Next Article in Journal
Unrevealing the Potential of Sansevieria trifasciata Prain Fraction for the Treatment of Androgenetic Alopecia by Inhibiting Androgen Receptors Based on LC-MS/MS Analysis, and In-Silico Studies
Previous Article in Journal
Solubility Optimization of Loxoprofen as a Nonsteroidal Anti-Inflammatory Drug: Statistical Modeling and Optimization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Modulation of Melanogenesis in B16 Cells Upon Treatment with Plant Extracts and Isolated Plant Compounds

1
Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland
2
Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland
3
Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
4
Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland
5
Chair of Gerontology, Geriatrics and Social Work at the Faculty of Pedagogy, Ignatianum Academy in Cracow, 31-501 Cracow, Poland
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(14), 4360; https://doi.org/10.3390/molecules27144360
Submission received: 2 June 2022 / Revised: 22 June 2022 / Accepted: 5 July 2022 / Published: 7 July 2022

Abstract

:
Plants are a rich source of secondary metabolites that exhibit numerous desired properties. The compounds may influence the biology of melanocytes, pigment cells that produce melanin, by modulating numerous signaling pathways, including cAMP/PKA, MAPKs and PI3K/AKT. Its downstream target is microphthalmia-associated transcription factor, responsible for the expression of the tyrosinase enzyme, which plays a major role in melanogenesis. Therefore, this literature review aims to provide insights related to melanogenesis modulation mechanisms of plant extracts and isolated plant compounds in B16 cells. Database searches were conducted using online-based library search instruments from 2012 to 2022, such as NCBI-PubMed and Google Scholar. Upregulation or downregulation of signaling pathways by phytochemicals can influence skin hypo- and hyperpigmentation by changing the level of melanin production, which may pose a significant cosmetic issue. Therefore, plant extracts or isolated plant compounds may be used in the therapy of pigmentation disorders.

Graphical Abstract

1. Introduction

Melanocytes, the melanin-producing cells, are located in the basal layer of the skin. Melanin accumulates in the melanosomes, which are then transferred to keratinocytes. The epidermal-melanin unit is composed of a single melanocyte and neighboring keratinocytes. Keratinocytes may also modulate melanocyte function and melanin production (melanogenesis): a complex biochemical process that begins with the transformation of the amino acid tyrosine by tyrosinase (TYR). The type and amount of melanin produced by melanocytes is genetically determined, but production is also influenced by many other internal and external factors, including ultraviolet (UV) radiation. The keratinocyte-derived paracrine factors, including melanocyte-stimulating hormone (α-MSH) and stem cell factor (SCF), may regulate melanocyte biology through receptor-mediated signaling pathways [1].
One of the major regulators of melanogenesis is microphthalmia-associated transcription factor (MITF), which is responsible for the activation of TYR expression. It is also the downstream target of many signaling pathways, including cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), mitogen-activated protein kinases (MAPKs), and phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT). These pathways are induced in melanocytes by the activation of melanocortin 1 receptor (MCR1) and receptor tyrosine kinase (c-KIT) as a result of attachment of the corresponding ligand, i.e., α-MSH and SCF [2].
Plants are a rich source of secondary metabolites, which can be classified in various groups including phenolics, alkaloids, saponins, terpenes, lipids, and carbohydrates ac-cording to their chemical structure. It is estimated that more than 200,000 such molecules may exist in the plant kingdom [3]. Compounds of natural origin have several beneficial properties, including modulation of different signaling pathways. The pathways involved in the regulation of melanogenesis may also be subject to such modulation. That process can be both upregulated and downregulated [4]. Thus, owing to their properties plant extracts and isolated plant compounds may play a very important role in counteracting hyperpigmentation and hypopigmentation skin disorders [5].
This literature review examines the role of plant extracts and isolated plant compounds in the process of melanogenesis regulation in B16 cells, a well-established model for the discovery of melanogenic principles. The mechanisms of regulation are discussed in detail from the viewpoint of intracellular signal transduction pathways.

2. Study Design

Published data in the time range 2012–2022 were explored using widely-recognized databases such as NCBI-PubMed, Google Scholar, Scopus, and ScienceDirect. The following keywords were used: plant extract, plant-derived compound, melanogenesis, B16 cells, signaling pathways, ultraviolet radiation. This literature review included studies on plant extracts and pure compounds with available information on their modulatory effects on signaling pathways in B16 cells. Only compounds isolated directly from plants were included. Studies involving non-B16 cells, and synthetic compounds were excluded, as well as studies reporting only the final effects of extracts or compounds on melanogenesis without clarifying their molecular background. Items published in languages other than English or with only an abstract available were also rejected. In order to standardize the scientific names of plants, the “Medicinal Plant Names Services” (https://mpns.science.kew.org/mpns-portal/searchName?) (accessed on 10 May 2022) was used. PuBChem (https://pubchem.ncbi.nlm.nih.gov) (accessed on 10 May 2022) was used to obtain IUPAC names of pure compounds.

3. Melanocyte Biology

While melanocytes are mostly found in the human skin, they can be also present elsewhere in the human body. In human skin, melanocytes are stationed in the basal layer of the epidermis and account for 1% of epidermal cells. The cells originate from neural crest cells, then mature and produce melanin in specialized organelles named melanosomes. The melanosomes are transferred to keratinocytes followed by pigment cell death. These cells express specific proteins including TYR, tyrosinase-related protein 1 and 2 (TYRP1, TYRP2) and MITF, among others. They form characteristic melanin units, in which one melanocyte is neighbored by 30–40 keratinocytes. Cross-talk exists between these two cell types. Keratinocytes influence the growth and activity of melanocytes through the action of growth factors and target adhesion molecules. Examples of compounds that are secreted by keratinocytes after UV radiation exposure and affect melanocytes include α-MSH, SCF, nerve growth factor (NGF), prostaglandin E2 (PGE2), endothelin (ET-1), granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR) and basic fibroblast growth factor (bFGF). Melanocyte biology is also controlled by fibroblasts, which secrete SCF and neuregulin 1 (NRG1), among others. Upon stimulation, melanocytes also secrete a number of signaling molecules such as pro-inflammatory cytokines including IL-1α, IL-2, IL-3, IL-6, IL-10 and tumor necrosis factor α (TNF-α), as well as chemokines including IL-8, CCL2 and transforming growth factor (TGF-β), α-MSH, catecholamines, eicosanoids, serotonin and nitric oxide (NO) [1,6,7].
Melanocytes synthesize melanin through a biochemical pathway called melanogenesis. The process takes place in separate cytoplasmic organelles called melanosomes. Melanin is typically found as yellow-–red pheomelanin and dark brown–black eumelanin, whose production is determined by enzyme action and substrate availability. TYR catalyzes the hydroxylation of tyrosine to L-3,4-dihydroxyphenylalanine (DOPA). DOPA is then oxidized to DOPAquinone. DOPAquinone in the presence of cysteine leads to the formation of 3- or 5-cysteinylDOPAs, which then yield pheomelanin by conversion. In the absence of thiol substrates, DOPAquinone undergoes cyclization to DOPAchrome. DOPAchrome leads to the formation of 5,6-dihydroxyindole, which upon transformation forms eumelanin. However, in the presence of DOPAchrome tautomerase (TYRP2), 5,6-dihydroxyindole-2-carboxylic acid is formed from DOPAchrome. Further transformations involving TYR and TYRP1 finally produce the brown color of melanin (Figure 1).
Human skin contains a mixture of all types of melanin, and visible pigmentation is determined by their ratio. In addition, melanin has numerous properties that are beneficial to the body; while the most important one is the absorbance and scattering of UV radiation, the molecule also plays a part in the neutralization of free radicals [4].
Melanogenesis is regulated by more than 125 genes [8]. Many factors, including external agents (UV radiation), and both internal and paracrine factors produced by keratinocytes and fibroblasts, stimulate specific intracellular signaling pathways involved in melanogenesis, including cAMP/PKA, MAPKs and PI3K/AKT. The most important regulator of melanogenesis is MITF. It affects the activation of key melanogenesis-related genes such as TYR, TYRP1 and TYRP2, and is a common downstream target of many signal transduction pathways that may be modulated by plant extracts and isolated plant compounds (Figure 2) [2].
For terminally differentiated cells, their proliferation is in turn inhibited [9]. Despite their very low proliferative capacity, melanocytes undergo telomerase-dependent senescence. Senescence results in characteristic morphological and functional changes. This is an irreversible process and cells cannot be stimulated to proliferate again by agents such as growth factors. A crucial role in senescence is played by the retinoblastoma protein (pRb). It acts as a repressor of genes involved in DNA replication, resulting in cell cycle arrest. Along with pRb, other cell cycle inhibitors such as p16INK4a and p21Waf1 and their homologs interact to inhibit cyclin-dependent kinases (CDKs) activity and thus prevent phosphorylation of pRB, keeping it in an activated state [10]. For example, Spartium junceum flowers inhibit melanogenesis in B16 melanoma cells by inducing senescence caused by cell cycle arrest in the G2/M phase [11].
Furthermore, it has been shown that melanocytes can also undergo stress-induced senescence. UVB radiation exposure was confirmed to affect melanocyte proliferation. Exposure to low and repeated doses of UVB radiation increased β-galactosidase activity associated with senescence, and changes in the expression of other markers, including p21, p53 and lamin B1. In addition, UVB radiation was shown to contribute to impairment of the proteasome, intensification of autophagy processes in melanocytes, and increased intracellular melanin levels. For example, autophagy may also be induced by hydroxydaidzein from fermented soybean paste [12]. Aging melanocytes accumulating in the skin are also thought to impair the proliferation of neighboring keratinocytes [13,14].

4. Natural Skin Agents against Hyper and Hypo-Pigmentation Disease

Alterations in skin pigmentation might become an esthetic problem. Common hyperpigmentation disorders include melasma, solar lentigines, post-inflammatory hyperpigmentation, and chloasma. UV radiation exposure can exacerbate all of these conditions. Clinically, it manifests as a brown or blue discoloration of the skin, whose location depends on the site of melanin deposition, i.e., in the epidermis and dermis. Hypopigmentation disorders include vitiligo which is related to genetic and environmental factors. Vitiligo is clinically manifested by the presence of white patches on the skin; the loss of melanocyte activity in these areas is probably the result of melanocyte destruction [5]. Plants serve as a reservoir for ingredients that can be used to improve the appearance of the skin. Metabolites of plant origin are classified as primary and secondary based on their role. The former are involved in the basic physiological functions of the cell, while the latter are intended to provide defense against adverse environmental conditions, herbivores, and pathogens [15].
Secondary metabolites can be divided into three main groups depending on the pathway of their synthesis: terpenes formed via the mevalonate pathway, phenolic com-pounds formed via the szikimic or mevalonate pathway, nitrogen-containing secondary metabolites formed mainly from aliphatic amino acids via the tricarboxylic acid pathway, and aromatic acids derived from the szikimic acid pathway [16]. Due to their chemical structure, secondary metabolites are divided into phenolics, alkaloids, saponins, terpenes, lipids or carbohydrates [17].
In the search for new depigmenting and pigmenting agents, studies of plant extracts have led to the identification of many potentially active compounds. Depigmenting agents may act at different levels of melanin production, many are activators of tyrosinase, a key enzyme involved in melanogenesis and pigment production. Others may affect the expression of this enzyme, in addition to the transport of pigment from melanocytes to keratinocytes. On the other hand, due to the lack of pigment cells, attempts have been made to find compounds that influence the differentiation and migration of melanoblasts or would enable melanin dispersion and induce skin pigmentation.
A great number of plant extracts and isolated plant compounds have been found to bear adequate anti-melanogenic or melanogenic potential. They may even have an additional protective potential due to their antioxidant properties, thanks to which they protect the cell’s macromolecules from the damaging effects of free radicals generated by UV radiation, among others. In conclusion, plant-derived compounds may form part of therapeutic interventions against skin abnormalities, including hyperpigmentation and hypopigmentation [18,19,20,21,22].

5. Mechanisms of Melanogenesis-Related Signaling Pathway Modulation by Plant Extracts and Isolated Compounds in B16 Cells

Many genes that encode diverse proteins are implicated in different steps of pigmentation such as melanocyte formation from the neural crest, formation of melanosome components, pigment inclusion, and the transfer of melanosomes from melanocytes to keratinocytes [23]. Plant extracts and isolated plant compounds may be able to modulate pigment formation. Most previous studies are based simply on the assessment of melanin content and tyrosinase enzyme activity in melanocytes exposed to phytochemicals. Others examine changes in gene expression and protein production that are particularly important for melanogenesis. These include MITF, a master regulator of melanogenesis gene expression; this activates the genes encoding the pigmentation enzyme TYR and tyrosinase-related proteins (TYRPs) by binding to their promotors. Data on their expression and its modulation by plant extracts without and with identified compounds (Table 1 and Table 2) and single-derived compounds (Table 3) are presented. Additionally, the roles of phytochemicals in various signaling pathways are also analyzed. Activation of melanocortin 1 receptor (MC1R) or receptor tyrosine kinase (c-KIT) by α-MSH or SCF ligands, respectively, activate signaling pathways in melanoma cells that may be modulated by plant extract or isolated plant compounds. Natural chemicals may have the effect of both stimulating and inhibiting melanogenesis. Global expression analysis by Villareal et al. identified MITF modulation in B16 melanoma cells treated with the Cymbopogon schoenanthus extract, as well as 44 other pigmentation-related genes [24].

