κB and such inflammatory cytokines as IL-1β, IL-6, IL-8, and TNF-α. Kim et al. [39] point to the fact that quercetin in propolis reduces PDK-1 and AKT phosphorylation, which suggests its efficacy in preventing UV-induced photoaging. Furthermore, when combined with caffeic acid ester and apigenin, quercetin reduces PI3K activity, further enhancing its protective effect.
Findings from the clinical study conducted by Nebus et al. [40], in which oak quercetin formulated into an SPF 15 protective cream was used, demonstrated its protective effect on collagen and elastin with sustained proper degradation of waste proteins in aging skin. Patients in the study observed considerable improvement in skin parameters such as wrinkling, elasticity, smoothness, skin radiance and moisture.
The anti-aging effects of quercetin and its derivative, quercetin caprylate, were demonstrated in studies conducted by Chondrogianni et al. [41]. The substances showed the ability to activate the proteasome, which is responsible for the degradation of damaged proteins in human cells. Its activation is crucial for increasing resistance to oxidative stress and improving the viability of HFL-1 fibroblasts. Both compounds contributed to the rejuvenation of aging fibroblasts. which indicates their potential as anti-aging ingredients.
The research into using quercetin for UV protection started with studies of its function in plant tissues [42,43]. Choquenet et al. [43] described the increased level of photoprotection within the UVA range displayed by quercetin and its derivative (rutin) used in oil-in-water emulsion with 10% concentration. The effect was fortified when the flavonoids were used in association with titanium dioxide. Rajnochová-Svobodová et al. [44] demonstrated that quercetin and its derivative, taxifolin (dihydroquercetin), effectively reduce UVA-induced damage in skin fibroblasts and epidermal keratinocytes. These antioxidants prevent the formation of reactive oxygen species (ROS), depletion of GSH, activation of CASP3, and increase the expression of antioxidant proteins: HO-1, NQO1 and CAT. In a direct comparison, quercetin proved to be more effective than taxifolin, although at the highest concentration, it exhibited pro-oxidant properties. Additionally, Liu et al. [45] highlight the potential applications of dihydroquercetin in the treatment and prevention of skin diseases, including those caused by solar radiation, further emphasizing the importance of this substance in dermatology.
The protective effects against UVB radiation were the subject of studies by Vicentini et al. [38]. When quercetin was topically applied to the skin in the form of a water-in-oil microemulsion, it effectively penetrated the deeper layers of the epidermis without causing irritation. It significantly limited the depletion of glutathione induced by UVB exposure and reduced the activity of ultraviolet-induced metalloproteinase. The findings were subsequently confirmed in Casagrande et al. [46], where topically applied quercetin also mitigated glutathione depletion and reduced the activity of metalloproteinase and myeloperoxidase. Furthermore, research conducted by Zhu et al. [47] demonstrated that quercetin affected UVB-induced cytotoxicity in epidermal keratinocytes. The antioxidant blocked the production of reactive oxygen species (ROS) caused by radiation. By scavenging reactive oxygen species, quercetin protected the cell membrane and mitochondria, and slowed the leakage of cytochrome c, inhibiting keratinocyte apoptosis.

3. Anti-Melanogenesis and Skin Whitening

The primary mechanism responsible for pigmentation and skin color is melanogenesis; thus, halting, slowing and counteracting this process is fundamental for skin lightening and whitening efforts [48]. Based on in vivo and in vitro studies, Choi and Shin [36] did not demonstrate a whitening effect of quercetin in cosmetic products. However, quercetin plays a crucial role in blocking melanogenesis by inhibiting tyrosinase, a key enzyme that activates the pigmentation process [49,50,51]. Nonetheless, other studies indicate that quercetin can increase tyrosinase activity and promote melanogenesis [52,53,54], which may result from the concentration-dependent activity. Findings of the above studies seem to suggest that pure quercetin at concentrations higher than 20 μM reduces melanin concentration, while at concentrations of 10–20 μM, it increases melanin content. Melanin content also decreases at concentrations of 50–100 μM, but this fact seems to be associated with increased cellular toxicity, which can be reduced by using quercetin in combination with vitamin C and arbutin [36]. Additionally, the anti-melanogenic effect depends on the position of hydroxyl groups (-OH) in the compound structure and the position and type of sugar residues in quercetin derivatives [55,56,57,58].
Conversely, studies by Chondrogianni et al. [41] on quercetin and its derivative, quercetin caprylate, showed that both compounds induce changes in the physiological characteristics of cells, including a localized whitening effect.

