Next Article in Journal
Changes in Lipidomics, Metabolomics, and the Gut Microbiota in CDAA-Induced NAFLD Mice after Polyene Phosphatidylcholine Treatment
Next Article in Special Issue
Immunopharmacological Activities of Luteolin in Chronic Diseases
Previous Article in Journal
Dimethyl Fumarate Treatment Reduces the Amount but Not the Avidity of the Epstein–Barr Virus Capsid-Antigen-Specific Antibody Response in Multiple Sclerosis: A Pilot Study
Previous Article in Special Issue
β-Caryophyllene Acts as a Ferroptosis Inhibitor to Ameliorate Experimental Colitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer

1
Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
2
School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
3
Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1498; https://doi.org/10.3390/ijms24021498
Submission received: 25 December 2022 / Revised: 8 January 2023 / Accepted: 10 January 2023 / Published: 12 January 2023

Abstract

:
The skin is the main barrier between the body and the environment, protecting it from external oxidative stress induced by ultraviolet rays. It also prevents the entrance of infectious agents such as viruses, external antigens, allergens, and bacteria into our bodies. An overreaction to these agents causes severe skin diseases, including atopic dermatitis, pruritus, psoriasis, skin cancer, and vitiligo. Members of the flavonoid family include apigenin, quercetin, luteolin, and kaempferol. Of these, apigenin has been used as a dietary supplement due to its various biological activities and has been shown to reduce skin inflammation by downregulating various inflammatory markers and molecular targets. In this review, we deal with current knowledge about inflammatory reactions in the skin and the molecular mechanisms by which apigenin reduces skin inflammation.

1. Introduction

The skin may be divided into several layers according to their main functions, including the epidermis, dermis, and hypodermis [1]. The epidermis is the outermost layer of the skin and acts as a barrier to prevent disruption by external stimuli [2,3]. Moreover, it acts as a defense system to retain immune homeostasis against many pathogens, including bacteria, antigens, and viruses [4]. The epidermis consists largely of keratinocytes and some Langerhans cells (LC), melanocytes, and Merkel cells [5]. Among these, the Langerhans cell is a type of dendritic cell (DC) that can present the necessary antigen for the innate immune response. When LCs are activated by ultraviolet (UV) irradiation, E-cadherin is expressed at low levels in keratinocytes. As a result, migration of LCs into the lymph nodes occurs, and regulatory T cells can be further matured [6,7]. Idoyaga et al. [8] revealed that skin DCs can be targeted for immunomodulatory therapies. In the outer layers of the epidermis, the skin microbiome retains skin acidity to protect against external infections [9,10,11]. Organisms in the microbiome also interact with each other to form a community in the skin, and these interactions ensure immune homeostasis in the skin. On the other hand, dysregulation of the inflammatory equilibrium can induce skin immunological diseases [12,13,14], including atopic dermatitis (AD), pruritus, psoriasis, skin cancer, and vitiligo [15,16,17]. As part of the efforts to attenuate inflammatory symptoms, steroidal and non-steroidal drugs are mostly used as classical treatments [18,19,20,21,22]. However, because of the side effects of anti-inflammatory drugs, studies investigating natural compounds to replace chemical drugs have been more actively performed [23].
Apigenin (4′,5,7-trihydroxyflavone, Figure 1) is a natural compound that belongs to the subclass of flavonoids [24,25]. In previous studies, Tanacetum, Achillea, Artemisia, and Matricaria genera belonging to the Artemisia family of plants have been reported as the main source plants of apigenin [26,27,28,29]. It has been presented that apigenin has the potential to attenuate skin inflammatory conditions, such as AD, pruritus, and psoriasis, and the tumorigenic response of skin cancers (Figure 2). In addition, its anti-apoptotic [30], anti-inflammatory [31], and anti-hyperglycemic effects [32] have been researched. In this review, we focus on describing the functional properties of apigenin and its potential for pharmacological effects.

2. Anti-Inflammatory Effects of Apigenin on UV-Irradiated Skin

UV light is the main cause of skin inflammation and can be divided into UVA (320–400 nm), UVB (280–320 nm), and UVC (100–280 nm), according to the wavelength. Especially, UVA and UVB penetrate the skin layers, and can induce skin inflammation and aging in keratinocytes and fibroblasts [33,34]. UVB exposure causes an acute inflammation response in the epidermis by promoting the synthesis and secretion of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α and interleukin (IL)-6 from epidermal cells [35]. Moreover, UVA penetrates the dermal layers and indirectly causes DNA damage with degradation of the collagen and elastin fibers [34].
In previous studies, apigenin attenuated skin inflammation by downregulating the expression of cyclooxygenase-2 (COX-2) [36]. Another finding suggests its potential as a novel target for reducing skin inflammation. This compound works in the keratinocyte by targeting the non-receptor tyrosine kinase (e.g., Src) and COX-2 [37]. Apigenin prevented cyclobutene pyrimidine dimers, which are generated by UV exposure. Due to this, apigenin restored the lower level of nucleotide excision repair proteins and affected cell apoptosis [38,39]. Apigenin downregulated the level of metalloproteinase-1 by interfering with the Ca2+ influx-dependent mitogen-activated protein kinase (MAPK) and activator protein-1 (AP-1) pathways in HaCaT or normal human dermal fibroblast cells under UVA-irradiated conditions. Especially, the levels of c-Jun and c-Fos were decreased by apigenin treatment, which suppressed the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 [40,41]. As a result, apigenin could attenuate UV-mediated inflammation by decreasing the transcription of inflammatory cytokines via the downregulation of the AP-1, MAPK, and apoptotic signaling pathways (Figure 3).

3. Effect of Apigenin on Attenuating AD

AD is a chronic inflammatory disease that affects 80% of patients in infancy or childhood. Its severe symptoms include itching, dry skin, eczema, and swallowing. The cytokines and chemokines secreted in AD are summarized in Table 1; however, the exact mechanism of the stimulus is still unclear. There are many hypotheses to explain AD pathogenesis, including: (I) disproportion of skin microbiomes [3,42,43,44]; (II) weakness of skin barrier junctions [45,46]; (III) dysregulation between pro- and anti-inflammatory cytokines [47,48]; and (IV) excessive immunoglobulin E (IgE) secretion. Among these explanations, apigenin reduced IgE and interferon (IFN)-γ levels in serum in an NC/Nga mouse model. Moreover, apigenin attenuated damaging skin lesions induced by picrylchloride. Considering the protein levels, apigenin suppressed the phosphorylation of the signal transducer and activator of transcription 6 (STAT6) in IL-4–stimulated mouse spleen cells [49]. In addition, apigenin showed a low expression of IL-31 in messenger RNA in a human mast cell line (HMC-1). In HMC-1 cells, apigenin downregulated nuclear factor-κB (NF-κB) pathway proteins, including the inhibitor of κB kinase, inhibitor of κB, and the p65/NF-κB, and MAPK pathway factors of c-Jun N-terminal kinase, ERK, and p38 [50]. In summary, it is thought that apigenin can ameliorate the symptoms of AD by decreasing the levels of pro-inflammatory cytokines and inflammatory mediators by downregulating the MAPK, NF-κB, and Jak/STAT signaling pathways (Figure 4).