5.1. cAMP/PKA Signaling Pathway

The neuromodulating peptide α-MSH is released by keratinocytes after stimulation with pro-inflammatory cytokines or UV light. The junction of ligand to MCR1 located on the melanocyte cell surface activates adenylyl cyclase, responsible for the synthesis of cAMP. Increased cAMP levels cause stimulation of PKA. Both the cAMP level and melanin production are increased following treatment of B16 melanoma cells with 1,5-dicaffeoylquinic acid isolated from Vernonia anthelmintica seeds [93]. Phyllostachys nigra stem extracts were found to have an anti-melanogenic effect in B16 melanoma cells by decreasing intracellular cAMP and PKA levels [94], as did Dendropanax morbiferus leaves [95] and Lotus seedpod extract [96]. Among single-derived compounds, bisabolangelone, a sesquiterpene derivative, isolated from Angelica koreana roots [97] and arctigenin, a lignan, from Arctium lappa seeds inhibit cAMP-dependent PKA activation [98].
PKA entails phosphorylation at Ser133 of cAMP-response element binding protein (CREB) and subsequent activation of MITF expression. The extracts that up-regulate pigment formation are Melia azedarach bark [99] and Dalbergia odorifera fruit [100]. In B16 melanoma cells, they increase the intracellular cAMP level and PKA activity, translating to increased phosphorylation of CREB, its downstream signaling protein, followed by up-regulation of MITF and TYR expression. However, no effects on MAPKs were observed. Moreover, CREB was activated by cirsimaritin, a dimethoxyflavone, isolated from the branches of Lithocarpus dealbatus [101], hesperetin, a flavanone glycoside, from Citrus sinensis, Citrus aurantium and Citrus reticulata extract [102] and scopoletin, a hydroxycoumarin, from the aerial parts of Cirsium setidens [103]. The opposite effect on phosphorylation of PKA and CREB are interfered with by Nelumbo nucifera leaf extract [104]. Rhodiola rosea root extract [105], Elaeagnus umbellate branches and leaves [106], Oenothera laciniata extracts [107] and Kadsura coccinea roots, stems, leaves, fruits [108] extracts inhibited the phosphorylation of CREB followed by downregulation of MITF and TYR expression. Similar inhibitory effects on CREB exerts isoorientin, a flavone glycoside, derived from Gentiana veitchiorum flowers [109], polysaccharide from Morchella esculenta fruits [110], loganin, an iridoid monoterpenoid, from Cornus officinalis [111], kaempferol-7-O-β-D-glucuronide, flavonol glucoside, and tilianin, a flavonoid glycoside, isolated from aerial parts Cryptotaenia japonica [112], moracin J, a 2-arylbenzofuran flavonoid, from leaves of Morus alba [113], and 6-O-isobutyrylbritannilactone from the flowers of Inula britannica [114]. This pathway is also suppressed by Phoenix dactylifera seed extract that decreased the expression of MC1R [115].

5.2. MAPKs Signaling Pathway

Both α-MSH-MC1R and SCF-c-KIT junction may modulate the MAPKs pathway. MAPKs regulate cell proliferation, differentiation, motility and survival by converting extracellular signals into intracellular cellular responses. The best-studied Ser/Thr kinases in this family are extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun amino-terminal kinases 1–3 (JNK 1 to -3), and p38 (α, β, γ, and δ). Each of these groups consists of a set of three sequentially acting kinases: MAPK, MAPK kinase (MAPKK) and MAPKK kinase (MAPKKK). Activation of MAPKKK leads to phosphorylation and activation of MAPKK, which in turn activates MAPK through dual phosphorylation of Thr residues within the Thr-X-Thr motif located in the activation loop of the kinase domain [116].
It is assumed that the increased level of cAMP triggered by MC1R activation is followed by induction of ERK in a cell-type seemed to be mediated by B-RAF, such as melanoma cells [117]. cAMP levels seemed to have the opposite effect on p38 activity. The effect of cAMP on the JNK signaling cascades has been less explored; however, in the majority of cells, increased cAMP levels appeared to modulate JNK activity [118].
SCF is a growth factor secreted by human keratinocytes and fibroblasts under UV radiation [119,120]. It is believed to be involved in the dimerization and autophosphorylation of the c-KIT receptor by binding to the extracellular domain, thus activating it. Phosphorylation at Y703 and Y936 induce MAPKs. Following activation, the c-KIT phosphorylates the small GTPase (Ras), which in turn induces proto-oncogene serine/threonine-protein kinase (Raf-1) followed by the MAPKs signaling pathway [121].
Activated p38 induces melanogenesis via CREB phosphorylation followed by MITF activation and TYR expression. Activation of ERK phosphorylates MITF at Ser73, leading to its degradation via ubiquitination and thus inhibiting melanogenesis. JNK activation may modulate melanogenesis through phosphorylation of CREB-regulated transcription co-activator 3 (CRTC3)-dependent MITF expression [122,123].
P38 positively regulates melanin synthesis in the B16 melanoma cell line [2]. In this line, Polygonum multiflorum root extract was found to up-regulate p38 phosphorylation, thus activating melanogenesis [124]; the same effects were observed for Vernonia anthelmintica seed extract [125] and Annona squamosa leaf extract [126]. In contrast, treatment with Cuscuta japonica seed extract [127], Morinda citrifolia fruit and leaf extract [128], Dendropanax morbiferus leaf extract [95] or Lotus seedpod extract [96] were found to downregulate p38 phosphorylation and thus inhibit melanogenesis. Similar effects are demonstrated by the polysaccharide from Morchella esculenta fruits [110], methyl 3,5-di-caffeoylquinate from the stems and leaves of Erigeron annuus [129] and moracin J, a 2-arylbenzofuran flavonoid, from Morus alba leaves [113].
ERK negatively regulates melanin synthesis in the B16 melanoma cell line. ERK may be suppressed by Ardisia crenata leaf and small branch extract [130] and by lupenone, a triterpenoid, isolated from Erica multiflora leaf [131]. In contrast, ERK may be upregulated, and melanogenesis suppressed, by Astragalus membranaceus whole plant extract [132], Aster yomena callus pellet extract [133], Melochia corchorifolia whole plant extract [134], Artemisia capillaris whole plant extract [135], Orostachys japonicus whole plant extract [136], and Aster spathulifolius leaf extract treatment [137]. Similar effects are also triggered by 2-[4-(3-hydroxypropyl)-2-methoxyphenoxy]-1,3-propanediol isolated from Juglans mandshurica fruits [138], 1-O-acetylbritannilactone from Inula britannica flowers [139], zerumbone, a sesquiterpenoid, from Zingiber officinale [140], octaphlorethol A, a phenolic compound, from Ishige foliacea [141], 2-[4-(3-hydroxypropyl)-2-methoxyphenoxy]-1,3-propanediol from Juglans mandshurica fruits [138], loganin, an iridoid monoterpenoid, from Cornus officinalis [111], neobavaisoflavone, a 7-hydroxyisoflavone, from aerial parts of Pueraria lobata [142], 6-O-isobutyrylbritannilactone from Inula britannica flowers [128] and quercitrin, a quercetin O-glycoside, from Lindera obtusiloba leaves [143].
Arctium lappa leaf extract was found to inhibit melanogenesis in B16 melanoma cells by regulating JNK phosphorylation [144].
Many studies have examined the influence of plant extracts and isolated compounds on various MAPKs in B16 melanoma cells. Extracts derived from Artocarpus communis heartwood [145], Phyla nodiflora aerial part [146] and Oenothera laciniata [107] were found to modulate ERK and JNK phosphorylation, resulting in anti-melanogenic effects. Similar results were obtained for kaempferol-7-O-β-D-glucuronide and tilianin, a flavonoid glycoside isolated from the aerial parts of Cryptotaenia japonica [126]. Penthorum chinense whole plant extract [147], Kaempferia galanga whole plant extract [148] and Phragmites communis leaf extract [149] were found to modulate both p38 and JNK phosphorylation, followed by reduced melanin production, in B16 melanoma cells. This phosphorylation was also modulated by norartocarpetin, a flavone, obtained from Artocarpus communis heartwood [150]. Argania spinosa fruit extract [151] upregulates melanogenesis through the modulation of p38 and ERK signaling. Among single-derived compounds this effect was obtained for cynarine, a hydroxycinnamic acid derivative, extracted from Vernonia anthelmintica [93] and hesperetin, a flavanone glycoside, from Citrus sinensis, Citrus aurantium and Citrus reticulata extract [102]. Eupafolin, a flavone, isolated from the aerial part of Phyla nodiflora was found to suppress melanogenesis via p38 and ERK modulation in B16 cells [152]. Paederia foetida whole plant extract was found to modulate the activity of all MAPKs, resulting in inhibited melanin production [153]; this was also observed for Rosa gallica petal extract [154] as well as dihydromyricetin, a flavanonol, isolated from the leaves and stems of Ampelopsis grossedentata [155].

5.3. PI3K/AKT Signaling Pathway

MC1R activated by α-MSH, as well as c-KIT activated by SCF, can also modulate the PI3K/AKT pathway: a very important process regulating cell proliferation and survival. It has been shown that MC1R activation followed by induction of cAMP leads to the inhibition of PI3K activity and of AKT phosphorylation and activity. Inactive AKT is unable to phosphorylate glycogen synthase kinase 3 beta (GSK3β) [156,157,158]; this phosphorylation is needed for β-catenin accumulation, followed by translocation to the nucleus. This in turn stimulates MITF activity and TYR gene expression. SCF is believed to be involved in the dimerization and autophosphorylation of the c-KIT receptor by binding to the extracellular domain, thus activating it, phosphorylation at Y721 and PI3K pathway induction. Activated AKT phosphorylate GSK3β becomes inactivated and targeted for proteasomal degradation [121,159].
In B16 melanoma cells, Musa sapientum peel extract [160], Aster yomena callus pellet extract [133] and Orostachys japonicus whole plant [136] treatment reduced phosphorylation of AKT, and these might act as anti-melanogenesis agents. Upregulation of AKT by Phragmites communis leaf extract [149], Aster spathulifolius leaf extract treatment [137] and Ginkgo biloba leaf extract [161] may also inhibit melanin synthesis. Among plant-derived compounds, 1-O-acetylbritannilactone obtained from Inula britannica flowers [139], eupafolin, a flavone, isolated from aerial part of Phyla nodiflora [152] and 6-O-isobutyrylbritannilactone from the flowers of Inula britannica have been found to increase AKT signaling in B16 cells [114]. GSK3β activation is stimulated by hesperetin, a flavanone glycoside, from Citrus sinensis, Citrus aurantium and Citrus reticulata extract [102], an agent with melanogenic potential; and is also inactivated by isoorientin, a flavone glycoside, derived from Gentiana veitchiorum flowers, and neobavaisoflavone, a 7-hydroxyisoflavone, from aerial parts of Pueraria lobata [142], agents with whitening potential [109].

5.4. In Vivo Studies

In some studies, given above, in vivo analyses were also conducted in addition to the in vitro study and evaluation of signaling pathway modulation by plant extracts and compounds isolated from plants. The melanocyte activity and the distribution of melanin granules were decreased in UVB-irradiated C57BL/6 mice treated with Aster spathulifolius leaf extract. Mice were exposed to UVB radiation at a dose of 100 mJ/cm2 for 10 days. Extract was orally administered for 9 weeks at 35, 70 and 140 mg/kg concentrations [137]. Nelumbo nucifera leaf extract reduced skin melanogenesis induced by UVB radiation in guinea pigs. The animals were exposed to UVB radiation three times a week for two weeks. The total UVB dose was 500 mJ/cm2 per exposure. Then, 1 or 2% of extract was given topically to the UVB-irradiated regions the next day [96]. Treatment of subjects with skin pigmentation with Aster yomena callus pellet extract-containing cream formulations resulted in 3.33%, 7.06%, and 8.68% improvements in melanin levels at 2, 4, and 8 weeks, respectively. These results suggest that Aster spathulifolius, Nelumbo nucifera and Aster yomena extracts might be vulnerable and promising therapeutics as agents for hyperpigmentation [133].
Studies based on single compounds indicate that arctigenin from Fructus arctii demonstrated antimelanogenic activity using zebrafish embryo. Arctigenin was added directly in fish water at final concentrations of 1, 10, and 100 𝜇M [98]. The same result was obtained for the zebrafish embryo treated with 10, 50, or 100 µM 6-O-isobutyrylbritannilactone from Inula britannica [114]. In addition, a similar result was also observed for the zebrafish embryo treated with 75, 150 and 300 μg/mL heteropolysaccharide from Morchella esculenta [110]. The embryos had significantly reduced pigmentation in the arctigenin, 6-O-isobutyrylbritannilactone and heteropolysaccharide-treated specimens. In conclusion, the results suggest that these compounds have potential as a skin-whitening agent for hyperpigmentation.

