Skip to main content
International Journal of Molecular Sciences logoLink to International Journal of Molecular Sciences
. 2009 Dec 10;10(12):5326–5349. doi: 10.3390/ijms10125326

The Hunt for Natural Skin Whitening Agents

Nico Smit 1,*, Jana Vicanova 2, Stan Pavel 3
PMCID: PMC2801997  PMID: 20054473

Abstract

Skin whitening products are commercially available for cosmetic purposes in order to obtain a lighter skin appearance. They are also utilized for clinical treatment of pigmentary disorders such as melasma or postinflammatory hyperpigmentation. Whitening agents act at various levels of melanin production in the skin. Many of them are known as competitive inhibitors of tyrosinase, the key enzyme in melanogenesis. Others inhibit the maturation of this enzyme or the transport of pigment granules (melanosomes) from melanocytes to surrounding keratinocytes. In this review we present an overview of (natural) whitening products that may decrease skin pigmentation by their interference with the pigmentary processes.

Keywords: whitening, tyrosinase inhibitors, natural agents, cosmetics

1. Introduction

In the skin, melanocytes are situated on the basal layer which separates dermis and epidermis. One melanocyte is surrounded by approximately 36 keratinocytes. Together, they form the so-called epidermal melanin unit. The melanin produced and stored inside the melanocyte in the melanosomal compartment is transported via dendrites to the overlaying keratinocytes. The melanin pigment is a polymer produced inside the melanosomes and synthesised from the amino acid l-tyrosine that is converted by the enzyme tyrosinase to dopaquinone [1]. This reaction continues spontaneously via dopachrome to the monomeric indolic precursors (5,6-dihydroxyindole and 5,6-dihydroxyindole 2-carboxylic acid) of the black-brown pigment eumelanin. However, some other enzymes, like the tyrosinase related proteins (TRP-1 and dopachrome tautomerase (TRP-2) may also play an important role in melanogenesis in vivo. Upon reaction with cysteine, dopaquinone forms 2- or 5-S-cysteinyldopa that generates the benzothiazine precursors of the red/yellow pheomelanin polymer. In general, a mixed type of pheo- and eumelanin polymer is produced and deposited onto the melanosomal matrix proteins. Considering the many colour variations that can be seen in the skin and hair, one may expect that the composition of the mixed melanins is regulated in many different ways. However, altered production of cutaneous melanin may cause considerable problems of esthetic nature, especially in hyperpigmentary conditions, like melasma, postinflammatory hyperpigmentation, freckles or lentigines. But also depigmenting conditions, like vitiligo, have high impact on the quality of life of the patients.

In the Western culture it is still considered desirable to obtain a (bronze) tan. Despite warnings about the consequences of excessive sun or UV exposure, the artificial tanning business has expanded strongly in the last decades. In the Eastern world, however, a centuries long tradition exists whereby a light complexion is regarded as equivalent to youth and beauty. Development of preparations for bleaching hyperpigmented lesions or to safely achieve overall whitening is one of the challenges for cosmetic industry. In recent years, the interest in skin whitening has grown tremendously.

2. Targeting Tyrosinase as the Key Enzyme of Melanogenesis

One of the most obvious cellular targets for depigmenting agents is the enzyme tyrosinase. The scientific literature on tyrosinase inhibition shows that a large majority of the work has been conducted since 2000 and has mostly been devoted to the search for new depigmenting agents. Notably, many of these studies deal with tyrosinase inhibitors from natural sources and are mostly of Asian origin (see Tables 1 and 2). However, early pioneering work in the field has been performed much earlier using 4-hydroxyanisole. This compound could serve as an alternative substrate for tyrosinase causing depigmentation both in vivo and in vitro [2,3]. Since this and various other substituted phenolic compound can generate potentially toxic quinone products they were used in various studies aimed at the induction of toxicity mediated by tyrosinase in melanoma cells [4,5].

Table 1.

Compounds selected as tyrosinase inhibitors by extraction from natural sources and the (possible) isolation and characterization of the active ingredients.

Source Compounds (type) Mode of action tested* Refs.
TI comments

Chouji and Yakuchi extracts, crude drugs eugenol, yakuchinone A, ferulic acid, curcumin and yakuchinone B TI (c) [18]

Anacardium occidentale, cashew fruit 6-[8(Z),11(Z),14-pentadecatrienyl]-salicylic acid and 5-[8(Z),11(Z),14-pentadecatrienyl]resorcinol TI (c) [19]

Bolivian medicinal plants, Buddleia coriacea, Gnaphalium cheiranthifolium, and Scheelea princeps. phenolic TI [20]

Artocarpus gomezianus. among eight other compounds norartocarpetin (5) and resveratrol (8) were isolated 5,8 were most potent TI [21]

Artocarpus incisus flavonoids, stilbenes and related 4-substituted resorcinols TI 4-substituted recorcinol increases TI (c) [15]

Stryphnodendron barbatimao, Portulaca pilosa, Cariniana brasiliensis, Entada africana and Prosopis africana. Five plants out of 67 tropical plants unknown strong TI TI comparable to Morus alba as positive control [22]

Pulsatilla cernua 3,4-dihydroxycinnamic acid (1) 4-hydroxy-3-methoxycinnamic acid (2) 2 > other TI
> 1
1,2 (nc)
[23]

galls of Rhus javanica Tannic acid TI - [24]

Sophora flavescens prenylated flavonoids; kuraridin, kurarinone and norkurarinol strong TI > KA C8 and C5 substitutionis essential for TI [16]

Sophora flavescens sophoraflavanone G, kuraridin, and kurarinone TI > KA [25]

Veratrum patulum hydroxystilbene compounds; resveratrol, oxyresveratrol, and their analogs potent TI cellulase treatment improved TI [17]

Phellinus linteus cerebroside B (1), protocate-chualdehyde (2), 5-hydroxymethyl-2-furaldehyde (HMF) (3), succinic acid (4), fumaric acid (5) 2,3 TI
2 > 3
2 (c)
3 (nc)
[26]

Ecklonia stolonifera. edible brown alga out of 17 seaweed extracts phloroglucinol derivatives [phloroglucinol (1), eckstolonol (2), eckol (3), phlorofucofuroeckol A (4), and dieckol (5)]. 1,2 TI (c) 3–5 (nc) TI similar to arbutin [27]

39 seashore plant species, Japan: Hibiscus tiliaceus, Carex pumila, and Garcinia subelliptica GS contained 2 biflavonoids; 2R,3S-5, 7,4′,5″,7″,3‴,4‴-heptahydroxy-flavanone[3-8″] flavone (1) and 5,7,4′,5″,7″,3‴,4‴-heptahydroxy[3–8″] biflavanone (2) both strong TI 1 > KA [28]

Glycyrrhiza uralensis Glycyrrhiza inflate Licorice liquiritin(1), licuraside (2), isoliquiritin(3), liquiritigenin(4) and licochalcone A (5) 2,3 and 5 potent TI (c) [29]

Trifolium balansae three steroids, stigmast-5-ene-3 beta,26-diol (2), stigmast-5-ene-3-ol (3) and campesterol (4) 2,3 and 4 potent TI 2 > 3,4 [30]

Amberboa ramosa Jafri cycloartane type triterpenoids; eight compounds identified. 3β,21,22,23-tetrahydroxycycloart-24 (31),25(26)-diene (cmpd. 7) 7 most potent TI > KA SAR studies [31]

Sake lees triacylglycerols; triolein (1) and trilinolein (2) TI 1,2 (nc) TI 2 > 1 PI in E coli (2) [32]

Garcinia kola screening 21 families of medicinal plants from West and Central Africa. 5 extracts selected with G. kola showing good TI; five biflavanones identified TI > 60% IC50 > KA [33]

Marrubium velutinum and Marrubium cylleneum Screening of 45 metabolites. Flavonoids (1), phenylethanoid glycosides (2), phenolic acids (3) 1,2 moderate TI, 3 < 2 [34]

Lichen species; Graphidaceae family(1) Usnea ghattensis (2), Heterodermia podocarpa, Arthothelium awasthii (3) and Parmotrema tinctorum unknown TI (1)
30–78%
antioxidant, antimicrobial, antityrosinase IC50 (2,3) similar or less than other TIs [35,36]

Sophora flavescens sophoraflavanone G (1), kurarinone (2) and kurarinol (3) strong TI
1,2 (nc)
3 (c)
1,2 antibacterial 3 PI in SB MMS on 3 [37]

50 crude drugs Glycyrrhiza glabra, Morus alba, Syzygium aromaticum, Citrus aurantifolia, Cypreae moneta, Punica granatum and Citrus aurantium yes
All < KA
[38]

Ganoderma lucidum yes [39]

Arbutus andrachne Arbutin, hydroquinone, β-sitosterol and ursolic acid present in extracts yes [40]

Morus alba L. and Morus rotundiloba Koidz Mulberry betulinic acid (present) yes anti inflammatory [41]

Guioa villosa sesquiterpene diglycosides; crenulatosides E, F and G (1 – 3) betulin (14), lupeol (15) and soyacerebroside I (16) no
strong TI
[42]

Broussonetia kazinoki. 1,3-diphenylpropanes: kazinol C (1), D (2), F (3), broussonin C (4), kazinol S (5) and kazinol T (6) 1,3–5 (c)
4; max TI
-
-
[43]

Artocarpus heterophyllus 15 compounds. norartocarpetin (4) and artocarpesin (6) yes 5 cmpds > KA -
-
[44,45]

Paeonia suffruticosa kaempferol (I), quercetin (II), mudanpioside B (III), benzoyl-oxypaeoniflorin (IV), mudanpioside H (V), and pentagalloyl-β-d-glucose (VI) yes
I to V (c)
VI (nc)
-
-
[46]

Table 2.

New whitening agents from natural sources and their mode of action as tyrosinase inhibitor (TI), inhibitor of pigment synthesis (PI) or by other mechanisms. Azelaic acid, Kojic acid, Arbutin and Aloesin are often used as positive skin whitening agents.

