You are on page 1of 16

Cent. Eur. J. Biol.

• 8(4) • 2013 • 315-330


DOI: 10.2478/s11535-013-0141-1

Central European Journal of Biology

Heterogeneity of neural crest-derived


melanocytes
Review Article

Miroslawa Cichorek*, Malgorzata Wachulska, Aneta Stasiewicz

Department of Embryology,
Medical University of Gdansk,
80-210 Gdansk, Poland
Received 05 July 2012; Accepted 27 December 2012

Abstract: The majority of melanocytes originate from the neural crest cells (NCC) that migrate, spread on the whole embryo’s body
to form elements of the nervous system and skeleton, endocrinal glands, muscles and melanocytes. Human melanocytes
differentiate mainly from the cranial and trunk NCC. Although melanocyte development has traditionally been associated with the
dorsally migrating trunk NCC, there is evidence that a part of melanocytes arise from cells migrating ventrally. The ventral NCC
differentiate into neurons and glia of the ganglia or Schwann cells. It has been suggested that the precursors for Schwann cells
differentiate into melanocytes. As melanoblasts travel through the dermis, they multiply, follow the process of differentiation and
invade the forming human fetal epidermis up to third month. After birth, melanocytes lose the ability to proliferate, except the
hair melanocytes that renew during the hair cycle. The localization of neural crest-derived melanocytes in non-cutaneous places
e.g. eye (the choroid and stroma of the iris and the ciliary body), ear (cells of the vestibular organ, cochlear stria vascularis),
meninges of the brain, heart seems to indicate that repertoire of melanocyte functions is much wider than we expected e.g. the
protection of tissues from potentially harmful factors (e.g. free radicals, binding toxins), storage ions, and anti-inflammatory action.

Keywords: Neural crest cells • Melanocytes origin • Skin melanocytes • Ocular melanocytes • Ear melanocytes • Epidermal melanin unit
© Versita Sp. z o.o.

tube during neurulation (formation of the neural tube,


1. Introduction primordium for central nervous system) [6,7]. Neural
The melanocyte biology has fascinated scientists of crest cells (NCC) leave the epithelium of the neural
different disciplines for years, especially according to tube, migrate to many specific places in the body, where
the basic function of these cells – production of the they differentiate into a remarkable variety of cells as
skin, hair and eye pigment [1-3]. Nowadays these cells neurons, glial cells, endocrine cells, chondroblasts,
seemed to play many other functions in the organism. osteoblasts, smooth muscles and melanocytes
The cells broadly classified as melanocytes (cells [7-10]. Migration occurs along the well-defined
able to synthesize melanins) form a heterogeneous pathways which determines in part what type of cells
group of cells referring to their embryonic origin and NCC will form [8,10].
anatomical location. There are attempts to group Melanocytes differentiate from all NCC populations
pigment-producing cells into classical (cutaneous) but the molecular pathways of development are well
and non-classical (non-cutaneous) melanocytes [4,5]. characterized only for the trunk NCC [8,11,12]. Except
The embryonic origin of these two groups is different, the skin and eye melanocytes are present in ear,
cutaneous melanocytes develop from the neural crest nervous system and probably other places e.g. heart
cells (NCC) while non-cutaneous from NCC and neural [5]. The remarkable feature of the melanocytes is
tube cells (e.g. retinal pigment epithelium, RPE). the melanin production. Melanin protects cells in the
This review will focus on a group of melanocytes epidermis against sun-induced damage by ultraviolet
differentiating from the NCC – an embryonic population radiation (UVR) but what is/are its function/s in other
that develops from the dorsal part of the forming neural place e.g. ear, meninges is not clear [11,12].

* E-mail: cichorek@gumed.edu.pl
315
Heterogeneity of neural crest-derived melanocytes

Apart from being the melanin factories, melanocytes 2. The origin and development of
are immunocompetent, immunomodulatory and melanocytes
secretory cells [13-15]. They produce and release
neurotransmitters (acetylocholine, catecholamines), 2.1 Neural crest – embryonic structure of the
hormones (adrenocorticotropin hormone, ACTH; melanocyte origin precursors
melanocyte stimulating hormone, α-MSH) and many Precursors for melanocytes develop from the neural
cytokines [16-21]. Melanocytes as cells presenting crest [6]. The basic biology of the neural crest
antigens and secreting cytokines for lymphocytes, are development is very similar among model organisms
active parts of the immune response in the skin [13]. but small differences do exist. Specifically, we know very
They cooperate with neighboring cells as epidermal little about the molecular development of neural crest
keratinocytes, dermal fibroblasts and work in an during the early human embryogenesis [7,9]. Neural
organized regulatory network for the maintenance of crest originates from neuroectoderm, developing from
epidermal homeostasis [22]. How this process looks ectoderm under inductive influence of the notochord,
like in other than epidermis places e.g. stria vascularis during the neurulation process (Figure 1). Differentiation
cells of the cochlear duct secreting endolymph or of the neuroectoderm gives neural tube (future central
cardiomyocytes, is completely not understood. nervous system elements) and neural crest cells, so
Melanocytes in the inner ear are necessary for the named because of their site of origin, the crest of the
hearing process although the mechanism is not well closing neural tube formed from two lateral strips of the
defined [23]. Melanocyte presence, not only in the neural plate (Figure 1) [7,8]. NCC change the morphology
epidermis and hair but also in the nervous system and from epithelial to mesenchyme and as the result they get
in some organs, emphasizes the alternative functions of ability to movement at a 4-week old human embryo [7,9].
these cells that are still poorly understood [24,25]. The They migrate out from neuroepithelium and locate above
developing knowledge of the melanocytes biology give the neural tube underneath surface ectoderm. NCC
ability to understanding the melanoma development, are initially multipotent but gradually become lineage–
human pigmentation disorders as vitiligo, greying of restricted in developmental potential (Figure 1) [10].
hair or even problems with hearing. In this review, the This potential is determined by anatomical localization
current knowledge about human melanocytes of the along the anterior-posterior axis. Traditionally there
neural crest-origin and their heterogeneity is described. are distinguishable four NCC populations: cranial,

Figure 1. Differentiation of the ectoderm into surface ectoderm (future epidermis) and neuroectoderm (neural tube and neural crest cells) at
4-weeks human embryo. The neuroectodermal cells proliferate, form neural plate that folds and changes into neural groove (A). The
neural groove fuses from the middle part into both ends and the neural tube forms (B). During this neurulation process, cells from edges
of the neural groove separate from neural tube as independent population of embryonic cells - neural crest cells (NCC), located above
the neural tube (future brain and spinal cord) and beneath surface ectoderm (future epidermis) (B).

316
M. Cichorek et al.

trunk, vagal and sacral, cardiac. Multipotency of NCC on the extracellular matrix molecules and cells they
is well known e.g. cranial NCC form most of cartilages meet on the migratory pathways [7-9].
and bones of the face and head, whereas trunk NCC The dorsolaterally migrating cells follow a path between
form elements of the peripheral nervous system, ectoderm and somites and become melanocytes
medulla of the adrenal glands. These cells proliferate, (Figure 2). The ventrally migrating cells follow a way
and start to express distinct molecular markers. Each between the neural tube and somites giving rise to the
NCC population activates different molecular pathways peripheral nervous system and some endocrine cells
that produce glial cells, neurons, mesenchyme cells, e.g. adrenal medulla (Figure 2) [7,9]. Which of these
melanocytes [9,11,12]. There are conflicting results of two NCC populations migrate first in human is not
experiments concerning the problem – at what stage clear, in mouse first migrate dorsal NCC but in chick
NCC get features of melanoblasts – precursors to ventral NCC [8]. Although melanocyte development
melanocytes [8,9,12] is not clear. has traditionally been associated with this dorsal
way of NCC migration, there is evidence that a part
2.2 NCC groups for melanoblast development of melanocytes arise from cells migrating ventrally
NCC of all populations can give rise to melanocytes and then along the nerves that terminate in the skin
although the trunk NCC give most of melanocytes [26-28].
in the skin. In the region of the future embryo’s head Recent studies pointed that Schwann cells, forming
melanocytes come from the cranial NCC population myelin sheath around peripheral nerves, when cultured
that biology is different from others. Head NCC with melanocyte medium develop into precursors able
together with head mesoderm form the mesenchyme to give glial cells and melanocytes [29]. NCC migrating
(ectomesenchyme), which gives the skull, muscles, ventrally either differentiate into neurons and glial cells
dermis of the head [7]. The cranial NCC begin the of the spinal and autonomic ganglia or Schwann cells.
migration before the closure of the cranial neuropore, It has been suggested that a population of NCC cells,
while in the spinal cord of the neural tube (future trunk), nerve sheath stem cells (precursors for Schwann cells),
NCC detach after the neural groove fusion. There are could differentiate into melanocytes of the epidermis
two basic ways of the trunk NCC migration dorsally [28,30]. The recent finding that in human fetuses
or ventrally away from the neural tube (Figure 2). The the prenatal nevi develop from dermal not epidermis
different ways of NCC migration and their fate depend melanocytes supports this thesis [30].These findings

