You are on page 1of 14

The official journal of

INTERNATIONAL FEDERATION OF PIGMENT CELL SOCIETIES · SOCIETY FOR MELANOMA RESEARCH

PIGMENT CELL & MELANOMA


Research
Recent progress in melasma pathogenesis
Ai-Young Lee

DOI: 10.1111/pcmr.12404
Volume 28, Issue 6, Pages 648–660

If you wish to order reprints of this article,


please see the guidelines here

EMAIL ALERTS
Receive free email alerts and stay up-to-date on what is published
in Pigment Cell & Melanoma Research – click here

Submit your next paper to PCMR online at http://mc.manuscriptcentral.com/pcmr

Subscribe to PCMR and stay up-to-date with the only journal committed to publishing
basic research in melanoma and pigment cell biology

As a member of the IFPCS or the SMR you automatically get online access to PCMR. Sign up as
a member today at www.ifpcs.org or at www.societymelanomaresarch.org

To take out a personal subscription, please click here


More information about Pigment Cell & Melanoma Research at www.pigment.org
Pigment Cell Melanoma Res. 28; 648–660 REVIEW

Recent progress in melasma pathogenesis


Ai-Young Lee

Department of Dermatology, Dongguk University Ilsan Hospital, Ilsandong-gu, Goyang-si, Gyeonggi-do, South KEYWORDS melasma/pathogenesis/UV/estrogen/
Korea UV exposure-independent factors/dermal factors/
microRNAs
CORRESPONDENCE Ai-Young Lee, e-mails: lay5604@naver.com; leeay@dumc.org
PUBLICATION DATA Received 17 April 2015, revised
and accepted for publication 27 July 2015, published
online 31 July 2015

doi: 10.1111/pcmr.12404

Summary
Melasma is a common skin pigmentation condition. Given therapeutic difficulty as one of the biggest concerns,
understanding of the etiology and pathogenesis of melasma becomes essential. UV irradiation, female sex
hormones, and inflammatory processes are addressed as triggering factors with genetic predisposition. The
mechanism of UV-induced melanogenesis has been extensively investigated as a model system to study
melasma pathogenesis. Hitherto, treatment modalities for melasma are similar to other hyperpigmentation
disorders. However, individual triggering factors induce a separate pigmentation disease, whose pathogenic
mechanisms and clinical phenotypes are different from the ones encountered in melasma. Fortunately, there
have been ongoing updates on melasma pathogenesis with regard to major triggering factors. Presence of
certain factors working independently of UV exposure and role of dermal factors and microRNAs are being
identified as novel discoveries about melasma pathogenesis. In this review, the melasma pathogenesis is
reviewed in association with updated and new findings.

hyperpigmentation disorders. Genes that encode tyrosi-


Introduction
nase, a key enzyme in melanogenesis, and the tyrosi-
Melasma is a pattern of acquired hyperpigmentation and nase-related proteins (TRPs) TRP-1 and TRP-2 are
one of the most common pigmentation disorders involv- commonly involved in the pigmentation disorders induced
ing the face. Three types of melasma exist, as classified by many exogenous and endogenous factors, and
according to their distribution on the face, which include microphthalmia-associated transcription factor (MITF)
centrofacial, malar, and mandibular patterns (Mandry plays a fundamental role in the transcriptional regulation
Pagan and Sanchez, 2000; Sanchez et al., 1981). Another of these genes. A thorough understanding of the etiology
classification of melasma according to the location of the and pathogenesis of melasma in particular and skin
pigment as epidermal, dermal, or mixed type (Gilchrest hyperpigmentation disorders in general is crucial to the
et al., 1977) may perhaps facilitate in predicting the appropriate management of melasma.
treatment outcome, which is the most imperative issue in Genetic backgrounds, chronic exposure to ultraviolet
melasma, particularly in dermal type of melasma (Cayce (UV) radiation, and female sex hormones have been
et al., 2004). With particular emphasis on facial involve- implicated as the main causes of melasma. Recently,
ment and therapeutic difficulty, melasma has consider- involvement of inflammatory processes in melasma
able impact on the quality of life of affected individuals. development has also been considered (Handel et al.,
Although hyperpigmentation results from diverse disor- 2014a; Noh et al., 2014). Each of these factors simulta-
ders, to date, no differences in treatment modalities for neously induces a separate pigmentation disease, such
these disorders have been presented (Grimes, 2009; as pigmentation induced by chronic UV irradiation (pho-
Guerrero, 2012; Lynde et al., 2006; Molinar et al., 2014; toaging), drug (oral contraceptives)-induced pigmentation,
Perez-Bernal et al., 2000; Stratigos and Katsambas, or post-inflammatory hyperpigmentation (PIH). In fact,
2004). This phenomenon suggests that common patho- PIH is one of the common hyperpigmentation disorders in
genic mechanisms could be involved in various skin company with melasma and lentigines (Cayce et al.,

648 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Melasma pathogenesis

2004; Nicolaidou et al., 2007). There is a possibility of co- 2002), dilated dermal microvasculature (Kim et al., 2007),
existence of these hyperpigmentations in the same and basement membrane disruption with protrusion of
patients. Therefore, these conditions should be differen- melanocytes into dermis (Torres-Alvarez  et al., 2011)
tiated to investigate the pathogenesis of melasma. Their have been observed in melasma (Table 1). Similarities in
clinical characteristics are different from melasma the microscopic findings between skin with chronic UV
(Table 1), with the exception of pigmentation due to oral exposure and melasma skin emphasize a potential role of
contraceptives, which frequently induce skin pigmenta- cutaneous photoaging due to cumulative sun exposure in
tion of the melasma type (Baker, 1969; Resnik, 1967). melasma (Hern andez-Barrera et al., 2008).
Post-inflammatory hyperpigmentation develops at any However, a role of UV exposure-independent factors in
age on the site of inflammation without specific site melasma pathogenesis has been identified (Kim et al.,
predilection caused by physical or chemical injury, skin 2010, 2013a). Our understanding of the mechanism
irritation, contact dermatitis, or dermatosis (Eimpunth concerning the effect of estrogen on skin pigmentation
et al., 2013; Stulberg et al., 2003). The pigmentation and/or melasma has also been updated (Kim et al., 2012;
induced by chronic UV exposures could be accompanied Kippenberger et al., 1998; McLeod et al., 1994). Recently,
by characteristic findings of photoaging, such as wrinkles, novel cellular and molecular mechanisms for skin pigmen-
tactile roughness, and loss of skin tone/resilience. The tation including melasma have been identified in the
vital difference lies in the pigmentary change in these context of cell-to-cell interaction between melanocytes
disorders, which tends to be recovered, whereas that in and dermal fibroblasts (Cardinali et al., 2009; Choi et al.,
melasma does not (Table 1). A complex interplay of these 2010; Kang et al., 2006; Kim et al., 2013a,b; Poon et al.,
factors may make the outcome of melasma more 2005; Shin et al., 2012; Yamaguchi et al., 2004) and
difficult. Microscopically, increased melanin in epidermal microRNAs (Dynoodt et al., 2013; Kim et al., 2014a,b,c).
keratinocytes, dermal macrophages, or both is a common In this review, factors implicated in melasma development
finding in hyperpigmentation disorders including melasma are reviewed with focus particularly on the updated action
(Callender et al., 2011; Dereure, 2001; Grimes et al., mechanism of UV exposure and estrogens, along with the
2005; Pathak and Fanselow, 1983). On the other hand, newly identified mechanism pertaining to UV exposure-
hyperpigmentation from repeated UV irradiation is usually independent factors, dermal factors, and microRNAs.
accompanied by changes from chronic sun exposure,
such as massive elastosis, collagen degeneration, and
Update on factors implicated as main
twisted dilated dermal microvasculature (Gilchrest, 1996).
causes in melasma
However, these changes are not specific for repeated UV
exposure. Despite no obvious signs of photoaging in Genetic factors in melasma development
appearance, more prominent solar elastosis (Grimes Genetic predisposition is considered one of the main
et al., 2005; Hernandez-Barrera et al., 2008; Kang et al., causes implicated in melasma development, with sun

Table 1. Characteristics of melasma versus related hyperpigmentation disorders

Characteristic
Disorder clinical finding Microscopic finding Outcome References

Melasma Facial pigmentation Increased melanin in Chronic course with Sanchez et al. (1981),
with centrofacial, epidermal keratinocytes, exacerbation Mandry Pag an
malar, and mandibular dermal macrophages, or and Sanchez (2000),
distribution both Kang et al. (2002),
Elastosis, collagen Grimes et al. (2005),
degeneration, and dilated Hernandez-
dermal microvasculature Barrera et al. (2008),
Kim et al. (2007),

Torres-Alvarez et al. (2011)
UV-induced Pigmentation, with Normalize after Gilchrest (1996)
hyperpigmentation wrinkles, tactile elimination of specific
roughness, and/or cause
loss of skin tone/
resilience
Oral contraceptives- Similar to those Increased melanin in epidermal Resnik (1967),
induced of melasma keratinocytes, dermal Baker (1969)
hyperpigmentation macrophages, or both
Post-inflammatory No specific site Stulberg et al. (2003),
hyperpigmentation predilection, Eimpunth et al. (2013)
pigmentation on the
site of inflammation