6. Mechanisms of Melanogenesis-Related Signaling Pathway Modulation by Plant Extracts and Single-Derived Compounds in B16 Cells Stimulated by UV Radiation

Exposure to UV radiation has deleterious effects on human skin followed by acute and chronic skin damage. It also generates the production of ROS and proinflammatory cytokines. Both experimental and epidemiological data indicate that the melanin present in skin plays an important role in photoprotection.
UV radiation interferes directly with macromolecules, including proteins, lipids and nucleic acids. Interactions with nucleic acids in skin cells may contribute to DNA damage. If these mutations occur in genes responsible for the regulation of repair processes, cell cycle or apoptosis, they may subsequently initiate oncogenic transformation.
UVA can generate ROS, which are capable of inducing oxidative DNA damage: singlet oxygen activity and type 1 photosensitization reactions result in oxidative modifications of nitrogenous bases, mainly guanine. If not repaired, 7,8-dihydro-8-oxoguanine lesions are formed leading to mutations. The major mutations are the G -> A transition and the G -> T transversion. Cyclobutanopyrimidine dimers (CPDs) may also form.
UVB leads to the formation of photoproducts such as CPDs and pyrimidine 6-4 pyrimidones in DNA due to the activation of a photochemical reaction, usually occurring between adjacent pyrimidine nucleotides. If left unrepaired, they can lead to mutations, including C -> T and CC -> TT transition mutations, and oncogenesis [162].
UV radiation exposure induces DNA damage in keratinocytes and activates the p53 tumor suppressor protein, which can bind to and activate the pro-opiomelanocortin (POMC) promoter. It can also induce production of subunit melanogenic peptides, including α-MSH which binds to the MCR1 on melanocytes, thus stimulating the expression of genes involved in melanin production [163].
Certain plant extracts and isolated plant compounds are able to inhibit UV-induced melanogenesis, as observed in stimulated B16 melanoma cells. Psidium guajava leaf [164] and Foeniculum vulgare fruit [165] extracts block the activity of TYR and calcium release-activated calcium channel protein 1 (ORAI1). The ORAI1 channel is also inhibited by valencene, a sesquiterpene, isolated from the rhizomes of Cyperus rotundus [166].
UV radiation directly induces intracellular calcium signaling in melanocytes, mediated in part by the ORAI1 channel. In addition, UV radiation stimulates melanocytes through compounds such as endothelin 1 (ET-1) released by keratinocytes. Activation of the ET-1 receptor also triggers intracellular calcium signaling, mediated by ORAI-1. Increased calcium levels activate TYR, resulting in melanin production. Therefore, ORAC channel antagonists play a key role in inhibiting UV-induced melanogenesis [167,168].

7. Conclusions

Plant extracts or isolated plant compounds, including in particular phenolics and terpenes may act as activators or inhibitors of key signaling pathways, such as cAMP/PKA, MAPKs and PI3K/AKT in melanocytes. Such modulation influences the expression of proteins, including master regulator of melanogenesis-MITF. Therefore, natural chemicals may serve as useful ingredients for reducing skin pigmentation or activating pigment formation. However, despite the very high potential of plant-derived molecules, proposals for the future work include improved exploration of the signaling pathways that may be modulated by phytochemicals in the melanogenesis process as well as better evaluation of their effects on living organisms.

Author Contributions

Conceptualization, A.M.-S. and P.S.; Formal analysis, E.K., K.Z. and R.Z.; Data curation, E.K., K.Z. and R.Z.; Writing—original draft preparation, A.M-S.; Writing—review and editing, A.M.-S., P.S. and T.K.; Supervision, R.Z. 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.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are available from the authors.