4. Treatment with Apigenin for Alleviating Pruritus

Pruritus or itching negatively affects quality of life [65,66]. Many different factors play a role in this event, but the exact pathogenetic mechanisms are not known [67,68]. Histamine, serotonin, cytokines, peptides, and phospholipid metabolites are included as mediators of pruritus [69]. Among these, cytokines are strong players that manage itching by activating receptors [70]. One example, IL-31, which is derived from the IL-6 family, acts as the therapeutic target of pruritus in the Th2 cell-mediated response. IL-31 has been reported to induce chemokines such as CCL1, CCL17, and CCL22 [71,72]. Additionally, IL-33 has also been considered a pathophysiologically important cytokine that manages innate immune responses [73] and Th2 cell differentiation by promoting the expression of chemokines and pro-inflammatory cytokines, and by activating natural killer cells and dendritic cells [74]. Apigenin weakened the expression of IL-31 in human mast cells and mouse skin through downregulation of MAPK and NF-κB signaling [50]. In an ovalbumin-induced BALB/c mouse model, apigenin regulated the balance of Th1/Th2 cells by downregulating the NF-κB pathway and reducing histamine, IgE, and STAT1 expression. Moreover, apigenin improved the Th1 response by controlling the expression of IFN-γ and T-box protein expressed in T cells [75]. Furthermore, apigenin-treated microglial cells lowered the expression levels of IL-31 and IL-33 without displaying cytotoxicity. This expression was verified by polymerase chain reaction as well as Western blotting via the inhibition of the ERK and JNK pathways [76]. In experiments using astrocytes, apigenin significantly suppressed IL-31 and IL-33 messenger RNA expression. Pre-treatment with apigenin in astrocytes decreased the expression levels of IL-31 and IL-33 at the protein level. In astrocytes, apigenin also inhibited the phosphorylation of MAPK and NF-κB signaling proteins [77]. Taken together, these studies suggest that apigenin can ameliorate pruritus by inhibiting IL-31 and IL-33 secretion through suppression of the NF-κB and MAPK pathways (Figure 5).

5. The Mechanism of Apigenin for the Amelioration of Psoriasis

Psoriasis is a chronic, immunological skin disease affecting about 125 million patients in America [78]. These patients and others motivate the study of psoriasis to improve pathophysiological knowledge of the condition. Psoriasis is a sustained inflammatory disease caused by the hyperproliferation of keratinocytes and dysfunctional differentiation. In addition, the infiltration of Th17 cells secreting inflammatory cytokines, such as IL-23, into keratinocytes, dominantly occurs in psoriasis [79].
Apigenin showed the greatest effects in a psoriasis model by decreasing cytokine levels. Skin barrier recovery effects were observed in apigenin-treated skin. Apigenin also improved the skin’s condition by increasing the hydration level of the stratum corneum. Meanwhile, apigenin influenced the synthesis of skin structural proteins such as filaggrin, involucrin, and loricrin in mouse models [80]. With co-treatment of apigenin and lipopolysaccharide (LPS) in DCs, this compound significantly inhibited TNF-α messenger RNA expression. Moreover, apigenin suppressed the level of pro-inflammatory cytokines, including IFN-γ, IL-6, IL-1β, IL-23, and IL-10, in both LPS treatment and non-treatment groups. Meanwhile, apigenin affected naïve T cell differentiation by modulating the function of DCs [81]. Overall, apigenin treatment may ameliorate psoriasis symptoms by regulating the transcription of inflammatory cytokines via regulation of the Toll-like receptor 4 pathway (Figure 6).

6. The Suppressive Activity of Apigenin on Skin Cancer

The inflammation response promotes cell proliferation to renew damaged cell tissues, so it plays a pivotal role in retaining tissue homeostasis [82]. However, chronic inflammation is also known to induce tumorigenesis. The tumor microenvironment is initiated by the excessive production of inflammatory cytokines. Thus, this phenomenon merits study. Many cytokines and chemokines can be induced in hypoxic conditions in this tumorigenic environment [83]. Skin cancer is a malignant tumor, particularly in Caucasians, with about 1 million cases occurring annually in the United States [84,85,86]. Skin tumors have been named according to their involved cells and clinical behavior. There are three types of skin tumors: basal cell carcinoma; cutaneous malignant melanoma (CM); and non-melanocytic skin cancer (NMSC), which is also known as squamous cell carcinoma [84,87]. Chronic UV exposure most commonly leads to skin cancer among all known risk environments and affects gene mutation, immunosuppression, and oxidative stress [88,89,90]. From various studies, there are several approaches to suppress skin cancer, including through the PI3K/Akt/mTOR, TNF-related apoptosis-inducing ligand, JAK/STAT, and MAPK signaling pathways [91,92,93,94].

6.1. NMSC

Apigenin downregulates the Akt signaling pathway in UVB-irradiated keratinocytes, blocking the mammalian target of rapamycin (mTOR) activation and suppressing the cell cycle and cell proliferation in mouse skin and keratinocytes. Meanwhile, it promotes autophagy via mTOR inhibition, which inhibits keratinocyte proliferation [95,96]. In primary human epidermal keratinocytes and a skin cutaneous squamous cell carcinoma cell line (COLO-16), treatment with apigenin decreased the conversion of the microtubule-associated protein 1 light chain 3 (LC3) and GFP-LC3 puncta [97]. Apigenin also inhibited skin carcinogenesis by downregulating the COX-2 expression level in UVB-irradiated mouse skins [98]. It is commonly known that 12-O-tetradecanoylphorbol-13-acetate (TPA) can induce a tumor by binding and activating the protein kinase C signaling pathways [99,100]. Apigenin treatment suppressed PKC activity dose-dependently and inhibited TPA-mediated carcinogenesis in mouse skin [101,102]. In summary, it is speculated that apigenin treatment can reduce tumorigenic responses by inducing autophagy and via the inactivation of Akt and PKC in keratinocytes (Figure 7).

6.2. CMs

Apigenin inhibited the proliferation of melanoma cell lines by downregulating the AKT signaling pathway, which promotes cell apoptosis [103]. Moreover, apigenin treatment suppressed melanoma metastasis to the lungs in C57BL/6 mice and inhibited the phosphorylation of STAT3 in melanoma cells [104]. The presence of apigenin induced anti-melanoma effects by triggering the apoptosis of A375SM cells. Apoptotic proteins, including the caspases, p53, Bcl-2-associated X protein, and poly ADP-ribose polymerase (PARP), were upregulated by apigenin treatment. The compound also downregulated the levels of Akt, STAT3, and MAPK in melanoma cells [105]. Therefore, it is suggested that apigenin could inhibit carcinogenesis by inducing apoptosis in melanoma cells and downregulating the activities of some important survival factors, such as STAT3, Akt, and MAPK proteins (Figure 8).

7. The Therapeutic Effects of Apigenin on Vitiligo

Vitiligo has been reported as a pigmentary disorder impacting about 1% of the world’s population [106]. The symptoms of this disease include inconsistent and various sizes of white spots found on the skin and a change of hair color to white [107,108,109]. The main cause of vitiligo is concerned with the autoimmune chronic destruction of melanocytes. The death of pigment cells expands the white lesions on the skin. Therefore, the therapeutic strategy has focused on preventing the apoptosis of melanocytes from oxidative stress and suppressing the proinflammatory response [110]. Of many different compounds, apigenin was reported to upregulate antioxidant enzyme activities, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). Moreover, apigenin was found to promote the gene expression level that is involved in the antioxidant process the at mRNA and protein levels [111]. Meanwhile, it was revealed that apigenin can affect dopamine (DA)-triggered apoptosis in melanocytes by downregulating cleaved PARP and cleaved caspase 3 levels [112]. Apigenin also protected melanocytes from apoptosis by blocking the phosphorylation of Akt, p38, and JNK, which are induced by DA [112] (Figure 9).