Source Compounds (type) Mode of action tested(*)(**) Refs.
TI PI other
Pityrosporum ovale Azelaic acid; C9-dicarboxylic acid yes yes - [75]
Aspergillus niger and Aspergillus penicillium Kojic acid; 5-hydroxy-2-(hydroxymethyl)-γ-pyrone Yes (c,m) - - [76,77]
Arctostaphylos uva-ursi bearberry Arbutin; hydroquinone glucoside β-d-gluconopyranoside yes (c,m,nc) - - [13,78] [79]
Aloe vera Aloesin; C-glycosylated chromone yes (nc) - - [9,13,80]
Artocarpus incisus (best of) 23 heart wood species from Papua New Guinea. (+)-dihydromorin, chlorophorin, (+)-norartocarpanone, 4-prenyl-oxyresveratrol, artocarbene, artocarpesin and isoarto-carpesin yes ≈ KA yes (B16 and GP) - [81]
Morus alba Rheum undulatum 1. Oxyresveratrol
2. Hydroxystilbene
yes > KA (nc)
yes
- 1. no effect on expression or synthesis [82]
Morus alba Mulberroside F (moracin M-6, 3′-di-O-beta-d-glucopyranoside yes yes (melan-a) mild anti-oxidant SO scavenger <KA [83]
Citrus fruit peel 3′,4′,5,6,7,8-hexamethoxy-flavone (nobiletin) yes > KA antimutagenic [84]
Ramulus mori (young twigs of Morus alba) 2,3′,4,5′-tetrahydroxy-stilbene (2-oxyresveratrol) yes (c) yes (GP + UV) no effect on expression or synthesis non-toxic [85]
Glycyrrhiza glabra Licorice extract glabrene and 2′,4′,4-tri-hydroxychalcone yes yes [86]
Grape seed proanthocyanidin yes yes (B16, GP + UV) antioxidant activity, 8OHdG [56]
Aspergillus fumigatus and Saccharomyces cerevisiae melanin degrading enzymes - - [70]
Carthamus tinctorius safflower seeds 1) N-feruloylserotonin,2) N-(p-coumaroyl)serotonin, and 3) acacetin yes, 1,2 > arbutin yes (SB, B16). 1,2 > arbutin [49]
Glycyrrhiza uralensis Glycyrrhisoflavone (1) and glyasperin C(2) yes yes (B16) 1 > 2 [87]
Punica granatum Pomegranate ellagic acid yes ≈ Arb yes (GP + UV) ≈ AA [57]
Fish, Poultry vitamin B3 derivative, niacinamide no no MT inh. Mc/Kc cocult. CT [53]
Piper longum piperlonguminine no yes (B16 + msh) Tyr mRNA red. cAMP pathway via MITF inh. [88,89]
Angelica dahurica isoimperatorin imperatorin no yes (B16) Tyr protein + mRNA red. [90]
Artocarpus lakoocha heartwood oxyresveratrol yes Nd CT (female volunteers) > KA > licorice [60]
Astragalus taschkendicus askendoside B yes Nd [91]
Spatholobus suberectus Dunn (Leguminosae) Chinese herb Butin (most effective compound) yes yes (nHEM) Tyr,Trp-1 and Trp-2 reduced (WB,qPCR) [64]
Sophora japonica and Spatholobus suberectus out of 25 Chinese Herbs high phenolic content, e.g., gallic acid yes yes (nHEM) AO activity (DPPH) [92]
Galla Chinensis Radix Clematidis out of 90 Chinese Herbs unknown yes yes (Mel-ab, melan-a, melan-a/SP1 cocult.) Effects on Tyr, Trp-1 and Trp-2expression [52]
Kaempferia pandurata chalcone compounds, isopanduratin A and 4-hydroxypanduratin A yes > PTU yes (melan-a) > PTU Tyr protein reduced [93]
Corn bran Polyamine conjugates, N,N′-dicoumaroylputrescine (DCP), N-p-coumaroyl-N′-feruloylputrescine (CFP), and N,N′-diferuloyl-putrescine (DFP) yes DCP > AA yes (B16) DFP > Arb AO activity (DPPH) [94]
Podocarpus macrophyllus 2,3-dihydro-4′,4‴-di-O-methylamentoflavone yes yes (nHEM) Trp-2 mRNA reduced [95]
Longan seed corilagin, gallic acid and ellagic acid or other phenolic/flavonoid glycosides and ellagitannins yes n.d. AO activity (DPPH and ORAC assays) [71]
Gastrodia elata Blume Orchidaceae (synthetic) p-hydroxybenzyl alcohol yes (Irrev) yes (B16, mouse MC-KC cocult.) AO; radical scavenger [51,96]
Sophora flavescens 1) kurarinol, 2) kuraridinol, and 3) trifolirhizin yes 1,2 > KA 1,2 (nc) yes (B16) [97]
Cucumis sativus Lutein no yes (B16) Tyr protein reduced [98]
Lespedeza cyrtobotrya Haginin A yes (nc) yes (melan-a) GP (+UV) zebra fish MITF, Tyr, Trp-1 reduced. Erk induced [59]
Malpighia emarginata Acerola fruit cyanidin-3-alpha-O-rhamnoside. pelargonidin-3-α-O-rhamnoside yes yes (B16) GP (+UV) [11]
Coccoloba uvifera Sea grape unknown yes (nHEM) AO; reduces IL-1alpha, TNF-alpha and alpha-MSH in nHEM + UV [74]
Salicornia herbacea, halophyte yes yes (B16) AO activity [99]
Allium species such as garlic and onions. 1-propylmercaptan yes ≈ KA yes ≈ KA [100]
Willughbeia coriacea
Phyllanthus urinaria out of 14 medicinal plants Central Kalimantan
unknown yes
yes
yes (B16)
yes (B16)
AO assay DPPH [101]
Rhus Ghinensis; Chinese galls 3 Gallotannins; 2,3,4,6-tetra-O-galloyl-d-glucopyranose, 1,2,3,6-tetra-O-galloyl-β-d-glucopyranose, and 1,2,3,4,6-penta-O-galloyl-β-d-glucopyranose yes (nc) Yes (B16 + UVA; MSH) [102]
Rhus succedanea 10′(Z)-heptadecenyl-hydroquinone yes > HQ yes > HQ (B16) [103]
Polygonum cuspidatum.
Paris polyphylla
Vitex negundo
Physcion (anthraquinone + anthraquinone analog)
(+)-Lyoniresinol
yes ≈ KA
yes > KA
yes > KA
n.d.
n.d.
n.d,
Good skin permeation [10,104]
[105]
Raspberry Tiliroside yes Yes (B16) >Arb [106]
Erigeron breviscapus Chinese herb (2Z,8Z)-matricaria acid methyl ester no yes (B16, elan-a > Arb Tyr protein reduced? [107]
Alpinia galanga and Curcuma aromatica medicinal plants eugenol and curcuminoids possible active ingredients yes yes (G361 ma cells + UVA) AO defence [108]
Grape seed oligomeric proanthocyanidins - yes, nHEM + UV effects on TE, Trp-1 and Trp-2 expression AO activity [65]
*

Modes of action tested; TI; tyrosinase inhibition, (c)competitive (u) uncompetitive (nc) non-competitive and (m) mixed mode; PI; pigment inhibition, SB; Streptomyces bikiniensis, B16 or other melanoma cultures, melan-a mouse melanocytes, nHEM; normal human epidermal melanocytes, SEM; skin equivalent model, (α)-msh; (α)-melanocyte stimulating hormone, UV; ultraviolet, GP; guinea pig + msh or uv induced pigmentation; CT; tested in clinical trial.

**

Comparison of effects on tyrosinase inhibition (TI) and pigmentation inhibition (PI) are mostly done in comparison to Arbutin (Arb), Kojic acid (KA) Ascorbic Acid (AA) and phenylthiourea (PTU). Other modes of action; AO; antioxidant; TE; tyrosinase expression (mRNA), MT; melanosome transport; 8OHdg = 8 hydroxy deoxy guanosine.

Considerable interest in tyrosinase inhibitors exists also in the food industry because the activity of this enzyme is responsible for the browning of fruit and vegetables. Cysteine or ascorbic acid can be used to prevent the enzymatic browning of fruit and vegetables by binding the o-dopaquinone intermediates. More recently also 4-hexylresorcinol has been utilized for this purpose [69]. Since safety considerations are very strict in food industry, the search for new, natural tyrosinase inhibitors without negative side effects is of utmost importance in this field of research.

Work on synthetic and natural tyrosinase inhibitors has been recently reviewed in several papers [7,9,10]. The tyrosinase inhibitors can be classified as competitive, uncompetitive, mixed type and non-competitive inhibitors [10]. The nature of tyrosinase inhibition can be disclosed by measuring enzyme inhibition kinetics using Lineweaver-Burk plots with varying concentrations of l-DOPA as the substrate. This can be seen on example of polyphenol extracts from acerola (West Indian cherry) or a chalcone derivative isolated from Morus nigra (black mulberry) which has been described in recent work of Hanamura et al. and Zhang et al. [11,12]. Knowledge of the type of inhibition may be important in order to achieve better skin lightening effects since combined treatments may result in synergistic effects. This has been shown in case of the competitive tyrosinase inhibitor, arbutin and the noncompetitive inhibitor, aloesin [9,13].

A 2009 paper by Chang states that a large majority of tyrosinase inhibitors show reversible inhibition [10]. In irreversible inhibition, covalent binding with the enzyme may cause its inactivation by altering the active site of the enzyme and/or by conformational changes to the protein molecule. Irreversible inhibition may also occur via the so-called suicide inhibition mechanism as described in the model by Land et al. [14]. Also, two 8-hydroxy isoflavones isolated from soygerm koji showed suicide inhibition of tyrosinase and have been tested with promising results in an in vivo assay with 60 volunteers [10]. In Table 1 we summarize the large number of studies using tyrosinase inhibitors from natural sources that have appeared, mostly in the last decade. In many of the investigations, the active ingredients from extracts of various species have been isolated and identified. In case the mode of tyrosinase inhibition was established, a comparison with IC50 values of well known inhibitors such as kojic acid and arbutin was often made. In some of the studies specific side groups (with substitutions to C4, C5 or C8 position) of recorcinols isolated from the breadfruit (Artocarpus incisus) or from a ‘bitter root’ (Sophora flavescens) proved of great importance to their inhibitory potential [15,16]. In some cases modifications to the natural compounds were made, e.g., the deglycosylation of stilbene compounds by cellulase treatment of the Veratrum patulum extract resulted in improved tyrosinase inhibition [17]. Thus, exact knowledge on enzyme inhibition mechanisms is helpful for designing new whitening products based on targeting the key enzyme of melanogenesis, tyrosinase. Although tyrosinase plays a major role in melanin synthesis, one should realize that the regulation of skin pigmentation exists at various levels and therefore, different modes of interference are possible. There are indications that combined approaches could be more successful than targeting tyrosinase only.