References

  1. Cichorek, M.; Wachulska, M.; Stasiewicz, A.; Tymińska, A. Skin melanocytes: Biology and development. Postep. Dermatol. Alergol. 2013, 30, 30–41. [Google Scholar] [CrossRef] [PubMed]
  2. D’Mello, S.A.N.; Finlay, G.J.; Baguley, B.C.; Askarian-Amiri, M.E. Signaling pathways in melanogenesis. Int. J. Mol. Sci. 2016, 17, 1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wink, M. Plant secondary metabolism: Diversity, function and its evolution. Nat. Prod. Commun. 2008, 3, 1205–1216. [Google Scholar] [CrossRef] [Green Version]
  4. Videira, I.F.D.S.; Moura, D.F.L.; Magina, S. Mechanisms regulating melanogenesis. An. Bras. Dermatol. 2013, 88, 76–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nicolaidou, E.; Katsambas, A.D. Pigmentation disorders: Hyperpigmentation and hypopigmentation. Clin. Dermatol. 2014, 32, 66–72. [Google Scholar] [CrossRef] [PubMed]
  6. Tang, H.; Yang, L.; Wu, L.; Wang, H.; Chen, K.; Wu, H.; Li, Y. Kaempferol, the melanogenic component of Sanguisorba officinalis, enhances dendricity and melanosome maturation/transport in melanocytes. J. Pharmacol. Sci. 2021, 147, 348–357. [Google Scholar] [CrossRef]
  7. Brenner, M.; Hearing, V.J. The protective role of melanin against UV damage in human skin. Photochem. Photobiol. 2008, 84, 539–549. [Google Scholar] [CrossRef] [Green Version]
  8. Bennett, D.C.; Lamoreux, M.L. The color loci of mice-A genetic century. Pigment Cell Res. 2003, 16, 333–344. [Google Scholar] [CrossRef] [Green Version]
  9. Bennett, D.C.; Medrano, E.E. Molecular regulation of melanocyte senescence. Pigment Cell Res. 2002, 15, 242–250. [Google Scholar] [CrossRef]
  10. Becker, T.; Haferkamp, S. Molecular Mechanisms of Cellular Senescence. In Senescence and Senescence-Related Disorders; Wang, Z., Inuzuka, H., Eds.; IntechOpen Limited: London, UK, 2013; pp. 25–50. [Google Scholar]
  11. Nanni, V.; Canuti, L.; Gismondi, A.; Canini, A. Hydroalcoholic extract of Spartium junceum L. flowers inhibits growth and melanogenesis in B16-F10 cells by inducing senescence. Phytomedicine 2018, 46, 1–10. [Google Scholar] [CrossRef]
  12. Kim, E.S.; Shin, J.H.; Seok, S.H.; Kim, J.B.; Chang, H.; Park, S.J.; Jo, Y.K.; Choi, E.S.; Park, J.S.; Yeom, M.H.; et al. Autophagy mediates anti-melanogenic activity of 3′-ODI in B16F1 melanoma cells. Biochem. Biophys. Res. Commun. 2013, 442, 165–170. [Google Scholar] [CrossRef] [PubMed]
  13. Low, E.; Alimohammadiha, G.; Smith, L.A.; Costello, L.F.; Przyborski, S.A.; von Zglinicki, T.; Miwa, S. How good is the evidence that cellular senescence causes skin ageing? Ageing Res. Rev. 2021, 71, 101456. [Google Scholar] [CrossRef] [PubMed]
  14. Martic, I.; Wedel, S.; Jansen-Dürr, P.; Cavinato, M. A new model to investigate UVB-induced cellular senescence and pigmentation in melanocytes. Mech. Ageing Dev. 2020, 190, 111322. [Google Scholar] [CrossRef] [PubMed]
  15. Erb, M.; Kliebenstein, D.J. Plant Secondary Metabolites as Defenses, Regulators, and Primary Metabolites: The Blurred Functional Trichotomy. Plant Physiol. 2020, 184, 39–52. [Google Scholar] [CrossRef] [PubMed]
  16. Parsaeimehr, A.; Sargsyan, E.; Vardanyan, A. Expression of secondary metabolites in plants and their useful perspective in animal health. Int. J. Bioflux Soc. 2011, 3, 129–134. [Google Scholar]
  17. Hussein, A.R.; El-Anssary, A. Plants Secondary Metabolites: The Key Drivers of the Pharmacological Actions of Medicinal Plants. In Herbal Medicine; Builders, P.F., Ed.; IntechOpen Limited: London, UK, 2019; pp. 11–30. [Google Scholar]
  18. Solano, F.; Briganti, S.; Picardo, M.; Ghanem, G. Hypopigmenting agents: An updated review on biological, chemical and clinical aspects. Pigment Cell Res. 2006, 19, 550–571. [Google Scholar] [CrossRef]
  19. Szczurko, O.; Boon, H.S. A systematic review of natural health product treatment for vitiligo. BMC Dermatol. 2008, 8, 2. [Google Scholar] [CrossRef] [Green Version]
  20. Fisk, W.A.; Agbai, O.; Lev-Tov, H.A.; Sivamani, R.K. The use of botanically derived agents for hyperpigmentation: A systematic review. J. Am. Acad. Dermatol. 2014, 70, 352–365. [Google Scholar] [CrossRef]
  21. Smit, N.; Vicanova, J.; Pavel, S. The hunt for natural skin whitening agents. Int. J. Mol. Sci. 2009, 10, 5326–5349. [Google Scholar] [CrossRef]
  22. Zhu, W.; Gao, J. The use of botanical extracts as topical skin-lightening agents for the improvement of skin pigmentation disorders. J. Investig. Dermatol. Symp. Proc. 2008, 13, 20–24. [Google Scholar] [CrossRef] [Green Version]
  23. Praetorius, C.; Grill, C.; Stacey, S.N.; Metcalf, A.M.; Gorkin, D.U.; Robinson, K.C.; Van Otterloo, E.; Kim, R.S.Q.; Bergsteinsdottir, K.; Ogmundsdottir, M.H.; et al. XA polymorphism in IRF4 affects human pigmentation through a tyrosinase-dependent MITF/TFAP2A pathway. Cell 2013, 155, 1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Villareal, M.O.; Kume, S.; Neffati, M.; Isoda, H. Upregulation of Mitf by Phenolic Compounds-Rich Cymbopogon schoenanthus Treatment Promotes Melanogenesis in B16 Melanoma Cells and Human Epidermal Melanocytes. Biomed Res. Int. 2017, 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jeong, D.; Lee, J.; Jeong, S.G.; Hong, Y.H.; Yoo, S.; Han, S.Y.; Kim, J.H.; Kim, S.; Kim, J.S.; Chung, Y.S.; et al. Artemisia asiatica ethanol extract exhibits anti-photoaging activity. J. Ethnopharmacol. 2018, 220, 57–66. [Google Scholar] [CrossRef] [PubMed]
  26. Dissanayake, C.Y.; Moon, H.H.; Yang, K.M.; Lee, Y.; Han, C.H. The effects of green tea (Camellia sinensis) flower extract on melanin synthesis in B16-F10 melanoma cells. Korean J. Vet. Res. 2018, 58, 66–72. [Google Scholar] [CrossRef]
  27. Kim, J.H.; Choi, G.N.; Kwak, J.H.; Jeong, C.H.; Jeong, H.R.; Lee, U.; Kim, M.J.; Heo, H.J. Inhibitory effects of chestnut inner skin extracts on melanogenesis. Food Sci. Biotechnol. 2012, 21, 1571–1576. [Google Scholar] [CrossRef]
  28. Lee, S.C.; Chen, C.H.; Yu, C.W.; Chen, H.L.; Huang, W.T.; Chang, Y.S.; Hung, S.H.; Lee, T.L. Inhibitory effect of Cinnamomum osmophloeum Kanehira ethanol extracts on melanin synthesis via repression of tyrosinase expression. J. Biosci. Bioeng. 2016, 122, 263–269. [Google Scholar] [CrossRef]
  29. Huang, H.C.; Hsieh, W.Y.; Niu, Y.L.; Chang, T.M. Inhibitory effects of adlay extract on melanin production and cellular oxygen stress in B16F10 melanoma cells. Int. J. Mol. Sci. 2014, 15, 16665–16679. [Google Scholar] [CrossRef] [Green Version]
  30. Chatatikun, M.; Yamauchi, T.; Yamasaki, K.; Aiba, S.; Chiabchalard, A. Anti melanogenic effect of Croton roxburghii and Croton sublyratus leaves in α-MSH stimulated B16F10 cells. J. Tradit. Complement. Med. 2019, 9, 66–72. [Google Scholar] [CrossRef]
  31. Ko, Y.J.; Yang, S.K.; Song, S.M.; Yoon, W.J.; Bae, K.H. Effect of Dendrobium moniliforme on Melanogenic Protein Expression in B16F10 Melanoma Cells. J. Biol. Act. Prod. Nat. 2015, 5, 12–17. [Google Scholar] [CrossRef]
  32. Park, S.A.; Lee, H.M.; Ha, J.H.; Jeon, S.H.; Park, S.N. Inhibitoiy effects of Dendropanax morbifera leaf extracts on melanogenesis through down-regulation of tyrosinase and TRP-2. Appl. Chem. Eng. 2014, 25, 468–473. [Google Scholar] [CrossRef]
  33. Li, P.H.; Chiu, Y.P.; Shih, C.C.; Wen, Z.H.; Ibeto, L.K.; Huang, S.H.; Chiu, C.C.; Ma, D.L.; Leung, C.H.; Chang, Y.N.; et al. Biofunctional Activities of Equisetum ramosissimum Extract: Protective Effects against Oxidation, Melanoma, and Melanogenesis. Oxid. Med. Cell. Longev. 2016, 2016, 1–9. [Google Scholar] [CrossRef] [Green Version]
  34. Baek, S.H.; Nam, I.J.; Kwak, H.S.; Kim, K.C.; Lee, S.H. Cellular anti-melanogenic effects of a Euryale ferox seed extract ethyl acetate fraction via the lysosomal degradation machinery. Int. J. Mol. Sci. 2015, 16, 9217–9235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kim, M.; Shin, S.; Lee, J.A.; Park, D.; Lee, J.; Jung, E. Inhibition of melanogenesis by Gaillardia aristata flower extract. BMC Complement. Altern. Med. 2015, 15, 449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Hamid, M.A.; Sarmidi, M.R.; Park, C.S. Mangosteen leaf extract increases melanogenesis in B16F1 melanoma cells by stimulating tyrosinase activity in vitro and by up-regulating tyrosinase gene expression. Int. J. Mol. Med. 2012, 29, 209–217. [Google Scholar] [CrossRef] [Green Version]
  37. Shim, E.; Song, E.; Choi, K.S.; Choi, H.J.; Hwang, J. Inhibitory effect of Gastrodia elata Blume extract on alpha-melanocyte stimulating hormone-induced melanogenesis in murine B16F10 melanoma. Nutr. Res. Pract. 2017, 11, 173–179. [Google Scholar] [CrossRef] [Green Version]
  38. Qiao, Z.; Koizumi, Y.; Zhang, M.; Natsui, M.; Flores, M.J.; Gao, L.; Yusa, K.; Koyota, S.; Sugiyama, T. Anti-melanogenesis effect of Glechoma hederacea L. extract on B16 murine melanoma cells. Biosci. Biotechnol. Biochem. 2012, 76, 1877–1883. [Google Scholar] [CrossRef] [Green Version]
  39. Bodurlar, Y. Inhibitory Activity of Soy Cell Culture Extract on Tyrosinase Activity and Melanin Formation in ⍺-MSH Induced B16-F10 Melanoma Cells 2022, 2022, 1–14. Res. Sq. 2022, 1–14. [Google Scholar] [CrossRef]
  40. Chao, H.C.; Najjaa, H.; Villareal, M.O.; Ksouri, R.; Han, J.; Neffati, M.; Isoda, H. Arthrophytum scoparium inhibits melanogenesis through the down-regulation of tyrosinase and melanogenic gene expressions in B16 melanoma cells. Exp. Dermatol. 2013, 22, 131–136. [Google Scholar] [CrossRef]
  41. Lee, J.Y.; Cho, Y.R.; Park, J.H.; Ahn, E.K.; Jeong, W.; Shin, H.S.; Kim, M.S.; Yang, S.H.; Oh, J.S. Anti-melanogenic and anti-oxidant activities of ethanol extract of Kummerowia striata: Kummerowia striata regulate anti-melanogenic activity through down-regulation of TRP-1, TRP-2 and MITF expression. Toxicol. Rep. 2019, 6, 10–17. [Google Scholar] [CrossRef]
  42. Park, J.-H.; Lee, J.Y.; Cho, Y.-R.; Ahn, E.-K.; Jeong, W.; Shin, H.S.; Kim, M.-S.; Oh, J.S. Anti-Melanogenic and Anti-Oxidant Activities of an Ethanolic Extract of Kummerowia Striata and Its Active Compounds, P-Coumaric Acid and Quercetin. Preprints 2018, 2018060341. [Google Scholar] [CrossRef]
  43. Yao, C.; Jin, C.L.; Oh, I.G.; Park, C.H.; Chung, J.H. Melia azedarach extract stimulates melanogenesis through increase of tyrosinase-related protein 1 expression in B16F10 mouse melanoma cells. Int. J. Mol. Med. 2015, 35, 1761–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Tayarani-Najaran, Z.; Akaberi, M.; Vatani, M.; Emami, S.A. Evaluation of antioxidant and anti-melanogenic activities of different extracts from aerial parts of nepeta binaludensis jamzad in murine melanoma B16F10 cells. Iran. J. Basic Med. Sci. 2016, 19, 662–669. [Google Scholar]
  45. Akaberi, M.; Emami, S.A.; Vatani, M.; Tayarani-Najaran, Z. Evaluation of antioxidant and anti-melanogenic activity of different extracts of aerial parts of N. Sintenisii in murine melanoma B16F10 cells. Iran. J. Pharm. Res. 2018, 17, 225–235. [Google Scholar] [PubMed]
  46. Park, H.J.; Lee, E.H.; Jung, H.Y.; Kang, I.K.; Cho, Y.J. Effects of phenolics from oplismenus undulatifolius in α-MSH-stimulated B16F10 melanoma cells. J. Appl. Biol. Chem. 2020, 63, 89–93. [Google Scholar] [CrossRef] [Green Version]
  47. Han, J.H.; Byeon, S.H.; Hyun, C.G.; Lee, N.H. Melanogenesis inhibitory activity in the extracts of Oreocnide fruticosa (Gaudich.) Hand.-Mazz. branches. J. Appl. Pharm. Sci. 2014, 4, 166–169. [Google Scholar] [CrossRef]
  48. Wang, Y.C.; Haung, X.Y.; Chiu, C.C.; Lin, M.Y.; Lin, W.H.; Chang, W.T.; Tseng, C.C.; Wang, H.M.D. Inhibitions of melanogenesis via Phyllanthus emblica fruit extract powder in B16F10 cells. Food Biosci. 2019, 28, 177–182. [Google Scholar] [CrossRef]