8. Conclusions

This review described the attenuating effects of apigenin on skin inflammatory conditions and cancer as summarized in Table 2. A natural flavonoid, apigenin showed the greatest activity by attenuating the symptoms of skin inflammatory diseases and tumorigenic responses. Apigenin downregulated inflammatory cytokine expression by suppressing the AP-1, MAPK, and NF-κB pathways in keratinocytes. In addition, apigenin induced autophagy by decreasing mTOR activity and inactivating Akt and PKC activities. Moreover, apigenin protects the cell from oxidative stress-induced cell death. Through treatment, apigenin could prevent skin inflammatory responses to retain the proper regulation of inflammatory cells. Furthermore, apigenin could affect the synthesis of skin barrier factors and Ca2+ influx. Based on this review, apigenin could be applied to treat skin inflammatory diseases and cancer.

9. Perspective

Apigenin is a bioactive compound used as a therapeutic agent for various diseases, such as diabetes, Alzheimer’s disease, cancer, and amnesia [113,114,115,116]. Treatment with apigenin has led to decreased levels of many inflammatory cytokines. According to study results, apigenin could inhibit the inflammatory response in the skin by downregulating transcription factors, such as AP-1, NF-κB, and STAT. These mechanisms not only back up the excellence of apigenin, but also suggest the possibility of using it as a drug for inflammatory skin diseases. There are now many trials assessing active natural compounds as substitutes for chemically synthetic drugs because of severe side effects associated with the latter [117,118]. Previous studies administered apigenin by applying it to damaged skin or cells to attenuate skin inflammation. Nowadays, apigenin products manufactured with chamomile extracts are being sold in markets to attenuate stress hormones, and bad dreams, as well as in the form of commercially available capsules that provide powerful antioxidant supplements to promote healthy aging and skin health [119]. However, flavonoids can be degraded by high temperatures [120], thus studies aimed at stabilizing apigenin therapeutics should be performed. We suggest that apigenin can not only be used as a therapeutic material, but also as a health supplement for skin diseases, based upon the various studies discussed in this review.

Author Contributions

J.H.Y., M.-Y.K. and J.Y.C. conceptualized this review; J.H.Y. visualized the figure for the paper; J.H.Y., M.-Y.K. and J.Y.C. wrote the manuscript; M.-Y.K. and J.Y.C. administrated the writing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by grant from the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Korean Ministry of Education (Grant No.: 2017R1A6A1A03015642).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

LCLangerhans cells
UVUltraviolet radiation
TregRegulatory T cells
ADAtopic dermatitis
TNF-αTumor necrosis factor-α
IL-6Interleukin-6
COX-2Cyclooxygenase-2
SrcNon-receptor tyrosine kinase
CPDCyclobutene pyrimidine dimers
NERNucleotide excision repair
MMP-1Metalloproteinases-1
MAPKMitogen-activated protein kinase
AP-1Activator protein 1
nHDFNormal Human Dermal Fibroblasts
IgEImmunoglobulin E
IFN-γInterferon-gamma
PiClPicrylchloride
STAT6Signal transducers and activators of transcription 6
IL-4Interleukin-4
IL-31Interleukin-31
HMC-1Human mast cells
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
IKKInhibitor of NF-κB kinase
IκBInhibitor of kappaB
JNKc-Jun N-terminal kinase
ERKExtracellular signal-regulated kinase
CCL26Eosinophil chemokine
IL-31RAInterleukin-31 receptor A
ILC2Innate lymphoid cells-2
CXCR3C-X-C Motif Chemokine Receptor 3
TSLPThymic stromal lymphopoietin
CCLC-C motif chemokine ligand
NK cellsNatural killer cells
OVAOvalbumin
T-betT-box protein expressed in T cells
IL-23Interleukin-23
LPSLipopolysaccharide
TLR4Toll-like receptor 4
TMETumor microenvironments
BCCsBasal cell carcinomas
CMsCutaneous malignant melanomas
NMSCNonmelanocytic skin cancer
SCCsSquamous cell carcinomas
TRAILTNF-related apoptosis-inducing ligand
mTORMammalian target of rapamycin
HEKsHuman epidermal keratinocytes
COLO-16Skin cutaneous squamous cell carcinoma cell line
LC3Light chain 3
TPA12-O-tetradecanoylphorbol-13-acetate
BaxBcl-2-associated X protein
PARPPoly ADP-ribose polymerase
SODSuperoxide dismutase
CATCatalase
GSH-PxGlutathione peroxidase