TI; tyrosinase inhibition, (c) competitive mode (nc) non competitive mode of inhibition. SB; Streptomyces bikiniensis [47]. MMS; molecular modeling studies on TI. SAR; structure activity relationship. PI; pigment inhibition.

Tyrosinase inhibition among different studies is difficult to compare for several reasons (see also Chang [10]) because of different sources of tyrosinase used (see Parvez, [9]) and IC50 values that are found using either tyrosinase or l-DOPA as the substrate. In the table comparison to kojic acid (KA) for some of the component (number) is indicated as < or > or compounds are compared among each other (1 > 2).

Extraction procedures for isolation and identification are highly important for good yield of the active ingredients. Many of the papers in Table 1 describe different extraction procedures. An overview of TI from natural and synthetic sources has been presented earlier in the review by Kim and Ujama [7].

3. Different Modes of Reducing Melanin Production in Melanocytes and Skin

As proposed by Briganti et al. all depigmenting agents may be divided on the basis of interference in melanin synthesis, transport and removal by skin turnover [48]. In Table 2, we sum up a large number of studies that describe new whitening agents from natural sources with some extra information on their mode of action besides the inhibition of tyrosinase. Next to tyrosinase inhibition (TI) the extracts or their isolated active components were demonstrated to exhibit pigment inhibition (PI). For this purpose, some studies make use of the pigment-producing S. bikiniensis (SB) system [37,49] or transformed E.coli [32]. In most cases, however B16 melanoma cells are used for demonstrating PI. In addition, PI is demonstrated in the mouse melan-a or mel-ab melanocyte cultures or in normal human melanocytes (nHEM). Obviously, the use of the nHEM may better simulate the in vivo situation. On the other hand, the melanocytes are more difficult to maintain in culture. These cells, also show variations in melanin content from donor to donor and from one passage to the other [50]. Cocultures of melanocytes and keratinocytes from mouse [51,52] or human skin [53] more closely mimic the in vivo situation and, eventually, a skin equivalent model (SEM) may be the preferred in vitro system for testing skin whitening agents [54]. In this respect, recently commercially available SEM have already been applied for skin whitening studies [55]. Next to this, the brownish guinea pig (GP) model is used in several studies (Table 2) where the pigmentation is induced by either UV or α-MSH. In case of in vivo studies, prevention of the induction of pigment by the whitening agents could be demonstrated using a Minolta chromameter or by histochemical investigations showing a decrease in DOPA positive cells [56,57]. Another animal model used for whitening studies is the zebrafish that also proved useful for demonstrating the in vivo toxicity of the whitening agents [58,59]. So far, only limited numbers of clinical trials (CT) with skin whitening agents or formulations have been performed [10,60].

Preventing the maturation or intracellular trafficking of tyrosinase is an alternative way to reduce the effect of the enzyme on pigmentation [6163]. Various natural extracts can also influence tyrosinase mRNA at the transcription level; also mRNA of the other tyrosinase-related proteins or microphtalmia transcription factor (MITF) can be affected (see refs. [59,64,65] and others in Table 2). From the work of Sharlow et al. [66] and Seiberg [67] we learned that the protease activated receptor 2 (PAR-2) is important for melanosomal transfer from melanocytes to keratinocytes and that this transfer can be used as a target for skin lightening [68]. The vitamin B3 derivative niacinamide is one of the agents used for inhibiting melanosomal transfer [53]. Melanocytes express high levels of sAPP, the soluble N-terminal ectodomain of the β-amyloid precursor protein [69]. sAPP may play a role in the release of melanin particles via dendritic tips. Blocking the sAPP signalling could thus be another way to influence melanosome transport.

Mammone et al. [70] (Estee Lauder) proposed that melanin can be degraded enzymatically in keratinocytes and application of melanin degrading enzymes could be used to prevent UVB induced pigmentation in human skin.

Reduction of ROS levels in melanocytes may prevent activation of melanogenesis. In various studies, extracts from plants or fruit or other species were tested for their antioxidant capacity by using the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical-scavenging assay or the oxygen radical absorbance capacity (ORAC) (e.g., Rangkadilok et al. [71]). Fujiwara and colleagues [72] showed that daily oral administration of vitamin C (ascorbic acid) and vitamin E and cysteine to brownish guinea pigs reduced UVB-induced pigmentation. Ascorbic acid is considered a skin whitening agent and more stable derivatives such as ascorbyl glucoside and ascorbyl palmitate are already being used in different skin whitening formulations [73]. As known from many cases of post-inflammatory hyperpigmentation, melanogenesis can be stimulated by some inflammatory mediators. Inhibition of the production of inflammatory mediators (Il1α and TNF-α) was reported for sea grape extracts [74]. Via this indirect way stimulation of melanogenesis in the pigment cells could be prevented [48].

4. Induction of Pigmentation

For the development of effective skin whitening, we also need to understand processes that regulate the induction of pigmentation. Constitutive pigmentation is reflected by the phenotypes of the different skin types with varying pigmentation based on their genetic diversity. The facultative pigmentation acquired on top of the constitutive level can be obtained via different stimuli of which ultraviolet radiation (UVR) is well known as provoking the “tanning response”. An overview of the signalling pathways and intrinsic and extrinsic factors (inclusive UV) that influence melanocyte proliferation or metabolism can be found in the paper by Brenner and Hearing [109]. In brief, the UV response increases the microphtalmia-associated transcription factor (MITF) that is on its turn regulated by another transcription factor SOX9 [110]. MI is the main switch for induction of the melanogenic proteins responsible for the final increase of the melanin content in skin after UV exposure. Various pathways can be induced by the signalling through basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), stem cell factor (SCF), endothelin-1 (ET-1), adrenocorticotropic hormone and α-melanocyte stimulating hormone (ACTH and α-MSH) via their respective receptors present on melanocytes and thus stimulating their pigment production. These signalling pathways could also serve as a means of specific targeting the melanogenic pathway. In this way, the presence of melanocortin-1 receptor (MC1R) on (B16) melanoma cells has been often used for induction of pigmentation and for testing the depigmenting effects of natural skin lighteners (see examples in Table 2).

Several authors focus on factors that were not directly involved in melanin synthesis but could affect proteins indirectly connected with skin pigmentation. For instance, the endothelin-1 induction of pigmentation in melanocytes could be prevented by 3’antisense S-oligo for tyrosinase that also reduced UV induced pigmentation [111].

The Wnt/β-catenin pathway is known to play an important role in developmental processes [112]. Binding of Wnt proteins to their receptors (the frizzled family of transmembrane proteins)can be inhibited by Dikkopf 1 (DKK1), a factor secreted by fibroblasts which can suppress growth of melanocytes and strongly inhibit melanin production [109,113]. Thus, some of the natural whitening agents presented in Table 1 or 2 are not direct inhibitors of tyrosinase but downregulate expression of melanogenic proteins and in this way they may interfere with the complex regulation of melanocyte signalling cascades. Stem cell factor is a cytokine that binds to the c-kit receptor (CD117) and the activation of c-Kit leads to the activation of multiple signaling cascades, including the RAS/ERK, PI3-Kinase, Src kinase, and JAK/STAT pathways [114]. Na and coworkers [115] have used the signalling via SCF/c-kit for the evaluation of new whitening agents by high throughput screening with approximately 10.000 synthetic compounds. They found that phenyl-imidazole sulfonamide derivatives prevented stem cell factor induced c-kit phosphorylation in (501mel) human melanoma cells and also the UV induced pigmentation on brownish guinea pigs. Furthermore, the SCF/c-kit pathway was used to induce pigmentation in case of vitiligo. Geniposide (from the fruit of Gardenia jasminoides Ellis) is used in traditional Chinese medicine for treatment of generalized vitiligo. This compound was shown to increase pigmentation via SCF/c-kit in normal human melanocytes where melanogenesis was suppressed by norepinephrine [116]. In the case of SOX9 and MITF, signalling is mediated via cAMP and PKA [110], but also the stimulation of PKC (via diacylglycerol and calcium) may results in activation of tyrosinase. Inhibition of PKC by the specific PKC inhibitor bisindolylmaleimide (bis) resulted in a reduced tanning response in pigmented guinea pigs and in a marked lightening of freshly depilated hairs in mice [117].

Furthermore, Yaar et al. [118] proposed bone morphogenetic proteins (BMPs) to be involved in modulating melanogenesis since melanocytes express the BMP receptors and produce BMP-4, that is able to decrease melanin synthesis in human melanocytes in culture. Another mechanism of pigment regulation is suggested for the peroxisome proliferator- activated receptor (PPAR) since binding of octadecenedioic acid to this PPAR leads to reduced melanogenesis and tyrosinase expression. The same was found for a known pharmaceutical PPAR agonist rosiglitazone [119].

Another pathway indicated in several papers by Kim et al. is the signalling via extracellular signal-regulated kinases (ERK). This pathway can be triggered by different stimuli, like growth factors (bFGF and HGF) and cytokines (SCF), as indicated above. The authors first reported that c2-ceramide inhibits melanogenesis by activation of ERK and they showed that inhibition of ERK (and AKT/PKB) caused an increase in pigmentation in human melanocytes [120]. As a follow-up they described a new 2-imino-1,3-thiazoline derivative that decreased melanin production in B16 melanoma cells via induction of ERK [121]. More recently they showed that terrein, which acted on ERK and downregulated MITF, in combination with a new tyrosinase inhibitor, KI-063, caused additive effects on depigmentation in the Mel-ab melanocytes [122]. They also described a new imidazole derivative AVS-1357 that reduced pigmentation by activation of ERK and the downregulation of MITF and tyrosinase [123]. Similar results were achieved with haginin A that inhibited tyrosinase and also activated ERK and thus downregulated MITF and tyrosinase and TRP-1. Haginin A effectively reduced pigmentation in the brownish guinea pig and the zebrafish model systems [59].