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 649
Melasma pathogenesis

A new secreted protein that binds to Wnt proteins and inhibits their melanogenesis via MITF as a direct target. J. Invest. Dermatol.
activities. Nature 398, 431–436. 134, 1075–1082.
Im, S., Kim, J., On, W.Y., and Kang, W.H. (2002). Increased Kim, N.H., Choi, S.H., Lee, T.R., and Lee, A.Y. (2014c). Cadherin 11, a
expression of alpha-melanocyte-stimulating hormone in the miR-675 target, induces N-cadherin expression and epithelial-
lesional skin of melasma. Br. J. Dermatol. 146, 165–167. mesenchymal transition in melasma. J. Invest. Dermatol. 134,
Imokawa, G., Miyagishi, M., and Yada, Y. (1995). Endothelin-1 as a 2967–2976.
new melanogen: coordinated expression of its gene and the Kippenberger, S., Loitsch, S., Solano, F., Bernd, A., and Kaufmann, R.
tyrosinase gene in UVB-exposed human epidermis. J. Invest. (1998). Quantification of tyrosinase, TRP-1, and Trp-2 transcripts in
Dermatol. 105, 32–37. human melanocytes by reverse transcriptase-competitive multi-
Ing, N.H., and Tornesi, M.B. (1997). Estradiol up-regulates estrogen plex PCR–regulation by steroid hormones. J. Invest. Dermatol.
receptor and progesterone receptor gene expression in specific 110, 364–367.
ovine uterine cells. Biol. Reprod. 56, 1205–1215. KrupaShankar, D.S., Somani, V.K., Kohli, M. et al. (2014). A cross-
Jang, Y.H., Lee, J.Y., Kang, H.Y., Lee, E.S., and Kim, Y.C. (2010). sectional, multicentric clinico-epidemiological study of melasma in
Oestrogen and progesterone receptor expression in melasma: an India. Dermatol. Ther. (Heidelb) 4, 71–81.
immunohistochemical analysis. J. Eur. Acad. Dermatol. Venereol. Kunisada, T., Lu, S.Z., Yoshida, H. et al. (1998). Murine cutaneous
24, 1312–1316. mastocytosis and epidermal melanocytosis induced by ker-
Jee, S.H., Lee, S.Y., Chiu, H.C., Chang, C.C., and Chen, T.Y. (1994). atinocyte expression of transgenic stem cell factor. J. Exp. Med.
Effects of estrogen and estrogen receptor in normal human 187, 1565–1573.
melanocytes. Biochem. Biophys. Res. Commun. 199, 1407–1412. Lieberman, R., and Moy, L. (2008). Estrogen receptor expression in
Jian, D., Jiang, D., Su, J., Chen, W., Hu, X., Kuang, Y., Xie, H., Li, J., melasma: results from facial skin of affected patients. J. Drugs
and Chen, X. (2011). Diethylstilbestrol enhances melanogenesis via Dermatol. 7, 463–465.
cAMP-PKA-mediating up-regulation of tyrosinase and MITF in Longley, B.J. Jr, Morganroth, G.S., Tyrrell, L., Ding, T.G., Anderson,
mouse B16 melanoma cells. Steroids 76, 1297–1304. D.M., Williams, D.E., and Halaban, R. (1993). Altered metabolism
Jo, H.Y., Kim, C.K., Suh, I.B., Ryu, S.W., Ha, K.S., Kwon, Y.G., and of mast-cell growth factor (c-kit ligand) in cutaneous mastocytosis.
Kim, Y.M. (2009). Co-localization of inducible nitric oxide synthase N. Engl. J. Med. 328, 1302–1307.
and phosphorylated Akt in the lesional skins of patients with Longstaff, C. (1994). Studies on the mechanisms of action of
melasma. J. Dermatol. 36, 10–16. aprotinin and tranexamic acid as plasmin inhibitors and antifibri-
Kanechorn Na Ayuthaya, P., Niumphradit, N., Manosroi, A., and nolytic agents. Blood Coagul. Fibrinolysis 5, 537–542.
Nakakes, A. (2012). Topical 5% tranexamic acid for the treatment Lynde, C.B., Kraft, J.N., and Lynde, C.W. (2006). Topical treatments
of melasma in Asians: a double-blind randomized controlled clinical for melasma and postinflammatory hyperpigmentation. Skin Ther-
trial. J. Cosmet. Laser Ther. 14, 150–154. apy Lett. 11, 1–6.
Kang, W.H., Yoon, K.H., Lee, E.S., Kim, J., Lee, K.B., Yim, H., Sohn, Mahmoud, B.H., Ruvolo, E., Hexsel, C.L., Liu, Y., Owen, M.R.,
S., and Im, S. (2002). Melasma: histopathological characteristics in Kollias, N., Lim, H.W., and Hamzavi, I.H. (2010). Impact of long-
56 Korean patients. Br. J. Dermatol. 146, 228–237. wavelength UVA and visible light on melanocompetent skin. J.
Kang, H.Y., Hwang, J.S., Lee, J.Y., Ahn, J.H., Kim, J.Y., Lee, E.S., Invest. Dermatol. 130, 2092–2097.
and Kang, W.H. (2006). The dermal stem cell factor and c-kit are Mandry Pag an, R., and S
anchez, J.L. (2000). Mandibular melasma. P.
overexpressed in melasma. Br. J. Dermatol. 154, 1094–1099. R. Health Sci. J. 19, 231–234.
Kasinski, A.L., and Slack, F.J. (2011). MicroRNAs en route to the McLeod, S.D., Ranson, M., and Mason, R.S. (1994). Effects of
clinic: progress invalidating and targeting microRNAs for cancer estrogens on human melanocytes in vitro. J. Steroid Biochem.
therapy. Nat. Rev. Cancer 11, 849–864. Mol. Biol. 49, 9–14.
Kato, H., Araki, J., Eto, H., Doi, K., Hirai, R., Kuno, S., Higashino, T., Mione, M., and Bosserhoff, A. (2015). MicroRNAs in melanocyte
and Yoshimura, K. (2011). A prospective randomized controlled and melanoma biology. Pigment Cell Melanoma Res. 28, 340–
study of oral tranexamic acid for preventing postinflammatory 354.
hyperpigmentation after Q-switched ruby laser. Dermatol. Surg. Miot, L.D., Miot, H.A., Polettini, J., Silva, M.G., and Marques, M.E.
37, 605–610. (2010). Morphologic changes and the expression of alpha-me-
Kim, E.H., Kim, Y.C., Lee, E.S., and Kang, H.Y. (2007). The vascular lanocyte stimulating hormone and melanocortin-1 receptor in
characteristics of melasma. J. Dermatol. Sci. 46, 111–116. melasma lesions: a comparative study. Am. J. Dermatopathol.
Kim, N.H., Lee, C.H., and Lee, A.Y. (2010). H19 RNA downregulation 32, 676–682.
stimulated melanogenesis in melasma. Pigment Cell Melanoma Moin, A., Jabery, Z., and Fallah, N. (2006). Prevalence and awareness
Res. 23, 84–92. of melasma during pregnancy. Int. J. Dermatol. 45, 285–288.
Kim, N.H., Cheong, K.A., Lee, T.R., and Lee, A.Y. (2012). PDZK1 Molinar, V.E., Taylor, S.C., and Pandya, A.G. (2014). What’s new in
upregulation in estrogen-related hyperpigmentation in melasma. objective assessment and treatment of facial hyperpigmentation?
J. Invest. Dermatol. 132, 2622–2631. Dermatol. Clin. 32, 123–135.
Kim, J.E., Chang, S.E., Yeo, U.C., Haw, S., and Kim, I.H. (2013a). Muallem, M.M., and Rubeiz, N.G. (2006). Physiological and biological
Histopathological study of the treatment of melasma lesions using skin changes in pregnancy. Clin. Dermatol. 24, 80–83.
a low-fluence Q-switched 1064-nm neodymium:yttrium-alu- Na, J.I., Choi, S.Y., Yang, S.H., Choi, H.R., Kang, H.Y., and Park, K.C.
minium-garnet laser. Clin. Exp. Dermatol. 38, 167–171. (2013). Effect of tranexamic acid on melasma: a clinical trial with
Kim, J.Y., Lee, T.R., and Lee, A.Y. (2013b). Reduced WIF-1 histological evaluation. J. Eur. Acad. Dermatol. Venereol. 27, 1035–
expression stimulates skin hyperpigmentation in patients with 1039.
melasma. J. Invest. Dermatol. 133, 191–200. Nicolaidou, E., Antoniou, C., and Katsambas, A.D. (2007). Origin,
Kim, K.H., Bin, B.H., Kim, J. et al. (2014a). Novel inhibitory function of clinical presentation, and diagnosis of facial hypermelanoses.
miR-125b in melanogenesis. Pigment Cell Melanoma Res. 27, Dermatol. Clin. 25, 321–326.
140–144. Noh, T.K., Choi, S.J., Chung, B.Y., Kang, J.S., Lee, J.H., Lee, M.W.,
Kim, N.H., Choi, S.H., Kim, C.H., Lee, C.H., Lee, T.R., and Lee, A.Y. and Chang, S.E. (2014). Inflammatory features of melasma lesions
(2014b). Reduced MiR-675 in exosome in H19 RNA-related in Asian skin. J. Dermatol. 41, 788–794.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 659
Melasma pathogenesis

Figure 1. Schematic view of melanogenesis


induced by external stimuli, particularly UV
radiation. Direct effects on melanocytes and
indirect effects on keratinocytes/fibroblasts
releasing melanogenic factors, such as
proopiomelanocortin (POMC)-derived peptides
(MSH, ACTH), ET-1, SCF, bFGF, or NGF, are
involved in melanogenesis. Protein kinase C
(PKC), NO, and cAMP are major intracellular
signal transduction pathways. The black ovals
indicate melanosomes (DAG, 1,2-
diacylglycerol; NO, nitric oxide; MSH,
melanocyte-stimulating hormone; ACTH,
adrenocorticotrophic hormone; ET-1,
endothelin-1; SCF, stem cell factor; bFGF,
basic fibroblast growth factor; NGF, nerve
growth factor; MC1R, melanocortin-1 receptor;
TYR, tyrosinase; TRP, tyrosinase-related
protein).