References

  1. Hsu, Y.-C.; Li, L.; Fuchs, E. Emerging interactions between skin stem cells and their niches. Nat. Med. 2014, 20, 847–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Belkaid, Y.; Tamoutounour, S. The influence of skin microorganisms on cutaneous immunity. Nat. Rev. Immunol. 2016, 16, 353–366. [Google Scholar] [CrossRef] [PubMed]
  3. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef] [PubMed]
  4. Nestle, F.O.; Di Meglio, P.; Qin, J.-Z.; Nickoloff, B.J. Skin immune sentinels in health and disease. Nat. Rev. Immunol. 2009, 9, 679–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Abdo, J.M.; Sopko, N.A.; Milner, S.M. The applied anatomy of human skin: A model for regeneration. Wound Med. 2020, 28, 100179. [Google Scholar] [CrossRef]
  6. Tang, A.; Amagai, M.; Granger, L.G.; Stanley, J.R.; Uddy, M.C. Adhesion of epidermal Langerhans cells to keratinocytes mediated by E-cadherin. Nature 1993, 361, 82–85. [Google Scholar] [CrossRef]
  7. Schwarz, A.; Noordegraaf, M.; Maeda, A.; Torii, K.; Clausen, B.E.; Schwarz, T. Langerhans cells are required for UVR-induced immunosuppression. J. Investig. Dermatol. 2010, 130, 1419–1427. [Google Scholar] [CrossRef] [Green Version]
  8. Idoyaga, J.; Fiorese, C.; Zbytnuik, L.; Lubkin, A.; Miller, J.; Malissen, B.; Mucida, D.; Merad, M.; Steinman, R.M. Specialized role of migratory dendritic cells in peripheral tolerance induction. J. Clin. Investig. 2013, 123, 844–854. [Google Scholar] [CrossRef] [Green Version]
  9. Scharschmidt, T.C.; Fischbach, M.A. What lives on our skin: Ecology, genomics and therapeutic opportunities of the skin microbiome. Drug Discov. Today: Dis. Mech. 2013, 10, e83–e89. [Google Scholar] [CrossRef] [Green Version]
  10. Belkaid, Y.; Segre, J.A. Dialogue between skin microbiota and immunity. Science 2014, 346, 954–959. [Google Scholar] [CrossRef]
  11. Grice, E.A. The intersection of microbiome and host at the skin interface: Genomic-and metagenomic-based insights. Genome Res. 2015, 25, 1514–1520. [Google Scholar] [CrossRef] [PubMed]
  12. McLoughlin, I.J.; Wright, E.M.; Tagg, J.R.; Jain, R.; Hale, J.D. Skin microbiome—The next frontier for probiotic intervention. Probiotics Antimicrob. Proteins 2022, 14, 630–647. [Google Scholar] [CrossRef] [PubMed]
  13. Iebba, V.; Totino, V.; Gagliardi, A.; Santangelo, F.; Cacciotti, F.; Trancassini, M.; Mancini, C.; Cicerone, C.; Corazziari, E.; Pantanella, F. Eubiosis and dysbiosis: The two sides of the microbiota. New Microbiol 2016, 39, 1–12. [Google Scholar]
  14. Di Meglio, P.; Perera, G.K.; Nestle, F.O. The multitasking organ: Recent insights into skin immune function. Immunity 2011, 35, 857–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Robert, C.; Kupper, T.S. Inflammatory Skin Diseases, T Cells, and Immune Surveillance. New Engl. J. Med. 1999, 341, 1817–1828. [Google Scholar] [CrossRef]
  16. Balato, A.; Cacciapuoti, S.; Di Caprio, R.; Marasca, C.; Masarà, A.; Raimondo, A.; Fabbrocini, G. Human Microbiome: Composition and Role in Inflammatory Skin Diseases. Arch. Immunol. Et Ther. Exp. 2019, 67, 1–18. [Google Scholar] [CrossRef]
  17. Maru, G.B.; Gandhi, K.; Ramchandani, A.; Kumar, G. The role of inflammation in skin cancer. Adv. Exp. Med. Biol. 2014, 816, 437–469. [Google Scholar]
  18. Abdulla, A.; Adams, N.; Bone, M.; Elliott, A.M.; Gaffin, J.; Jones, D.; Knaggs, R.; Martin, D.; Sampson, L.; Schofield, P. Guidance on the management of pain in older people. Age Ageing 2013, 42, i1–i57. [Google Scholar] [CrossRef] [Green Version]
  19. Breivik, H.; Collett, B.; Ventafridda, V.; Cohen, R.; Gallacher, D. Survey of chronic pain in Europe: Prevalence, impact on daily life, and treatment. Eur. J. Pain 2006, 10, 287–333. [Google Scholar] [CrossRef]
  20. Conaghan, P.G. A turbulent decade for NSAIDs: Update on current concepts of classification, epidemiology, comparative efficacy, and toxicity. Rheumatol. Int. 2012, 32, 1491–1502. [Google Scholar] [CrossRef] [Green Version]
  21. Melmed, S.; Polonsky, K.S.; Larsen, P.R.; Kronenberg, H.M. Williams Textbook of Endocrinology E-Book; Elsevier Health Sciences: Amsterdam, The Netherlands, 2015. [Google Scholar]
  22. Emery, P. Treatment of rheumatoid arthritis. Bmj 2006, 332, 152–155. [Google Scholar] [CrossRef] [Green Version]
  23. Borquaye, L.S.; Darko, G.; Laryea, M.K.; Roberts, V.; Boateng, R.; Gasu, E.N. Anti-inflammatory activities of extracts from Oliva sp., Patella rustica, and Littorina littorea collected from Ghana’s coastal shorelines. Cogent Biol. 2017, 3, 1364063. [Google Scholar] [CrossRef]
  24. Tang, D.; Chen, K.; Huang, L.; Li, J. Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Expert Opin. Drug Metab. Toxicol. 2017, 13, 323–330. [Google Scholar] [CrossRef]
  25. Wang, M.; Firrman, J.; Liu, L.; Yam, K. A Review on Flavonoid Apigenin: Dietary Intake, ADME, Antimicrobial Effects, and Interactions with Human Gut Microbiota. BioMed Res. Int. 2019, 2019, 7010467. [Google Scholar] [CrossRef] [PubMed]
  26. Ornano, L.; Venditti, A.; Donno, Y.; Sanna, C.; Ballero, M.; Bianco, A. Phytochemical analysis of non-volatile fraction of Artemisia caerulescens subsp. densiflora (Viv.)(Asteraceae), an endemic species of La Maddalena Archipelago (Sardinia–Italy). Nat. Prod. Res. 2016, 30, 920–925. [Google Scholar] [CrossRef] [PubMed]
  27. Venditti, A.; Maggi, F.; Vittori, S.; Papa, F.; Serrilli, A.M.; Di Cecco, M.; Ciaschetti, G.; Mandrone, M.; Poli, F.; Bianco, A. Antioxidant and α-glucosidase inhibitory activities of Achillea tenorii. Pharm. Biol. 2015, 53, 1505–1510. [Google Scholar] [CrossRef] [PubMed]
  28. Sharifi-Rad, M.; Nazaruk, J.; Polito, L.; Morais-Braga, M.F.B.; Rocha, J.E.; Coutinho, H.D.M.; Salehi, B.; Tabanelli, G.; Montanari, C.; del Mar Contreras, M. Matricaria genus as a source of antimicrobial agents: From farm to pharmacy and food applications. Microbiol. Res. 2018, 215, 76–88. [Google Scholar] [CrossRef]
  29. Venditti, A.; Frezza, C.; Sciubba, F.; Serafini, M.; Bianco, A.; Cianfaglione, K.; Lupidi, G.; Quassinti, L.; Bramucci, M.; Maggi, F. Volatile components, polar constituents and biological activity of tansy daisy (Tanacetum macrophyllum (Waldst. et Kit.) Schultz Bip.). Ind. Crops Prod. 2018, 118, 225–235. [Google Scholar] [CrossRef]
  30. Zhou, Z.; Zhang, Y.; Lin, L.; Zhou, J. Apigenin suppresses the apoptosis of H9C2 rat cardiomyocytes subjected to myocardial ischemia-reperfusion injury via upregulation of the PI3K/Akt pathway. Mol. Med. Rep. 2018, 18, 1560–1570. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, J.; Liu, Y.-T.; Xiao, L.; Zhu, L.; Wang, Q.; Yan, T. Anti-Inflammatory Effects of Apigenin in Lipopolysaccharide-Induced Inflammatory in Acute Lung Injury by Suppressing COX-2 and NF-kB Pathway. Inflammation 2014, 37, 2085–2090. [Google Scholar] [CrossRef]