Figure 2. The basic pathways of the trunk neural crest cell (NCC) migration during early embryonic time. Dorsal cells (1) migrate between the
surface ectoderm and somites, finally develop mainly into melanocytes of the epidermis and hair. Ventral cells (2) migrate between
neural tube and somites, give elements of the peripheral nervous system, medulla of the adrenal glands and melanocytes of the skin.

317
Heterogeneity of neural crest-derived melanocytes

suggested that the precursors for Schwann cells arrive is secreted by dermal and epidermal cells during
near epidermis along cutaneous nerves, may respond melanoblast migration from the region of the neural
to factors secreted by epidermal cells and differentiate tube through the dermis into epidermis. SCF is a factor
into melanocytes [28,30]. The control of melanocyte required for survival of melanoblasts [35,38]. Except
functions by the cutaneous nervous system has been for the MAPK-signaling pathway, the Notch-signaling
poorly understood although communication between the and Wnt-signaling are essential for melanocyte
nervous system and epidermal melanocytes has been development [38,39]. Like ephrins, Notch receptor
shown [31]. The increasing evidence that epidermal requires the contact with another cell because the
melanocytes molecularly differ from dermal melanocytes ligand for this receptor is a plasma membrane protein
might reflects dual origin of the skin melanocytes [4]. (Delta protein, Jagged, Serrate). After ligand binding
intracellular domain of Notch receptor is cleaved by
2.3 Melanoblast differentiation into protease and migrates to nucleus where regulates
melanocytes transcription factors activity [39]. Studies in several
As melanoblasts travel through the dermis, they multiply model organisms suggest that components of the
and follow the process of differentiation. These cells Wnt/Frizzled protein/β-catenin-signaling pathway are
finally except the skin (epidermis, hair follicles) reside required for induction of melanocyte fate [40]. The
the eye (choroid, iris) and some internal sites, such ephrin and endothelin receptors allow melanoblasts to
as meninges, the inner ear (stria vascularis), mucous migrate along extracellular matrix containing ephrins
membranes (oral cavity) [29]. What do we know about and endothelins secreted by embryo’s cells [8].
human melanoblast migration into all these places? Spaces between tissue layers and cells are filled
The only knowledge about this process comes from the with a rich extracellular matrix (ECM) formed by many
epidermal melanoblasts. During the human development fibrillar proteins as collagens, fibronectin. Cell adhere to
melanoblast migration into embryo’s surface ectoderm one another or ECM using adhesion molecules such as
takes place at a 6-8-weeks old embryo and after the cadherins and integrins. These elements also participate
additional month most of these cells are located in the in the melanocyte development [41,42].
epidermis and hair follicles [30,32,33]. Thus after this Most of the above mentioned main melanocytic
time majority of melanoblasts has been suggested to differentiation elements induce expression of the
enter the skin. However, some authors do not exclude MITF, a protein functioning as a master of melanocyte
the possibility that a part of melanoblasts might stay in proliferation, differentiation and survival. MITF induces
the dermis [33,34]. expression of many genes that products are necessary
Epidermal melanoblasts proliferate, differentiate for melanocytes e.g. antiapoptotic BCL-2 protein
together with the fetus growth when the surface of the necessary for melanocytes survival, tyrosinase main
epidermis increases but the proliferation of epidermal enzyme for melanin production [35,43].
melanocytes has not yet been fully characterized. The The main melanocyte differentiation process is the
basic molecular elements of melanocyte differentiation creation of melanogenesis pathways. As the result
are: of this process, in the melanocyte cytoplasm new
- special receptors in the plasma membrane as ephrin organelles called melanosomes (dark vesicles) appear
receptors (EphR), endothelin receptors (especially and a cell forms many dendritic processes, gets a
EdnRB2), tyrosine kinase receptor KIT (c-Kit), receptor star-like shape. Melanogenesis is a cytotoxic process
for the beta catenins (Frizzled receptor), and as a way of defense against the death possibility
- transcription factors as SOX10, PAX3 and MITF melanocyte increases the level of antiapoptotic protein
(Microphthalmia-associated transcription factor) BCL-2 [44,45].
- melanogenic enzymes e.g. tyrosinase-related Melanocyte is a highly differentiated cell and as
protein 2 (TRP2), tyrosinase (TYR) [9,12,35]. such loose the ability to proliferate during the maturation
The most important growth factors regulating process. In the adult skin, the only renewing melanocyte
melanoblasts development from the NCC are groups are the hair melanocytes which cyclically, as hair
Wnt proteins, stem cell factor (SCF, c-Kit ligand), are lost, rebuilt the number [46]. Epidermal melanocytes
endothelins, ephrins, bone morphogenetic proteins are a very stable population which proliferate extremely
(BMP) [35-38]. Ephrins and SCF bind to receptors rare at the physiological conditions [46-48]. One
that have a cytoplasmic tyrosine kinase domain and known reason for this longevity is the resistance of
activate the MAPK- signaling pathway (Ras/Raf/ melanocytes to apoptosis, particularly resulting from a
MEK/ERK). Early melanoblast development requires high constitutive level of antiapoptotic BCL-2 [44]. It is
the presence of the SCF and its receptor c-Kit. SCF consider that in terminally differentiated cells the cell

318
M. Cichorek et al.

cycle is inhibited by several mechanisms: increased level but also for photoprotection (UV light absorption),
of cyclin dependent kinase inhibitors (p16, p27, p21), reactive oxygen species (ROS) neutralization and ions
retinoblastoma protein (pRB) hypophosphorylation, storage [50,56-59].
decreased level of cyclin D1 [47,48]. Melanosomes produce three kinds of melanin –
Most pigment cells, in all parts of the body, seem to pheomelanin and two types of eumelanins. Pheomelanin
be constant in number and function until approximately is red to yellow and it is responsible for red hair and
the middle ages (the fourth or fifth decade of life) [49]. freckles. The most abundant form of melanin is a highly
Thereafter, the number of melanocytes in the skin and heterogenous dark brown to black, insoluble – eumelanin
hair decreases, approximately 10 percent per decade [57,60]. Eumelanin has antioxidant properties, while
[49,50]. Melanocytes from older people are larger, more pheomelanin exhibits phototoxic prooxidant action
dendritic and have decreased tyrosinase activity [45]. [55]. Melanogenesis requires a number of specific
In vitro, fetal melanocytes proliferate better than adults enzymes for melanin synthesis and structural proteins
and melanocytes from patients with the prematurely for building the fibrillar stroma for this process in the
aging disorder [48,51]. Furthermore, the reason of melanosomes. Critical enzymes are tyrosinase (TYR),
decreased number of melanocytes in older people could tyrosinase-related protein 1 (TRP1) and tyrosinase-
be apoptosis of terminally differentiated melanocytes related protein 2 (TRP2, DOPAchrome tautomerase
[48,52]. It is stressed that senescence maintenance DCT) [55,61,62]. The most important structural proteins
melanocytes with damaged DNA and simultaneously inside melanosomes are Pmel17 (gp100) and MART1
can produce viable melanocytes predisposed to (melanoma-associated antigen recognized by T cells)
malignant transformation into melanoma cells [48,53]. [45,63].
The process of hair greying is the result of melanocyte The first step, common for all melanin kinds, involve
stem cells death [46]. hydroxilation of tyrosine to
3-4-dihydroxyphenylalanine (L-DOPA) and
oxidation of L-DOPA to DOPAquinone. Both of which
3. Melanogenesis are catalyzed by tyrosinase, a crucial enzyme for the
melanin biosynthetic pathway [50,55,64]. The availability
3.1 Melanin synthesis of substrates and the function of melanogenetic
Melanogenesis is a process of pigment melanin enzymes decide which kind of melanin is produced.
synthesis within membrane-bound organelles termed DOPAquinone in the presence of cysteine creates 3- or
melanosomes [54,55]. A distribution of melanin is 5-cysteinylDOPA which then oxidize and polymerize,
responsible not only for skin, hair and eye pigmentation, giving rise to yellow/red pheomelanin (Figure 3) [45,65].

