Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/225152725

Mechanism of skin pigmentation

Article  in  Biotechnology and Bioprocess Engineering · August 2008


DOI: 10.1007/s12257-008-0143-z

CITATIONS READS
32 1,590

3 authors, including:

Lam Do Dung Nguyen


Ho Chi Minh City University of Technology (HCMUT) Institute of Tropical Biology
1 PUBLICATION   32 CITATIONS    31 PUBLICATIONS   162 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Dung Nguyen on 30 May 2020.

The user has requested enhancement of the downloaded file.


Biotechnology and Bioprocess Engineering 2008, 13: 383-395
DOI/10.1007/s12257-008-0143-z

Mechanism of Skin Pigmentation


=
i~ã=aç=mÜìçåÖ=róÉåI=aìåÖ=eç~åÖ=kÖìóÉåI=~åÇ=bìåJhá=háãG=
Department of Biological Engineering, Inha University, Incheon 402-751, Korea =
Abstract Melanin is a pigment that plays an important role in providing coloration and protecting human skin from the harm-
ful effects of UV light radiation. Human skin color is determined by the type and amount of melanins that are syn-
thesized and deposited within the melanosomes. In addition, the transfer of these specialized membrane-bound
organelles from melanocytes to surrounding keratinocytes also plays a role in dictating human skin color. In order
to investigate the principle features of skin pigmentation, the origin, function, and production ability of melanin
should be highly understood in terms of biological and pathophysiological aspects. Furthermore, a deep under-
standing of melanin synthesis will also contribute to cosmetics and drugs development. In this review, the proc-
esses of melanin biosynthesis, such as survival, proliferation, and differentiation of melanin cells, as well as the
biological regulation of human pigmentation were described. © KSBB

hÉóïçêÇëW=ãÉä~åáåI=âÉê~íáåçÅóíÉI=êÉÖìä~íáçåI=íóêçëáå~ëÉI=ìäíê~îáçäÉí=ê~Çá~íáçå=
=
=
=
=
INTRODUCTION MELANIN PIGMENT IN HUMAN SKIN
PHYSIOLOGY
Melanin plays an important role in protecting human skin
from the harmful effects of UV sun radiation and in scaveng- Epidermal melanin has important evolutionary and
ing toxic drugs and chemicals. Visible pigmentation of the physiological implications, particularly for unclothed hu-
skin, hair, and eyes depends primarily on the different func- mans. Racial pigmentation has a high content of melanin
tions of melanocytes, a minor subset of cells that specialize content that can protect the skin from ultraviolet radiation
in the synthesis and distribution of the pigmented biopoly- (UV) − induced skin damage through its optical and chemi-
mer melanin. Melanocytes are derived from precursor cells cal filtering properties [1]. Melanin can absorb UV light,
(called melanoblasts) during embryological development, scavenge free radicals generated within the cytoplasm, and
and melanoblasts destined for the skin originate from the shield the host from various types of ionizing radiation as
neural crest. The accurate migration, distribution, and func- well as determine skin color when combine with other pig-
tion of melanoblasts/melanocytes determine the visible phe- ments. In humans, the variation in skin color occurs at the
notype of organisms ranging from simple fungi to the most function level of the epidermal melanin unit. Basically, skin
complex animal species. color is defined by the density of melanocytes, the number,
In this review, we will report the most recent findings in size, and dispersion of melanosomes transferred to epidermal
the pigmentation system of the skin. Understanding the keratinocytes, the nature of the pigment and its degradation
process of producing melanin will provide the basic steps for rate [2].
elucidating the mechanism of pigment-related diseases such
as melasma. Recently there has been a surge in the market
for pharmaceutical or functional cosmetics. This review em- CELL ORIGIN AND DEVELOPMENT OF
phasizes the mechanism of the pigmenting process and the MELANOCYTES
target genes that controlling melanin production and melano-
some transfer. Signal transduction inside melanocytes and Melanocytes are highly dendritic cells that originate in the
cytokine-mediated signaling between the mealnocyte and neural crest, which is a transient population of cells localized
keratonocyte is also reviewed in relation to melanin produc- in the dorsal portion of the closing neural tube. It derives the
tion. melanocyte lineage by expressing the Microphthalmia-
associated transcription factor (MITF). Melanoblasts origi-
*Corresponding author nate in the neural crest and migrate to the epidermis where
Tel: +82-32-860-7514 Fax: +82-32-872-4046 hair follicle bulges promote the differentiation of melano-
e-mail: ekkim@inha.ac.kr blasts into melanocytes [3-5].
384

The early signals that induce formation of the neural crest


include members of the Wnt, fibroblast growth factor (FGF),
bone morphogenetic protein (BMP) families, etc. The Wnt/β
-catenin signaling pathway is the first activated, followed by
KIT, ET3/endothelin B, and then the hepatocyte growth fac-
tor (HGF)/MET. It is also possible that simultaneous activa-
tion of multiple receptors is required for the differentiation of
the melanocyte precursor [6,7].

Wnts

Wnts are secreted glycoproteins that play a critical role in


melanocyte determination by activating the Frizzled receptor Fig. 1. Binding of the growth factor to the receptor on melano-
family. Wnt signaling also stimulates quiescent melanocyte cytes.
stem cells to enter the cell cycle for the generation of new
stem cells [3]. Wnt controls β-catenin via its influence on the
Ser/Thr kinase activity of the key enzyme glycogen synthase melanoblasts [12]. Consequently, the survival and migration
kinase 3β (GSK3β). In the absence of Wnt, β-Catenin is of neural crest-derived cells during embryogenesis depends
recruited to a multimolecular protein complex that includes upon interactions between specific receptors (C-KIT recep-
Axin, APC, and the kinase GSK-3β. β-Catenin is pho- tor − a tyrosinase kinase receptor) on the cell surface and
sphorylated by GSK-3β, and subsequently ubiquitinated and their extracellular ligands (steel factor − mast/stem cell
targeted for proteasomal degradation. If Wnt binds to its growth factor) (Fig. 1).
receptor Frizzled and potential co-receptor LRP-5/6, GSK- The C-KIT receptor is a tyrosine kinase that belongs to the
3β phosphorylation of β-Catenin is suppressed, which results platelet-derived growth factor (PDGF) receptor family. The
in the accumulation of β-Catenin that is then translocated to steel factors interact with KIT receptors on the cell mem-
the nucleus. In the nucleus, it binds to LEF/TCF transcrip- brane surface, which leads to the replacement of the phos-
tion factors, which results in the activation of Wnt target phate group on tyrosine residues, tyrosine transfer from the
genes [8]. inactive to the active form, and the control of the growth and
The survival of melanoblasts during development as well differentiation of embryonic melanoblasts [6].
as the proliferation and differentiation of the melanocyte
stem cells is most likely controlled by MITF. MITF is a Wnt HGF/MET
target gene that encodes a critical regulator of the melano-
cyte specific genes and has been shown to increase the in- During development, melanocyte viability and prolifera-
duction of melanoblast [9]. MITF binds the canonical E-box tion depends on activation of MET by its ligand HGF (hepa-
sequence CACGTG as well as the nonpalindromic sequence tocyte growth factor). Targeted ablation of the MET gene in
CACATG. Its expression is upregulated by MSH through mice caused a complete loss of melanoblasts, beginning at
cAMP signaling followed by CREB phosphorylation and the late stages of neural crest migration. In contrast, overex-
activation of the melanocyte-specific MITF promoter and pression of HGF in various tissues or specifically in kertino-
may regulate multiple pigmentation genes by upregulating cytes enhanced the ectopic localization of melanoblast and
MITF expression. The three major pigmentation enzymes increased the melanocyte population in the dermis [13,14].
tyrosinase, Trp-1, and Trp-2, all contain consensus MITF
DNA binding elements in their promoter region [8]. In addi- Endothelins (ET)/Endothelin Receptor B (EDNRB)
tion to activating the expression of p16INK4a and p21Cip1
to induce cell cycle arrest, MITF is also required for the pro- Endothelins are growth factors that originate from endo-
liferation of melanoma cells. The effects of MITF on prolif- thelial cells. Mutations in these growth factors produce sig-
eration most likely depends on the status of its post- nificant neural crest defects, including melanocyte defi-
translational modifications that will in turn dictate its ability ciency. There are three endothelin proteins, ET1, ET2, and
to regulate the transcription of different target genes [10,11]. ET3, and two endothelin receptors, EDNRA and EDNRB.
Mutation will affect many of the factors that are involved in ET3 binds to EDNRB on melanoblast ganglion cell precur-
regulating the melanocyte-specific MITF-M promoter, sors, which is essential for the survival and migration of
which can lead to reduced numbers of melanoblasts. melanoblast. Mutations in either ET3 or EDNRB produce a
substantial loss of melanocytes in human and mice [15,16].
Steel Factor/C-kit Receptor
Cadherins and Migration to the Skin
The melanoblasts have expression markers that are cha-
racteristic of the melanocyte lineage, most notably is the re- Cadherins are calcium-dependent surface receptors that
ceptor tyrosine kinase Kit (C-KIT) [9]. C-KIT plays a pivotal mediate cell adhesion, homing, and invasion into different
role in the normal growth and differentiation of embryonic layers. The expression of cadherins dynamically changes as
Biotechnol. Bioprocess Eng. 385=