  32. Jung, U.J.; Cho, Y.-Y.; Choi, M.-S. Apigenin Ameliorates Dyslipidemia, Hepatic Steatosis and Insulin Resistance by Modulating Metabolic and Transcriptional Profiles in the Liver of High-Fat Diet-Induced Obese Mice. Nutrients 2016, 8, 305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mesa-Arango, A.C.; Flórez-Muñoz, S.V.; Sanclemente, G. Mechanisms of skin aging. Iatreia 2017, 30, 160–170. [Google Scholar] [CrossRef]
  34. Lephart, E.D. Equol’s anti-aging effects protect against environmental assaults by increasing skin antioxidant defense and ECM proteins while decreasing oxidative stress and inflammation. Cosmetics 2018, 5, 16. [Google Scholar] [CrossRef] [Green Version]
  35. Barker, J.N.; Griffiths, C.E.; Nickoloff, B.J.; Mitra, R.; Dixit, V.M. Keratinocytes as initiators of inflammation. Lancet 1991, 337, 211–214. [Google Scholar] [CrossRef] [Green Version]
  36. Tong, X.; Dross, R.T.V.; Abu-Yousif, A.; Morrison, A.R.; Pelling, J.C. Apigenin Prevents UVB-Induced Cyclooxygenase 2 Expression: Coupled mRNA Stabilization and Translational Inhibition. Mol. Cell. Biol. 2007, 27, 283–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Byun, S.; Park, J.; Lee, E.; Lim, S.; Yu, J.G.; Lee, S.J.; Chen, H.; Dong, Z.; Lee, K.W.; Lee, H.J. Src kinase is a direct target of apigenin against UVB-induced skin inflammation. Carcinogenesis 2012, 34, 397–405. [Google Scholar] [CrossRef] [Green Version]
  38. Britto, S.M.; Shanthakumari, D.; Agilan, B.; Radhiga, T.; Kanimozhi, G.; Prasad, N.R. Apigenin prevents ultraviolet-B radiation induced cyclobutane pyrimidine dimers formation in human dermal fibroblasts. Mutat. Res. /Genet. Toxicol. Environ. Mutagen. 2017, 821, 28–35. [Google Scholar] [CrossRef]
  39. García Forero, A.; Villamizar Mantilla, D.A.; Núñez, L.A.; Ocazionez, R.E.; Stashenko, E.E.; Fuentes, J.L. Photoprotective and Antigenotoxic Effects of the Flavonoids Apigenin, Naringenin and Pinocembrin. Photochem. Photobiol. 2019, 95, 1010–1018. [Google Scholar] [CrossRef]
  40. Hwang, Y.P.; Oh, K.N.; Yun, H.J.; Jeong, H.G. The flavonoids apigenin and luteolin suppress ultraviolet A-induced matrix metalloproteinase-1 expression via MAPKs and AP-1-dependent signaling in HaCaT cells. J Derm. Sci 2011, 61, 23–31. [Google Scholar] [CrossRef]
  41. Choi, S.; Youn, J.; Kim, K.; Joo da, H.; Shin, S.; Lee, J.; Lee, H.K.; An, I.S.; Kwon, S.; Youn, H.J.; et al. Apigenin inhibits UVA-induced cytotoxicity in vitro and prevents signs of skin aging in vivo. Int J Mol Med 2016, 38, 627–634. [Google Scholar] [CrossRef] [Green Version]
  42. Meisel, J.S.; Sfyroera, G.; Bartow-McKenney, C.; Gimblet, C.; Bugayev, J.; Horwinski, J.; Kim, B.; Brestoff, J.R.; Tyldsley, A.S.; Zheng, Q. Commensal microbiota modulate gene expression in the skin. Microbiome 2018, 6, 1–15. [Google Scholar] [CrossRef] [PubMed]
  43. Kong, H.H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; Murray, P.R. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012, 22, 850–859. [Google Scholar] [CrossRef] [PubMed]
  44. Shi, B.; Bangayan, N.J.; Curd, E.; Taylor, P.A.; Gallo, R.L.; Leung, D.Y.; Li, H. The skin microbiome is different in pediatric versus adult atopic dermatitis. J. Allergy Clin. Immunol. 2016, 138, 1233–1236. [Google Scholar] [CrossRef] [Green Version]
  45. Kim, B.E.; Leung, D.Y. Epidermal barrier in atopic dermatitis. Allergy Asthma Immunol. Res. 2012, 4, 12–16. [Google Scholar] [CrossRef] [Green Version]
  46. Rerknimitr, P.; Otsuka, A.; Nakashima, C.; Kabashima, K. The etiopathogenesis of atopic dermatitis: Barrier disruption, immunological derangement, and pruritus. Inflamm. Regen. 2017, 37, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. David Boothe, W.; Tarbox, J.A.; Tarbox, M.B. Atopic dermatitis: Pathophysiology. Manag. Atopic Dermat. 2017, 21–37. [Google Scholar]
  48. Tsakok, T.; Woolf, R.; Smith, C.; Weidinger, S.; Flohr, C. Atopic dermatitis: The skin barrier and beyond. Br. J. Dermatol. 2019, 180, 464–474. [Google Scholar] [CrossRef] [PubMed]
  49. Yano, S.; Umeda, D.; Yamashita, S.; Yamada, K.; Tachibana, H. Dietary apigenin attenuates the development of atopic dermatitis-like skin lesions in NC/Nga mice. J. Nutr. Biochem. 2009, 20, 876–881. [Google Scholar] [CrossRef]
  50. Che, D.N.; Cho, B.O.; Shin, J.Y.; Kang, H.J.; Kim, J.-S.; Oh, H.; Kim, Y.-S.; Jang, S.I. Apigenin Inhibits IL-31 Cytokine in Human Mast Cell and Mouse Skin Tissues. Molecules 2019, 24, 1290. [Google Scholar] [CrossRef] [Green Version]
  51. Oetjen, L.K.; Mack, M.R.; Feng, J.; Whelan, T.M.; Niu, H.; Guo, C.J.; Chen, S.; Trier, A.M.; Xu, A.Z.; Tripathi, S.V. Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch. Cell 2017, 171, 217–228. e213. [Google Scholar] [CrossRef] [Green Version]
  52. Gandhi, N.A.; Bennett, B.L.; Graham, N.M.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2 inflammation in disease. Nat. Rev. Drug Discov. 2016, 15, 35–50. [Google Scholar] [CrossRef] [PubMed]
  53. Cornelissen, C.; Marquardt, Y.; Czaja, K.; Wenzel, J.; Frank, J.; Lüscher-Firzlaff, J.; Lüscher, B.; Baron, J.M. IL-31 regulates differentiation and filaggrin expression in human organotypic skin models. J. Allergy Clin. Immunol. 2012, 129, 426–433. e428. [Google Scholar] [CrossRef] [PubMed]
  54. Yosipovitch, G.; Berger, T.; Fassett, M. Neuroimmune interactions in chronic itch of atopic dermatitis. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 239–250. [Google Scholar] [CrossRef] [PubMed]
  55. Kezic, S.; O’Regan, G.M.; Lutter, R.; Jakasa, I.; Koster, E.S.; Saunders, S.; Caspers, P.; Kemperman, P.M.J.H.; Puppels, G.J.; Sandilands, A.; et al. Filaggrin loss-of-function mutations are associated with enhanced expression of IL-1 cytokines in the stratum corneum of patients with atopic dermatitis and in a murine model of filaggrin deficiency. J. Allergy Clin. Immunol. 2012, 129, 1031–1039. e1031. [Google Scholar] [CrossRef] [Green Version]
  56. Archer, N.K.; Jo, J.-H.; Lee, S.K.; Kim, D.; Smith, B.; Ortines, R.V.; Wang, Y.; Marchitto, M.C.; Ravipati, A.; Cai, S.S. Injury, dysbiosis, and filaggrin deficiency drive skin inflammation through keratinocyte IL-1α release. J. Allergy Clin. Immunol. 2019, 143, 1426–1443. e1426. [Google Scholar] [CrossRef] [Green Version]
  57. Roan, F.; Obata-Ninomiya, K.; Ziegler, S.F. Epithelial cell–derived cytokines: More than just signaling the alarm. J. Clin. Investig. 2019, 129, 1441–1451. [Google Scholar] [CrossRef] [Green Version]
  58. Ryu, W.-I.; Lee, H.; Bae, H.C.; Ryu, H.J.; Son, S.W. IL-33 down-regulates filaggrin expression by inducing STAT3 and ERK phosphorylation in human keratinocytes. J. Dermatol. Sci. 2016, 82, 131–134. [Google Scholar] [CrossRef]