The effects of ceramide on pigmentation is of interest as well since it has been reported that glycosylation of lipids could be of importance for proper sorting of the melanogenic proteins to the melanosomes [124]. A glycosphingolipid-deficient melanoma culture was not pigmented and by transfection with ceramide glucosyltransferase, pigmentation could be restored [124]. We found that reducing the levels of glucosylceramide may affect pigment production in normal human melanocytes. In this respect it is interesting to note that 1-deoxynojirimycin (DNJ) is a glycosidase inhibitor and one of the main components in mulberry leaves (from Morus alba) [125] and personal communication Aerts JMG, Academic Medical Center, University of Amsterdam). As shown in Tables 1 and 2 the compounds isolated from Morus alba (oxyresveratrol, mulberroside F and betulinic acid) inhibited tyrosinase [41,82,83] but the effect of DNJ on lipid glycosylation could inhibit melanin synthesis as well (N. Smit, manuscript in preparation).

5. Cosmetic Use of (natural) Agents for Skin Whitening

In cosmetic formulations hydroquinone (HQ) has been widely used as an effective whitening agent but it has been banned recently because of serious safety concerns: its use has been connected with mutagenicity and the increased incidence of ochronosis in African countries. Other compounds often used are kojic acid, arbutin and azelaic acid (see top of Table 2). Arbutin is a glycosylated form of HQ that is present in bearberry extracts but it can also be synthesized from HQ by glucosidation. A new derivative, deoxyarbutin was prepared by removal of all hydroxyl groups from the glucose side chain of arbutin and showed much lower cytotoxicity than arbutin [126,127]. In the large variety of whitening products, nowadays commercially available the use of different natural whitening agents is noticeable. Although the information on the exact formulations for all the whitening products is not easily accessible on the internet, we made an attempt to summarize the active whitening ingredients for some of them (Table 3). The utilization of kojic acid and arbutin is still common because these agents have repeatedly been demonstrated to be effective whitening agents. The use of bearberry extracts (a natural source of β-arbutin) may strengthen the effect of α-arbutin in Meladerm and Lucederm preparations. Among the natural extracts, mulberry and licorice are popular components added to the skin whiteners. The isolation of their active components and their ffect on tyrosinase inhibition (TI) and pigment reduction (PI) has been described (see Tables 1 and 2). Also lemon extract is used in the preparations like Skin Bright, Lucederm and Meladerm as a potent skin bleaching ingredient. However, it can only be used at low concentrations because it easily causes skin irritation. In Tables 1 and 2 several studies are included describing Sophora species from which several active compounds have been isolated that act as potent inhibitors of tyrosinase and pigment production. Also in the product Synerlight from LiBiol an extract from Sophora species is present. In this case it is combined with Kiwi fruit (Actinidia Chinensis) which contains flavonoids (e.g., quercetin) that may be responsible for tyrosinase inhibition [10]. Niacinamide, which besides inhibition of tyrosinase, interferes in melanosome transfer to keratinocytes is used in the formulations of Meladerm and Lucederm. The Revitol product Skin Brightener contains Lumiskin with some patented ingredient, diacetyl boldine, that influences tyrosinase at the expression level. The Mandresy extract of Bayer contains two compounds luteolin and verbascoside that do not only inhibit tyrosinase and pigment production but also influence the interaction between keratinocytes and melanocytes by reducing formation of dendrites. Some of the products (Meladerm and Tosseki whitening cream) contain a mixture of many extracts with the obvious tyrosinase inhibitors (Mulberry, Licorice, Sophora and Peonia) but also other extracts that may act as antioxidant or anti-inflammatory. One of the components of the Meladerm preparation is TegoCosmo which contains a guanidine compound that acts on tyrosinase activity. Another component is Gigawhite that contains various plant extracts from the Alps and that has been tested on 10 subjects of Asian origin. Its bleaching effects may partly be attributed to tyrosinase inhibition. The question arises whether the increasing amounts of potentially active whitening ingredients will cause additive effects or will reduce the effects of the most potent ingredients (by competitive inhibition).

Table 3.

Limited selection of whitening products available on the market with some information on active ingredients.

Company Product Ingredients Documentation; in Vitro/in Vivo Effect
Revitol Skin Brightener Arbutin, Lumiskin (diacetyl boldine), Z Whitener (new natural ingredient, unknown) + vitamins A,C and E and other natural extracts (antioxidants) Lumiskin TM: action on tyrosinase expression based on principle described by Fuller 2000 [128] http://naturalskincareformula.com/
Premium Naturals Skinbright Arbutin, Kojic Acid, Lemon Extract www.whiterskin.com/
Sisquoc Lucederm Niacinamide, α-Arbutin, Kojic Acid, Mulberry, Bearberry, Licorice, Lemon www.whiterskin.com/ [29,53,82,83]
LIBiol Synerlight Actinidia Chinensis (Kiwi) Fruit, Sophora Angustifolia Root http://www.gattefossecanada.ca/
Bayer HealthCare Mandresy extract Buddleja axillaris leaves; extract rich in orthocinnamic compounds and flavonoids, verbascoside & luteolin TI (mushroom); PI (nHEM + UV); reduces dendricity; in vivo brightening 8 volunteers (Chromameter) www.serdex-plantextracts.com: United States Patent Application 20090028969
Civant Skin care Meladerm Kojic Acid, α-Arbutin, Niacinamide, Mulberry, Bearberry, Licorice, Tego® Cosmo C250, Gigawhite, Lemon Juice, Emblica
TegoCosmo; a natural amino acid derivative that belongs to the class of guanidine compounds
Giga white:plant extracts from the Swiss alps; Malva Sylvestris, Mentha Piperita, Primula Veris, Alchemilla Vulgaris, Veronica Officinalis, Melissa Officinalis, Achillea Millefolium
niacinamide, mulberry and licorice (refs. [29,53,82,83])
www.whiterskin.com/;http://www.whiterskin.com/studies/cosmo.pdfhttp://www.whiterskin.com/studies/giga.pdf
Juju Cosmetics Tosekki whitening cream Glycyrrhetinic acid, Ginseng, Houttuynia, Yeast, Coix, Horse Chestnut, Arica, Grape Leaf, Ypericum, Ivy, Witch Hazel, Sophora Root, Mulberry Bark, Peony Root, Japanese Angelica Root, Rose Fruit and other ingredients. Glycyrrhetinc acid; Sophora Root; Peony Root; Mulberry Bark (refs. [25,29,38,46,82,83,86])
http://beautyknot.wordpress.com/2009/02/27/juju-cosmetics-tosekki-whitening-cream/
Lipotec Chromabright dimetylmethoxy chromanyl palmitate TI (mushroom + human); PI (nHEM) photoprotective; in vivo brightening 20 Asian females (Chromameter) www.lipotec.com ; Patent; http://www.maxworth.co.th/max/pdf/ES291%20Chromabriht.pdf

Some companies still use single synthetic compounds. For instance Lipotec uses dimetylmethoxy chromanyl palmitate in its product Chromabright. This exhibited lightening activity in a group of 20 Asian volunteers after 30 and 60 days. Sederma company makes use of a new mechanism of action targeting the peroxisome proliferator- activated receptor (PPAR). Their active ingredient named O.D.A. White is able to reduce tyrosinase mRNA expression [119].

Thus, approaches for skin whitening have broadened widely in the recent years. The utilization of single agents inhibiting tyrosinase is in many cases extended to the use of complex mixtures that target different mechanism like tyrosinase expression, transfer of melanosomes, antioxidant and anti-inflammatory effects.

Abbreviations

AA

Ascorbic Acid

ACTH

adrenocorticotropic hormone

AO

antioxidant

Arb

Arbutin

bFGF

basic fibroblast growth factor

BMP

bone morphogenetic proteins

cAMP

cyclic AMP

CT

clinical trials

DNJ

1-deoxynojirimycin

DPPH

1,1-diphenyl-2-picrylhydrazyl

ET-1

endothelin-1

ERK

extracellular signal-regulated kinases

GP

guinea pig

HGF

hepatocyte growth factor

HQ

hydroquinone

IC50

half maximal inhibitory concentration

Il1α

interleukin 1α

KA

kojic acid

l-DOPA

l-dihydroxyphenylalanine

MC1R

melanocortin-1 receptor

MITF

microphtalmia transcription factor

(α)-msh

(α)-melanocyte stimulating hormone

MT

melanosome transport

nHEM

normal human epidermal melanocytes

8OHdg

8 hydroxy deoxy guanosine

ORAC

oxygen radical absorbance capacity

PKA

protein kinase A

PKC

protein kinase C

PPAR

peroxisome proliferator- activated receptor

PTU

phenylthiourea

SAR

structure activity relationship

sAPP

soluble N-terminal ectodomain of the beta-amyloid precursor protein

SCF

stem cell factor

SEM

skin equivalent model

Sox

Sry-related HMG box

TE

tyrosinase expression

TI

tyrosinase inhibition

(c)

competitive mode

(nc)

non competitive mode

(m)