neural crest cells emerge from the neural tube. Cadherin ex-
pression is temporarily upregulated just prior to entering the
epidermis and is subsequently suppressed as migration con-
tinues out of the epidermis into hair follicles. Cadherins are
also regulators of invasion or metastatic behavior in melano-
cytic neoplasms [17,18].

LOCALIZATION, STRUCTURE, AND FUNCTION


OF MELANOCYTES

Melanocytes exist in various tissues in the body. They are


localized in the skin at the dermal-epidermal interface, in the
hair matrix, in the retinal pigment epithelium of the eye, the
uveal tract, the stria vascularis of the ear, the vestibular re-
gion of the inner ear, the leptomeninges, and mucous mem-
branes. In normal skin, melanocytes are located in the epi-
dermal layer and their dendrites are projected into the epi-
dermis where they transfer melanosomes to keratinocytes [4].
Approximately every tenth cell in the basal layer is a Fig. 2. “Epidermal melanin unit” of melanocytes located in the
melanocyte [6]. One melanocyte is associated with approxi- stratum basal layer of the skin and associated with the
mately 30~40 surrounding keratinocytes through its den- surrounding keratinocytes. Melanin is located inside
drites. The entire unit is referred to as the “epidermal mela- melanocyte and keratinocytes. The accumulation and
nin unit” (Fig. 2) [19]. distribution of melanin to the basal layer will determine
The density of epidermal melanocytes varies at different the human skin color.
sites in the body. There are approximately 2,000 epidermal
melanocytes per square millimeter on the skin of the head
and forearm and approximately 1,000 on the rest of the body. such as α-Melanocyte Stimulating Hormone (α-MSH) [4,6].
Individuals will experience an apparent change in the colora-
tion of their skin through the course of their life. Almost all
infants, including black infants, are lighter at birth and be- BIOSYNTHESIS, DISTRIBUTION, AND
come darker during the first week of postnatal life. The dor- TRANSPORT OF MELANOSOMES
sal skin of the hand becomes mottled in color in old age as a
result of an age-induced decline in the number of epidermal Melanosomes are specialized members of the lysosomal
melanocytes. In addition, there is approximately an 8 to 10 lineage, which originate in the smooth endoplasmic reticu-
percent reduction in the density of melanocytes for each dec- lum. These membrane-bound granules, which are approxi-
ade of life in areas that are not exposed to sun light, except mately 200 × 900 nm, are formed within melanocytes in the
for genital sites [20]. skin and other places of the body. They are most closely
During embryogenesis, melanocytes diffuse throughout related to lysosomes, primarily because both of them contain
the dermis. They first appear in the head and neck region at similar marker proteins and are positioned within the cyto-
approximately 10 weeks of gestation. However, by the end plasm of melanocytes. The lysosome-associated membrane
of gestation, active dermal melanocytes “disappear”, except proteins (Lamp) are also present in the outer membrane of
in the head, neck, dorsal aspects of distal extremities, and melanosomes [22]. Lysosomes protect against proenzymes
the presacral area. Therefore, at this stage a portion of the such as proteinases and melanosomes protect against mela-
dermal melanocytes have clearly migrated into the epider- nin precursors (phenols, quinones) that can oxidize lipid
mis or died. membranes. The melanosome contains both matrix proteins,
The major determinant of normal human skin color is the which form scaffolds for melanin deposition, and proteins
melanogenic activity within the melanocytes and the quan- (primarily enzymes) that regulate the biosynthesis of mela-
tity and quality of pigment production, but not melanocyte nin [4,6].
density [21]. Several factors play a role in determining the During development, melanosomes acquire some factors
level of melanocyte activity, including the specific charac- and three genes-related melanogenic metalloenzyme, such as
teristics of the individual melanosomes (size, shape, type, tyrosinase, tyrosinase-related protein 1 (TRP-1), and tyrosi-
and color), baseline (constitutive) and stimulated (faculta- nase-related protein 2 (TRP-2), which become fully func-
tive) levels of activity of the enzymes involved in the tional and properly folded through glycosylation.
melanin biosynthetic pathway, the mode in which melano-
somes are transferred to the keratinocytes, and their distri- The Glycosylation of Tyrosinase
bution in the keratinocytes. The latter are influenced by
receptor-mediated interactions with extracellular ligands Tyrosinase is the rate-limiting copper-containing enzyme
386

that contains virtually all of the catalytic activity necessary


for melanosome maturation and melanin synthesis. It con-
tains a signal peptide that is 529 amino acids, a C-terminal
transmembrane domain containing a 29 amino acid cytoso-
lic tail, 7 N-linked glycosylation sites (branched structure),
three cysteine (filled circles) clusters, and two copper-
binding sites that are essential for catalytic activity and for
the cellular trafficking of tyrosinase, which facilitates
transport from the rough endoplasmic reticulum (RER)
through the Golgi complex to the melanosome. This en-
zyme is highly important enzyme because of its ability to
induce melanin biosynthesis when expressed in heterolo-
gous cellular systems, even in the absence of any other
melanocyte specific proteins [23,24].
In addition to tyrosinase, Trp-1 and Trp-2 are tyrosinase
gene family proteins that are highly glycosylated and pri-
marily N-linked. They must be correctly and fully glycosy-
lated in the ER and Golgi complex before being transported Fig. 3. The role of calnexin in melanosome biogenesis.
from the trans-Golgi network (TGN) to melanosomes [22].
Tyrosinase glycosylation is a post-translational process that
occurs within the endoplasmic reticulum (ER) and the glycoproteins are recognized by glucosyl transferase, which
Golgi apparatus and is catalyzed by oligosaccharyltrans- then targets them for ER retention [4,24].
ferase (OT), a protein complex localized in the lumen of The immature misfolded tyrosinases that remain bound to
the ER. The normal structure and function of tyrosinase calnexin and calreticulin are repaired and eventually attain
requires the attachment of sugars that are essential for the their correct three-dimensional structure. Calnexin has a spe-
correct folding [25]. This process is initiated on the cytoso- cific affinity for monoglucosylated asparagine-like oligosac-
lic surface of the ER membrane. The ER contains many charides (Glu1Man9-8GlucNAc2), which are formed from the
soluble molecular chaperone and folding enzymes that in- removal of the first and second glucoses (G) in the original
teract with tyrosinase to achieve correct folding. N-linked triglucosylated core oligosaccharide (Glu3Man9GlucNAc2)
glycans are attached to the protein in the ER and are bound by the sequential action of glucosidase I and II. Only prop-
to the polypeptide chain through an N-glycosidic bond. 2 erly folded glycoproteins are transported to TGN, from
N-acetylglucosamines and 5 mannoses are then added onto which newly synthesized glycoproteins are selectively trans-
the oligosaccharide and the oligosaccharide is transferred ported to secretory granules, such as lysosome, endosome,
to the Asn side chain in the Asn−Xaa−Thr/Ser sequence and melanosome through a M6P recognition system. By
motif by the oligosaccharyltransferase enzyme complex. virtue of a specific cargo-recognition system Lamp-1, -2, and
This process begins when the ER α-glucosidase removes 3 -3 are associated with the outer membrane of late endosomes,
glucose residues in the core glycan and 2 terminal α-1, 2 lysosomes and melanosomes, and tyrosinase and Trp-1 are
linked mannose residues. The trimming of the glycan chain transported to melanosomes and late endosomes (Fig. 3)
by ER α-mannosidase I and II to create Man9 oligoman- [22,26].
nose and Man7 glycoprotein is then folded and transported The quality control test in the ER may also involve the
to the Golgi [25]. production of the tyrosinase cofactor DOPA, which is pro-
In the Golgi apparatus, 2 mannoses from the Man7GlcNAc2 duced by the oxidation of tyrosine. DOPA can bind to and
glycans are remove by the Golgi α-mannosidase I to become stabilize nascent tyrosinase, making it competent for trans-
Man5GlcNAc2 glycans. Through N-acetyl glucosamine I port. Other proteins, such as Trp1 and Trp-2, can stabilize
(NAG I) activity, these glycans transfer 1 N-acetyl glucosa- tyrosinase in the ER and increase its activity. Therefore, the
mine to form a hybrid structure of Man5GlcNAc3 glycans. presence of melanogenic complexes is important not only for
Two mannose residues are then removed from this hybrid enzymatic optimization but also for the trafficking of pro-
structure resulting in the formation of Man3GlcNAc3 glycans. teins out of the ER [4,26].
Finally the addition of N-acetyl glucosamine, galactoses, It has been well established that the cytoplasmic domain is
fucose, and sialic acids forms the complex structure which critical for melanogenic function and for the cellular traffick-
escapes terminal glycosylation [23,24]. ing of tyrosinase. The cytoplasmic domain facilitates trans-
The newly synthesized tyrosinase polypeptide is then sub- port from the RER through the Golgi complex to the
jected to the quality control system in the ER, which moni- melanosome. Protein kinase C-β (PKC-β) is a signal trans-
tors protein maturation to ensure that defective proteins are duction enzyme that is required for the activation of tyrosi-
not transported throughout the cell. The system includes nase. PKC-β co-localizes with tyrosinase at the melanosomal
calnexin, calreticulin, which are lectin chaperones that bind membrane and activates tyrosinase by phosphorylating the
specifically to glycoproteins containing monoglucosylated serine residues on the C-terminal of the cytoplasmic domain
glycans. Finally, incorrectly folded and misassembled [27,28].
Biotechnol. Bioprocess Eng. 387=