  49. Lee, S.-H.; Jang, T.-W.; Choi, J.-S.; Mun, J.-Y.; Park, J.-H. Inhibitory Effects of Pine Cone (Pinus densiflora) on Melanogenesis in B16F10 Melanoma Cells. Korean J. Plant Resour. 2019, 32, 275–281. [Google Scholar]
  50. Ghali, S.K.; Chakraborty, A.; Pallapolu, P.; Rafeeqi, T.A.; Husain, G.M.; Javed, G.; Waheed, M.A.; Kazmi, M.H. Expression of MITF Gene in Melanogenesis with Psoralea corylifolia, Zingiber officinale, Terminalia chebula, Punica granatum and Eclipta alba Based on Poly-Herbal Formulation. J. Biol. Act. Prod. Nat. 2021, 11, 82–96. [Google Scholar] [CrossRef]
  51. Junlatat, J.; Fangkrathok, N.; Sripanidkulchai, B. Antioxidative and melanin production inhibitory effects of syzygium cumini extracts. Songklanakarin J. Sci. Technol. 2018, 40, 1136–1143. [Google Scholar] [CrossRef]
  52. Dong, Y.; Woo, Y.M.; Cha, J.H.; Cha, J.Y.; Lee, N.W.; Back, M.W.; Park, J.; Lee, S.; Ha, J.; Kim, A. The Effect of Inhibition of Uncaria rhynchophylla as an Inhibitor of Melanogenesis and an Antioxidant in B16F10 Melanoma Cells. J. Life Sci. 2020, 30, 1033–1041. [Google Scholar] [CrossRef]
  53. Zhou, S.; Riadh, D.; Sakamoto, K. Grape extract promoted α-msh-induced melanogenesis in b16f10 melanoma cells, which was inverse to resveratrol. Molecules 2021, 26, 5959. [Google Scholar] [CrossRef] [PubMed]
  54. Ma, H.; Xu, J.; DaSilva, N.A.; Wang, L.; Wei, Z.; Guo, L.; Johnson, S.L.; Lu, W.; Xu, J.; Gu, Q.; et al. Cosmetic applications of glucitol-core containing gallotannins from a proprietary phenolic-enriched red maple (Acer rubrum) leaves extract: Inhibition of melanogenesis via down-regulation of tyrosinase and melanogenic gene expression in B16F10 melanoma ce. Arch. Dermatol. Res. 2017, 309, 265–274. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, S.Y.; Won, K.J.; Kim, D.Y.; Kim, M.J.; Won, Y.R.; Kim, N.Y.; Lee, H.M. Wound healing-promoting and melanogenesis-inhibiting activities of Angelica polymorpha maxim. Flower absolute in vitro and its chemical composition. Molecules 2021, 26, 6172. [Google Scholar] [CrossRef] [PubMed]
  56. Bourhim, T.; Villareal, M.O.; Couderc, F.; Hafidi, A.; Isoda, H.; Gadhi, C. Melanogenesis promoting effect, antioxidant activity, and uplc-esi-hrms characterization of phenolic compounds of argan leaves extract. Molecules 2021, 26, 371. [Google Scholar] [CrossRef]
  57. Woo, S.M.; Choi, W.R.; Lee, D.R.; Kim, H.S.; Yi, C.; Kim, K.H.; Kim, H.L.; Cheng, J.; Le, B.; Yang, S.H.; et al. Leukodin isolated from Artemisia capillaris inhibits alpha-melanocyte stimulating hormone induced melanogenesis in B16F10 melanoma cells. Eur. J. Integr. Med. 2019, 25, 85–91. [Google Scholar] [CrossRef]
  58. Panichakul, T.; Rodboon, T.; Suwannalert, P.; Tripetch, C.; Rungruang, R.; Boohuad, N.; Youdee, P. Additive effect of a combination of Artocarpus Lakoocha and Glycyrrhiza Glabra extracts on tyrosinase inhibition in melanoma B16 cells. Pharmaceuticals 2020, 13, 1–17. [Google Scholar] [CrossRef]
  59. Yamahara, M.; Sugimura, K.; Kumagai, A.; Fuchino, H.; Kuroi, A.; Kagawa, M.; Itoh, Y.; Kawahara, H.; Nagaoka, Y.; Iida, O.; et al. Callicarpa longissima extract, carnosol-rich, potently inhibits melanogenesis in B16F10 melanoma cells. J. Nat. Med. 2016, 70, 28–35. [Google Scholar] [CrossRef]
  60. Lall, N.; Kishore, N.; Momtaz, S.; Hussein, A.; Naidoo, S.; Nqephe, M.; Crampton, B. Extract from Ceratonia siliqua exhibits depigmentation properties. Phyther. Res. 2015, 29, 1729–1736. [Google Scholar] [CrossRef]
  61. Kang, M.H.; Jang, G.Y.; Ji, Y.J.; Lee, J.H.; Choi, S.J.; Hyun, T.K.; Kim, H.D. Antioxidant and anti-melanogenic activities of heat-treated licorice (Wongam, glycyrrhiza glabra × g. uralensis) extract. Curr. Issues Mol. Biol. 2021, 43, 1171–1187. [Google Scholar] [CrossRef]
  62. Choi, J.H.; Jung, J.G.; Kim, J.E.; Bang, M.A. Anti-melanogenic effects of Hordeum vulgare L. Barely sprout extract in murine B16F10 melanoma cells. J. Nutr. Health 2019, 52, 168–175. [Google Scholar] [CrossRef]
  63. Jegal, J.; Chung, K.W.; Chung, H.Y.; Jeong, E.J.; Yang, M.H. The standardißed extract of juniperus communis alleviates hyperpigmentation in vivo HRM-2 hairless mice and in vitro murine B16 melanoma cells. Biol. Pharm. Bull. 2017, 40, 1381–1388. [Google Scholar] [CrossRef] [Green Version]
  64. Pedrosa, T.D.N.; Barros, A.O.; Nogueira, J.R.; Fruet, A.C.; Rodrigues, I.C.; Calcagno, D.Q.; Smith, M.D.A.C.; de Souza, T.P.; Barros, S.B.D.M.; de Vasconcellos, M.C.; et al. Anti-wrinkle and anti-whitening effects of jucá (Libidibia ferrea Mart.) extracts. Arch. Dermatol. Res. 2016, 308, 643–654. [Google Scholar] [CrossRef]
  65. Lee, S.G.; Karadeniz, F.; Seo, Y.; Kong, C.S. Anti-melanogenic effects of flavonoid glycosides from limonium tetragonum (thunb.) bullock via inhibition of tyrosinase and tyrosinase-related proteins. Molecules 2017, 22, 1480. [Google Scholar] [CrossRef] [Green Version]
  66. Juang, L.J.; Gao, X.Y.; Mai, S.T.; Lee, C.H.; Lee, M.C.; Yao, C.L. Safety assessment, biological effects, and mechanisms of Myrica rubra fruit extract for anti-melanogenesis, anti-oxidation, and free radical scavenging abilities on melanoma cells. J. Cosmet. Dermatol. 2019, 18, 322–332. [Google Scholar] [CrossRef] [Green Version]
  67. Li, H.; Dasilva, N.A.; Liu, W.; Xu, J.; Dombi, G.W.; Dain, J.A.; Li, D.; Chamcheu, J.C.; Seeram, N.P.; Ma, H. Thymocid®, a standardized black cumin (Nigella sativa) seed extract, modulates collagen cross-linking, collagenase and elastase activities, and melanogenesis in murine B16F10 melanoma cells. Nutrients 2020, 12, 2146. [Google Scholar] [CrossRef]
  68. Pliego-Arreaga, R.; Regalado, C.; Amaro-Reyes, A.; García-Almendárez, B.E. Changes in the phenolic compounds profile, antioxidant and anti-melanogenic activity from organs of Petasites japonicas under di erent extraction methods. Rev. Mex. Ing. Química 2013, 12, 505–511. [Google Scholar]
  69. Sripanidkulchai, B.; Junlatat, J. Bioactivities of alcohol based extracts of Phyllanthus emblica branches: Antioxidation, antimelanogenesis and anti-inflammation. J. Nat. Med. 2014, 68, 615–622. [Google Scholar] [CrossRef]
  70. Han, E.B.; Chang, B.Y.; Kim, D.S.; Cho, H.K.; Kim, S.Y. Melanogenesis inhibitory effect of aerial part of Pueraria thunbergiana in vitro and in vivo. Arch. Dermatol. Res. 2014, 307, 57–72. [Google Scholar] [CrossRef] [Green Version]
  71. Gao, D.; Kim, J.H.; Kim, C.T.; Jeong, W.S.; Kim, H.M.; Sim, J.; Kang, J.S. Evaluation of anti-melanogenesis activity of enriched pueraria lobata stem extracts and characterization of its phytochemical components using hplc–pda–esi–ms/ms. Int. J. Mol. Sci. 2021, 22, 8105. [Google Scholar] [CrossRef]
  72. Kim, M.J.; Kim, S.S.; Yun, S.H.; Kim, S.Y.; Hyun, K.H.; Lee, J.; Lee, N.H.; Hyun, C.G. Melanogenesis inhibitory activity of Rhododendron weyrichii in mouse B16 melanoma cells. Orient. J. Chem. 2016, 32, 1899–1907. [Google Scholar] [CrossRef] [Green Version]
  73. Han, H.J.; Park, S.K.; Kang, J.Y.; Kim, J.M.; Yoo, S.K.; Heo, H.J. 2020 nutrients Anti-Melanogenic Effect of Ethanolic Extract of Sorghum bicolor on IBMX–Induced Melanogenesis in B16/F10 melanoma cells. Nutrients 2020, 12, 832. [Google Scholar] [CrossRef] [Green Version]
  74. Tuerxuntayi, A.; Liu, Y.Q.; Tulake, A.; Kabas, M.; Eblimit, A.; Aisa, H.A. Kaliziri extract upregulates tyrosinase, TRP-1, TRP-2 and MITF expression in murine B16 melanoma cells. BMC Complement. Altern. Med. 2014, 14, 166. [Google Scholar] [CrossRef] [Green Version]
  75. Yoon, W.J.; Ham, Y.M.; Yoon, H.S.; Lee, W.J.; Lee, N.H.; Hyun, C.G. Acanthoic acid inhibits melanogenesis through tyrosinase down-regulation and melanogenic gene expression in B16 melanoma cells. Nat. Prod. Commun. 2013, 8, 1359–1362. [Google Scholar] [CrossRef] [Green Version]
  76. Yim, S.H.; Tabassum, N.; Kim, W.H.; Cho, H.; Lee, J.H.; Batkhuu, G.J.; Kim, H.J.; Oh, W.K.; Jung, D.W.; Williams, D.R. Isolation and Characterization of Isofraxidin 7-O-(6′-O-p-Coumaroyl)-β-glucopyranoside from Artemisia capillaris Thunberg: A Novel, Nontoxic Hyperpigmentation Agent That Is Effective In Vivo. Evid.-Based Complement. Altern. Med. 2017, 2017, 867494. [Google Scholar] [CrossRef] [Green Version]
  77. Tabassum, N.; Lee, J.H.; Yim, S.H.; Batkhuu, G.J.; Jung, D.W.; Williams, D.R. Isolation of 4,5-O-Dicaffeoylquinic Acid as a Pigmentation Inhibitor Occurring in Artemisia capillaris Thunberg and Its Validation In Vivo. Evid.-Based Complement. Altern. Med. 2016, 2016, 1–11. [Google Scholar] [CrossRef] [Green Version]
  78. Chang, T.S.; Chao, S.Y.; Ding, H.Y. Melanogenesis inhibition by homoisoflavavone sappanone a from Caesalpinia sappan. Int. J. Mol. Sci. 2012, 13, 10359–10367. [Google Scholar] [CrossRef]
  79. Hashemi-Shahri, S.H.; Golshan, A.; Mohajeri, S.A.; Baharara, J.; Amini, E.; Salek, F.; Sahebkar, A.; Tayarani-Najaran, Z. ROS-scavenging and Anti-tyrosinase Properties of Crocetin on B16F10 Murine Melanoma Cells. Anticancer. Agents Med. Chem. 2017, 18, 1064–1069. [Google Scholar] [CrossRef]
  80. Liu, Z.J.; Wang, Y.L.; Li, Q.L.; Yang, L. Improved antimelanogenesis and antioxidant effects of polysaccharide from cuscuta Chinensis lam seeds after enzymatic hydrolysis. Brazilian J. Med. Biol. Res. 2018, 51, e7256. [Google Scholar] [CrossRef]
  81. Kim, I.S.; Yoon, S.J.; Park, Y.J.; Lee, H.B. Inhibitory effect of ephedrannins A and B from roots of Ephedra sinica STAPF on melanogenesis. Biochim. Biophys. Acta-Gen. Subj. 2015, 1850, 1389–1396. [Google Scholar] [CrossRef]
  82. Han, S.K.; Kim, Y.G.; Kang, H.C.; Huh, J.R.; Kim, J.Y.; Baek, N.I.; Lee, D.K.; Lee, D.G. Oleanolic acid from Fragaria ananassa calyx leads to inhibition of α-MSH-induced melanogenesis in B16-F10 melanoma cells. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 735–742. [Google Scholar] [CrossRef]
  83. Manse, Y.; Ninomiya, K.; Okazaki, A.; Okada-Nishida, E.; Imagawa, T.; Imamura-Mizushima, M.; Yamano, Y.; Kaname, K.; Nakamura, S.; Morikawa, T. Melanogenesis inhibitory activity of diterpenoid and triterpenoid constituents from the aerial part of Isodon trichocarpus. Natural Product. Commun. 2017, 12, 1185–1188. [Google Scholar] [CrossRef] [Green Version]
  84. Woo, S.Y.; Wong, C.P.; Win, N.N.; Lae, K.Z.W.; Woo, B.; Elsabbagh, S.A.; Liu, Q.Q.; Ngwe, H.; Morita, H. Anti-melanin deposition activity and active constituents of Jatropha multifida stems. J. Nat. Med. 2019, 73, 805–813. [Google Scholar] [CrossRef]
  85. Ninomiya, K.; Matsumoto, T.; Chaipech, S.; Miyake, S.; Katsuyama, Y.; Tsuboyama, A.; Pongpiriyadacha, Y.; Hayakawa, T.; Muraoka, O.; Morikawa, T. Simultaneous quantitative analysis of 12 methoxyflavones with melanogenesis inhibitory activity from the rhizomes of Kaempferia parviflora. J. Nat. Med. 2016, 70, 179–189. [Google Scholar] [CrossRef]
  86. Lee, Y.K.; Hwang, B.S.; Hwang, Y.; Lee, S.Y.; Oh, Y.T.; Kim, C.H.; Nam, H.J.; Jeong, Y.T. Melanogenesis inhibitory activity of epicatechin-3-O-gallate isolated from Polygonum amphibium L. Microbiol. Biotechnol. Lett. 2021, 49, 24–31. [Google Scholar] [CrossRef]
  87. Kuroi, A.; Sugimura, K.; Kumagai, A.; Kohara, A.; Nagaoka, Y.; Kawahara, H.; Yamahara, M.; Kawahara, N.; Takemori, H.; Fuchino, H. The Importance of 11α-OH, 15-oxo, and 16-en Moieties of 11α-Hydroxy-15-oxo-kaur-16-en-19-oic Acid in Its Inhibitory Activity on Melanogenesis. Skin Pharmacol. Physiol. 2017, 30, 205–215. [Google Scholar] [CrossRef] [Green Version]