4. Wound Healing, Reduction of Skin Irritation and Scarring Prevention

One of the earliest analyses regarding the impact of quercetin on reducing dermal wound healing time in rats was conducted by Gomathi et al. [59]. The authors divided the studied rats into a control group and a group treated with quercetin incorporated into collagenous matrix. The group subjected to quercetin treatment exhibited a greater potential to quench free radicals in the active inflammatory processes in skin wounds. Consequently, the study pointed out the quercetin potential as a wound healing-promoting agent when used in dressing material.
Gopalakrishnan et al. [60] emphasized the significant role of TGF-β1 and VEGF activation by quercetin in the wound healing acceleration process. Two groups of rats: a control group and a Que-treated group were subject to evaluation for the period of 14 days. The quercetin-treated group exhibited a faster rate of wound closure compared to the control group. In addition to activating the above-mentioned compounds, quercetin significantly attenuated TNF-α activity, supported fibroblast proliferation processes and collagen activity, and induced IL-10 levels. A similar study was conducted by Kant et al. [61] where 80 rats divided into a 4 groups (including a control group) were treated with dimethyl sulfoxide solutions and Que concentrations ranging from 0.03% to 0.3%. After 20 days, it was found that the administration of the polyphenol at the highest dose of 0.3% resulted in faster formation of granulation tissue, allowing for quicker wound closure by supporting angiogenic and proliferative processes within fibroblasts. Quercetin also contributed to the induction of, e.g., IL-10, VEGF, TGF-β1, simultaneously reducing TNF-α activity.
The mechanism of quercetin impact on the improvement in wound healing quality, was investigated by Doersch and Newell-Rogers [62]. Quercetin was found to exhibit the potential to reduce fibrosis without affecting the rate of healing. Quercetin reduced the level of integrin αV while increasing the expression of integrin β1. These changes in transmembrane receptor expression may affect processes such as cell proliferation and extracellular matrix production. Additionally, the presence of quercetin reduces the demand for extracellular matrix, thus contributing to easier wound healing and scar reduction.
Moravvej et al. [63] points out quercetin’s potential for scar treatments. Long et al. [64] showed that quercetin combined with X-ray radiation reduces collagen synthesis in fibroblasts, both healthy and scarred. Continuing their research on the properties of quercetin, Hosnuter et al. [65], demonstrated in a randomized controlled clinical trial the efficacy of onion extract containing quercetin in reducing scar hypertrophy, while not affecting itchiness. Further studies conducted by Ramakrishnan et al. [66] focused on the synergistic action of quercetin with vitamin D3 on isolated keloid fibroblasts. Quercetin was found to reduce cell proliferation and collagen synthesis while inducing apoptosis. In addition, Si et al. [67] demonstrated that quercetin can suppress keloid resistance to radiotherapy by inhibiting the expression of the HIF-1 and interacting with the PI3K/AKT pathway, reducing AKT phosphorylation.
Yin et al. [68] demonstrated the effectiveness of quercetin in the process of pressure ulcer healing. Their in vitro and in vivo analyses of induced pressure ulcers in mice provided evidence as to quercetin participation in stimulating processes responsible for wound edge closure by inhibiting interference with the MAPK pathway. Additionally, quercetin was found to reduce cellular infiltration and decrease the concentration of inflammatory cytokines.
As suggested by Karuppagounder et al. [69], Beken et al. [70], and Hou et al. [71], the anti-inflammatory and antioxidant properties of quercetin can be utilized in the treatment of atopic dermatitis (AD). The course and development of AD are associated with the action of epithelial-derived cytokines [72]. In studies conducted by Beken et al. [70], the use of quercetin in prepared keratinocytes resulted in a decrease in the expression of cytokines such as IL-1β, IL-6, IL-8, and TSLP, and a simultaneous increase in the expression of SOD1, SOD2, GPX, CAT, and IL-10. An increase in mRNA expression of E-cadherin and occludin was also observed, along with a decrease in the expression of matrix metalloproteinases: MMP-1, MMP-2, and MMP-9. Additionally, inhibition of ERK1/2 phosphorylation, and MAPK was noted, as well as decreased expression of nuclear transcription factor NF-κB, with no effect on STAT6. The mechanism of action of quercetin had been previously determined by Cheng et al. [73], who focused on its impact on retinal pigment epithelial cells. Hou et al. [71] demonstrated that quercetin effectively lowers expression levels of cytokines such as CCL17, CCL22, IL-4, IL-6, IFN-γ, and TNF-α.
Quercetin demonstrates the ability to inhibit pro-inflammatory cytokines induced by Propionibacterium acnes bacteria. Lim et al. [33] showed that it suppresses TLR-2 production and inhibits phosphorylation of p38, ERK, and JNK MAPK kinases. Additionally, a decrease in mRNA levels for MMP-9 was observed. In vivo studies showed quercetin-induced reduction in thickness of erythema and swelling.
Liu et al. [74] developed quercetin-loaded liposomes in gels (QU-LG) to investigate their possible therapeutic effect on cutaneous eczema due to favorable antioxidant activity and anti-inflammatory effects of quercetin. Que was encapsulated in liposomes and evenly dispersed in sodium carboxymethyl cellulose hydrogels in order to enhance its bioavailability and efficiency of its dermal delivery. The research demonstrated that quercetin-containing liposomes-in-gel (QU-LG) applied to the skin of mice suffering from skin eczema exhibited good stability and adhesion to the skin. In the antioxidant test, QU-LG inhibited the production of malondialdehyde (MDA) in the liver better than the commercially available drug, dexamethasone acetate cream. Compared to untreated mice, mice treated with QU-LG showed a statistically significant reduction in dermatopathological symptoms. The results suggest that QU-LG exhibits good antioxidant activity in vivo and in vitro and that it can be used in the prevention and treatment of cutaneous eczema.
Maramaldi et al. [34] and Kurek-Górecka [75] investigated the use of phytosomes, which are alternatives to liposomes, as formulations enhancing the bioavailability of active ingredients. Quercetin in the form of phospholipids significantly reduced erythema and wheal diameter [34]. The study of Lu et al. [76] into quercetin-loaded niosomes described improved solubility, photostability, and skin penetration ability compared to conventional methods of delivering active ingredients. Li et al. [77] developed a xerogel utilizing polyvinyl alcohol (PVA) and quercetin-borate nanoparticles as the crosslinking agent. The produced xerogel films exhibited high bacteriostatic properties, high antioxidant potential, and accelerated skin regeneration.
Nalini et al. [78] compared the effects of quercetin and quercetin-loaded chitosan nanoparticles on the healing processes of open wounds. An accelerated healing process was observed in the studied rodents, due to inhibition of inflammatory cytokines, promotion of angiogenesis, and inhibition of free radicals. Increased levels of hydroxyproline and hexalin indicated enhanced reepithelialization. Quercetin used in monotherapy showed lower effectiveness than the mixture of quercetin-loaded alginate. Chitosan nanoparticles with a concentration of 0.075% showed the highest efficacy.
Yang et al. [79] investigated the association between quercetin and histamine, a compound that triggers inflammation. The study demonstrated a direct interaction between histamine H4 receptors and quercetin. It was found that quercetin inhibits IL-8 mRNA expression in keratinocyte and the scratching behavior-induced compound 48/80. Additionally, quercetin reduces calcium influx (Ca2+) induced by the H4 receptor through the TRPV1 channel, which limits itching, inflammation, and discomfort sensation.
Studies conducted by Katsarou et al. [80] did not show a beneficial effect of quercetin on sodium-lauryl-sulfate-induced skin irritation. It was observed that quercetin did not restore the protective barrier function of the skin, normalize transepidermal water loss, nor reduce erythema to levels observed before irritation.
Due to the more challenging wound healing process in cases of diabetes, studies have investigated the wound-healing potential of quercetin in diabetic rats [81,82]. Fu et al. [81] administered quercetin-containing medication to diabetic rats at various concentrations and observed reduced levels of inflammatory cytokines and the number of iNOS-positive cells, as well as increased activity of CD206-positive cells and intensified angiogenesis processes. The researchers attributed these effects to the influence of quercetin on inducing a shift in macrophage phenotype towards M2. Kant et al. [82] also evaluated the effectiveness of quercetin on wounds in diabetic rats. The applied quercetin (0.3%) ointment accelerated wound healing time and induced, e.g., VEGF and TGF-β, at the same time reducing levels of MMP-9 and TNF-α. The researchers also observed higher levels of GAP-43 resulting from polyphenol application.

5. Protection against Skin Oxidation, Anti-Inflammatory and Antiseptic Properties

Numerous studies have described not only the anti-inflammatory and antioxidant effect of quercetin on skin but also its general antioxidant activity [32,83,84]. Experimental studies by Tang et al. [85] confirmed the effectiveness of quercetin in inhibiting TNF-α, IL-1β, and IL-6 cytokines. The theoretical calculations illustrated that the oxygen atom on B rings may be the main site of electron cloud density changes, which results in ROS scavenging effects of quercetin. Ha et al. [86] demonstrated, that quercetin 3-O-β-D-glucuronide possesses protective effects on skin, including anti-inflammatory and antioxidant actions against UVB- or H2O2-induced oxidative stress. It reduces the expression of pro-inflammatory genes (COX-2, TNF-α) in stressed HaCaT cells as well as increasing Nrf2 expression and inhibiting melanin production in α-MSH-treated B16F10 cells.
Quercetin, combined with rutin and curcumin, loaded on porous copper oxide nanorods, exhibits not only anti-inflammatory properties but also bacteriostatic and bactericidal properties. Mansi et al. [87] demonstrated Que nanocomposite’s effective antagonism against bacteria such as Staphylococcus aureus, Bacillus subtilis, Salmonella typhi, Pseudomonas aeruginosa, Escherichia coli, and Klebsiella pneumoniae. The inhibitory action against Pseudomonas aeruginosa and Staphylococcus aureus was confirmed by Chittasupho et al. [88]. Ramzan et al. [89] utilized nanoparticles composed of quercetin and its copper complex, employing polycaprolactone (PCL) as a structural material, for treating skin infections. Studies showed that the nanoparticles have strong bactericidal effect against Staphylococcus aureus and stimulate epidermal regeneration without causing skin irritation. This points to the potential use of quercetin as an active agent in the treatment of impetigo and as an alternative to widely used ciprofloxacin. Quercetin delivered in the form of oil-based nanostructured lipid carriers also showed efficacy against Staphylococcus aureus [90]. Lúcio et al. [91] described the synergistic action of quercetin with omega-3 fatty acids, where bioactives in the form of nanostructured lipid carriers and hydrogels were exhibited with high stability and skin permeability.
Quercetin, combined with other antioxidants, occurring in the extract of sumac (Rhus coriaria), shows strong antibacterial activity. Gabr and Alghadir [92] demonstrated the antibacterial effect of the extract against Staphylococcus aureus and Pseudomonas aeruginosa. Additionally, the extract reduced inflammation, regulated the activity of MMP-8 and MPO enzymes, supported wound contraction, and promoted collagen and hydroxyproline deposition. Extracts isolated from Syncarpia hillii leaves, containing the quercetin glycoside (quercitrin), also exhibited high antibacterial activity against both Gram-positive and Gram-negative bacteria, including staphylococci and Enterococcus faecalis. The leaves are used in traditional herbal medicine for treating wounds and skin infections [93]. Extracts rich in quercetin and its glycosides from the Opuntia genus (Opuntia Spp.) also show antibacterial properties in wound treatment [94], as do extracts from Bridelia ferruginea [95] and Spermacoce princeae, which additionally exhibit UV-protective effects [96].
Quercetin exhibits antibiofilm effectiveness. Biofilms are bacterial aggregations that can grow on different surfaces. Biofilms colonize wounds, protect the pathogen from host defenses and obstruct antibiotic delivery, thereby weakening wound healing [97]. Mu et al. [98] claim that quercetin and other secondary metabolites isolated from plants have demonstrated varying levels of biofilm inhibition in Gram-negative pathogens. In their studies [98,99] quercetin and extracts rich in this bioactive compound exhibited a concentration-dependent reduction in Staphylococcus epidermidis biofilm formation. Quercetin reduced biofilm formation up to 95.3% at the concentration of 500 μg mL−1. Gopu et al. [100] tested quercetin in case of biofilm formation of different pathogens, responsible for food spoilage. Their studies, conducted for quercetin at different concentration (20–80 μg mL−1), showed 13–72%, 8–80%, and 10–61% reduction in biofilm biomass of K. pneumoniae, P. aeruginosa, and Y. enterocolitica, respectively. In their studies, Musini et al. [97] highlighted the antibiofilm activity of quercetin against the drug-resistant pathogen S. aureus. Since S. aureus is an important virulence factor influencing its persistence in both the environment and the host organism and is responsible for biofilm-associated infections, the antibiofilm activity of quercetin is a promising solution for antibiotic-resistant strains of this bacteria.

