The official journal of INTERNATIONAL FEDERATION OF PIGMENT CELL SOCIETIES · SOCIETY FOR MELANOMA RESEARCH

PIGMENT CELL & MELANOMA Research Skin phototype: a new perspective Vittoria Maresca, Enrica Flori and Mauro Picardo

DOI: 10.1111/pcmr.12365 Volume 28, Issue 4, Pages 378–389

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

REVIEW

Pigment Cell Melanoma Res. 28; 378–389

Skin phototype: a new perspective Vittoria Maresca, Enrica Flori and Mauro Picardo Laboratory of Cutaneous Physiopathology and Integrated Centre of Metabolomics Research, San Gallicano Dermatologic Institute, Rome, Italy

KEYWORDS phototype/melanocytes/MC1R/a-melanocyte-stimulating hormone/ultraviolet radiation

CORRESPONDENCE: M. Picardo, e-mail: [email protected]

PUBLICATION DATA Received 9 January 2015, revised and accepted for publication 16 March 2015, published online 18 March 2015 doi: 10.1111/pcmr.12365

Summary Cutaneous phototype is considered mainly related to cutaneous pigmentation and to the eumelanin/ pheomelanin ratio, which is mostly genetically determined by the melanocortin 1 receptor (MC1R) polymorphisms. However, data in literature indicate that, in addition to stimulation of eumelanin synthesis, the MC1R signalling activates antioxidant, DNA repair and survival pathways. New emerging aspects regarding photoprotection and skin phototypes are going beyond those features connected to the melanin content in the skin. Important new findings link the MC1R to nuclear receptors activation, shedding light on new extramelanogenic effects dependent on the a-melanocyte-stimulating hormone (a-MSH) activity and new ways through which such functions are modulated. These evidences indicate that several factors including melanin play a part in defining the basis for individual sun sensitivity, suggesting that the cutaneous phototype represents a ‘biochemical fingerprint’.

Introduction Skin phototype is a clinical classification system, developed by Thomas B. Fitzpatrick, based on self-reported erythema sensitivity and tanning ability (Fitzpatrick, 1988). The phototype is graded on a number (from I to VI) which, to some extent, reflects the degree of colour in the skin and its level of sensitivity to the damage generated by ultraviolet radiation (UV) (Del Bino and Bernerd, 2013; Fitzpatrick, 1988). Such classification has proven to be useful in indicating photoinduced skin cancer risk (Del Bino and Bernerd, 2013; Fitzpatrick, 1988; van der Leest  et al., 2011; Leiter and Garbe, 2008; Li et al., 2012; Mahe et al., 2011; dos Santos Silva et al., 2009). Constitutive skin colour designates a genetically determined level, type and distribution of melanin (Rouzaud et al., 2005; Sturm, 2009; Sturm et al., 2003), which is considered the main photoprotective agent against UV-induced deleterious effects (Kadekaro et al., 2003; Meredith and Sarna, 2006; Prota, 1997; Rouzaud et al., 2005). Several experimental data demonstrated that cutaneous pigmentation is regulated by a complex intercellular network, in which both keratinocytes and fibroblasts synthesize hormones, growth factors and cytokines, able to modulate melano cyte activity (Cardinali et al., 2008, 2012; Cario-Andre et al., 2006; Kovacs et al., 2010; Nordlund, 2007). More-

378

over, evidence suggests that skin phototype and the degree of photoprotection associated with it, is a complex strategy, in which melanins represent one of the elements (Bessou-Touya et al., 1998; Briganti et al., 2012;  et al., 1999, 2005; Maresca et al., 2006, Cario-Andre 2008, 2009; Picardo et al., 1999; Schallreuter and Wood, 2001). This review attempts to shed light on new aspects relating to the concepts of photoprotection and skin phototype, going beyond those aspects connected to the melanin content in skin and the filter effect carried out by it. Skin pigmentation: an adaptive characteristic of human skin One adaptive characteristic which is peculiar to human skin has been the gradual loss of hair on the body surface, linked to the appearance of skin pigmentation in the hairless areas (Jablonski and Chaplin, 2000, 2013). This event coincided with the transfer of Homo erectus from the African forests to the savannah. Travelling over long distances in sunny regions, these hominins needed to modify their way of thermoregulating. The first requirement was met by the number of sweat glands spread all over the body and the reduction of the body surface covered by hair. The second requirement of protecting the skull and therefore the brain from overheating, again due to solar exposure, was met by the concentration of ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

A new point of view of phototype

hair on the head (Ibraimov, 2007; Jablonski and Chaplin, 2000; Juzeniene et al., 2009; Pagel and Bodmer, 2003). After the hair had fallen out, human skin, which was initially light like that of current great apes (Jablonski and Chaplin, 2000; Westerhof, 2007), gradually began to pigment (Elias et al., 2009; Jablonski and Chaplin, 2000; Juzeniene et al., 2009). Various hypotheses, not mutually exclusive, have been formulated to explain which selective pressure caused the change. Some of them proposed that the gradual levels of pigmentation in different geographical areas of the world would be the evolutionary result of a compromise between skin which is light enough to allow the synthesis of vitamin D, essential for calcium absorption, yet still dark enough to protect the inner layers of skin from damage caused by UV (destruction of folic acid, one of the B-group vitamins) (Jablonski and Chaplin, 2000, 2013, 2014; Juzeniene et al., 2009; Yuen and Jablonski, 2010). Another hypothesis proposed was that human pigmentation developed to provide an effective permeability barrier, a requirement for life in desiccating terrestrial environment (Elias and Williams, 2013; Elias et al., 2009, 2010; Man et al., 2014). Experimental evidence shows that darkly pigmented skin possess a more efficacious skin barrier than lightly pigmented skin (Gunathilake et al., 2009; Liu et al., 2010; Man et al., 2014). Moreover, in depigmented areas of vitiligo, the epidermal barrier recovery after stripping is significantly delayed (Liu et al., 2010). Finally, pigmented hairless mice (Skh2) display a superior permeability barrier in comparison with the non-pigmented albino (Skh1) mice (Man et al., 2014). The barrier competence reflects a more cohesive structure of adjoining corneocytes, coupled with its low water content and its acidic pH, which encourages the growth of normal flora, while providing a formidable distal layer of the innate immune system that combats the invasion of pathogens. (Brogden et al., 2012; Drake et al., 2008; Elias, 2007; Elias and Williams, 2013; Gunathilake et al., 2009; Mackintosh, 2001). According to these disparate hypotheses, skin pigmentation is therefore an element which is closely linked to general skin homeostasis: not only a filter which protects the body from UV, but constituting an efficient barrier which prevents xerosis, protects against pathogens and indirectly contributes to thermoregulation. Melanogenesis: a biosynthetic means of providing protection, not only through the production of melanin Regulation of melanogenesis is a quite complex mechanism. Locally, keratinocytes and fibroblasts are involved through the production of proopiomelanocortins (POMCs), growth factors, cytokines and reactive oxygen species. Systemic hormones such as corticosteroids and oestrogens also influence the pigmentation process (Imokawa, 2004; Kondo and Hearing, 2011; Schallreuter et al., 2008; Yamaguchi and Hearing, 2009). The a-MSH/ MC1R interaction triggers cAMP/PKA signalling, the most ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

relevant pathway for melanin synthesis. Moreover, a-MSH is able to control melanogenesis also independently of MC1R, possibly by acting directly in melanosomes (Schallreuter et al., 2008). Among the other melanocortins, ACTH, the most abundant melanocortin in the human epidermis, interacts with MC1R and stimulates the phosphatidylinositol (PI(4,5)P2/PLC-beta) pathway and the intracellular release of calcium from the RER. The calcium release promotes the active transport of L-phenylalanine via large amino acid transporter 1 (LAT1) and its turnover via phenylalanine hydroxylase activity (PAH), in the presence of the cofactor 6BH4, providing sufficient concentrations of L-tyrosine to sustain melanogenesis (Schallreuter and Wood, 1999; Schallreuter et al., 2004). The correlation between epidermal PAH activities and 6BH4 levels with skin phototypes I-VI supports their close relationship with skin pigmentation (Schallreuter et al., 1994, 1997). The PI(4,5) P2/PLC-beta pathway controls also the activation of PKCb which in turn activates tyrosinase by phosphorylating serine residues in its cytoplasmic domain (Park et al., 1999). The same pathway is also activated by catecholamines (Grando et al., 2006). Following UV irradiation, p53 increases melanogenesis by inducing the expression of POMC in keratinocytes and by directly stimulating the genes encoding tyrosinase and tyrosinase-related protein-1 (TYRP-1) in melanocyte (Cui et al., 2007; Khlgatian et al., 2002; Kondo and Hearing, 2011; Nylander et al., 2000). Moreover, a mechanism involving H2O2 in the regulation of tyrosinase via p53 through the transcription of hepatocyte nuclear factor 1a has been proposed (Schallreuter et al., 2003). The nitric oxide (NO) produced in melanocytes after UV is considered as a major intra- and extracellular messenger molecule. NO elicits its effects through the activation of a soluble guanylate cyclase, leading to an increase in intracellular cGMP content and the activation of cGMPdependent protein kinase, stimulating melanogenesis (Busc a and Ballotti, 2000). Also H2O2, in the micromolar range, is able to act as a signal mediator, up-regulating a plethora of transcription factors including p53, MITF, NF-kB and the antioxidant enzymes catalase, thioredoxin reductase, glutathione reductase and methionine sulfoxide reductase (Schallreuter et al., 2008). DNA damage and DNA repair play relevant roles in the UV-induced melanogenic response of melanocytes. In particular, thymidine dimmers (pT-pT) increase p53 expression and up-regulate the expression of tyrosinase and TRP1 (Arad et al., 2006; Gilchrest and Eller, 1999). Finally, a number of new factors that are involved in melanogenesis have recently been reported, such as factors regulating melanosome pH and ion transport (Bellono and Oancea, 2014; Cheli et al., 2009; Fuller et al., 2001; Kondo and Hearing, 2011; Schallreuter et al., 2008). Research in the fields of pigmentation and dermatology essentially focuses on melanin as a filter, types of melanin (whether eu or pheomelanin) and the correlation 379

Maresca et al.