  88. Shim, S.Y.; Lee, Y.E.; Song, H.Y.; Lee, M. P-hydroxybenzoic acid β-d-glucosyl ester and cimidahurinine with antimelanogenesis and antioxidant effects from Pyracantha angustifolia via bioactivity-guided fractionation. Antioxidants 2020, 9, 258. [Google Scholar] [CrossRef] [Green Version]
  89. Seong, Z.K.; Kim, H.S.; Won, Y.M.; Kim, J.L.; Song, H.H.; Kim, D.Y.; Oh, S.R.; Cho, H.W.; Cho, J.H.; Lee, H.K. Phenylacylphenol derivatives with anti-melanogenic activity from Stewartia pseudocamellia. Arch. Pharm. Res. 2016, 39, 636–645. [Google Scholar] [CrossRef]
  90. Park, H.J.; Cho, J.H.; Hong, S.H.; Kim, D.H.; Jung, H.Y.; Kang, I.K.; Cho, Y.J. Whitening and anti-wrinkle activities of ferulic acid isolated from Tetragonia tetragonioides in B16F10 melanoma and CCD-986sk fibroblast cells. J. Nat. Med. 2018, 72, 127–135. [Google Scholar] [CrossRef]
  91. Zhang, J.; Kurita, M.; Ebina, K.; Ukiya, M.; Tokuda, H.; Yasukawa, K.; Masters, E.T.; Shimizu, N.; Akihisa, M.; Feng, F.; et al. Melanogenesis-inhibitory activity and cancer chemopreventive effect of glucosylcucurbic acid from shea (vitellaria paradoxa) kernels. Chem. Biodivers. 2015, 12, 547–558. [Google Scholar] [CrossRef]
  92. Won, Y.M.; Seong, Z.K.; Kim, J.L.; Kim, H.S.; Song, H.H.; Kim, D.Y.; Kim, J.H.; Oh, S.R.; Cho, H.W.; Cho, J.H.; et al. Triterpene glycosides with stimulatory activity on melanogenesis from the aerial parts of Weigela subsessilis. Arch. Pharm. Res. 2015, 38, 1541–1551. [Google Scholar] [CrossRef]
  93. Mamat, N.; Lu, X.Y.; Kabas, M.; Aisa, H.A. Potential anti-vitiligo properties of cynarine extracted from Vernonia anthelmintica (L.) Willd. Int. J. Mol. Med. 2018, 42, 2665–2675. [Google Scholar] [CrossRef] [PubMed]
  94. Choi, M.H.; Jo, H.G.; Yang, J.H.; Ki, S.H.; Shin, H.J. Antioxidative and anti-melanogenic activities of bamboo stems (Phyllostachys nigra variety henosis) via PKA/CREB-mediated MITF downregulation in B16F10 melanoma cells. Int. J. Mol. Sci. 2018, 19, 409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Park, J.U.; Yang, S.Y.; Guo, R.H.; Li, H.X.; Kim, Y.H.; Kim, Y.R. Anti-Melanogenic Effect of Dendropanax morbiferus and Its Active Components via Protein Kinase A/Cyclic Adenosine Monophosphate-Responsive Binding Protein- and p38 Mitogen-Activated Protein Kinase-Mediated Microphthalmia−Associated Transcription Factor Do. Front. Pharmacol. 2020, 11, 507. [Google Scholar] [CrossRef] [PubMed]
  96. Hsu, J.Y.; Lin, H.H.; Li, T.S.; Tseng, C.Y.; Wong, Y.; Chen, J.H. Anti-melanogenesis effects of Guinea Pigs in vitro and in vivo. Nutrients 2020, 12, 3535. [Google Scholar] [CrossRef]
  97. Roh, E.; Yun, C.Y.; Yun, J.Y.; Park, D.; Doo Kim, N.; Yeon Hwang, B.; Jung, S.H.; Park, S.K.; Kim, Y.B.; Han, S.B.; et al. CAMP-binding site of PKA as a molecular target of bisabolangelone against melanocyte-specific hyperpigmented disorder. J. Invest. Dermatol. 2013, 133, 1072–1079. [Google Scholar] [CrossRef] [Green Version]
  98. Park, H.; Song, K.H.; Jung, P.M.; Kim, J.E.; Ro, H.; Kim, M.Y.; Ma, J.Y. Inhibitory effect of arctigenin from fructus arctii extract on melanin synthesis via repression of tyrosinase expression. Evid.-Based Complement. Altern. Med. 2013, 2013, 965312. [Google Scholar] [CrossRef] [Green Version]
  99. Kim, M.O.; Park, S.J.; Park, S.H.; Oh, S.W.; Lee, S.E.; Yoo, J.A.; Kwon, K.; Kim, J.; Kim, M.H.; Cho, J.Y.; et al. Ethanolic extract of Melia azedarach L. induces melanogenesis through the cAMP-PKA-CREB signaling pathway. Mol. Cell. Toxicol. 2019, 15, 75–83. [Google Scholar] [CrossRef]
  100. Oh, S.W.; Park, S.H.; Lee, H.S.; Kang, M.; Lee, S.E.; Yoo, J.A.; Cho, J.Y.; Lee, J. Melanogenic mechanism of ethanolic extract of Dalbergia odorifera. Mol. Cell. Toxicol. 2017, 13, 453–459. [Google Scholar] [CrossRef]
  101. Kim, H.J.; Kim, I.S.; Dong, Y.; Lee, I.S.; Kim, J.S.; Kim, J.S.; Woo, J.T.; Cha, B.Y. Melanogenesis-inducing effect of cirsimaritin through increases in microphthalmia-associated transcription factor and tyrosinase expression. Int. J. Mol. Sci. 2015, 16, 8772–8788. [Google Scholar] [CrossRef] [Green Version]
  102. Huang, Y.C.; Liu, K.C.; Chiou, Y.L. Melanogenesis of murine melanoma cells induced by hesperetin, a Citrus hydrolysate-derived flavonoid. Food Chem. Toxicol. 2012, 50, 653–659. [Google Scholar] [CrossRef]
  103. Ahn, M.J.; Hur, S.J.; Kim, E.H.; Lee, S.H.; Shin, J.S.; Kim, M.K.; Uchizono, J.A.; Whang, W.K.; Kim, D.S. Scopoletin from Cirsium setidens increases melanin synthesis via CREB phosphorylation in B16F10 cells. Korean J. Physiol. Pharmacol. 2014, 18, 307–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lai, P.-J.; Kao, E.-S.; Chen, S.-R.; Huang, Y.-T.; Wang, C.-J.; Huang, H.-P. Nelumbo nucifera Leaf Extracts Inhibit Melanogenesis in B16 Melanoma Cells and Guinea Pigs through Downregulation of CREB/MITF Activation. J. Food Nutr. Res. 2020, 8, 459–465. [Google Scholar] [CrossRef]
  105. Chiang, H.M.; Chien, Y.C.; Wu, C.H.; Kuo, Y.H.; Wu, W.C.; Pan, Y.Y.; Su, Y.H.; Wen, K.C. Hydroalcoholic extract of Rhodiola rosea L. (Crassulaceae) and its hydrolysate inhibit melanogenesis in B16F0 cells by regulating the CREB/MITF/tyrosinase pathway. Food Chem. Toxicol. 2014, 65, 129–139. [Google Scholar] [CrossRef] [PubMed]
  106. Lee, J.H.; Lee, B.; Jeon, Y.D.; Song, H.W.; Lee, Y.M.; Song, B.J.; Kim, D.K. Inhibitory effect of elaeagnus umbellata fractions on melanogenesis in α-MSH-stimulated B16-F10 melanoma cells. Molecules 2021, 26, 1308. [Google Scholar] [CrossRef]
  107. Ko, H.H.; Chang, Y.T.; Kuo, Y.H.; Lin, C.H.; Chen, Y.F. Oenothera laciniata Hill Extracts Exhibits Antioxidant Effects and Attenuates Melanogenesis in B16-F10 Cells via Downregulating CREB/MITF/Tyrosinase and Upregulating p-ERK and p-JNK. Plants 2021, 10, 727. [Google Scholar] [CrossRef]
  108. Jeon, J.S.; Kang, H.M.; Park, J.H.; Kang, J.S.; Lee, Y.J.; Park, Y.H.; Je, B.I.; Park, S.Y.; Choi, Y.W. A comparative study on photo-protective and anti-melanogenic properties of different kadsura coccinea extracts. Plants 2021, 10, 1633. [Google Scholar] [CrossRef]
  109. Wu, Q.Y.; Wong, Z.C.F.; Wang, C.; Fung, A.H.Y.; Wong, E.O.Y.; Chan, G.K.L.; Dong, T.T.X.; Chen, Y.; Tsim, K.W.K. Isoorientin derived from Gentiana veitchiorum Hemsl. flowers inhibits melanogenesis by down-regulating MITF-induced tyrosinase expression. Phytomedicine 2019, 57, 129–136. [Google Scholar] [CrossRef]
  110. Cai, Z.N.; Li, W.; Mehmood, S.; Pan, W.J.; Wu, Q.X.; Chen, Y.; Lu, Y.M. Effect of polysaccharide FMP-1 from Morchella esculenta on melanogenesis in B16F10 cells and zebrafish. Food Funct. 2018, 9, 5007–5015. [Google Scholar] [CrossRef]
  111. Jung, H.J.; Bang, E.; Kim, B.M.; Jeong, S.H.; Lee, G.H. Loganin Inhibits α-MSH and IBMX-induced Melanogenesis by Suppressing the Expression of Tyrosinase in B16F10 Melanoma Cells. J. Life Sci. 2019, 29, 1200–1207. [Google Scholar] [CrossRef]
  112. Seong, Z.K.; Lee, S.Y.; Poudel, A.; Oh, S.R.; Lee, H.K. Constituents of Cryptotaenia japonica inhibit melanogenesis via CREB- and MAPK-associated signaling pathways in murine B16 melanoma cells. Molecules 2016, 21, 1296. [Google Scholar] [CrossRef] [Green Version]
  113. Li, H.X.; Park, J.U.; Su, X.D.; Kim, K.T.; Kang, J.S.; Kim, Y.R.; Kim, Y.H.; Yang, S.Y. Identification of anti-melanogenesis constituents from morus alba L. Leaves. Molecules 2018, 23, 2559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Jang, D.K.; Pham, C.H.; Lee, I.S.; Jung, S.H.; Jeong, J.H.; Shin, H.S.; Yoo, H.M. Anti-melanogenesis activity of 6-O-isobutyrylbritannilactone from inula britannica on B16F10 melanocytes and in vivo zebrafish models. Molecules 2020, 25, 3887. [Google Scholar] [CrossRef] [PubMed]
  115. Huang, H.C.; Wang, S.S.; Tsai, T.C.; Ko, W.P.; Chang, T.M. Phoenix dactylifera L. Seed extract exhibits antioxidant effects and attenuates melanogenesis in B16F10 murine melanoma cells by downregulating PKA signaling. Antioxidants 2020, 9, 1270. [Google Scholar] [CrossRef] [PubMed]
  116. Cargnello, M.; Roux, P.P. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef] [Green Version]
  117. Buscà, R.; Abbe, P.; Mantoux, F.; Aberdam, E.; Peyssonnaux, C.; Eychène, A.; Ortonne, J.P.; Ballotti, R. Ras mediates the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes. EMBO J. 2000, 19, 2900–2910. [Google Scholar] [CrossRef] [Green Version]
  118. Gerits, N.; Kostenko, S.; Shiryaev, A.; Johannessen, M.; Moens, U. Relations between the mitogen-activated protein kinase and the cAMP-dependent protein kinase pathways: Comradeship and hostility. Cell. Signal. 2008, 20, 1592–1607. [Google Scholar] [CrossRef]
  119. Lee, A.Y. Recent progress in melasma pathogenesis. Pigment Cell Melanoma Res. 2015, 28, 648–660. [Google Scholar] [CrossRef]
  120. Kim, M.; Shibata, T.; Kwon, S.; Park, T.J.; Kang, H.Y. Ultraviolet-irradiated endothelial cells secrete stem cell factor and induce epidermal pigmentation. Sci. Rep. 2018, 8, 4235. [Google Scholar] [CrossRef]
  121. Li, P.H.; Liu, L.H.; Chang, C.C.; Gao, R.; Leung, C.H.; Ma, D.L.; David Wang, H.M. Silencing stem cell factor gene in fibroblasts to regulate paracrine factor productions and enhance c-Kit expression in melanocytes on melanogenesis. Int. J. Mol. Sci. 2018, 19, 19. [Google Scholar] [CrossRef] [Green Version]
  122. Kim, J.H.; Hong, A.R.; Kim, Y.H.; Yoo, H.; Kang, S.W.; Chang, S.E.; Song, Y. JNK suppresses melanogenesis by interfering with CREB-regulated transcription coactivator 3-dependent MITF expression. Theranostics 2020, 10, 4017–4029. [Google Scholar] [CrossRef]
  123. Ahn, J.H.; Jin, S.H.; Kang, H.Y. LPS induces melanogenesis through p38 MAPK activation in human melanocytes. Arch. Dermatol. Res. 2008, 300, 325–329. [Google Scholar] [CrossRef] [PubMed]
  124. Kim, D.; Kim, H.J.; Jun, H.S. Polygonum multiflorum Thunb. Extract Stimulates Melanogenesis by Induction of COX2 Expression through the Activation of p38 MAPK in B16F10 Mouse Melanoma Cells. Evid.-Based Complement. Altern. Med. 2020, 2020, 1–10. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, J.; Shang, J.; Ping, F.; Zhao, G. Alcohol extract from Vernonia anthelmintica (L.) willd seed enhances melanin synthesis through activation of the p38 MAPK signaling pathway in B16F10 cells and primary melanocytes. J. Ethnopharmacol. 2012, 143, 639–647. [Google Scholar] [CrossRef] [PubMed]
  126. Ko, G.A.; Kang, H.R.; Moon, J.Y.; Ediriweera, M.K.; Eum, S.; Bach, T.T.; Cho, S.K. Annona squamosa L. leaves inhibit alpha-melanocyte-stimulating hormone (α-MSH) stimulated melanogenesis via p38 signaling pathway in B16F10 melanoma cells. J. Cosmet. Dermatol. 2020, 19, 1785–1792. [Google Scholar] [CrossRef]
  127. Jang, J.Y.; Kim, H.N.; Kim, Y.R.; Choi, Y.H.; Kim, B.W.; Shin, H.K.; Choi, B.T. Aqueous fraction from Cuscuta japonica seed suppresses melanin synthesis through inhibition of the p38 mitogen-activated protein kinase signaling pathway in B16F10 cells. J. Ethnopharmacol. 2012, 141, 338–344. [Google Scholar] [CrossRef]
  128. Masuda, M.; Itoh, K.; Murata, K.; Naruto, S.; Uwaya, A.; Isami, F.; Matsuda, H. Inhibitory Effects of Morinda citrifolia Extract and Its Constituents on Melanogenesis in Murine B16 Melanoma Cells. Biol. Pharm. Bull. 2012, 35, 78–83. [Google Scholar] [CrossRef] [Green Version]
  129. Kim, H.J.; Kim, J.S.; Woo, J.T.; Lee, I.S.; Cha, B.Y. Hyperpigmentation mechanism of methyl 3,5-di-caffeoylquinate through activation of p38 and MITF induction of tyrosinase. Acta Biochim. Biophys. Sin. 2015, 47, 548–556. [Google Scholar] [CrossRef] [Green Version]