6. Bioavailability, Safety of Use, and Potential Harmful Effects

The effectiveness of quercetin and its derivatives in treating skin diseases may be compromised by its absorbability [101]. Studies conducted by Hung et al. have demonstrated that quercetin is better absorbed through photoaged skin, likely because UV exposure disrupts the barrier functions of the skin [102]. Lin et al. [103] compared the transdermal absorption levels of quercetin and its derivatives, including polymethoxylated quercetin (QM). It was shown that the structure of the compound has a crucial impact on transdermal absorption. Derivatives with higher lipophilicity more easily penetrated the skin barrier. Furthermore, the sugar moiety of glycosides significantly affects skin permeability, with those having -OH groups potentially forming hydrogen bonds with ceramides in the epidermis. QM demonstrated the highest level of permeation, suggesting it as the best delivery form of quercetin for topical applications.
Quercetin is considered safe for daily consumption by food safety authorities, including the U.S. FDA—Food & Drug Administration [104]. The average daily intake of quercetin from food is approximately 20–40 mg. The daily dosage of quercetin in dietary supplements ranges from 50 to 500 mg. Detection of this compound in plasma is possible about 15–30 min after consuming a 250 mg or 500 mg chewable quercetin preparation. The maximum concentration is reached after 120–180 min (levels return to baseline after 24 h) [105]. In studies, doses of 500–1000 mg of quercetin are typically used. It has been proven that such an amount of quercetin, up to 1000 mg, taken over several months does not adversely affect blood parameters, liver and kidney function, or serum electrolyte levels [106].
In addition to many health benefits, potential health risks associated with the use of quercetin have also been noted [107,108]. Studies have shown a negative impact of quercetin supplementation for neurodegenerative prevention, aimed at protecting nerve cells. It has been demonstrated that high exposure to quercetin can lead to a reduction in intracellular glutathione levels, as well as changes in the genes responsible for intracellular processes [107]. Different studies suggest potential cytotoxic activity of quercetin, induced by inhibiting the action of specific genes in the presence of gamma radiation doses [108]. In conclusion, it is important to emphasize that the risks associated with quercetin use are minimal compared to its benefits. Long-term consumption of quercetin is not recommended, however, for individuals prone to hypotension (low blood pressure) and those with impaired blood clotting [109].

7. Summary

Quercetin exhibits many health-promoting, antioxidant, and therapeutic properties (Table 3). While research confirms the potential use of its various forms (e.g., extract, emulsion, aqueous extracts) in skin therapy, the variety of methods for obtaining quercetin even broadens its possible application in medicine. The cited studies suggest that research on the use of natural sources of quercetin in treatment of skin diseases, specifically reducing oxidation processes, aging, melanogenesis, scarring, accelerating wound healing, and protection against UV radiation, is well founded. Advances in the research will undoubtedly contribute to the development of new dermatological preparations and therapies. Among many studies in the field of development new materials, many of them mention incorporating quercetin or extracts containing this compound into wound healing and treatment of skin diseases [110,111,112]. This direction seems to be the closest to commercialization and therefore the appearance of products (hydrogels, patches, wound dressings etc.) should take place in the near future. Obtaining quercetin from waste from the food industry and constantly improving technologies and extraction techniques will allow for greater availability of this compound, not only in medicine, but also in pharmaceuticals and the food sector.