with skin tumours in response to UV exposure. In particular, eumelanin, characteristic of darkly pigmented skin, acts as a filter against UV (d’Ischia et al., 2013; Kadekaro et al., 2003; Meredith and Sarna, 2006) and possesses scavenger properties towards the UV-induced free radical species (Kadekaro et al., 2003; Meredith and Sarna, 2006; Prota, 1997). On the other hand, pheomelanin, predominantly present in individuals with pale skin and red or fair hair, presents different properties, being a less effective filter against UV (Prota, 1997), and acting as an endogenous photosensitizer (Hill and Hill, 2000; d’Ischia et al., 2013; Kadekaro et al., 2003; Maresca et al., 2006; Prota, 1997; Rouzaud et al., 2005). Moreover, the interest in colourless intermediates of melanogenesis has traditionally been focused on their role as pigment precursors. However, specific melanin intermediates may exert, per se, a protective function not necessarily related to their capacity to act as filters (d’Ischia et al., 2013). The diffusible major intermediate in eumelanin biosynthesis 5,6-dihydroxyindole (DHICA) exerts an inhibitory effect against lipid peroxidation (Gauden et al., 2008; Memoli et al., 1997; Novellino et al., 1998, 1999; Zhang et al., 2000). On human keratinocytes, DHICA induces the antioxidant defence systems and differentiation and significantly reduces the UV-induced apoptosis (Kovacs et al., 2012). Therefore, DHICA mediates cell protection mechanisms in melanocytes and in the surrounding keratinocytes, functioning as a diffusible chemical messenger in the context of the paracrine interactions between epidermal cells, regulating skin homeostasis and defence. Moreover, melanin may protect against H2O2-induced DNA strand breaks in both melanocytes and keratinocytes and through its ability to bind Ca2+ (Hoogduijn et al., 2004).

MC1R: a key regulator of pigmentation The Melanocortin 1 receptor (MC1R) is the only member of the family of seven-transmembrane G-protein-coupled melanocortin receptors (MCRs) expressed on the membrane of melanocytes, with a key role in the regulation of €hm et al., pigmentation (Abdel-Malek et al., 2014; Bo n et al., 2014; Nasti and Timares, 2006; Garcıa-Borro 2014; Rouzaud and Hearing, 2005; Slominski et al., 2004; Sturm, 2002, 2009; Sturm et al., 2003). The activation of this receptor by a-melanocyte-stimulating hormone (a-MSH) or adrenocorticotropic hormone (ACTH), two peptidic hormones produced by keratinocytes and deriving from the processing of proopiomelanocortin, triggers a mechanism of signal transduction which involves the adenylyl-cyclase via Gs protein and leads to increased intracellular cAMP levels and activation of protein kinase-A (PKA). The cAMP/PKA signal transduction pathway is the main pathway responsible for the melanogenic actions of a-MSH (Busca and Ballotti, 2000; n et al., 2014; Im D’Orazio and Fisher, 2011; Garcıa-Borro et al., 1998). PKA phosphorylates the cAMP-responsive 380

element-binding protein (CREB) which in turn induces the microphthalmia-associated transcription factor (MITF), a master regulator of melanocyte development, survival and differentiation (Cheli et al., 2010; Levy et al., 2006) and a key transcription factor for the expression of tyrosinase and TRP-1/TRP-2 (Bertolotto et al., 1998; Busc a and Ballotti, 2000; Cheli et al., 2010; Yamaguchi et al., 2007). In addition, upon a-MSH stimulation, MC1R activates the extracellular signal-regulated kinases (ERK) ERK1/ERK2 mitogen-activate protein kinase (MAPK) n et al., 2014; pathway (Busc a et al., 2000; Garcıa-Borro Herraiz et al., 2009), which also plays a key role in melanin synthesis, at least in part, through the regulation of MITF activation and stability (Englaro et al., 1998; n et al., 2014; Hemesath et al., 1998; Herraiz Garcıa-Borro et al., 2011; Wu et al., 2000). a-MSH-induced PKA activation also triggers the p38MAPK pathway, implicated in the regulation of important steps in melanocyte differentiation (Newton et al., 2007; Singh et al., 2005; Smalley and Eisen, 2000, 2002). Moreover, a-MSH signalling impinges on the peroxisome proliferator-activated receptor gamma coactivator proteins (PGC-1s), transcriptional coactivators of most of the nuclear receptors, and thereby playing important roles in numerous metabolic processes (Chinsomboon et al., 2009; Spiegelman, 2007; Yoon et al., 2001). The PKA-dependent activation of PGC1s is required for the induction of MITF transcription, providing an interesting link between metabolism and pigmentation (Ronai, 2013; Shoag et al., 2013). The finding that MITF also contributes to PGC-1a transcription suggests the existence of a feedback loop for regulation of the PGC-1a-MITF axis (Haq et al., 2013; Ronai, 2013; Vazquez et al., 2013). It is noteworthy that the transduction through cAMP/PKA pathway does not include only the activated MCIR but also other receptors such as b2adrenoceptor, the muscarinic receptors (M1, M3, M5), the a- and b-oestrogen receptors and the CRF/CRF-R1 signal (Schallreuter et al., 2008). Stimulation of MC1R induces the synthesis of photoprotective eumelanin. MC1R is highly polymorphic and its variants can explain differences in the wide range of human skin pigmentation. Melanocytes that express the consensus sequence for MC1R are darkly pigmented. However, more than 100 gene polymorphisms have been n et al., 2014; described (Chen et al., 2014; Garcıa-Borro rez Oliva et al., 2009), some of which have been Pe demonstrated to be loss-of-function variants (Herraiz et al., 2012; Sturm, 2009; Sturm et al., 2003). Some of these variants are closely associated with the red hair and fair skin phenotype (RHC-variants), a condition that is determined by the prevalent level of pheomelanin synthesis. This can place the individual at higher risk of malignant skin melanomas. MC1R: not only a regulator of pigmentation The information available about MC1R and its ligand a-MSH points to an interaction capable of coordinating ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

A new point of view of phototype

several homeostatic skin functions, which go well beyond the mere control of quantitatively and qualitatively photoprotective melanin (Abdel-Malek et al., 2014; Garcıan et al., 2014). The MCRs family actually represents Borro an old system of receptors, which are stimulated by derivatives of proopiomelanocortin (Abdel-Malek et al., €hm et al., 2006; Garcıa-Borro n et al., 2005, 2014; Bo 2014; Rouzaud and Hearing, 2005; Sturm, 2009). They are expressed in several tissues in man and other mammals. The members of this receptor family are involved in an extraordinarily diverse number of physiological functions, including pigmentation, energy homeostasis, exocrine secretion, inflammation and €hm et al., 2006; Catania, cardiovascular regulation (Bo 2007; Rodrigues et al., 2014; Slominski et al., 2004). Even though they are expressed in different cells and tissues, as G-protein-coupled receptors, they share the same ancestor pathways: the cAMP/PKA signal pathway and the phosphatidylinositol signal pathway, which do not always necessarily coexist under the same receptor (Collins et al., 1992; Dennis et al., 1991; Gilman, 1995; Irvine, 1992; Krumins and Gilman, 2006; Yang, 2011). Through these different transduction pathways, expressed in different tissues, they can promote similar protective strategies. Exposure of the skin to UV radiation induces the production of a-MSH by the keratinocytes, which, in turn, stimulate eumelanin synthesis and melanocyte survival and proliferation by binding and activating n et al., MC1R (Abdel-Malek et al., 2014; Garcıa-Borro 2014). a-MSH is also able to stimulate protective biological effects in non-melanocytic populations, such as keratinocytes and fibroblasts. These amelanotic systems provide information about potential photoprotective properties of a-MSH that go beyond its ability to regulate melanogenesis. In dermal fibroblasts, a-MSH binding to MC1R reduces the transforming growth factor (TGF-b1)€hm et al., induced synthesis of collagen types I and III (Bo 2004, 2006) and reduces skin fibrosis in mouse models of scleroderma (Kokot et al., 2009b). In keratinocytes, a-MSH enhances UV-induced DNA repair (Dong et al., 2010) and increases the expression of the transcription factors Nrfs, which exert a key regulatory role in the expression of ROS-detoxifying enzyme (Kokot et al., 2009a). Furthermore, the activation of the a-MSH/MC1R interaction has been implicated in the regulation of extracellular matrix homeostasis (Muffley et al., 2011). Finally, a-MSH is also known to exert anti-inflammatory effects in different cell populations, not only in the skin (Brzoska et al., 2008; Catania, 2007; Kokot et al., 2009a; Mastrofrancesco et al., 2010; Raap et al., 2003; Scholzen et al., 2003). MC1R: a regulator of oxidative stress and DNA repair Further emphasis has been given to the role of MC1R in controlling oxidative stress (Kadekaro et al., 2010; Maresca et al., 2010; Song et al., 2009) and in maintaining genomic integrity (Denat et al., 2014; Hauser et al., 2006; ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Kadekaro et al., 2012; Yin et al., 2014) in melanocytes. These two biological aspects are closely linked because oxidative stress, particularly when generated following UV exposure, is one of the main causes of DNA damage and one of the principal mechanisms which trigger cancer. a-MSH is able to counteract UV-induced oxidative damage by decreasing intracellular levels of hydrogen peroxide (Haycock et al., 2000; Kadekaro et al., 2005, 2010; Song et al., 2009) and enhances the repair of DNA photoproducts in primary cultures of human melanocytes €hm et al., 2005; Kadekaro et al., 2005, 2010; Smith (Bo et al., 2008; Song et al., 2009; Swope et al., 2014). Moreover, a-MSH increases phosphorylation of p53 in UVR-irradiated melanocytes, which, in turn, leads to p53 accumulation and activation. The increase in p53 is significant in reducing ROS generation, and consequently oxidative DNA damage, as well as in regulating the expression of enzymes that have a key role in base excision repair (BER), the main repair pathway for oxidative DNA damage (Denat et al., 2014; Kadekaro et al., 2012). Furthermore, the induction of the NR4A subfamily of nuclear receptors by MC1R signalling exerts a key role in promoting DNA repair in UV-irradiated melanocytes (Jagirdar et al., 2013; Smith et al., 2008; Yin et al., 2014). In addition, wild-type MC1R, but not its RHC-variants, significantly stabilizes the phosphatase and tensin homologue (PTEN) in UV-irradiated cells (Cao et al., 2013). In non-melanoma skin cancer, PTEN acts as an essential genomic gatekeeper through the regulation of the UV-induced DNA repair (Ming et al., 2011). Moreover, the interaction between MC1R and the b-arrestin (ARRB) family of cytosolic multifunctional adaptor proteins has been recently described (Abrisqueta et al., 2013), and b-arrestins are implicated in modulation of DNA damage signalling pathways (Hara et al., 2011). In association with melanin, the antioxidant defence system has to act in an integrated manner in order to minimize the damage caused by UV-generated free radicals within the cell (Briganti and Picardo, 2003). Some experimental evidence has shown a correlation between pigmentation and antioxidants. Thioredoxin reductase and catalase activities directly correlate with cutaneous phototype in different experimental models (Picardo et al., 1999; Schallreuter and Wood, 2001). In an ex vivo model of epidermal reconstructs, lightly pigmented reconstructs showed lower levels of catalase enzymatic activity and a higher concentration of peroxidable substrates in the cell membranes, in comparison with highly pigmented ones, making the first ones more susceptible to UV damage  et al., 1999; (Bessou-Touya et al., 1998; Cario-Andre Maresca et al., 2006). Among UV-generated reactive oxygen species, H2O2 plays a key role, both for its capacity to diffuse across cell membranes, reaching all cellular regions, and for its high reactivity (Bienert et al., 2006). In association with catalase, other antioxidant enzymes participate in the protection against oxidative stress. Thioredoxin reductase together with its electron 381