  130. Yao, C.; Jin, C.L.; Oh, J.H.; Oh, I.G.; Park, C.H.; Chung, J.H. Ardisia crenata extract stimulates melanogenesis in B16F10 melanoma cells through inhibiting ERK1/2 and Akt activation. Mol. Med. Rep. 2015, 11, 653–657. [Google Scholar] [CrossRef] [Green Version]
  131. Villareal, M.O.; Han, J.; Matsuyama, K.; Sekii, Y.; Smaoui, A.; Shigemori, H.; Isoda, H. Lupenone from erica multiflora leaf extract stimulates melanogenesis in B16 murine melanoma cells through the inhibition of ERK1/2 activation. Planta Med. 2013, 79, 236–243. [Google Scholar] [CrossRef]
  132. Tsao, Y.T.; Kuo, C.Y.; Kuan, Y.D.; Lin, H.C.; Wu, L.H.; Lee, C.H. The extracts of astragalus membranaceus inhibit melanogenesis through the ERK signaling pathway. Int. J. Med. Sci. 2017, 14, 1049–1053. [Google Scholar] [CrossRef] [Green Version]
  133. Lee, J.I.; Seo, J.H.; Ko, E.S.; Cho, S.M.; Kang, J.R.; Jeong, J.H.; Jeong, Y.J.; Kim, C.Y.; Cha, J.D.; Kim, W.S.; et al. Inhibition of melanogenesis by aster yomena callus pellet extract in melanoma cells and patients with skin pigmentation. Int. J. Med. Sci. 2021, 18, 3299–3308. [Google Scholar] [CrossRef] [PubMed]
  134. Yuan, X.H.; Tian, Y.D.; Oh, J.H.; Bach, T.T.; Chung, J.H.; Jin, Z.H. Melochia corchorifolia extract inhibits melanogenesis in B16F10 mouse melanoma cells via activation of the ERK signaling. J. Cosmet. Dermatol. 2020, 19, 2421–2427. [Google Scholar] [CrossRef] [PubMed]
  135. Saba, E.; Oh, M.J.; Lee, Y.Y.; Kwak, D.; Kim, S.; Rhee, M.H. Artemisia capillaris Thunb. inhibits melanin synthesis activity via ERK-dependent MITF pathway in B16/F10 melanoma cells. Korean J. Vet. Res. 2018, 58, 1–7. [Google Scholar] [CrossRef]
  136. Im, D.S.; Lee, J.M.; Lee, J.; Shin, H.J.; No, K.T.; Park, S.H.; Kim, K. Inhibition of collagenase and melanogenesis by ethanol extracts of Orostachys japonicus A. Berger: Possible involvement of Erk and Akt signaling pathways in melanoma cells. Acta Biochim. Biophys. Sin. 2017, 49, 945–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Hwang, G.Y.; Choung, S.Y. Anti-melanogenic effects of Aster spathulifolius extract in UVB-exposed C57BL/6J mice and B16F10 melanoma cells through the regulation of MAPK/ERK and AKT/GSK3β signalling. J. Pharm. Pharmacol. 2016, 68, 503–513. [Google Scholar] [CrossRef]
  138. Kim, J.Y.; Lee, E.J.; Ahn, Y.; Park, S.J.; Kim, S.H.; Oh, S.H. A chemical compound from fruit extract of Juglans mandshurica inhibits melanogenesis through p-ERK-associated MITF degradation. Phytomedicine 2019, 57, 57–64. [Google Scholar] [CrossRef]
  139. Choo, S.J.; Ryoo, I.J.; Kim, K.C.; Na, M.; Jang, J.H.; Ahn, J.S.; Yoo, I.D. Hypo-pigmenting effect of sesquiterpenes from Inula britannica in B16 melanoma cells. Arch. Pharm. Res. 2014, 37, 567–574. [Google Scholar] [CrossRef]
  140. Oh, T.I.; Jung, H.J.; Lee, Y.M.; Lee, S.; Kim, G.H.; Kan, S.Y.; Kang, H.; Oh, T.; Ko, H.M.; Kwak, K.C.; et al. Zerumbone, a tropical ginger sesquiterpene of Zingiber officinale Roscoe, attenuates α-MSH-induced melanogenesis in B16F10 cells. Int. J. Mol. Sci. 2018, 19, 3149. [Google Scholar] [CrossRef] [Green Version]
  141. Kim, K.N.; Yang, H.M.; Kang, S.M.; Kim, D.; Ahn, G.; Jeon, Y.J. Octaphlorethol A isolated from Ishige foliacea inhibits α-MSH-stimulated induced melanogenesis via ERK pathway in B16F10 melanoma cells. Food Chem. Toxicol. 2013, 59, 521–526. [Google Scholar] [CrossRef]
  142. Kim, D.E.; Chang, B.Y.; Ham, S.O.; Kim, Y.C.; Kim, S.Y. Neobavaisoflavone inhibits melanogenesis through the regulation of Akt/GSK-3β and MEK/ERK pathways in B16F10 cells and a reconstructed human 3D skin model. Molecules 2020, 25, 2683. [Google Scholar] [CrossRef]
  143. Hong, C.O.; Lee, H.A.; Rhee, C.H.; Choung, S.Y.; Lee, K.W. Separation of the antioxidant compound quercitrin from Lindera obtusiloba blume and its antimelanogenic effect on B16F10 melanoma cells. Biosci. Biotechnol. Biochem. 2013, 77, 58–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Lee, C.J.; Park, S.K.; Kang, J.Y.; Kim, J.M.; Yoo, S.K.; Han, H.J.; Kim, D.O.; Heo, H.J. Melanogenesis regulatory activity of the ethyl acetate fraction from Arctium lappa L. leaf on α-MSH–induced B16/F10 melanoma cells. Ind. Crops Prod. 2019, 138, 111581. [Google Scholar] [CrossRef]
  145. Fu, Y.T.; Lee, C.W.; Ko, H.H.; Yen, F.L. Extracts of artocarpus communis decrease-melanocyte stimulating hormone-induced melanogenesis through activation of ERK and JNK signaling pathways. Sci. World J. 2014, 2014, 1–8. [Google Scholar] [CrossRef]
  146. Yen, F.L.; Wang, M.C.; Liang, C.J.; Ko, H.H.; Lee, C.W. Melanogenesis inhibitor(s) from Phyla nodiflora extract. Evidence-Based Complement. Altern. Med. 2012, 2012, 867494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Jeong, D.; Lee, J.; Park, S.H.; Kim, Y.A.; Park, B.J.; Oh, J.; Sung, G.H.; Aravinthan, A.; Kim, J.H.; Kang, H.; et al. Antiphotoaging and antimelanogenic effects of penthorum chinense pursh ethanol extract due to antioxidant- and autophagy-inducing properties. Oxid. Med. Cell. Longev. 2019, 2019, 1–14. [Google Scholar] [CrossRef] [PubMed]
  148. In, M.-H.; Jeon, B.K.; Mun, Y.-J.; Woo, W.-H. Hexane Extract of Kaempferia galanga L. Suppresses Melanogenesis via p38, JNK and Akt. Korean J. Orient. Physiol. Pathol. 2016, 30, 47. [Google Scholar] [CrossRef]
  149. Sim, M.O.; Ham, J.R.; Lee, M.K. Young leaves of reed (Phragmites communis) suppress melanogenesis and oxidative stress in B16F10 melanoma cells. Biomed. Pharmacother. 2017, 93, 165–171. [Google Scholar] [CrossRef]
  150. Ko, H.H.; Tsai, Y.T.; Yen, M.H.; Lin, C.C.; Liang, C.J.; Yang, T.H.; Lee, C.W.; Yen, F.L. Norartocarpetin from a folk medicine Artocarpus communis plays a melanogenesis inhibitor without cytotoxicity in B16F10 cell and skin irritation in mice. BMC Complement. Altern. Med. 2013, 13, 348. [Google Scholar] [CrossRef] [Green Version]
  151. Makbal, R.; Villareal, M.O.; Gadhi, C.; Hafidi, A.; Isoda, H. Argania spinosa fruit shell extract-induced melanogenesis via cAMP signaling pathway activation. Int. J. Mol. Sci. 2020, 21, 2539. [Google Scholar] [CrossRef] [Green Version]
  152. Ko, H.H.; Chiang, Y.C.; Tsai, M.H.; Liang, C.J.; Hsu, L.F.; Li, S.Y.; Wang, M.C.; Yen, F.L.; Lee, C.W. Eupafolin, a skin whitening flavonoid isolated from Phyla nodiflora, downregulated melanogenesis: Role of MAPK and Akt pathways. J. Ethnopharmacol. 2014, 151, 386–393. [Google Scholar] [CrossRef]
  153. Chung, Y.C.; Lee, J.N.; Kim, B.S.; Hyun, C.G. Anti-melanogenic effects of Paederia foetida L. Extract via mapk signaling-mediated mitf downregulation. Cosmetics 2021, 8, 22. [Google Scholar] [CrossRef]
  154. Song, Y.R.; Lim, W.C.; Han, A.; Lee, M.H.; Shin, E.J.; Lee, K.M.; Nam, T.G.; Lim, T.G. Rose Petal Extract (Rosa gallica) Exerts Skin Whitening and Anti-Skin Wrinkle Effects. J. Med. Food 2020, 23, 870–878. [Google Scholar] [CrossRef] [PubMed]
  155. Huang, H.C.; Liao, C.C.; Peng, C.C.; Lim, J.M.; Siao, J.H.; Wei, C.M.; Chen, C.C.; Wu, C.S.; Chang, T.M. Dihydromyricetin from Ampelopsis grossedentata inhibits melanogenesis through down-regulation of MAPK, PKA and PKC signaling pathways. Chem. Biol. Interact. 2016, 258, 166–174. [Google Scholar] [CrossRef]
  156. Khaled, M.; Larribere, L.; Bille, K.; Aberdam, E.; Ortonne, J.P.; Ballotti, R.; Bertolotto, C. Glycogen synthase kinase 3β is activated by cAMP and plays an active role in the regulation of melanogenesis. J. Biol. Chem. 2002, 277, 33690–33697. [Google Scholar] [CrossRef] [Green Version]
  157. Wang, C.; Zhao, L.; Su, Q.; Fan, X.; Wang, Y.; Gao, S.; Wang, H.; Chen, H.; Chan, C.B.; Liu, Z. Phosphorylation of MITF by AKT affects its downstream targets and causes TP53-dependent cell senescence. Int. J. Biochem. Cell Biol. 2016, 80, 132–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Mosca, S.; Cardinali, G.; Flori, E.; Briganti, S.; Bottillo, I.; Mileo, A.M.; Maresca, V. The PI3K pathway induced by αMSH exerts a negative feedback on melanogenesis and contributes to the release of pigment. Pigment Cell Melanoma Res. 2021, 34, 72–88. [Google Scholar] [CrossRef] [PubMed]
  159. Duda, P.; Akula, S.M.; Abrams, S.L.; Steelman, L.S.; Martelli, A.M.; Cocco, L.; Ratti, S.; Candido, S.; Libra, M.; Montalto, G.; et al. Targeting GSK3 and Associated Signaling Pathways Involved in Cancer. Cells 2020, 9, 1110. [Google Scholar] [CrossRef]
  160. Phacharapiyangkul, N.; Thirapanmethee, K.; Sa-Ngiamsuntorn, K.; Panich, U.; Lee, C.H.; Chomnawang, M.T. The ethanol extract of musa sapientum linn. Peel inhibits melanogenesis through akt signaling pathway. Cosmetics 2021, 8, 70. [Google Scholar] [CrossRef]
  161. Ku, B.; Kim, D.; Choi, E.M. Anti-melanogenic effect of the aqueous ethanol extract of Ginkgo biloba leaf in B16F10 cells. Toxicol. Environ. Health Sci. 2020, 12, 287–295. [Google Scholar] [CrossRef]
  162. Chen, H.; Weng, Q.Y.; Fisher, D.E. UV signaling pathways within the skin. J. Investig. Dermatol. 2014, 134, 2080–2085. [Google Scholar] [CrossRef] [Green Version]
  163. Box, N.F.; Terzian, T. The role of p53 in pigmentation, tanning and melanoma. Pigment Cell Melanoma Res. 2008, 21, 525–533. [Google Scholar] [CrossRef] [PubMed]
  164. Lee, D.U.; Weon, K.Y.; Nam, D.Y.; Nam, J.H.; Kim, W.K. Skin protective effect of guava leaves against UV-induced melanogenesis via inhibition of ORAI1 channel and tyrosinase activity. Exp. Dermatol. 2016, 25, 977–982. [Google Scholar] [CrossRef] [PubMed]
  165. Nam, J.H.; Lee, D.U. Foeniculum vulgare extract and its constituent, trans-anethole, inhibit UV-induced melanogenesis via ORAI1 channel inhibition. J. Dermatol. Sci. 2016, 84, 305–313. [Google Scholar] [CrossRef] [PubMed]
  166. Nam, J.H.; Nam, D.Y.; Lee, D.U. Valencene from the Rhizomes of Cyperus rotundus Inhibits Skin Photoaging-Related Ion Channels and UV-Induced Melanogenesis in B16F10 Melanoma Cells. J. Nat. Prod. 2016, 79, 1091–1096. [Google Scholar] [CrossRef]
  167. Stanisz, H.; Vultur, A.; Herlyn, M.; Roesch, A.; Bogeski, I. The role of Orai-STIM calcium channels in melanocytes and melanoma. J. Physiol. 2016, 594, 2825–2835. [Google Scholar] [CrossRef] [Green Version]
  168. Zhang, H.; Chen, Z.; Zhang, A.; Gupte, A.A.; Hamilton, D.J. The Role of Calcium Signaling in Melanoma. Int. J. Mol. Sci. 2022, 23, 1010. [Google Scholar] [CrossRef]
Figure 1. Schematic presentation of melanin synthesis during melanogenesis in melanocytes. [Created by BioRender].
Figure 1. Schematic presentation of melanin synthesis during melanogenesis in melanocytes. [Created by BioRender].
Molecules 27 04360 g001
Figure 2. Selected signaling pathways involved in melanogenesis that are modulated by several isolated plant compounds. [Created by BioRender].
Figure 2. Selected signaling pathways involved in melanogenesis that are modulated by several isolated plant compounds. [Created by BioRender].
Molecules 27 04360 g002
Table 1. Modulation on gene expression related to melanogenesis in B16 melanoma cells by plant extracts without identified compounds.
Table 1. Modulation on gene expression related to melanogenesis in B16 melanoma cells by plant extracts without identified compounds.
Name of Species/FamilyPart of PlantType of SolventConcentrationMethodsEffectsRef.
Artemisia asiatica Nakai ex Pamp./Asteraceaewhole plantethanol25–50 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[25]
Camellia sinensis (L.) Kuntze/Theaceaeflowerethanol20–40 µg/mLRT-PCRReduced expression: TYR[26]
Castanea crenata Siebold & Zucc./Fagaceaeinner skinethyl acetate10–100 µg/mLWestern blottingReduced expression: TYR[27]
Cinnamomum osmophloeum Kaneh./Lauraceaeleavesethanol21.25 µg/mLRT-PCRReduced expression: MITF, TYR[28]