Author Contributions

Conceptualization, M.K.Z., A.D. and K.L.; writing—original draft preparation, M.K.Z. and K.L.; writing—review and editing, B.B., J.S. and A.D., visualization, M.K.Z., B.B. and J.S.; supervision, A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hatalska, N. Mapa Trendów. 2024. Available online: https://infuture.institute/mapa-trendow/ (accessed on 17 May 2024).
  2. Szulc, J.; Błaszak, B.; Wenda-Piesik, A.; Gozdecka, G.; Zary-Sikorska, E.; Bąk, M.; Bauza-Kaszewska, J. Zero Waste Technology of Soybeans Processing. Sustainability 2023, 15, 14873. [Google Scholar] [CrossRef]
  3. Hejna, A.; Szulc, J.; Błaszak, B. Brewers’ Spent Grain—Simply Waste or Potential Ingredient of Functional Food? Zywnosc Nauka Technol. Jakosc/Food Sci. Technol. Qual. 2024, 30, 5–23. [Google Scholar] [CrossRef]
  4. Hogervorst, J.C.; Miljić, U.; Puškaš, V. 5—Extraction of Bioactive Compounds from Grape Processing By-Products. In Handbook of Grape Processing By-Products; Galanakis, C.M., Ed.; Academic Press: Cambridge, MA, USA, 2017; pp. 105–135. ISBN 978-0-12-809870-7. [Google Scholar]
  5. Abbas, M.; Saeed, F.; Anjum, F.M.; Afzaal, M.; Tufail, T.; Bashir, M.S.; Ishtiaq, A.; Hussain, S.; Suleria, H.A.R. Natural Polyphenols: An Overview. Int. J. Food Prop. 2017, 20, 1689–1699. [Google Scholar] [CrossRef]
  6. Salehi, B.; Machin, L.; Monzote, L.; Sharifi-Rad, J.; Ezzat, S.M.; Salem, M.A.; Merghany, R.M.; El Mahdy, N.M.; Kılıç, C.S.; Sytar, O.; et al. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health. ACS Omega 2020, 5, 11849–11872. [Google Scholar] [CrossRef] [PubMed]
  7. Abraham, M.H.; Acree, W.E. On the Solubility of Quercetin. J. Mol. Liq. 2014, 197, 157–159. [Google Scholar] [CrossRef]
  8. Rich, G.T.; Buchweitz, M.; Winterbone, M.S.; Kroon, P.A.; Wilde, P.J. Towards an Understanding of the Low Bioavailability of Quercetin: A Study of Its Interaction with Intestinal Lipids. Nutrients 2017, 9, 111. [Google Scholar] [CrossRef] [PubMed]
  9. Antonio Tamayo-Ramos, J.; Martel, S.; Barros, R.; Bol, A.; Atilhan, M.; Aparicio, S. On the Behavior of Quercetin + Organic Solvent Solutions and Their Role for C60 Fullerene Solubilization. J. Mol. Liq. 2022, 345, 117714. [Google Scholar] [CrossRef]
  10. Lakhanpal, P.; Rai, D.K. Quercetin: A Versatile Flavonoid. Internet J. Med. Update 2007, 2. [Google Scholar] [CrossRef]
  11. Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem 2023 Update. Nucleic Acids Res. 2023, 51, D1373–D1380. [Google Scholar] [CrossRef]
  12. Oku, S.; Aoki, K.; Honjo, M.; Muro, T.; Tsukazaki, H. Temporal Changes in Quercetin Accumulation and Composition in Onion (Allium cepa L.) Bulbs and Leaf Blades. Hortic. J. 2021, 90, 326–333. [Google Scholar] [CrossRef]
  13. Mogren, L.M.; Olsson, M.E.; Gertsson, U.E. Quercetin Content in Field-Cured Onions (Allium cepa L.):  Effects of Cultivar, Lifting Time, and Nitrogen Fertilizer Level. J. Agric. Food Chem. 2006, 54, 6185–6191. [Google Scholar] [CrossRef]
  14. Heimler, D.; Romani, A.; Ieri, F. Plant Polyphenol Content, Soil Fertilization and Agricultural Management: A Review. Eur. Food Res. Technol. 2017, 243, 1107–1115. [Google Scholar] [CrossRef]
  15. Kazimierczak, R.; Średnicka-Tober, D.; Barański, M.; Hallmann, E.; Góralska-Walczak, R.; Kopczyńska, K.; Rembiałkowska, E.; Górski, J.; Leifert, C.; Rempelos, L.; et al. The Effect of Different Fertilization Regimes on Yield, Selected Nutrients, and Bioactive Compounds Profiles of Onion. Agronomy 2021, 11, 883. [Google Scholar] [CrossRef]
  16. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef]
  17. Nishimuro, H.; Ohnishi, H.; Sato, M.; Ohnishi-Kameyama, M.; Matsunaga, I.; Naito, S.; Ippoushi, K.; Oike, H.; Nagata, T.; Akasaka, H.; et al. Estimated Daily Intake and Seasonal Food Sources of Quercetin in Japan. Nutrients 2015, 7, 2345–2358. [Google Scholar] [CrossRef]
  18. Häkkinen, S.H.; Kärenlampi, S.O.; Heinonen, I.M.; Mykkänen, H.M.; Törrönen, A.R. Content of the Flavonols Quercetin, Myricetin, and Kaempferol in 25 Edible Berries. J. Agric. Food Chem. 1999, 47, 2274–2279. [Google Scholar] [CrossRef]
  19. Stecher, G.; Huck, C.W.; Popp, M.; Bonn, G.K. Determination of Flavonoids and Stilbenes in Red Wine and Related Biological Products by HPLC and HPLC-ESI-MS-MS. Fresenius J. Anal. Chem. 2001, 371, 73–80. [Google Scholar] [CrossRef] [PubMed]
  20. Eftekhari, M.; Alizadeh, M.; Ebrahimi, P. Evaluation of the Total Phenolics and Quercetin Content of Foliage in Mycorrhizal Grape (Vitis Vinifera L.) Varieties and Effect of Postharvest Drying on Quercetin Yield. Ind. Crops Prod. 2012, 38, 160–165. [Google Scholar] [CrossRef]
  21. Ruberto, G.; Renda, A.; Daquino, C.; Amico, V.; Spatafora, C.; Tringali, C.; Tommasi, N.D. Polyphenol Constituents and Antioxidant Activity of Grape Pomace Extracts from Five Sicilian Red Grape Cultivars. Food Chem. 2007, 100, 203–210. [Google Scholar] [CrossRef]
  22. Osojnik Črnivec, I.G.; Skrt, M.; Šeremet, D.; Sterniša, M.; Farčnik, D.; Štrumbelj, E.; Poljanšek, A.; Cebin, N.; Pogačnik, L.; Smole Možina, S.; et al. Waste Streams in Onion Production: Bioactive Compounds, Quercetin and Use of Antimicrobial and Antioxidative Properties. Waste Manag. 2021, 126, 476–486. [Google Scholar] [CrossRef]
  23. Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef] [PubMed]
  24. Aghababaei, F.; Hadidi, M. Recent Advances in Potential Health Benefits of Quercetin. Pharmaceuticals 2023, 16, 1020. [Google Scholar] [CrossRef]
  25. Wang, G.; Wang, Y.; Yao, L.; Gu, W.; Zhao, S.; Shen, Z.; Lin, Z.; Liu, W.; Yan, T. Pharmacological Activity of Quercetin: An Updated Review. Evid. Based Complement. Altern. Med. 2022, 2022, 3997190. [Google Scholar] [CrossRef] [PubMed]
  26. Naidu, N.; Biyani, D.; Umekar, M.; Burley, V. An Overview of a Versatile Compound: Quercetin. Int. J. Pharm. Sci. Rev. Res. 2021, 69, 248–257. [Google Scholar] [CrossRef]
  27. Jan, A.T.; Kamli, M.; Murtaza, I.; Singh, J.; Ali, A.; Haq, Q. Dietary Flavonoid Quercetin and Associated Health Benefits—An Overview. Food Rev. Int. 2010, 26, 302–317. [Google Scholar] [CrossRef]
  28. Cui, Z.; Zhao, X.; Amevor, F.K.; Du, X.; Wang, Y.; Li, D.; Shu, G.; Tian, Y.; Zhao, X. Therapeutic Application of Quercetin in Aging-Related Diseases: SIRT1 as a Potential Mechanism. Front. Immunol. 2022, 13, 943321. [Google Scholar] [CrossRef] [PubMed]
  29. Amić, A.; Lučić, B.; Stepanić, V.; Marković, Z.; Marković, S.; Dimitrić Marković, J.M.; Amić, D. Free Radical Scavenging Potency of Quercetin Catecholic Colonic Metabolites: Thermodynamics of 2H+/2e− Processes. Food Chem. 2017, 218, 144–151. [Google Scholar] [CrossRef] [PubMed]
  30. Aliaga, C.; Lissi, E.A. Comparison of the Free Radical Scavenger Activities of Quercetin and Rutin An Experimental and Theoretical Study. Can. J. Chem. 2004, 82, 1668–1673. [Google Scholar] [CrossRef]
  31. Sato, S.; Mukai, Y. Modulation of Chronic Inflammation by Quercetin: The Beneficial Effects on Obesity. J. Inflamm. Res. 2020, 13, 421–431. [Google Scholar] [CrossRef]