Figure 3. The simplified scheme of the melanin synthesis in melanocytes during melanogenesis. The basic substrate tyrosine under influence of
the basic enzymes as tyrosinase (TYR), tyrosine-related protein 1 (TRP1) and 2 (TRP2) changes into a polymer of melanin, a mixture of
pigments named eumelanin and pheomelanin.

319
Heterogeneity of neural crest-derived melanocytes

In the absence of cysteine, DOPAquinone undergoes receptor (MC1R) [58,71,72]. In regulation of the melanin
spontaneously non-enzymatic intramolecular cyclization synthesis signal molecules and transcription factors,
and futher rearrangment, yielding DOPAchrome. This such as MITF and cAMP, play an important roles [64,73].
process happens very fast [66-68]. DOPAchrome gives In the group of melanogenic inhibitors are sphingolipids,
eumelanins by two pathways – nonenzymatic and BMP-4 (Bone Morphogenetic Protein) and
enzymatic, with use tyrosine related proteins (TRPs). tetrahydrobiopterin [64,74,75]. During melanin synthesis
In the first case DOPAchrome undergoes spontaneous many cytotoxic agents are formed e.g. hydrogen
decarboxylation, becoming 5,6-dihydroxyindole (DHI), peroxide, quinones. To avoid the interaction between
which rapidly oxidizes and polymerizes to form dark them and other cytosolic components, the process of
brown/black, DHI-melanin (Figure 3) [66,68,69]. melanization takes place in the encircled by membrane
In the second pathway TRP2 (DCT, DOPAchrome structures – melanosomes [54].
tautomerase) tautomerizes DOPAchrome to form DHI-
2-carboxylic acid (DHICA) [70]. Next, TRP1 catalyzes 3.2 Melanosome biogenesis
DHICA oxidation and polymerization and form light Melanosome biogenesis include four stages (I–IV),
brown, DHICA-melanin. When DOPAchrome is which are determined by their quality, quantity, structure
present, TRP-2 or metal ion (as copper or zinc) favor and arrangement of the melanin produced (Figure 4).
the formation of DHICA instead of DHI (Figure 3) They are often separated into “early” (Stage I and II) and
[65,69]. “late” melanosomes (Stage II and IV) according to lack
The pigmentation is mostly genetically determined, but or presence of pigment [50,58,76,77].
the skin melanocytes activity is under control of signals The formation of stage I is still an open question [64].
from neighboring keratinocytes as well as autocrine The early melanosomes show the content of proteins
signals and environmental factors such as UV radiation derived from the endoplasmatic reticulum, coated vesicles,
[58,71]. The secretion of keratinocyte-derived factors is lysosomes and endosomes [76,78]. These melanosomes
increased by UV radiation, but it is also evident that UV are spherical vacuoles with no internal structural
can directly stimulate melanin production and melanocyte components. In stage II, the visible fibrillar matrix formed
dendricity [58,71]. Melanin production is stimulated mainly by glycoproteins (Pmel17, MART-1), melanosomes
by alpha melanocyte stimulating hormone (α-MSH) that elongates, tyrosinase is present [71,77,79]. In stage III,
influences on melanocyte through the melanocortin 1 melanosomes begin melanin production that polymerizes

Figure 4. The melanosome formation and maturation during the melanin production by a melanocyte. The melanogenesis takes place in a special
organelles named melanosomes. As first develops a vesicle (Stage I) which builds inside a fibrillar matrix formed by glycoproteins
(Pmel17, MART-1) and gets tyrosinase and other enzymes of melanogenesis (Stage II). The melanosome produces melanin, that
polymerizes and settles on the internal fibrils (Stage III). In the last Stage IV melanosome fulfills melanin. Each type of the melanin is
synthesized in a separated melanosomes.

320
M. Cichorek et al.

and settle on the internal fibrills net. Melanosomes have 4. Melanocytes in the human skin
elliptical or ellipsoidal shapes. In the last stage IV melanin
fulfills melanosome. Each type of melanin is synthesized in 4.1 Melanocytes in the epidermis
a separated melanosomes [58,72]. The eumelanosomes The mature epidermal melanocytes, as small dendritic
are large, elliptical, and contain a fibrillar matrix required cells with melanosomes, are located in a basal layer of
for eumelanin synthesis. The pheomelanosomes are the epidermis (Figure 5) [81]. Melanocyte cooperates
smaller, spherical in shape, and their matrix has loose with 30–40 associated keratinocytes and form the
structure. Both types of melanosomes might be present in epidermal melanin unit together [58,82,83]. About
a single melanocyte [50]. Most of melanosomal proteins 1200 melanocytes exist per mm2 of the skin independent
are glycosylated. They undergo early glycosylation in of the human race [84]. Cutaneous pigmentation
the endoplasmic reticulum (ER) and finally in the Golgi does not depend on melanocyte number, but the
apparatus. In the next step, proteins are trafficked to early intensity of melanogenesis and melanin distribution in
or late endosomes and then to the melanosomes. The keratinocytes. Homeostasis between these two type
tyrosinase uses this mechanism to reach melanosomes of cells is maintained by cell-cell adhesion structures
[76,77,79]. Aforementioned melanogenesis process and by auto- and paracrine factors secreted by skin
concerns the melanin production in the epidermal cells [15,58,64,77]. The result of different disturbances
melanocytes but whether it goes the same way in of these interactions can be uncontrolled proliferation
melanocytes at other places is not known. of the melanocytes and the nevi or melanoma
The skin melanocytes (epidermal, hair) transfer development [85]. Under the normal conditions all of
mature melanosomes to the dendrical processes ends the melanin produced is carried away from melanocytes
and to keratinocytes. Transport is arranged on the long into keratinocytes. Joshi et al. described the physical
axis of the dentrites under control of cytoskeleton [64,77]. contact between melanocyte and keratinocytes as the
The exact mechanism of the melanosomes transfer to ligand-receptor type interaction causing intracellular
keratinocytes is not well understood, it could follow by calcium signal in keratinocytes necessary for melanin
many ways: fusion of plasma membranes, exocytosis, transfer [86]. Melanin granules are accumulated in
transfer by membrane vesicles or cytophagocytosis keratinocytes as a cup above the nucleus, it protects
[64,77]. Human skin contains mixture of all types of keratinocytes’ DNA against UV radiation and gives
melanins [45,80]. color of the skin [50,58,87].

Figure 5. Melanocytes localization in the epidermis and hair. The epidermal melanocytes are located among the basal layer (stratum basalis) of a
stratified squamos keratinized epithelium (A). The hair melanocytes are between cells covering the hair papilla in the hair bulb (B). Stem
cells for melanocytes are located in the region named the hair bulge.