Coix lacryma-jobi L./Poaceaeseedsethanol20–40 mg/mLRT-PCR, Western blotting Reduced expression: MITF, TYR, TYRP-1, TYRP-2[29]
Croton roxburghii N.P.Balakr. and Croton sublyratus Kurz/Euphorbiaceaeleavesethanol25–100 µg/mLRT-PCR,
Western blotting
Reduced expression: MITF, TYR, TYRP-1, TYRP-2[30]
Dendrobium moniliforme (L.) Sw./Orchidaceaeleavesethanol12.5–50 µg/mLWestern blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[31]
Dendropanax morbiferus H.Lév./Araliaceae leavesethanol12.5–50 µg/mLWestern blottingReduced expression: TYR, TYRP-2[32]
Equisetum ramosissimum Desf./Equisetaceaewhole plantethyl acetate, dichloromethane10–100 µg/mLWestern blotting
Western blotting
ethyl acetate: Reduced expression: MITF, TYR, TYRP-1, TYRP-2; dichloromethane: Increased expression: MITF, TYR, TYRP-1, TYRP-2[33]
Euryale ferox Salisb./Nymphaeaceaeseedsethyl acetate30 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[34]
Gaillardia aristata Pursh/Asteraceaeflowersethanol10–20 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[35]
Garcinia mangostana L./Clusiaceaeleaveswater4–32 µg/mLWestern blottingIncreased expression: TYR[36]
Gastrodia elata Blume/Orchidaceaewhole plantwater0.5–5 mg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[37]
Glechoma hederacea L./Lamiaceaewhole plantwater0.1–1 mg/mLRT-PCR, Western blottingReduced expression: TYR[38]
Glycine max (L.) Merr./Fabaceaeseeds cell cultureethanol0.5–1 mg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[39]
Haloxylon scoparium Pomel/Amaranthaceae stemsethanol0.017%(w/v)RT-PCR, Western blottingReduced expression: MC1R, TYR, TYRP-1[40]
Kummerowia striata (Thunb.) Schindl./Fabaceaeaerial partsethanol100–400 µg/mLRT-PCR,
Western blotting
Reduced expression: MITF, TYR, TYRP-1, TYRP-2[41]
Kummerowia striata (Thunb.) Schindl./Fabaceaeaerial partsethanol100–400 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[42]
Melia azedarach L./Meliaceaewhole plantethanol20 µg/mLRT-PCR, Western blottingIncreased expression: TYRP-1[43]
Nepeta binaludensis Jamzad/Lamiaceaeaerial partsmethanol50 µg/mLWestern blottingReduced expression: TYR[44]
Nepeta sintenisii Bornm./Lamiaceaeaerial partsn-hexane, methanol, water50 µg/mLWestern blottingReduced expression: MITF[45]
Oplismenus undulatifolius (Ard.) P.Beauv./Poaceaewhole plantethanol5–15 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[46]
Oreocnide fruticosa (Gaudich.) Hand. Mazz./Urticaceaebranchesethyl acetate25–100 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[47]
Phyllanthus emblica L./Phyllanthaceaefruitswater0.05–1 mg/mLRT-PCR,
Western blotting
Reduced expression: MITF, TYR, TYRP-1, TYRP-2[48]
Pinus densiflora Siebold & Zucc./Pinaceaepine coneethyl acetate12.5–50 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[49]
Psoralea corylifolia (Babchi)/Fabaceae and Zingiber officinale Roscoe/Zingiberaceae; Psoralea corylifolia (Babchi)/Fabaceae and Eclipta prostrata (L.) L./Asteraceaewhole plantsmethanol10–100 µg/mLRT-PCR, Western blottingIncreased expression: MITF[50]
Syzygium cumini (L.) Skeels/Myrtaceaeleaves and branchethanol25–100 µg/mLRT-PCRReduced expression: TYR, TYRP-1, TYRP-2[51]
Uncaria rhynchophylla (Miq.) Miq./Rubiaceaestems and hooksethanol0.1–1 mg/mLRT-PCRReduced expression: TYR[52]
Vitis vinifera L./Vitaceaepericarp, seed, flesh, and grape stemethanol100 µg/mLWestern blottingIncreased expression: MITF, TYR, TYRP-1, TYRP-2[53]
Table 2. Modulation on gene expression related to melanogenesis in B16 melanoma cells by plant extracts with identified compounds.
Table 2. Modulation on gene expression related to melanogenesis in B16 melanoma cells by plant extracts with identified compounds.
Name of Species/FamilyPart of PlantType of SolventIdentified CompoundsConcentrationMethodsEffectsRef.
Acer rubrum L./Sapindaceaeleavesethanolphenolic compounds10 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[54]
Angelica polymorpha Maxim./Apiaceaeflowershexanearomadendrene, methoxsalen, bergapten, isopimpinellin, nonadencane0.1–100 µg/mLWestern blottingReduced expression: MITF, TYR[55]
Argania spinosa L.) Skeels/Sapotaceaeleavesethanol14 compounds30 µg/mLWestern blottingIncreased expression: TYR, TYRP-1,[56]
Artemisia capillaris Thunb./Asteraceaewhole plantethanolleukodin12.5–50 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[57]
Artocarpus lacucha Buch.-Ham./Moraceae and Glycyrrhiza glabra L./Fabaceaeheartwood and rootethanolgallic acid, oxyresveratrol, resveratrol and glabridin0.1 mg/mLWestern blottingReduced expression: MITF, TYRP-2[58]
Callicarpa longissima (Hemsl.) Merr./Lamiaceaewhole plantethanolcarnosol and carnosic acid0.1–10 µg/mLRT-PCRReduced expression: MITF[59]
Ceratonia siliqua L./Fabaceaeleaves, bark and fruitsethanolepicatechin-3-O-gallate, 1,2,3,6-tetra-O-galloyl-ß-D-glucose
 and gallocatechin-3-O-gallate
100 µg/mLRT-PCRReduced expression: TYR[60]
Glycyrrhiza glabra L. and Glycyrrhiza uralensis Fisch. ex DC./Fabaceaewhole plant/heat treatedethanolisoliquiritigenin100 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[61]
Hordeum vulgare L./Poaceaebarely sproutwaterp-coumaric, ferulic, and vanillic acids50–250 µg/mLWestern blottingReduced expression: MITF, TYR[62]
Juniperus communis L./Cupressaceaefruitsethanolhypolaetin-7-O-β-D-xylopyranoside and isoscutellarein-7-O-β-D-xylopyranoside50 µg/mLWestern blottingReduced expression: TYR[63]
Libidibia ferrea (Mart. ex Tul.) L.P.Queiroz/Fabaceaebark and podsethanol18 compounds25 µg/mLRT-PCR, Western blottingReduced expression: TYR[64]
Limonium tetragonum (Thunb.) Bullock/Plumbaginaceaewhole plantwater, methanol, buthanolmyricetin 3-galactoside and quercetin 3-O-
 -galactopyronaside
5–20 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[65]
Myrica rubra (Lour.) Siebold & Zucc./Myricaceaefruitswatermyricetin-O-deoxyhexoside, quercetin-O-deoxyhexoside,
 and aempferol-O-hexoside
0.5–2 mg/mLRT-PCR, Western blottingReduced expression: MITF, TYRP-1,[66]
Nigella sativa L./RanunculaceaeseedThymocid®thymoquinone20 µg/mLRT-PCR, Western blottingReduced expression: MITF, TYRP-1, TYRP-2[67]
Petasites japonicus (Siebold & Zucc.) Maxim./Asteraceaeleaves, stems, and rootswaterleaf extract-isorhamnetin (main)
 root extract-p-coumaric acid (main)
50–200 µg/mLRT-PCR, Western blottingReduced expression: TYR[68]
Phyllanthus emblica L./Phyllanthaceaebranchethanolgallic acid and
 vanillic acid
6.25–25 µg/mLRT-PCRReduced expression: TYR, TYRP-1, TYRP-2[69]
Pueraria montana (Lour.) Merr./Fabaceae aerial partsethanoldaidzein, daidzin, glycitein, glycitin, genistein, genistin 10–100 µg/mLRT-PCR, Western blottingReduced expression: TYR[70]
Pueraria montana (Lour.) Merr./Fabaceaestemsn-hexane12 compounds50 µg/mLRT-PCRReduced expression: TYR[71]
Rhododendron weyrichii Maxim./Ericaceae Durande flowersethanolp-coumaric acid25–200 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[72]
Sorghum bicolor (L.) Moench/Poaceaewhole plantethanol1-O-ca
eoylglycerol, dica
eoylglycerides, 1,3-O-dica
eoylglycerol, p-coumaroyl-ca
eoylglycerol, feruloyl-ca
eoylglycerol, Tricin, 9-hydroxyoctadecadienoic acid
2–10 µg/mLWestern blottingReduced expression: MITF, TYRP-1,[73]
Vernonia anthelmintica (L.) Willd./Asteraceaewhole plantethanol15 compounds (mainly flavonoids)20 µg/mLWestern blottingIncreased expression: TYR[74]
Table 3. Modulation on gene expression related to melanogenesis in B16 melanoma cells by isolated plant compounds.
Table 3. Modulation on gene expression related to melanogenesis in B16 melanoma cells by isolated plant compounds.
Name of Species/FamilyPart of PlantCompoundsConcentrationMethodsEffectsRef.
Acanthopanax koreanum Nakai/Araliaceaerootsacanthoic acid25–100 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[75]
Artemisia capillaris Thunb./Asteraceaewhole plantleukodin37.5–150 µg/mLWestern blottingReduced expression: TYRP-1, TYRP-2[57]
Artemisia capillaris Thunb./Asteraceaewhole plantisofraxidin 7-O-(6′-O-p-coumaroyl)-𝛽-glucopyranoside25 µg/mLRT-PCRIncreased expression: MITF, TYR[76]
Artemisia capillaris Thunb./Asteraceaeleaves and stems4,5-𝑂-dicaffeoylquinic acid25 µg/mLRT-PCRReduced expression: TYRP-1[77]
Caesalpinia sappan L./Fabaceaeheartwoodsappanone A4.4 µg/mLRT-PCRReduced expression: TYR[78]
Crocus sativus L./Iridaceaestigmascrocetin0.5–32 µg/mLWestern blottingReduced expression: MITF[79]
Cuscuta chinensis Lam./Convolvulaceaewhole plantpolysaccharide40–160 µg/mLWestern blottingReduced expression: MITF, TYR, TYRP-1[80]
Ephedra sinica Stapf/Ephedraceaerootsephedrannins A and BA: 18–72 µg/mL;
 B: 1.85–7.4 µg/mL
RT-PCRReduced expression: TYR[81]
Fragaria × ananassa (Duchesne ex Weston) Duchesne ex Rozier/Rosaceaecalyxoleanolic acid12.5 µg/mLWestern blottingReduced expression: TYR, TYRP-1, TYRP-2[82]
Isodon trichocarpus (Maxim.) Kudô./Lamiaceaeaerial partsenmein, isodocarpin, nodosin, oridonin1–3 µg/mLRT-PCRReduced expression: TYR, TYRP-1, TYRP-2[83]
Jatropha multifida L./EuphorbiaceaestemsSecoisolariciresinol6.25–200 µg/mLRT-PCRReduced expression: TYR[84]
Kaempferia parviflora Wall. ex Baker/Zingiberaceaerhizomes5-hydroxy-7,3′,4′-trimethoxyflavone, 5,7,3′,4′-tetramethoxyflavone, 5,3′-
 dihydroxy-3,7,4′-trimethoxyflavone and 5-hydroxy-3,7,3′,4′-tetramethoxyflavone
3–30 µg/mLRT-PCRReduced expression: TYR, TYRP-1, TYRP-2[85]
Limonium tetragonum (Thunb.) Bullock/Plumbaginaceae whole plantmyricetin 3-galactoside and quercetin 3-O-galactopyronaside10 µg/mLWestern blottingReduced expression: TYRP-1, TYRP-2[65]
Persicaria amphibia (L.) Delarbre/Polygonaceaewhole plantepicatechin-3-O-gallate25–200 µg/mLWestern blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[86]
Pteris dispar Kunze/Pteridaceaeleavesent -11α-hydroxy-15-oxo-kaur-16-en-19-oic acid10 µg/mLRT-PCR, Western blottingReduced expression: TYR[87]
Pyracantha angustifolia (Franch.) C.K.Schneid./Rosaceaeleaves, twigs, and fruits
 β-D-glucosylester and cimidahurinine
10–100 µg/mLWestern blottingReduced expression: TYRP-1, TYRP-2[88]
Stewartia pseudocamellia Maxim.twigsstewartianol and stewartianol-
 3-O-glucoside
20–80 µg/mLWestern blottingReduced expression: MITF[89]
Tetragonia tetragonoides (Pall.) Kuntze/Aizoaceaewhole plantferulic acid5–20 µg/mLWestern blottingReduced expression: MITF, TYR[90]
Vitellaria paradoxa C.F.Gaertn./Sapotaceaefruitglucosylcucurbic acid and cucurbic acid30–100 µg/mLWestern blottingReduced expression: MITF, TYR, TYRP-1, TYRP-2[91]
Weigela subsessilis (Nakai) L.H.Bailey/Caprifoliaceae aerial partsloniceroside A, loniceroside
 L
1–20 µg/mLWestern blottingIncreased expression: MITF, TYR[92]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Merecz-Sadowska, A.; Sitarek, P.; Kowalczyk, T.; Zajdel, K.; Kucharska, E.; Zajdel, R. The Modulation of Melanogenesis in B16 Cells Upon Treatment with Plant Extracts and Isolated Plant Compounds. Molecules 2022, 27, 4360. https://doi.org/10.3390/molecules27144360

AMA Style

Merecz-Sadowska A, Sitarek P, Kowalczyk T, Zajdel K, Kucharska E, Zajdel R. The Modulation of Melanogenesis in B16 Cells Upon Treatment with Plant Extracts and Isolated Plant Compounds. Molecules. 2022; 27(14):4360. https://doi.org/10.3390/molecules27144360

Chicago/Turabian Style

Merecz-Sadowska, Anna, Przemysław Sitarek, Tomasz Kowalczyk, Karolina Zajdel, Ewa Kucharska, and Radosław Zajdel. 2022. "The Modulation of Melanogenesis in B16 Cells Upon Treatment with Plant Extracts and Isolated Plant Compounds" Molecules 27, no. 14: 4360. https://doi.org/10.3390/molecules27144360

Article Metrics

Back to TopTop