  32. Xu, D.; Hu, M.-J.; Wang, Y.-Q.; Cui, Y.-L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef]
  33. Lim, H.-J.; Kang, S.-H.; Song, Y.-J.; Jeon, Y.-D.; Jin, J.-S. Inhibitory Effect of Quercetin on Propionibacterium Acnes-Induced Skin Inflammation. Int. Immunopharmacol. 2021, 96, 107557. [Google Scholar] [CrossRef] [PubMed]
  34. Maramaldi, G.; Togni, S.; Pagin, I.; Giacomelli, L.; Cattaneo, R.; Eggenhöffner, R.; Burastero, S.E. Soothing and Anti-Itch Effect of Quercetin Phytosome in Human Subjects: A Single-Blind Study. Clin. Cosmet. Investig. Dermatol. 2016, 9, 55–62. [Google Scholar] [CrossRef]
  35. Shin, E.J.; Lee, J.S.; Hong, S.; Lim, T.-G.; Byun, S. Quercetin Directly Targets JAK2 and PKCδ and Prevents UV-Induced Photoaging in Human Skin. Int. J. Mol. Sci. 2019, 20, 5262. [Google Scholar] [CrossRef] [PubMed]
  36. Choi, M.-H.; Shin, H.-J. Anti-Melanogenesis Effect of Quercetin. Cosmetics 2016, 3, 18. [Google Scholar] [CrossRef]
  37. Zhang, X.; Tang, Y.; Lu, G.; Gu, J. Pharmacological Activity of Flavonoid Quercetin and Its Therapeutic Potential in Testicular Injury. Nutrients 2023, 15, 2231. [Google Scholar] [CrossRef] [PubMed]
  38. Vicentini, F.T.M.C.; He, T.; Shao, Y.; Fonseca, M.J.V.; Verri, W.A.; Fisher, G.J.; Xu, Y. Quercetin Inhibits UV Irradiation-Induced Inflammatory Cytokine Production in Primary Human Keratinocytes by Suppressing NF-NF-κB pathway. J. Dermatol. Sci. 2011, 61, 162–168. [Google Scholar] [CrossRef]
  39. Kim, D.H.; Auh, J.-H.; Oh, J.; Hong, S.; Choi, S.; Shin, E.J.; Woo, S.O.; Lim, T.-G.; Byun, S. Propolis Suppresses UV-Induced Photoaging in Human Skin through Directly Targeting Phosphoinositide 3-Kinase. Nutrients 2020, 12, 3790. [Google Scholar] [CrossRef] [PubMed]
  40. Nebus, J.; Vassilatou, K. Clinical Improvements in Facial Photoaged Skin Using a Novel Oak Quercetin Topical Preparation. J. Am. Acad. Dermatol. 2011, 64, AB73. [Google Scholar] [CrossRef]
  41. Chondrogianni, N.; Kapeta, S.; Chinou, I.; Vassilatou, K.; Papassideri, I.; Gonos, E.S. Anti-Ageing and Rejuvenating Effects of Quercetin. Exp. Gerontol. 2010, 45, 763–771. [Google Scholar] [CrossRef] [PubMed]
  42. Singh, P.; Arif, Y.; Bajguz, A.; Hayat, S. The Role of Quercetin in Plants. Plant Physiol. Biochem. 2021, 166, 10–19. [Google Scholar] [CrossRef] [PubMed]
  43. Choquenet, B.; Couteau, C.; Paparis, E.; Coiffard, L.J.M. Quercetin and Rutin as Potential Sunscreen Agents: Determination of Efficacy by an in Vitro Method. J. Nat. Prod. 2008, 71, 1117–1118. [Google Scholar] [CrossRef]
  44. Rajnochová Svobodová, A.; Ryšavá, A.; Čížková, K.; Roubalová, L.; Ulrichová, J.; Vrba, J.; Zálešák, B.; Vostálová, J. Effect of the Flavonoids Quercetin and Taxifolin on UVA-Induced Damage to Human Primary Skin Keratinocytes and Fibroblasts. Photochem. Photobiol. Sci. 2022, 21, 59–75. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Z.; Qiu, D.; Yang, T.; Su, J.; Liu, C.; Su, X.; Li, A.; Sun, P.; Li, J.; Yan, L.; et al. Research Progress of Dihydroquercetin in the Treatment of Skin Diseases. Molecules 2023, 28, 6989. [Google Scholar] [CrossRef] [PubMed]
  46. Casagrande, R.; Georgetti, S.R.; Verri, W.A.; Dorta, D.J.; dos Santos, A.C.; Fonseca, M.J.V. Protective Effect of Topical Formulations Containing Quercetin against UVB-Induced Oxidative Stress in Hairless Mice. J. Photochem. Photobiol. B 2006, 84, 21–27. [Google Scholar] [CrossRef] [PubMed]
  47. Zhu, X.; Li, N.; Wang, Y.; Ding, L.; Chen, H.; Yu, Y.; Shi, X. Protective Effects of Quercetin on UVB Irradiation-induced Cytotoxicity through ROS Clearance in Keratinocyte Cells. Oncol. Rep. 2017, 37, 209–218. [Google Scholar] [CrossRef] [PubMed]
  48. Qian, W.; Liu, W.; Zhu, D.; Cao, Y.; Tang, A.; Gong, G.; Su, H. Natural Skin-Whitening Compounds for the Treatment of Melanogenesis (Review). Exp. Ther. Med. 2020, 20, 173–185. [Google Scholar] [CrossRef] [PubMed]
  49. Fan, M.; Zhang, G.; Hu, X.; Xu, X.; Gong, D. Quercetin as a Tyrosinase Inhibitor: Inhibitory Activity, Conformational Change and Mechanism. Food Res. Int. 2017, 100, 226–233. [Google Scholar] [CrossRef] [PubMed]
  50. Cho, H.W.; Jung, W.S.; An, B.; Cho, J.H.; Jung, S.Y. Isolation of Compounds Having Inhibitory Activity toward Tyrosinase from Receptaculum Nelumbinis. Korean J. Pharmacogn. 2013, 44, 1–5. [Google Scholar]
  51. Morlière, P.; Mazière, J.-C.; Patterson, L.K.; Conte, M.-A.; Dupas, J.-L.; Ducroix, J.-P.; Filipe, P.; Santus, R. On the Repair of Oxidative Damage to Apoferritin: A Model Study with the Flavonoids Quercetin and Rutin in Aerated and Deaerated Solutions. Free Radic. Res. 2013, 47, 463–473. [Google Scholar] [CrossRef]
  52. Niu, C.; Aisa, H.A. Upregulation of Melanogenesis and Tyrosinase Activity: Potential Agents for Vitiligo. Molecules 2017, 22, 1303. [Google Scholar] [CrossRef]
  53. Takekoshi, S.; Matsuzaki, K.; Kitatani, K. Quercetin Stimulates Melanogenesis in Hair Follicle Melanocyte of the Mouse. Tokai J. Exp. Clin. Med. 2013, 38, 129–134. [Google Scholar]
  54. Takeyama, R.; Takekoshi, S.; Nagata, H.; Osamura, R.Y.; Kawana, S. Quercetin-Induced Melanogenesis in a Reconstituted Three-Dimensional Human Epidermal Model. J. Mol. Histol. 2004, 35, 157–165. [Google Scholar] [CrossRef]
  55. Nagata, H.; Takekoshi, S.; Takeyama, R.; Homma, T.; Yoshiyuki Osamura, R. Quercetin Enhances Melanogenesis by Increasing the Activity and Synthesis of Tyrosinase in Human Melanoma Cells and in Normal Human Melanocytes. Pigment. Cell Res. 2004, 17, 66–73. [Google Scholar] [CrossRef]
  56. Yamauchi, K.; Mitsunaga, T.; Batubara, I. Novel Quercetin Glucosides from Helminthostachys Zeylanica Root and Acceleratory Activity of Melanin Biosynthesis. J. Nat. Med. 2013, 67, 369–374. [Google Scholar] [CrossRef]
  57. Yamauchi, K.; Mitsunaga, T.; Batubara, I. Synthesis of Quercetin Glycosides and Their Melanogenesis Stimulatory Activity in B16 Melanoma Cells. Bioorg. Med. Chem. 2014, 22, 937–944. [Google Scholar] [CrossRef] [PubMed]
  58. Fujii, T.; Saito, M. Inhibitory Effect of Quercetin Isolated from Rose Hip (Rosa canina L.) against Melanogenesis by Mouse Melanoma Cells. Biosci. Biotechnol. Biochem. 2009, 73, 1989–1993. [Google Scholar] [CrossRef]
  59. Gomathi, K.; Gopinath, D.; Rafiuddin Ahmed, M.; Jayakumar, R. Quercetin Incorporated Collagen Matrices for Dermal Wound Healing Processes in Rat. Biomaterials 2003, 24, 2767–2772. [Google Scholar] [CrossRef] [PubMed]
  60. Gopalakrishnan, A.; Ram, M.; Kumawat, S.; Tandan, S.; Kumar, D. Quercetin Accelerated Cutaneous Wound Healing in Rats by Increasing Levels of VEGF and TGF-Β1. Indian J. Exp. Biol. 2016, 54, 187–195. [Google Scholar]
  61. Kant, V.; Jangir, B.L.; Kumar, V.; Nigam, A.; Sharma, V. Quercetin Accelerated Cutaneous Wound Healing in Rats by Modulation of Different Cytokines and Growth Factors. Growth Factors 2020, 38, 105–119. [Google Scholar] [CrossRef] [PubMed]
  62. Doersch, K.M.; Newell-Rogers, M.K. The Impact of Quercetin on Wound Healing Relates to Changes in αV and Β1 Integrin Expression. Exp. Biol. Med. 2017, 242, 1424–1431. [Google Scholar] [CrossRef]
  63. Moravvej, H.; Memariani, M.; Memariani, H. Quercetin: A Potential Treatment for Keloids. Sultan Qaboos Univ. Med. J. 2019, 19, e372–e373. [Google Scholar] [CrossRef] [PubMed]
  64. Long, X.; Zeng, X.; Zhang, F.; Wang, X. Influence of Quercetin and X-Ray on Collagen Synthesis of Cultured Human Keloid-Derived Fibroblasts. Chin. Med. Sci. J. 2006, 21, 179–183. [Google Scholar]
  65. Hosnuter, M.; Payasli, C.; Isikdemir, A.; Tekerekoglu, B. The Effects of Onion Extract on Hypertrophic and Keloid Scars. J. Wound Care 2007, 16, 251–254. [Google Scholar] [CrossRef] [PubMed]
  66. Mathangi Ramakrishnan, K.; Babu, M.; Lakshmi Madhavi, M.S. Response of Keloid Fibroblasts to Vitamin D3 and Quercetin Treatment—In Vitro Study. Ann. Burn. Fire Disasters 2015, 28, 187–191. [Google Scholar]
  67. Si, L.-B.; Zhang, M.-Z.; Han, Q.; Huang, J.-N.; Long, X.; Long, F.; Zhao, R.C.-H.; Huang, J.-Z.; Liu, Z.-F.; Zhao, R.; et al. Sensitization of Keloid Fibroblasts by Quercetin through the PI3K/Akt Pathway Is Dependent on Regulation of HIF-1α. Am. J. Transl. Res. 2018, 10, 4223–4234. [Google Scholar]
  68. Yin, G.; Wang, Z.; Wang, Z.; Wang, X. Topical Application of Quercetin Improves Wound Healing in Pressure Ulcer Lesions. Exp. Dermatol. 2018, 27, 779–786. [Google Scholar] [CrossRef] [PubMed]
  69. Karuppagounder, V.; Arumugam, S.; Thandavarayan, R.A.; Sreedhar, R.; Giridharan, V.V.; Watanabe, K. Molecular Targets of Quercetin with Anti-Inflammatory Properties in Atopic Dermatitis. Drug Discov. Today 2016, 21, 632–639. [Google Scholar] [CrossRef] [PubMed]
  70. Beken, B.; Serttas, R.; Yazicioglu, M.; Turkekul, K.; Erdogan, S. Quercetin Improves Inflammation, Oxidative Stress, and Impaired Wound Healing in Atopic Dermatitis Model of Human Keratinocytes. Pediatr. Allergy Immunol. Pulmonol. 2020, 33, 69–79. [Google Scholar] [CrossRef]
  71. Hou, D.-D.; Zhang, W.; Gao, Y.-L.; Sun, Y.-Z.; Wang, H.-X.; Qi, R.-Q.; Chen, H.-D.; Gao, X.-H. Anti-Inflammatory Effects of Quercetin in a Mouse Model of MC903-Induced Atopic Dermatitis. Int. Immunopharmacol. 2019, 74, 105676. [Google Scholar] [CrossRef]
  72. Ukleja-Sokołowska, N.; Bartuzi, Z. IL 25, IL 33 i TSLP w chorobach atopowych—Obecny stan wiedzy. Alerg. Astma Immunol. 2020, 25, 59. [Google Scholar]
  73. Cheng, S.-C.; Huang, W.-C.; S. Pang, J.-H.; Wu, Y.-H.; Cheng, C.-Y. Quercetin Inhibits the Production of IL-1β-Induced Inflammatory Cytokines and Chemokines in ARPE-19 Cells via the MAPK and NF-κB Signaling Pathways. Int. J. Mol. Sci. 2019, 20, 2957. [Google Scholar] [CrossRef]
  74. Liu, C.; Cheng, X.; Wu, Y.; Xu, W.; Xia, H.; Jia, R.; Liu, Y.; Shen, S.; Xu, Y.; Cheng, Z. Antioxidant Activity of Quercetin-Containing Liposomes-in-Gel and Its Effect on Prevention and Treatment of Cutaneous Eczema. Pharmaceuticals 2023, 16, 1184. [Google Scholar] [CrossRef] [PubMed]
  75. Kurek-Górecka, A.; Górecki, M. Fitosomy—Fosfolipidowe Nośniki Jako Alternatywa Dla Liposomów w Nowoczesnych Kosmetykach. Pol. J. Cosmetol. 2015, 18, 117–118. [Google Scholar]
  76. Lu, B.; Huang, Y.; Chen, Z.; Ye, J.; Xu, H.; Chen, W.; Long, X. Niosomal Nanocarriers for Enhanced Skin Delivery of Quercetin with Functions of Anti-Tyrosinase and Antioxidant. Molecules 2019, 24, 2322. [Google Scholar] [CrossRef] [PubMed]
  77. Li, X.; Yang, X.; Wang, Z.; Liu, Y.; Guo, J.; Zhu, Y.; Shao, J.; Li, J.; Wang, L.; Wang, K. Antibacterial, Antioxidant and Biocompatible Nanosized Quercetin-PVA Xerogel Films for Wound Dressing. Colloids Surf. B Biointerfaces 2022, 209, 112175. [Google Scholar] [CrossRef] [PubMed]
  78. Nalini, T.; Khaleel Basha, S.; Mohamed Sadiq, A.; Sugantha Kumari, V. Fabrication and Evaluation of Nanoencapsulated Quercetin for Wound Healing Application. Polym. Bull. 2023, 80, 515–540. [Google Scholar] [CrossRef]
  79. Yang, C.-C.; Hung, Y.-L.; Li, H.-J.; Lin, Y.-F.; Wang, S.-J.; Chang, D.-C.; Pu, C.-M.; Hung, C.-F. Quercetin Inhibits Histamine-Induced Calcium Influx in Human Keratinocyte via Histamine H4 Receptors. Int. Immunopharmacol. 2021, 96, 107620. [Google Scholar] [CrossRef] [PubMed]
  80. Katsarou, A.; Davoy, E.; Xenos, K.; Armenaka, M.; Theoharides, T.C. Effect of an Antioxidant (Quercetin) on Sodium-Lauryl-Sulfate-Induced Skin Irritation. Contact Dermat. 2000, 42, 85–89. [Google Scholar] [CrossRef]
  81. Fu, J.; Huang, J.; Lin, M.; Xie, T.; You, T. Quercetin Promotes Diabetic Wound Healing via Switching Macrophages from M1 to M2 Polarization. J. Surg. Res. 2020, 246, 213–223. [Google Scholar] [CrossRef]
  82. Kant, V.; Jangir, B.L.; Sharma, M.; Kumar, V.; Joshi, V.G. Topical Application of Quercetin Improves Wound Repair and Regeneration in Diabetic Rats. Immunopharmacol. Immunotoxicol. 2021, 43, 536–553. [Google Scholar] [CrossRef]
  83. Zhou, Y.; Qian, C.; Tang, Y.; Song, M.; Zhang, T.; Dong, G.; Zheng, W.; Yang, C.; Zhong, C.; Wang, A.; et al. Advance in the Pharmacological Effects of Quercetin in Modulating Oxidative Stress and Inflammation Related Disorders. Phytother. Res. 2023, 37, 4999–5016. [Google Scholar] [CrossRef] [PubMed]
  84. Alizadeh, S.R.; Ebrahimzadeh, M.A. Quercetin Derivatives: Drug Design, Development, and Biological Activities, a Review. Eur. J. Med. Chem. 2022, 229, 114068. [Google Scholar] [CrossRef] [PubMed]
  85. Tang, J.; Diao, P.; Shu, X.; Li, L.; Xiong, L. Quercetin and Quercitrin Attenuates the Inflammatory Response and Oxidative Stress in LPS-Induced RAW264.7 Cells: In Vitro Assessment and a Theoretical Model. BioMed Res. Int. 2019, 2019, 7039802. [Google Scholar] [CrossRef] [PubMed]
  86. Ha, A.T.; Rahmawati, L.; You, L.; Hossain, M.A.; Kim, J.-H.; Cho, J.Y. Anti-Inflammatory, Antioxidant, Moisturizing, and Antimelanogenesis Effects of Quercetin 3-O-β-D-Glucuronide in Human Keratinocytes and Melanoma Cells via Activation of NF-κB and AP-1 Pathways. Int. J. Mol. Sci. 2021, 23, 433. [Google Scholar] [CrossRef]
  87. Mansi, K.; Kumar, R.; Narula, D.; Pandey, S.K.; Kumar, V.; Singh, K. Microwave-Induced CuO Nanorods: A Comparative Approach between Curcumin, Quercetin, and Rutin to Study Their Antioxidant, Antimicrobial, and Anticancer Effects against Normal Skin Cells and Human Breast Cancer Cell Lines MCF-7 and T-47D. ACS Appl. Bio Mater. 2022, 5, 5762–5778. [Google Scholar] [CrossRef] [PubMed]
  88. Chittasupho, C.; Manthaisong, A.; Okonogi, S.; Tadtong, S.; Samee, W. Effects of Quercetin and Curcumin Combination on Antibacterial, Antioxidant, In Vitro Wound Healing and Migration of Human Dermal Fibroblast Cells. Int. J. Mol. Sci. 2021, 23, 142. [Google Scholar] [CrossRef] [PubMed]
  89. Ramzan, N.; Abbas, G.; Mahmood, K.; Aziz, M.; Rasul, S.; Ahmed, N.; Shah, S.; Uzair, M.; Usman, M.; Khan, W.S.; et al. Concomitant Effect of Quercetin and Its Copper Complex in the Development of Sustained-Release Nanoparticles of Polycaprolactone, Used for the Treatment of Skin Infection. Mol. Pharm. 2023, 20, 1382–1393. [Google Scholar] [CrossRef] [PubMed]