  59. Savinko, T.; Matikainen, S.; Saarialho-Kere, U.; Lehto, M.; Wang, G.; Lehtimäki, S.; Karisola, P.; Reunala, T.; Wolff, H.; Lauerma, A. IL-33 and ST2 in atopic dermatitis: Expression profiles and modulation by triggering factors. J. Investig. Dermatol. 2012, 132, 1392–1400. [Google Scholar] [CrossRef] [Green Version]
  60. Albanesi, C.; Scarponi, C.; Sebastiani, S.; Cavani, A.; Federici, M.; De Pità, O.; Puddu, P.; Girolomoni, G. IL-4 enhances keratinocyte expression of CXCR3 agonistic chemokines. J. Immunol. 2000, 165, 1395–1402. [Google Scholar] [CrossRef] [Green Version]
  61. Wang, Y.H.; Liu, Y.J. Thymic stromal lymphopoietin, OX40-ligand, and interleukin-25 in allergic responses. Clin. Exp. Allergy 2009, 39, 798–806. [Google Scholar] [CrossRef] [Green Version]
  62. Cianferoni, A.; Spergel, J. The importance of TSLP in allergic disease and its role as a potential therapeutic target. Expert Rev. Clin. Immunol. 2014, 10, 1463–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Xie, Y.; Takai, T.; Chen, X.; Okumura, K.; Ogawa, H. Long TSLP transcript expression and release of TSLP induced by TLR ligands and cytokines in human keratinocytes. J. Dermatol. Sci. 2012, 66, 233–237. [Google Scholar] [CrossRef] [PubMed]
  64. Chieosilapatham, P.; Ogawa, H.; Niyonsaba, F. Current insights into the role of human β-defensins in atopic dermatitis. Clin. Exp. Immunol. 2017, 190, 155–166. [Google Scholar] [CrossRef] [PubMed]
  65. Ständer, S.; Steinhoff, M.; Schmelz, M.; Weisshaar, E.; Metze, D.; Luger, T. Neurophysiology of pruritus: Cutaneous elicitation of itch. Arch. Dermatol. 2003, 139, 1463–1470. [Google Scholar] [CrossRef] [PubMed]
  66. Weisshaar, E.; Apfelbacher, C.; Jäger, G.; Zimmermann, E.; Bruckner, T.; Diepgen, T.; Gollnick, H. Pruritus as a leading symptom: Clinical characteristics and quality of life in German and Ugandan patients. Br. J. Dermatol. 2006, 155, 957–964. [Google Scholar] [CrossRef] [PubMed]
  67. Lyell, A. The itching patient: A review of the causes of pruritus. Scott. Med. J. 1972, 17, 334–347. [Google Scholar] [CrossRef]
  68. Lavery, M.J.; Kinney, M.O.; Mochizuki, H.; Craig, J.; Yosipovitch, G. Pruritus: An overview. What drives people to scratch an itch? Ulst. Med. J. 2016, 85, 164. [Google Scholar]
  69. Schmelz, M. Itch—Mediators and mechanisms. J. Dermatol. Sci. 2002, 28, 91–96. [Google Scholar] [CrossRef]
  70. Shibuya, R.; Takimoto-Ito, R.; Kambe, N.; Kabashima, K. A new era with the development of cytokine-based therapy for pruritus. J. Investig. Dermatol. 2022, 142, 47–52. [Google Scholar] [CrossRef]
  71. Zhang, Q.; Putheti, P.; Zhou, Q.; Liu, Q.; Gao, W. Structures and biological functions of IL-31 and IL-31 receptors. Cytokine Growth Factor Rev. 2008, 19, 347–356. [Google Scholar] [CrossRef] [Green Version]
  72. Takamori, A.; Nambu, A.; Sato, K.; Yamaguchi, S.; Matsuda, K.; Numata, T.; Sugawara, T.; Yoshizaki, T.; Arae, K.; Morita, H. IL-31 is crucial for induction of pruritus, but not inflammation, in contact hypersensitivity. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef]
  73. Hudson, C.A.; Christophi, G.P.; Gruber, R.C.; Wilmore, J.R.; Lawrence, D.A.; Massa, P.T. Induction of IL-33 expression and activity in central nervous system glia. J. Leukoc. Biol. 2008, 84, 631–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Yasuoka, S.; Kawanokuchi, J.; Parajuli, B.; Jin, S.; Doi, Y.; Noda, M.; Sonobe, Y.; Takeuchi, H.; Mizuno, T.; Suzumura, A. Production and functions of IL-33 in the central nervous system. Brain Res. 2011, 1385, 8–17. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, F.; He, D.; Yan, B. Apigenin Attenuates Allergic Responses of Ovalbumin-Induced Allergic Rhinitis Through Modulation of Th1/Th2 Responses in Experimental Mice. Dose-Response 2020, 18. [Google Scholar] [CrossRef]
  76. Che, D.N.; Cho, B.O.; Kim, J.-S.; Shin, J.Y.; Kang, H.J.; Jang, S.I. Effect of Luteolin and Apigenin on the Production of Il-31 and Il-33 in Lipopolysaccharides-Activated Microglia Cells and Their Mechanism of Action. Nutrients 2020, 12, 811. [Google Scholar] [CrossRef] [Green Version]
  77. Che, D.N.; Cho, B.O.; Kim, J.-S.; Shin, J.Y.; Kang, H.J.; Jang, S.I. Luteolin and Apigenin Attenuate LPS-Induced Astrocyte Activation and Cytokine Production by Targeting MAPK, STAT3, and NF-κB Signaling Pathways. Inflammation 2020, 43, 1716. [Google Scholar] [CrossRef]
  78. Armstrong, A.W.; Read, C. Pathophysiology, Clinical Presentation, and Treatment of Psoriasis: A Review. JAMA 2020, 323, 1945–1960. [Google Scholar] [CrossRef] [PubMed]
  79. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int J Mol Sci 2019, 20, 1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Hou, M.; Sun, R.; Hupe, M.; Kim, P.L.; Park, K.; Crumrine, D.; Lin, T.-K.; Santiago, J.L.; Mauro, T.M.; Elias, P.M.; et al. Topical apigenin improves epidermal permeability barrier homoeostasis in normal murine skin by divergent mechanisms. Exp. Dermatol. 2013, 22, 210–215. [Google Scholar] [CrossRef] [Green Version]
  81. Ginwala, R.; Bhavsar, R.; Moore, P.; Bernui, M.; Singh, N.; Bearoff, F.; Nagarkatti, M.; Khan, Z.K.; Jain, P. Apigenin Modulates Dendritic Cell Activities and Curbs Inflammation Via RelB Inhibition in the Context of Neuroinflammatory Diseases. J Neuroimmune Pharm. 2021, 16, 403–424. [Google Scholar] [CrossRef]
  82. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef]
  83. Naylor, M.; Stamp, G.W.; Foulkes, W.D.; Eccles, D.; Balkwill, F.R. Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J. Clin. Investig. 1993, 91, 2194–2206. [Google Scholar] [CrossRef] [Green Version]
  84. D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Rogers, H.W.; Weinstock, M.A.; Harris, A.R.; Hinckley, M.R.; Feldman, S.R.; Fleischer, A.B.; Coldiron, B.M. Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch. Dermatol. 2010, 146, 283–287. [Google Scholar] [CrossRef] [PubMed]
  86. Geller, A.C.; Annas, G.D. Epidemiology of Melanoma and Nonmelanoma Skin Cancer. In Seminars in Oncology Nursing; Elsevier: Amsterdam, The Netherlands, 2003; pp. 2–11. [Google Scholar]
  87. Armstrong, B.K.; Kricker, A. The epidemiology of UV induced skin cancer. J. Photochem. Photobiol. B Biol. 2001, 63, 8–18. [Google Scholar] [CrossRef]
  88. Saladi, R.N.; Persaud, A.N. The causes of skin cancer: A comprehensive review. Drugs Today 2005, 41, 37–54. [Google Scholar] [CrossRef]
  89. Nikolaou, V.; Stratigos, A. Emerging trends in the epidemiology of melanoma. Br. J. Dermatol. 2014, 170, 11–19. [Google Scholar] [CrossRef] [PubMed]