Transport of the Tyrosinase Gene Family from the


Golgi to Melanosomes

After glycosylation, proper folding, stabilizing, and possi-


bly oligomerizing within the Golgi apparatus, tyrosinases are
packaged into COPII-coated transport vesicles that bud from
the smooth ER. The vesicles traffic through the ER-Golgi
intermediate compartment (ERGIC) and combine with ma-
trix proteins to transport tyrosinase and Tyrp1 through late
endosomes to early melanosomes.
The sorting of tyrosinase and other melanocyte-specific
proteins from the Golgi to melanosomes is determined by
recognition signals in the cytoplasmic segment of the protein,
which are typically short stretches of six amino acid residues
including a di-leucine motif [29].
There are two distinct types of sorting signals for intracel- Fig. 4. Trafficking and sorting of melanosomal proteins to
lular localization of tyrosines to melanosome, a tyrosinase- melanosomes correlate with the maturation stages of
based signal and a di-leucine motif signal in the tyrosine tail, melanosomes from I to IV. This process involves not only
which acquires the presence of Trp-1, silver (Pmel17 and gp gp100 but also the AP family. Cleavage of the amino
100) and P-protein through the binding of an adapter-like terminus plays a critical role in the maturation of the
protein complex AP3. amorphous vesicular stage I to the fibrillar stage II of
The sorting system has adapter proteins, such as clathrin, melanosome.
which is a coat protein that assembles under the presence of
two distinct but closely related adapter protein (AP) com-
plexes. Clathins bind to different sorting signals on the cyto- these proteins have a high affinity for the QPLL signal at the
plasmic tails of protein and functions at various subcellular cytoplasmic tail of the AP-3. Cytoplasmic tail with di-
locations for the transport of proteins between different or- leucine-based signals, will direct these glycoproteins to
ganelle components [30,31]. AP-1, which specifically binds melanosomes. This tail is important for the intracellular
to tyrosinase-based signals, is a distinct sorting pathwayand transport of the tyrosinase gene family proteins from the
that involves transport from the TGN to early endosomes. TGN to melanosomes [34].
Proteins that use the AP1 pathway need to subsequently use The processing of gp100, which results in the cleavage of
AP3 or another sorting mechanism to transport from early the amino terminus, plays a critical role in the maturation of
endosomes to melanosomes. AP2 is involved in sorting and the amorphous vesicular stage I melanosome to the fibrillar
transporting proteins from the plasma membrane back into stage II. The gp100 has been shown to localize in the ER,
the cell and to early endosomes. In order to use this system, AP3 vesicles, cis-Golgi, and trans-Golgi network. In addi-
proteins need to first be sorted out of the melanocyte and tion, gp100 does not transport to early and late endosomes,
then brought back in. AP-1 and AP-2 are heterotetramers but rather traffics directly to Stage I melanosomes. Except
that provide clathrins with membrane specificity at their as- for gp100, the other melanosomal proteins, such as tyrosi-
sembly by enabling them to recognize the sorting motifs in nase, are further glycosylated and processed through the
the cytoplasmic domains of trafficking membrane proteins. trans-Golgi network (TGN). From that point they can poten-
Cargo proteins, which are sorted by AP-1 and AP-2 into tially use the AP3 sorting system, which traffics proteins to
clathrin-coated vesicles, are subsequently delivered to the early or late endosomes and then to lysosomes and melano-
endocytic lysosomal system. The third AP complex (AP-3) somes (Fig. 4) [1].
is more distantly related to AP-1 and AP-2. AP3 is involved
in tyrosinase sorting, which also transports cargo from the Melanosome Maturation
TGN to the plasma membrane. From an immune perspective,
a number of melanosome proteins must have at least a tran- Melanosomes develop through a series of morphologically
sient expression on the cell surface. In additional, AP-3 does defined stages (Fig. 6) from an unpigmented (stage I) to a
not bind to the cytoplasmic tails of other non-melanosomal striated organelle enriched in melanin (stage IV). In the sub-
membrane proteins that contain di-leucine and tyrosine- sequent stages of maturation, melanosomes are relatively
based sorting signals. This finding indicates that AP-3 must nondescript membrane-bound vesicles (stage I) found in the
only transport a subset of cargo proteins, specifically the peri-nuclear area. Melanosomes under eumelanogenesis
tyrosinase gene family proteins, through the di-leucine-based (eumelanosomes) are always ellipsoidal in shape and are
motifs [32,33]. Finally, AP4 may also play a role in the traf- located in well-organized lamellae filaments, elongated fi-
ficking of melanosome-specific proteins. Sorting signals are brillar and membrane bound organelles termed a premelano-
typically found at the carboxyl termini of proteins and sev- some (stage II). Once the fibrillar matrix had been generated,
eral types are dileucine and tyrosine-based sorting signals. melanin synthesis begins and the electron dense pigment is
Tyrosinases only weakly interact with AP-1 and AP-2, but deposited on the fibrils, which become electron dense upon
388

Fig. 6. The melanin biosynthetic pathway according to Raper-


Mason [44].