321
Heterogeneity of neural crest-derived melanocytes

Epidermal melanocytes cooperate not only with [45,88]. Thus, paracrine and autocrine molecules
keratinocytes but also with fibroblasts located in secreted by keratinocytes, fibroblast and melanocytes
the dermis [76,88]. Melanocytes, keratinocytes and are necessary for the regulation of pigment production
fibroblasts communicate with each other by secreted in response to environmental factors and also to control
factors e.g. growth factors, hormones, inflammatory topological differences in the skin color [88,92]. In
mediators and cell-cell contacts (E-cadherin, integrins) addition, these factors influence not only the growth
[45,83,89,90]. Hormones secreted by keratinocytes and pigmentation of melanocytes, but also their shape,
regulate melanocyte proliferation, melanogenesis dendricity, mobility and adhesive properties [50,93].
and dendrites growth [50,58]. The signaling pathways It is stressed that in the melanocyte-keratinocytes
cross-talk between keratinocytes and melanocytes is unit the pigment cell is very active. Melanocytes are
a part of epidermal complex network involved in the able to secrete a number of signal molecules (cytokines,
maintenance of skin homeostatis and its basic elements growth factors, hormones) targeting keratinocytes and
are presented in Figure 6 [35,50,58,91]. Proliferation other epidermal cells [15,50,58]. Melanocytes are
and pigmentation of melanocytes are under influence important components of the skin immune system [13].
of the factors secreted also by fibroblasts [50,88]. Inflammatory mediators can directly affect melanocytes
In vitro studies pointed that secreted by the dermis functions and may cause hypo- or hyperpigmentation of
fibroblasts Dickkopf-related protein 1 (DKK1) decreases the skin [94].
melanocytes growth and melanin production while The neuroendocrine capabilities of melanocytes are
neuroregulin (NRG1) increases melanin production of special importance in the human skin homeostasis

Figure 6. The graphical presentation of the basic cross-talk between melanocytes and keratinocytes in the epidermis. The melanocytes
proliferation, differentiation, melanogenesis are under control of surrounding keratinocytes. Melanocyte and up to 40 keratinocytes
form the epidermal melanin unit. CF stem cell factor; bFGF basic fibroblast growth factor; GM-CSF granulocyte-macrophage colony-
stimulating factor; ET-1 endothelin 1; a-MSH melanocyte-stimulating hormones; PGE2 prostaglandin E2; PGF2a prostaglandin
F2a; NGF nerve growth factor; c-Kit tyrosine kinase receptor; FGFR1/2 fibroblast growth factor receptor; GM-CSFR granulocyte-
macrophage colony-stimulating factor receptor; ETBR endothelin B receptor; MC1R melanocortin 1 receptor; EP1/EP3/FP prostanoid
receptors; NGFR nerve growth factor receptor; MAPK mitogen-activated protein (MAP) kinases ; PKC Protein kinase C; PKA Protein
kinase A; PLC phospholipase C; TYR tyrosinase; TRP1 tyrosinase-related protein 1; TRP2 tyrosinase-related protein 2; MITF-M
Melanocyte-specific MITF (Microphthalmia-associated transcription factor) isoform; CRE cAMP response elements; CREB cAMP
response element-binding.

322
M. Cichorek et al.

[14,15]. Cumulative evidence provided that these cells cells with inactive way of melanin biosynthesis also
are elements of cholinergic system and hypothalamic- exist, the melanocyte stem cells (MelSCs) [46,96,101].
pitiutary-thyroid axis [16,17,22,95]. Thus, skin MelSC are located in the hair follicle bulge together
melanocytes, as the mediators of the humoral and with hair follicle stem cells (HFSCs; precursors for
neural signals, coordinate not only cutaneous but keratinocytes). The bulge area is a part of outer root
also a systemic (peripherial, global) homeostasis. The sheath that provides the insertion point for arector pili
melanocytes with melanosomes – unique organelle/ muscle and points the bottom of the permanent portion
messengers – participate in the regulation of skin of hair follicles (Figure 5). Each time a hair is lost the
functions including its protective, bioregulatory and hair follicle regenerates and MelSC are activated
sensory properties [17,21,22,95]. under control of Wnt signaling [102]. After stimulation
these cells migrate from bulge into a new hair bulb and
4.2 Hair follicle melanocytes differentiate into mature melanocytes [33,103]. MelSCs
Melanocytes are responsible for either skin or hair are a melanocyte reservoir not only for hair but also for
pigmentation. They are located in the proximal bulb of epidermis [46]. The understanding of MelSCs biology
each hair follicle among the epithelial cells of the hair is the key for explanation the mechanisms of hair
papilla at the apex. Melanocytes dendritic processes pigmentation, greying and skin disorders e.g. vitiligo
enter between hair keratinocytes and form the hair [46,97].
melanin units (Figure 5) [96,97]. Hair pigmentation is the
result of the structural and functional interactions between
follicular melanocytes, matrix keratinocytes and dermal 5. Melanocytes in places other than
papilla fibroblasts. Process of hair pigmentation includes skin
the melanogenic activity of follicular melanocytes, the
transfer of melanin granules into keratinocytes that form As the result of prenatal migration melanocytes are
the pigmented hair shaft [96-98]. Hair melanocytes are located in the ear, eye, nervous system, and heart. In
larger and more dendritic than the epidermal ones, these organs melanocytes are not related to keratinocytes
including mainly mature Stage IV melanosomes but it – the main cells affected pigment cells of the skin.
is considered that melanin transport to the keratinocytes Pigment cells functioning in the microenvironment of
in the growing hair shaft is similar to the epidermal [97]. ear (stria vascularis of cochlear duct), eye (choroid, iris,
The mechanism of the melanin synthesis and its ciliary body), brain (leptomeninges) and heart are linked
control in the hair is similar to epidermal melanocytes probably with their impact with local cells/structures. It
but it is stated that hair follicles melanocytes are must be emphasized that biology of these melanocytes
more sensitive to aging influences than the epidermal is less known than skin melanocytes, especially in the
melanocytes, leading to hair graying [97]. It seems that context of the cell cooperation [50].
substances derived from fibroblast of dermal papilla (e.g.
insulin-like growth factor IGF-1, keratinocyte growth 5.1 Eye
factor KGF, hepatocyte growth factor HGF, noggin Melanocytes of NCC origin exist in the choroid and
and stem cell factor SCF) have special significance stroma of the iris and the ciliary body. Pigment is also
for control division and differentiation of the hair matrix produced in the retinal pigment epithelium (RPE) and
keratinocytes and differentiation, proliferation and epithelia of the iris and ciliary body, but the origin of
activity of melanocytes during the anagen hair growth these cells is the optic cup not the neural crest. Unlike
phase [96,99,100]. epidermal melanocytes, ocular melanocytes are in
Epidermal melanocytes proliferate rarely, while hair contact only with each other, and they do not transfer their
melanocytes divide repeatedly, migrate and undergo melanosomes. Mature melanocytes remain quiescent
maturation in every hair cycle [46]. Melanogenesis and do not produce melanin in adults [5]. Ocular
process takes place only during the anagen stage melanocytes are embeded in the connective tissue
(growing phase) of the hair growth cycle; pigment and are probably under influence of different cells e.g.
formation is turned-off in catagen (regressing phase) fibroblasts, endothelial cells, smooth muscles, neurons
and absent in telogen (resting phase). Additionally it is [104-106].These localizations probably determines the
marked that during catagen completely differentiated role of ocular melanocytes. In the anterior part of the
bulbar melanocytes die through apoptosis but with the iris, which is exposed to sunlight and UV radiation, the
development of the new hair new melanocytes appear. photoprotecting effect of melanin dominates. Deeply
So, except for high melanogenic melanocytes in the hair localized ocular melanocytes are exposed to high oxygen
bulb a minor population of poorly differentiated pigment tension and melanin seems to protect cells against