  90. Gordon, R. Skin Cancer: An overview of Epidemiology and Risk Factors. In Seminars in Oncology Nursing: 2013; Elsevier: Amsterdam, The Netherlands, 2003; pp. 160–169. [Google Scholar]
  91. Fresno Vara, J.A.; Casado, E.; de Castro, J.; Cejas, P.; Belda-Iniesta, C.; González-Barón, M. PI3K/Akt signalling pathway and cancer. Cancer Treat. Rev. 2004, 30, 193–204. [Google Scholar] [CrossRef] [PubMed]
  92. Johnstone, R.W.; Frew, A.J.; Smyth, M.J. The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nat. Rev. Cancer 2008, 8, 782–798. [Google Scholar] [CrossRef] [PubMed]
  93. Brooks, A.J.; Putoczki, T. JAK-STAT Signalling Pathway in Cancer. Cancers 2020, 12, 1971. [Google Scholar] [CrossRef]
  94. Guo, Y.J.; Pan, W.W.; Liu, S.B.; Shen, Z.F.; Xu, Y.; Hu, L.L. ERK/MAPK signalling pathway and tumorigenesis (Review). Exp. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bridgeman, B.B.; Wang, P.; Ye, B.; Pelling, J.C.; Volpert, O.V.; Tong, X. Inhibition of mTOR by apigenin in UVB-irradiated keratinocytes: A new implication of skin cancer prevention. Cell. Signal. 2016, 28, 460–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jangdey, M.S.; Gupta, A.; Saraf, S.; Saraf, S. Development and optimization of apigenin-loaded transfersomal system for skin cancer delivery: In vitro evaluation. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1452–1462. [Google Scholar] [CrossRef] [Green Version]
  97. Gilardini Montani, M.S.; Cecere, N.; Granato, M.; Romeo, M.A.; Falcinelli, L.; Ciciarelli, U.; D’orazi, G.; Faggioni, A.; Cirone, M. Mutant p53, stabilized by its interplay with HSP90, activates a positive feed-back loop between NRF2 and p62 that induces chemo-resistance to apigenin in pancreatic cancer cells. Cancers 2019, 11, 703. [Google Scholar] [CrossRef] [PubMed]
  98. Mirzoeva, S.; Tong, X.; Bridgeman, B.B.; Plebanek, M.P.; Volpert, O.V. Apigenin Inhibits UVB-Induced Skin Carcinogenesis: The Role of Thrombospondin-1 as an Anti-Inflammatory Factor. Neoplasia 2018, 20, 930–942. [Google Scholar] [CrossRef]
  99. Berenblum, I. A re-evaluation of the concept of cocarcinogenesis. Carcinog. Carcinog. Test. 1969, 11, 21–30. [Google Scholar]
  100. Boutwell, R.K. The function and mechanism of promoters of carcinogenesis. CRC Crit. Rev. Toxicol. 1974, 2, 419–443. [Google Scholar] [CrossRef]
  101. Wei, H.; Tye, L.; Bresnick, E.; Birt, D.F. Inhibitory effect of apigenin, a plant flavonoid, on epidermal ornithine decarboxylase and skin tumor promotion in mice. Cancer Res 1990, 50, 499–502. [Google Scholar]
  102. Huang, Y.T.; Kuo, M.L.; Liu, J.Y.; Huang, S.Y.; Lin, J.K. Inhibitions of protein kinase C and proto-oncogene expressions in NIH 3T3 cells by apigenin. Eur. J. Cancer 1996, 32a, 146–151. [Google Scholar] [CrossRef]
  103. Zhao, G.; Han, X.; Cheng, W.; Ni, J.; Zhang, Y.; Lin, J.; Song, Z. Apigenin inhibits proliferation and invasion, and induces apoptosis and cell cycle arrest in human melanoma cells. Oncol. Rep. 2017, 37, 2277–2285. [Google Scholar] [CrossRef] [Green Version]
  104. Cao, H.-H.; Chu, J.-H.; Kwan, H.-Y.; Su, T.; Yu, H.; Cheng, C.-Y.; Fu, X.-Q.; Guo, H.; Li, T.; Tse, A.K.-W.; et al. Inhibition of the STAT3 signaling pathway contributes to apigenin-mediated anti-metastatic effect in melanoma. Sci. Rep. 2016, 6, 21731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Woo, J.S.; Choo, G.S.; Yoo, E.S.; Kim, S.H.; Lee, J.H.; Han, S.H.; Kim, H.J.; Jung, S.H.; Park, Y.S.; Kim, B.S.; et al. Apigenin induces apoptosis by regulating Akt and MAPK pathways in human melanoma cell A375SM. Mol Med Rep 2020, 22, 4877–4889. [Google Scholar] [CrossRef] [PubMed]
  106. Whitton, M.E.; Pinart, M.; Batchelor, J.; Leonardi-Bee, J.; González, U.; Jiyad, Z.; Eleftheriadou, V.; Ezzedine, K. Interventions for vitiligo. Cochrane Database Syst. Rev. 2015, 2, Cd003263. [Google Scholar] [CrossRef]
  107. Halder, R.M.; Chappell, J.L. Vitiligo Update. In Seminars in Cutaneous Medicine and Surgery; Elsevier: Amsterdam, The Netherlands, 2009; pp. 86–92. [Google Scholar]
  108. Mahmoud, F.; Abul, H.; Al-Saleh, Q.; Haines, D.; Burleson, J.; Morgan, G. Peripheral T-cell activation in non-segmental vitiligo. J. Dermatol. 1998, 25, 637–640. [Google Scholar] [CrossRef] [PubMed]
  109. Mahmoud, F.; Abul, H.; Haines, D.; Al-Saleh, C.; Khajeji, M.; Whaley, K. Decreased total numbers of peripheral blood lymphocytes with elevated percentages of CD4+ CD45RO+ and CD4+ CD25+ of T-helper cells in non-segmental vitiligo. J. Dermatol. 2002, 29, 68–73. [Google Scholar] [CrossRef] [PubMed]
  110. Taïeb, A. Vitiligo as an inflammatory skin disorder: A therapeutic perspective. Pigment Cell Melanoma Res. 2012, 25, 9–13. [Google Scholar] [CrossRef]
  111. Zhang, B.; Wang, J.; Zhao, G.; Lin, M.; Lang, Y.; Zhang, D.; Feng, D.; Tu, C. Apigenin protects human melanocytes against oxidative damage by activation of the Nrf2 pathway. Cell Stress Chaperones 2020, 25, 277–285. [Google Scholar] [CrossRef]
  112. Lin, M.; Lu, S.-s.; Wang, A.-x.; Qi, X.-y.; Zhao, D.; Wang, Z.-h.; Man, M.-Q.; Tu, C.-x. Apigenin attenuates dopamine-induced apoptosis in melanocytes via oxidative stress-related p38, c-Jun NH2-terminal kinase and Akt signaling. J. Dermatol. Sci. 2011, 63, 10–16. [Google Scholar] [CrossRef]
  113. Malik, S.; Suchal, K.; Khan, S.I.; Bhatia, J.; Kishore, K.; Dinda, A.K.; Arya, D.S. Apigenin ameliorates streptozotocin-induced diabetic nephropathy in rats via MAPK-NF-κB-TNF-α and TGF-β1-MAPK-fibronectin pathways. Am. J. Physiol. -Ren. Physiol. 2017, 313, F414–F422. [Google Scholar] [CrossRef] [Green Version]
  114. Torkin, R.; Lavoie, J.-F.; Kaplan, D.R.; Yeger, H. Induction of caspase-dependent, p53-mediated apoptosis by apigenin in human neuroblastoma. Mol. Cancer Ther. 2005, 4, 1–11. [Google Scholar] [CrossRef]
  115. Zhao, L.; Wang, J.-L.; Liu, R.; Li, X.-X.; Li, J.-F.; Zhang, L. Neuroprotective, anti-amyloidogenic and neurotrophic effects of apigenin in an Alzheimer’s disease mouse model. Molecules 2013, 18, 9949–9965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Popović, M.; Caballero-Bleda, M.; Benavente-García, O.; Castillo, J. The flavonoid apigenin delays forgetting of passive avoidance conditioning in rats. J. Psychopharmacol. 2014, 28, 498–501. [Google Scholar] [CrossRef] [PubMed]
  117. Beg, S.; Swain, S.; Hasan, H.; Barkat, M.A.; Hussain, M.S. Systematic review of herbals as potential anti-inflammatory agents: Recent advances, current clinical status and future perspectives. Pharm. Rev 2011, 5, 120–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Drew, A.K.; Myers, S.P. Safety issues in herbal medicine: Implications for the health professions. Med. J. Aust. 1997, 166, 538–541. [Google Scholar] [CrossRef] [PubMed]