Fig. 5. Electron microscopy of melanosome development during tyrosinase and Trp1 are misrouted to other sites. P-proteins,
eumelanogenesis (a~f) and pheomelanogensis (g~j) in in trafficking or sorting of melanocyte-specific proteins, exit
normal melanocytes. the Golgi apparatus and are transported to premelanosomes
[37,38].
Stage IV melanosomes, after complete opacification of the
melanin deposition (stage III). Finally in the case of darkly melanosomal content by melanin deposition, will be trans-
pigmented tissues, melanin synthesis and deposition contin- ported to dendritic tips [39].
ues until fully pigmented (stage IV). In contrast, melano-
somes under pheomelanogenesis (pheome-lanosomes) are Melanin Biosynthesis
always spherical in shape and contain only internal granular
materials at all four stages of their maturation (Fig. 5) Melanin is the major product of melanocytes and is the
[35,36]. main determinant of differences in skin color. Melanin is
Stage I melanosomes is the initial site for transport of the synthesized in two main forms: eumelanin and the pheome-
tyrosinase gene family proteins. In this stage, melanosomes lanin. The amino acid tyrosine is the starting material for the
are aspherical organelles that have an irregular structure and production of melanin in the synthesis of both the brown-
contain matrix filaments and internal membranous vesicles, black eumelanin and the yellow-red pheomelanin. The key
which are formed by the invagination of the outer limiting regulatory enzyme in this pathway is tyrosinase, which has
membrane. They likely correspond to the late-coated en- tyrosine hydroxylase, DOPA oxidase, and dihydroxyindole
dosomal multivesicular bodies found in nonmelanogenic oxidase activity, therefore, it can catalyze multiple steps in
cells. the biosynthesis of melanin (Fig. 6) [6,40,41].
At stage II, the melanosomes become elongated and form Tyrosinase controls the initial chemical reaction, an im-
ordered striations which act as templates for melanin polym- portant step in melanin biosynthesis, which is the hydrolysis
erization, which commences in stage III melanosomes. of tyrosine to L-DOPA (3,4-dihydroxy-phenylalanin). L-
Stage III melanosomes is characterized by the deposition DOPA enhances tyrosinase activity by allowing oxygen to
of electron dense material on the matrix. This stage pos- bind to the active site of tyrosinase. The oxygen-bound form
sesses functional oxidoreductases, which triggers the melan- of tyrosine uses tyrosine and DOPA as substrates. Subse-
somes to darken. Silver is a type I membrane glycoprotein quently, L-DOPA is catalytically oxidized to L-DOPAquinone
that plays a structural role in the formation of these striations (3,4-dihydroxy-phenylalanin quinone) by the met-form of the
because silver expression in nonmelanogenic cells support enzyme. L-DOPA is a reactive intermediate, which is either
the formation of striations within multivescular bodies. processed into eumelanin or pheomelanin. In the absence of
Melanin biosynthesis is dependent on proper pH homeosta- thiol compounds, DOPAquinone undergoes cyclization and is
sis, osmotic pressure, transport of the substrate into the converted DOPAchrome. TRP-2 (DOPAchrome tautomerase)
melanosome lumen and other uncharacterized activities such catalyses the tautomerization of DOPAchrome into 5, 6-
as that controlled by the OA1 protein. Proper pH and os- dihydroxyindole-2-carboxylic acid (DHICA). This step leads
motic pressure are likely to require the activity of multiple to a much slower oxidation and polymerization, which results
spanning membrane transport proteins (pink-eyed dilution in a more soluble, lower molecular weight and lighter colored
protein, P-protein, and antigen in melanoma-1/membrane- melanin known as DHICA-melanin. At the same time, the
associated transporter protein (AIM-1/MATP) that have a decarboxylation of DOPAchrome leads to the formation of
role in ionic regulation in the melanosomes and possibly indole-5,6-quinine from 5, 6-dihydroxyindole (DHI) and oxi-
even in the ER and the Golgi [8,15,52]. If the normal P- date DHI. Trp1 and tyrosinase catalyze the conversion of
protein is absent, the melanosomal structure is disrupted and DHICA to carboxylated indole-quinone for the final produc-
Biotechnol. Bioprocess Eng. 389=

microtubules in the dendrites until they are captured at the


dendrited tips (Fig. 7) [4].
This process requires protein motors, cytoskeletal tracks,
and adapters or effectors that attach the organelles to the
motors. There are two mechanisms involved in the centrifu-
gal movement of melanosomes to the microtubules and cell
periphery. Long-range movement by kinesin and dynein or
short-range movement at the cell periphery by the Rab 27a,
melanophilin, myosin Va complex [43].
The long-range movement of melanosomes proceeds to-
wards the plus ends of microtubules; therefore it may be
powered by motors of the kinesin superfamily. Kinesins are
molecular motors that are involved in microtubule and ATP
dependent transport of organelles, protein complexes and
mRNA [43,46,47]. Kinesins consist of two globular heads,
which is formed by two kinesin heavy chains (KHCs), and a
Fig. 7. The molecular machinery that is involved in melanosome fan-like end, which is formed by two kinesin light chains.
movement in melanocytes. Anterograde movement, to- The KHC possesses a motor domain, which binds to the
wards the plus end of microtubules at the cell periphery is microtubules, a-helical coiled-coil stalk domain, which is
achieved by kinesin. According to the formation of com- involved in dimer formation, and a C-terminal tail domain.
plex Rab27a, melanophilin and myosin Va. In the periph- Conventional kinesin (kinesin I) is highly expressed in
ery region of the cell, melanosomes display short-range mammalian melanocytes, and has been implicated in
movements on actin filaments. The microtubule- and ac- melanosome transport. Anti-sense oligonucleotides that in-
tin-dependent motors are coordinated to achieve the in- terfered with the synthesis of kinesin I inhibited the bi-
tracellular dispersion of melanosomes. Retrograde move- directional movement of melanosomes along microtubules
ment, towards the minus end of microtubules in the cen- and promoted perinuclear aggregation [48]. Retrograde
ter of the cell is dependent on dynein. Single black ar- movement towards the microtubules’ minusends is achieved
rows indicate direction of melanosome movement. by cytoplasmic dynein, which associates to its cargo via the
multisubunit complex dynactin. Cytoplasmic dynein was
implicated in both human and frog melanosome movement.
tion of brown-black eumelanins. DHICA-melanins have re- This microtubule-activated ATPase is a member of the
duced photoabsorption, no phototoxicity and less cytotoxicity dynein superfamily of proteins and is composed of two
[31,41-43]. heavy chains, multiple intermediate chains, light intermedi-
The formation of pheomelanin requires less tyrosinase ac- ate chains, and light chains. Another associated subunit is the
tivity than does the formation of eumelanin. They are pro- dynactin complex, which may serve in general regulatory
duced from cysteinyldopa and benzothiazine metabolites roles [46,47].
when DOPAquinone combines with cysteine or glutathione In short-range movements, melanosomes still undergo
to form the intermediate products cysteinyl-DOPA and rapid bi-directional and microtubule-dependent movements
alanyl-hydroxyl-benzothiazine. Pheomelanins have a yellow- between the center and the periphery of the cell. This mode
ish-red color, are soluble in alkali and have a low molecular of movement provides a means of transporting the melano-
weight. In contrast, pheomelanin has very little photoabsorp- somes to the cell periphery. In the cell periphery, the acto-
tion, a high phototoxic potential, and low cytotoxicity. The myosin system functions to ensure that the mature melano-
ratios of pheomelanic and eumelanic monomers determine somes are captured and stay in position for subsequent trans-
the final color of the skin and hair [44]. fer. The Rab family is involved in the transport of the vesicle
During the induction of new melanogenesis, tyrosinase carriers along microtubules and actin filaments, tethering or
and TRP-1 coordinate together to upregulate Lamp-1, which docking of vesicles to the acceptor membrane, and fusion of
is continuously coated on the inner surface of the melano- the vesicles with the membrane of the acceptor compartment.
somal membrane. Their high content of N- and O-linked In melanocytes, Rab27a associates with the cytosolic leaflet
oligosaccharides serve as a scavenger to toxic melanin in- of the melanosome membrane and appears to be the key
termediates that are produced by tyrosinase [45]. melanosome-associated protein of the tethering complex.
The association of Rab27a with melanosomes does not de-
Melanosome Transport to Dendritic Tips pend on melanophilin and myosin Va. In fact, the mecha-
nism of Rab27a − melanosome association is currently un-
Melanosomes become enriched with pigment in the cell known, however, unraveling these targeting mechanisms
body at stage IV and travel to the dendritic tip in a centrifu- will help in fully understanding this process. Melanophilin
gal manner. At the dendritic tip, melanosomes are subse- contains at least three functional domains. The N-terminal
quently transferred to neighboring keratinocytes in the epi- domain binds to Rab27a and is conserved throughout the Slp
dermis. Melanosomes move in a bi-directional manner on family. Over-expression of this domain in melanocytes in-
390