Maresca et al.

acceptors thioredoxin, thioredoxin peroxidases and glutathione reductase/glutathione coupled to glutathione peroxidase are involved in the removal of hydrogen peroxide deriving from enzymatic dismutation of superoxide anion r, 2001; Schallrecatalysed by SOD (Nordberg and Arne uter and Wood, 2001; Schallreuter et al., 2008). Nevertheless, in melanocytes, the role of Cat is critical because it is the first enzyme devoted to the neutralization of H2O2 (Yohn et al., 1991). The enzyme is very susceptible to peroxidizing agents, recovers slowly when UV damaged (Shindo and Hashimoto, 1997; Shindo et al., 1994) and it can be considered a marker of both acute and chronic oxidative stress. In primary cultures of human melanocytes, catalase expression and activity are directly correlated with the total melanin content and tyrosinase expression (Maresca et al., 2008). The close relationship between melanogenic activity and protection from hydrogen peroxide provided by catalase suggested MC1R as the element capable of coordinating the two strategies. (Maresca et al., 2010). a-MSH increases the expression of catalase in unirradiated as well as UV-irradiated cells (Maresca et al., 2010; Song et al., 2009). Moreover, in response to a-MSH treatment, catalase is rapidly induced and colocalizes with the melanin in the melanosomes. The association with melanosomes suggests a role of this enzyme in the protection of melanocytes themselves and possibly of the whole skin against UV, through the transfer of melanosomes into keratinocytes (Maresca et al., 2010). The protective effects of a-MSH are absent in melanocytes expressing loss-of-function MC1R (AbdelMalek et al., 2014; Denat et al., 2014; Kadekaro et al., 2012; Smith et al., 2008; Song et al., 2009), establishing the significance of the activated MC1R in protection of melanocytes from oxidative stress. The a-MSH/nuclear receptors connection: a link of physiological relevance Relatively few genetic targets of MC1R signalling have been identified independent of the pigmentation pathway (Smith et al., 2008). Emerging evidence suggests that the nuclear receptors family of transcriptional regulators has a role in the context of melanocyte biology. MC1R activation induces the transcription of the NR4A subfamily of nuclear receptors (Jagirdar et al., 2013; Smith et al., 2008; Yin et al., 2014), implicated in a wide range of biological processes including cell proliferation, differentiation, metabolism, inflammation and vascular diseases (Pearen and Muscat, 2010; Smith et al., 2008; Yin et al., 2014). More recently, the involvement of NR4A proteins in promoting DNA repair in several cell types has been documented (Malewicz et al., 2011; Ramirez-Herrick et al., 2011; Smith et al., 2008). The induction of these nuclear receptors by MC1R activation in melanocytic cells seems to be a key component of MC1R-mediated DNA repair following UVR (Smith et al., 2008). We recently identified an activating connection between a-MSH and the peroxisome proliferator-activated receptor-c (PPAR-c) 382

(Maresca et al., 2013) in melanocytic cells. PPAR-c is a transcription factor belonging to the nuclear receptor family of peroxisome proliferator-activated receptors (PPARs). These transcription factors are activated by lipid mediators, such as linoleic acid or lipid products of lipoxygenases (Montagner et al., 2011; Varga et al., 2011; Wahli and Michalik, 2012). Three PPAR isoforms have been discovered and described in literature: a, b/d and c (Varga et al., 2011), all of which are expressed in human and mouse epidermis (Michalik and Wahli, 2007; Sertznig et al., 2008). In the skin, PPARs and corresponding ligands have been shown to regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses, redox state and wound healing (Dubrac and Schmuth, 2011; Park et al., 2013; Polvani et al., 2012; Sertznig et al., 2008; Varga et al., 2011). In particular, PPARc induces differentiation and increases cell antioxidant defence in response to natural or pharmacological agonists, in melanocytes and melanoma cells (Flori et al., 2011; Grabacka et al., 2008; Kang et al., 2004; Lee et al., 2007; Okuno et al., 2010; Polvani et al., 2012). Moreover, PPARc activation increases cell antioxidant defence and counteracts senescence-like phenotype in primary cultures of human fibroblasts (Briganti et al., 2013, 2014). Mice lacking epidermal PPARc exhibit a marked increase in photocarcinogenesis and heightened UVB-induced apoptosis, inflammation and barrier dysfunction (Sahu et al., 2012). Accordingly, mice with loss of the PPARc heterodimerization partner RXRa exhibit increased keratinocytes apoptosis and augmented DNA damage in response to UVB exposure (Wang et al., 2011). Due to its key role in the control of cell proliferation and differentiation, natural or pharmacological inductors of PPAR-c are used in innovative therapeutic models for various forms of cancer, including melanomas (Botton et al., 2009; Grommes €ssner et al., 2002; Robbins and Nie, 2012; et al., 2004; Mo Toyoda et al., 2002). The a-MSH/PPAR-c connection appears to be a phylogenetically preserved link. In fact, in the presence of a wild-type MC1R, it is independent of the animal species of origin and it is not correlated with the transformation condition of the cell (Maresca et al., 2013). Many of the functions modulated by PPAR-c pharmacological inductors overlap with functions modulated by a-MSH (e.g. control of inflammation, proliferation and redox state) for which the activation mechanism is not known. The detailed analysis of the a-MSH/nuclear receptors connection and the underlying pathways could shed light on new extra-melanogenic effects dependent on the a-MSH/MC1R interaction and new ways through which such functions are modulated (Figure 1). MC1R: a regulator of lipid signalling Research on the role of lipids in cell and tissue biology is a booming, groundbreaking field, especially because it has been understood that these molecules can initiate and regulate signalling events that will decisively influence ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

A new point of view of phototype POMCs

Figure 1. Summary of the protective effects induced by POMCs in melanocytes. The activation of MC1R by POMCs triggers different mechanisms of signal transduction (cAMP/PKA, ERK1/2, p38, PI(4,5)P2/PLCb), promoting the activation of key transcription factors (MITF and PPAR-c). This cascade of events leads to DNA repair and antioxidant defence promotion, in addition to stimulating pigmentation.

Activated signal transduction pathways

Activated transcription factors

POMCs-mediated protective effects

development, cellular differentiation, metabolism and related function through the regulation of gene expression (Varga et al., 2011; Wymann and Schneiter, 2008). Growing evidence suggests the involvement of lipids mediators in skin homeostasis. For example, the endocannabinoid system (ECS) plays a key role in the control of proliferation, differentiation, apoptosis and cytokine, mediator and hormone production in various cell types  et al., 2009). As regards of the skin and appendages (Bıro lipid-mediated signalling and melanocyte function, a fully functional ECS is present in human melanocytes and it stimulates melanogenesis through the activation of p38and p42/44 MAPK pathways (Pucci et al., 2012). Moreover, the sphingolipid-enriched total lipid fraction from human placenta induces pigmentation in melanocytic cells through p38 MAPK via induction of tyrosinase gene expression (Saha et al., 2006, 2009; Singh et al., 2005). However, the link between lipid mediators and MC1R is an unexplored topic. We demonstrated that the activation of PPAR-c in response to a-MSH depends on the induction of the phosphatidylinositol (PI(4,5)P2/PLCb) signal pathway (Maresca et al., 2013). This finding sums up a series of sporadic studies conducted in the past, in which the lipid pathway was indirectly postulated, but never clearly described, after the stimulation of cells with a-MSH (Buffey et al., 1992; Eves et al., 2006; Haycock et al., 2000). Lipids generated in the (PI(4,5)P2/PLCb) signal pathway, such as linoleic and arachidonic acids, are the main lipid mediators involved in signal transduction mechanisms (Robbins and Nie, 2012; Varga et al., 2011). When they are detached from the glycerol backbone of membrane phospholipids, they trigger cascade reactions mainly connected with the control of proliferation and inflammation (Yuri et al., 2007). Therefore, the (PI(4,5)P2/ PLCb) signal pathway could subtend the capacity of MC1R to influence, through lipid mediators, melanogenesis and other functions which include proliferation rates, control and inflammation. Moreover, the influence of fatty acid composition of the cell membranes has been, so far, only partially considered in association with the pigmentation levels and MC1R functionality (Picardo, 2009; Pucci ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

cAMP/PKA

ERK1/2

MITF

PIGMENTATION

p38

PI(4,5)P2/PLC

PPAR-γγ

DNA REPAIR

ANTIOXIDANT DEFENCE SYSTEM

et al., 2012; Saha et al., 2009), but instead it could be fundamental in driving manifold biological aspects. Finally, in recent years, alterations in lipid metabolism seem to be a common metabolic feature of malignant cells, including melanoma. These modifications are thought to promote pathology by generating substrates for energy production and membrane synthesis, as well as signalling lipids that €ller-Decker and trigger pro-tumorigenic cascades (Mu €rstenberger, 2007; Sertznig et al., 2008; Wymann and Fu Schneiter, 2008).