In melasma, DNA damage from direct effect of UV melasma as in other skin hyperpigmentation conditions,
photons (Eller et al., 1996) or DAG/arachidonic acid including UV-induced melanogenesis. In melasma
pathways released from melanocyte membranes (Cars- patients, the expression of certain keratinocyte-derived
berg et al., 1995) are not delineated. On the other hand, paracrine factors is elevated in hyperpigmented lesional
cross talk between melanocytes and keratinocytes has skin relative to normally pigmented skin (Table 3). Con-
been considered as a melanogenic mechanism in focal microscopic examination and Western blotting have

Table 3. Expression of paracrine melanogenic factors in melasma that have been identified in other skin hyperpigmentation conditions

Conditions in which the References in the


Origin Factors factor has been detected context of melasma

Keratinocytes Neural endopeptidase UV-induced hyperpigmentation Bak et al. (2009)


NGF UV-induced hyperpigmentation Kim et al. (2013a,b)
Alpha-MSH UV-induced hyperpigmentation Im et al. (2002),
Miot et al. (2010)
bFGF UV-induced hyperpigmentation No report identified
ET-1 UV-induced hyperpigmentation No report identified
iNOS UV-induced hyperpigmentation Jo et al. (2009)
Mast cell Histamine UV-induced hyperpigmentation, PIH, No report identified
Urticaria pigmentosa
Fibroblasts SCF UV-induced hyperpigmentation, Kang et al. (2006),
post-laser hyperpigmentation, 
Torres-Alvarez et al. (2011)
familial hyperpigmentation
bFGF Post-laser hyperpigmentation
HGF Post-laser hyperpigmentation No report identified
KGF PIH No report identified
DKK1 Physiological No report identified
Neuregulain-1 Physiological No report identified
VGEF UV-induced hyperpigmentation Kim et al. (2007)

NGF, nerve growth factor; MSH, melanocyte-stimulating hormone; MC1R, melanocortin-1 receptor; bFGF, basic fibroblast growth factor; ET-1,
endothelin-1; iNOS, inducible nitric oxide synthase; WIF-1, Wnt inhibitory factor 1; SCF, stem cell factor; HGF, hepatocyte growth factor; KGF,
keratinocyte growth factor; DKK1, dickkopf 1; VEGF, vascular endothelial growth factor; PIH, post-inflammatory hyperpigmentation; Familial
hyperpigmentation, familial progressive hyperpigmentation-like disorder.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 651
Lee

shown greater expression of NGF receptor with neural also identified Wnt inhibitory factor1 (WIF-1) (Figure 2)
endopeptidase in the hyperpigmented lesional skin in six (Kim et al., 2013a,b), as another factor involved in melasma
patients (Bak et al., 2009). Increased NGF expression in pathogenesis without any expression changes by UV
melasma skin of eight patients has also been examined irradiation. This will be dealt with in detail in the later
immunohistochemically (Kim et al., 2013a,b). Similar section examining dermal factors.
immunohistochemistry result of increased alpha-MSH
expression in lesional epidermis has been reported in ten Role of female sex hormones in melasma
Korean women (Im et al., 2002) and in 24 Brazilian development
women (Miot et al., 2010). Another UV-induced paracrine Melasma is considered as a common physiological skin
melanogenic factor from keratinocytes is NO (Rome ro- change during pregnancy (Elling and Powell, 1997;
Graillet et al., 1997). The role of NO has also been Muallem and Rubeiz, 2006; Sodhi and Sausker, 1988),
suggested in melasma based on the immunohistochem- and as an undesirable cutaneous effect of oral contra-
ical observation of increased inducible NO synthase ceptives (Foldes, 1988). Epidemiological data revealed
expression in basal layer keratinocytes of hyperpig- the occurrence of melasma in 14.5–56% of pregnant
mented lesional skin from nine patients and RT-PCR women and in 11.3–46% of individuals who used oral
from additional three patients (Jo et al., 2009). Paracrine contraceptives in different countries, including Singapore
melanogenic factors could also be released from dermal (Goh and Dlova, 1999), Iran (Moin et al., 2006), Tunisian
fibroblasts in melasma as in UV irradiation (Table 3); (Guinot et al., 2010), India (Achar and Rathi, 2011;
immunohistochemistry has shown increased SCF in KrupaShankar et al., 2014), and Brazil (Handel et al.,
hyperpigmented dermis when compared to normally 2014b; Tamega Ade et al., 2013). Despite a large differ-
pigmented dermis in melasma patients (Kang et al., ence in the prevalences of melasma among different

2006; Torres-Alvarez et al., 2011). ethnic groups and skin phototypes, the preferential
A complex interaction of causative factors, but not a development of melasma during the reproductive lifespan
single factor, is likely to be involved in melasma develop- of women and the association of melasma with oral
ment. Although the role of UV exposure has been well contraceptives suggest that female sex hormones are
established, the mechanism of UV-induced pigmentation another important precipitating factor for the develop-
may not be the same for skin pigmentation conditions ment and aggravation of this disease. In contrast, the
induced by different causes. In addition, UV irradiation may dissenting opinion that hormones only weakly influence
not be always necessary in melasma development. We melasma has also been offered (Passeron, 2013).
have identified that downregulation in the expression of With respect to female sex hormones, estrogens and
H19 RNA, which has been detected in microarray analysis progesterones have been implicated in the development
for comparison of hyperpigmented skin specimens with of melasma. The activities of estrogens and proges-
normally pigmented biopsied skin specimens from patients terones are mediated by specific receptors expressed in
with melasma, stimulates melanogenesis and melano- human skin, including the estrogen receptors (ERs) ER-
some transfer (Kim et al., 2010). H19 RNA downregulation alpha/ER-beta and progesterone receptors (PRs), respec-
has been examined in the hyperpigmented skin of the tively (Pelletier and Ren, 2004; Thornton, 2002). Conflict-
patients, but not in the UV-exposed hyperpigmented skin ing results regarding the effects of these hormones,
(Figure 2). In addition, no alterations were observed in H19 particularly progesterones, have been obtained. Melasma
RNA knockdown-induced melanogenesis in the presence has been reported as an adverse reaction of contraceptives
of UV irradiation, although UV irradiation alone and H19 containing the synthetic progestin levonorgestrel (Chom-
knockdown alone increased tyrosinase expression. We pootaweep et al., 1996), and the expression of PR

Postinflammatory hyperpigmentation

Keratinocyte-derived
paracrine melagenic
factors

Fibroblast-derived
paracrine melagenic
factors

Figure 2. Factors involved in e melanogenesis


H19 RNA ↓ in melasma versus other hyperpigmentation
disorders. Decreased expression of H19 RNA
Melasma
WIF-1 ↓ and WIF-1 is involved in the pathogenesis of
melasma, but not post-inflammatory
hyperpigmentation and UV-induced
UV-induced pigmentation pigmentation (WIF-1, Wnt inhibitory factor 1).

652 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Melasma pathogenesis

proteins is increased in hyperpigmented skin in melasma


(Jang et al., 2010; Tamega Ade et al., 2015), suggesting
that progesterone plays a role in the development of
melasma. Conversely, the prevention of melasma by
progesterone components in oral contraceptives has
been suggested based on findings that progesterone
reduces proliferation without significant effects on tyrosi-
nase activity, counteracting the stimulatory effects of
estrogen in cultured melanocytes (Wiedemann et al.,
2009). An increase in male melasma patients due to the
advent of finasteride, an anti-androgen (Famenini et al.,
2014), also suggests that increased progesterone is
unrelated to the development of melasma, because
finasteride inhibits progesterone synthesis (Freeman
et al., 1993) without changing estrogen levels (Vermeulen
et al., 1991). Certain factors which could alter the sex
hormone metabolism, including interindividual variation in
the enzymes (Orme et al., 1989), may contribute to
generate the conflicting result. Other cellular and para-
crine regulatory factors have been also suggested to be
involved in estrogen responses (Feucht et al., 1988; Ing
and Tornesi, 1997), which may require a careful research
design and an analysis of the result for in vitro studies.
Immunohistochemistry revealed increased ER expression
in the affected skin (Lieberman and Moy, 2008), although
significant immunohistochemical expression of ER-beta
has been presented in melasma-affected dermis, but not
in epidermis (Jang et al., 2010). Increased ER expression
indicates a potential role of estrogen in melasma. Estro-
gens are considered to stimulate melanogenesis in
cultured human melanocytes by inducing synthesis of
melanogenic enzymes such as tyrosinase, TRP-1, TRP-2,
and MITF (Jian et al., 2011; Kim et al., 2012; Kippen-
berger et al., 1998). Melanocytes express ERs (Jee et al., Figure 3. Mechanism involved in estrogen-induced melanogenesis
1994; Kim et al., 2012), which are involved in estrogen- and PDZK1 role in melasma. (A) By binding to ERs, estrogen
induced melanogenesis, because the inhibition of ER by enhances cAMP levels and upregulates CREB, MITF, and tyrosinase
its antagonist leads to reduction in melanogenesis (Kim family protein expression, with the involvement of the PKA pathway.
et al., 2012). In addition, estrogen-induced melanogene- (B) PDZK1 could facilitate the estrogen action by interaction with
other proteins including ion exchangers, resulting in the stimulation
sis could be associated with the activation of the cAMP-
of melanogenesis and melanosome transfer in melasma patients
PKA pathway, because estrogens enhance cAMP levels (ER, estrogen receptor; PKA, protein kinase A; CREB, cAMP
and upregulation of tyrosinase and MITF attenuation by responsive-element-binding protein; CBP, CREB-binding protein;
blocking the PKA pathway, which has previously been MITF, micropthalmia-associated transcription factor; TYR, tyrosinase;
reported (Jian et al., 2011) (Figure 3A). TRP, tyrosinase-related protein; PDZK1, PDZ domain protein kidney
We have identified upregulation of PDZ domain protein 1; NHE, sodium–hydrogen exchanger; CFTR, cystic fibrosis
transmembrane conductance regulator).
kidney 1 (PDZK1) expression in the hyperpigmented skin
of melasma patients using real-time PCR and immuno-
histochemical staining (Kim et al., 2012). Cultured normal tyrosinase expression in melanocyte monocultures and
human keratinocytes and melanocytes express PDZK1, melanocyte–keratinocyte co-cultures. In addition, PDZK1
and PDZK1 overexpression of keratinocytes alone, overexpression increases estrogen-stimulated tyrosinase
melanocytes alone, or both increases the expression of expression and melanosome transfer with ER-alpha and
tyrosinase, CREB, and MITF in melanocyte monocultures ER-beta expression (Kim et al., 2012). As the name
or melanocyte–keratinocyte co-cultures. PDZK1 is a NHERF suggests, PDZK1 interacts with ion exchangers
member of the sodium–hydrogen exchanger regulatory such as NHE, cystic fibrosis transmembrane conductance
factor (NHERF) family, called NHERF-3. As estrogen regulator (CFTR), and SLC26A family with potential
exerts actions through NHERF1 or PDZK1 regulation in relationship between each ER subtype (ER-alpha or ER-
ER-containing breast cancer cells (Ediger et al., 1999; beta) and NHE3 or CFTR expression, respectively.
Ghosh et al., 2000), estrogen increases PDZK1 with Sodium–hydrogen exchanger regulatory factor family

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 653
Lee

proteins mediate the most abundant protein–protein increase melanogenic factors, such as SCF, bFGF, and
interactions; therefore, PDZK1 could facilitate the estro- hepatocyte growth factor (HGF), resulting in post-laser
gen action by interaction with other proteins including hyperpigmentation (Poon et al., 2005). Increased expres-
ion exchangers, resulting in the stimulation of melano- sion of SCF, HGF, and KGF in dermal fibroblasts of
genesis and melanosome transfer in melasma patients hyperpigmented skin also suggests a role of fibroblast-
(Figure 3B). derived paracrine factors in familial progressive hyperpig-
mentation-like disorder (Cardinali et al., 2009).
With reference to melasma, there are only few data
Newly discovered factors in melasma
related to fibroblast-derived dermal factors (Table 3).
pathogenesis
Immunohistochemistry data from melasma patients sug-
Dermal factors implicated in melasma gest a role of increased SCF in dermis along with
The dermal resident cells, mast cells, and fibroblasts have increased c-KIT in epidermis in pigmentation (Kang et al.,
been presented with regard to skin pigmentation. Given 
2006; Torres-Alvarez et al., 2011). Meanwhile, we
urticaria pigmentosa is a cutaneous mastocytosis charac- demonstrated that reduced WIF-1 expression in the
terized by skin pigmentation, the role of mast cells in skin hyperpigmented skin of melasma patients increases
pigmentation has been considered. Although SCF, also melanognesis and melanosome transfer, irrespective of
known as mast cell growth factor, is involved in cuta- UV irradiation (Kim et al., 2013a,b). WIF-1, as a secreted
neous mastocytosis and melanogenesis in urticarial antagonist of Wnt signaling, belongs to the secreted
pigmentosa (Costa et al., 1996; Kunisada et al., 1998; Frizzled-related protein class, which inhibits both the
Longley et al., 1993), it is not originated from mast cells. canonical and non-canonical Wnt pathway (Hsieh et al.,
Meanwhile, histamine released from mast cells is iden- 1999). WIF-1 is expressed in both cultured normal human
tified to be involved in melanogenesis in urticarial keratinocytes and fibroblasts, but not in melanocytes. The
pigmentosa as well as post-inflammatory pigmentation decreased expression of WIF-1 not only in keratinocytes
and UV-induced pigmentation (Tomita et al., 1989; but also in fibroblasts reduces WIF-1 binding to Wnts in
Yoshida et al., 2002). As in melanogenesis induced by melanocytes, exerting the action through the canonical
interaction between POMC-derived peptides and MC1R Wnt/beta-catenin pathway resulting in MITF upregulation
(Abdel-Malek et al., 1999; Chakraborty et al., 1999), and the non-canonical pathway resulting in translocation
histamine increases melanin synthesis by binding to of nuclear factor of activated T cells (NFAT) to nucleus
histamine receptor in melanocytes, particularly H2, (Figure 4).
through intracellular cAMP and subsequent PKA activa- The potential role of altered dermal vasculature has also
tion (Yoshida et al., 2000). been considered in melasma patients and UV-irradiated
Role of histamine in melasma has not been presented individuals (Yano et al., 2005). Immunohistochemistry
(Table 3), although an increase in the number of mast data revealed an increase in dermal blood vessel number
cells has been detected in pigmented lesional dermis and vascular endothelial growth factor (VEGF) expression
of melasma patients (Hernandez-Barrera et al., 2008; in pigmented lesion of melasma (Kim et al., 2007)

Torres-Alvarez et al., 2011). (Table 3). Although VEGF can increase the number and
The role of dermal fibroblasts in skin pigmentation has diameter of peripheral blood vessels (Chen et al., 2014),
been presented in both physiological and pathological skin the role of VEGF in skin pigmentation has not been clearly
conditions. In case of physiological conditions, dickkopf 1 elucidated. In addition, the therapeutic outcomes of
(DKK1) has been identified to play a role in hypopig- copper bromide combined with a yellow laser that emits
mented phenotype of palmoplantar skin (Yamaguchi dual wavelengths, enabling the selective and simultane-
et al., 2004); DKK1 is secreted by fibroblasts. As an ous destruction of both blood vessels and melanin-
inhibitor of the canonical Wnt signaling pathway, DKK1 containing cells, have not been encouraging (Eimpunth
suppresses melanocyte function and growth through the et al., 2014). Therapeutic effect of tranexamic acid, which
regulation of MITF and beta-catenin. Role of fibroblast- reduces pigmentation and vessel number (Na et al.,
derived factors in the regulation of human skin color has 2013), may support a role of altered dermal vasculature
also been identified; Fibroblasts in dark skin secrete in melasma. However, tranexamic acid acts as a plasmin
neuregulin-1, which exerts its action on melanocytes to inhibitor and an antifibrinolytic agent (Longstaff, 1994).
increase melanogenesis (Choi et al., 2010). In patholog- Research has not demonstrated the inhibition of vascular
ical skin pigmentation conditions, the role of paracrine ectasia by tranexamic acid. Although randomized, com-
factors derived from dermal fibroblasts has been pre- parative clinical trials have indicated that tranexamic acid
sented; increased SCF secretion from fibroblasts treatment produces favorable outcomes for PIH and
exposed to UV radiation is involved in skin hyperpigmen- melasma (Budamakuntla et al., 2013; Cho et al., 2013;
tation induced by chronic sunlight exposure (Shin et al., Shin et al., 2013), other trials have not supported this
2012). Keratinocyte growth factor (KGF) is mentioned as finding (Kanechorn Na Ayuthaya et al., 2012; Kato et al.,
fibroblast-derived growth factor in PIH (Cardinali et al., 2011). Therefore, the mechanism of action of tranexamic
2012). Fibroblasts exposed to sublethal laser fluence acid in melasma treatment remains unclear.

654 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Melasma pathogenesis

Figure 4. Role of WIF-1 in fibroblasts (or


keratinocytes) in melanogenesis. Reduced
WIF-1 expression in fibroblasts (or
keratinocytes) increases Wnt expression and
action in melanocytes through the canonical
Wnt/beta-catenin pathway with
microphthalmia-associated transcription factor
(MITF) upregulation and the non-canonical
pathway with NFATc2 translocation to nucleus
(WIF-1, Wnt inhibitory factor 1; LEF-1,
lymphocyte enhancer binding factor 1; NFAT,
nuclear factor of activated T cells).

MicroRNAs and their targets in melasma of human melanocytes under the external stimuli of
development forskolin and UV irradiation through Myo5a as a target
MicroRNAs (miRNAs) are small, 20–24 nucleotide, (Dynoodt et al., 2013). In addition, miR-125b is identified
endogenously expressed non-coding RNAs. MiRNAs as a regulator of steady-state melanogenesis in the
anneal to the 30 untranslated region of mRNAs in a absence of external stimuli, such as forskolin treatment
sequence-specific fashion and then either block transla- and UV irradiation (Kim et al., 2014a).
tion or promote transcript degradation, thus playing a Potential role of miRNAs has been presented in
major role in post-transcriptional regulation of gene melasma; in the process of examining the mechanism
expression (Filipowicz et al., 2008). Based on the asso- of H19 non-coding RNA downregulation in melasma (Kim
ciation between miRNAs and a broad range of patholog- et al., 2010), we identified a potential role of miRNA in
ical conditions, such as cancer, cardiovascular diseases, melanogenesis and melanosome transfer with its targets
diabetes, liver diseases, respiratory diseases, psychiatric (Figure 5; Kim et al., 2014a,b). Expression of a H19 RNA-
diseases, neurological diseases, and inflammatory and derived miRNA, miR-675, is reduced in the hyperpig-
autoimmune diseases, the discovery of miRNAs mented skin of melasma patients (Kim et al., 2014a). In
becomes one of the major scientific breakthroughs in addition, miR-675 overexpression decreases the expres-
the current field of cell biology and medical science (Ha, sion of tyrosinase, TRP-1, and TRP-2, whereas its
2011). In pigmentation field, the role of miRNAs is knockdown increases their expression. Although melano-
continuously emerging (Mione and Bosserhoff, 2015). genesis occurs in melanocytes, miR-675 is released
MiR-25 in alpacas reduces melanogenesis through MITF from keratinocytes. Exosomes, which are small mem-
as a direct target, with its abundance in alpacas turning up brane vesicles of endocytic origin and released into the
white hair (Zhu et al., 2010). MiR-137 also targets MITF extracellular environment, contain intact and functional
with downregulation of tyrosinase, TRP-1, and TRP-2 miR-675 as reported (Valadi et al., 2007) as a means of
expression in transgenic mice with miR-137 overexpres- cell–cell communication between keratinocytes and
sion (Dong et al., 2012). In addition to miRNAs’ induction, melanocytes (Kim et al., 2014a,b,c). MiRNAs exert action
their silencing affects skin pigmentation. In fish skin through targets, and we identified MITF as a target of
pigmentation, miR-429 targets Foxd3, and its silencing miR-675, with a potential role of miR-675 reduction in
increases Foxd3 expression and thereby reduction in MITF upregulation in melasma patients (Kim et al.,
MITF (Yan et al., 2013). Role of miRNAs in skin pigmen- 2014a). Individual miRNAs can target multiple mRNAs
tation is also identified in human melanocytes; MiR-145 (Kasinski and Slack, 2011), and cadherin 11 (CDH11) is
regulates melanogenesis and melanosome translocation identified as another target of miR-675 (Kim et al.,

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 655
Lee

Figure 5. Schematic view of the role of H19-


derived miR-675 in keratinocytes in melasma.
MiR-675 is delivered from keratinocytes to
melanocytes and fibroblasts via membrane-
bound exosomes. MiR-675 could exert the
action through either microphthalmia-
associated transcription factor (MITF) in
melanocytes or CDH11 in fibroblasts/
keratinocytes as its target. CDH11 in
fibroblasts or keratinocytes is involved in
melanogenesis via the canonical Wnt and AKT
activation pathways in neighboring
melanocytes through the induction of N-
cadherin (CDH, cadherin; AKT, apoptosis
signal-regulating kinase; TYR, tyrosinase; TRP,
tyrosinase-related protein).

2014b). Increased CDH expression is also examined in Table 4. Working mechanisms and current therapeutic modalities
hyperpigmented skin of melasma patients, suggesting for melasma
the role of CDH11 in melasma. Although CDH11 expres-
Therapeutic
sion is not detectable in melanocytes, CDH11 in fibrob- Groups Targets modalities
lasts or keratinocytes stimulated melanogenesis via the
canonical Wnt and apoptosis signal-regulating kinase Common to Upregulation of Skin lightening
(AKT) activation pathways in co-cultured melanocytes various skin tyrosinase/MITF agents, such as
through the induction of N-cadherin. hyperpigmentation through intracellular hydroquinone with
disorders signaling pathways or without tretinoin
and corticosteroids,
Perspectives azelaic acid, and kojic
acid, among others
Melasma is a common skin pigmentation condition. Interventions, such
Published articles regarding melasma have largely as chemical peels,
focused on the treatment of this condition. This review lasers, and IPL
notes that multiple signaling pathways, including the Triggering factor UV radiation Strict use of
sunscreen
MSH/cAMP, KIT, and Wnt pathways, are involved in the
Dermal vessel Tranexamic acid
upregulation of tyrosinase and MITF, resulting in the ectasia (with attention
stimulation of melanogenesis and the development of devoted to potential
melasma. The concurrent activation of these pathways is thrombus formation)
a cornerstone of most hyperpigmentation syndromes Female sex Not clearly
(Speeckaert et al., 2014). Therefore, it is somewhat hormones recommended
understandable that the same therapeutic modalities Inflammation Corticosteroids
(but not clearly
(e.g. skin lightening agents with or without certain
recommended)
interventions) are used for melasma (Gupta et al., 2006)
and for other hyperpigmentation disorders (Table 4). IPL, intense pulsed light therapy; MITF, microphthalmia-associated
An effort to inhibit triggering factors or signaling transcription factor.
pathways involved in melasma could help to improve
the effects of popular therapeutic modalities (Table 4).
The results of epidemiological studies examining the roles Dlova, 1999; Guinot et al., 2010; Handel et al., 2014b;
of family history, UV irradiation, and pregnancy/contra- Hexsel et al., 2014; KrupaShankar et al., 2014; Moin
ceptives in melasma (Achar and Rathi, 2011; Goh and et al., 2006; Tamega Ade et al., 2013) differ across broad

656 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Melasma pathogenesis

ranges, suggesting that these findings are of limited or proteomic analysis and validation of patient skin
value. Experimental data from melasma patients have specimens (which are diagnosed correctly) would be
largely been obtained from microscopic or immunohisto- required. Identified causative factors could be exploited
chemical studies. Nonetheless, in association with UV as therapeutic agents (particularly if the expression or
exposure, melasma and skin changes due to chronic UV function of these factors was reduced) or as biological
irradiation exhibit similar histopathologic findings (Grimes targets used to develop therapeutic compounds for
et al., 2005; Hernandez-Barrera et al., 2008; Kang et al., melasma. They could also be utilized to identify the

2002; Torres-Alvarez et al., 2011); reports have also important contributing factors for individual patients.
stated that paracrine melanogenic factors are derived Causative factors would be more useful, if they could be
from both keratinocytes (Bak et al., 2009; Im et al., 2002; classified as melasma specific, which needs more steps
Jo et al., 2009; Miot et al., 2010) and fibroblasts (Kang for validation in other hyperpigmentation conditions as

et al., 2006; Torres-Alvarez et al., 2011). These ongoing well as in different types of melasma. It is apparent that
updates emphasize the role of UV radiation and propose the identification of melasma-specific triggering factors
strict use of sunscreen for the management of melasma, will pave desirable way to pursue personalized therapy
although more stable and intense effect of visible light in future.
irradiation on skin pigmentation may provide a clue in
explaining partial protective effect of sunscreens (Mah-
moud et al., 2010). With respect to the role of female sex Acknowledgements
hormones in the progression of melasma, the effects of This work was supported by the National Research Foundation of
estrogen, such as increased ER expression in hyperpig- Korea (NRF) grant funded by the Korea Government (MSIP) (No.
mented lesions (Jang et al., 2010; Lieberman and Moy, NRF-2014R1A2A2A09051812).
2008), and PDZK1, a factor that facilitates estrogen
activity by acting as a scaffold in interactions with other References
proteins (Kim et al., 2012), have been more consistent
than the effects of progesterones; however, the question Abdel-Malek, Z., Suzuki, I., Tada, A., Im, S., and Akcali, C. (1999). The
of which contraceptives are appropriate remains unre- melanocortin-1 receptor and human pigmentation. Ann. N. Y. Acad.
Sci. 885, 117–133.
solved. Based on the ectasia of dermal vessels in
Achar, A., and Rathi, S.K. (2011). Melasma: a clinico-epidemiological
melasma, tranexamic acid has been clinically tested in study of 312 cases. Indian J. Dermatol. 56, 380–382.
recent years; however, tranexamic acid should be used Bak, H., Lee, H.J., Chang, S.E., Choi, J.H., Kim, M.N., and Kim, B.J.
cautiously in the treatment of melasma because this (2009). Increased expression of nerve growth factor receptor and
compound can potentially promote thrombus formation neural endopeptidase in the lesional skin of melasma. Dermatol.
(Sperzel and Huetter, 2007) and because its administra- Surg. 35, 1244–1250.
Baker, H. (1969). Adverse cutaneous reactions to oral contraceptives.
tion has generated conflicting therapeutic results. With
Br. J. Dermatol. 81, 946–949.
respect to the PIH implicated in the development of Budamakuntla, L., Loganathan, E., Suresh, D.H., Shanmugam, S.,
melasma, inflammatory features have been examined in Suryanarayan, S., Dongare, A., Venkataramiah, L.D., and Prabhu,
melasma patients (Handel et al., 2014a; Noh et al., 2014); N. (2013). A randomised, open-label, comparative study of
however, the roles of histamine from mast cells and KGF tranexamic acid microinjections and tranexamic acid with micro-
from dermal fibroblasts have been identified in PIH but needling in patients with melasma. J. Cutan. Aesthet. Surg. 6,
not in melasma. A great deal of additional work may be 139–143.
Callender, V.D., St Surin-Lord, S., Davis, E.C., and Maclin, M. (2011).
necessary to identify the role of PIH in melasma.
Postinflammatory hyperpigmentation: etiologic and therapeutic
Certain triggering factors independent of UV exposure, considerations. Am. J. Clin. Dermatol. 12, 87–99.
such as H19 RNA and WIF-1 (Kim et al., 2010, 2013a,b), Cardinali, G., Kovacs, D., Giglio, M.D., Cota, C., Aspite, N., Amantea,
and the roles of miRNAs, particularly miRNAs derived A., Girolomoni, G., and Picardo, M. (2009). A kindred with familial
from the H19 non-coding RNA (Kim et al., 2014a,b), progressive hyperpigmentation-like disorder: implication of fibrob-
have also been identified in melasma via their direct last-derived growth factors in pigmentation. Eur. J. Dermatol. 19,
targets. Despite recent progress in understanding the 469–473.
Cardinali, G., Kovacs, D., and Picardo, M. (2012). Mechanisms
pathogenesis of melasma, the associations between
underlying post-inflammatory hyperpigmentation: lessons from
melasma and each triggering factor have only been solar lentigo. Ann. Dermatol. Venereol. 139(Suppl 4), S148–S152.
partially identified. It remains unclear which triggering Carsberg, C.J., Ohanian, J., and Friedmann, P.S. (1995). Ultraviolet
factors play more important roles in this pathogenesis. radiation stimulates a biphasic pattern of 1,2-diacylglycerol forma-
In addition, not every patient with melasma exhibits the tion in cultured human melanocytes and keratinocytes by activation
same clinical and histological features, suggesting that of phospholipases C and D. Biochem. J. 305(Pt 2), 471–477.
Cayce, K.A., Feldman, S.R., and McMichael, A.J. (2004). Hyperpig-
melasma could involve various mechanisms. For
menation: a review of common treatment options. J. Drugs
instance, relatively severe actinic damage is observed Dermatol. 3, 668–673.
in mandibular melasma (Mandry Pagan and S anchez, Chakraborty, A.K., Funasaka, Y., Slominski, A., Bolognia, J., Sodi, S.,
2000). To develop desirable treatment modalities, the Ichihashi, M., and Pawelek, J.M. (1999). UV light and MSH
identification of causative factors based on genomic and/ receptors. Ann. N. Y. Acad. Sci. 885, 100–116.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 657
Lee

Chen, M., Wang, W.P., Jia, W.R., Tang, L., Wang, Y., Zhan, W.W., Freeman, D.A., Gocze, P.M., and Porpaczy, Z. (1993). Finasteride
and Fei, X.C. (2014). Three-dimensional contrast-enhanced sonog- blocks progesterone synthesis in MA-10 Leydig tumor cells.
raphy in the assessment of breast tumor angiogenesis: correlation Endocrinology 133, 1915–1917.
with microvessel density and vascular endothelial growth factor Fuller, B.B., Lunsford, J.B., and Iman, D.S. (1987). Alpha-melanocyte-
expression. J. Ultrasound Med. 33, 835–846. stimulating hormone regulation of tyrosinase in Cloudman S-91
Chhajlani, V., and Wikberg, J.E. (1992). Molecular cloning and mouse melanoma cell cultures. J. Biol. Chem. 262, 4024–4033.
expression of the human melanocyte stimulating hormone recep- Ghosh, M.G., Thompson, D.A., and Weigel, R.J. (2000). PDZK1 and
tor cDNA. FEBS Lett. 309, 417–420. GREB1 are estrogen-regulated genes expressed in hormone-
Cho, H.H., Choi, M., Cho, S., and Lee, J.H. (2013). Role of oral responsive breast cancer. Cancer Res. 60, 6367–6375.
tranexamic acid in melasma patients treated with IPL and low Gilchrest, B.A. (1996). A review of skin ageing and its medical
fluence QS Nd:YAG laser. J. Dermatolog. Treat. 24, 292–296. therapy. Br. J. Dermatol. 135, 867–875.
Choi, W., Wolber, R., Gerwat, W., Mann, T., Batzer, J., Smuda, C., Gilchrest, B.A., Fitzpatrick, T.B., Anderson, R.R., and Parrish, J.A.
Liu, H., Kolbe, L., and Hearing, V.J. (2010). The fibroblast-derived (1977). Localization of melanin pigmentation in the skin with
paracrine factor neuregulin-1 has a novel role in regulating the Wood’s lamp. Br. J. Dermatol. 96, 245–248.
constitutive color and melanocyte function in human skin. J. Cell Gilchrest, B.A., Park, H.Y., Eller, M.S., and Yaar, M. (1996).
Sci. 123, 3102–3111. Mechanisms of ultraviolet light-induced pigmentation. Photochem.
Chompootaweep, S., Kochagarn, E., Sirisumpan, S., Tang-usaha, J., Photobiol. 63, 1–10.
Theppitaksak, B., and Dusitsin, N. (1996). Effectiveness of Goh, C.L., and Dlova, C.N. (1999). A retrospective study on the
Norplant implants among Thai women in Bangkok. Contraception clinical presentation and treatment outcome of melasma in a
53, 33–36. tertiary dermatological referral centre in Singapore. Singapore
Costa, J.J., Demetri, G.D., Harrist, T.J., Dvorak, A.M., Hayes, D.F., Med. J. 40, 455–458.
Merica, E.A., Menchaca, D.M., Gringeri, A.J., Schwartz, L.B., and Grimes, P.E. (2009). Management of hyperpigmentation in darker
Galli, S.J. (1996). Recombinant human stem cell factor (kit ligand) racial ethnic groups. Semin. Cutan. Med. Surg. 28, 77–85.
promotes human mast cell and melanocyte hyperplasia and Grimes, P.E., Yamada, N., and Bhawan, J. (2005). Light microscopic,
functional activation in vivo. J. Exp. Med. 183, 2681–2686. immunohistochemical, and ultrastructural alterations in patients
Dereure, O. (2001). Drug-induced skin pigmentation. Epidemiology, with melasma. Am. J. Dermatopathol. 27, 96–101.
diagnosis and treatment. Am. J. Clin. Dermatol. 2, 253–262. Guerrero, D. (2012). Dermocosmetic management of hyperpigmen-
Dong, C., Wang, H., Xue, L., Dong, Y., Yang, L., Fan, R., Yu, X., Tian, tations. Ann. Dermatol. Venereol. 139(Suppl 4), S166–S169.
X., Ma, S., and Smith, G.W. (2012). Coat color determination by Guinot, C., Cheffai, S., Latreille, J., Dhaoui, M.A., Youssef, S., Jaber,
miR-137 mediated down-regulation of microphthalmia-associated K., Nageotte, O., and Doss, N. (2010). Aggravating factors for
transcription factor in a mouse model. RNA 18, 1679–1686. melasma: a prospective study in 197 Tunisian patients. J. Eur.
Dynoodt, P., Mestdagh, P., Van Peer, G., Vandesompele, J., Acad. Dermatol. Venereol. 24, 1060–1069.
Goossens, K., Peelman, L.J., Geusens, B., Speeckaert, R.M., Gupta, A.K., Gover, M.D., Nouri, K., and Taylor, S. (2006). The
Lambert, J.L., and Van Gele, M.J. (2013). Identification of miR-145 treatment of melasma: a review of clinical trials. J. Am. Acad.
as a key regulator of the pigmentary process. J. Invest. Dermatol. Dermatol. 55, 1048–1065.
133, 201–209. Ha, T.Y. (2011). The role of microRNAs in regulatory T cells and in the
Ediger, T.R., Kraus, W.L., Weinman, E.J., and Katzenellenbogen, B.S. immune response. Immune Netw. 11, 11–41.
(1999). Estrogen receptor regulation of the Na+/H+ exchange Hachiya, A., Kobayashi, A., Yoshida, Y., Kitahara, T., Takema, Y., and
regulatory factor. Endocrinology 140, 2976–2982. Imokawa, G. (2004). Biphasic expression of two paracrine
Eimpunth, S., Wanitphadeedecha, R., and Manuskiatti, W. (2013). A melanogenic cytokines, stem cell factor and endothelin-1, in
focused review on acne-induced and aesthetic procedure-related ultraviolet B-induced human melanogenesis. Am. J. Pathol. 165,
postinflammatory hyperpigmentation in Asians. J. Eur. Acad. 2099–2109.
Dermatol. Venereol. 27(Suppl 1), 7–18. Handel, A.C., Miot, L.D., and Miot, H.A. (2014a). Melasma: a clinical
Eimpunth, S., Wanitphakdeedecha, R., Triwongwaranat, D., Varothai, and epidemiological review. An. Bras. Dermatol. 89, 771–782.
S., and Manuskiatti, W. (2014). Therapeutic outcome of melasma Handel, A.C., Lima, P.B., Tonolli, V.M., Miot, L.D., and Miot, H.A.
treatment by dual-wavelength (511 and 578 nm) laser in patients (2014b). Risk factors for facial melasma in women: a case–control
with skin phototypes III–V. Clin. Exp. Dermatol. 39, 292–297. study. Br. J. Dermatol. 171, 588–594.
Eller, M.S., Ostrom, K., and Gilchrest, B.A. (1996). DNA damage Hearing, V.J., and Tsukamoto, K. (1991). Enzymatic control of
enhances melanogenesis. Proc. Natl Acad. Sci. USA 93, 1087– pigmentation in mammals. FASEB J. 5, 2902–2909.
1092. Hernandez-Barrera, R., Torres-Alvarez, B., Castanedo-Cazares, J.P.,
Elling, S.V., and Powell, F.C. (1997). Physiological changes in the skin Oros-Ovalle, C., and Moncada, B. (2008). Solar elastosis and
during pregnancy. Clin. Dermatol. 15, 35–43. presence of mast cells as key features in the pathogenesis of
Famenini, S., Gharavi, N.M., and Beynet, D.P. (2014). Finasteride melasma. Clin. Exp. Dermatol. 33, 305–308.
associated melasma in a caucasian male. J. Drugs Dermatol. 13, Hexsel, D., Arellano, I., and Rendon, M. (2006). Ethnic considerations
484–486. in the treatment of Hispanic and Latin-American patients with
Feucht, K.A., Walker, M.J., Das Gupta, T.K., and Beattie, C.W. hyperpigmentation. Br. J. Dermatol. 156(Suppl 1), 7–12.
(1988). Effect of 17 beta-estradiol on the growth of estrogen Hexsel, D., Lacerda, D.A., Cavalcante, A.S. et al. (2014). Epidemiol-
receptor-positive human melanoma in vitro and in athymic mice. ogy of melasma in Brazilian patients: a multicenter study. Int. J.
Cancer Res. 48, 7093–7101. Dermatol. 53, 440–444.
Filipowicz, W., Bhattacharyya, S.N., and Sonenberg, N. (2008). Ho, S.G., and Chan, H.H. (2009). The Asian dermatologic patient:
Mechanisms of post-transcriptional regulation by microRNAs: are review of common pigmentary disorders and cutaneous diseases.
the answers in sight? Nat. Rev. Genet. 9, 102–114. Am. J. Clin. Dermatol. 10, 153–168.
Foldes, E.G. (1988). Pharmaceutical effect of contraceptive pills on Hsieh, J.C., Kodjabachian, L., Rebbert, M.L., Rattner, A., Smallwood,
the skin. Int. J. Clin. Pharmacol. Ther. Toxicol. 26, 356–359. P.M., Samos, C.H., Nusse, R., Dawid, I.B., and Nathans, J. (1999).

658 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Melasma pathogenesis

A new secreted protein that binds to Wnt proteins and inhibits their melanogenesis via MITF as a direct target. J. Invest. Dermatol.
activities. Nature 398, 431–436. 134, 1075–1082.
Im, S., Kim, J., On, W.Y., and Kang, W.H. (2002). Increased Kim, N.H., Choi, S.H., Lee, T.R., and Lee, A.Y. (2014c). Cadherin 11, a
expression of alpha-melanocyte-stimulating hormone in the miR-675 target, induces N-cadherin expression and epithelial-
lesional skin of melasma. Br. J. Dermatol. 146, 165–167. mesenchymal transition in melasma. J. Invest. Dermatol. 134,
Imokawa, G., Miyagishi, M., and Yada, Y. (1995). Endothelin-1 as a 2967–2976.
new melanogen: coordinated expression of its gene and the Kippenberger, S., Loitsch, S., Solano, F., Bernd, A., and Kaufmann, R.
tyrosinase gene in UVB-exposed human epidermis. J. Invest. (1998). Quantification of tyrosinase, TRP-1, and Trp-2 transcripts in
Dermatol. 105, 32–37. human melanocytes by reverse transcriptase-competitive multi-
Ing, N.H., and Tornesi, M.B. (1997). Estradiol up-regulates estrogen plex PCR–regulation by steroid hormones. J. Invest. Dermatol.
receptor and progesterone receptor gene expression in specific 110, 364–367.
ovine uterine cells. Biol. Reprod. 56, 1205–1215. KrupaShankar, D.S., Somani, V.K., Kohli, M. et al. (2014). A cross-
Jang, Y.H., Lee, J.Y., Kang, H.Y., Lee, E.S., and Kim, Y.C. (2010). sectional, multicentric clinico-epidemiological study of melasma in
Oestrogen and progesterone receptor expression in melasma: an India. Dermatol. Ther. (Heidelb) 4, 71–81.
immunohistochemical analysis. J. Eur. Acad. Dermatol. Venereol. Kunisada, T., Lu, S.Z., Yoshida, H. et al. (1998). Murine cutaneous
24, 1312–1316. mastocytosis and epidermal melanocytosis induced by ker-
Jee, S.H., Lee, S.Y., Chiu, H.C., Chang, C.C., and Chen, T.Y. (1994). atinocyte expression of transgenic stem cell factor. J. Exp. Med.
Effects of estrogen and estrogen receptor in normal human 187, 1565–1573.
melanocytes. Biochem. Biophys. Res. Commun. 199, 1407–1412. Lieberman, R., and Moy, L. (2008). Estrogen receptor expression in
Jian, D., Jiang, D., Su, J., Chen, W., Hu, X., Kuang, Y., Xie, H., Li, J., melasma: results from facial skin of affected patients. J. Drugs
and Chen, X. (2011). Diethylstilbestrol enhances melanogenesis via Dermatol. 7, 463–465.
cAMP-PKA-mediating up-regulation of tyrosinase and MITF in Longley, B.J. Jr, Morganroth, G.S., Tyrrell, L., Ding, T.G., Anderson,
mouse B16 melanoma cells. Steroids 76, 1297–1304. D.M., Williams, D.E., and Halaban, R. (1993). Altered metabolism
Jo, H.Y., Kim, C.K., Suh, I.B., Ryu, S.W., Ha, K.S., Kwon, Y.G., and of mast-cell growth factor (c-kit ligand) in cutaneous mastocytosis.
Kim, Y.M. (2009). Co-localization of inducible nitric oxide synthase N. Engl. J. Med. 328, 1302–1307.
and phosphorylated Akt in the lesional skins of patients with Longstaff, C. (1994). Studies on the mechanisms of action of
melasma. J. Dermatol. 36, 10–16. aprotinin and tranexamic acid as plasmin inhibitors and antifibri-
Kanechorn Na Ayuthaya, P., Niumphradit, N., Manosroi, A., and nolytic agents. Blood Coagul. Fibrinolysis 5, 537–542.
Nakakes, A. (2012). Topical 5% tranexamic acid for the treatment Lynde, C.B., Kraft, J.N., and Lynde, C.W. (2006). Topical treatments
of melasma in Asians: a double-blind randomized controlled clinical for melasma and postinflammatory hyperpigmentation. Skin Ther-
trial. J. Cosmet. Laser Ther. 14, 150–154. apy Lett. 11, 1–6.
Kang, W.H., Yoon, K.H., Lee, E.S., Kim, J., Lee, K.B., Yim, H., Sohn, Mahmoud, B.H., Ruvolo, E., Hexsel, C.L., Liu, Y., Owen, M.R.,
S., and Im, S. (2002). Melasma: histopathological characteristics in Kollias, N., Lim, H.W., and Hamzavi, I.H. (2010). Impact of long-
56 Korean patients. Br. J. Dermatol. 146, 228–237. wavelength UVA and visible light on melanocompetent skin. J.
Kang, H.Y., Hwang, J.S., Lee, J.Y., Ahn, J.H., Kim, J.Y., Lee, E.S., Invest. Dermatol. 130, 2092–2097.
and Kang, W.H. (2006). The dermal stem cell factor and c-kit are Mandry Pag an, R., and S
anchez, J.L. (2000). Mandibular melasma. P.
overexpressed in melasma. Br. J. Dermatol. 154, 1094–1099. R. Health Sci. J. 19, 231–234.
Kasinski, A.L., and Slack, F.J. (2011). MicroRNAs en route to the McLeod, S.D., Ranson, M., and Mason, R.S. (1994). Effects of
clinic: progress invalidating and targeting microRNAs for cancer estrogens on human melanocytes in vitro. J. Steroid Biochem.
therapy. Nat. Rev. Cancer 11, 849–864. Mol. Biol. 49, 9–14.
Kato, H., Araki, J., Eto, H., Doi, K., Hirai, R., Kuno, S., Higashino, T., Mione, M., and Bosserhoff, A. (2015). MicroRNAs in melanocyte
and Yoshimura, K. (2011). A prospective randomized controlled and melanoma biology. Pigment Cell Melanoma Res. 28, 340–
study of oral tranexamic acid for preventing postinflammatory 354.
hyperpigmentation after Q-switched ruby laser. Dermatol. Surg. Miot, L.D., Miot, H.A., Polettini, J., Silva, M.G., and Marques, M.E.
37, 605–610. (2010). Morphologic changes and the expression of alpha-me-
Kim, E.H., Kim, Y.C., Lee, E.S., and Kang, H.Y. (2007). The vascular lanocyte stimulating hormone and melanocortin-1 receptor in
characteristics of melasma. J. Dermatol. Sci. 46, 111–116. melasma lesions: a comparative study. Am. J. Dermatopathol.
Kim, N.H., Lee, C.H., and Lee, A.Y. (2010). H19 RNA downregulation 32, 676–682.
stimulated melanogenesis in melasma. Pigment Cell Melanoma Moin, A., Jabery, Z., and Fallah, N. (2006). Prevalence and awareness
Res. 23, 84–92. of melasma during pregnancy. Int. J. Dermatol. 45, 285–288.
Kim, N.H., Cheong, K.A., Lee, T.R., and Lee, A.Y. (2012). PDZK1 Molinar, V.E., Taylor, S.C., and Pandya, A.G. (2014). What’s new in
upregulation in estrogen-related hyperpigmentation in melasma. objective assessment and treatment of facial hyperpigmentation?
J. Invest. Dermatol. 132, 2622–2631. Dermatol. Clin. 32, 123–135.
Kim, J.E., Chang, S.E., Yeo, U.C., Haw, S., and Kim, I.H. (2013a). Muallem, M.M., and Rubeiz, N.G. (2006). Physiological and biological
Histopathological study of the treatment of melasma lesions using skin changes in pregnancy. Clin. Dermatol. 24, 80–83.
a low-fluence Q-switched 1064-nm neodymium:yttrium-alu- Na, J.I., Choi, S.Y., Yang, S.H., Choi, H.R., Kang, H.Y., and Park, K.C.
minium-garnet laser. Clin. Exp. Dermatol. 38, 167–171. (2013). Effect of tranexamic acid on melasma: a clinical trial with
Kim, J.Y., Lee, T.R., and Lee, A.Y. (2013b). Reduced WIF-1 histological evaluation. J. Eur. Acad. Dermatol. Venereol. 27, 1035–
expression stimulates skin hyperpigmentation in patients with 1039.
melasma. J. Invest. Dermatol. 133, 191–200. Nicolaidou, E., Antoniou, C., and Katsambas, A.D. (2007). Origin,
Kim, K.H., Bin, B.H., Kim, J. et al. (2014a). Novel inhibitory function of clinical presentation, and diagnosis of facial hypermelanoses.
miR-125b in melanogenesis. Pigment Cell Melanoma Res. 27, Dermatol. Clin. 25, 321–326.
140–144. Noh, T.K., Choi, S.J., Chung, B.Y., Kang, J.S., Lee, J.H., Lee, M.W.,
Kim, N.H., Choi, S.H., Kim, C.H., Lee, C.H., Lee, T.R., and Lee, A.Y. and Chang, S.E. (2014). Inflammatory features of melasma lesions
(2014b). Reduced MiR-675 in exosome in H19 RNA-related in Asian skin. J. Dermatol. 41, 788–794.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 659
Lee