duces melanosome clustering, which serves as a Rab27a- A


binding region. The Rab27a binding domain consists of two M
stretches of amino acids referred to as the Sl phomology
domain-1 (SHD1) and the Sl phomology domain-2 SHD2.
The SHD1 is both necessary and sufficient for Rab27a spe-
cific binding, whereas the SHD2 is involved in enhancing K
the Rab27a binding affinity to melanophilin. Melanophilin
specifically interacts with the globular tail of myosin Va via B C D
a coiled coil domain within a central domain. The C-terminal
domain immunoprecipitates with actin and may be required
for peripheral melanosome distribution. Myosin Va is an M
actin-based motor that belongs to the large myosin super-
family and shares a common N-terminal motor domain that
binds to actin and generates force through the hydrolysis of
ATP. The tail domain of myosin is postulated to direct the K
interaction with the cargo, which ultimately determines the
functional specificity of the motor. Myosin Va is also found
to colocalize with F-actin in the dendrites and respective tips Fig. 8. Different modes of melanin transfer [50].
where the melanosomes normally accumulate. This suggests
that the interaction of myosin Va with cortical actin is re-
sponsible for the peripheral capture of melanosomes. Myosin will form a phagolysosome by fusion with lysosomes result-
Va, containing exons D and F, is expressed in skin and other ing in the degradation of the melanocyte membranes. The
tissues and are essential for the interaction of myosin Va phagolysosome will then be transported to the supranuclear
with melanophilin and consequently for the association of region and disintegrates into smaller vesicles, followed by
myosin Va with melanosomes [44]. An increase in micro- dispersion into the cytoplasm.
tubule based transport is observed in the absence of compo- The second mechanism is based on regulated exocytosis.
nents needed for actomyosin based transport. Myosin V con- In this process, the membranes of cytoplasmic organelles
tributes to the dispersion of melanosomes by counteracting fuse with the melanocyte plasma membrane and release
the action of dynein, particularly by shortening the length of melanin into the intercellular space. Then keratinocytes take
dynein-driven runs and by ensuring that the regular motion up the melanin by phagocytosis (Fig. 8B) [53].
of melanosomes is towards the microtubule plus-end. My- The third mechanism proposes that the presence of cog-
osin Va operates in concert with actin filaments that line the nate SNARE protein on melanocyte and keratinocyte plasma
cell periphery to move granules to dendritic tips. In the ab- membrane is responsible for the transportation to keratino-
sence of Myosin Va, the bi-directional movement of cytes. In this mechanism, SNARE proteins initiate the direct
melanosomes on microtubules is normal. However, under fusion of the two outer membranes. The resultant filopodia
these conditions the melanosomes fail to accumulate at the then extends from the dendrite tips and cell body of melano-
dendritic tips. Rab proteins are adapter proteins that are lo- cytes, adheres to the surface of neighbouring keratinocytes
calized on the melanosome membrane and are involved in and creates a channel that will support the transfer of
the regulation of intracellular vesicular transport in concert melanosomes from one cell to the other cell (Fig. 8C).
with effector molecules. They link to melanophilin and in The final mechanism postulates that the transfer of mela-
turn link to myosin Va and F-actin, thus, providing specific- nin from melanocytes to keratinocytes occurs through mem-
ity in the trafficking steps of intracellular vesicles by promot- brane vesicles (Fig. 8D). Proteins and lipids destined for
ing the binding and fusion of vesicles destined for specific transfer are concentrated in the plasma membrane and con-
locations within the cell [4,32,49]. tribute to the formation of extracellular vesicles, which sheds
the melanosomes and travels to distant cells. After this proc-
Melanosome Transfer to Keratinocytes ess is complete, the vesicles will be ingested by the keratino-
cytes through phagocytosis or fuse with the keratinocyte
Melanosomes that are enriched in pigment at the dendritic plasma membrane. Once inside the keratinocyte, melanins
tips are translocated to adjacent keratinocytes where melanin are free and ultimately determine skin color.
forms a photoprotective cap over the keratinocyte nuclei [50]. In these mechanisms, PAR-2, which has an important role
Four mechanisms of the transfer have been proposed (Fig. in phagocytosis, is a major regulator of melanosome transfer.
8) [51-54]. The first mechanism suggests that the dendrite PAR-2 is expressed in keratinocytes and enhances the
tips of the melanocytes are phagocytosed by the keratinocyte phagocytosis rate of keratinocytes, which leads to increased
in the presence of keratinocyte receptors (Fig. 8A). melanosome transfer.
In this mechanism it has been postulated that melanocytes PAR-2 can potentially affects pigment modulation
extend their dendrite to contact with a keratinocyte. Then the through keratinocyte phagocytosis (Fig. 9). UVB-induced
dendrite tip is squeezed and pinched off and the cytoplasmic PAR-2 activation could provide immediate photo-protection
reticulum is filled with melanosomes. These melansomes by the rapid transfer of available melanosomes. This would
Biotechnol. Bioprocess Eng. 391=

Fig. 9. PAR-2 activation and inhibition and the effects on kerati-


nocyte phagocytosis and melanosome transfer. The acti-
vation of PAR-2 by UV radiation and the presence of
trypsin result in an enhanced melanosome transfer, Fig. 10. Factor-induced skin pigmentation. UV irradiation, growth
which leads to pigmentation. The presence of trypsin in- factors or endothelin-1, drugs and the activation of pro-
hibitors follow by inhibiting PAR-2 activation and reduced tein kinase A by α-MSH and its receptor MC1-R as well
melanosome transfer, which leads to depigmentation. as protein kinase C by TPA and Ca2+ can induce skin
pigmentation.

increase the ability of keratinocytes to ingest melanosomes


through a process of keratinocyte-melanocyte interaction. include direct damage to DNA, apoptosis, growth arrest, and
PAR-2 activation leads to skin darkening, but inhibition of stimulation of melanogenesis. Long-term effects of UV in-
PAR-2 activation by serine protease inhibitors reduces pig- clude photoaging and photocarcinogenesis. Melanin, particu-
ment transfer and leads to depigmentation. The soybean larly eumelanin, represents the major photoprotective
trypsin inhibitor (STI) have been shown to inhibit PAR-2 mechanism of the skin. Melanin limits the extent of UV
cleavage, and reduce phagocytosis and melanosome transfer; penetration through the epidermal layers and scavenges reac-
thereby completely inhibiting UVB-induced pigmentation tive oxygen radicals that may lead to oxidative DNA damage.
[55]. The extent of UV-induced DNA damage and the incidence
Other factors, such as keratinocyte growth factor receptor of skin cancer are inversely correlated with the total melanin
(KGFR), α-MSH, Rab3, Rab27a, cadherins, lectin, ect. also content of the skin [52].
contribute to events that enhance the phagocytosis rate of When UV radiation reaches the skin, reflection, scattering,
keratinocyte, such as regulating melanosome exocytosis, and absorption takes place. UV rays penetrate into the deep
effect the docking of melanosomes at the plasma membrane, layers of the subcutaneous tissue, particularly by the corneo-
and mediate melanocytes-keratinocytes adhesion and expres- cytes. UV ray absorption is increased by the content of aro-
sion of genes associated with exocytosis. All of these proc- matic acids, such as tyrosine, tryptophan, and phenylalanine
esses are essential for the transfer of melanocytes to kerati- and urocanic acid, which forms in the keratinocyte by the
nocytes [51]. process of keratinisation. Urocanic acid is presence mainly
in the stratum corneum and is important for skin moisture
mainternance, and stimulation of stratum corneum thicken-
FACTORS THAT INFLUENCE SKIN ing and melanin synthesis by melanocytes [52].
PIGMENTATION Facultative pigmentation is often divided into immediate
pigment darkening (IPD) and delayed pigment darkening
Skin pigmentation is influenced by many factors. These (DPD). IPD is a transitory darkening of the skin, which is
factores belong to three groups: ultraviolet light (UVR), observed within seconds of UVA (320~400 nm) exposure
drugs and genetic componensts (α-melanocyte stimulating and is typically resolved within 1~3 days. IPC involves
hormone (α-MSH), agouti signal protein (ASP), basic fibro- structural changes in melanocytes and keratinocytes and a
blast growth factor (β-FGF), and endothelin-1 (ET-1)) (Fig. chemical modification of pre-existing melanin. The mecha-
10) [32,56,57]. nism for IPD include photo-oxidation of melanin, changes in
cytoskeletal distribution, translocation of melanosomes from
Role of Ultraviolet Radiation in Melanogenesis the perinuclear region to the dendrites, increased melano-
somes transfer, and changes in the melanosome distribution
Solar ultraviolet radiation (UV) is a major environmental pattern within keratinocytes [58]. DPD results from an in-
factor that dramatically alters the homeostasis of the skin by crease in the number of melanocytes in addition to an in-
affecting the survival, proliferation, and differentiation of crease in the number of melanosomes in melanocytes and
various cutaneous cell types. The effects of UV on the skin keratinocytes. DPD typically occurs within 2~3 days after
392

vation of protein kinase C (PKC). In addition, an enhance-


ment in pigment production will result from melanocytes
exposure to agents that increase intracytoplasmic levels of
cAMP such as cholera toxin, dibutyryl cAMP, and α-MSH.
The interactions between α-MSH and ASP are critical for
the switch to produce eumelanin or pheomelanin (Fig. 11)
[63,64]. α-MSH produced from UVR stimulated keratino-
cytes promotes eumelanin synthesis, whereas ASP promotes
pheomelanin synthesis. The effects of α-MSH are mediated
by the MSH receptor, which is known as the melanocortin 1
receptor (MCR-1) and is expressed at high levels in melano-
cytes. α-MSH binding to MC1-R enhances the expression of
the MSH receptor in melanocytes, activates the PKA path-
way, stimulates melanocyte proliferation and differentiation,
and results in an increased melanogenesis by the melanocyte
[11,65]. α-MSH binds to the MC1-R and this protein pair
interacts with a complex of G proteins, which uses
guanosine triphosphate (GTP) and guanosine diphosphate
(GDP) as intermediary messengers. The GTP-Gsα subunit
then activates adenylate cyclase, which leads to an increase
in production of cyclic adenosine monophosphate (cAMP)
Fig. 11. Interactions that control pigment production. α-MSH and within the melanocyte. An increase in the intracellular con-
ASP interact with MC1-R. Activity of the MC1-R is en- centration of cAMP then causes an increase in tyrosinase
hanced by binding with α-MSH, which results in eume- activity and eumelanin production. If the MC1-R receptor is
lanogenesis. The dysfunction of the MC1-R or the com- dysfunctional and fails to initiate a significant rise in the in-
bination of ASP blocking MC1-R can lead to pheome- tracellular level of cAMP, pheomelanins are produced [9,66].
lanogenesis.
Agouti Signal Protein (ASP)

UVB (290~320 nm) exposure. A major stimulant for faculta- Agouti signal protein (ASP) acts as a competitive antago-
tive pigmentation is UVR, which is the most potent stimu- nist of α-MSH for MSH-R binding. MSH-R in melanocytes
lant for growth and differentiation of melanocytes. Mito- is considered to be a control point for pigmentation. MSH-R
genic signals, in addition to UVR, have been shown to regu- is also present on other cells such as monocytes, endothelial
late melanocytic proliferation. Melanin not only functions as cells, and keratinocytes. When ASP is present, as a result of
a sunscreen to absorb UV and prevent DNA damage, but its inhibition of MC1-R function and down-regulation of the
also an antioxidant and radical scavenger, which play impor- PKA pathway, melanocytes switch into their pheomelano-
tant roles in protecting cells from such damage [32,59,60]. genic mode and express melanosomal proteins at basal levels
The activation and differentiation of melanocytes can be except for tyrosinase, which continues to be expressed at
induced directly by UVR or indirectly through their interac- very low levels [67,68].
tion with surrounding UV-irradiated keratinocytes. UVR can
increase the synthesis of β-fibroblast growth factor (β-FGF) Activation of Tyrosinase
in keratinocytes, which in turn stimulates proliferation and
melanogenesis of epidermal melanocytes [61,62]. The expression of only tyrosinase still results in a low
In addition to increased synthesis of β-FGF, exposure of level of melanin production. The sulfhydryl content of
keratinocytes to UVR results in the upregulation of other melanosomes, in the form of cysteine, is not exhausted faster
keratinocyte-derived cytokines such as endothelin-1, which than it can be transported into the melanosome and the
is a small peptide originally isolated from endothelial cells. dopaquinone is converted to cysteinyldopa only to produce
Endothelin-1 plays an important role in stimulating melano- pheomelanin. Furthermore, since the other melanosomal
cyte proliferation and melanization through the G protein- proteins are not expressed, the melanosome never matures
coupled endothelin B receptor-mediated signal transduction beyond stage I. Therefore, the melanin produced does not
pathway and can also lead to an increase in tyrosinase activ- have a physical substrate to polymerize; hence the pheome-
ity and increase in melanin production. lanosomes have splotchy irregular deposits of melanin. In
contrast, upon stimulation of differentiation by UV or α-
MSH, expression of all known melanosomal proteins is up-
THE ACTIVATION OF PROTEIN KINASE A OR C regulated and melanosomes then undergo normal maturation
to the fibrillar stage II and beyond [68]. The high levels of
Exposure of melanocytes to tetradecanol phorbol acetate tyrosinase result in an increased synthesis of melanin. In
(TPA), can lead to increased melanin formation via the acti- addition, the exhaustion of intra-melanosomal sulfhydryl
Biotechnol. Bioprocess Eng. 393=

A B C genes, such as tyrosinase, TRP-1, TRP-2, and melanosomal


matrix components are induced and synthesized. Tyrosinase
undergoes posttranslational processing and glycosylation,
which controls its activity and directs its transport to melano-
cytes. In melanocyte, tyrosinase is localized to the melan-
somes, where its functions in concert with other melanogenic
Fig. 12. Drug-induced skin pigmentation. (A) Minocyline pigmen- proteins to generate melanin. Melanosomes act as carriers of
tation, (B) flagellate pigmentation from bleomycin, (C) tyrosinase, where melanin synthesis is initiated and eventually
amiodarone photosensitivity. transported to keratinocytes. Melanin is distributed within the
epidermis and determines skin color. However, many more
important questions remain unanswered, such as how the den-
cause the dopaquinone generated to cyclize and form drite interacts with the keratinocyte membrane at the site of
dopachrome. This process results in the predominant synthe- attachment and what is the mechanism of regulation of the
sis of eumelanin. The eumelanin is then deposited on the microtubule and actin based motors. Moreover, further studies
melanosomal matrix followed by increased levels of visible should aim at a better understanding of keratinocyte phagocy-
pigmentation in the tissue. tosis, the regulation of PAR-2 expression and signaling in
keratinocytes, the natural activators of PAR-2 in skin, the ter-
Drug-induced Skin Pigmentation mination of PAR-2 signaling as well as the identification of
key molecules that are involved in the dendrite-keratinocyte
Drug-induced skin pigmentation is quite common and ac- interaction. Therefore, a further and better understanding of
counts for 10~20% of all cases of acquired hyperpigmentation. these issues will continue to provide important insights about
Pigmentation may be induced by a wide variety of drugs; the the skin pigmentation process.
most common ones include non-steroidal anti-inflammatory
drugs (NSAIDs), phenytoin, antimalarials, amiodarone, antip-
sychotic drugs, cytotoxic drugs, tetracyclines, and heavy met- Acknowledgements This work was supported by the
als (Fig. 12) [8]. Korea Science and Engineering Foundation (KOSEF) grant
Drug-induced skin pigmentation may result from in- funded by the Korea Government (MEST) R0A-2007-000-
creased melanin synthesis, increased lipofuscin synthesis or 10015-0.
cutaneous deposition of drug-related material.
Certain heavy metals, such as iron, may be deposited in
the dermis following damage to dermal vessels. If deposited Received May 19, 2008; accepted June 30, 2008
in sufficient quantities a distinctive change in skin color may
be observed without any significant increase in melanin.
Some drugs react with melanin in different ways, includ- REFERENCES
ing to form a drug-pigment complex, induce accumulation of
melanin as a non-specific post-inflammatory change in pre- 1. Slominski, A., D. J. Tobin, S. Shibahara, and J. Worts-
disposed individuals or induce pigmentation directly by ac- man (2004) The pigmentation in mammalian skin and
cumulating and reacting with other substances in the skin its hormonal regulation. Physiol. Rev. 84: 1155-1228.
[8,69]. 2. Solano, F., S. Briganti, M. Picardo, and G. Ghanem
(2006) Hypopigmenting agents: an updated review on
biological, chemical and clinical aspects. Pigment Cell
CONCLUSION AND FINAL REMARKS Res. 19: 550-571.
3. Goding, C. R. (2007) Melanocytes: the new Black. Int. J.
Over the past few years, significant advances have been Biochem. Cell Biol. 39: 275-279.
made in the understanding of the molecular mechanisms of 4. Freedberg, I. M., A. Z. Eisen, K. Wolff, K. F. Austen, L.
skin pigmentation. A better understanding of these mecha- A. Goldsmith, and S. Katz (2003) Fitzpatrick’s Derma-
nisms could result in better control of human skin color. In tology in General Medicine. 6th ed. McGraw-Hill, Co-
addition, this insight could be used to discover new depig- lumbus, USA.
menting agents and validate their efficacy and safety. This 5. Osawa, M., G. Egawa, S. S. Mak, M. Moriyama, R.
review summarizes the current understanding of pigmenta- Freter, S. Yonetani, F. Beermann, and S. I. Nishikawa
tion in mammalian skin. This is a process that is carryied out (2005) Molecular characterization of melanocyte stem
within melanosomes via a series of oxidative reactions in- cells in their niche. Development 132: 5589-5599.
volving the substrate tyrosine and melanogenic tyrosinase 6. Bolognia, J. L., J. L. Jorizzo, and R. P. Rapini (2007)
enzymes. The first event in this process is the development Dermatology. 2nd ed. Elsevier, Amsterdam, The Nether-
and migration of dendritic precursor cells, known as lands.
melanoblast, which originate in the neural crest. After migra- 7. Alhaidari, Z., T. Olivry, and J. P. Ortonne (1999)
tion, melanoblasts differentiate into melanocytes in the pres- Melanocytogenesis and melanogenesis: genetic regula-
ence of MITF, Sox, Bcl-2, CREB, etc. Then, melanogenic tion and comparative clinical diseases. Vet. Dermatol.
394