mixed mode of inhibition

TNF-α

tumor necrosis factor-α

TRP

tyrosinase related protein

UV

ultraviolet

UVA

ultraviolet A

UVB

ultraviolet B

UVR

ultraviolet radiation

References

  • 1.Cooksey CJ, Garratt PJ, Land EJ, Pavel S, Ramsden CA, Riley PA, Smit NP. Evidence of the indirect formation of the catecholic intermediate substrate responsible for the autoactivation kinetics of tyrosinase. J. Biol. Chem. 1997;272:26226–26235. doi: 10.1074/jbc.272.42.26226. [DOI] [PubMed] [Google Scholar]
  • 2.Riley PA. Hydroxyanisole depigmentation: In-vitro studies. J. Pathol. 1969;97:193–206. doi: 10.1002/path.1710970203. [DOI] [PubMed] [Google Scholar]
  • 3.Riley PA. Hydroxyanisole depigmentation: In-vivo studies. J. Pathol. 1969;97:185–191. doi: 10.1002/path.1710970202. [DOI] [PubMed] [Google Scholar]
  • 4.Naish-Byfield S, Cooksey CJ, Latter AM, Johnson CI, Riley PA. In vitro assessment of the structure-activity relationship of tyrosinase-dependent cytotoxicity of a series of substituted phenols. Melanoma. Res. 1991;1:273–287. doi: 10.1097/00008390-199111000-00007. [DOI] [PubMed] [Google Scholar]
  • 5.Smit NP, Peters K, Menko W, Westerhof W, Pavel S, Riley PA. Cytotoxicity of a selected series of substituted phenols towards cultured melanoma cells. Melanoma Res. 1992;2:295–304. doi: 10.1097/00008390-199212000-00002. [DOI] [PubMed] [Google Scholar]
  • 6.Friedman M. Food browning and its prevention: An overview. J. Agric. Food Chem. 1996;44:631–653. [Google Scholar]
  • 7.Kim YJ, Uyama H. Tyrosinase inhibitors from natural and synthetic sources: Structure, inhibition mechanism and perspective for the future. Cell Mol. Life Sci. 2005;62:1707–1723. doi: 10.1007/s00018-005-5054-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mcevily AJ, Iyengar R, Otwell S. Sulfite alternative prevents shrimp melanosis. Food Technol.—Chicago. 1991;45:80–86. [Google Scholar]
  • 9.Parvez S, Kang M, Chung HS, Bae H. Naturally occurring tyrosinase inhibitors: Mechanism and applications in skin health, cosmetics and agriculture industries. Phytother. Res. 2007;21:805–816. doi: 10.1002/ptr.2184. [DOI] [PubMed] [Google Scholar]
  • 10.Chang TS. An updated review of tyrosinase inhibitors. Int. J. Mol. Sci. 2009;10:2440–2475. doi: 10.3390/ijms10062440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hanamura T, Uchida E, Aoki H. Skin-lightening effect of a polyphenol extract from Acerola (Malpighia emarginata DC.) fruit on UV-induced pigmentation. Biosci. Biotechnol. Biochem. 2008;72:3211–3218. doi: 10.1271/bbb.80421. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang X, Hu X, Hou A, Wang H. Inhibitory effect of 2,4,2′,4′-tetrahydroxy-3-(3-methyl-2-butenyl)-chalcone on tyrosinase activity and melanin biosynthesis. Biol. Pharm. Bull. 2009;32:86–90. doi: 10.1248/bpb.32.86. [DOI] [PubMed] [Google Scholar]
  • 13.Jin YH, Lee SJ, Chung MH, Park JH, Park YI, Cho TH, Lee SK. Aloesin and arbutin inhibit tyrosinase activity in a synergistic manner via a different action mechanism. Arch. Pharm. Res. 1999;22:232–236. doi: 10.1007/BF02976355. [DOI] [PubMed] [Google Scholar]
  • 14.Land EJ, Ramsden CA, Riley PA, Stratford MR. Evidence consistent with the requirement of cresolase activity for suicide inactivation of tyrosinase. Tohoku J. Exp. Med. 2008;216:231–238. doi: 10.1620/tjem.216.231. [DOI] [PubMed] [Google Scholar]
  • 15.Shimizu K, Kondo R, Sakai K. Inhibition of tyrosinase by flavonoids, stilbenes and related 4-substituted resorcinols: Structure-activity investigations. Planta Med. 2000;66:11–15. doi: 10.1055/s-2000-11113. [DOI] [PubMed] [Google Scholar]
  • 16.Son JK, Park JS, Kim JA, Kim Y, Chung SR, Lee SH. Prenylated flavonoids from the roots of Sophora flavescens with tyrosinase inhibitory activity. Planta Med. 2003;69:559–561. doi: 10.1055/s-2003-40643. [DOI] [PubMed] [Google Scholar]
  • 17.Kim DH, Kim JH, Baek SH, Seo JH, Kho YH, Oh TK, Lee CH. Enhancement of tyrosinase inhibition of the extract of Veratrum patulum using cellulase. Biotechnol. Bioeng. 2004;87:849–854. doi: 10.1002/bit.20189. [DOI] [PubMed] [Google Scholar]
  • 18.Shirota S, Miyazaki K, Aiyama R, Ichioka M, Yokokura T. Tyrosinase inhibitors from crude drugs. Biol. Pharm. Bull. 1994;17:266–269. doi: 10.1248/bpb.17.266. [DOI] [PubMed] [Google Scholar]
  • 19.Kubo I, Kinst-Hori I, Yokokawa Y. Tyrosinase inhibitors from Anacardium occidentale fruits. J. Nat. Prod. 1994;57:545–551. doi: 10.1021/np50106a021. [DOI] [PubMed] [Google Scholar]
  • 20.Kubo I, Yokokawa Y, Kinst-Hori I. Tyrosinase inhibitors from Bolivian medicinal plants. J. Nat. Prod. 1995;58:739–743. doi: 10.1021/np50119a013. [DOI] [PubMed] [Google Scholar]
  • 21.Likhitwitayawuid K, Sritularak B, De-Eknamkul W. Tyrosinase inhibitors from Artocarpus gomezianus. Planta Med. 2000;66:275–277. doi: 10.1055/s-2000-8656. [DOI] [PubMed] [Google Scholar]
  • 22.Baurin N, Arnoult E, Scior T, Do QT, Bernard P. Preliminary screening of some tropical plants for anti-tyrosinase activity. J. Ethnopharmacol. 2002;82:155–158. doi: 10.1016/s0378-8741(02)00174-5. [DOI] [PubMed] [Google Scholar]
  • 23.Lee HS. Tyrosinase inhibitors of Pulsatilla cernua root-derived materials. J. Agric. Food Chem. 2002;50:1400–1403. doi: 10.1021/jf011230f. [DOI] [PubMed] [Google Scholar]
  • 24.Kubo I, Kinst-Hori I, Nihei K, Soria F, Takasaki M, Calderon JS, Cespedes CL. Tyrosinase inhibitors from galls of Rhus javanica leaves and their effects on insects. Z. Naturforsch. C. 2003;58:719–725. doi: 10.1515/znc-2003-9-1022. [DOI] [PubMed] [Google Scholar]
  • 25.Kim SJ, Son KH, Chang HW, Kang SS, Kim HP. Tyrosinase inhibitory prenylated flavonoids from Sophora flavescens. Biol. Pharm. Bull. 2003;26:1348–1350. doi: 10.1248/bpb.26.1348. [DOI] [PubMed] [Google Scholar]
  • 26.Kang HS, Choi JH, Cho WK, Park JC, Choi JS. A sphingolipid and tyrosinase inhibitors from the fruiting body of Phellinus linteus. Arch. Pharm. Res. 2004;27:742–750. doi: 10.1007/BF02980143. [DOI] [PubMed] [Google Scholar]
  • 27.Kang HS, Kim HR, Byun DS, Son BW, Nam TJ, Choi JS. Tyrosinase inhibitors isolated from the edible brown alga Ecklonia stolonifera. Arch. Pharm. Res. 2004;27:1226–1232. doi: 10.1007/BF02975886. [DOI] [PubMed] [Google Scholar]
  • 28.Masuda T, Yamashita D, Takeda Y, Yonemori S. Screening for tyrosinase inhibitors among extracts of seashore plants and identification of potent inhibitors from Garcinia subelliptica. Biosci. Biotechnol. Biochem. 2005;69:197–201. doi: 10.1271/bbb.69.197. [DOI] [PubMed] [Google Scholar]
  • 29.Fu B, Li H, Wang X, Lee FS, Cui S. Isolation and identification of flavonoids in licorice and a study of their inhibitory effects on tyrosinase. J. Agric. Food Chem. 2005;53:7408–7414. doi: 10.1021/jf051258h. [DOI] [PubMed] [Google Scholar]
  • 30.Sabudak T, Tareq Hassan KM, Iqbal CM, Oksuz S. Potent tyrosinase inhibitors from Trifolium balansae. Nat. Prod. Res. 2006;20:665–670. doi: 10.1080/14786410500196821. [DOI] [PubMed] [Google Scholar]
  • 31.Khan MT, Khan SB, Ather A. Tyrosinase inhibitory cycloartane type triterpenoids from the methanol extract of the whole plant of Amberboa ramosa Jafri and their structure-activity relationship. Bioorg. Med. Chem. 2006;14:938–943. doi: 10.1016/j.bmc.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 32.Jeon HJ, Noda M, Maruyama M, Matoba Y, Kumagai T, Sugiyama M. Identification and kinetic study of tyrosinase inhibitors found in sake lees. J. Agric. Food Chem. 2006;54:9827–9833. doi: 10.1021/jf062315p. [DOI] [PubMed] [Google Scholar]
  • 33.Okunji C, Komarnytsky S, Fear G, Poulev A, Ribnicky DM, Awachie PI, Ito Y, Raskin I. Preparative isolation and identification of tyrosinase inhibitors from the seeds of Garcinia kola by high-speed counter-current chromatography. J. Chromatogr. A. 2007;1151:45–50. doi: 10.1016/j.chroma.2007.02.085. [DOI] [PubMed] [Google Scholar]
  • 34.Karioti A, Protopappa A, Megoulas N, Skaltsa H. Identification of tyrosinase inhibitors from Marrubium velutinum and Marrubium cylleneum. Bioorg. Med. Chem. 2007;15:2708–2714. doi: 10.1016/j.bmc.2007.01.035. [DOI] [PubMed] [Google Scholar]
  • 35.Behera BC, Adawadkar B, Makhija U. Tyrosinase-inhibitory activity in some species of the lichen family Graphidaceae. J. Herb. Pharmacother. 2006;6:55–69. [PubMed] [Google Scholar]
  • 36.Behera BC, Verma N, Sonone A, Makhija U. Tissue culture of some lichens and screening of their antioxidant, antityrosinase and antibacterial properties. Phytother. Res. 2007;21:1159–1170. doi: 10.1002/ptr.2228. [DOI] [PubMed] [Google Scholar]
  • 37.Ryu YB, Westwood IM, Kang NS, Kim HY, Kim JH, Moon YH, Park KH. Kurarinol, tyrosinase inhibitor isolated from the root of Sophora flavescens. Phytomedicine. 2008;15:612–618. doi: 10.1016/j.phymed.2007.09.022. [DOI] [PubMed] [Google Scholar]
  • 38.Adhikari A, Devkota HP, Takano A, Masuda K, Nakane T, Basnet P, Skalko-Basnet N. Screening of Nepalese crude drugs traditionally used to treat hyperpigmentation: In vitro tyrosinase inhibition. Int. J. Cosmet. Sci. 2008;30:353–360. doi: 10.1111/j.1468-2494.2008.00463.x. [DOI] [PubMed] [Google Scholar]
  • 39.Chien CC, Tsai ML, Chen CC, Chang SJ, Tseng CH. Effects on tyrosinase activity by the extracts of Ganoderma lucidum and related mushrooms. Mycopathologia. 2008;166:117–120. doi: 10.1007/s11046-008-9128-x. [DOI] [PubMed] [Google Scholar]
  • 40.Issa RA, Afifi FU, Amro BI. Studying the anti-tyrosinase effect of Arbutus andrachne L. extracts. Int. J. Cosmet. Sci. 2008;30:271–276. doi: 10.1111/j.1468-2494.2008.00439.x. [DOI] [PubMed] [Google Scholar]
  • 41.Nattapong S, Omboon L. A new source of whitening agent from a Thai Mulberry plant and its betulinic acid quantitation. Nat. Prod. Res. 2008;22:727–734. doi: 10.1080/14786410601130794. [DOI] [PubMed] [Google Scholar]
  • 42.Magid AA, Voutquenne-Nazabadioko L, Bontemps G, Litaudon M, Lavaud C. Tyrosinase inhibitors and sesquiterpene diglycosides from Guioa villosa. Planta Med. 2008;74:55–60. doi: 10.1055/s-2007-993780. [DOI] [PubMed] [Google Scholar]
  • 43.Baek YS, Ryu YB, Curtis-Long MJ, Ha TJ, Rengasamy R, Yang MS, Park KH. Tyrosinase inhibitory effects of 1,3-diphenylpropanes from Broussonetia kazinoki. Bioorg. Med. Chem. 2009;17:35–41. doi: 10.1016/j.bmc.2008.11.022. [DOI] [PubMed] [Google Scholar]
  • 44.Zheng ZP, Cheng KW, To JT, Li H, Wang M. Isolation of tyrosinase inhibitors from Artocarpus heterophyllus and use of its extract as antibrowning agent. Mol. Nutr. Food Res. 2008;52:1530–1538. doi: 10.1002/mnfr.200700481. [DOI] [PubMed] [Google Scholar]
  • 45.Zheng ZP, Chen S, Wang S, Wang XC, Cheng KW, Wu JJ, Yang D, Wang M. Chemical components and tyrosinase inhibitors from the twigs of Artocarpus heterophyllus. J. Agric. Food Chem. 2009;57:6649–6655. doi: 10.1021/jf9014685. [DOI] [PubMed] [Google Scholar]
  • 46.Ding HY, Lin HC, Chang TS. Tyrosinase inhibitors isolated from the roots of Paeonia suffruticosa. J. Cosmet. Sci. 2009;60:347–352. [PubMed] [Google Scholar]
  • 47.Tomita K, Oda N, Ohbayashi M, Kamei H, Miyaki T, Oki T. A new screening method for melanin biosynthesis inhibitors using Streptomyces bikiniensis. J. Antibiot. (Tokyo) 1990;43:1601–1605. doi: 10.7164/antibiotics.43.1601. [DOI] [PubMed] [Google Scholar]
  • 48.Briganti S, Camera E, Picardo M. Chemical and instrumental approaches to treat hyperpigmentation. Pigment Cell Res. 2003;16:101–110. doi: 10.1034/j.1600-0749.2003.00029.x. [DOI] [PubMed] [Google Scholar]
  • 49.Roh JS, Han JY, Kim JH, Hwang JK. Inhibitory effects of active compounds isolated from safflower (Carthamus tinctorius L.) seeds for melanogenesis. Biol. Pharm. Bull. 2004;27:1976–1978. doi: 10.1248/bpb.27.1976. [DOI] [PubMed] [Google Scholar]
  • 50.Smit NP, Kolb RM, Lentjes EG, Noz KC, van der Meulen H, Koerten HK, Vermeer BJ, Pavel S. Variations in melanin formation by cultured melanocytes from different skin types. Arch. Dermatol. Res. 1998;290:342–349. doi: 10.1007/s004030050315. [DOI] [PubMed] [Google Scholar]
  • 51.Liu SH, Chu IM, Pan IH. Effects of hydroxybenzyl alcohols on melanogenesis in melanocyte-keratinocyte co-culture and monolayer culture of melanocytes. J. Enzyme Inhib. Med. Chem. 2008;23:526–534. doi: 10.1080/14756360701654894. [DOI] [PubMed] [Google Scholar]
  • 52.Zhong S, Wu Y, Soo-Mi A, Zhao J, Wang K, Yang S, Jae-Ho Y, Zhu X. Depigmentation of melanocytes by the treatment of extracts from traditional Chinese herbs: A cell culture assay. Biol. Pharm. Bull. 2006;29:1947–1951. doi: 10.1248/bpb.29.1947. [DOI] [PubMed] [Google Scholar]
  • 53.Greatens A, Hakozaki T, Koshoffer A, Epstein H, Schwemberger S, Babcock G, Bissett D, Takiwaki H, Arase S, Wickett RR, Boissy RE. Effective inhibition of melanosome transfer to keratinocytes by lectins and niacinamide is reversible. Exp. Dermatol. 2005;14:498–508. doi: 10.1111/j.0906-6705.2005.00309.x. [DOI] [PubMed] [Google Scholar]
  • 54.Duval C, Smit NP, Kolb AM, Regnier M, Pavel S, Schmidt R. Keratinocytes control the pheo/eumelanin ratio in cultured normal human melanocytes. Pigment Cell Res. 2002;15:440–446. doi: 10.1034/j.1600-0749.2002.02055.x. [DOI] [PubMed] [Google Scholar]
  • 55.Ni-Komatsu L, Leung JK, Williams D, Min J, Khersonsky SM, Chang YT, Orlow SJ. Triazine-based tyrosinase inhibitors identified by chemical genetic screening. Pigment Cell Res. 2005;18:447–453. doi: 10.1111/j.1600-0749.2005.00273.x. [DOI] [PubMed] [Google Scholar]
  • 56.Yamakoshi J, Otsuka F, Sano A, Tokutake S, Saito M, Kikuchi M, Kubota Y. Lightening effect on ultraviolet-induced pigmentation of guinea pig skin by oral administration of a proanthocyanidin-rich extract from grape seeds. Pigment Cell Res. 2003;16:629–638. doi: 10.1046/j.1600-0749.2003.00093.x. [DOI] [PubMed] [Google Scholar]
  • 57.Yoshimura M, Watanabe Y, Kasai K, Yamakoshi J, Koga T. Inhibitory effect of an ellagic acid-rich pomegranate extract on tyrosinase activity and ultraviolet-induced pigmentation. Biosci. Biotechnol. Biochem. 2005;69:2368–2373. doi: 10.1271/bbb.69.2368. [DOI] [PubMed] [Google Scholar]
  • 58.Choi TY, Kim JH, Ko DH, Kim CH, Hwang JS, Ahn S, Kim SY, Kim CD, Lee JH, Yoon TJ. Zebrafish as a new model for phenotype-based screening of melanogenic regulatory compounds. Pigment Cell Res. 2007;20:120–127. doi: 10.1111/j.1600-0749.2007.00365.x. [DOI] [PubMed] [Google Scholar]
  • 59.Kim JH, Baek SH, Kim DH, Choi TY, Yoon TJ, Hwang JS, Kim MR, Kwon HJ, Lee CH. Downregulation of melanin synthesis by haginin A and its application to in vivo lightening model. J. Invest Dermatol. 2008;128:1227–1235. doi: 10.1038/sj.jid.5701177. [DOI] [PubMed] [Google Scholar]
  • 60.Tengamnuay P, Pengrungruangwong K, Pheansri I, Likhitwitayawuid K. Artocarpus lakoocha heartwood extract as a novel cosmetic ingredient: Evaluation of the in vitro anti-tyrosinase and in vivo skin whitening activities. Int. J. Cosmet. Sci. 2006;28:269–276. doi: 10.1111/j.1467-2494.2006.00339.x. [DOI] [PubMed] [Google Scholar]
  • 61.Francis E, Wang N, Parag H, Halaban R, Hebert DN. Tyrosinase maturation and oligomerization in the endoplasmic reticulum require a melanocyte-specific factor. J. Biol. Chem. 2003;278:25607–25617. doi: 10.1074/jbc.M303411200. [DOI] [PubMed] [Google Scholar]
  • 62.Halaban R, Pomerantz SH, Marshall S, Lambert DT, Lerner AB. Regulation of tyrosinase in human melanocytes grown in culture. J. Cell Biol. 1983;97:480–488. doi: 10.1083/jcb.97.2.480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Petrescu SM, Petrescu AJ, Titu HN, Dwek RA, Platt FM. Inhibition of N-glycan processing in B16 melanoma cells results in inactivation of tyrosinase but does not prevent its transport to the melanosome. J. Biol. Chem. 1997;272:15796–15803. doi: 10.1074/jbc.272.25.15796. [DOI] [PubMed] [Google Scholar]
  • 64.Lee MH, Lin YP, Hsu FL, Zhan GR, Yen KY. Bioactive constituents of Spatholobus suberectus in regulating tyrosinase-related proteins and mRNA in HEMn cells. Phytochemistry. 2006;67:1262–1270. doi: 10.1016/j.phytochem.2006.05.008. [DOI] [PubMed] [Google Scholar]
  • 65.Zi SX, Ma HJ, Li Y, Liu W, Yang QQ, Zhao G, Lian S. Oligomeric proanthocyanidins from grape seeds effectively inhibit ultraviolet-induced melanogenesis of human melanocytes in vitro. Int. J. Mol. Med. 2009;23:197–204. [PubMed] [Google Scholar]
  • 66.Sharlow ER, Paine CS, Babiarz L, Eisinger M, Shapiro S, Seiberg M.The protease-activated receptor-2 upregulates keratinocyte phagocytosis J Cell Sci 2000113(Pt 17),3093–3101. [DOI] [PubMed] [Google Scholar]
  • 67.Seiberg M. Keratinocyte-melanocyte interactions during melanosome transfer. Pigment Cell Res. 2001;14:236–242. doi: 10.1034/j.1600-0749.2001.140402.x. [DOI] [PubMed] [Google Scholar]
  • 68.Seiberg M, Paine C, Sharlow E, Ndrade-Gordon P, Costanzo M, Eisinger M, Shapiro SS. Inhibition of melanosome transfer results in skin lightening. J. Invest Dermatol. 2000;115:162–167. doi: 10.1046/j.1523-1747.2000.00035.x. [DOI] [PubMed] [Google Scholar]
  • 69.Quast T, Wehner S, Kirfel G, Jaeger K, De Luca M, Herzog V. sAPP as a regulator of dendrite motility and melanin release in epidermal melanocytes and melanoma cells. FASEB J. 2003;17:1739–1741. doi: 10.1096/fj.02-1059fje. [DOI] [PubMed] [Google Scholar]
  • 70.Mammone T, Marenus K, Muizzuddin N, Maes D. Evidence and utility of melanin degrading enzymes. J. Cosmet. Sci. 2004;55:116–117. [PubMed] [Google Scholar]
  • 71.Rangkadilok N, Sitthimonchai S, Worasuttayangkurn L, Mahidol C, Ruchirawat M, Satayavivad J. Evaluation of free radical scavenging and antityrosinase activities of standardized longan fruit extract. Food Chem. Toxicol. 2007;45:328–336. doi: 10.1016/j.fct.2006.08.022. [DOI] [PubMed] [Google Scholar]
  • 72.Fujiwara Y, Sahashi Y, Aritro M, Hasegawa S, Akimoto K, Ninomiya S, Sakaguchi Y, Seyama Y. Effect of simultaneous administration of vitamin C, l-cysteine and vitamin E on the melanogenesis. Biofactors. 2004;21:415–418. doi: 10.1002/biof.552210182. [DOI] [PubMed] [Google Scholar]
  • 73.Balaguer A, Chisvert A, Salvador A. Environmentally friendly LC for the simultaneous determination of ascorbic acid and its derivatives in skin-whitening cosmetics. J. Sep. Sci. 2008;31:229–236. doi: 10.1002/jssc.200700414. [DOI] [PubMed] [Google Scholar]
  • 74.Silveira JE, Pereda MC, Eberlin S, Dieamant GC, Di Stasi LC. Effects of Coccoloba uvifera L. on UV-stimulated melanocytes. Photodermatol. Photoimmunol. Photomed. 2008;24:308–313. doi: 10.1111/j.1600-0781.2008.00382.x. [DOI] [PubMed] [Google Scholar]
  • 75.Nazzaro-Porro M, Passi S. Identification of tyrosinase inhibitors in cultures of pityrosporum. J. Invest Dermatol. 1978;71:205–208. doi: 10.1111/1523-1747.ep12547184. [DOI] [PubMed] [Google Scholar]
  • 76.Lim JT. Treatment of melasma using kojic acid in a gel containing hydroquinone and glycolic acid. Dermatol. Surg. 1999;25:282–284. doi: 10.1046/j.1524-4725.1999.08236.x. [DOI] [PubMed] [Google Scholar]
  • 77.Chen JS, Wei CI, Marshall MR. Inhibition-mechanism of kojic acid on polyphenol oxidase. J. Agric. Food Chem. 1991;39:1897–1901. [Google Scholar]
  • 78.Maeda K, Fukuda M. Arbutin: Mechanism of its depigmenting action in human melanocyte culture. J. Pharmacol. Exp. Ther. 1996;276:765–769. [PubMed] [Google Scholar]
  • 79.Funayama M, Arakawa H, Yamamoto R, Nishino T, Shin T, Murao S. Effects of alpha-arbutin and beta-arbutin on activity of tyrosinases from mushroom and mouse melanoma. Biosci. Biotechnol. Biochem. 1995;59:143–144. doi: 10.1271/bbb.59.143. [DOI] [PubMed] [Google Scholar]
  • 80.Yagi A, Kanbara T, Morinobu N. Inhibition of mushroom-tyrosinase by aloe extract. Planta Med. 1987;53:515–517. doi: 10.1055/s-2006-962798. [DOI] [PubMed] [Google Scholar]
  • 81.Shimizu K, Kondo R, Sakai K, Lee SH, Sato H. The inhibitory components from Artocarpus incisus on melanin biosynthesis. Planta Med. 1998;64:408–412. doi: 10.1055/s-2006-957470. [DOI] [PubMed] [Google Scholar]
  • 82.Kim YM, Yun J, Lee CK, Lee H, Min KR, Kim Y. Oxyresveratrol and hydroxystilbene compounds. Inhibitory effect on tyrosinase and mechanism of action. J. Biol. Chem. 2002;277:16340–16344. doi: 10.1074/jbc.M200678200. [DOI] [PubMed] [Google Scholar]
  • 83.Lee SH, Choi SY, Kim H, Hwang JS, Lee BG, Gao JJ, Kim SY. Mulberroside F isolated from the leaves of Morus alba inhibits melanin biosynthesis. Biol. Pharm. Bull. 2002;25:1045–1048. doi: 10.1248/bpb.25.1045. [DOI] [PubMed] [Google Scholar]
  • 84.Sasaki K, Yoshizaki F. Nobiletin as a tyrosinase inhibitor from the peel of Citrus fruit. Biol. Pharm. Bull. 2002;25:806–808. doi: 10.1248/bpb.25.806. [DOI] [PubMed] [Google Scholar]
  • 85.Lee KT, Lee KS, Jeong JH, Jo BK, Heo MY, Kim HP. Inhibitory effects of Ramulus mori extracts on melanogenesis. J. Cosmet. Sci. 2003;54:133–142. [PubMed] [Google Scholar]
  • 86.Nerya O, Vaya J, Musa R, Izrael S, Ben-Arie R, Tamir S. Glabrene and isoliquiritigenin as tyrosinase inhibitors from licorice roots. J. Agric. Food Chem. 2003;51:1201–1207. doi: 10.1021/jf020935u. [DOI] [PubMed] [Google Scholar]
  • 87.Kim HJ, Seo SH, Lee BG, Lee YS. Identification of tyrosinase inhibitors from Glycyrrhiza uralensis. Planta Med. 2005;71:785–787. doi: 10.1055/s-2005-871232. [DOI] [PubMed] [Google Scholar]
  • 88.Min KR, Kim KS, Ro JS, Lee SH, Kim JA, Son JK, Kim Y. Piperlonguminine from Piper longum with inhibitory effects on alpha-melanocyte-stimulating hormone-induced melanogenesis in melanoma B16 cells. Planta Med. 2004;70:1115–1118. doi: 10.1055/s-2004-835836. [DOI] [PubMed] [Google Scholar]
  • 89.Kim KS, Kim JA, Eom SY, Lee SH, Min KR, Kim Y. Inhibitory effect of piperlonguminine on melanin production in melanoma B16 cell line by downregulation of tyrosinase expression. Pigment Cell Res. 2006;19:90–98. doi: 10.1111/j.1600-0749.2005.00281.x. [DOI] [PubMed] [Google Scholar]
  • 90.Cho YH, Kim JH, Park SM, Lee BC, Pyo HB, Park HD. New cosmetic agents for skin whitening from Angelica dahurica. J. Cosmet. Sci. 2006;57:11–21. [PubMed] [Google Scholar]
  • 91.Khan MT, Choudhary MI, Atta UR, Mamedova RP, Agzamova MA, Sultankhodzhaev MN, Isaev MI. Tyrosinase inhibition studies of cycloartane and cucurbitane glycosides and their structure-activity relationships. Bioorg. Med. Chem. 2006;14:6085–6088. doi: 10.1016/j.bmc.2006.05.002. [DOI] [PubMed] [Google Scholar]
  • 92.Wang KH, Lin RD, Hsu FL, Huang YH, Chang HC, Huang CY, Lee MH. Cosmetic applications of selected traditional Chinese herbal medicines. J. Ethnopharmacol. 2006;106:353–359. doi: 10.1016/j.jep.2006.01.010. [DOI] [PubMed] [Google Scholar]
  • 93.Yoon JH, Shim JS, Cho Y, Baek NI, Lee CW, Kim HS, Hwang JK. Depigmentation of melanocytes by isopanduratin A and 4-hydroxypanduratin A isolated from Kaempferia pandurata ROXB. Biol. Pharm. Bull. 2007;30:2141–2145. doi: 10.1248/bpb.30.2141. [DOI] [PubMed] [Google Scholar]
  • 94.Choi SW, Lee SK, Kim EO, Oh JH, Yoon KS, Parris N, Hicks KB, Moreau RA. Antioxidant and antimelanogenic activities of polyamine conjugates from corn bran and related hydroxycinnamic acids. J. Agric. Food Chem. 2007;55:3920–3925. doi: 10.1021/jf0635154. [DOI] [PubMed] [Google Scholar]
  • 95.Cheng KT, Hsu FL, Chen SH, Hsieh PK, Huang HS, Lee CK, Lee MH. New constituent from Podocarpus macrophyllus var. macrophyllus shows anti-tyrosinase effect and regulates tyrosinase-related proteins and mRNA in human epidermal melanocytes. Chem. Pharm. Bull. (Tokyo) 2007;55:757–761. doi: 10.1248/cpb.55.757. [DOI] [PubMed] [Google Scholar]
  • 96.Liu SH, Pan IH, Chu IM. Inhibitory effect of p-hydroxybenzyl alcohol on tyrosinase activity and melanogenesis. Biol. Pharm. Bull. 2007;30:1135–1139. doi: 10.1248/bpb.30.1135. [DOI] [PubMed] [Google Scholar]
  • 97.Hyun SK, Lee WH, Jeong dM, Kim Y, Choi JS. Inhibitory effects of kurarinol, kuraridinol, and trifolirhizin from Sophora flavescens on tyrosinase and melanin synthesis. Biol. Pharm. Bull. 2008;31:154–158. doi: 10.1248/bpb.31.154. [DOI] [PubMed] [Google Scholar]
  • 98.Kai H, Baba M, Okuyama T. Inhibitory effect of Cucumis sativus on melanin production in melanoma B16 cells by downregulation of tyrosinase expression. Planta Med. 2008;74:1785–1788. doi: 10.1055/s-0028-1088338. [DOI] [PubMed] [Google Scholar]