323
Heterogeneity of neural crest-derived melanocytes

oxidative damage as free radical scavenger [105,107]. indicated that noise induces cochlea hyperpigmentation
Studies of patients with the iris melanogenesis defects and melanosomes transfer to marginal cells [5,23,115].
pointed to abnormal adrenergic innervation of the iris It seems that melanocytes prevent noise-induced
[108]. Relationship of melanocytes with blood vessels, damage of the inner ear but the molecular mechanism
nerve fibers and the presence of melanocytes groups of the stress-induced melanization is unknown. The
around and along an extraocular muscle, may indicate melanin in ear, as a biological reservoir for ions, can
a functional role as an ion reservoir [104]. Sharif et al., bind to ototoxic drugs [116]. Noise and ototoxic drugs
mentioning about intercellular communication within iris induce oxidative stress; high level of the reactive
melanogenic process, indicate the histamine receptor oxygen substances (ROS) can destroy sensory cells in
expression on the human isolated iridial fibroblasts and the cochlea. Thus melanin as the scavenger of ROS
the presence of endothelin-1 (ET-1), prostaglandin E2 protects sensory cells from damage [23,117].
(PGE2) on the ocular melanoblast [106]. Higa et al.
described the presence of melanocytes in an additional 5.3 Heart
section of the eye, corneal limbus, transitional area Recently, in animals studies, the presence of
between cornea and sclera [109]. These pigment cells melanocytes in the heart has been described [24,118].
were MART-1 (melanoma antigen recognized by T-cells) During mammalian embryogenesis neural crest cells
and tyrosinase positive, have multiple processes and are involved in the outflow part of the heart formation.
exist as sporadic cells among basal epithelial cells. Thus, the presence of the melanocyte cells of the NCC
Thus, melanocytes and epithelial cells in the limbus may origin is not a surprise [118]. Melanocytes are situated
form a functional network similar to the melanin unit of in valves, atrial and intraventricular septa [24]. These
the skin [109]. melanocytes in heart have some structural differences
in comparison to other melanocytes e.g. have a lot of
5.2 Ear caveloae along the plasma membranes [5]. Cardiac
The relationship between pigmentation and hearing melanocytes are present only in four-chambered hearts
has been noticed by Alfonso Corti in 19th century [118]. The functions of the heart melanocytes remain
[110]. The presence of melanocytes are required unclair.
for normal ear development, a deficiency of these In human heart tissue in the atrium and pulmonary
cells in Waardenburg’s Syndrome (WS) resulted in vein walls are cells with dopachrometautomerase activity
hypopigmentation and deafness. In another situation, (DCT, TRP2; melanogenesis enzyme, melanoblast’s
albinism, the melanin absence does not influence the marker) [119]. Detailed analysis showed that these
hearing process. Thus, melanocytes not melanin is DCT-positive cells also express tyrosinase and MITF,
necessary for the ear development [23]. It is known that but further examinations are necessary to explain if
melanocytes appear in almost all parts of the inner ear, these cells are melanocytes or cardiomyocytes able
such as the cochlear duct, stria vascularis, vestibular to melanin synthesis [119]. Levin et al. observed that
organ in the region surrounding the cristae and maculae, the presence of DCT-positive cardiac melanocyte-like
semicircular canals [5]. cells within the pulmonary veins and atrium contribute
The stria vascularis of the cochlear duct is composed to atrialarrhythmogenesis in mouse [119]. This incidate
of a stratified epithelium, between the cells of which a new research direction for human atrial arrhythmias
runs a dense capillary network. The melanocytes and melanocytes.
form intermediate layer in the stria vascularis. They
are the starry-shaped cells, have globular melanin 5.4 Nervous system
granules [23]. The key function of stria vascularis is the In the central nervous system melanocytes of the NCC
production of endolymph and the creation of the positive origin are distributed in the leptomeninges over the
endocochlear potential through the control of the ionic ventrolateral surface of the medulla oblongata of the
balance. Melanocytes seem to participate in this last human brain [25,120]. These dendritic cells contain
function. Melanocytes as a biological reservoir for melanosomes [5]. It is indicated that these melanocytes
calcium and potassium ions could regulate the content could play neuroendocrine functions e.g. secretion
of these ions in the endolymph, that is important for the of opioids. The authors suggest the possibility that
hearing physiology [5,59,111]. The strial melanocytes melanocytes are involved in the control of the respiratory
activity is controlled by the same factors as in the rhytm and sleeping regulation [120]. The human
epidermis e.g. MITF, c-Kit, SOX 10, PAX3 [23,112,113]. brain has some places with neurons able to produce
It has been observed that in humans the cochlear neuromelanin but these neurons developed from neural
melanin level increases with age [114]. The animal studies tube not NCC [120,121].

324
M. Cichorek et al.

Summarizing, main functions of the melanocytes arising from migration of a multipotent precursor cell
localized outside skin are: the protection of tissues from along nerve projections. It is probable that there are
potentially harmful factors (e.g. free radicals, binding cells with stem cell properties located in the cutaneous
toxins), storage ions and anti-inflammatory action. nerves [26,28]. These cells are retained in a stem
cell-like state until the signal send by the end of the
cutaneous nerve promotes these cells to differentiate
6. Stem cells for human melanocytes into melanocytes [125]. But there are also observations
in adults that some melanoblasts from the dorsal way of NCC
migration stay in the dermis after the end of epidermis
Melanocytes proliferate rarely but some situations point inoculation, up to the second trimester during fetal time
that in the skin are present precursors for melanocytes [33,126,127]. Whether they survive in the human adult
e.g. repigmentation of the skin of vitiligo patients after dermis is still an open problem.
phototherapy [122]. There are difficulties in the identification of
What do we know about the presence of melanoblasts melanoblasts in the adults skin because of the lack
in the adult skin? of a defined set of melanoblast markers. There are
The first documented reservoir for melanocytes stem some markers determining the melanocyte fate of
cells was the bulge area of the hair follicles either in NCC e.g. dopachrometautomerase (TRP2/DCT),
mouse or human (Figure 5) [103,123]. The bulge region MITF, c-Kit.
contains pluripotential, morphologically undifferentiated Epidermis and dermis are places for the melanocytes
cells which develop into keratinocyte progenitors and reservoir and give opportunity for a new look at the
melanoblasts not only for hair but also for epidermis melanoma heterogeneity, a tumor which develops
[46]. from melanocytes. There is no agreement if the mature
However, there are growing evidence that the dermis melanocytes or cells from earlier developmental stages
is also a reservoir of melanocytes [33,122,124]. Where may follow the tumoricidal transformation [34,128-130].
are the melanoblasts located in the dermis? Answer Whiteman et al. divides melanoma(s) into melanoma
to this question is not satisfactory enough. Recent arising from epithelial melanocytes (origin from dorsal
developmental studies using model organisms and migration way) and not associated with epithelia (origin
lineage tracing have been able to trace melanocytes from Schwann precursors cells) [129].

References

[1] Fitzpatrick T., Becker S.W., Lerner A.B., Montgomery [5] Colombo S., Berlin I., Delmas V., Larue L., Classical
H., Tyrosinase in human skin: Demonstration of its and nonclassical melanocytes in vertebrates,
presence and its role in human melanin formation, In: Borovansky J., Riley P.A., (Eds.), Melanins
Science, 1950, 112, 223–225 and melanosomes. Biosynthesis, biogenesis,
[2] Nordlund J.J., Boissy R.E., Hearing V.J., King R.A, physiological and pathological functions, Willey-
Ortonne J.P., The Pigmentary System. Physiology Blackwell, Weinheim, 2011
and Pathophysiology, Oxford University Press, [6] Rawles M.E., Origin of pigment cells from neural
New York, 1998 crest in the mouse embryo, Physiol. Zool., 1947,
[3] Halaban R., Hebert D.N., Fisher D.E., Biology of 20, 248-270
Melanocytes, In: Freedberg I.M., Wolff K., Austen [7] O’Rahilly R., Müller F., The development of the neural
K.F., Goldsmith L.A., Katz S.I. (Eds.), Fitzpatrick’s crest in the human, J. Anat., 2007, 211, 335-351
Dermatology in General Medicine, McGraw-Hill, [8] Harris M.L., Erickson C.A., Lineage specification in
New York, 2003 neural crest cell path finding, Dev. Dyn., 2007, 236,
[4] Aoki H., Yamada Y., Hara A., Kunisada T., Two 1-19
distinct types of mouse melanocyte: differential [9] Betters E., Liu Y., Kjaeldgaard A., Sundström E.,
signaling requirement for the maintenance of García-Castro MI., Analysis of early human neural
non-cutaneous and dermal versus epidermal crest development, Dev. Biol., 2010, 344, 578-592
melanocytes, Development., 2009, 136, 2511- [10] Theveneau E., Mayor R., Neural crest delamination
2521 and migration: From epithelium-to-mesenchyme

325
Heterogeneity of neural crest-derived melanocytes

transition to collective cell migration, Dev. Biol., melanocytes in the leptomeninges of the human
2012, 366, 34-54 brain., J. Invest. Dermatol., 1984, 82, 235-238
[11] Le Douarin N.M., Creuzet S., Couly G., Dupin [26] Cramer S.F., The histogenesis of acquired
E., Neural Crest Cell plasticity and its limits, melanocytic nevi-based on a new concept of
Development, 2004, 131, 4637-4650 melanocytic differentiation, Am. J. Dermatopathol.,
[12] Ernfors P., Cellular origin and developmental 1984, 6, 289-298
mechanisms during the formation of skin [27] Cramer S.F., Stem cells for epidermal melanocytes-
melanocytes, Exp. Cell Res., 2010, 316, 1397- a challenge for students of dermatology, Am. J.
1407 Dermatopathol., 2009, 31, 331-341
[13] Lu Y., Zhu W.Y., Tan C., Yu G.H., Gu J.X., [28] Adameyko I., Lallemend F., Aquino J.B., Pereira J.A.,
Melanocytes are potential immunocompetent Topilko P., Muller T., et al., Schwann cell precursors
cells: evidence from recognition of immunological from nerve innervation are a cellular origin of
characteristics of cultured human melanocytes, melanocytes in skin, Cell, 2009, 139, 366–379
Pigment Cell Res., 2002, 15, 454-460 [29] Dupin E., Real C., Glavieux-Pardanaud C., Vaigot
[14] Slominski A., Paus R., Are L-tyrosine and L-dopa P., LeDourain N.M., Reversal of developmental
hormone-like bioregulators?, J. Theor. Biol., 1990, restrictions in neural crest cells lineages; transition
143, 123-138 from Schwann cells to glial-melanocytic precursors
[15] Slominski A., Paus R., Schadendorf D., in vitro, Proc. Natl. Acad. Sci. USA, 2003, 100,
Melanocytes as “sensory” and regulatory cells in 5229-5233
the epidermis, J. Theor. Biol., 1993, 164, 103-120 [30] Cramer S.F., Fesyuk A., On the development
[16] Schallreuter K.U., Lemke K.R., Pittelkow M.R., of neurocutaneous units-implications for the
Wood J.M., Körner C., Malik R., Catecholamines histogenesis of congenital, acquired, and dysplastic
in human keratinocyte differentiation, J. Invest. nevi, Am. J. Dermatopathol., 2012, 34, 60-81
Dermatol., 1995, 104, 953-957 [31] Hara M., Toyoda M., Yaar M., Bhawan J., Avila
[17] Grando S.A., Pittelkow M.R., Schallreuter K.U., E.M., Penner I.R., et al., Innervation of melanocytes
Adrenergic and cholinergic control in the biology of in human skin, J. Exp. Med., 1996, 184, 1385-1395
epidermis: physiological and clinical significance., [32] Holbrook K.A., Underwood R.A., Vogel A.M., Gown
J. Invest. Dermatol., 2006, 126, 1948-1965 A.M., Kimball H., The appearance, density and
[18] Slominski A., Wortsman J., Luger T., Paus R., distribution of melanocytes in human embryonic
Solomon S., Corticotropin releasing hormone and and fetal skin revealed by anti melanoma
proopiomelanocortin involvement in the cutaneous monoclonal antibody HMB-45, Anat. Embryol.,
response to stress, Physiol. Rev., 2000, 80, 979-1020 1989, 180, 443-455
[19] Slominski A., Wortsman J., Neuroendocrinology of [33] Gleason B.C., Crum C.P., Murphy G.F.,
the skin, Endocr. Rev., 2000, 21, 457-487 Expression patterns of MITF during human
[20] Slominski A., Wortsman J., Tobin D.J., The cutaneous embryogenesis: evidence for bulge
cutaneous serotoninergic/melatoninergic system: epithelial expression and persistence of dermal
securing a place under the sun, FASEB J., 2005, melanoblasts, J. Cutan. Pathol., 2008, 35, 615-622
19, 176-194 [34] Zabierowski S.E., Fukunaga-Kalabis M., Li L.,
[21] Slominski A., Neuroendocrine activity of the Herlyn M., Dermis-derived stem cells: a source of
melanocyte, Exp. Dermatol., 2009, 18, 760-763 epidermal melanocytes and melanoma? Pigment
[22] Slominski A.T., Zmijewski M.A., Skobowiat C., Cell Melanoma Res., 2011, 24, 422-429
Zbytek B., Slominski R.M., Steketee J.D., Sensing [35] Hirobe T., How are proliferation and differentiation of
the environment: regulation of local and global melanocytes regulated?, Pigment Cell Melanoma
homeostasis by the skin’s neuroendocrine system, Res., 2011, 24, 462-478
Adv. Anat. Embryol. Cell Biol., 2012, 212, 1-115 [36] Cooper C.D., Raible D.W., Mechanisms for
[23] Tachibana M., Sound needs sound melanocytes to reaching the differentiated state: Insights from
be heard, Pigment Cell Res., 1999, 12, 344-354 neural crest-derived melanocytes, Semin. Cell
[24] Mjaatvedt C.H., Kern C.B., Norris R.A., Fairey S., Dev. Biol., 2009, 20, 105-110
Cave C.L., Normal distribution of melanocytes in [37] Sommer L., Generation of melanocytes from neural
the mouse heart, Anat. Rec. Discov. Mol. Cell. crest cells, Pigment Cell Melanoma Res., 2011, 24,
Evol. Biol., 2005, 285, 748–757 411-421
[25] Goldgeier M,H,, Klein L.E., Klein-Angerer S., [38] Hari L., Miescher I., Shakhova O., Suter U., Chin L.,
Moellmann G., Nordlund J.J., The distribution of Taketo M., et al., Temporal control of neural crest

326
M. Cichorek et al.

lineage generation by Wnt/β-catenin signaling, [53] Campisi J., The role of cellular senescence in skin
Development, 2012, 139, 2107-2117 aging, J. Investig. Dermatol. Symp. Proc., 1998, 3,
[39] Cornell R.A., Eisen J.S., Notch in the pathway: 1-5
the roles of Notch signaling in neural crest [54] Seiji H., Fitzpatrick T.B., The reciprocal relationship
development, Semin. Cell Dev. Biol., 2005, 16, between melanization and tyrosinase activity in
663-672 melanosomes (melanin granules), J. Biochem.,
[40] Raible D.W., Ragland J.W., Reiterated Wnt and 1961, 49, 700-706
BMP signals in neural crest development, Semin. [55] Simon D.J., Peles D., Wakamatsu K., Ito S., Current
Cell Dev. Biol., 2005, 16, 673-682 challenges in understanding melanogenesis:
[41] Lin J.Y., Fisher D.E., Melanocyte biology and skin bridging chemistry, biological control, morphology
pigmentation, Nature, 2007, 445, 843-850 and function, Pigment Cell Melanoma Res., 2009,
[42] Clay M.R., Halloran M.C., Regulation of cell 22, 563-579
adhesions and motility during initiation of neural [56] Pathak M.A., Rilley F.C., Fitzpatrick T.B.,
crest migration, Curr. Opin. Neurobiol., 2011, 21, Melanogenesis in human skin following exposure
17-22 to long-wave ultraviolet and visible light., J. Invest.
[43] Vance K.W., Goding C.R., The transcription Dermatol., 1962, 39, 435-443
network regulating melanocyte development and [57] Riley P.A., Melanin., Int. J. Biochem. Cell Biol.,
melanoma, Pigment Cell Res., 2004, 17, 318- 1997, 29, 1235-1239
325 [58] Slominski A., Tobin D.J., Shibahara S., Wortsman
[44] McGill G.G., Horstmann M., Widlund H.R., Du J., J., Melanin pigmentation in mammalian skin and its
Motyckova G., Nishimura E.K. et al., Bcl2 regulation hormonal regulation, Physiol. Rev., 2004, 84, 1155-
by the melanocyte master regulator Mitf modulates 1228
lineage survival and melanoma cell viability, Cell, [59] Bush W.D., Simon J.D., Quantification of Ca(2+)
2002, 109, 707-718 binding to melanin supports the hypothesis that
[45] Yamaguchi Y., Brenner M., Hearing V.J., The melanosomes serve a functional role in regulating
regulations of skin pigmentation, J. Biol. Chem., calcium homeostasis, Pigment Cell Res., 2007,
2007, 13, 1-11 20, 134–139
[46] Nishimura E.K., Melanocyte stem cells: a [60] Ito S., High-performance liquid chromatography
melanocyte reservoir in hair follicles for hair and (HPLC) analysis of eu- and pheomelanin in
skin pigmentation, Pigment Cell Melanoma Res., melanogenesis control, J. Invest. Dermatol., 1993,
2011, 24, 401-404 100, 166–171
[47] Bennet D.C., Medrano E.E., Molecular regulation [61] Fitzpatrick T.B., Miyamoto M., Ishikawa K., The
of melanocyte senescence, Pigment Cell Res., evolution of concepts of melanin biology, Arch.
2002, 15, 242-250 Dermatol., 1967, 96, 305-323
[48] Rizos H., Becker T.M., Holland E.A., Cell cycle [62] Hearing V.J., Tsukamoto K., Enzymatic control
regulation in the melanocyte, In: Thomson J.F., of pigmentation in mammals, FASEB J., 1991, 5,
Morton D.L., Kroon B.B.R., Textbook of Melanoma, 2902-2909
Martin Dunitz Taylor & Francis Group, London, [63] Hoashi T., Watabe H., Muller J., Yamaguchi Y.,
2004 Vieira W.D., Hearing V.J., MART-1 is required
[49] Nordlund J.J., The lives of pigment cells, Dermatol. for the function of the melanosomal matrix
Clin., 1986, 4, 407-418 protein PMEL17/GP100 and the maturation of
[50] Costin G.E., Hearing V.J., Human skin pigmentation: melanosomes, J. Biol. Chem., 2005, 280, 14006-
melanocytes modulate skin color in response to 14016
stress, FASEB J., 2007, 21, 976-994 [64] Park H.Y., Kasmadaki M., Gilchrest Y.B.A., Cellular
[51] Haddad M.M., Xu W., Medrano E.E., Aging in mechanisms regulating melanogenesis, Cell Mol.
epidermal melanocytes: cell cycle genes and Life Sci., 2009, 66, 1493-1506
melanins, J. Investig. Dermatol. Symp. Proc., [65] Ito S., The IFPCS presidential lecture: a chemist’s
1998, 3, 36-40 view of melanogenesis, Pigment Cell Res., 2003,
[52] Selzer E., Schlagbauer-Wadl H., Okamoto I., 16, 230–236
Pehamberger H., Pötter R., Jansen B., Expression [66] Pawelek J.M., After dopachrome?, Pigment Cell
of Bcl-2 family members in human melanocytes, in Res., 1991, 4, 53–62
melanoma metastases and in melanoma cell lines, [67] Olivares C., Jiménez-Cervantes C., Lozano
Melanoma Res., 1998, 8, 197-203 J.A. Solano F., García-Borrón J.C., The

327
Heterogeneity of neural crest-derived melanocytes

5,6-dihydroxyindole-2-carboxylic acid (DHICA) [81] Busam K.J., Charles C., Lee G., Halpern A.C.,
oxidase activity of human tyrosinase, Biochem. J., Morphologic features of melanocytes, pigment
2001, 354, 131-139 keratinocytes, and melanophages by in vivo
[68] Ito S., Wakamatsu K., Chemistry of mixed confocal scanning laser microscopy, Mod. Pathol.,
melanogenesis-pivotal roles of dopaquinone, 2001, 14, 862-868
Photochem. Photobiol., 2008, 84, 582-592 [82] Fitzpatrick T.B., Breathnach A.S., The epidermal
[69] Del Marmol V., Beerman F., Tyrosinase and related melanin unit system, Dermatol. Wochenschr.,
proteins in mammalian pigmentation, FEBS Lett., 1963, 147, 481-489
1996, 381, 165-168 [83] Haass N.K., Smalley K.S., Li L., Herlyn M.,
[70] Korner A.M., Pawelek J., DOPAchrome Adhesion, migration and communications in
conversion: a possible control point in melanin melanocytes and melanoma, Pigment Cell Res.,
biosynthesis, J. Invest. Dermatol., 1980, 75, 192– 2005, 18, 150-159
195 [84] Miot L.D., Miot H.A., Silva M.G., Marques M.E.,
[71] Hearing V.J., Determination of melanin synthetic Physiopathology of melasma, An. Bras. Dermatol.,
pathway, J. Invest. Dermatol., 2011, 131, 8-11 2009, 84, 623-635
[72] Schiaffino M.V., Signalling pathways in melanosome [85] Rickelt S., Franke W.W., Doerflinger Y., Goerdt
biogenesis and pathology, Int. J. Biochem. Biol., S., Brandner J.M., Peitsch W.K., Subtypes of
2010, 42, 1094-1104 melanocytes and melanoma cells distinguished by
[73] Goding C.R., Mitf from neural crest to melanoma: their intercellular contacts: heterotypic adherent
signal transduction and transcription in the junctions, adhesive associations, and dispersed
melanocyte lineage, Genes. Dev., 2000, 14, 1712- desmoglein 2 glycoproteins, Cell Tissue Res.,
1728 2008, 334, 401-422
[74] Park H.Y., Wu C., Yaar M., Stachur C.M., Kosmadaki [86] Joshi P.G., Nair N., Begum G., Joshi N.B., Sinkar
M., Gilchrest B.A., Role of BMP-4 and its signaling V.P., Vora S., Melanocyte-keratinocyte interaction
pathways in cultured human melanocytes, Int. J. induces calcium signaling and melanin transfer to
Cell Biol., 2009, 2009, doi:10.1155/2009/750482 keratinocytes, Pigment Cell Res., 2007, 20, 380-
[75] Schallreuter K.U., Wood J.M., Pittelkow M.R., 384
Gütlich M., Lemke K.R., Rödl W., et al., Regulation [87] Plonka P.M., Passeron T., Brenner D.J., Tobin S.,
of melanin biosynthesis in the human epidermis Shibahara S., Thomas A., What are melanocytes
by tetrahydrobiopterin, Science, 1994, 263, 1444- really doing all day long…?, Exp. Dermatol., 2009,
1446 18, 799-819
[76] Hearing V.J., Biogenesis of pigment granules: a [88] Choi W., Kolbe L., Hearing V.J., Characterization
sensitive way to regulate melanocyte function, J. of the bioactive motif of neuregulin-1, a fibroblast-
Dermatol. Sci., 2005, 37, 3-14 derived paracrine factor that regulates the
[77] Watabe H., Valencia J.C., Le Pape E., Yamaguchi constitutive color and the function of melanocytes
Y., Nakamura M., Rouzaud F., Involvement of in human skin, Pigment Cell Melanoma Res., 2012,
dynein and spectrin with early melanosome 25, 1-5
transport and melanosomal protein trafficking, J. [89] Lee A.Y., Role of keratinocytes in the development
Invest. Dermatol., 2008, 128, 162-173 of vitiligo, Ann. Dermatol., 2012, 24, 115-125
[78] Raposo G., Tenza D., Murphy D.M., Berson [90] Tabone-Eglinger S., Wehrle-Haller M., Aebischer
J.F., Marks M.S., Distinct protein sorting and N., Jacquier M.C., Wehrle-Haller B., Membrane
localization to premelanosomes, melanosomes, bound Kit ligand regulates melanocyte adhesion
and lysosomes in pigmented melanocytic cells, J. and survival, providing physical interaction with an
Cell Biol., 2001, 152, 809-824 intraepithelial niche, FASEB J., 2012, 26, 3738-
[79] Kushimoto T., Basrur V., Valencia J., Matsunaga 3753
J., Vieira W. D., Ferrans V.J., et al., A model for [91] Hirobe T., Role of keratinocyte-derived
melanosome biogenesis based on the purification factors involved in regulating the proliferation
and analysis of early melanosomes, Proc. Natl. and differentiation of mammalian epidermal
Acad. Sci. USA., 2001, 98, 10698–10703 melanocytes, Pigment Cell Res., 2004, 18, 2-12
[80] Kondo T., Hearing V.J., Update on the regulation [92] Imokawa G., Autocrine and paracrine
of mammalian melanocyte function and skin regulation of melanocytes in human skin and
pigmentation, Expert. Rev. Dermatol., 2011, 6, 97- in pigmentary disorders, Pigment Cell Res.,
108 2004, 17, 96-110

328
M. Cichorek et al.

[93] Wang Z., Coleman D.J., Bajaj G., Liang X., [107] Sarna T.J., Properties and function of the ocular
Ganguli-Indra G., Indra AK., RXRα ablation in melanin - a photobiophysical view, Photochem.
epidermal keratinocytes enhances UVR-induced Photobiol. B., 1992, 12, 215-258
DNA damage, apoptosis, and proliferation [108] Melamed S., Lahav M., Sandbank U., Yassur Y.,
of keratinocytes and melanocytes, J. Invest. Ben-Sira I., Fuch’s heterochromic iridocyclitis:
Dermatol., 2011, 131, 177-187 an electron microscopic study of the iris, Invest.
[94] Taylor S., Grimes P., Lim J., Im S., Lui H., Ophthalmol. Vis. Sci., 1978, 17, 1193-1199
Postinflammatory hyperpigmentation, J. Cutan. [109] Higa K., Shimmura S., Miyashita H., Shimazaki
Med. Surg., 2009, 13, 183-191 J., Tsubota K., Melanocytes in the corneal limbus
[95] Slominski A., Zmijewski M.A., Pawelek J., interact with K19-positive basal epithelial cells,
L-tyrosine and L-dihydroxyphenylalanine as Exp. Eye Res., 2005, 81, 218-223
hormone-like regulators of melanocyte functions, [110] Corti A., Recherche sur l`organe des l`ouie des
Pigment Cell Melanoma Res., 2012, 25, 14-27 mammiferes. Premiere partie 2 Z. wiss, Zoologie,
[96] Commo S., Bernard B. A., Melanocyte 1851, 3, 109-169
subpopulation turnover during the human hair [111] Meyer zum Gottesberge A.M., Calcium dependent
cycle: an immunohistochemical study, Pigment intercellular interaction of the neural crest
Cell Res., 2000, 13, 253-259 derivate-melanocytes and the epithelial cells of
[97] Slominski A., Wortsman J., Plonka P.M., the vestibular organ, Acta. Otolaryngol., 1995,
Schallreuter K.U., Paus R., Tobin D.J., Hair 520, 360-361
Follicle Pigmentation, J. Invest. Dermatol., 2005, [112] Tachibana M., Cochlear melanocytes and MITF
124, 13-21 signaling, J. Invest. Dermatol. Symp. Proc., 2001,
[98] Commo S., Gaillard O., Thibaut S., Bernard B.A., 6, 95-98
Absence of TRP-2 in melanogenic melanocytes of [113] Price E.R., Fisher D.E., Sensorineural deafness
human hair, Pigment Cell Res., 2004, 17, 488-497 and pigmentation genes: melanocytes and the
[99] Randall V.A., Androgens and hair growth, Mitf transcriptional network, Neuron, 2001, 30,
Dermatol. Ther., 2008, 21, 314-328 15-18
[100] Slominski A., Paus R., Melanogenesis is coupled [114] Suzuki T., Nomoto Y., Nakagawa T., Kuwahat
to murine anagen: toward new concepts for N., Ogawa H., Suzuki Y., et al., Age-dependent
the role of melanocytes and the regulation degeneration of the stria vascularis in human
of melanogenesis in hair growth, J. Invest. cochleae, Laryngoscope, 2006, 116, 1846-1850
Dermatol., 1993, 101, 90-97 [115] Wassif G.A., El Begermy M., Age-related changes
[101] Slominski A., Paus R., Plonka P., Chakraborty A., of the cochlear lateral wall (stria vascularis) in the
Maurer M., Pruski D., et al., Melanogenesis during guinea pigs: an ultrastructural study, Egypt J.
the anagen-catagen-telogen transformation of Histol., 2008, 31, 332-340
the murine hair cycle, J. Invest. Dermatol., 1994, [116] Meyer zum Gottesberge A.M., Physiology and
102, 862-869 pathophysiology of inner ear melanin, Pigment
[102] Goding C.R., Melanocytes; the new black, Int. J. Cell Res., 1988, 1, 238-249
Biochem. Cell Biol., 2007, 39, 275-279 [117] Henderson D., McFadden S.L., Liu C.C., Hight N.,
[103] Nishimura E., Jordan S.A., Oshima H., Yoshida Zheng X.Y., The role of antioxidants in protection
H., Osawa M., Moriyama M., et al., Dominant from impulse noise, Ann. N. Y. Acad. Sci., 1999,
role of the niche in melanocyte stem-cell fate 884, 368-380
determination, Nature, 2002, 416, 854-860 [118] Brito F.C., Kos L., Timeline and distribution of
[104] Wan der Werf F., Baljet B., Otto A.J., Pigment- melanocyte precursors in the mouse heart,
containing cells in extraocular tissues of the Pigment Cell Melanoma Res., 2008, 21, 464-470
primate, Doc. Ophthalmol., 1992, 81, 357-368 [119] Levin M.D., Lu M.M., Petrenko N.B., Hawkins
[105] Hu D.N., Simon J., Sarna T., Role of ocular B.J., Gupta T.H., Lang D., et al., Melanocyte-like
melanin in ophtalmic physiology and pathology, cells in the heart and pulmonary veins contribute
Phatochem. Phatobiol., 2008, 84, 639-644 to atrial arrhythmia triggers, J. Clin. Invest., 2009,
[106] Sharif N.A., Crider J.Y., Intracellular signaling in 119, 3420-3436
human iridial fibroblasts and iridial melanocytes [120] Takeda K., Takahashi N.H., Shibahara S.,
in response to prostaglandins, endothelin, Neuroendocrine function of melanocytes: beyond
isoproterenol, and other pharmacological agents, the skin-deep melanin maker, Tohoku J. Exp.
Curr. Eye Res., 2011, 36, 310-320 Med., 2007, 211, 201-221

329
Heterogeneity of neural crest-derived melanocytes

[121] Zecca L., Bellei Ch., Costi P., Albertini A., Monzani progenitor and melanocyte lineages in the
E., Casella L., New melanic pigments in the cranial neural crest, Development, 2012, 139,
human brain that accumulate in aging and block 397-410
environmental toxic metals, Proc. Natl. Acad. Sci. [126] Suder E., Bruzewicz S., Melanocytes of fetal
USA, 2008, 105, 17567-17572 dermis - studies with anti-HMB-45 antibody, Med.
[122] Davids L.M., du Toit E., Kidson S.H., Todd G., A Sci. Monit., 2004, 10, 229-232
rare repigmentation pattern in a vitiligo patient: [127] Dupin E., Sommer L., Neural crest progenitors
a clue to an epidermal stem-cell reservoir of and stem cells: From early development to
melanocytes?, Clin. Exp. Dermatol., 2009, 34, adulthood, Dev. Biol., 2012, 366, 83-95
246-248 [128] Schatton T., Frank M.H., Cancer stem cells and
[123] Nishimura E.K., Granter S.R., Fisher D.E., human malignant melanoma, Pigment Cell
Mechanisms of hair graying; incomplete Melanoma Res., 2007, 21, 39-55
melanocyte stem cell maintance in the niche, [129] Whiteman D.C., Pavan W.J., Bastian B.C., The
Science, 2005, 307, 720-724 melanomas: a synthesis of epidemiological,
[124] Toma J.G., McKenzie I.A., Bagli D., Miller F.D., clinical, histopathological, genetic, and biological
Isolation and characterization of multipotent skin- aspects, supporting distinct subtypes, casual
derived precursors from human skin, Stem Cells, pathways, and cells of origin, Pigment Cell
2005, 23, 727-737 Melanoma Res., 2011, 24, 879-897
[125] Adameyko I., Lallemend F., Furlan A., Zinin [130] Hoek K.S., Goding C.R., Cancer stem cells versus
N., Aranda S., Kitambi S.S., et al., Sox2 and phenotype-switching in melanoma, Pigment Cell
Mitf cross-regulatory interactions consolidate Melanoma Res., 2010, 23, 746-759

330

You might also like