Orme, M.L., Back, D.J., and Ball, S. (1989). Interindividual variation in tinea versicolor, and postinflammatory hyperpigmentation. Am.
the metabolism of ethynylestradiol. Pharmacol. Ther. 43, 251–260. Fam. Physician 68, 1963–1968.
Park, H.Y., and Gilchrest, B.A. (1999). Signaling pathways mediating Suzuki, I., Im, S., Tada, A., Scott, C., Akcali, C., Davis, M.B., Barsh,
melanogenesis. Cell. Mol. Biol. 45, 919–930. G., Hearing, V., and Abdel-Malek, Z. (1999). Participation of the
Passeron, T. (2013). Melasma pathogenesis and influencing factors – melanocortin-1 receptor in the UV control of pigmentation. J.
an overview of the latest research. J. Eur. Acad. Dermatol. Investig. Dermatol. Symp. Proc. 4, 29–34.
Venereol. 27(Suppl 1), 5–6. Tamega Ade, A., Miot, L.D., Bonfietti, C., Gige, T.C., Marques, M.E.,
Pathak, M.A., and Fanselow, D.L. (1983). Photobiology of melanin and Miot, H.A. (2013). Clinical patterns and epidemiological
pigmentation: dose/response of skin to sunlight and its contents. J. characteristics of facial melasma in Brazilian women. J. Eur. Acad.
Am. Acad. Dermatol. 9, 724–733. Dermatol. Venereol. 27, 151–156.
Pawelek, J.M., Chakraborty, A.K., Osber, M.P., Orlow, S.J., Min, Tamega Ade, A., Miot, H.A., Mocßo, N.P., Silva, M.G., Marques, M.E.,
K.K., Rosenzweig, K.E., and Bolognia, J.L. (1992). Molecular and Miot, L.D. (2015). Gene and protein expression of oestrogen-b
cascades in UV-induced melanogenesis: a central role for melan- and progesterone receptors in facial melasma and adjacent healthy
otropins? Pigment Cell Res. 5, 348–356. skin in women. Int. J. Cosmet. Sci. 37, 222–228.
Pelletier, G., and Ren, L. (2004). Localization of sex steroid receptors Thornton, M.J. (2002). The biological actions of estrogens on skin.
in human skin. Histol. Histopathol. 19, 629–636. Exp. Dermatol. 11, 487–502.
rez-Bernal, A., Mun
Pe ~oz-Pe
rez, M.A., and Camacho, F. (2000). Tomita, Y., Maeda, K., and Tagami, H. (1989). Mechanisms for
Management of facial hyperpigmentation. Am. J. Clin. Dermatol. hyperpigmentation in postinflammatory pigmentation, urticaria
1, 261–268. pigmentosa and sunburn. Dermatologica 179(Suppl 1), 49–53.
Poon, V.K., Huang, L., and Burd, A. (2005). Biostimulation of dermal 
Torres-Alvarez, B., Mesa-Garza, I.G., Castanedo-C azares, J.P.,
fibroblast by sublethal Q-switched Nd:YAG 532 nm laser: collagen Fuentes-Ahumada, C., Oros-Ovalle, C., Navarrete-Solis, J., and
remodeling and pigmentation. J. Photochem. Photobiol., B 81, Moncada, B. (2011). Histochemical and immunohistochemical
1–8. study in melasma: evidence of damage in the basal membrane.
Resnik, S.S. (1967). Melasma and other skin manifestations or oral Am. J. Dermatopathol. 33, 291–295.
contraceptives. Trans. N. Engl. Obstet. Gynecol. Soc. 21, 101–107. Valadi, H., Ekstro €m, K., Bossios, A., Sjo €strand, M., Lee, J.J., and
Rome ro-Graillet, C., Aberdam, E., Biagoli, N., Massabni, W., Ortonne, Lo€tvall, J.O. (2007). Exosome-mediated transfer of mRNAs and
J.P., and Ballotti, R. (1996). Ultraviolet B radiation acts through the microRNAs is a novel mechanism of genetic exchange between
nitric oxide and cGMP signal transduction pathway to stimulate cells. Nat. Cell Biol. 9, 654–659.
melanogenesis in human melanocytes. J. Biol. Chem. 271, 28052– Vermeulen, A., Giagulli, V.A., De Schepper, P., and Buntinx, A.
28056. (1991). Hormonal effects of a 5 alpha-reductase inhibitor (finas-
Rome ro-Graillet, C., Aberdam, E., Cle ment, M., Ortonne, J.P., and teride) on hormonal levels in normal men and in patients with
Ballotti, R. (1997). Nitric oxide produced by ultraviolet-irradiated benign prostatic hyperplasia. Eur. Urol. 20(Suppl 1), 82–86.
keratinocytes stimulates melanogenesis. J. Clin. Invest. 99, 635– Wiedemann, C., N€ agele, U., Schramm, G., and Berking, C. (2009).
642. Inhibitory effects of progestogens on the estrogen stimulation of
Sanchez, N.P., Pathak, M.A., and Sato, S. (1981). Melasma: a clinical, melanocytes in vitro. Contraception 80, 292–298.
light microscopic, ultrastructural, and immunofluorescence study. Yamaguchi, Y., Itami, S., Watabe, H., Yasumoto, K., Abdel-Malek,
J. Am. Acad. Dermatol. 4, 698–709. Z.A., Kubo, T., Rouzaud, F., Tanemura, A., Yoshikawa, K., and
Shin, J., Kim, J.H., and Kim, E.K. (2012). Repeated exposure of Hearing, V.J. (2004). Mesenchymal-epithelial interactions in the
human fibroblasts to UVR induces secretion of stem cell factor and skin: increased expression of dickkopf1 by palmoplantar fibroblasts
senescence. J. Eur. Acad. Dermatol. Venereol. 26, 1577–1580. inhibits melanocyte growth and differentiation. J. Cell Biol. 165,
Shin, J.U., Park, J., Oh, S.H., and Lee, J.H. (2013). Oral tranexamic 275–285.
acid enhances the efficacy of low-fluence 1064-nm quality- Yan, B., Liu, B., Zhu, C.D., Li, K.L., Yue, L.J., Zhao, J.L., Gong, X.L.,
switched neodymium-doped yttrium aluminum garnet laser treat- and Wang, C.H. (2013). microRNA regulation of skin pigmentation
ment for melasma in Koreans: a randomized, prospective trial. in fish. J. Cell Sci. 126, 3401–3408.
Dermatol. Surg. 39, 435–442. Yano, K., Kadoya, K., Kajiya, K., Hong, Y.K., and Detmar, M. (2005).
Sodhi, V.K., and Sausker, W.F. (1988). Dermatoses of pregnancy. Ultraviolet B irradiation of human skin induces an angiogenic
Am. Fam. Physician 37, 131–138. switch that is mediated by upregulation of vascular endothelial
Speeckaert, R., Van Gele, M., Speeckaert, M.M., Lambert, J., and growth factor and by downregulation of thrombospondin-1. Br. J.
van Geel, N. (2014). The biology of hyperpigmentation syndromes. Dermatol. 152, 115–121.
Pigment Cell Melanoma Res. 27, 512–524. Yoshida, M., Takahashi, Y., and Inoue, S. (2000). Histamine induces
Sperzel, M., and Huetter, J. (2007). Evaluation of aprotinin and melanogenesis and morphologic changes by protein kinase A
tranexamic acid in different in vitro and in vivo models of activation via H2 receptors in human normal melanocytes. J.
fibrinolysis, coagulation and thrombus formation. J. Thromb. Invest. Dermatol. 114, 334–342.
Haemost. 5, 2113–2118. Yoshida, M., Hirotsu, S., Nakahara, M., Uchiwa, H., and Tomita, Y.
Stratigos, A.J., and Katsambas, A.D. (2004). Optimal management of (2002). Histamine is involved in ultraviolet B-induced pigmentation
recalcitrant disorders of hyperpigmentation in dark-skinned of guinea pig skin. J. Invest. Dermatol. 118, 255–260.
patients. Am. J. Clin. Dermatol. 5, 161–168. Zhu, Z., He, J., Jia, X. et al. (2010). MicroRNA-25 functions in
Stulberg, D.L., Clark, N., and Tovey, D. (2003). Common hyperpig- regulation of pigmentation by targeting the transcription factor
mentation disorders in adults: part II. Melanoma, seborrheic MITF in Alpaca (Lama pacos) skin melanocytes. Domest. Anim.
keratoses, acanthosis nigricans, melasma, diabetic dermopathy, Endocrinol. 38, 200–209.

660 ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

You might also like