  90. de Barros, D.P.C.; Santos, R.; Reed, P.; Fonseca, L.P.; Oliva, A. Design of Quercetin-Loaded Natural Oil-Based Nanostructured Lipid Carriers for the Treatment of Bacterial Skin Infections. Molecules 2022, 27, 8818. [Google Scholar] [CrossRef] [PubMed]
  91. Lúcio, M.; Giannino, N.; Barreira, S.; Catita, J.; Gonçalves, H.; Ribeiro, A.; Fernandes, E.; Carvalho, I.; Pinho, H.; Cerqueira, F.; et al. Nanostructured Lipid Carriers Enriched Hydrogels for Skin Topical Administration of Quercetin and Omega-3 Fatty Acid. Pharmaceutics 2023, 15, 2078. [Google Scholar] [CrossRef]
  92. Gabr, S.A.; Alghadir, A.H. Evaluation of the Biological Effects of Lyophilized Hydrophilic Extract of Rhus Coriaria on Myeloperoxidase (MPO) Activity, Wound Healing, and Microbial Infections of Skin Wound Tissues. Evid. Based Complement. Altern. Med. 2019, 2019, 5861537. [Google Scholar] [CrossRef]
  93. Perera, M.M.N.; Dighe, S.N.; Katavic, P.L.; Collet, T.A. Antibacterial Potential of Extracts and Phytoconstituents Isolated from Syncarpia Hillii Leaves In Vitro. Plants 2022, 11, 283. [Google Scholar] [CrossRef] [PubMed]
  94. Madrigal-Santillán, E.; Portillo-Reyes, J.; Madrigal-Bujaidar, E.; Sánchez-Gutiérrez, M.; Izquierdo-Vega, J.A.; Izquierdo-Vega, J.; Delgado-Olivares, L.; Vargas-Mendoza, N.; Álvarez-González, I.; Morales-González, Á.; et al. Opuntia Spp. in Human Health: A Comprehensive Summary on Its Pharmacological, Therapeutic and Preventive Properties. Part 2. Plants 2022, 11, 2333. [Google Scholar] [CrossRef] [PubMed]
  95. Yeboah, G.N.; Owusu, F.W.A.; Archer, M.-A.; Kyene, M.O.; Kumadoh, D.; Ayertey, F.; Mintah, S.O.; Atta-Adjei Junior, P.; Appiah, A.A. Bridelia Ferruginea Benth.; An Ethnomedicinal, Phytochemical, Pharmacological and Toxicological Review. Heliyon 2022, 8, e10366. [Google Scholar] [CrossRef] [PubMed]
  96. Sekandi, P.; Namukobe, J.; Byamukama, R.; Nagawa, C.B.; Barbini, S.; Bacher, M.; Böhmdorfer, S.; Rosenau, T. Antimicrobial, Antioxidant, and Sun Protection Potential of the Isolated Compounds from Spermacoce Princeae (K. Schum). BMC Complement. Med. Ther. 2023, 23, 201. [Google Scholar] [CrossRef] [PubMed]
  97. Musini, A.; Singh, H.N.; Vulise, J.; Pammi, S.S.S.; Giri, A. Quercetin’s Antibiofilm Effectiveness against Drug Resistant Staphylococcus Aureus and Its Validation by in Silico Modeling. Res. Microbiol. 2024, 175, 104091. [Google Scholar] [CrossRef] [PubMed]
  98. Mu, Y.; Zeng, H.; Chen, W. Quercetin Inhibits Biofilm Formation by Decreasing the Production of EPS and Altering the Composition of EPS in Staphylococcus Epidermidis. Front. Microbiol. 2021, 12, 1058. [Google Scholar] [CrossRef] [PubMed]
  99. Mu, Y.Q.; Xie, T.T.; Zeng, H.; Chen, W.; Wan, C.X.; Zhang, L.L. Streptomyces-Derived Actinomycin D Inhibits Biofilm Formation via Downregulating Ica Locus and Decreasing Production of PIA in Staphylococcus Epidermidis. J. Appl. Microbiol. 2020, 128, 1201–1207. [Google Scholar] [CrossRef] [PubMed]
  100. Gopu, V.; Meena, C.K.; Shetty, P.H. Quercetin Influences Quorum Sensing in Food Borne Bacteria: In-Vitro and In-Silico Evidence. PLoS ONE 2015, 10, e0134684. [Google Scholar] [CrossRef]
  101. Aljuffali, I.A.; Hsu, C.-Y.; Lin, Y.-K.; Fang, J.-Y. Cutaneous Delivery of Natural Antioxidants: The Enhancement Approaches. Curr. Pharm. Des. 2015, 21, 2745–2757. [Google Scholar] [CrossRef]
  102. Hung, C.-F.; Fang, C.-L.; Al-Suwayeh, S.A.; Yang, S.-Y.; Fang, J.-Y. Evaluation of Drug and Sunscreen Permeation via Skin Irradiated with UVA and UVB: Comparisons of Normal Skin and Chronologically Aged Skin. J. Dermatol. Sci. 2012, 68, 135–148. [Google Scholar] [CrossRef]
  103. Lin, C.-F.; Leu, Y.-L.; Al-Suwayeh, S.A.; Ku, M.-C.; Hwang, T.-L.; Fang, J.-Y. Anti-Inflammatory Activity and Percutaneous Absorption of Quercetin and Its Polymethoxylated Compound and Glycosides: The Relationships to Chemical Structures. Eur. J. Pharm. Sci. 2012, 47, 857–864. [Google Scholar] [CrossRef] [PubMed]
  104. US Food and Drug Administration. Gras Notice for High-Purity Quercetin 2010. Available online: https://web.archive.org/web/20171031050012/http://www.fda.gov/downloads/food/ingredientspackaginglabeling/gras/noticeinventory/ucm269540.pdf (accessed on 16 May 2024).
  105. Bazzucchi, I.; Patrizio, F.; Ceci, R.; Duranti, G.; Sgrò, P.; Sabatini, S.; Di Luigi, L.; Sacchetti, M.; Felici, F. The Effects of Quercetin Supplementation on Eccentric Exercise-Induced Muscle Damage. Nutrients 2019, 11, 205. [Google Scholar] [CrossRef] [PubMed]
  106. Kumar, R.; Subramanian, V.; Nadanasabapathi, S. Health Benefits of Quercetin. Def. Life Sci. J. 2017, 2, 142. [Google Scholar] [CrossRef]
  107. Jazvinšćak Jembrek, M.; Čipak Gašparović, A.; Vuković, L.; Vlainić, J.; Žarković, N.; Oršolić, N. Quercetin Supplementation: Insight into the Potentially Harmful Outcomes of Neurodegenerative Prevention. Naunyn Schmiedebergs Arch. Pharmacol. 2012, 385, 1185–1197. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, R.; Lin, J.; Hong, J.; Han, D.; Zhang, A.D.; Lan, R.; Fu, L.; Wu, Z.; Lin, J.; Zhang, W.; et al. Potential Toxicity of Quercetin: The Repression of Mitochondrial Copy Number via Decreased POLG Expression and Excessive TFAM Expression in Irradiated Murine Bone Marrow. Toxicol. Rep. 2014, 1, 450–458. [Google Scholar] [CrossRef] [PubMed]
  109. Mieszkowski, J.; Pałys, A.; Budzisz, E. Quercetin—Structure, Function and Clinical Usage. Farm. Pol. 2011, 67, 18–23. [Google Scholar]
  110. Wangsawangrung, N.; Choipang, C.; Chaiarwut, S.; Ekabutr, P.; Suwantong, O.; Chuysinuan, P.; Techasakul, S.; Supaphol, P. Quercetin/Hydroxypropyl-β-Cyclodextrin Inclusion Complex-Loaded Hydrogels for Accelerated Wound Healing. Gels 2022, 8, 573. [Google Scholar] [CrossRef] [PubMed]
  111. Li, J.; Li, Y.; Guo, C.; Wu, X. Development of Quercetin Loaded Silk Fibroin/Soybean Protein Isolate Hydrogels for Burn Wound Healing. Chem. Eng. J. 2024, 481, 148458. [Google Scholar] [CrossRef]
  112. Zawani, M.; Maarof, M.; Tabata, Y.; Motta, A.; Fauzi, M.B. Quercetin-Embedded Gelastin Injectable Hydrogel as Provisional Biotemplate for Future Cutaneous Application: Optimization and In Vitro Evaluation. Gels 2022, 8, 623. [Google Scholar] [CrossRef]
Table 1. Chosen quercetin sources.
Table 1. Chosen quercetin sources.
ReferenceAmount
[mg 100 g−1]
FormProduct
[16]4.0 *Fresh cropApple
[16,17]14.0–23.6 *Fresh cropAsparagus
[16,18]2.4–14.6 *Fresh cropBlueberry
[18]15.8 *Fresh cropWhortleberry
[18]14.6 *Fresh cropLingonberry
[18]8.9 *Fresh cropChokeberry
[18]6.3 *Fresh cropRowanberry
[18]5.6 *Fresh cropCrowberry
[18]0.6 ***JuiceElderberry
[16,18]12.1–25.0 *Fresh cropCranberry
[16]17.4 *Fresh cropCherry
[19,20]0.46 ***Juice from fresh Italian grape (Chianti)Grape
[21]54–119 **Grape waste (pomace): Methanolic extract from waste (skins, stalks, seeds)
[20]0.62–1.02 **Dried grape leaves
[20]0.68–0.82 **Dried grape stems
[16]3.16 ***BeverageRed wine
[16]22.6 *Fresh cropKale
[16]14.7 *Fresh cropLettuce
[17]12.0 *Fresh cropRomaine lettuce
[17]30.6–10.3 *Fresh cropRed leaf lettuce
[17]1.6 *Fresh cropTomato
[16,17]11.0–45.0 *Fresh cropYellow onion
[22]792.0 **Waste (peels): water and ethanol extracts
[22]41.9 **Edible part water and ethanol extractsRed onion
[22]1625.0 *Waste (peels) water and ethanol extracts
[22]40.0 **Edible part water and ethanol extractsRed shallot
[22]1161.0 *Waste (peels) water and ethanol extracts
* Amount presented in fresh weight (FW). ** Amount presented in dry weight (DW). *** Amount presented in mg/100 mL.
Table 2. List of abbreviations and their descriptions.
Table 2. List of abbreviations and their descriptions.
Full NameAbbreviation
Kelch-like ECH-associated protein 1KEAP1
nuclear respiratory factor 2NRF2
antioxidant response elementARE
4-hydroxynonenal4-HNE
3,4-methylenedioxyamphetamineMDA
superoxide dismutaseSOD
GlutathioneGSH
CatalaseCAT
QuercetinQUE
cyclooxygenase-2COX-2
inducible nitric oxide synthaseINOS
activator protein-1AP-1
p38 mitogen-activated protein kinasesp38PAPK
nuclear factor-κBNF-κB
NLR family pyrin domain containing 3NLRP3
tumor necrosis factor-αTNF-α
monocyte chemoattractant protein-1MCP-1
interleukin-10IL-10
interleukin-6IL-6
interleukin-1βIL-1β
interleukin-8IL-8
protein kinase BAKT
mammalian target of rapamycinmTOR
extracellular signal-regulated kinase familyERK
extracellular signal-related kinases 1 and 2ERK1/2
kinase c-Jun N terminalJNK
signal transducers and activator of transcription 3STAT3
protein kinase CδPKCδ
Janus kinase-2JAK-2
phosphoinositide-dependent protein kinase-1PDK-1
heme oxygenase-1HO-1
human fetal lung fibroblast-1HFL-1
transforming growth factor-β1TGB-β1
vascular endothelial growth factorVEGF
transforming growth factor-β1TGF-β1
hypoxia-inducible factor-1HIF-1
phosphoinositide 3-kinasePI3K
mitogen-activated protein kinasesMAPK
matrix metalloproteinase-1MMP-1
matrix metalloproteinase-2MMP-2
matrix metalloproteinase-9MMP-9
extracellular signal-regulated kinase 1ERK1
signal transducers and activator of transcription 6STAT6
CC chemokine ligand 17CCL17
CC chemokine ligand 22CCL22
interleukin-4IL-4
interferon gammaIFN-γ
Toll-like receptor-2TLR-2
growth-associated protein-43GAP-43
matrix metalloproteinase-8MMP-8
MyeloperoxidaseMPO
caspase 3CASP3
NAD(P)H quinone dehydrogenase 1NQO1
Thymic stromal lymphopoietinTSLP
Superoxide dismutase 1SOD1
Superoxide dismutase 2SOD2
glutathione peroxidaseGPX
transient receptor potential cation channel subfamily V member 1TRPV1
Table 3. Mechanism of action of quercetin.
Table 3. Mechanism of action of quercetin.
ReferenceMOACompound
Anti-aging effects and UV protection
[35]Inhibition of MMP-1 and COX-2 expressionquercetin
[35,38]Inhibition of AP-1 and NF-κB activityquercetin
[38]Inhibition of IL-1β, IL-6, IL-8, and TNF-α inflammatory cytokinesquercetin
[39]Inhibition of PDK-1 and AKT phosphorylationquercetin, caffeic acid ester and apigenin
[43]Reduction of UVA radiation effectsquercetin + rutin + titanium dioxide
[44]Prevention of ROS creation, GSH depletion, CASP3 activation.
Increased HO-1, NQO1, CAT expression
quercetin
[38,46]Reduction in GSH depletion, reduction in metalloproteinase activityquercetin
[47]Blocked ROS production, slowed leakage of cytochrome c, inhibition of keratinocyte apoptosisquercetin
[41]Activation of proteasome, improvement in HFL-1 fibroblasts viabilityquercetin, quercetin caprylate
Anti-melanogenesis and skin-whitening
[49,50,51]Inhibition of tyrosinasequercetin
[36]Reduction in melanin contentquercetin + vitamin C + arbutin
[52,53,54]Upregulation of tyrosinase activityquercetin
Wound healing, reduction in skin irritation and scarring prevention
[59]Inhibition of free radicals’ activityquercetin
[60]TGB-β1 and VEGF activation,
Promotion of fibroblast proliferation, TNF-α attenuation, inducing IL-10 levels
quercetin
[61]Increasing IL-10, VEGF, TGF-1 expressions, and decreasing expressions of TNF-αquercetin in DMSO
[62]Reduction in integrin αV level;
increasing integrin β1 expressions
quercetin
[66]Reduction in cell proliferation and collagen synthesis; inducing apoptosisquercetin + vitamin D
[64]Inhibition of collagen synthesis in fibroblastsquercetin + X-ray
[67]Inhibition of HIF-1 expression
Reduction in AKT phosphorylation
quercetin
[65]Improvement in hypertrophic and keloid scarsquercetin
[68]Inhibition of MAPK signaling pathway;
Reduction in inflammatory cytokines concentration
quercetin
[70,73]Reduction in IL-1β, IL-6, IL-8 expressions
Increasing SOD1, SOD2, GPx, catalase expressions, IL-10
Increasing mRNA, E-cadherin, occludin expressions
Decreasing MMP1, MMP2 and MMP9 expressions
Inhibition of ERK1/2 MAPK phosphorylation
Decreasing NF-κB expression
quercetin
[71]Reduction in CCL17, CCL22, IL-4, IL-6, IFN-γ and TNF-α expressionsquercetin
[33]Reduction in TLR-2 production
Inhibition of p38, ERK and JNK MAPK
Decreasing mRNA level for MMP-9
quercetin
[74]Inhibition of malondialdehyde productionquercetin encapsulated in liposomes
[78]Inhibition of inflammatory cytokines,
enhancing free radical scavenging ability, increasing reepithelialization
quercetin, chitosan, alginate
[79]Decreasing histamine 4 receptor-induced calcium influx through the TRPV1 channelquercetin
[81]Decreasing inflammatory cytokines levels and INOS-positive cells
Increasing CD206-positive cells activity
Induction of macrophage polarization to M2
quercetin
[82]Induction of VEGF, TGF-β
Increasing GAP-43 concentration
Reduction in MMP-9 and TNF-α
quercetin
Protection against skin oxidation, anti-inflammatory, and antiseptic properties
[85]Changing electron cloud density,
Inhibition of TNF-α, IL-1β and IL-6 cytokines
quercetin
[87]Antimicrobial activityquercetin on CuO nanorods
[89]Antimicrobial activityquercetin + copper complex + polycaprolactone
[90]Antimicrobial activityquercetin-loaded lipid carriers
[92]Antimicrobial activity regulation of MMP-8 and MPO activity
Increasing collagen and hydroxyproline deposition
extract of Rhus coriaria (quercetin)
[93]Antimicrobial activityExtract of Syncarpia hillii (Querciturone)
[94]Antimicrobial activityExtract of Opuntia spp. (quercetin and its glycosides)
[95]Antimicrobial activityExtract of Bridelia ferruginea (quercetin)
[96]Antimicrobial activity UV protectionExtract of z Spermacoce princeae (quercetin)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zaborowski, M.K.; Długosz, A.; Błaszak, B.; Szulc, J.; Leis, K. The Role of Quercetin as a Plant-Derived Bioactive Agent in Preventive Medicine and Treatment in Skin Disorders. Molecules 2024, 29, 3206. https://doi.org/10.3390/molecules29133206

AMA Style

Zaborowski MK, Długosz A, Błaszak B, Szulc J, Leis K. The Role of Quercetin as a Plant-Derived Bioactive Agent in Preventive Medicine and Treatment in Skin Disorders. Molecules. 2024; 29(13):3206. https://doi.org/10.3390/molecules29133206

Chicago/Turabian Style

Zaborowski, Michał Kazimierz, Anna Długosz, Błażej Błaszak, Joanna Szulc, and Kamil Leis. 2024. "The Role of Quercetin as a Plant-Derived Bioactive Agent in Preventive Medicine and Treatment in Skin Disorders" Molecules 29, no. 13: 3206. https://doi.org/10.3390/molecules29133206

Article Metrics

Back to TopTop