10: 3-16. 23. Helenius, A. and M. Aebi (2001) Intracellular functions


8. Anne, L. and T. John (2006) Adverse Drug Reactions. of N-linked glycans. Science 291: 2364-2369.
2nd ed. Pharmaceutical Press, London, UK. 24. Branza-Nichita, N., A. J. Petrescu, G. Negroiu, R. A.
9. Steingrímsson, E., N. G. Copeland, and N. A. Jenkins Dwek, and S. M. Petrescu (2000) N-Glycosylation proc-
(2004) Melanocytes and the microphthalmia transcrip- essing and glycoprotein folding-lessons from the tyrosi-
tion factor network. Annu. Rev. Genet. 38: 365-411. nase-related proteins. Chem. Rev. 100: 4697-4712.
10. Carreira, S., J. Goodall, I. Aksan, S. A. La Rocca, M. D. 25. Olivares, C., F. Solano, and J. C. García-Borrón (2003)
Galibert, L. Denat, L. Larue, and C. R. Goding (2005) Conformation-dependent post-translational glycosyla-
Mitf cooperates with Rb1 and activates p21Cip1 expres- tion of tyrosinase. Requirement of a specific interaction
sion to regulate cell cycle progression. Nature 433: 764- involving the CuB metal binding site. J. Biol. Chem.
769. 278: 15735-15743.
11. Haskell-Luevano, C., H. Miwa, C. Dickinson, V. J. 26. Halaban, R., E. Cheng, Y. Zhang, G. Moellmann, D.
Hruby, T. Yamada, and I. Gantz (1994) Binding and Hanlon, M. Michalak, V. Setaluri, and D. N. Hebert
cAMP studies of melanotropin peptides with the cloned (1997) Aberrant retention of tyrosinase in the endoplas-
human peripheral melanocortin receptor, hMC1R. Bio- mic reticulum mediates accelerated degradation of the
chem. Biophys. Res. Commun. 204: 1137-1142. enzyme and contributes to the dedifferentiated pheno-
12. Sulaimon, S. S. and B. E. Kitchell (2003) The biology of type of amelanotic melanoma cells. Proc. Natl. Acad.
melanocytes. Vet. Dermatol. 14: 57-65. Sci. USA 94: 6210-6215.
13. Jouneau, A., Y. Q. Yu, M. Pasdar, and L. Larue (2000) 27. Park, H. Y. and B. A. Gilchrest (1999) Signaling pathway
Plasticity of cadherin-catenin expression in the melano- mediating melanogenesis. Cell. Mol. Biol. 45: 919-930.
cyte lineage. Pigment Cell Res. 13: 260-272. 28. Choe, T., I. Park, and S. Hong (2002) Determination of
14. Kunisada, T., H. Yamazaki, T. Hirobe, S. Kamei, M. tyrosinase mRNA in melanoma by reverse transcription-
Omoteno, H. Tagaya, H. Hemmi, U. Koshimizu, T. Na- PCR and optical mirror resonance biosensor. Biotechnol.
kamura, and S. I. Hayashi (2000) Keratinocyte expres- Bioprocess Eng. 7: 212-215.
sion of transgenic hepatocyte growth factor affects 29. Setaluri, V. (2000) Sorting and targeting of melano-
melanocyte development, leading to dermal melanocy- somal membrane proteins: signals, pathways, and
tosis. Mech. Dev. 94: 67-78. mechanisms. Pigment Cell Res. 13: 128-134.
15. Pla, P. and L. Larue (2003) Involvement of endothelin 30. Raposo, G. and M. S. Marks (2002) The dark side of
receptors in normal and pathological development of lysosome-related organelles: specialization of the endo-
neural crest cells. Int. J. Dev. Biol. 47: 315-325. cytic pathway for melanosome biogenesis. Traffic 3:
16. Lahav, R., E. Dupin, L. Lecoin, C. Glavieux, D. Cham- 237-248.
peval, C. Ziller, and N. M. Le Douarin (1998) Endo- 31. Seabra, M. C., E. H. Mules, and A. N. Hume (2002) Rab
thelin 3 selectively promotes survival and proliferation GTPases, intracellular traffic and disease. Trends Mol.
of neural crest-derived glial and melanocytic precursors Med. 8: 23-30.
in vitro. Proc. Natl. Acad. Sci. USA 95: 14214-14219. 32. Hearing, V. J. (2005) Biogenesis of pigment granules: a
17. Nishimura, E. K., H. Yoshida, T. Kunisada, and S. I. sensitive way to regulate melanocyte function. J. Der-
Nishikawa (1999) Regulation of E- and P-cadherin matol. Sci. 37: 3-14.
exression correlated with melanocyte migration and di- 33. García-Borrón, J. C. and F. Solano (2002) Molecular
versification. Dev. Biol. 215: 155-166. anatomy of tyrosinase and its related proteins: beyond
18. Herlyn, M., C. Berking, G. Li, and K. Satyamoorthy the histidine-bound metal catalytic center. Pigment Cell
(2000) Lessons from melanocyte development for un- Res. 15: 162-173.
derstanding the biological events in naevus and mela- 34. Berens, W., K. Van Den Bossche, T. J. Yoon, W. West-
noma formation. Melanoma Res. 10: 303-312. broek, J. C. Valencia, C. J. Out, J. Marie Naeyaert, V. J.
19. Jimbow, K., W. C. Quevedo, Jr. T. B. Fitzpatrick, and G. Hearing, and J. Lambert (2005) Different approaches for
Szabo (1976) Some aspects of melanin biology: 1950- assaying melanosome transfer. Pigment Cell Res. 18:
1975. J. Invest. Dermatol. 67: 72-89. 370-381.
20. Szabo, G. (1967) The regional anatomy of the human 35. Slominski, A., D. J. Tobin, S. Shibahara, and J. Worts-
integument with special reference to the distribution of man (2004) Melanin pigmentation in mammalian skin
hair follicles, sweat glands and melanocytes. Philos. and its hormonal regulation. Physiol. Rev. 84: 1155-
Trans. R. Soc. Lond. B 252: 447-485. 1228.
21. Bolognia, J. L. and J. M. Pawelek (1988) Biology of 36. Kushimoto, T., J. C. Valencia, G.-E. Costin, K. Toyo-
hypopigmentation. J. Am. Acad. Dermatol. 19: 217-255. fuku, H. Watabe, K.-I. Yasumoto, F. Rouzaud, W. D.
22. Jimbow, K., P. F. Gomez, K. Toyofuku, D. Chang, S. Vieira, and V. J. Hearing (2003) The melanosome: an
Miura, H. Tsujiya, and J. S. Park (1997) Biological role ideal model to study cellular differentiation. Pigment
of tyrosinase related protein and its biosynthesis and Cell Res. 16: 237-244.
transport from TGN to stage I melanosome, late en- 37. Ancans, J., M. J. Hoogduijn, and A. J. Thody (2001)
dosome, through gene transfection study. Pigment Cell Melanosomal pH, pink locus protein and their roles in
Res. 10: 206-213. melanogenesis. J. Invest. Dermatol. 117: 158-159.
Biotechnol. Bioprocess Eng. 395=