  119. DeRango-Adem, E.F.; Blay, J. Does Oral Apigenin Have Real Potential for a Therapeutic Effect in the Context of Human Gastrointestinal and Other Cancers? Front. Pharmacol. 2021, 12, 1477. [Google Scholar] [CrossRef]
  120. Barnes, J.S.; Foss, F.W., Jr.; Schug, K.A. Thermally accelerated oxidative degradation of quercetin using continuous flow kinetic electrospray-ion trap-time of flight mass spectrometry. J. Am. Soc. Mass Spectrom. 2013, 24, 1513–1522. [Google Scholar] [CrossRef]
Figure 1. The structure of apigenin (4′,5,7-trihydroxyflavone). This figure was made using JChemPaint software.
Figure 1. The structure of apigenin (4′,5,7-trihydroxyflavone). This figure was made using JChemPaint software.
Ijms 24 01498 g001
Figure 2. Scheme of the therapeutic efficacies of apigenin on the skin. This figure was created with BioRender.com.
Figure 2. Scheme of the therapeutic efficacies of apigenin on the skin. This figure was created with BioRender.com.
Ijms 24 01498 g002
Figure 3. The molecular interactions of apigenin due to UV irradiation.
Figure 3. The molecular interactions of apigenin due to UV irradiation.
Ijms 24 01498 g003
Figure 4. The role of apigenin in protein regulation in AD-irritated skin cells.
Figure 4. The role of apigenin in protein regulation in AD-irritated skin cells.
Ijms 24 01498 g004
Figure 5. A schema of the efficacy of apigenin against pruritus.
Figure 5. A schema of the efficacy of apigenin against pruritus.
Ijms 24 01498 g005
Figure 6. The mechanism of apigenin in LPS-treated DCs.
Figure 6. The mechanism of apigenin in LPS-treated DCs.
Ijms 24 01498 g006
Figure 7. Mechanism of anti-carcinogenesis effects according to apigenin treatment in an NMSC model.
Figure 7. Mechanism of anti-carcinogenesis effects according to apigenin treatment in an NMSC model.
Ijms 24 01498 g007
Figure 8. The molecular process of apigenin in melanoma cells.
Figure 8. The molecular process of apigenin in melanoma cells.
Ijms 24 01498 g008
Figure 9. Antioxidant response of apigenin to protect melanocytes against oxidative stress.
Figure 9. Antioxidant response of apigenin to protect melanocytes against oxidative stress.
Ijms 24 01498 g009
Table 1. The cytokines and chemokines that stimulate AD.
Table 1. The cytokines and chemokines that stimulate AD.
CytokinesClassificationRoleReference
IL-4Th2 cell-derived cytokines
  • Interacts with keratinocytes to produce eosinophil chemokine (CCL26) in the acute phase.
  • Regulates IgE secretion from B cells.
  • Directly acts on itch sensory neurons to promote pruritus.
[51,52]
IL-13
IL-31Th2 cell-derived cytokines
  • Causes pruritus by binding the IL-31 receptor A (IL31RA).
  • Downregulates barrier-associated protein expression.
  • Inhibits keratinocyte differentiation.
[53,54]
IL-1α IL-1 family
  • Recruits leukocytes to inflammation sites.
  • Decreases the natural moisturizing factors in stratum corneum layers.
[55,56]
IL-1β
IL-33IL-1 family
  • Regulates the activation of mast cells, ILC2, and basophils.
  • Secretes pruritic cytokines from Th2 cells or keratinocytes.
[57,58,59]
IFN-γTh1 cytokines
  • Recruits CXCR3 agonistic chemokines, which induce the T cell into the inflammation site.
[60]
TNF-α
TSLPIL-7-like cytokines
  • Promotes dendritic cells to differentiate into naïve T cells.
  • Binds to the TSLP receptor which is placed in cutaneous sensory neurons to induce pruritus.
[61,62,63,64]
Table 2. Mechanism of apigenin against inflammatory skin diseases and cancers.
Table 2. Mechanism of apigenin against inflammatory skin diseases and cancers.
The Type of Skin Disease Mechanisms of ApigeninTest ModelDose References
UV-mediated inflammationDownregulates Src and COX-2 levels.In vitro10, 20, 40, 50 μM[36,37]
Regulates the level of apoptotic proteins and anti-apoptotic proteins.In vitro7, 15 μM[38,39]
Inhibits MMP-1 expression by suppressing Ca2+ influx.[40,41]
Suppresses the MAPK and AP-1 signaling pathways.In vitro1, 5, 10, 20 μM
Atopic dermatitisSuppresses phosphorylation of STAT6 in IL-4 stimulated mouse spleen cells.Ex vivo25 μM[49]
Ameliorates damaged skin lesions induced by picrylchloride(piCl).In vivo0.05% feed to mice
Downregulates the protein levels of the NF-κB, MAPK pathways.In vitro10, 20, 30 μM[50]
PruritusSuppresses IL-31 levels by inhibiting the NF-κB and MAPK signaling pathways.In vitro 10, 30 µM[50]
Regulates Th1/Th2 balance by inhibiting the NF-κB pathway, and levels of histamine, IgE, and STAT1 expression. In vivo5, 10, 20 mg/kg of mice[75]
Enhances the Th1 response by decreasing the expression of IFN-γ, and T-box proteins in T cells.
Shows low expression of IL-31, IL-33 in apigenin-treated microglial cells via downregulating ERK and JNK expression.In vitro5, 10, 20, 40, 60, 80, 100 µM[76]
Inactivates MAPK and NF-κB proteins.In vitro30, 60 µM [77]
PsoriasisPromotes the synthesis of skin barrier factors.In vivo60 µL of 0.1% apigenin in 100% ethanol[80]
Downregulates the mRNA expression of inflammatory cytokines in LPS-treated DCs.In vitro8, 20 μM[81]
Skin cancerDownregulates mTOR and AKT signaling pathways.In vitro 25 μM[95,96]
In vivo5 μM in 0.2 mL DMSO/acetone (1:9) vehicle mix of mice
Induces autophagy by inhibiting mTOR expression and the conversion of LC3.In vitro6, 12, 25, 50 μM [97]
Decreases carcinogenesis in TPA-mediated mouse skin and PKC activity,.In vivo5, 25 μM to mice
10, 50, 100 μM
[101,102]
In vitro
Attenuates melanoma metastases to the lung by decreasing STAT3 levels.In vivo150 mg/kg of mice[104]
Promotes the expression of apoptotic proteins in A375SM cells.
Inactivates the Akt and MAPK pathway proteins.
In vitro25, 50, 75, 100 µM[103,105]
In vivo25, 50 mg/kg of mice
VitiligoPromotes antioxidant enzyme activity in dose-dependent ways.
Increases the expression of antioxidant genes at the mRNA and protein levels.
In vitro1, 5, 10, 20 µM[111]
Protects pigment cells from DA-induced apoptosis by decreasing the level of apoptotic agents.In vitro10 µM[112]
Inactivates p38, JNK, and Akt levels in the presence of DA.
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

Yoon, J.H.; Kim, M.-Y.; Cho, J.Y. Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer. Int. J. Mol. Sci. 2023, 24, 1498. https://doi.org/10.3390/ijms24021498

AMA Style

Yoon JH, Kim M-Y, Cho JY. Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer. International Journal of Molecular Sciences. 2023; 24(2):1498. https://doi.org/10.3390/ijms24021498

Chicago/Turabian Style

Yoon, Ji Hye, Mi-Yeon Kim, and Jae Youl Cho. 2023. "Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer" International Journal of Molecular Sciences 24, no. 2: 1498. https://doi.org/10.3390/ijms24021498

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop