You are on page 1of 27

https://www.britannica.

com/science/melanocyte

Melanocyte, specialized skin cell that produces the protective skin-


darkening pigment melanin. Birdsand mammals possess these pigment cells, which are found
mainly in the epidermis, though they occur elsewhere—e.g., in the matrix of the hair.
Melanocytes are branched, or dendritic, and their dendrites are used to transfer pigment
granules to adjacent epidermal cells.
All melanocytes, whether resident in the basal epidermis or in the matrix of the hair, have
migrated there during embryonic life from a region known as the neural crest. Each epidermal
melanocyte is associated with a group of neighbouring keratinocytes (keratin-synthesizing
epidermal cells) into which its dendrites transfer pigment. This structure is known as
an epidermal melanocyte unit. The melanin produced by melanocytes is of two kinds: dark
brown eumelanin and pale red or yellowish phaeomelanin. Both are formed within the
melanocytes by the initial oxidation of the amino acidtyrosine with the aid of
the enzyme tyrosinase; subsequently their synthetic pathways diverge.
An increase in melanin pigmentation may be caused by an increased density of melanocytes, by
abnormal packaging of melanin, or by increased melanin production.
Pigmented birthmarks usually reflect local increases in melanocyte numbers, but in certain rare
congenital pigmentary disorders, such as von Recklinghausen neurofibromatosis, there is
abnormal packaging of melanin within the melanocytes. Pigment production in the skin is
regulated by a pituitary gland peptide hormonecalled melanocyte-stimulating hormone, and
the increase in melanin pigmentation seen with pituitary tumours may reflect overproduction
of this hormone by the pituitary. Both suntans and postinflammatory pigmentation result from
the overproduction of melanin.
The absence of melanocytes, which occurs in vitiligo, results in a loss of melanin pigmentation.
Conditions such as albinism and phenylketonuria are caused by reduced or absent synthesis of
melanin by melanocytes.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3834696/

In the human skin, melanocytes are present in the epidermis and hair follicles. The basic
features of these cells are the ability to melanin production and the origin from neural crest
cells. This last element is important because there are other cells able to produce melanin but
of different embryonic origin (pigmented epithelium of retina, some neurons, adipocytes). The
life cycle of melanocyte consists of several steps including differentiation of melanocyte
lineage/s from neural crest, migration and proliferation of melanoblasts, differentiation of
melanoblasts into melanocytes, proliferation and maturation of melanocytes at the target
places (activity of melanogenic enzymes, melanosome formation and transport to
keratinocytes) and eventual cell death (hair melanocytes). Melanocytes of the epidermis and
hair are cells sharing some common features but in general they form biologically different
populations living in unique niches of the skin.

Melanocytes form a heterogeneous group of cells in the human body. Although all of them
have ability to produce melanin and originate from embryonic cells named neural crest cells
(NCC), their particular functions in all target places are much wider than the melanin synthesis
only [1]. In the human body melanocytes’ presence does not confirm only epidermis, hair and
iris where they give a color of these structures. Melanocytes have been also found in the inner
ear, nervous system, heart and probably it is not the end of a list where these cells exist [2, 3]. It
is necessary to stress that not only melanocytes have ability to produce melanin but also other
cells e.g. cells of pigmented epithelium of retina, epithelia of iris and ciliary body of the eye,
some neurons, adipocytes [4, 5].
The life cycle of melanocytes consists of several steps including lineage specification from
embryonic neural crest cells (melanoblasts), migration and proliferation of melanoblasts,
differentiation of melanoblasts into melanocytes, maturation of melanocytes (melanin
production in special organelles – melanosomes, dendritic morphology), transport of mature
melanosomes to keratinocytes and eventual cell death. Several populations of neural crest cells
(cranial, dorsal trunk, ventral trunk) give melanocytes of the skin. The embryonic origin of
epidermal and hair melanocytes is the same but development is different [6, 7]. The problem of
melanocyte stem cells’ localization in the adult skin is still a matter of debate. The first such
place was a hair bulge, but if only…? [8]. Experimental data clearly show that the trunk NCC
migrating through a ventral pathway could remain in a myelin sheath of the cutaneous nerves
and in particular situations give melanoblasts [9, 10]. The embryonic development of
melanocytes give an opportunity to better understand the skin diseases e.g. melanoma and its
heterogeneity, vitiligo. Thus, in this review the epidermal and hair melanocytes’ biology and
development are characterized.
Melanocytes molecularly are recognizable by identification of melanocyte-specific proteins as
tyrosinase (TYR), tyrosinase-related protein 1 and 2 (TYRP1, TYRP2/DCT), melanosomal matrix
proteins (Pmel17, MART-1), microphthalmia transcription factor (MITF) [1]. The microscopic
analysis indicates that mature melanocytes are oval or fusiform, dendritic cells, smaller than
keratinocytes. In the cytoplasm there are present special membrane-bound organelles
producing melanin, melanosomes [11]. Melanocytes reside in the basal layer of epidermis
where they form the epidermal melanin units as a result of the relationship between one
melanocyte and 30-40 associated keratinocytes [12]. The ratio of melanocytes to keratinocytes
is 1: 10 in the epidermal basal layer (Figure 1). This balance is maintained through the human
live but the exact mechanism is unknown [13]. About 1200 melanocytes exist per mm2 of the
skin independently of the human race [14]. Adhesion molecules such as E- and P-cadherins
participate in building cell-cell contact structures [15]. The contact between the dendritic
processes of differentiated melanocyte and keratinocytes is necessary for the melanin transfer
into keratinocytes determining the skin color and is involved in the skin cells’ photoprotection.
Melanin granules are accumulated above the keratinocyte nucleus and are removed with the
shed epidermal cells [1]. The molecular mechanisms of the melanosomes transfer from
melanocyte to keratinocytes is still a subject of investigations. Recently, Ando et al. proposed a
model of melanosomes’ transport via the shedding vesicle system through the following stages
[16, 17]:

 surrounded by the membrane pigment globules (PG) containing multiple melanosomes


and a few mitochondria are formed in the filopodia of melanocyte dendrites,
 PG are released from different areas of the dendrites into extracellular space,
 PG are captured by microvilli of keratinocytes, which incorporate them in a protease-
activated receptor-2 (PAR-2)-dependent way,
 membrane-surrounded PG is degraded,
 single melanosomes are released in a keratinocyte cytosol and reach the perinuclear
area.
 The size of the epidermal melanin units is similar independently of the human race, but
varies between body areas. Racial distinctions are manifested in an arrangement of the
melanocytic dendrites and in the melanogenesis intensity [18].
 The precise mechanisms that control the organization and number of melanocytes in
the epidermis are unknown. Melanocytes, keratinocytes and dermal fibroblasts
communicate with each other by secreted factors and by cell-cell contacts [19].
Keratinocytes control melanocyte growth and activity through a system of paracrine
growth factors and cell adhesion molecules [13, 20]. Melanocytes and keratinocytes are
also the local source of the different hormones which regulate melanocyte proliferation,
melanogenesis and melanocytic dendrites’ formation [21]. The cross-talking of different
signaling pathways between keratinocytes and melanocytes is a part of an epidermal
complex network involved in the maintenance of skin homeostasis. The basic elements
of keratinocytes’ influence on melanocytes’ biology are illustrated in Figure 2 (based on
[22–24]). In vitro, under ultraviolet radiation (UVR) keratinocytes increase the secretion
of factors that influence many of melanocytes’ biological activities summarized in Table
1 (based on [24, 25]).
Melanocytes’ biology is controlled also by dermal fibroblasts secreted factors, e.g. stem cell
factor (SCF), neuregulin 1 (NRG1) [23, 26]. These cytokines influence not only the growth and
pigmentation of melanocytes, but also their shape, dendricity, mobility and adhesive properties
[23, 27]. In the epidermal melanin unit melanocyte is a very active element that secrets a
number of signal molecules targeting not only keratinocytes but also skin immunological system
cells [28, 29]. The proinflammatory cytokines (IL-1α, IL-2, IL-3, IL-6, IL-10 and TNF-α),
chemokines (IL-8, CCL2), transforming growth factor (TGF-β), catecholamines, eicosanoids,
serotonin, melanocyte stimulating factor (α-MSH) and nitric oxide (NO) are included as factors
released by stimulated melanocytes [28, 29]. Secreted substances act also as autocrine factors,
e.g. IL-1, IL-6 and TNF-α inhibit melanogenesis while under the influence of eicosanoids and α-
MSH the level of melanin synthesis is elevated [30]. Thus, melanocytes and cooperating
keratinocytes form well-organized units in the epidermis. The stable element in each unit is the
melanocyte that lives long, keratinocytes die and are shedding. It is an open question how long
melanocyte lives in the human skin.
Melanocytes are located in the proximal bulb of each hair follicle and also near hair, e.g. in the
sebaceous gland [31]. The bodies of bulbar melanocytes are located at the apex on the dermal
papilla. Melanocyte dendrites enter between the cortical and medullar keratinocytes [32]. The
ratio of melanocytes to keratinocytes is 1: 5, it is more dense than in the epidermis (Figure 3)
[33]. Follicular pigmentation is a result of structural and functional interactions between
follicular melanocytes, matrix keratinocytes and dermal papilla fibroblasts. This tripartite
system is described as the hair melanin unit or follicular melanin unit. The process of hair
pigmentation includes the melanogenic activity of follicular melanocytes, the transfer of
melanin granules into keratinocytes and the formation of pigmented hair shafts [32–34]. It is
considered that a transport of melanin granules to keratinocytes in the growing hair shaft is
similar to the epidermal phagocytosis of melanosomes mediated by receptor PAR2 on
keratinocytes. But differences concern degradation of melanosomes and their quality. Hair
melanocytes, in contrast to epidermal ones, include mainly bigger mature Stage IV
melanosomes (melanogenesis and stages of melanosomes are described further in this work)
and more expanded Golgi apparatus and rough endoplasmatic reticulum (RER). These pigment
cells are larger and more dendritic than epidermal ones [31, 33]. Moreover, in epidermis almost
whole transported melanin is degraded in the differentiating keratinocytes, but in hair cortical
keratinocytes pigment granules are digested only slightly [33]. Diversity of hair color arises
mostly from the quantity and ratio of the brown-black eumelanin and the yellow-red
pheomelanin [35].
Melanin synthesis in the hair occurs under control of products secreted by neighboring cells as
keratinocytes, fibroblasts and endothelial cells, which act through paracrine or autocrine
mechanisms and may be modified by hormonal signals. In pigmentation determining hair color,
the following elements are involved: melanocortin receptor 1 (MCR1) and its α-MSH,
adrenocorticotropic hormone (ACTH), receptor c-Kit and its ligand SCF, endothelins, different
neurotransmitters, cytokines, growth factor and other regulators similar as for epidermal
melanogenesis control [31, 33]. The biochemical pathway of pigment formation and
melanosomes biogenesis run likewise in the epidermis, but it is stressed that hair follicle
melanocytes are more sensitive to aging influences than epidermal melanocytes, what results
in hair greying [33]. It seems that fibroblast of dermal papilla derived factors: insulin growth
factor (IGF-1), keratinocyte growth factor (KGF), noggin, SCF have special significance for
control the hair matrix keratinocyte and melanocyte proliferation and activity during the hair
growth [32, 36]. Epidermal melanocytes are long-living cells, while hair melanocytes die at the
end of the hair cycle which lasts 3-8 years [31]. The melanogenesis process takes place only
during the anagen stage (growing phase) of the hair growth cycle; pigment formation is turned
off in the catagen stage (regressing phase) and absent in the telogen stage (resting phase) [31].
Additionally, it is marked that during catagen completely differentiated bulbar melanocytes die
through apoptosis, but new melanocytes develop from melanoblasts residing in the hair bulge
[8, 32]. Summarizing, survival, proliferation and differentiation of melanocytes are regulated by
microenvironment of the hair follicles.

Melanogenesis
Melanogenesis is a biochemical pathway responsible for melanin synthesis [37]. It takes place in
melanocytes, in separate cytoplasmic organelles called melanosomes [11]. Two major types of
melanin are produced – pheomelanin and eumelanin. They differ in color and the way of
synthesis. Melanin has numerous properties which are beneficial to the body: UV light
absorption and scattering, free radical scavenging, coupled oxidation-reduction reactions and
ion storage [23, 38, 39]. The availability of substrates and the function of melanogenesis
enzymes decide about the types of melanins produced (Figure 4). Tyrosinase (TYR) carries out
tyrosine hydroxylation to L-3,4-dihydroxyphenylalanine (DOPA) which is rapidly oxidized to
DOPAquinone [40]. In the presence of cysteine DOPAquinone react with it, yielding 3- or 5-
cysteinylDOPAs, which then oxidize and polymerize, giving rise to yellow-red soluble melanin –
pheomelanin [37, 41]. In the absence of thiols (cysteine, glutathione or thioredoxin) brown-
black eumelanin is produced. DOPAquinone spontaneously undergoes cyclization to
DOPAchrome [42]. The DOPAchrome spontaneously loses carboxylic acid and generates 5,6-
dihydroxyindole (DHI), which rapidly oxidizes and polymerizes to form dark brown-black,
insoluble DHI-melanin. However, if DOPAchrome tautomerase (TYRP2/DCT) is present,
DOPAchrome will form DHI-2-carboxylic acid (DHICA) [43]. Tyrosinase and TYRP1 catalyze
further conversions obtaining finally a lighter brown color DHICA-melanin [30, 37]. Human skin
contains a mixture of all melanin types, and the ratio of those in part determines visible
pigmentation [19]. Diversity of skin pigmentation among different ethnic groups is preserved
and depends on eumelanin content. The ratio of eumelanin to total melanin decide about skin
color [30]. Pheomelanin does not correlate with skin pigmentation, a similar amount of this
pigment is observed in the dark and light skin. While in hair, the ratio of eumelanin to
pheomelanin decides about the color [35]. Eumelanin comparing to pheomelanin has better
photoprotecting properties – higher resistance to degradation and ability to reactive oxygen
species (ROS) neutralization [44]. Eumelanins are considered to be more effective in terms of
photoprotection than the reddish pheomelanin. As a consequence, the risk of skin cancer in
lighter skin is 30-40-fold higher than in the darker one [41]. Products of genes regulating
melanogenesis act at subcellular, cellular, tissue and environmental levels [21]. During
melanogenesis, as intermediate products, cytotoxic molecules are synthesized (quinones,
hydrogen peroxide). Thus, melanocyte protects itself by separating areas of melanogenesis in
melanosomes and increases the level of antiapoptotic protein Bcl-2 [1, 21].
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3996377/

Human melanocytes are distributed not only in the epidermis and in hair follicles but also in
mucosa, cochlea (ear), iris (eye), and mesencephalon (brain) among other tissues. Melanocytes,
which are derived from the neural crest, are unique in that they produce eu-/pheo-melanin
pigments in unique membrane-bound organelles termed melanosomes, which can be divided
into four stages depending on their degree of maturation. Pigmentation production is
determined by three distinct elements: enzymes involved in melanin synthesis, proteins
required for melanosome structure, and proteins required for their trafficking and distribution.
Many genes are involved in regulating pigmentation at various levels, and mutations in many of
them cause pigmentary disorders, which can be classified into three types: hyperpigmentation
(including melasma), hypopigmentation (including oculocutaneous albinism [OCA]), and mixed
hyper-/hypopigmentation (including dyschromatosis symmetrica hereditaria). We briefly review
vitiligo as a representative of an acquired hypopigmentation disorder.

Pigments that determine human skin colors include melanin, hemoglobin (red), hemosiderin
(brown), carotene (yellow), and bilin (yellow). Among those, melanins play key roles in
determining human skin (and hair) pigmentation. Melanin pigments can be classified into two
major types based on their biosynthetic pathways, as updated and reviewed elsewhere:
eumelanin (dark brown and black) and pheomelanin (yellow, red, and light brown) (Simon et al.
2009; Hearing 2011; Kondo and Hearing 2011). Eu-/pheo-melanin pigments are produced and
deposited in melanosomes, which belong to the LRO (lysosome-related organelle) family in that
they contain acid-dependent hydrolases and lysosomal-associated membrane proteins (Raposo
and Marks 2007). Melanosomes can be divided into four stages depending on their degree of
maturation. Early melanosomes, especially stage I melanosomes, are similar to lysosomes
whereas late melanosomes contain a structured matrix and highly dense melanin deposits.
Studies of melanosomes are not only performed in medicine but also in archaeology because
various morphologies of melanosomes remaining in fossils serve as clues to hypothesize the
colors of dinosaurs (Li et al. 2012).
Melanocytes can be defined as cells that possess the unique capacity to synthesize melanins
within melanosomes. Factors related to melanin production within melanocytes can be divided
into three groups as previously reviewed: structural proteins of melanosomes, enzymes
required for melanin synthesis, and proteins required for melanosome transport and
distribution (Yamaguchi and Hearing 2009). We briefly update the recent findings regarding
pigmentation-related factors.
Disruptions of the functions of many pigmentation-related factors are known to cause
pigmentary disorders and a curated list of those are summarized and updated at the homepage
of the European Society for Pigment Cell Research (www.espcr.org/micemut). Those disorders
include hyperpigmentation, hypopigmentation, and mixed hyper-/hypopigmentation disorders,
which are subdivided into congenital or acquired status (Table 1). Their diagnosis depends on
the size, location (involved site(s) of the body), and morphology (isolated, multiple, map-like,
reticular, or linear) of the lesions. Hypopigmentation disorders are subclassified into those
associated with complete or incomplete depigmentation.

As recently summarized (Kawakami and Fisher 2011; Sommer 2011), melanocytes in the skin
are exclusively derived from the neural crest. Melanocytes used to be thought to derive directly
from neural crest cells migrating via a dorsolateral path (between the ectoderm and
dermamyotome of somites) during embryogenesis, whereas neurons and glial cells were
thought to derive from neural crest cells migrating via a ventral path between the neural tube
and somites. Adameyko et al. (2009) recently challenged this idea and reported that
melanocytes migrate and differentiate from nerve-derived Schwann cell precursors, whose fate
is determined by the loss of Hmx1 homeobox gene function in the ventral path. Schwann cell
precursors detaching from the nerve differentiate into melanocytes, whereas precursors that
stay in contact with nerves eventually differentiate into Schwann cells. Those authors also
showed that Schwann cells remain competent to form melanocytes using Krox20 (early growth
response 2 or Egr2)-Cre loci crossed to YFP reporter strains. They also showed that Neuregulin-
1 (also known as glial growth factor, Heregulin or Neu differentiation factor) regulates the
survival and proliferation of Schwann cell precursors and determines the fate of Schwann cells
and melanocytes depending on high and low expression levels, respectively, and that secreted
signals, including IGF (insulin-like growth factor) and PDGF (platelet-derived growth factor)
enhance melanocyte development (Adameyko et al. 2009). Those findings may explain the facts
that patients with neurofibromatosis type 1, who develop neurofibromas consisting mainly of
Schwann cells, are hyperpigmented, and that segmental vitiligo mostly occurs along with the
affected innervation zones or dermatomes.
Taken together, melanocytes in the skin eventually derive from the neural crest and either
differentiate directly from neural crest cells via a dorsolateral path or derive from Schwann cell
precursors via a ventral path after detaching from the nerve. Various transcription factors,
including Hmx1 and Krox20, act as intrinsic factors that regulate the fate of these cell types,
which are modulated by extrinsic factors including Neuregulin-1, IGF, and PDGF.
Human melanocytes reside not only in the epidermis and in hair follicles but also in mucosa,
cochlea of the ear, iris of the eye, and mesencephalon of the brain as well as other tissues
(Plonka et al. 2009). As far as mouse melanocytes are concerned, Aoki et al. reported that
noncutaneous (ear, eye, and harderian gland) and dermal melanocytes are different from
epidermal melanocytes in that the former do not respond to KIT stimulation but respond well
to ET3 (endothelin 3) or HGF (hepatocyte growth factor) signals (Aoki et al. 2009), suggesting
the heterogeneity of mouse melanocytes. They also reported that noncutaneous or dermal
melanocytes cannot participate in regenerating follicular melanocytes using the hair
reconstitution assay, unlike epidermal melanocytes (Aoki et al. 2011). Studies by Tobin’s group
also support the hypothesis that follicular and epidermal melanocytes in human skin are
different regarding their responses to various biological response modifiers, including αMSH
(Tobin 2011). Additionally, Li et al. (2010) also reported that dermal melanocyte stem cells
derived from glabrous human foreskin (i.e., with no hair follicles) can differentiate into
functional epidermal melanocytes using a three-dimensional skin equivalent model.
These results make us wonder whether human fetal and/or adult melanocytes are
heterogeneous. Human adult melanocytes in skin on the palms and soles may be different from
melanocytes derived from other sites of the skin based on the facts that melanocyte migration
stops at the palms and soles during embryogenesis and that skin on the palms and soles is
hypopigmented and contains a fivefold lower density of melanocytes than at other skin areas.
Additionally, fibroblasts in the dermis of the palms and soles secrete high levels of DKK1
(dickkopf1), which is an inhibitor of the Wnt signaling pathway and suppresses the proliferation
and differentiation of those melanocytes (Yamaguchi et al. 2004). Preliminary results obtained
from human fetal melanocytes cultured from four different body sites (scalp, back, abdomen,
and sole) indicate that palmoplantar melanocytes express high levels of DKK1, TBX4, WIF1,
FGF7, and CHI3L1 (Nakamura et al. 2012). Although the relevance to melanocytes has not been
elucidated, a series of studies from Chang’s group showed that the expression patterns of
homeotic genes (HOX genes, which are expressed in a nested pattern along developmental
axes) determine positional identities within the human body (Chang 2009) and that a long
noncoding RNA, which used to be considered to have nonspecific roles, has site-specificity (Rinn
et al. 2007) and maintains active chromatin to coordinate HOX gene expression (Wang et al.
2011). Additionally, the expression levels of distal-specific HOXA13 are up-regulated in adult
fibroblasts in the skin of paws of mice, thereby inducing the expression of Wnt5A, a morphogen
required for distal development, in fibroblasts and of keratin 9, a distal specific marker of
epidermis, in keratinocytes (Rinn et al. 2008). These results obtained in studies of mice support
the hypothesis that mesenchymal-epithelial interactions play important roles in maintaining the
site-specificity of the skin (Yamaguchi et al. 2005).
Taken together, human fetal and adult melanocytes may be heterogeneous/site-specific
because those melanocytes are also regulated and maintained by site-specific HOX genes,
whose expression patterns are eventually determined by chromatins and long noncoding RNAs.
That melanocyte heterogeneity may be affected both by intrinsic factors, including a site-
specific transcription factor, HOX, and by extrinsic factors secreted by surrounding resident cell
types: fibroblasts and keratinocytes. The fact that acral melanoma is different from other types
of melanoma (Curtin et al. 2005), especially in that AMP kinase-related NUAK2 expression levels
are high in patients with poor prognosis acral melanoma (Namiki et al. 2011), may also support
the idea that melanocytes are heterogeneous.

FACTORS THAT REGULATE MELANIN PRODUCTION WITHIN MELANOCYTES


Pigment-specific factors that modulate melanin production within melanocytes are usually
located within, on, or close to melanosomes and can be divided into three types (Fig. 1):
proteins involved in melanosome structure, proteins that regulate melanin synthesis, and
proteins involved in the intracellular trafficking of melanosome components and the transport
of melanosomes to the cell’s periphery (Yamaguchi and Hearing 2009). Transcription factors
specifically expressed by melanocytes and by melanoma cells (especially MITF) regulate the
expression and function of many of those pigment-specific factors. We briefly update several
important findings since our review published several years ago (Yamaguchi and Hearing 2009).

Melanosomal Structural Proteins


Three important structural proteins that form melanosomes include PMEL17/Silv/GP100,
MART-1 (melanoma antigen recognized by T cell-1), and GPNMB (glycoprotein non-metastatic
melanoma protein b)/DC-HIL/osteoactivin. PMEL17 forms an amyloidogenic fibril depending on
the critical acidic pH (5.0 ± 0.5) within melanosomes (Pfefferkorn et al. 2010). Studying PMEL17
mutations can be useful to investigate the conversion between physiological/benign and
pathological amyloid proteins (Watt et al. 2011). MART-1, which is abundant in early
melanosomes, is required for the maturation of PMEL17 (Hoashi et al. 2005). GPNMB, which is
highly homologous to Pmel17 but lacks the RPT domain (imperfect repeats of proline, serine,
and threonine-rich motifs), is a melanosome-specific and proteolytically released protein, which
is abundant in late melanosomes (Hoashi et al. 2010). GPNMB is critical for the formation of
melanosomes in a MITF-independent fashion (Zhang et al. 2012). These structural melanosomal
proteins provide scaffold materials to the enzymes required for melanin deposition and on
which the melanins are deposited.

Enzymes Involved in Melanin Synthesis


There are three key enzymes that play critical roles in melanin synthesis within melanosomes:
TYR (tyrosinase), TYRP1 (tyrosinase related protein-1), and TRP2/DCT (dopachrome
tautomerase). Many factors, including BLOC-1, OA1, P, and SLC45A2 (previously known as
MATP) influence the trafficking and thus the function of these enzymes. Of note, metal ions
including zinc and copper serve as catalysts and/or chelators when melanin is synthesized
(Simon et al. 2009) and metal remnants in fossils enable us to predict the colors of dinosaurs
(Wogelius et al. 2011), in addition to determining the configurations of melanosomes (Li et al.
2012). Cystinosin, a cysteine/H+ symporter that exports cysteine out of lysosomes, is the gene
associated with cystinosis, a rare autosomal recessive disorder with multiple organ dysfunctions
including hypopigmentation (Chiaverini et al. 2012). Additionally, NAD(P)H:quinone
oxidoreductase-1 enhances melanogenesis by increasing the levels of TYR protein (Choi et al.
2010). The regulation of intramelanosomal pH may play important roles for regulating the
appropriate enzymatic functions in melanin synthesis as well as the processing and function of
melanosomal structural proteins.

Melanosome Trafficking Proteins


Melanin granules are transported from the perinuclear area to the periphery of melanocytes
and are eventually transferred to adjacent keratinocytes (Yamaguchi and Hearing 2009). Early
melanosomes, produced via the trans-Golgi network and/or endocytosis, originate in the
perinuclear area and then mature to late (pigmented) melanosomes as they move toward the
periphery of the melanocyte (i.e., the dendrites). In this trafficking pathway, kinesin (prograde)
and dynein (retrograde) act like wheels for melanosomal cargo and microtubules function like
rails. The clathrin adaptor AP-1 is reported to interact with the kinesin motor KIF13A,
suggesting the role of adaptor proteins in melanosome trafficking (Delevoye et al. 2009). The
melanosomal cargo is transferred from microtubules to F-actin, which acts as a rail, at the
periphery of the melanocyte. Semi-automated analysis of organelle movement and membrane
content reveals the involvement of Rab27a and its complex in the regulation of this transfer
(Hume et al. 2011). Rab27a, melanophilin, and myosin Va are connected to melanosomes in
that order and function like wheels. Finally, Slp2-a may regulate melanosomal cargo exocytosis.
Recent findings show that melanoregulin regulates retrograde melanosome transport via the
interaction with RILP (Rab-interacting lysosomal protein) and its complex (dynactin subunit 1)
(Ohbayashi et al. 2012). Ciliobrevins were discovered as small-molecule inhibitors of the
AAA+ (ATPases associated with diverse cellular activities) ATPase dynein (Firestone et al. 2012).
Studies further elucidating melanosome trafficking might be enhanced in the near future
characterizing these factors.

Melanocyte-Specific Transcription Factors


MITF has been investigated intensively among the many transcription factors known to regulate
melanocyte function. MITF itself is regulated by many other transcription factors, including
PAX3 (a neural-crest-associated transcription factor), SOX9, SOX10, LEF-1/TCF (a downstream
regulator of Wnt signaling pathway), and CREB (cAMP responsive-element-binding protein,
which is phosphorylated by signals via MC1R, melanocortin-1 receptor) (Yamaguchi and Hearing
2009).
Fisher’s group recently reported that MITF directly up-regulates DICER, an endoribonuclease in
the RNase III family that cleaves double-stranded RNA and pre-microRNA into short RNA
fragments (20–25 nucleotides long), on melanocyte differentiation (Levy et al. 2010). Enhanced
DICER expression plays a crucial role in melanocyte survival via the posttranscriptional
processing of the pre-microRNA-17 ∼ 92 cluster, which leads to the down-regulation of BIM, a
proapoptotic factor.
Go to:

PIGMENTARY DISORDERS
As summarized above, the regulation of pigmentation involves many factors required for
development, heterogeneity, regeneration, and senescence of melanocytes and their
precursors, as well as those involved in determining melanosome structure, melanin synthesis,
the trafficking of melanosomal components and the transport and distribution of
melanosomes, and melanocyte-specific transcription factors that control the expression and
function of all those genes. More than 350 loci are currently known to be directly or indirectly
involved with those processes in mice and mutations of many of those genes have been
associated with human pigmentary disorders. Those include hyperpigmentation,
hypopigmentation, and mixed hyper-/hypopigmentation disorders and can be diagnosed by
size (systemic or localized), comorbidities, site of the involvement, and patterns/shapes of the
lesions (Table 1). Those are subclassified into congenital and acquired disorders and in addition
hypopigmentation disorders can also be subclassified into complete and incomplete
depigmentation. Among the many pigmentary disorders summarized in Table 1, we focus on
vitiligo because of space limitations.

Congenital Hyperpigmentation Disorders


Congenital hyperpigmentation disorders include those involving epidermal hyperpigmentation
(nevus cell nevus, Spitz nevus, and nevus spilus), dermal hyperpigmentation (blue nevus, nevus
Ohta, dermal melanosis, nevus Ito, and Mongolian spot), ephelides, acropigmentatio reticularis,
Spitzenpigment/acropigmentation, and lentiginosis (generalized lentiginosis, LEOPARD
syndrome, inherited patterned lentiginosis, Carney complex, Peutz–Jeghers syndrome, Laugier–
Hunziker–Baran syndrome, and Cronkhite–Canada syndrome).
The responsible locus for generalized lentiginosis, characterized by widespread lentigines
without systemic involvement, has been localized to chromosome 4q21.1-q22.3 (Xing et al.
2005). LEOPARD syndrome is characterized by multiple lentigines, congenital cardiac
abnormalities, ocular hypertelorism, and retardation of growth, and many reports have shown
its association with mutations in the PTPN11 (protein tyrosine phosphatase, nonreceptor type
11) gene located at chromosome 12q24 since Legius et al. (2002)first reported the association
with Noonan syndrome. Carney complex, a multiple neoplasia syndrome characterized by
spotty skin pigmentation, cardiac and other myxomas, endocrine tumors, and psammomatous
melanotic schwannomas, has been shown to be caused by mutations in PRKAR1A (protein
kinase A regulatory subunit 1α), a tumor-suppressor gene (Kirschner et al. 2000). Peutz-Jeghers
syndrome, which predisposes to benign and malignant tumors of many organ systems, has
been reported to be associated with mutations in STK11 (serine/threonine protein kinase)/LKB1
(Hemminki et al. 1998).

Acquired Hyperpigmentation Disorders


Acquired hyperpigmentation disorders include senile lentigines/lentigo, melasma/chloasma,
Riehl’s melanosis, labial melanotic macule, penile/vulvovaginal melanosis, erythromelanosis
follicularis faciei Kitamura, UV-induced pigmentation (tanning and pigmentation petaloides
actinica), postinflammatory pigmentation (friction melanosis and ashy dermatosis),
chemical/drug-induced pigmentation (polychlorinated biphenyl, arsenic, 5-FU, bleomycin,
cyclophosphamide, methotrexate, chlorpromazine, phenytoin, tetracycline, and chloroquine),
pigmentary demarcation lines, foreign material deposition (such as carotene, silver, gold,
mercury, bismuth, and tattoos). Hyperpigmentation related with systemic disorders includes
metabolism/enzyme disorders (hemochromatosis, Wilson’s disease, Gaucher’s disease,
Niemann–Pick’s disease, amyloidosis, ochronosis, acanthosis nigricans, and porphyria cutanea
tarda), endocrine disorders (Addison’s disease, Cushing syndrome, and hyperthyroidism),
nutritional disorders (pellagra, vitamin B12 deficiency, folic acid deficiency, vagabond’s disease,
and prurigo pigmentosa), mastocytosis, collagen diseases, liver dysfunction, and kidney
dysfunction. Hyperpigmentation can also be related with infectious diseases (measles, syphilis,
and Malassezia furfur) and syndromes (von Recklinghausen’s disease, Sotos syndrome, POEMS
syndrome, Naegeli syndrome, Cantu syndrome, McCune–Albright syndrome, Watson
syndrome, and Bloom syndrome).
Bioinformatics studies have shown that genes responsible for melasma include Wnt and lipid
metabolism-related genes in addition to melanogenic markers (Kang et al. 2011) and that
reduced expression levels of WIF-1 (Wnt inhibitory factor-1) in keratinocytes and/or fibroblasts
may play roles in the development of melasma (Kim et al. 2013). Patients with mastocytosis
usually carry the D816 V KIT mutation and a bioinformatics study shows that those patients
show the up-regulation of genes involved in innate and inflammatory immune responses,
including interferon-induced genes and genes involved in cellular responses to viral antigens,
together with complement inhibitory molecules and genes involved in lipid metabolism and
protein processing. Aggressive mastocytosis additionally shows deregulation of apoptosis and
cell cycle-related genes, whereas patients with indolent mastocytosis display increased
expression levels of adhesion-related molecules (Teodosio et al. 2013). Sotos syndrome,
characterized by childhood overgrowth with advanced bone age, craniofacial dysmorphic
features including macrocephaly and learning difficulties, results from the haploinsufficiency of
NSD1 (nuclear receptor binding SET domain protein 1) (Kurotaki et al. 2002). POEMS syndrome
(polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes) is a
multisystem disorder associated with plasma cell dyscrasia, and patients with POEMS syndrome
show up-regulated serum levels of angiogenetic factors including VEGF (vascular endothelial
growth factor) and HGF (hepatocyte growth factor) (Yamada et al. 2013). Cantu syndrome,
characterized by hypertrichosis, macrosomia, osteochondrodysplasia, and cardiomegaly, is
reported to be caused by mutations in ABCC9 (ATP-binding cassette, subfamily C, member 9)
(van Bon et al. 2012). McCune–Albright syndrome results from somatic mutations of the GNAS
gene (G-protein α-subunit) especially mutations in Gsα (stimulatory G protein) (Dumitrescu and
Collins 2008). The causative gene for Bloom syndrome, with photosensitivity and increased risk
of malignancy, is BLM (Bloom syndrome, RecQ helicase-like), localized at chromosome 15q26.1
(German et al. 1994).

Congenital Hypopigmentation Disorders


Congenital hypopigmentation disorders include various types of oculocutaneous albinism
(OCA1–4, Hermansky–Pudlak syndrome, Chediak–Higashi syndrome, and Griscelli syndrome),
Cross–McKusick–Breen syndrome, phenylketonuria, piebaldism, Waardenburg syndrome,
nevus depigmentosus, hypomelanosis of Ito, Hirschprung’s disease, Charcot–Marie–Tooth
disease, Menkes disease, and Wilson disease.
Many of the genes responsible for those hypopigmentary disorders have been identified and in
general are involved with the intracellular trafficking of proteins to LROs (including
melanosomes), in the transport of organelles to the peripheries of the cell and in their transfer
to surrounding keratinocytes. Figure 2 outlines the diagnostic decision tree for representative
congenital hypopigmentation disorders, and we recommend that interested readers check the
many references shown in the ESPCR webpage for further details and updates of genes
associated with congenital hypopigmentary diseases. As recently reviewed, Hermansky-Pudlak
syndrome, characterized by oculocutaneous albinism, prolonged bleeding times, and
pulmonary fibrosis, is triggered by syndromic dysgenesis of specialized LROs including
melanosomes, platelet granules, synaptic vesicles, lytic granules, Azurophil granules, and
lamellar bodies (Wei and Li 2013). Its responsible dysfunctional proteins include the biogenesis
of lysosome-related organelles complex 1 (BLOC-1), BLOC-2, BLOC-3, and AP-3. For example,
the responsible gene for Hermansky-Pudlak syndrome type 7 is DTNBP1 (dysbindin), a
component of BLOC-1 (Li et al. 2003). Hypomelanosis of Ito may be derived from duplication of
Xp11.3-p11.4 and random X-inactivation (Zou and Milunsky 2009).
https://www.derm101.com/inflammatory/embryologic-histologic-and-anatomic-
aspects/melanocytes/

EMBRYOLOGIC, HISTOLOGIC, AND ANATOMIC ASPECTS


Melanocytes

Melanocytes synthesize melanin. From precursors in the neural crest, melanocytes migrate
normally to the epidermis, the bulb of hair follicles, epithelia of various mucous membranes,
leptomeninges, inner ear, and a few other tissues (Fig. 1.61). Primitive melanocytes first appear
in the skin during the eighth week of fetal life, but at that time they produce little melanin. Only
in postnatal life do normal melanocytes begin to function fully, except in certain cutaneous loci,
such as nipples and genitalia, and in the bulb of follicles, where in the fetus they make melanin
copiously.

Figure 1.61

Melanocytes (black dots) are derived from primordial neural crest and migrate especially to
skin, mucous membranes, ocular structures, and leptomeninges.

When scrutinized through a conventional microscope, melanocytes appear as “clear cells” in


and immediately beneath the row of epidermal basal cells, that is, the dermoepidermal
junction (Fig. 1.62). The space characteristic of melanocytes, and for which they were named
“clear cells” by Masson, actually is an artifact of fixation during which cytoplasm shrinks and
becomes concentrated around the nucleus, thereby creating a lacuna. In fact, melanocytes are
not clear cells; they possess abundant translucent cytoplasm. The nucleus of a melanocyte is
smaller and more deeply basophilic than that of a basal keratocyte, and the cytoplasm of it is
dendritic, unlike that of a keratocyte (Fig. 1.63). Dendrites of melanocytes are revealed more
effectively with silver salts that stain melanin black; they then are seen to arborize in all
directions among neighboring keratocytes and even extend into the uppermost part of the
dermis (Fig. 1.64). It has been estimated that in this manner a single melanocyte is connected
to about 36 keratocytes, a relationship that has been termed, imprecisely, “the epidermal
melanin unit.” The unit really consists of a melanocyte and keratocytes that are supplied by it.
In sections stained by hematoxylin and eosin, the average ratio of melanocytes to basal
keratocytes is about 1 to 10, but the concentration of melanocytes varies according to the
region of the body, being more numerous in the skin of the face (up to 2,900 melanocytes per
mm2) than on the trunk (up to 1,250 melanocytes per mm2).

Figure 1.62

Epidermal melanocytes, when viewed through a conventional microscope, appear as “clear


cells” in and immediately beneath the basal layer. Vacuity of melanocytes is an artifact of
fixation caused by the collapse of cytoplasm around the nucleus. A tad of stellate-shaped
cytoplasm still can be seen attached to the nucleus. Note that nuclei of melanocytes are smaller
and darker than those of contiguous keratocytes. (x1350)

Figure 1.63

Dendrites of melanocytes are exaggerated in a split-skin preparation from a rhesus monkey that
was irradiated for 28 days prior to biopsy. (Dopa stain.) (Courtesy of William Montagna, Ph.D.)

Figure 1.64

Dendrites of a melanocyte extend in all directions, especially along the basal layer and between
keratocytes in the spinous zone. Once melanin has been formed, it is transferred from
melanocytes to keratocytes by a process called apocopation.
Melanosomes within the cytoplasm of melanocytes are factories for manufacture of melanin
and warehouses for storage of it (Fig. 1.65). They are spherical or ellipsoid membrane-bound
particles with a highly organized internal structure composed of longitudinally oriented
concentric lamellae that possess a characteristic periodicity (Figs. 1.66 and 1.67). Once
melanosomes are formed, melanized, and transported to the tip of dendrites, they are
transferred to keratocytes by apocopation, i.e., a process in which the tip of dendrites is
snipped off and engulfed by keratocytes. That phenomenon enables collections of
melanosomes to be housed in epidermal and follicular keratocytes, and there to serve their
purpose. In the epidermis, melanosomes become concentrated in umbrella-like array above the
nucleus of keratocytes, on the side of a nucleus closest to the surface of the skin. After transfer
to keratocytes, fully melanized melanosomes are conveyed outward as those keratocytes
mature, eventually becoming degraded by lysosomal enzymes and shed along with
desquamated corneocytes.

Figure 1.65

A melanocyte contains melanosomes but no tonofibrils. (x17,000) (Courtesy of Ken Hashimoto,


M.D.)

Figure 1.66

In a melanocyte, spherical and elliptical membrane-bound melanosomes are present at various


stages of development. (Courtesy of Alvin Zelickson, M.D.)
Figure 1.67

Melanosomes in a melanocyte have a distinctive structure formed of concentric lamellae with


characteristic periodicity. (x929,000) (Courtesy of Alvin Zelickson, M.D.)

Because the absolute number of melanocytes in human skin is approximately the same for both
sexes and all races, the amount of melanin in keratocytes and the distribution of it determines
the degree of pigmentation of skin. Differences in color among the races result from differences
in the number, size, degree of melanization, distribution, and rate of degradation of
melanosomes within keratocytes. In dark-skinned peoples, melanosomes within keratocytes
are numerous, large, melanized markedly, distributed as solitary units, and degraded slowly. In
addition, the epidermis of dark-skinned peoples contains melanocytes whose dendrites are
larger and more elongated. In contrast, melanosomes within epidermal keratocytes of light-
skinned peoples are fewer, smaller, less abundantly melanized, and distributed as aggregates
within phagosomes where they are degraded more rapidly; dendrites are shorter. When pale
skin is exposed to ultraviolet light, however, melanocytes increase in number and size, and
become more strikingly dendritic. Radiation by ultraviolet light accelerates synthesis,
melanization, and transfer of melanosomes to keratocytes. As melanosomes become larger and
melanized more copiously, melanin is synthesized actively. This series of events results in
tanning. Melanocytes situated in the bulb of anagen follicles impart to hair various colors, those
who bear them being known, colloquially, as blonds, brunettes, and redheads.

Aging is accompanied by a decrease in number and decline in activity of melanocytes


positioned in follicular bulbs, the result being progressive graying of hair. Although epidermal
melanocytes also decline in number with age, skin that has been exposed extensively and
repeatedly to ultraviolet light is marked by greater density of melanocytes than regions that
have been spared such exposure. Unlike the situation in an incipient stage of melanoma,
however, the melanocytes stationed at the dermoepidermal junction of skin damaged severely
by the effects of ultraviolet light, that being evidenced by an extraordinary amount of solar
elastosis (elastotic material), are equidistant from one another and monomorphous.

The principal function of melanin is to protect the skin from harmful effects of sunlight, a task
accomplished by its capability to scatter and absorb ultraviolet light.
https://www.sciencedirect.com/topics/neuroscience/melanocyte

Melanocyte
A melanocyte is a neuroectodermally derived dendritic cell that contains the intracellular
apparatus to manufacture melanin.

Melanocytes or their precursors melanoblasts are derived from the neural crest cells. After
extensive migration, they finally reside in the skin, inner ear, and uveal tract as highly dendritic,
heavily pigmented cells; they are generally located in the epidermal basal cell layer of these
areas, including hair follicles.1Genetic observations indicate that steel factor (SLF, also known as
MGF or stem cell factor)2 and endothelin 3 (ET3)3 are essential environmental factors for the
early stages of melanocytedevelopment.
Melanocytes and Pigmentation
Melanocytes are specialized cells that are distributed in the skin, other epithelial surfaces and
the eye (1). In the skin, melanocytesare typically distributed at infrequent but regular intervals
along the basal layer of the epidermis (Figure 36-1) and in hair follicles (2). A primary function
of melanocytes is the distribution of packages of the pigment melanin to neighboring
keratinocytes. Distribution of pigment is accomplished through the transfer of melanosomes, a
unique organelle where the chemical steps in melanin biosynthesis occur (3). These structures
are then transferred from the ends of the dendritic processes of melanocytes to adjacent
keratinocytes. Melanocytes in the skin are generally long-lived, nonproliferative cells that
constitutively express the anti-apoptotic molecule BCL2, but are able to proliferate in response
to inflammation during tissue damage or following excessive solar radiation.

Melanocytes
Melanocytes are neural crest-derived cells that reside in the stratum basale at the frequency of
1 in every 10 basal keratinocytes. Certain areas of the body including the face, shins, and
genitalia have an even greater density of melanocytes. In these areas the frequency approaches
1:1 [4]. The principal role of the melanocyte is in making melanosomes. Melanocytestransfer
melanosomes to keratinocytes by way of cytocrine secretion. Melanosomes are elongated,
membrane-bound, pigment-producing granules within the keratinocytes that determine
differences in skin color [12].

Melanocytes are dendritic cells with cellular processes, or dendrites that allow them to contact
many keratinocytes, over long distances. While theircell bodies may be between the stratum
basale and basement membrane, their dendritic processes extend to reach many keratinocytes
throughout the stratum basale and stratum spinosum [19]. Together, keratinocytes
and melanocytes form a melanin unit. Melanocytesproduce melanosomes in the golgi zone of
the cell. The melanosomes are then moved to the tips of their cellular processes. Keratinocytes
then ingest the tips of the melanocytic dendrites by phagocytosis, allowing them to take in the
melanosomes. This process is called apocopation [13]. Melanin granules then form a protective
cap over the nucleus of the keratinocyte protecting the nucleus from the photo damage of UV
light [4]. Tyrosinase acts on a variety of melanin precursors, including tyrosine, to make
melanin [10]. The melanocortin 1 receptor plays a prominent role in melanin production [4].

The number of melanocytes does not determine skin color. Instead, the number, size, and
distribution of the melanosomes determine skin color. People with pale skin have fewer
melanosomes that are packaged in membrane-bound complexes. People with darker skin have
a greater number of melanosomes that are larger and more widely distributed. Chronic sun
exposure stimulates melanocytes to make melanosomes in a pattern similar to what is found in
people with darker skin. In addition, melanocytes in people with red hair tend to be more
round and produce more phaeomelanin. Leukoderma, or diseases that cause whitening of the
skin, can be caused by a decrease in the number of melanocytes [4]. Vitiligo is a result of
autoimmune destruction of melanocytes. Albinism, on the other hand, is caused by a deficiency
of fully pigmented melanosomes. Increases in pigment can be caused by a variety of different
factors [12]. Freckles result when a normal number of melanocytes increase their production of
pigment. Black “sunburn” or “ink spots” result from basilar hyperpigmentation and increased
melanin content in the stratum corneum. Lastly, nevi are the result of benign proliferations
in melanocytes, while melanomas are the result of malignancies [4].
https://biologiedelapeau.fr/spip.php?article7

MELANOCYTES

Normal skin pigmentation is a complex process that, in the epidermis as in the hair follicles,
begins with the synthesis of melanin within melanosomes in the melanocytes, followed by
melanosome transfer to neighboring basal and suprabasal keratinocytes. In basal cells,
melanin granules are translocated to the upper pole of the nucleus, forming a melanin cap
that protect DNA from UVrays. Melanin granules are eventually degraded as the keratinocyte
undergoes terminal differentiation.

In humans, the melanocytes are localised either in the basal layers of the epidermis or in the
hair follicles. Whatever is their localisation in skin, the melanocytes are derived from
precursor cells (called melanoblasts) that originate from the neural crest.

Mammalian melanocytes produce in their melanosomes, two chemically distinct types of


melanin pigments: black-brown eumelanin and yellow-reddish pheomelanin. In the
melanocytes, eumelanosomes and pheomelanosomes cohabit.

Tyrosinase is the key-enzyme which regulates the first steps of eumelanin and pheomelanin
synthesis: the transformation of L-tyrosines into L-3,4 dihydroxyphénylalanine (DOPAs) and
then into DOPAquinones. From DOPAquinones, the synthesis pathways are different for the
pheomelanin or eumelanin.

In human, as in the others mammals, the skin and hair color is mainly determined by the
number, the size, the type, and the mode of repartition of the melanosomes. It is particularly
interesting to note that in normal conditions, the racial differences in skin pigmentation in
human do not depend of the melanocyte number in the epidermis. For a specific area, the
number of epidermal melanocytes is nearly the same in Caucasoids, Negroids and
Mongoloids.

Normal human skin color ranges from white to brown to black and results from the mixed
contribution of four pigments: oxygenated hemoglobin (red) in the capillaries, reduced
hemoglobin (blue) in the venules of the dermis, exogenously produced carotenoids (yellow) and
endogenously produced melanin (brown). However, it is the amount and type of melanin
pigment produced in melanosomes by cutaneous and follicular melanocytes which largely
determined the differences in skin and hair color between individuals.

Normal skin pigmentation is a complex process that, in the epidermis as in the hair follicles,
begins with the synthesis of melanin within melanosomes in the melanocytes, followed by
melanosome transfer to neighboring basal and suprabasal keratinocytes. In basal cells, melanin
granules are translocated to the upper pole of the nucleus, forming a melanin cap that protect
DNA from UV rays. Melanin granules are eventually degraded as the keratinocyte undergoes
terminal differentiation.

Melanocytes are specialized cells of the epidermis that produce the pigment melanin. There are
approximately 2000 or more melanocytes per square millimeter in the exposed skin of the
head, in the skin of the scrotum or in the foreskin and 1,000 to 1,500 melanocytes per square
millimeter on the rest of the body in Caucasoids, Negroids and Mongoloids. Accordingly, racial
differences in pigmentation are not due to a difference in the number of melanocytes.

Epidermal melanocytes are regularly dispersed, at an approximate ratio of 1:10, among basal
keratinocytes and distribute the melanin they produce to around 40 overlying suprabasal
keratinocytes via their elongated dendrites and cell/cell contacts. The anatomical relationship
between keratinocytes and melanocytes is known as “the epidermal melanin unit”. The
secretory melanocytes in hair follicles are associated with cells of the hair matrix in the same
way that the epidermal melanocytes are associated with the epidermal keratinocytes.

The epidermal melanocyte activity is continuous while the melanocytes of the hair follicle
follow its rhytmical activity; they are active during the anagen phase of growth and have no
detectable tyrosinase activity during the resting (telogen) phase.
Melanocyte proliferation occurs in non stimulated, unexposed human skin and is increased
after UV irradiation. In hair follicle, melanocyte proliferation and melanin synthesis are both
synchronized with hair growth.

Melanocytes are detected in histological sections by ammoniacal silver nitrate reaction of


Fontana-Masson which stains melanins and by DOPA reaction which revealed tyrosinase
activity. They are stained in immunohistology with various antibodies against melanocyte
antigens, such as tyrosinase, pigmentation associated protein and MART1/Melan-A antigen.
Embryonic melanocytes, hair bulb melanocytes and activated (but not resting) melanocytes of
adult epidermis express the premelanosome-associated glycoprotein gp100 recognised by the
monoclonal antibody HMB-45.

Ultrastructurally, they have an electron-lucent cytoplasm containing no tonofilaments and


no desmosomes but containing loose intermediate filaments (vimentin) and specific organelles,
the melanosomes at various maturation stages.

The melanocytes are derived from precursor cells (called melanoblasts) during embryological
development, and melanoblasts destined for the skin originate from the neural crest beginning
in the second month of human embryonic life. The melanoblasts are large round or oval cells
which differentiate into melanocytes by becoming dendritic and DOPA oxydase-positive. They
reach dermis between the 10thand the 12th week of development and 2 weeks later for the
epidermis and differentiate into melanocytes; melanocytes are already established at
epidermal-dermal junction sites at about the sixth month of fetal life. The melanocytes colonise
the dermis and the epidermis before the initial stage of hair production and distribute into the
hair without a specific localisation. It is only after the sixth month of gestation, that, in the hair,
the melanocytes will be strictly localised in the infundibulum and in the hair matrix. Dermal
melanocytes decrease in number during gestation and virtually disappear by birth, whereas
epidermal melanocytes established at the epidermal-dermal junction continue to proliferate
and start to produce melanosomes in which melanin is synthesized.

MELANOSOMES

Melanosomes are specific melanin-producing intracellular organelles that share several


features with lysosomes in that they contain acid dependent hydrolases and lysosomal-
associated membrane proteins (LAMPs). They belong to a family of cell-specific organelles,
termed lysosome-related organelles , which also include lytic granules observed in cytotoxic T
lymphocytes and natural killer cells, MHC class II compartments observed in antigen presenting
cells, platelet-dense granules, basophil granules, azurophil granules observed in neutrophils and
Weibel-Palade bodies observed in endothelial cells. The main structural component of
melanosomes is Pmel17/gp100/Silv.

Melanosomes result from the fusion of vesicles that contain tyrosinase (Tyr), dopachrome
tautomerase (Dct/Tyrp2) and DHCI oxydase (Tyrp1) and derive from Golgi apparatus with
vesicles that contain the structural components of melanosomes and originate from granular
endoplasmic reticulum. The trafficking of Tyr to melanosomes can be affected dramatically by
mutations in several melanogenic genes and by changes of intracellular pH that result in
oculocutaneous albinism or hypopigmentation, respectively.

2.1 Structure of melanosomes

The melanosome structure is different according to the type of melanin produced;


pheomelanosomes produce pheomelanin, remain round and contain vesiculoglobular matrix;
eumelanosomes produce eumelanin, are elliptical and have a fibrillar matrix.

Both types of melanosomes are typically divided into four maturation stages (I–IV) determined
by their structure and the quantity, quality, and arrangement of the melanin produced. Stage I
melanosomes have an early matrix organization, are spherical, do not contain tyrosinase
activity, and are localised at the periphery of the nucleus. Stage II melanosomes are tyrosinase-
containing elongated (for eumelanosomes) organelles with an organized filamentous matrix
and with no melanin deposition in eumelanosomes and already formed melanin and a vesiculo-
globular matrix in phaeomelanosomes.The production of the internal matrix fibers and the
maturation from stage I to stage II appear dependent of the presence of a structural protein
named Pmel17 (or gp 100/Silver). Just after its transfer into the stage I melanosome, Pmel17 is
cleaved into several fragments which constitute the fibrillar matrix of the organite. Another
protein, Mart-1, also known as Melan-A, localised in stage I and/or II, contributes to the
melanosome formation since it is necessary to the expression, the stability and the maturation
of Pmel17 and therefore to the critical step of the stage II melanosome formation.

In type III melanosomes, melanin is uniformly deposited. Type IV melanosomes are


melanosomes that are electron-opaque, fully melanized and have a low tyrosinase activity; they
are the melanosomes supplied from the dendrites to the neighboring keratinocytes.

2.2 The enzymes of pigmentation

Within melanosomes, at least three key-enzymes, named tyrosinase, Tyrosinase-related protein


1 (Tyrp1), and Tyrosinase-related protein 2 (Tyrp2/Dct) are absolutely required for the synthesis
of different types of melanin.

Tyrosinase is responsible for the critical initial rate limiting steps of melanogenesis. It is
synthetised as an inactive precursor that is activated when melanocytes are stimulated by
alpha-MSH via AMPc. The Agouti protein is an alpha-MSH antagonist. It blocks the alpha-MSH
fixation to its receptor and, therefore, the receptor activation and stabilises the inactive form of
the receptor. Tyrosinase is dependent upon the incorporation of copper ion to catalyze the
hydroxylation of tyrosine to β-3,4-dihydroxyphenylalanine (DOPA) and the subsequent
oxidation of DOPA to DOPAquinone. It is also involved downstream in the oxydation of 5,6-
dihydroxyindole (DHI) to indole-5,6-quinone.

Tyrp1 and Tyrp2/Dct have distinct catalytic functions in melanin synthesis downstream of Tyr.
The isomerisation of DOPAchrome (indolene-2-carboxylic acid-5,6-quinone) to 5,6-
dihydroxyindole-2-carboxylic acid (DHICA) is catalysed by DOPAchrome tautomerase
(Tyrp2/Dct) and the oxydation of DHICA is catalysed by DHICA-oxydase (Tyrp1). In addition to
having enzymatic functions, Tyrp1 and Tyrp2/Dct also stabilize Tyr. Tyrp1 is also a critical
enzyme for the correct trafficking of tyrosinase to melanosomes and Tyrp2/Dct seems to also
play an important role in detoxification processes within melanosomes.

2.3 Transport and transfer of melanosomes

During their maturation, melanosomes move from the perinuclear area of melanocytes where
they are produced towards the extremity of dendrites as they become more melanized. This
microtubule-dependent intracellular transport involves microtubules, dynein and kinesin, actin
filaments, Rab27a, melanophilin, myosin Va and Slp2-a. Melanin synthesized in melanosomes
within melanocytes is transferred to keratinocytes through the protease-activated receptor,
PAR-2.

MELANIN
Mammalian melanocytes produce two chemically distinct types of melanin pigments: black-
brown eumelanin and yellow-reddish pheomelanin .

Eumelanin is a highly heterogeneous polymer consisting of DHI (DiHydroxyIndole) and DHICA


((DiHydroxyIndole Carboxylic Acid) units in reduced or oxidized states. Pheomelanin consists
mainly of sulfur-containing benzothiazine derivatives. Melanins are synthesized from tyrosine
exogenously supplied by blood. Tyrosines are oxidized by tyrosinase and metabolised into
DOPAs and then into DOPAquinones which are automatically oxidized into indole compounds.
Indole compounds connect to each other to produce eumelanins.

The pheomelanin synthesis pathway involves sulfur compounds, the amino acid cystein or
glutathion that liberates cysteins through the action of a glutamyl-transpeptidase. In presence
of cysteins, DOPAquinones connect with cysteins to form 5-S-cysteinyl DOPA and 2-S-
cysteinylDOPA which give benzothiazin intermediates that polymerise to produce
pheomelanins.
The major role of melanins is to protect skin from the harmful effects of UV rays and to prevent
skin cancer. Besides this, both eumelanin and pheomelanin play an important protective role
within melanocytes and keratinocytes due to their ability to bind cations, anions, drugs, and
chemicals.
https://en.wikipedia.org/wiki/Melanocyte

Melanocytes are melanin-producing neural-crest derived[3] cells located in the bottom layer
(the stratum basale) of the skin's epidermis, the middle layer of the eye (the uvea),[4] the inner
ear,[5] meninges,[6]bones,[7] and heart.[8] Melanin is the pigment primarily responsible for skin
color. Once synthesised, melanin is contained in a special organelle called a melanosome and
moved along arm-like structures called dendrites, so as to reach the keratinocytes.

Melanogenesis[edit]
Through a process called melanogenesis, melanocytes produce melanin, which is a pigment
found in the skin, eyes, and hair. This melanogenesis leads to a long-lasting pigmentation,
which is in contrast to the pigmentation that originates from oxidation of already-existing
melanin.
There are both basal and activated levels of melanogenesis; in general, lighter-skinned people
have low basal levels of melanogenesis. Exposure to UV-B radiation causes an increased
melanogenesis. The purpose of the melanogenesis is to protect the hypodermis, the layer
under the skin, from the UV-B light that can damage it (DNA photodamage). The color of the
melanin is black, allowing it to absorb a majority of the UV-B light and block it from passing
through this skin layer.[9]
Since the action spectrum of sunburn and melanogenesis are virtually identical, they are
assumed to be induced by the same mechanism.[10]The agreement of the action spectrum with
the absorption spectrum of DNA points towards the formation of cyclobutane pyrimidine
dimers(CPDs) - direct DNA damage.
Typically, between 1000 and 2000 melanocytes per square millimeter of skin are found.
Melanocytes comprise from 5% to 10% of the cells in the basal layer of epidermis. Although
their size can vary, melanocytes are typically 7 μm in length.
The difference in skin color between lightly and darkly pigmented individuals is due not to the
number (quantity) of melanocytes in their skin, but to the melanocytes' level of activity
(quantity and relative amounts of eumelanin and pheomelanin). This process is under hormonal
control, including the MSH and ACTH peptides that are produced from the precursor
proopiomelanocortin.
People with oculocutaneous albinism typically have a very low level of melanin production.
Albinism is often but not always related to the TYR gene coding the tyrosinase enzyme.
Tyrosinase is required for melanocytes to produce melanin from the amino
acid tyrosine.[11] Albinism may be caused by a number of other genes as well,
like OCA2,[12] SLC45A2,[13]TYRP1,[14] and HPS1[15] to name some. In all, already 17 types of
oculocutaneous albinism have been recognized.[16] Each gene is related to different protein
having a role in pigment production.
People with Chédiak–Higashi_syndrome have a buildup of melanin granules due to abnormal
function of microtubules.

Stimulations[edit]
Main article: Melanocortin

Numerous stimuli are able to alter melanogenesis, or the production of melanin by cultured
melanocytes, although the method by which it works is not fully understood. Certain
melanocortins have been shown in laboratory testing to have effect on appetite and sexual
activity in mice.[17] Vitamin D metabolites, retinoids, melanocyte-stimulating
hormone, forskolin, cholera toxin, isobutylmethylxanthine, diacylglycerol analogues, and UV
irradiation all trigger melanogenesis and, in turn, pigmentation.[18] Increased melanin
production is seen in conditions where Adrenocorticotropic Hormone (ACTH) is elevated, such
as Cushing's disease. This is mainly a consequence of alpha-MSH being secreted along with the
hormone associated with reproductive tendencies in primates. Alpha-MSH is a cleavage
product of ACTH that has an equal affinity for the MC1 receptor on melanocytes as ACTH. [19]
Melanosomes are vesicles that package the chemical inside a plasma membrane.
The melanosomes are organized as a cap protecting the nucleus of the keratinocyte.
When ultraviolet rays penetrate the skin and damage DNA, thymidine dinucleotide (pTpT)
fragments from damaged DNA will trigger melanogenesis[20] and cause the melanocyte to
produce melanosomes, which are then transferred by dendrites to the top layer of
keratinocytes.

You might also like