The biochemical fingerprint of phototype Cutaneous pigmentation is considered the major photoprotective mechanism against sun-induced skin ageing and carcinogenesis. Clinical evaluation of skin photosusceptibility is essentially based on the classification of Fitzpatrick skin phototype and minimal erythema dose (MED), which reflect, to some extent, the degree of colour in the skin (Fitzpatrick, 1988). However, data in literature shows that in depigmented vitiligo skin, the (sub)erythematogenic UV doses correlated with skin type of the patients (Briganti et al., 2012; Caron-Schreinemachers et al., 2005) and that subjects affected by albinism do not show a higher incidence of melanoma compared to subjects with red phenotypic features (Greaves, 2014), suggesting that melanin is not the only factor responsible for UV sensitivity. All the evidences which have been discussed in this review indicate that factors other than melanin determine a network of metabolic interactions which work together for the photoprotection of the skin and the correct construction of the barrier function. From a global and up-to-date point of view, all these parameters play a part in defining a biochemical basis for individual sun sensitivity. Eumelanin and pheomelanin are obviously crucial, with their quantitative and qualitative contribution to photoprotection (Kadekaro et al., 2003; Maresca et al., 2006; Meredith and Sarna, 2006; Prota, 1997; Rosso et al., 2007; Vincensi et al., 1998; Wakamatsu et al., 2006). The final products of eumelanogenesis and pheomelanogenesis, and the chemical intermediates of the 383

Maresca et al.

two distinct biosynthetic pathways, contribute in different ways to promote photoprotection or alternatively to increase the phenomenon of cutaneous sun sensitivity (in the case of pheomelanin). Regarding eumelanogenesis, DHICA, due to its capacity to diffuse, is able to provide photoprotection also for keratinocytes, giving them antioxidant protection and promoting differentiation (Kovacs et al., 2012). In the case of pheomelanogenesis, different intermediates of this biosynthetic pathway in the hairs of red-haired subjects can be considered genetic markers associated with diverse gradations of ‘red’ phenotypes, with different photosensitizing potential. The endogenous photosensitising property commonly associated with pheomelanin reflects the capacity of this pigment to amplify the generation of free radicals, in response to exposure to UV radiation (Hill and Hill, 2000; Kadekaro et al., 2003; Maresca et al., 2006; Napolitano et al., 2014; Prota, 1997; Rouzaud et al., 2005). It is then the task of the chemical and enzymatic antioxidants, which collectively act as a ‘barrier’, to stop the reactivity of the free radicals which are produced in the skin following the impact of UV radiation and in particular of UVA (Briganti and Picardo, 2003; Natarajan et al., 2014). If, however, the UV dose is massive and/or the cutaneous antioxidant defence system is impaired, oxidative stress takes place, specific signal transduction pathways and cell cycle control elements are influenced and the initial steps of carcinogenesis are promoted (Godic et al., 2014; Sander et al., 2003). In association with melanins, antioxidants are therefore well placed to participate in skin photoprotection. The mechanisms to counteract, reduce or repair skin photodamage should also include DNA repair systems, extracellular matrix homeostasis and control of inflammatory effects. Although these biochemical parameters were not directly involved in determining skin colour nor in the filter effect against UV, they nonetheless influence a complete substrate responsible for photosensitivity.

Perspectives The information available in literature and the evidence, which will be added over time, are already sufficient to describe a point of view which is changing. For many years, diverse experimental evidence has been contributing to create a ‘biochemical fingerprint of cutaneous phototype’, which does not only include the contribution of melanin. In this respect, the study of MC1R may become extremely interesting, especially if it begins to be considered as more than just ‘the principle regulator of melanogenic enzymes’, but also as a member of the ‘MCR family’, that is a family of receptors which carry out different functions in cells and €hm et al., 2006; Catania, 2007; Rodrigues et al., tissues (Bo 2014; Slominski et al., 2004), functions which all produce cellular protection and well-being. As melanoma susceptibility is often related to the presence of ‘loss-of-function’ MC1R variants in some subjects’ genomes, what impor384

tance might the presence of one of these variants have in conditioning the response of PPAR-c, or in other aspects of cellular well-being? Knowledge of these aspects could broaden the concept of ‘loss of function’, related to MC1R, extending it to effects which are not exclusively pigmentary. Experimental evidences which have been reported in this review, together with other data present in literature and new evidence which will be added, are slowly modifying the concept of ‘phototype’, extending its meaning to biological and molecular parameters; therefore, no longer merely a clinical risk parameter for melanomas and other photoinducible diseases.

Acknowledgement lo€ıse Kerr-Wilson and Ms Lucia De Caprio for We thank Ms He language editing.

References Abdel-Malek, Z.A., Swope, V.B., Starner, R.J., Koikov, L., Cassidy, P., and Leachman, S. (2014). Melanocortins and the melanocortin 1 receptor, moving translationally towards melanoma prevention. Arch. Biochem. Biophys. 563, 4–12. rez Oliva, A.B., Sanchez-Laorden, B.L., Abrisqueta, M., Herraiz, C., Pe nez-Cervantes, C., and Garcıa-Borro n, J.C. Olivares, C., Jime (2013). Differential and competitive regulation of human melanocortin 1 receptor signaling by b-arrestin isoforms. J. Cell Sci. 126, 3724–3737. Arad, S., Konnikov, N., Goukassian, D.A., and Gilchrest, B.A. (2006). T-oligos augment UV-induced protective responses in human skin. FASEB J. 20, 1895–1897. Bellono, N.W., and Oancea, E.V. (2014). Ion transport in pigmentation. Arch. Biochem. Biophys. 563, 35–41. Bertolotto, C., Busc a, R., Abbe, P., Bille, K., Aberdam, E., Ortonne, J.P., and Ballotti, R. (1998). Different cis-acting elements are involved in the regulation of TRP1 and TRP2 promoter activities by cyclic AMP: pivotal role of M boxes (GTCATGTGCT) and of microphthalmia. Mol. Cell. Biol. 18, 694–702. Bessou-Touya, S., Picardo, M., Maresca, V., Surleve-Bazeille, J.E., Pain, C., and Taieb, A. (1998). Chimeric human epidermal reconstructs to study the role of melanocytes and keratinocytes in pigmentation and photo-protection. J. Invest. Dermatol. 111, 1103–1108. Bienert, G.P., Schjoerring, J.K., and Jahn, T.P. (2006). Membrane transport of hydrogen peroxide. Biochim. Biophys. Acta 1758, 994– 1003. , T., To th, B.I., Hasko , G., Paus, R., and Pacher, P. (2009). The Bıro endocannabinoid system of the skin in health and disease: novel perspectives and therapeutic opportunities. Trends Pharmacol. Sci. 30, 411–420. €hm, M., Raghunath, M., Sunderko €tter, C., Schiller, M., St€ Bo ander, S., €th, H.B., Schwarz, T., and Luger, T.A. Brzoska, T., Cauvet, T., Schio (2004). Collagen metabolism is a novel target of the neuropeptide alpha-melanocyte-stimulating hormone. J. Biol. Chem. 279, 6959– 6966. €hm, M., Wolff, I., Scholzen, T.E., Robinson, S.J., Healy, E., Luger, Bo T.A., Schwarz, T., and Schwarz, A. (2005). alpha-Melanocytestimulating hormone protects from ultraviolet-induced apoptosis and DNA damage. J. Biol. Chem. 280, 5795–5802. €hm, M., Luger, T.A., Tobin, D.J., and Garcıa-Borro n, J.C. (2006). Bo Melanocortin receptor ligands: new horizons for skin biology and clinical dermatology. J. Invest. Dermatol. 126, 1966–1975.

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

A new point of view of phototype Botton, T., Puissant, A., Bahadoran, P. et al. (2009). In vitro and in vivo anti-melanoma effects of ciglitazone. J. Invest. Dermatol. 129, 1208–1218. Briganti, S., and Picardo, M. (2003). Antioxidant activity, lipid peroxidation and skin diseases. What’s new. J. Eur. Acad. Dermatol. Venereol. 17, 663–669. Briganti, S., Caron-Schreinemachers, A.L., Picardo, M., and Westerhof, W. (2012). Anti-oxidant defence mechanism in vitiliginous skin increases with skin type. J. Eur. Acad. Dermatol. Venereol. 26, 1212– 1219. Briganti, S., Flori, E., Mastrofrancesco, A., Kovacs, D., Camera, E., Ludovici, M., Cardinali, G., and Picardo, M. (2013). Azelaic acid reduced senescence-like phenotype in photo-irradiated human dermal fibroblasts: possible implication of PPARc. Exp. Dermatol. 22, 41–47. Briganti, S., Flori, E., Bellei, B., and Picardo, M. (2014). Modulation of PPARc provides new insights in a stress induced premature senescence model. PLoS ONE 9, e104045. Brogden, N.K., Mehalick, L., Fischer, C.L., Wertz, P.W., and Brogden, K.A. (2012). The emerging role of peptides and lipids as antimicrobial epidermal barriers and modulators of local inflammation. Skin Pharmacol. Physiol. 25, 167–181. €hm, M. (2008). Brzoska, T., Luger, T.A., Maaser, C., Abels, C., and Bo Alpha-melanocyte-stimulating hormone and related tripeptides: biochemistry, antiinflammatory and protective effects in vitro and in vivo, and future perspectives for the treatment of immunemediated inflammatory diseases. Endocr. Rev. 29, 581–602. Buffey, J., Thody, A.J., Bleehen, S.S., and Mac Neil, S. (1992). Alphamelanocyte-stimulating hormone stimulates protein kinase C activity in murine B16 melanoma. J. Endocrinol. 133, 333–340. Busc a, R., and Ballotti, R. (2000). Cyclic AMP a key messenger in the regulation of skin pigmentation. Pigment Cell Res. 13, 60–69. Busc a, R., Abbe, P., Mantoux, F., Aberdam, E., Peyssonnaux, C., ne, A., Ortonne, J.P., and Ballotti, R. (2000). Ras mediates Eyche the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes. EMBO J. 19, 2900–2910. Cao, J., Wan, L., Hacker, E. et al. (2013). MC1R is a potent regulator of PTEN after UV exposure in melanocytes. Mol. Cell 51, 409–422. Cardinali, G., Bolasco, G., Aspite, N., Lucania, G., Lotti, L.V., Torrisi, M.R., and Picardo, M. (2008). Melanosome transfer promoted by keratinocyte growth factor in light and dark skin-derived keratinocytes. J. Invest. Dermatol. 128, 558–567. Cardinali, G., Kovacs, D., and Picardo, M. (2012). Mechanisms underlying post-inflammatory hyperpigmentation: lessons from solar lentigo. Ann. Dermatol. Venereol. 139, S148–S152. , M., Bessou, S., Gontier, E., Maresca, V., Picardo, M., Cario-Andre and Ta€ıeb, A. (1999). The reconstructed epidermis with melanocytes: new tool to study pigmentation and photo-protection. Cell. Mol. Biol. (Noisy-le-grand) 45, 931–942. , M., Briganti, S., Picardo, M., Nikaido, O., de Verneuil, H., Cario-Andre and Ta€ıeb, A. (2005). Polyunsaturated fatty acids partially reproduce the role of melanocytes in the epidermal melanin unit. Exp. Dermatol. 14, 194–201. , M., Pain, C., Gauthier, Y., Casoli, V., and Taieb, A. Cario-Andre (2006). In vivo and in vitro evidence of dermal fibroblasts influence on human epidermal pigmentation. Pigment Cell Res. 19, 434–442. Caron-Schreinemachers, A.L., Kingswijk, M.M., Bos, J.D., and Westerhof, W. (2005). UVB 311 nm tolerance of vitiligo skin increases with skin photo type. Acta Derm. Venereol. 85, 24–26. Catania, A. (2007). The melanocortin system in leukocyte biology. J. Leukoc. Biol. 81, 383–392. Cheli, Y., Luciani, F., Khaled, M., Beuret, L., Bille, K., Gounon, P., Ortonne, J.P., Bertolotto, C., and Ballotti, R. (2009). alphaMSH and Cyclic AMP elevating agents control melanosome pH through a

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

protein kinase A-independent mechanism. J. Biol. Chem. 284, 18699–18706. Cheli, Y., Ohanna, M., Ballotti, R., and Bertolotto, C. (2010). Fifteenyear quest for microphthalmia-associated transcription factor target genes. Pigment Cell Melanoma Res. 23, 27–40. Chen, H., Weng, Q.Y., and Fisher, D.E. (2014). UV signaling pathways within the skin. J. Invest. Dermatol. 134, 2080–2085. Chinsomboon, J., Ruas, J., Gupta, R.K., Thom, R., Shoag, J., Rowe, G.C., Sawada, N., Raghuram, S., and Arany, Z. (2009). The transcriptional coactivator PGC-1alpha mediates exercise-induced angiogenesis in skeletal muscle. Proc. Natl. Acad. Sci. USA 106, 21401–21406. Collins, S., Caron, M.G., and Lefkowitz, R.J. (1992). From ligand binding to gene expression: new insights into the regulation of Gprotein-coupled receptors. Trends Biochem. Sci. 17, 37–39. Cui, R., Widlund, H.R., Feige, E. et al. (2007). Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell 128, 853–864. Del Bino, S., and Bernerd, F. (2013). Variations in skin colour and the biological consequences of ultraviolet radiation exposure. Br. J. Dermatol. 169(Suppl 3), 33–40. Denat, L., Kadekaro, A.L., Marrot, L., Leachman, S.A., and AbdelMalek, Z.A. (2014). Melanocytes as instigators and victims of oxidative stress. J. Invest. Dermatol. 134, 1512–1518. Dennis, E.A., Rhee, S.G., Billah, M.M., and Hannun, Y.A. (1991). Role of phospholipase in generating lipid second messengers in signal transduction. FASEB J. 5, 2068–2077. Dong, L., Wen, J., Pier, E., Zhang, X., Zhang, B., Dong, F., Ziegler, N., Mysz, M., Armenta, R., and Cui, R. (2010). Melanocyte-stimulating hormone directly enhances UV-Induced DNA repair in keratinocytes by a xeroderma pigmentosum group A-dependent mechanism. Cancer Res. 70, 3547–3556. D’Orazio, J., and Fisher, D.E. (2011). Central role for cAMP signaling in pigmentation and UV resistance. Cell Cycle 10, 8–9. Drake, D.R., Brogden, K.A., Dawson, D.V., and Wertz, P.W. (2008). Thematic review series: skin lipids. Antimicrobial lipids at the skin surface. J. Lipid Res. 49, 4–11. Dubrac, S., and Schmuth, M. (2011). PPAR-alpha in cutaneous inflammation. Dermatoendocrinology 3, 23–26. Elias, P.M. (2007). The skin barrier as an innate immune element. Semin. Immunopathol. 29, 3–14. Elias, P.M., and Williams, M.L. (2013). Re-appraisal of current theories for the development and loss of epidermal pigmentation in hominins and modern humans. J. Hum. Evol. 64, 687–692. Elias, P.M., Menon, G., Wetzel, B.K., and Williams, J.J. (2009). Evidence that stress to the epidermal barrier influenced the development of pigmentation in humans. Pigment Cell Melanoma Res. 22, 420–434. Elias, P.M., Menon, G., Wetzel, B.K., and Williams, J. (2010). Barrier requirements as the evolutionary ‘driver’ of epidermal pigmentation in humans. Am. J. Hum. Biol. 22, 526–537. s, G., Englaro, W., Bertolotto, C., Busc a, R., Brunet, A., Page Ortonne, J.P., and Ballotti, R. (1998). Inhibition of the mitogenactivated protein kinase pathway triggers B16 melanoma cell differentiation. J. Biol. Chem. 273, 9966–9970. Eves, P.C., MacNeil, S., and Haycock, J.W. (2006). alpha-Melanocyte stimulating hormone, inflammation and human melanoma. Peptides 27, 444–452. Fitzpatrick, T.B. (1988). The validity and practically of sun-reactive skin type I through VI. Arch. Dermatol. 124, 869–871. Flori, E., Mastrofrancesco, A., Kovacs, D., Ramot, Y., Briganti, S., Bellei, B., Paus, R., and Picardo, M. (2011). 2,4,6-Octatrienoic acid is a novel promoter of melanogenesis and antioxidant defence in normal human melanocytes via PPAR-c activation. Pigment Cell Melanoma Res. 24, 618–630.

385

Maresca et al. Fuller, B.B., Spaulding, D.T., and Smith, D.R. (2001). Regulation of the catalytic activity of preexisting tyrosinase in black and Caucasian human melanocyte cell cultures. Exp. Cell Res. 262, 197–208. n, J.C., Sanchez-Laorden, B.L., and Jime nez-Cervantes, Garcıa-Borro C. (2005). Melanocortin-1 receptor structure and functional regulation. Pigment Cell Res. 18, 393–410. n, J.C., Abdel-Malek, Z., and Jime nez-Cervantes, C. Garcıa-Borro (2014). MC1R, the cAMP pathway, and the response to solar UV: extending the horizon beyond pigmentation. Pigment Cell Melanoma Res. 27, 699–720. Gauden, M., Pezzella, A., Panzella, L., Neves-Petersen, M.T., Skovsen, E., Petersen, S.B., Mullen, K.M., Napolitano, A., d’Ischia, €m, V. (2008). Role of solvent, pH, and molecular M., and Sundstro size in excited-state deactivation of key eumelanin building blocks: implications for melanin pigment photostability. J. Am. Chem. Soc. 130, 17038–17043. Gilchrest, B.A., and Eller, M.S. (1999). DNA photodamage stimulates melanogenesis and other photoprotective responses. J. Investig. Dermatol. Symp. Proc. 4, 35–40. Gilman, A.G. (1995). Nobel Lecture. G proteins and regulation of adenylyl cyclase. Biosci. Rep. 15, 65–97. Godic, A., Polj sak, B., Adamic, M., and Dahmane, R. (2014). The role of antioxidants in skin cancer prevention and treatment. Oxid. Med. Cell. Longev. 2014, 860479. Grabacka, M., Placha, W., Urbanska, K., Laidler, P., Płonka, P.M., and Reiss, K. (2008). PPAR gamma regulates MITF and beta-catenin expression and promotes a differentiated phenotype in mouse melanoma S91. Pigment Cell Melanoma Res. 21, 388–396. Grando, S.A., Pittelkow, M.R., and Schallreuter, K.U. (2006). Adrenergic and cholinergic control in the biology of epidermis: physiological and clinical significance. J. Invest. Dermatol. 126, 1948–1965. Greaves, M. (2014). Was skin cancer a selective force for black pigmentation in early hominin evolution? Proc. Biol. Sci. 281, 20132955. Grommes, C., Landreth, G.E., and Heneka, M.T. (2004). Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 5, 419–429. Gunathilake, R., Schurer, N.Y., Shoo, B.A., Celli, A., Hachem, J.P., Crumrine, D., Sirimanna, G., Feingold, K.R., Mauro, T.M., and Elias, P.M. (2009). pH-regulated mechanisms account for pigment-type differences in epidermal barrier function. J. Invest. Dermatol. 129, 1719–1729. Haq, R., Shoag, J., Andreu-Perez, P. et al. (2013). Oncogenic BRAF regulates oxidative metabolism via PGC1a and MITF. Cancer Cell 23, 302–315. Hara, M.R., Kovacs, J.J., Whalen, E.J. et al. (2011). A stress response pathway regulates DNA damage through b2-adrenoreceptors and b-arrestin-1. Nature 477, 349–353. Hauser, J.E., Kadekaro, A.L., Kavanagh, R.J., Wakamatsu, K., Terzieva, S., Schwemberger, S., Babcock, G., Rao, M.B., Ito, S., and Abdel-Malek, Z.A. (2006). Melanin content and MC1R function independently affect UVR-induced DNA damage in cultured human melanocytes. Pigment Cell Res. 19, 303–314. Haycock, J.W., Rowe, S.J., Cartledge, S., Wyatt, A., Ghanem, G., Morandini, R., Rennie, I.G., and MacNeil, S. (2000). Alpha-melanocyte-stimulating hormone reduces impact of proinflammatory cytokine and peroxide-generated oxidative stress on keratinocyte and melanoma cell lines. J. Biol. Chem. 275, 5629–5636. Hemesath, T.J., Price, E.R., Takemoto, C., Badalian, T., and Fisher, D.E. (1998). MAP kinase links the transcription factor Microphthalmia to c-Kit signalling in melanocytes. Nature 391, 298–301. nez-Cervantes, C., Zanna, P., and Garcıa-Borro n, J.C. Herraiz, C., Jime (2009). Melanocortin 1 receptor mutations impact differentially on signalling to the cAMP and the ERK mitogen-activated protein kinase pathways. FEBS Lett. 583, 3269–3274.

386

, F., Abdel-Malek, Z., Ghanem, G., Jime nezHerraiz, C., Journe n, J.C. (2011). Signaling from the Cervantes, C., and Garcıa-Borro human melanocortin 1 receptor to ERK1 and ERK2 mitogenactivated protein kinases involves transactivation of cKIT. Mol. Endocrinol. 25, 138–156. , F., Ghanem, G., Jime nez-Cervantes, C., and Herraiz, C., Journe n, J.C. (2012). Functional status and relationships of Garcıa-Borro melanocortin 1 receptor signaling to the cAMP and extracellular signal-regulated protein kinases 1 and 2 pathways in human melanoma cells. Int. J. Biochem. Cell Biol. 44, 2244–2252. Hill, H.Z., and Hill, G.J. (2000). UVA, pheomelanin and the carcinogenesis of melanoma. Pigment Cell Res. 13(Suppl 8), 140–144. Hoogduijn, M.J., Cemeli, E., Ross, K., Anderson, D., Thody, A.J., and Wood, J.M. (2004). Melanin protects melanocytes and keratinocytes against H2O2-induced DNA strand breaks through its ability to bind Ca2 + . Exp. Cell Res. 294, 60–67. Ibraimov, A.I. (2007). The evolution of body heat, conductivity, skin and brain size in human. J. Hum. Ecol. 21, 95–103. Im, S., Moro, O., Peng, F., Medrano, E.E., Cornelius, J., Babcock, G., Nordlund, J.J., and Abdel-Malek, Z.A. (1998). Activation of the cyclic AMP pathway by alpha-melanotropin mediates the response of human melanocytes to ultraviolet B radiation. Cancer Res. 58, 47–54. Imokawa, G. (2004). Autocrine and paracrine regulation of melanocytes in human skin and in pigmentary disorders. Pigment Cell Res. 17, 96–110. Irvine, R.F. (1992). Inositol lipids in cell signalling. Curr. Opin. Cell Biol. 4, 212–219. d’Ischia, M., Wakamatsu, K., Napolitano, A. et al. (2013). Melanins and melanogenesis: methods, standards, protocols. Pigment Cell Melanoma Res. 5, 616–633. Jablonski, N.G., and Chaplin, G. (2000). The evolution of human skin coloration. J. Hum. Evol. 39, 57–106. Jablonski, N.G., and Chaplin, G. (2013). Epidermal pigmentation in the human lineage is an adaptation to ultraviolet radiation. Hum. Evol. 65, 671–675. Jablonski, N.G., and Chaplin, G. (2014). The evolution of skin pigmentation and hair texture in people of African ancestry. Dermatol. Clin. 32, 113–121. Jagirdar, K., Yin, K., Harrison, M., Lim, W., Muscat, G.E., Sturm, R.A., and Smith, A.G. (2013). The NR4A2 nuclear receptor is recruited to novel nuclear foci in response to UV irradiation and participates in nucleotide excision repair. PLoS ONE 8, e78075. Juzeniene, A., Setlow, R., Porojnicu, A., Steindal, A.H., and Moan, J. (2009). Development of different human skin colors: a review highlighting photobiological and photobiophysical aspects. J. Photochem. Photobiol., B 96, 93–100. Kadekaro, A.L., Kavanagh, R.J., Wakamatsu, K., Ito, S., Pipitone, M.A., and Abdel-Malek, Z.A. (2003). Cutaneous photobiology. The melanocyte vs. the sun: who will win the final round? Pigment Cell Res. 16, 434–447. Kadekaro, A.L., Kavanagh, R., Kanto, H. et al. (2005). alpha-Melanocortin and endothelin-1 activate antiapoptotic pathways and reduce DNA damage in human melanocytes. Cancer Res. 65, 4292–4299. Kadekaro, A.L., Leachman, S., Kavanagh, R.J. et al. (2010). Melanocortin 1 receptor genotype: an important determinant of the damage response of melanocytes to ultraviolet radiation. FASEB J. 24, 3850–3860. Kadekaro, A.L., Chen, J., Yang, J., Chen, S., Jameson, J., Swope, V.B., Cheng, T., Kadakia, M., and Abdel-Malek, Z. (2012). Alphamelanocyte-stimulating hormone suppresses oxidative stress through a p53-mediated signaling pathway in human melanocytes. Mol. Cancer Res. 10, 778–786. Kang, H.Y., Chung, E., Lee, M., Cho, Y., and Kang, W.H. (2004). Expression and function of peroxisome proliferator-activated receptors in human melanocytes. Br. J. Dermatol. 150, 462–468.

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

A new point of view of phototype Khlgatian, M.K., Hadshiew, I.M., Asawanonda, P., Yaar, M., Eller, M.S., Fujita, M., Norris, D.A., and Gilchrest, B.A. (2002). Tyrosinase gene expression is regulated by p53. J. Invest. Dermatol. 118, 126–132. Kokot, A., Metze, D., Mouchet, N., Galibert, M.D., Schiller, M., €hm, M. (2009a). Alpha-melanocyte-stimulating Luger, T.A., and Bo hormone counteracts the suppressive effect of UVB on Nrf2 and Nrf-dependent gene expression in human skin. Endocrinology 150, 3197–3206. €tter, C., Kerkhoff, C., Kokot, A., Sindrilaru, A., Schiller, M., Sunderko €hm, M. Eckes, B., Scharffetter-Kochanek, K., Luger, T.A., and Bo (2009b). alpha-melanocyte-stimulating hormone suppresses bleomycin-induced collagen synthesis and reduces tissue fibrosis in a mouse model of scleroderma: melanocortin peptides as a novel treatment strategy for scleroderma? Arthritis Rheum. 60, 592–603. Kondo, T., and Hearing, V.J. (2011). Update on the regulation of mammalian melanocyte function and skin pigmentation. Expert Rev. Dermatol. 6, 97–108. Kovacs, D., Cardinali, G., Aspite, N., Cota, C., Luzi, F., Bellei, B., Briganti, S., Amantea, A., Torrisi, M.R., and Picardo, M. (2010). Role of fibroblast-derived growth factors in regulating hyperpigmentation of solar lentigo. Br. J. Dermatol. 163, 1020–1027. Kovacs, D., Flori, E., Maresca, V., Ottaviani, M., Aspite, N., Dell’Anna, M.L., Panzella, L., Napolitano, A., Picardo, M., and d’Ischia, M. (2012). The eumelanin intermediate 5,6-dihydroxyindole-2-carboxylic acid is a messenger in the cross-talk among epidermal cells. J. Invest. Dermatol. 132, 1196–1205. Krumins, A.M., and Gilman, A.G. (2006). Targeted knockdown of G protein subunits selectively prevents receptor-mediated modulation of effectors and reveals complex changes in non-targeted signaling proteins. J. Biol. Chem. 281, 10250–10262. Lee, J.S., Choi, Y.M., and Kang, H.Y. (2007). PPAR-gamma agonist, ciglitazone, increases pigmentation and migration of human melanocytes. Exp. Dermatol. 16, 118–123. van der Leest, R.J., de Vries, E., Bulliard, J.L. et al. (2011). The Euromelanoma skin cancer prevention campaign in Europe: characteristics and results of 2009 and 2010. J. Eur. Acad. Dermatol. Venereol. 25, 1455–1465. Leiter, U., and Garbe, C. (2008). Epidemiology of melanoma and nonmelanoma skin cancer: the role of sunlight. Adv. Exp. Med. Biol. 624, 89–103. Levy, C., Khaled, M., and Fisher, D.E. (2006). MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol. Med. 12, 406–414. Li, J., Uter, W., Pfahlberg, A., and Gefeller, O. (2012). A comparison of patterns of sun protection during beach holidays and everyday outdoor activities in a population sample of young German children. Br. J. Dermatol. 166, 803–810. Liu, J., Man, W.Y., Lv, C.Z., Song, S.P., Shi, Y.J., Elias, P.M., and Man, M.Q. (2010). Epidermal permeability barrier recovery is delayed in vitiligo-involved sites. Skin Pharmacol. Physiol. 23, 193–200. Mackintosh, J.A. (2001). The antimicrobial properties of melanocytes, melanosomes and melanin and the evolution of black skin. J. Theor. Biol. 211, 101–113. , E., Beauchet, A., de Paula Corre ^a, M., Godin-Beekmann, S., Mahe gou, F., Saiag, P., Haeffelin, M., Bruant, S., Fay-Chatelard, F., Je and Aegerter, P. (2011). Outdoor sports and risk of ultraviolet radiation-related skin lesions in children: evaluation of risks and prevention. Br. J. Dermatol. 165, 360–367. Malewicz, M., Kadkhodaei, B., Kee, N. et al. (2011). Essential role for DNA-PK-mediated phosphorylation of NR4A nuclear orphan receptors in DNA double-strand break repair. Genes Dev. 25, 2031–2040. Man, M.Q., Lin, T.K., Santiago, J.L. et al. (2014). Basis for enhanced barrier function of pigmented skin. J. Invest. Dermatol. 134, 2399– 2407.

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

, M., Ta€ıeb, Maresca, V., Flori, E., Briganti, S., Camera, E., Cario-Andre A., and Picardo, M. (2006). UVA-induced modification of catalase charge properties in the epidermis is correlated with the skin phototype. J. Invest. Dermatol. 126, 182–190. Maresca, V., Flori, E., Briganti, S., Mastrofrancesco, A., Fabbri, C., Mileo, A.M., Paggi, M.G., and Picardo, M. (2008). Correlation between melanogenic and catalase activity in in vitro human melanocytes: a synergic strategy against oxidative stress. Pigment Cell Melanoma Res. 21, 200–205. Maresca, V., Flori, E., Fabbri, C., Briganti, S., Mariani, G., Catrical a, C., and Picardo, M. (2009). Acidic catalase in human skin in vivo: a new marker of permanent damage. Melanoma Res. 19, 372–378. Maresca, V., Flori, E., Bellei, B., Aspite, N., Kovacs, D., and Picardo, M. (2010). MC1R stimulation by alpha-MSH induces catalase and promotes its re- distribution to the cell periphery and dendrites. Pigment Cell Melanoma Res. 23, 263–275. Maresca, V., Flori, E., Camera, E., Bellei, B., Aspite, N., Ludovici, M., Catrical a, C., Cardinali, G., and Picardo, M. (2013). Linking aMSH with PPARc in B16-F10 melanoma. Pigment Cell Melanoma Res. 26, 113–127. Mastrofrancesco, A., Kokot, A., Eberle, A., Gibbons, N.C., Schallreuter, K.U., Strozyk, E., Picardo, M., Zouboulis, C.C., Luger, T.A., €hm, M. (2010). KdPT, a tripeptide derivative of alphaand Bo melanocyte-stimulating hormone, suppresses IL-1 beta-mediated cytokine expression and signaling in human sebocytes. J. Immunol. 185, 1903–1911. Memoli, S., Napolitano, A., d’Ischia, M., Misuraca, G., Palumbo, A., and Prota, G. (1997). Diffusible melanin-related metabolites are potent inhibitors of lipid peroxidation. Biochim. Biophys. Acta 1346, 61–68. Meredith, P., and Sarna, T. (2006). The physical and chemical properties of eumelanin. Pigment Cell Res. 19, 572–594. Michalik, L., and Wahli, W. (2007). Peroxisome proliferator-activated receptors (PPARs) in skin health, repair and disease. Biochim. Biophys. Acta 1771, 991–998. Ming, M., Feng, L., Shea, C.R., Soltani, K., Zhao, B., Han, W., Smart, R.C., Trempus, C.S., and He, Y.Y. (2011). PTEN positively regulates UVB-induced DNA damage repair. Cancer Res. 71, 5287–5295. Montagner, A., Rando, G., Degueurce, G., Leuenberger, N., Michalik, L., and Wahli, W. (2011). New insights into the role of PPARs. Prostaglandins Leukot. Essent. Fatty Acids 85, 235–243. €ssner, R., Schulz, U., Kru €ger, U., Middel, P., Schinner, S., and Mo €zesi, L. (2002). Agonists of peroxisome proliferator-activated Fu receptor gamma inhibit cell growth in malignant melanoma. J. Invest. Dermatol. 119, 576–582. Muffley, L.A., Zhu, K.Q., Engrav, L.H., Gibran, N.S., and Hocking, A.M. (2011). Spatial and temporal localization of the melanocortin 1 receptor and its ligand a-melanocyte-stimulating hormone during cutaneous wound repair. J. Histochem. Cytochem. 59, 278–288. €ller-Decker, K., and Fu €rstenberger, G. (2007). The cyclooxygenMu ase-2-mediated prostaglandin signaling is causally related to epithelial carcinogenesis. Mol. Carcinog. 46, 705–710. Napolitano, A., Panzella, L., Monfrecola, G., and d’Ischia, M. (2014). Pheomelanin-induced oxidative stress: bright and dark chemistry bridging red hair phenotype and melanoma. Pigment Cell Melanoma Res. 27, 721–733. Nasti, T.H., and Timares, L. (2014). MC1R, eumelanin and pheomelanin: their role in determining the susceptibility to skin cancer. Photochem. Photobiol. 91, 188–200. Natarajan, V.T., Ganju, P., Ramkumar, A., Grover, R., and Gokhale, R.S. (2014). Multifaceted pathways protect human skin from UV radiation. Nat. Chem. Biol. 10, 542–551. Newton, R.A., Roberts, D.W., Leonard, J.H., and Sturm, R.A. (2007). Human melanocytes expressing MC1R variant alleles show impaired activation of multiple signaling pathways. Peptides 28, 2387–2396.

387

Maresca et al. r, E.S.J. (2001). Reactive oxygen species, Nordberg, J., and Arne antioxidants and the mammalian thioredoxin system. Free Radical Biol. Med. 31, 1287–1312. Nordlund, J.J. (2007). The melanocyte and the epidermal melanin unit: an expanded concept. Dermatol. Clin. 25, 271–281. Novellino, L., d’Ischia, M., and Prota, G. (1998). Nitric oxide-induced oxidation of 5,6-dihydroxyindole and 5,6-dihydroxyindole-2-carboxylic acid under aerobic conditions: non-enzymatic route to melanin pigments of potential relevance to skin (photo)protection. Biochim. Biophys. Acta 1425, 27–35. Novellino, L., Napolitano, A., and Prota, G. (1999). 5,6-Dihydroxyindoles in the fenton reaction: a model study of the role of melanin precursors in oxidative stress and hyperpigmentary processes. Chem. Res. Toxicol. 12, 985–992. Nylander, K., Bourdon, J.C., Bray, S.E., Gibbs, N.K., Kay, R., Hart, I., and Hall, P.A. (2000). Transcriptional activation of tyrosinase and TRP-1 by p53 links UV irradiation to the protective tanning response. J. Pathol. 190, 39–46. Okuno, Y., Matsuda, M., Miyata, Y., Fukuhara, A., Komuro, R., Shimabukuro, M., and Shimomura, I. (2010). Human catalase gene is regulated by peroxisome proliferator activated receptor-gamma through a response element distinct from that of mouse. Endocr. J. 57, 303–309. Pagel, M., and Bodmer, W. (2003). A naked ape would have fewer parasites. Proc. Biol. Sci. 270, S117–S119. Park, H.Y., Perez, J.M., Laursen, R., Hara, M., and Gilchrest, B.A. (1999). Protein kinase C-beta activates tyrosinase by phosphorylating serine residues in its cytoplasmic domain. J. Biol. Chem. 274, 16470–16478. Park, M.H., Park, J.Y., Lee, H.J. et al. (2013). The novel PPAR a/c dual agonist MHY 966 modulates UVB-induced skin inflammation by inhibiting NF-jB activity. PLoS ONE 8, e76820. Pearen, M.A., and Muscat, G.E. (2010). Minireview: nuclear hormone receptor 4A signaling: implications for metabolic disease. Mol. Endocrinol. 24, 1891–1903. rez Oliva, A.B., Ferne ndez, L.P., Detorre, C., Herraiz, C., MartınezPe n, Escribano, J.A., Benıtez, J., Lozano Teruel, J.A., Garcıa-Borro nez-Cervantes, C., and Ribas, G. (2009). Identification J.C., Jime and functional analysis of novel variants of the human melanocortin 1 receptor found in melanoma patients. Hum. Mutat. 30, 811–822. Picardo, M. (2009). Lipid-mediated signalling and melanocyte function. Pigment Cell Melanoma Res. 22, 152–153. Picardo, M., Maresca, V., Eibenschutz, L., De Bernardo, C., Rinaldi, R., and Grammatico, P. (1999). Correlation between antioxidants and phototypes in melanocyte cultures a possible link of physiologic and pathologic relevance. J. Invest. Dermatol. 113, 424–425. Polvani, S., Tarocchi, M., and Galli, A. (2012). PPARc and Oxidative Stress: Con(b) Catenating NRF2 and FOXO. PPAR Res. 2012, 641087. Prota, G. (1997). Pigment cell research: what directions? Pigment Cell Res. 10, 5–11. Pucci, M., Pasquariello, N., Battista, N. et al. (2012). Endocannabinoids stimulate human melanogenesis via type-1 cannabinoid receptor. J. Biol. Chem. 287, 15466–15478. Raap, U., Brzoska, T., Sohl, S., P€ath, G., Emmel, J., Herz, U., Braun, A., Luger, T., and Renz, H. (2003). Alpha-melanocyte-stimulating hormone inhibits allergic airway inflammation. J. Immunol. 171, 353–359. Ramirez-Herrick, A.M., Mullican, S.E., Sheehan, A.M., and Conneely, O.M. (2011). Reduced NR4A gene dosage leads to mixed myelodysplastic/myeloproliferative neoplasms in mice. Blood 117, 2681–2690. Robbins, G.T., and Nie, D. (2012). PPAR gamma, bioactive lipids, and cancer progression. Front. Biosci. (Landmark Ed) 17, 1816–1834. Rodrigues, A.R., Almeida, H., and Gouveia, A.M. (2014). Intracellular signaling mechanisms of the melanocortin receptors: current state of the art. Cell. Mol. Life Sci. 72, 1331–1345.

388

Ronai, Z. (2013). The masters talk: the PGC-1a-MITF axis as a melanoma energizer. Pigment Cell Melanoma Res. 26, 294–295. Rosso, S., Zanetti, E., S anchez, M.J. et al., Helios Working Group (2007). Is 2,3,5-Pyrroletricarboxylic acid in hair a better risk indicator for melanoma than traditional epidemiologic measures for skin phenotype? Am. J. Epidemiol. 165, 1170–1177. Rouzaud, F., and Hearing, V.J. (2005). Regulatory elements of the melanocortin 1 receptor. Peptides 26, 1858–1870. Rouzaud, F., Kadekaro, A.L., Abdel-Malek, Z.A., and Hearing, V.J. (2005). MC1R and the response of melanocytes to ultraviolet radiation. Mutat. Res. 571, 133–152. Saha, B., Singh, S.K., Sarkar, C., Bera, R., Ratha, J., Tobin, D.J., and Bhadra, R. (2006). Activation of the Mitf promoter by lipidstimulated activation of p38-stress signalling to CREB. Pigment Cell Res. 19, 595–605. Saha, B., Singh, S.K., Mallick, S., Bera, R., Datta, P.K., Mandal, M., Roy, S., and Bhadra, R. (2009). Sphingolipid-mediated restoration of Mitf expression and repigmentation in vivo in a mouse model of hair graying. Pigment Cell Melanoma Res. 22, 205–218. Sahu, R.P., DaSilva, S.C., Rashid, B. et al. (2012). Mice lacking epidermal PPARc exhibit a marked augmentation in photocarcinogenesis associated with increased UVB-induced apoptosis, inflammation and barrier dysfunction. Int. J. Cancer 131, E1055–E1066. Sander, C.S., Hamm, F., Elsner, P., and Thiele, J.J. (2003). Oxidative stress in malignant melanoma and non-melanoma skin cancer. Br. J. Dermatol. 148, 913–922. dos Santos Silva, I., Higgins, D.C., Abramsky, T. et al. (2009). Overseas sun exposure, nevus counts, and premature skin aging in young English women: a population-based survey. J. Invest. Dermatol. 129, 50–59. Schallreuter, K.U., and Wood, J.M. (1999). The importance of Lphenylalanine transport and its autocrine turnover to L-tyrosine for melanogenesis in human epidermal melanocytes. Biochem. Biophys. Res. Commun. 262, 423–428. Schallreuter, K.U., and Wood, J.M. (2001). Thioredoxin reductase its role in epidermal redox status. J. Photochem. Photobiol., B 64, 179–184. €tlich, M., Lemke, Schallreuter, K.U., Wood, J.M., Pittelkow, M.R., Gu €dl, W., Swanson, N.N., Hitzemann, K., and Ziegler, I. K.R., Ro (1994). Regulation of melanin biosynthesis in the human epidermis by tetrahydrobiopterin. Science 263, 1444–1446. €nz, A., Beazley, W., and Schallreuter, K.U., Schulz-Douglas, V., Bu €rner, C. (1997). Pteridines in the control of pigmentation. J. Ko Invest. Dermatol. 109, 31–35. Schallreuter, K.U., Kothari, S., Hasse, S., Kauser, S., Lindsey, N.J., Gibbons, N.C., Hibberts, N., and Wood, J.M. (2003). In situ and in vitro evidence for DCoH/HNF-1 alpha transcription of tyrosinase in human skin melanocytes. Biochem. Biophys. Res. Commun. 301, 610–616. Schallreuter, K.U., Wazir, U., Kothari, S., Gibbons, N.C., Moore, J., and Wood, J.M. (2004). Human phenylalanine hydroxylase is activated by H2O2: a novel mechanism for increasing the Ltyrosine supply for melanogenesis in melanocytes. Biochem. Biophys. Res. Commun. 322, 88–92. Schallreuter, K.U., Kothari, S., Chavan, B., and Spencer, J.D. (2008). Regulation of melanogenesis–controversies and new concepts. Exp. Dermatol. 17, 395–404. €tter, C., Kalden, D.H., Brzoska, T., Fastrich, Scholzen, T.E., Sunderko M., Fisbeck, T., Armstrong, C.A., Ansel, J.C., and Luger, T.A. (2003). Alpha-melanocyte stimulating hormone prevents lipopolysaccharide-induced vasculitis by down-regulating endothelial cell adhesion molecule expression. Endocrinology 144, 360–370. Sertznig, P., Seifert, M., Tilgen, W., and Reichrath, J. (2008). Peroxisome proliferator-activated receptors (PPARs) and the human skin:importance of PPARs in skin physiology and dermatologic diseases. Am. J. Clin. Dermatol. 9, 15–31.

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

A new point of view of phototype Shindo, Y., and Hashimoto, T. (1997). Time course of changes in antioxidant enzymes in human skin fibroblasts after UVA irradiation. J. Dermatol. Sci. 14, 225–232. Shindo, Y., Witt, E., Han, D., Tzeng, B., Aziz, T., Nguyen, L., and Packer, L. (1994). Recovery of antioxidants and reduction in lipid hydroperoxides in murine epidermis and dermis after acute ultraviolet radiation exposure. Photodermatol. Photoimmunol. Photomed. 10, 183–191. Shoag, J., Haq, R., Zhang, M. et al. (2013). PGC-1 coactivators regulate MITF and the tanning response. 1. Mol. Cell 49, 145–157. Singh, S.K., Sarkar, C., Mallick, S., Saha, B., Bera, R., and Bhadra, R. (2005). Human placental lipid induces melanogenesis through p38 MAPK in B16F10 mouse melanoma. Pigment Cell Res. 18, 113– 121. Slominski, A., Tobin, D.J., Shibahara, S., and Wortsman, J. (2004). Melanin pigmentation in mammalian skin and its hormonal regulation. Physiol. Rev. 84, 1155–1228. Smalley, K.S., and Eisen, T.G. (2000). The involvement of p38 mitogenactivated protein kinase in the alpha-melanocyte stimulating hormone (alpha-MSH)-induced melanogenic and anti-proliferative effects in B16 murine melanoma cells. FEBS Lett. 476, 198–202. Smalley, K.S., and Eisen, T.G. (2002). Differentiation of human melanoma cells through p38 MAP kinase is associated with decreased retinoblastoma protein phosphorylation and cell cycle arrest. Melanoma Res. 12, 187–192. Smith, A.G., Luk, N., Newton, R.A., Roberts, D.W., Sturm, R.A., and Muscat, G.E. (2008). Melanocortin-1 receptor signaling markedly induces the expression of the NR4A nuclear receptor subgroup in melanocytic cells. J. Biol. Chem. 283, 12564–12570. Song, X., Mosby, N., Yang, J., Xu, A., Abdel-Malek, Z., and Kadekaro, A.L. (2009). alpha-MSH activates immediate defense responses to UV-induced oxidative stress in human melanocytes. Pigment Cell Melanoma Res. 22, 809–818. Spiegelman, B.M. (2007). Transcriptional control of energy homeostasis through the PGC1 coactivators. Novartis Found. Symp. 286, 3–6. discussion 6–12. Sturm, R.A. (2002). Skin colour and skin cancer – MC1R, the genetic link. Melanoma Res. 12, 405–416. Sturm, R.A. (2009). Molecular genetics of human pigmentation diversity. Hum. Mol. Genet. 18, R9–R17. Sturm, R.A., Duffy, D.L., Box, N.F., Newton, R.A., Sheohered, A.G., Chen, W., Marks, L.H., Leonard, J.H., and Martin, N.G. (2003). Genetic association and cellular function of MC1R variant alleles in human pigmentation. Ann. N. Y. Acad. Sci. 994, 348–358. Swope, V., Alexander, C., Starner, R., Schwemberger, S., Babcock, G., and Abdel-Malek, Z.A. (2014). Significance of the melanocortin 1 receptor in the DNA damage response of human melanocytes to ultraviolet radiation. Pigment Cell Melanoma Res. 27, 601–610. Toyoda, M., Takagi, H., Horiguchi, N., Kakizaki, S., Sato, K., Takayama, H., and Mori, M. (2002). A ligand for peroxisome proliferator activated receptor gamma inhibits cell growth and induces apoptosis in human liver cancer cells. Gut 50, 563–567. Varga, T., Czimmerer, Z., and Nagy, L. (2011). PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim. Biophys. Acta 1812, 1007– 1022.

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

Vazquez, F., Lim, J.H., Chim, H. et al. (2013). PGC1a expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell 23, 287–301. Vincensi, M.R., d’Ischia, M., Napolitano, A., Procaccini, E.M., Riccio, G., Monfrecola, G., Santoianni, P., and Prota, G. (1998). Phaeomelanin versus eumelanin as a chemical indicator of ultraviolet sensitivity in fair-skinned subjects at high risk for melanoma: a pilot study. Melanoma Res. 8, 53–58. Wahli, W., and Michalik, L. (2012). PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol. Metab. 23, 351– 363. Wakamatsu, K., Kavanagh, R., Kadekaro, A.L., Terzieva, S., Sturm, R.A., Leachman, S., Abdel-Malek, Z., and Ito, S. (2006). Diversity of pigmentation in cultured human melanocytes is due to differences in the type as well as quantity of melanin. Pigment Cell Res. 19, 154–162. Wang, Z., Coleman, D.J., Bajaj, G., Liang, X., Ganguli-Indra, G., and Indra, A.K. (2011). RXRa ablation in epidermal keratinocytes enhances UVR-induced DNA damage, apoptosis, and proliferation of keratinocytes and melanocytes. J. Invest. Dermatol. 131, 177– 187. Westerhof, W. (2007). Evolutionary, biologic, and social aspects of skin color. Dermatol. Clin. 25, 293–302. Wu, M., Hemesath, T.J., Takemoto, C.M., Horstmann, M.A., Wells, A.G., Price, E.R., Fisher, D.Z., and Fisher, D.E. (2000). c-Kit triggers dual phosphorylations, which couple activation and degradation of the essential melanocyte factor Mi. Genes Dev. 14, 301–312. Wymann, M.P., and Schneiter, R. (2008). Lipid signalling in disease. Nat. Rev. Mol. Cell Biol. 9, 162–176. Yamaguchi, Y., and Hearing, V.J. (2009). Physiological factors that regulate skin pigmentation. BioFactors 35, 193–199. Yamaguchi, Y., Brenner, M., and Hearing, V.J. (2007). The regulation of skin pigmentation. J. Biol. Chem. 282, 27557–27561. Yang, Y. (2011). Structure, function and regulation of the melanocortin receptors. Eur. J. Pharmacol. 660, 125–130. Yin, K., Sturm, R.A., and Smith, A.G. (2014). MC1R and NR4A receptors in cellular stress and DNA repair: implications for UVR protection. Exp. Dermatol. 23, 449–452. Yohn, J.J., Norris, D.A., Yrastorza, D.G., Buno, I.J., Leff, J.A., Hake, S.S., and Repine, J.E. (1991). Disparate antioxidant enzyme activities in cultured human cutaneous fibroblasts, keratinocytes, and melanocytes. J. Invest. Dermatol. 97, 405–409. Yoon, J.C., Puigserver, P., Chen, G. et al. (2001). Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413, 131–138. Yuen, A.W., and Jablonski, N.G. (2010). Vitamin D: in the evolution of human skin colour. Med. Hypotheses 74, 39–44. Yuri, M., Sasahira, T., Nakai, K., Ishimaru, S., Ohmori, H., and Kuniyasu, H. (2007). Reversal of expression of 15-lipoxygenase-1 to cyclooxygenase-2 is associated with development of colonic cancer. Histopathology 51, 520–527. Zhang, X., Erb, C., Flammer, J., and Nau, W.M. (2000). Absolute rate constants for the quenching of reactive excited states by melanin and related 5,6-dihydroxyindole metabolites: implications for their antioxidant activity. Photochem. Photobiol. 71, 524–533.

389

Skin phototype: a new perspective.

Cutaneous phototype is considered mainly related to cutaneous pigmentation and to the eumelanin/pheomelanin ratio, which is mostly genetically determi...
8MB Sizes 3 Downloads 8 Views