38. Du, L. and D. E. Fisher (2002) Identification of Aim-1 as some biogenesis. Pigment Cell Res. 13: 110-117.
the underwhite mouse mutant and its transcriptional 55. Seiberg, M. (2001) Keratinocyte-melanocyte inter-
regulation by MITF. J. Biol. Chem. 277: 402-406. actions during melanosome transfer. Pigment Cell Res.
39. Zuberbühler, D. A. D. (2002) Influence of the Polyun- 14: 236-242.
saturated Fatty Acids Linoleic Acid, Arachidonic Acid, 56. Parvez, S., M. Kang, H. S. Chung, C. Cho, M. C. Hong,
α-linolenic Acid and γ-linolenic Acid on Melanogenesis M. K. Shin, and H. Bae (2006) Survey and mechanism
of B16 Mouse Melanoma Cells And Normal Human of skin depigmenting and lightening agents. Phytother.
Melanocytes. Ph.D. Thesis. University of Basel, Basel, Res. 20: 921-934.
Switzerland. 57. Villarama, C. D. and H. I. Maibach (2005) Glutathione
40. Jimenez-Cervantes, C., J. C. García-Borrón, P. Valverde, as a depigmenting agent: an overview. Int. J. Cosmet.
F. Solano, and J. A. Lozano (1993) Tyrosinase isoen- Sci. 27: 147-153.
zymes in mammalian melanocytes. 1. Biochemical 58. Chakraborty, A. K., Y. Funasaka, A. Slominski, J. Bo-
characterization of two melanosomal tyrosinases from lognia, S. Sodi, M. Ichihashi, and J. M. Pawelek (1999)
B16 mouse melanoma. Eur. J. Biochem. 217: 549-556. UV lights and MSH receptors. Ann. N. Y. Acad. Sci.
41. Petit, L. and G. E. Piérard (2003) Skin-lightening prod- 885: 100-116.
ucts revisited. Int. J. Cosmet. Sci. 25: 169-181. 59. Lee, H. J. and Y. Seo (2006) Antioxidant properties of
42. Ferguson, C. A. and S. H. Kidson (1997) The regulation Erigeron Annuus extract and its three phenolic constitu-
of tyrosinase gene transcription. Pigment Cell Res. 10: ents. Biotechnol. Bioprocess Eng. 11: 13-18.
127-138. 60. Kang, K. A., S. Chae, K. H. Lee, R. Zhang, M. S. Jung,
43. Barral, D. C. and M. C. Seabra (2004) The melanosome H. J. You, J. S. Kim, and J. W. Hyun (2005) Antioxidant
as a model to study organelle motility in mammals. effect of homogentisic acid on hydrogen peroxide in-
Pigment Cell Res. 17: 111-118. duced oxidative stress in human lung fibroblast cells.
44. Boissy, R. E., C. Sakai, H. Zhao, T. Kobayashi, and V. J. Biotechnol. Bioprocess Eng. 10: 556-563.
Hearing (1998) Human tyrosinase related protein-1 61. Bolognia, J., M. Murray, and J. Pawelek (1989) UVB-
(TRP-1) does not function as a DHICA oxidase activity induced melanogenesis may be mediated through the
in contrast to murine TRP-1. Exp. Dermatol. 7: 198-204. MSH-receptor system. J. Invest. Dermatol. 92: 651-656.
45. Jimbow, K., J. S. Park, F. Kato, K. Hirosaki, K. Toyo- 62. Halaban, R., B. S. Kwon, and S. Ghosh (1988) bFGF as
fuku, C. Hua, and T. Yamashita (2000) Assembly, tar- an autocrine growth factor for human melanomas. On-
get-signaling and intracellular transport of tyrosinase cogene Res. 3: 177-186.
gene family proteins in the initial stage of melanosome 63. Barsh, G. S. (1995) Pigmentation, pleiotropy, and ge-
biogenesis. Pigment Cell Res. 13: 222-229. netic pathways in humans and mice. Am. J. Hum. Genet.
46. Hirokawa, N. (1998) Kinesin and dynein superfamily 57: 743-747.
proteins and the mechanism of organelle transport. Sci- 64. Smith, R., E. Healy, S. Siddiqui, N. Flanagan, P. M.
ence 279: 519-526. Steijlen, I. Rosdahl, J. P. Jacques, S. Rogers, R. Turner, I.
47. Vale, R. D. (2003) The molecular motor toolbox for J. Jackson, M. A. Birch-Machin, and J. L. Rees (1998)
intracellular transport. Cell 112: 467-480. Melanocortin 1 receptor variants in an Irish population.
48. Hara, M., M. Yaar, H. R. Byers, D. Goukassian, R. E. J. Invest. Dermatol. 111: 119-122.
Fine, J. Gonsalves, and B. A. Gilchrest (2000) Kinesin 65. Rees, J. L. (2000) The melanocortin 1 receptor (MC1R):
participates in melanosomal movement along melano- more than just red hair. Pigment Cell Res. 13: 135-140.
cyte dendrites. J. Invest. Dermatol. 114: 438-443. 66. Valverde, P., E. Healy, I. Jackson, J. L. Rees, and A. J.
49. Marks, M. S. and M. C. Seabra (2001) The melano- Thody (1995) Variants of the melanocyte-stimulating
some: membrane dynamics in black and white. Nat. Rev. hormone receptor gene are associated with red hair and
Mol. Cell Biol. 2: 738-748. fair skin in humans. Nat. Genet. 11: 328-330.
50. Boissy, R. E. (2003) Melanosome transfer to and trans- 67. Furumura, M., C. Sakai, Z. Abdel-Malek, G. S. Barsh,
location in the keratinocyte. Exp. Dermatol. 12: 5-12. and V. J. Hearing (1996) The interaction of agouti signal
51. Van Den Bossche, K., J. M. Naeyaert, and J. Lambert protein and melanocyte stimulating hormone to regulate
(2006) The quest for the mechanism of melanin transfer. melanin formation in mammals. Pigment Cell Res. 9:
Traffic 7: 769-778. 191-203.
52. Stanojevic, M., Z. Stanojevic, D. Jovanovic, and M. 68. Sakai, C., M. Ollmann, T. Kobayashi, Z. Abdel-Malek, J.
Stojiljkovic (2004) Ultraviolet radiation and melano- Muller, W. D. Vieira, G. Imokawa, G. S. Barsh, and V. J.
genesis. Arch. Oncol. 12: 203-205. Hearing (1997) Modulation of murine melanocyte func-
53. Scott, G., S. Leopardi, S. Printup, and B. C. Madden tion in vitro by agouti signal protein. EMBO J. 16: 3544-
(2002) Filopodia are conduits for melanosome transfer 3552.
to keratinocytes. J. Cell Sci. 115: 1441-1451. 69. Housseau, F., A. Moorthy, D. A. Langer, P. F. Robbins,
54. Jimbow, K., C. Hua, P. F. Gomez, K. Hirosaki, K. Shi- M. I. Gonzales, and S. L. Topalian (2001) N-linked car-
noda, T. G. Salopek, H. Matsusaka, H. Y. Jin, and T. bohydrates in tyrosinase are required for its recognition
Yamashita (2000) Intracellular vesicular trafficking of by human MHC class II-restricted CD4+ T cells. Eur. J.
tyrosinase gene family protein in eu- and pheomelano- Immunol. 31: 2690-2701.

View publication stats

You might also like