  • 99.Sung JH, Park SH, Seo DH, Lee JH, Hong SW, Hong SS. Antioxidative and skin-whitening effect of an aqueous extract of Salicornia herbacea. Biosci. Biotechnol. Biochem. 2009;73:552–556. doi: 10.1271/bbb.80601. [DOI] [PubMed] [Google Scholar]
  • 100.Chu HL, Wang BS, Duh PD. Effects of selected organo-sulfur compounds on melanin formation. J. Agric. Food Chem. 2009;57:7072–7077. doi: 10.1021/jf9005824. [DOI] [PubMed] [Google Scholar]
  • 101.Arung ET, Kusuma IW, Christy EO, Shimizu K, Kondo R. Evaluation of medicinal plants from Central Kalimantan for antimelanogenesis. Nat. Med. (Tokyo) 2009;63:473–480. doi: 10.1007/s11418-009-0351-7. [DOI] [PubMed] [Google Scholar]
  • 102.Chen LG, Chang WL, Lee CJ, Lee LT, Shih CM, Wang CC. Melanogenesis inhibition by gallotannins from Chinese galls in B16 mouse melanoma cells. Biol. Pharm. Bull. 2009;32:1447–1452. doi: 10.1248/bpb.32.1447. [DOI] [PubMed] [Google Scholar]
  • 103.Chen YR, Chiou RY-Y, Lin TY, Huang CP, Tang WC, Chen ST, Lin SB. Identification of an alkylhydroquinone from rhus succedanea as an inhibitor of tyrosinase and melanogenesis. J. Agric. Food Chem. 2009;57:2200–2205. doi: 10.1021/jf802617a. [DOI] [PubMed] [Google Scholar]
  • 104.Leu YL, Hwang TL, Hu JW, Fang JY. Anthraquinones from polygonum cuspidatum as tyrosinase inhibitors for dermal use. Phytother. Res. 2008;22:552–556. doi: 10.1002/ptr.2324. [DOI] [PubMed] [Google Scholar]
  • 105.Azhar UH, Malik A, Khan MT, Anwar UH, Khan SB, Ahmad A, Choudhary MI. Tyrosinase inhibitory lignans from the methanol extract of the roots of Vitex negundo Linn. and their structure-activity relationship. Phytomedicine. 2006;13:255–260. doi: 10.1016/j.phymed.2004.09.001. [DOI] [PubMed] [Google Scholar]
  • 106.Lu YH, Chen J, Wei DZ, Wang ZT, Tao XY. Tyrosinase inhibitory effect and inhibitory mechanism of tiliroside from raspberry. J. Enzyme Inhib. Med. Chem. 2009;24:1154–1160. doi: 10.1080/14756360802694252. [DOI] [PubMed] [Google Scholar]
  • 107.Luo LH, Kim HJ, Nguyen DH, Lee HB, Lee NH, Kim EK. Depigmentation of melanocytes by (2Z,8Z)-matricaria acid methyl ester isolated from Erigeron breviscapus. Biol. Pharm. Bull. 2009;32:1091–1094. doi: 10.1248/bpb.32.1091. [DOI] [PubMed] [Google Scholar]
  • 108.Panich U, Kongtaphan K, Onkoksoong T, Jaemsak K, Phadungrakwittaya R, Thaworn A, Akarasereenont P, Wongkajornsilp A. Modulation of antioxidant defense by Alpinia galanga and Curcuma aromatica extracts correlates with their inhibition of UVA-induced melanogenesis. Cell Biol Toxicol. 2009. doi:10.1007/s10565-009-9121-2. [DOI] [PubMed]
  • 109.Brenner M, Hearing V. Modifying skin pigmentation- approaches through intrinsic biochemistry and exogenous agents. Drug Discov. Today: Dis. Mech. 2008;5:e189–e199. doi: 10.1016/j.ddmec.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Passeron T, Valencia JC, Bertolotto C, Hoashi T, Le PE, Takahashi K, Ballotti R, Hearing VJ. SOX9 is a key player in ultraviolet B-induced melanocyte differentiation and pigmentation. Proc. Natl. Acad. Sci. USA. 2007;104:13984–13989. doi: 10.1073/pnas.0705117104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhang YG, Hu QH, Wang XZ, Qi ZL, Lin XX, Fang JL, Dai CC. The regulating effect of antisense-s-oligo on TYR gene expression and melanin production of melanocytes. Zhonghua Zheng Xing Wai Ke Za Zhi. 2003;19:285–287. (in Chinese). [PubMed] [Google Scholar]
  • 112.Akiyama T. Wnt/beta-catenin signaling. Cytokine Growth Factor Rev. 2000;11:273–282. doi: 10.1016/s1359-6101(00)00011-3. [DOI] [PubMed] [Google Scholar]
  • 113.Yamaguchi Y, Brenner M, Hearing VJ. The regulation of skin pigmentation. J. Biol. Chem. 2007;282:27557–27561. doi: 10.1074/jbc.R700026200. [DOI] [PubMed] [Google Scholar]
  • 114.Ronnstrand L. Signal transduction via the stem cell factor receptor/c-Kit. Cell Mol. Life Sci. 2004;61:2535–2548. doi: 10.1007/s00018-004-4189-6. [DOI] [PubMed] [Google Scholar]
  • 115.Na YJ, Baek HS, Ahn SM, Shin HJ, Chang IS, Hwang JS. [4-t-Butylphenyl]-N-(4-imidazol-1-yl phenyl)sulfonamide (ISCK03) inhibits SCF/c-kit signaling in 501mel human melanoma cells and abolishes melanin production in mice and brownish guinea pigs. Biochem. Pharmacol. 2007;74:780–786. doi: 10.1016/j.bcp.2007.05.028. [DOI] [PubMed] [Google Scholar]
  • 116.Lan WJ, Wang HY, Lan W, Wang KY. Geniposide enhances melanogenesis by stem cell factor/c-Kit signalling in norepinephrine-exposed normal human epidermal melanocyte. Basic Clin. Pharmacol. Toxicol. 2008;103:88–93. doi: 10.1111/j.1742-7843.2008.00251.x. [DOI] [PubMed] [Google Scholar]
  • 117.Park HY, Lee J, Gonzalez S, Middelkamp-Hup MA, Kapasi S, Peterson S, Gilchrest BA. Topical application of a protein kinase C inhibitor reduces skin and hair pigmentation. J. Invest Dermatol. 2004;122:159–166. doi: 10.1046/j.0022-202X.2003.22134.x. [DOI] [PubMed] [Google Scholar]
  • 118.Yaar M, Wu C, Park HY, Panova I, Schutz G, Gilchrest BA. Bone morphogenetic protein-4, a novel modulator of melanogenesis. J. Biol. Chem. 2006;281:25307–25314. doi: 10.1074/jbc.M600580200. [DOI] [PubMed] [Google Scholar]
  • 119.Wiechers JW, Rawlings AV, Garcia C, Chesne C, Balaguer P, Nicolas JC, Corre S, Galibert MD. A new mechanism of action for skin whitening agents: Binding to the peroxisome proliferator-activated receptor. Int. J. Cosmet. Sci. 2005;27:123–132. doi: 10.1111/j.1467-2494.2004.00256.x. [DOI] [PubMed] [Google Scholar]
  • 120.Kim DS, Kim SY, Chung JH, Kim KH, Eun HC, Park KC. Delayed ERK activation by ceramide reduces melanin synthesis in human melanocytes. Cell Signal. 2002;14:779–785. doi: 10.1016/s0898-6568(02)00024-4. [DOI] [PubMed] [Google Scholar]
  • 121.Kim DS, Jeong YM, Park IK, Hahn HG, Lee HK, Kwon SB, Jeong JH, Yang SJ, Sohn UD, Park KC. A new 2-imino-1,3-thiazoline derivative, KHG22394, inhibits melanin synthesis in mouse B16 melanoma cells. Biol. Pharm. Bull. 2007;30:180–183. doi: 10.1248/bpb.30.180. [DOI] [PubMed] [Google Scholar]
  • 122.Kim DS, Lee S, Lee HK, Park SH, Ryoo IJ, Yoo ID, Kwon SB, Baek KJ, Na JI, Park KC. The hypopigmentary action of KI-063 (a new tyrosinase inhibitor) combined with terrein. J. Pharm. Pharmacol. 2008;60:343–348. doi: 10.1211/jpp.60.3.0009. [DOI] [PubMed] [Google Scholar]
  • 123.Kim DS, Lee HK, Park SH, Chae CH, Park KC. AVS-1357 inhibits melanogenesis via prolonged ERK activation. Pharmazie. 2009;64:532–537. [PubMed] [Google Scholar]
  • 124.Sprong H, Degroote S, Claessens T, van Drunen J, Oorschot V, Westerink BH, Hirabayashi Y, Klumperman J, van der SP, van Meer G. Glycosphingolipids are required for sorting melanosomal proteins in the Golgi complex. J. Cell Biol. 2001;155:369–380. doi: 10.1083/jcb.200106104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Konno K, Ono H, Nakamura M, Tateishi K, Hirayama C, Tamura Y, Hattori M, Koyama A, Kohno K. Mulberry latex rich in antidiabetic sugar-mimic alkaloids forces dieting on caterpillars. Proc. Natl. Acad. Sci. USA. 2006;103:1337–1341. doi: 10.1073/pnas.0506944103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hamed SH, Sriwiriyanont P, de Long MA, Visscher MO, Wickett RR, Boissy RE. Comparative efficacy and safety of deoxyarbutin, a new tyrosinase-inhibiting agent. J. Cosmet. Sci. 2006;57:291–308. [PubMed] [Google Scholar]
  • 127.Hu ZM, Zhou Q, Lei TC, Ding SF, Xu SZ. Effects of hydroquinone and its glucoside derivatives on melanogenesis and antioxidation: Biosafety as skin whitening agents. J. Dermatol. Sci. 2009;55:179–184. doi: 10.1016/j.jdermsci.2009.06.003. [DOI] [PubMed] [Google Scholar]
  • 128.Fuller BB, Drake MA, Spaulding DT, Chaudhry F. Downregulation of tyrosinase activity in human melanocyte cell cultures by yohimbine. J. Invest Dermatol. 2000;114:268–276. doi: 10.1046/j.1523-1747.2000.00860.x. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Molecular Sciences are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES