M. Osawa, G. Egawa, and S. Mak, Molecular characterization of melanocyte stem cells in their niche, Development, vol.132, issue.24, pp.5589-5599, 2005.

K. Schouwey, V. Delmas, and L. Larue, Notch1 and Notch2 receptors influence progressive hair graying in a dosedependent manner, Developmental Dynamics, vol.236, issue.1, pp.282-289, 2007.

Y. Aoki, N. Saint-germain, and M. Gyda, Sox10 regulates the development of neural crest-derived melanocytes in Xenopus, Developmental Biology, vol.259, issue.1, pp.19-33, 2003.

D. Lang, M. M. Lu, and L. Huang, Pax3 functions at a nodal point in melanocyte stem cell differentiation, Nature, vol.433, issue.7028, pp.884-887, 2005.

M. Moriyama, M. Osawa, and S. Mak, Notch signaling via Hes1 transcription factor maintains survival of melanoblasts and melanocyte stem cells, Journal of Cell Biology, vol.173, issue.3, pp.333-339, 2006.

S. Shibahara, K. Yasumoto, and S. Amae, Regulation of pigment cell-specific gene expression by MITF, Pigment Cell Research, vol.13, issue.8, pp.98-102, 2000.

K. Takeda, K. Yasumoto, and R. Takada, Induction of melanocyte-specific microphthalmia-associated transcription factor by Wnt-3a, Journal of Biological Chemistry, vol.275, issue.19, pp.14013-14016, 2000.

S. Agarwal and A. Ojha, Piebaldism: a brief report and review of the literature, Indian Dermatology Online Journal, vol.3, issue.2, pp.144-147, 2012.

J. Y. Lin and D. E. Fisher, Melanocyte biology and skin pigmentation, Nature, vol.445, issue.7130, pp.843-850, 2007.

S. Nishikawa and M. Osawa, Generating quiescent stem cells, Pigment Cell Research, vol.20, issue.4, pp.263-270, 2007.

T. Tumbar, G. Guasch, and V. Greco, Defining the epithelial stem cell Niche in skin, Science, vol.303, issue.5656, pp.359-363, 2004.

R. Cui, H. R. Widlund, and E. Feige, Central role of p53 in the suntan response and pathologic hyperpigmentation, Cell, vol.128, issue.5, pp.853-864, 2007.

Z. Abdel-malek, M. C. Scott, and I. Suzuki, The melanocortin-1 receptor is a key regulator of human cutaneous pigmentation, Pigment Cell Research, vol.13, issue.8, pp.156-162, 2000.

C. Bertolotto, P. Abbe, and T. J. Hemesath, Microphthalmia gene product as a signal transducer in cAMP-induced differentiation of melanocytes, Journal of Cell Biology, vol.142, issue.3, pp.827-835, 1998.

C. Gaggioli, R. Buscà, P. Abbe, J. Ortonne, and R. Ballotti, Microphthalmia-associated transcription factor (MITF) is required but is not sufficient to induce the expression of melanogenic genes, Pigment Cell Research, vol.16, issue.4, pp.374-382, 2003.

K. S. Hoek, N. C. Schlegel, and O. M. Eichhoff, Novel MITF targets identified using a two-step DNA microarray strategy, Pigment Cell & Melanoma Research, vol.21, issue.6, pp.665-676, 2008.

F. Vetrini, A. Auricchio, and J. Du, The microphthalmia transcription factor (Mitf) controls expression of the ocular albinism type 1 gene: link between melanin synthesis and melanosome biogenesis, Molecular and Cellular Biology, vol.24, issue.15, pp.6550-6559, 2004.

C. Bertolotto, K. Bille, J. Ortonne, and R. Ballotti, Regulation of tyrosinase gene expression by cAMP in B16 melanoma cells involves two CATGTG motifs surrounding the TATA box: implication of the microphthalmia gene product, Journal of Cell Biology, vol.134, issue.3, pp.747-755, 1996.

C. Bertolotto, R. Buscà, and P. Abbe, 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, Molecular and Cellular Biology, vol.18, issue.2, pp.694-702, 1998.

C. Chiaverini, L. Beuret, and E. Flori, Microphthalmiaassociated transcription factor regulates RAB27A gene expression and controls melanosome transport, Journal of Biological Chemistry, vol.283, issue.18, pp.12635-12642, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02358833

T. Passeron, P. Bahadoran, and C. Bertolotto, Cyclic AMP promotes a peripheral distribution of melanosomes and stimulates melanophilin/Slac2-a actin association, FASEB Journal, vol.18, issue.9, pp.989-991, 2004.
URL : https://hal.archives-ouvertes.fr/inserm-02531092

F. P. Noonan, T. Otsuka, S. Bang, M. R. Anver, and G. Merlino, Accelerated ultraviolet radiation-induced carcinogenesis in hepatocyte growth factor/scatter factor transgenic mice, Cancer Research, vol.60, issue.14, pp.3738-3743, 2000.

A. Van-schanke, M. J. Jongsma, R. Bisschop, G. M. Van-venrooij, H. Rebel et al., Single UVB overexposure stimulates melanocyte proliferation in murine skin, in contrast to fractionated or UVA-1 exposure, Journal of Investigative Dermatology, vol.124, issue.1, pp.241-247, 2005.

G. J. Walker, M. G. Kimlin, and E. Hacker, Murine neonatal melanocytes exhibit a heightened proliferative response to ultraviolet radiation and migrate to the epidermal basal layer, Journal of Investigative Dermatology, vol.129, issue.1, pp.184-193, 2009.

T. Hirobe, Role of keratinocyte-derived factors involved in regulating the proliferation and differentiation of mammalian epidermal melanocytes, Pigment Cell Research, vol.18, issue.1, pp.2-12, 2005.

R. Buscà, P. Abbe, and F. Mantoux, Ras mediates the cAMPdependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes, EMBO Journal, vol.19, issue.12, pp.2900-2910, 2000.

G. G. Mcgill, M. Horstmann, and H. R. Widlund, Bcl2 regulation by the melanocyte master regulator Mitf modulates lineage survival and melanoma cell viability, Cell, vol.109, issue.6, pp.707-718, 2002.

R. Haqa, S. Yokoyamab, and E. B. Hawryluk, BCL2A1 is a lineage-specific antiapoptotic melanoma oncogene that confers resistance to BRAF inhibition, Proceedings of the National Academy of Sciences of the United States of Amrica, vol.110, pp.4321-4326, 2013.

J. N. Dynek, S. M. Chan, J. Liu, J. Zha, W. J. Fairbrother et al., Microphthalmia-associated transcription factor is a critical transcriptional regulator of melanoma inhibitor of apoptosis in melanomas, Cancer Research, vol.68, issue.9, pp.3124-3132, 2008.

T. Strub, S. Giuliano, and T. Ye, Essential role of microphthalmia transcription factor for DNA replication, mitosis and genomic stability in melanoma, Oncogene, vol.30, issue.20, pp.2319-2332, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530755

M. F. Holick, Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers, and cardiovascular disease, The American Journal of Clinical Nutrition, vol.80, issue.6, pp.1678-1688, 2004.

M. F. Holick, Vitamin D: importance in the prevention of cancers, type 1 diabetes, heart disease, and osteoporosis, American Journal of Clinical Nutrition, vol.79, issue.3, pp.362-371, 2004.

J. A. Curtin, J. Fridlyand, and T. Kageshita, Distinct sets of genetic alterations in melanoma, New England Journal of Medicine, vol.353, issue.20, pp.2135-2147, 2005.

S. Krengel, A. Hauschild, and T. Schäfer, Melanoma risk in congenital melanocytic naevi: a systematic review, British Journal of Dermatology, vol.155, issue.1, pp.1-8, 2006.

C. J. Hussussian, J. P. Struewing, and A. M. Goldstein, Germline p16 mutations in familial melanoma, Nature Genetics, vol.8, issue.1, pp.15-21, 1994.

L. Zuo, J. Weger, and Q. Yang, Germline mutations in the p16(INK4a) binding domain of CDK4 in familial melanoma, Nature Genetics, vol.12, issue.1, pp.97-99, 1996.

B. Bressac-de-paillerets, M. Avril, A. Chompret, and F. Demenais, Genetic and environmental factors in cutaneous malignant melanoma, Biochimie, vol.84, issue.1, pp.67-74, 2002.

M. C. Fargnoli, S. Gandini, K. Peris, P. Maisonneuve, and S. Raimondi, MC1R variants increase melanoma risk in families with CDKN2A mutations: a meta-analysis, European Journal of Cancer, vol.46, issue.8, pp.1413-1420, 2010.

C. Bertolotto, F. Lesueur, and S. Giuliano, A SUMOylationdefective MITF germline mutation predisposes to melanoma and renal carcinoma, Nature, vol.480, issue.7375, pp.94-98, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01791271

P. Ghiorzo, L. Pastorino, and P. Queirolo, Prevalence of the E318K MITF germline mutation in Italian melanoma patients: associations with histological subtypes and family cancer history, Pigment Cell & Melanoma Research, vol.26, issue.2, pp.259-262, 2013.

R. A. Sturm, C. Fox, and P. Mcclenahan, Phenotypic characterization of nevus and tumor patterns in MITF E318K mutation carrier melanoma patients, Journal of Investigative Dermatology, 2013.

S. Yokoyama, S. L. Woods, and G. M. Boyle, A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma, Nature, vol.480, issue.7375, pp.99-103, 2011.

P. T. Bradford, D. M. Freedman, A. M. Goldstein, and M. A. Tucker, Increased risk of second primary cancers after a diagnosis of melanoma, Archives of Dermatology, vol.146, issue.3, pp.265-272, 2010.

F. P. Noonan, J. A. Recio, and H. Takayama, Neonatal sunburn and melanoma in mice, Nature, vol.413, issue.6853, pp.271-272, 2001.

J. L. Bulliard, Site-specific risk of cutaneous malignant melanoma and pattern of sun exposure in New Zealand, International Journal of Cancer, vol.85, issue.5, pp.627-632, 2000.

S. Mouret, C. Philippe, and J. Gracia-chantegrel, UVAinduced cyclobutane pyrimidine dimers in DNA: a direct photochemical mechanism?, Organic and Biomolecular Chemistry, vol.8, issue.7, pp.1706-1711, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00472822

P. Filipe, P. Morlière, and J. N. Silva, Plasma lipoproteins as mediators of the oxidative stress induced by UV light in human skin: a review of biochemical and biophysical studies on mechanisms of apolipoprotein alteration, lipid peroxidation, and associated skin cell responses, Oxidative Medicine and Cellular Longevity, vol.2013, 2013.

E. C. De-fabo, F. P. Noonan, T. Fears, and G. Merlino, Ultraviolet B but not ultraviolet A radiation initiates melanoma, Cancer Research, vol.64, issue.18, pp.6372-6376, 2004.

A. R. Lehmann, D. Mcgibbon, and M. Stefanini, Xeroderma pigmentosum, Orphanet Journal of Rare Diseases, vol.6, issue.1, p.70, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00436508

A. Kauffmann, F. Rosselli, and V. Lazar, High expression of DNA repair pathways is associated with metastasis in melanoma patients, Oncogene, vol.27, issue.5, pp.565-573, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00281489

G. Giglia-mari and A. Sarasin, TP53 mutations in human skin cancers, Human Mutation, vol.21, issue.3, pp.217-228, 2003.
URL : https://hal.archives-ouvertes.fr/hal-02148397

C. Greenman, P. Stephens, and R. Smith, Patterns of somatic mutation in human cancer genomes, Nature, vol.446, issue.7132, pp.153-158, 2007.

E. D. Pleasance, R. K. Cheetham, and P. J. Stephens, A comprehensive catalogue of somatic mutations from a human cancer genome, Nature, vol.463, issue.7278, pp.191-196, 2010.

K. C. Cheng, D. S. Cahill, H. Kasai, S. Nishimura, and L. A. Loeb, 8-Hydroxyguanine, an abundant form of oxidative DNA damage, causes G ? T and A ? C substitutions, Journal of Biological Chemistry, vol.267, issue.1, pp.166-172, 1992.

M. F. Berger, E. Hodis, and T. P. Heffernan, Melanoma genome sequencing reveals frequent PREX2 mutations, Nature, vol.485, issue.7399, pp.502-506, 2012.

E. Hodis, I. R. Watson, and G. V. Kryukov, A landscape of driver mutations in melanoma, Cell, vol.150, issue.2, pp.251-263, 2012.

M. Krauthammer, Y. Kong, and B. H. Ha, Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma, Nature Genetics, vol.44, issue.9, pp.1006-1014, 2012.

D. F. Easton and R. A. Eeles, Genome-wide association studies in cancer, Human Molecular Genetics, vol.17, issue.2, pp.109-115, 2008.

P. D. Pharoah, A. Antoniou, M. Bobrow, R. L. Zimmern, D. F. Easton et al., Polygenic susceptibility to breast cancer and implications for prevention, Nature Genetics, vol.31, issue.1, pp.33-36, 2002.

K. Laud, C. Marian, and M. F. , Comprehensive analysis of CDKN2A (p16INK4A/p14ARF) and CDKN2B genes in 53 melanoma index cases considered to be at heightened risk of melanoma, Journal of Medical Genetics, vol.43, issue.1, pp.39-47, 2006.

J. F. Thompson, R. A. Scolyer, and R. F. Kefford, Cutaneous melanoma in the era of molecular profiling, The Lancet, vol.374, issue.9687, pp.362-365, 2009.

A. A. Nelson and H. Tsao, Melanoma and genetics, Clinics in Dermatology, vol.27, issue.1, pp.46-52, 2009.

N. P. Pavletich, Mechanisms of cyclin-dependent kinase regulation: structures of Cdks, their cyclin activators, and Cip and INK4 inhibitors, Journal of Molecular Biology, vol.287, issue.5, pp.821-828, 1999.

D. C. Bennett, How to make a melanoma: what do we know of the primary clonal events, Pigment Cell & Melanoma Research, vol.21, issue.1, pp.27-38, 2008.

V. C. Gray-schopfer, S. C. Cheong, and H. Chong, Cellular senescence in naevi and immortalisation in melanoma: a role for p16, British Journal of Cancer, vol.95, issue.4, pp.496-505, 2006.

E. V. Sviderskaya, V. C. Gray-schopfer, and S. P. Hill, p16/cyclin-dependent kinase inhibitor 2A deficiency in human melanocyte senescence, apoptosis, and immortalization: possible implications for melanoma progression, Journal of the National Cancer Institute, vol.95, issue.10, pp.723-732, 2003.

M. Castellano, B. G. Gabrielli, C. J. Hussussian, N. C. Dracopoli, and N. K. Hayward, Restoration of CDKN2A into melanoma cells induces morphologic changes and reduction in growth rate but not anchorage-independent growth reversal, Journal of Investigative Dermatology, vol.109, issue.1, pp.61-68, 1997.

J. Ackermann, M. Frutschi, K. Kaloulis, T. Mckee, A. Trumpp et al., Metastasizing melanoma formation caused by expression of activated N-RasQ61K on an INK4a-deficient background, Cancer Research, vol.65, issue.10, pp.4005-4011, 2005.

H. Rizos, S. Puig, and C. Badenas, A melanoma-associated germline mutation in exon 1 inactivates p14ARF, Oncogene, vol.20, issue.39, pp.5543-5547, 2001.

C. Hewitt, C. L. Wu, and G. Evans, Germline mutation of ARF in a melanoma kindred, Human Molecular Genetics, vol.11, issue.11, pp.1273-1279, 2002.

J. A. Randerson-moor, M. Harland, and S. Williams, A germline deletion of p14ARF but not CDKN2A in a melanomaneural system tumour syndrome family, Human Molecular Genetics, vol.10, issue.1, pp.55-62, 2001.

D. E. Freedberg, S. H. Rigas, and J. Russak, Frequent p16-independent inactivation of p14ARF in human melanoma, Journal of the National Cancer Institute, vol.100, issue.11, pp.784-795, 2008.

L. Ha, T. Lchikawa, and M. Anver, ARF functions as a melanoma tumor suppressor by inducing p53-independent senescence, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.10968-10973, 2007.

N. E. Sharpless, K. Kannan, J. Xu, M. W. Bosenberg, and L. Chin, Both products of the mouse INK4a/ARF locus suppress melanoma formation in vivo, Oncogene, vol.22, issue.32, pp.5055-5059, 2003.

M. Daniotti, M. Oggionni, and T. Ranzani, BRAF alterations are associated with complex mutational profiles in malignant melanoma, Oncogene, vol.23, issue.35, pp.5968-5977, 2004.

N. Soufir, M. Avril, and A. Chompret, Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group, Human Molecular Genetics, vol.7, issue.2, pp.209-216, 1998.

S. G. Rane, S. C. Cosenza, R. V. Mettus, and E. P. Reddy, Germ line transmission of the Cdk4R24C mutation facilitates tumorigenesis and escape from cellular senescence, Molecular and Cellular Biology, vol.22, issue.2, pp.644-656, 2002.

R. Chawla, J. A. Procknow, R. V. Tantravahi, J. S. Khurana, J. Litvin et al., Cooperativity of Cdk4R24C and Ras in melanoma development, Cell Cycle, vol.9, issue.16, pp.3305-3314, 2010.

R. Sotillo, J. F. García, and S. Ortega, Invasive melanoma in Cdk4-targeted mice, Proceedings of the National Academy of Sciences of the United States of America, vol.98, pp.13312-13317, 2001.

E. Steingrímsson, N. G. Copeland, and N. A. Jenkins, Melanocytes and the Microphthalmia transcription factor network, Annual Review of Genetics, vol.38, pp.365-411, 2004.

Y. Cheli, M. Ohanna, R. Ballotti, and C. Bertolotto, Fifteen-year quest for microphthalmia-associated transcription factor target genes, Pigment Cell & Melanoma Research, vol.23, issue.1, pp.27-40, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-02530740

L. A. Garraway, H. R. Widlund, and M. A. Rubin, Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma, Nature, vol.436, issue.7047, pp.117-122, 2005.

S. Ugurel, R. Houben, and D. Schrama, Microphthalmiaassociated transcription factor gene amplification in metastatic melanoma is a prognostic marker for patient survival, but not a predictive marker for chemosensitivity and chemotherapy response, Clinical Cancer Research, vol.13, issue.21, pp.6344-6350, 2007.

S. Carreira, J. Goodall, and L. Denat, Mitf regulation of Dia1 controls melanoma proliferation and invasiveness, Genes and Development, vol.20, issue.24, pp.3426-3439, 2006.

Y. Cheli, S. Guiliano, and T. Botton, Mitf is the key molecular switch between mouse or human melanoma initiating cells and their differentiated progeny, Oncogene, vol.30, issue.20, pp.2307-2318, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530763

Y. Cheli, S. Giuliano, and N. Fenouille, Hypoxia and MITF control metastatic behaviour in mouse and human melanoma cells, Oncogene, vol.31, issue.19, pp.2461-2470, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02530643

K. S. Hoek and C. R. Goding, Cancer stem cells versus phenotype-switching in melanoma, Pigment Cell & Melanoma Research, vol.23, issue.6, pp.746-759, 2010.

S. Carreira, J. Goodall, and I. Aksan, Mitf cooperates with Rb1 and activates p21Cip1 expression to regulate cell cycle progression, Nature, vol.433, issue.7027, pp.764-769, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01068395

S. Giuliano, Y. Cheli, and M. Ohanna, Microphthalmiaassociated transcription factor controls the DNA damage response and a lineage-specific senescence program in melanomas, Cancer Research, vol.70, issue.9, pp.3813-3822, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-02530789

S. Giuliano, M. Ohanna, R. Ballotti, and C. Bertolotto, Advances in melanoma senescence and potential clinical application, Pigment Cell & Melanoma Research, vol.24, issue.2, pp.295-308, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530650

M. Ohanna, S. Giuliano, and C. Bonet, Senescent cells develop a PARP-1 and nuclear factor-B-associated secretome (PNAS), Genes and Development, vol.25, issue.12, pp.1245-1261, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02530715

T. J. Hemesath, E. R. Price, C. Takemoto, T. Badalian, and D. E. Fisher, MAP kinase links the transcription factor Microphthalmia to c-Kit signalling in melanocytes, Nature, vol.391, issue.6664, pp.298-301, 1998.

W. Xu, L. Gong, and M. M. Haddad, Regulation of microphthalmia-associated transcription factor MITF protein levels by association with the ubiquitin-conjugating enzyme hUBC9, Experimental Cell Research, vol.255, issue.2, pp.135-143, 2000.

K. A. Wilkinson and J. M. Henley, Mechanisms, regulation and consequences of protein SUMOylation, Biochemical Journal, vol.428, issue.2, pp.133-145, 2010.

J. Du, H. R. Widlund, and M. A. Horstmann, Critical role of CDK2 for melanoma growth linked to its melanocyte-specific transcriptional regulation by MITF, Cancer Cell, vol.6, issue.6, pp.565-576, 2004.

L. Beuret, E. Flori, and C. Denoyelle, Up-regulation of MET expression by -melanocyte-stimulating hormone and MITF allows hepatocyte growth factor to protect melanocytes and melanoma cells from apoptosis, Journal of Biological Chemistry, vol.282, issue.19, pp.14140-14147, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02358861

R. Buscà, E. Berra, and C. Gaggioli, Hypoxia-inducible factor 1 is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells, Journal of Cell Biology, vol.170, issue.1, pp.49-59, 2005.

F. Liu, Y. Fu, and F. L. Meyskens, MiTF regulates cellular response to reactive oxygen species through transcriptional regulation of APE-1/Ref-1, Journal of Investigative Dermatology, vol.129, issue.2, pp.422-431, 2009.

T. J. Hornyak, S. Jiang, and E. A. Guzmán, Mitf dosage as a primary determinant of melanocyte survival after ultraviolet irradiation, Pigment Cell & Melanoma Research, vol.22, issue.3, pp.307-318, 2009.

A. J. Miller, J. Du, S. Rowan, C. L. Hershey, H. R. Widlund et al., Transcriptional regulation of the melanoma prognostic marker melastatin (TRPM1) by MITF in melanocytes and melanoma, Cancer Research, vol.64, issue.2, pp.509-516, 2004.

R. Haq, J. Shoag, and P. Andreu-perez, Oncogenic BRAF regulates oxidative metabolism via PGC1alpha and MITF, Cancer Cell, vol.23, issue.3, pp.302-315, 2013.

F. Vazquez, J. Lim, and H. Chim, PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress, Cancer Cell, vol.23, issue.3, pp.287-301, 2013.

C. Handschin and B. M. Spiegelman, Peroxisome proliferatoractivated receptor coactivator 1 coactivators, energy homeostasis, and metabolism, Endocrine Reviews, vol.27, issue.7, pp.728-735, 2006.

J. L. Rees, The melanocortin 1 receptor (MC1R): more than just red hair, Pigment Cell Research, vol.13, issue.3, pp.135-140, 2000.

R. A. Sturm, Skin colour and skin cancer-MC1R, the genetic link, Melanoma Research, vol.12, issue.5, pp.405-416, 2002.

C. Kennedy, J. Huurne, and M. Berkhout, Melanocortin 1 receptor (MC1R) gene variants are associated with an increased risk for cutaneous melanoma which is largely independent of skin type and hair color, Journal of Investigative Dermatology, vol.117, issue.2, pp.294-300, 2001.

J. S. Palmer, D. L. Duffy, and N. F. Box, Melanocortin-1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype?, American Journal of Human Genetics, vol.66, issue.1, pp.176-186, 2000.

P. Valverde, E. Healy, I. Jackson, J. L. Rees, and A. J. Thody, Variants of the melanocyte-stimulating hormone receptor gene are associated with red hair and fair skin in humans, Nature Genetics, vol.11, issue.3, pp.328-330, 1995.

N. F. Box, D. L. Duffy, and W. Chen, MC1R genotype modifies risk of melanoma in families segregating CDKN2A mutations, American Journal of Human Genetics, vol.69, issue.4, pp.765-773, 2001.

P. A. Van-der-velden, L. A. Sandkuijl, and W. Bergman, Melanocortin-1 receptor variant R151C modifies melanoma risk in Dutch families with melanoma, American Journal of Human Genetics, vol.69, issue.4, pp.774-779, 2001.

M. C. Fargnoli, K. Pike, and R. M. Pfeiffer, MC1R variants increase risk of melanomas harboring BRAF mutations, Journal of Investigative Dermatology, vol.128, pp.2485-2490, 2008.

M. T. Landi, J. Bauer, and R. M. Pfeiffer, MC1R germline variants confer risk for BRAF-mutant melanoma, Science, vol.313, issue.5786, pp.521-522, 2006.

D. F. Gudbjartsson, P. Sulem, and S. N. Stacey, ASIP and TYR pigmentation variants associate with cutaneous melanoma and basal cell carcinoma, Nature Genetics, vol.40, issue.7, pp.886-891, 2008.

L. P. Fernandez, R. L. Milne, and G. Pita, SLC45A2: a novel malignant melanoma-associated gene, Human Mutation, vol.29, issue.9, pp.1161-1167, 2008.

M. Kvaskoff, D. C. Whiteman, and Z. Z. Zhao, Polymorphisms in nevus-associated genes MTAP, PLA2G6, and IRF4 and the risk of invasive cutaneous melanoma, Twin Research and Human Genetics, vol.14, issue.5, pp.422-432, 2011.

S. Horn, A. Figl, and P. S. Rachakonda, TERT promoter mutations in familial and sporadic melanoma, Science, vol.339, issue.6122, pp.959-961, 2013.

X. Wei, V. Walia, and J. C. Lin, Exome sequencing identifies GRIN2A as frequently mutated in melanoma, Nature Genetics, vol.43, issue.5, pp.442-448, 2011.

T. D. Prickett, N. S. Agrawal, and X. Wei, Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4, Nature Genetics, vol.41, issue.10, pp.1127-1132, 2009.

L. H. Palavalli, T. D. Prickett, and J. R. Wunderlich, Analysis of the matrix metalloproteinase family reveals that MMP8 is often mutated in melanoma, Nature Genetics, vol.41, issue.5, pp.518-520, 2009.

R. Ghai, M. Mobli, and S. J. Norwood, Phox homology band 4.1/ezrin/radixin/moesin-like proteins function as molecular scaffolds that interact with cargo receptors and Ras GTPases, Proceedings of the National Academy of Sciences of the United States of America, vol.108, issue.19, pp.7763-7768, 2011.

M. Cully, J. Shiu, R. P. Piekorz, W. J. Muller, S. J. Done et al., Transforming acidic coiled coil 1 promotes transformation and mammary tumorigenesis, Cancer Research, vol.65, issue.22, pp.10363-10370, 2005.

P. M. Pollock, U. L. Harper, and K. S. Hansen, High frequency of BRAF mutations in nevi, Nature Genetics, vol.33, issue.1, pp.19-20, 2003.

I. Yeh, A. Deimling, and B. C. Bastian, Clonal BRAF mutations in melanocytic nevi and initiating role of BRAF in melanocytic neoplasia, Journal of the National Cancer Institute, vol.105, issue.12, pp.917-919, 2013.

Y. Chudnovsky, A. E. Adams, P. B. Robbins, Q. Lin, and P. A. Khavari, Use of human tissue to assess the oncogenic activity of melanoma-associated mutations, Nature Genetics, vol.37, issue.7, pp.745-749, 2005.

B. Bedogni and M. B. Powell, Hypoxia, melanocytes and melanoma-survival and tumor development in the permissive microenvironment of the skin, Pigment Cell & Melanoma Research, vol.22, issue.2, pp.166-174, 2009.

S. S. Dadras, Molecular diagnostics in Melanoma: current status and perspectives, Archives of Pathology and Laboratory Medicine, vol.135, issue.7, pp.860-869, 2011.

C. Denoyelle, G. Abou-rjaily, and V. Bezrookove, Antioncogenic role of the endoplasmic reticulum differentially activated by mutations in the MAPK pathway, Nature Cell Biology, vol.8, issue.10, pp.1053-1063, 2006.

C. Michaloglou, L. C. Vredeveld, and M. S. Soengas, BRAFE600-associated senescence-like cell cycle arrest of human naevi, Nature, vol.436, issue.7051, pp.720-724, 2005.

D. Dankort, D. P. Curley, and R. A. Cartlidge, BrafV600E cooperates with Pten loss to induce metastatic melanoma, Nature Genetics, vol.41, issue.5, pp.544-552, 2009.

N. Dhomen, J. S. Reis-filho, S. Da-rocha, and . Dias, Oncogenic braf induces melanocyte senescence and melanoma in mice, Cancer Cell, vol.15, issue.4, pp.294-303, 2009.

W. E. Damsky, D. P. Curley, and M. Santhanakrishnan, catenin signaling controls metastasis in braf-activated ptendeficient melanomas, Cancer Cell, vol.20, issue.6, pp.741-754, 2011.

D. C. Whiteman, W. J. Pavan, and B. C. Bastian, The melanomas: a synthesis of epidemiological, clinical, histopathological, genetic, and biological aspects, supporting distinct subtypes, causal pathways, and cells of origin, Pigment Cell & Melanoma Research, vol.24, issue.5, pp.879-897, 2011.

U. Banerji, A. Affolter, I. Judson, R. Marais, and P. Workman, BRAF and NRAS mutations in melanoma: potential relationships to clinical response to HSP90 inhibitors, Molecular Cancer Therapeutics, vol.7, issue.4, pp.737-739, 2008.

J. A. Ellerhorst, V. R. Greene, S. Ekmekcioglu, C. L. Warneke, M. M. Johnson et al., Clinical correlates of NRAS and BRAF mutations in primary human melanoma, Clinical Cancer Research, vol.17, issue.2, pp.229-235, 2011.

M. C. Van-dijk, M. R. Bernsen, and D. J. Ruiter, Analysis of mutations in B-RAF, N-RAS, and H-RAS genes in the differential diagnosis of Spitz nevus and spitzoid melanoma, American Journal of Surgical Pathology, vol.29, issue.9, pp.1145-1151, 2005.

H. Davies, G. R. Bignell, and C. Cox, Mutations of the BRAF gene in human cancer, Nature, vol.417, issue.6892, pp.949-954, 2002.

J. A. Jakob, R. L. Bassett, and C. S. Ng, NRAS mutation status is an independent prognostic factor in metastatic melanoma, Cancer, vol.118, issue.16, pp.4014-4023, 2012.

B. Jovanovic, S. Egyhazi, and M. Eskandarpour, Coexisting NRAS and BRAF mutations in primary familial melanomas with specific CDKN2A germline alterations, Journal of Investigative Dermatology, vol.130, issue.2, pp.618-620, 2010.

C. Wellbrock, S. Rana, H. Paterson, H. Pickersgill, T. Brummelkamp et al., Oncogenic BRAF regulates melanoma proliferation through the lineage specific factor MITF, PLoS ONE, vol.3, issue.7, 2008.

B. Govindarajan, X. Bai, and C. Cohen, Malignant transformation of melanocytes to melanoma by constitutive activation of mitogen-activated protein kinase kinase (MAPKK) signaling, Journal of Biological Chemistry, vol.278, issue.11, pp.9790-9795, 2003.

B. Jansen, H. Schlagbauer-wadl, and H. Kahr, Novel Ras antagonist blocks human melanoma growth, Proceedings of the National Academy of Sciences of the United States of America, vol.96, pp.14019-14024, 1999.

S. R. Hingorani, M. A. Jacobetz, G. P. Robertson, M. Herlyn, and D. A. Tuveson, Suppression of BRAFV599E in human melanoma abrogates transformation, Cancer Research, vol.63, issue.17, pp.5198-5202, 2003.

K. P. Hoeflich, D. C. Gray, and M. T. Eby, Oncogenic BRAF is required for tumor growth and maintenance in melanoma models, Cancer Research, vol.66, issue.2, pp.999-1006, 2006.

A. Sharma, M. A. Tran, and S. Liang, Targeting mitogenactivated protein kinase/extracellular signal-regulated kinase kinase in the mutant (V600E) B-Raf signaling cascade effectively inhibits melanoma lung metastases, Cancer Research, vol.66, issue.16, pp.8200-8209, 2006.

J. Caramel, E. Papadogeorgakis, and L. Hill, A switch in the expression of embryonic EMT-inducers drives the Development of Malignant Melanoma, Cancer Cell, vol.24, issue.4, pp.466-480, 2013.

M. Chraybi, I. Alsamad, and C. Copie-bergman, Oncogene abnormalities in a series of primary melanomas of the sinonasal tract: NRAS mutations and cyclin D1 amplification are more frequent than KIT or BRAF mutations, Human Pathology, vol.44, issue.9, pp.1902-1911, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00872629

E. R. Sauter, U. Yeo, and A. V. Stemm, Cyclin D1 is a candidate oncogene in cutaneous melanoma, Cancer Research, vol.62, issue.11, pp.3200-3206, 2002.

K. S. Smalley, M. Lioni, and M. D. Palma, Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas, Molecular Cancer Therapeutics, vol.7, issue.9, pp.2876-2883, 2008.

K. Trunzer, Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma, Journal of Clinical Oncology, vol.31, issue.14, pp.1767-1774, 2013.

K. V. Bhatt, L. S. Spofford, G. Aram, M. Mcmullen, K. Pumiglia et al., Adhesion control of cyclin D1 and p27Kip1 levels is deregulated in melanoma cells through BRAF-MEK-ERK signaling, Oncogene, vol.24, issue.21, pp.3459-3471, 2005.

V. Alexeev and K. Yoon, Distinctive role of the cKit receptor tyrosine kinase signaling in mammalian melanocytes, Journal of Investigative Dermatology, vol.126, issue.5, pp.1102-1110, 2006.

S. Nishikawa, M. Kusakabe, and K. Yoshinaga, In utero manipulation of coat color formation by a monoclonal antic-kit antibody: two distinct waves of c-kit-dependency during melanocyte development, EMBO Journal, vol.10, issue.8, pp.2111-2118, 1991.

M. Okura, H. Maeda, S. Nishikawa, and M. Mizoguchi, Effects of monoclonal anti-c-Kit antibody (ACK2) on melanocytes in newborn mice, Journal of Investigative Dermatology, vol.105, issue.3, pp.322-328, 1995.

F. Janku, J. Novotny, and I. Julis, KIT receptor is expressed in more than 50% of early-stage malignant melanoma: a retrospective study of 261 patients, Melanoma Research, vol.15, issue.4, pp.251-256, 2005.

A. K. Mobley, Driving transcriptional regulators in melanoma metastasis, Cancer and Metastasis Reviews, vol.31, issue.3-4, pp.621-632, 2012.

M. Bar-eli, Role of AP-2 in tumor growth and metastasis of human melanoma, Cancer and Metastasis Reviews, vol.18, issue.3, pp.377-385, 1999.

L. A. Mcpherson, A. V. Loktev, and R. J. Weigel, Tumor suppressor activity of AP2 mediated through a direct interaction with p53, Journal of Biological Chemistry, vol.277, issue.47, pp.45028-45033, 2002.

C. R. Antonescu, K. J. Busam, and T. D. Francone, L576P KIT mutation in anal melanomas correlates with KIT protein expression and is sensitive to specific kinase inhibition, International Journal of Cancer, vol.121, issue.2, pp.257-264, 2007.

C. Beadling, E. Jacobson-dunlop, and F. S. Hodi, KIT gene mutations and copy number in melanoma subtypes, Clinical Cancer Research, vol.14, issue.21, pp.6821-6828, 2008.

J. A. Curtin, K. Busam, D. Pinkel, and B. C. Bastian, Somatic activation of KIT in distinct subtypes of melanoma, Journal of Clinical Oncology, vol.24, issue.26, pp.4340-4346, 2006.

C. Willmore-payne, J. A. Holden, S. Hirschowitz, and L. J. Layfield, BRAF and c-kit gene copy number in mutationpositive malignant melanoma, Human Pathology, vol.37, issue.5, pp.520-527, 2006.

S. Huang, D. Jean, M. Luca, M. A. Tainsky, and M. Bar-eli, Loss of AP-2 results in downregulation of c-KIT and enhancement of melanoma tumorigenicity and metastasis, EMBO Journal, vol.17, issue.15, pp.4358-4369, 1998.

K. S. Smalley, R. Contractor, and T. K. Nguyen, Identification of a novel subgroup of melanomas with KIT/cyclindependent kinase-4 overexpression, Cancer Research, vol.68, issue.14, pp.5743-5752, 2008.

K. H. Vousden and C. Prives, Blinded by the light: the growing complexity of p53, Cell, vol.137, issue.3, pp.413-431, 2009.

L. A. Donehower, M. Harvey, and B. L. Slagle, Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours, Nature, vol.356, issue.6366, pp.215-221, 1992.

N. Ibrahim and F. G. Haluska, Molecular pathogenesis of cutaneous melanocytic neoplasms, Annual Review of Pathology, vol.4, pp.551-579, 2009.

N. Bardeesy, B. C. Bastian, A. Hezel, D. Pinkel, R. A. Depinho et al., Dual inactivation of RB and p53 pathways in RASinduced melanomas, Molecular and Cellular Biology, vol.21, issue.6, pp.2144-2153, 2001.

M. Dovey, R. M. White, and L. I. Zon, Oncogenic NRAS cooperates with p53 loss to generate melanoma in zebrafish, Zebrafish, vol.6, issue.4, pp.397-404, 2009.

V. K. Goel, N. Ibrahim, and G. Jiang, Melanocytic nevus-like hyperplasia and melanoma in transgenic BRAFV600E mice, Oncogene, vol.28, issue.23, pp.2289-2298, 2009.

E. E. Patton, H. R. Widlund, and J. L. Kutok, BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma, Current Biology, vol.15, issue.3, pp.249-254, 2005.

M. S. Soengas, P. Capodieci, and D. Polsky, Inactivation of the apoptosis effector Apaf-1 in malignant melanoma, Nature, vol.409, issue.6817, pp.207-211, 2001.

H. Yu, R. Mcdaid, and J. Lee, The role of BRAF mutation and p53 inactivation during transformation of a subpopulation of primary human melanocytes, American Journal of Pathology, vol.174, issue.6, pp.2367-2377, 2009.

V. Muthusamy, C. Hobbs, and C. Nogueira, Amplification of CDK4 and MDM2 in malignant melanoma, Genes Chromosomes and Cancer, vol.45, issue.5, pp.447-454, 2006.

A. Gembarska, F. Luciani, and C. Fedele, MDM4 is a key therapeutic target in cutaneous melanoma, Nature Medicine, vol.18, issue.8, pp.1239-1247, 2012.

B. Bedogni, S. M. Welford, D. S. Cassarino, B. J. Nickoloff, A. J. Giaccia et al., The hypoxic microenvironment of the skin contributes to Akt-mediated melanocyte transformation, Cancer Cell, vol.8, issue.6, pp.443-454, 2005.

M. Kaluzová, S. Kaluz, M. I. Lerman, and E. J. Stanbridge, DNA damage is a prerequisite for p53-mediated proteasomal degradation of HIF-1 in hypoxic cells and downregulation of the hypoxia marker carbonic anhydrase IX, Molecular and Cellular Biology, vol.24, issue.13, pp.5757-5766, 2004.

N. H. Kim, H. S. Kim, and X. Li, A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition, Journal of Cell Biology, vol.195, issue.3, pp.417-433, 2011.

T. Kim, A. Veronese, and F. Pichiorri, p53 regulates epithelialmesenchymal transition through microRNAs targeting ZEB1 and ZEB2, Journal of Experimental Medicine, vol.208, issue.5, pp.875-883, 2011.

. Scientifica,

H. Tsao, M. B. Atkins, and A. J. Sober, Management of cutaneous melanoma, New England Journal of Medicine, vol.351, issue.10, pp.998-1042, 2004.

J. M. Stahl, M. Cheung, A. Sharma, N. R. Trivedi, S. Shanmugam et al., Loss of PTEN promotes tumor development in malignant melanoma, Cancer Research, vol.63, issue.11, pp.2881-2890, 2003.

J. M. Stahl, A. Sharma, and M. Cheung, Deregulated Akt3 activity promotes development of malignant melanoma, Cancer Research, vol.64, issue.19, pp.7002-7010, 2004.

A. Y. Shull, A. Latham-schwark, and P. Ramasamy, Novel somatic mutations to PI3K pathway genes in metastatic melanoma, PLoS ONE, vol.7, issue.8, 2012.

V. K. Goel, A. J. Lazar, C. L. Warneke, M. S. Redston, and F. G. Haluska, Examination of mutations in BRAF, NRAS, and PTEN in primary cutaneous melanoma, Journal of Investigative Dermatology, vol.126, issue.1, pp.154-160, 2006.

L. Larue and A. Bellacosa, Epithelial-mesenchymal transition in development and cancer: role of phosphatidylinositol 3 kinase/AKT pathways, Oncogene, vol.24, issue.50, pp.7443-7454, 2005.

P. Wang, X. Wang, and P. Wu, GM3 upregulation of matrix metalloproteinase-9 possibly through PI3K, AKT, RICTOR, RHOGDI-2, and TNF-a pathways in mouse melanoma B16 cells, Advances in Experimental Medicine and Biology, vol.705, pp.335-348, 2011.

I. Stamenkovic, Matrix metalloproteinases in tumor invasion and metastasis, Seminars in Cancer Biology, vol.10, issue.6, pp.415-433, 2000.

A. Biddle and I. C. Mackenzie, Cancer stem cells and EMT in carcinoma, Cancer and Metastasis Reviews, vol.31, issue.1-2, pp.285-293, 2012.

D. Olmeda, M. Jordá, H. Peinado, Á. Fabra, and A. Cano, Snail silencing effectively suppresses tumour growth and invasiveness, Oncogene, vol.26, issue.13, pp.1862-1874, 2007.

H. Peinado, D. Olmeda, and A. Cano, Snail, ZEB and bHLH factors in tumour progression: an alliance against the epithelial phenotype?, Nature Reviews Cancer, vol.7, issue.6, pp.415-428, 2007.

J. P. Thiery, H. Acloque, R. Y. Huang, and M. A. Nieto, Epithelial-mesenchymal transitions in development and disease, Cell, vol.139, issue.5, pp.871-890, 2009.

J. Yang and R. A. Weinberg, Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis, Developmental Cell, vol.14, issue.6, pp.818-829, 2008.

S. R. Alonso, L. Tracey, and P. Ortiz, A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis, Cancer Research, vol.67, issue.7, pp.3450-3460, 2007.

E. Batlle, E. Sancho, and C. Franci, The transcription factor Snail is a repressor of E-cadherin gene expression in epithelial tumour cells, Nature Cell Biology, vol.2, issue.2, pp.84-89, 2000.

K. M. Hajra, D. Y. , .. C. David, Y. Chen, and E. R. Fearon, The SLUG zinc-finger protein represses E-cadherin in breast cancer, Cancer Research, vol.62, issue.6, pp.1613-1618, 2002.

J. Yang, S. A. Mani, and J. L. Donaher, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis, Cell, vol.117, issue.7, pp.927-939, 2004.

P. B. Gupta, C. Kuperwasser, and J. Brunet, The melanocyte differentiation program predisposes to metastasis after neoplastic transformation, Nature Genetics, vol.37, issue.10, pp.1047-1054, 2005.

I. K. Bukholm, J. M. Nesland, and A. L. Borresen-dale, Reexpression of E-cadherin, alpha-catenin and beta-catenin, but not of gamma-catenin, in metastatic tissue from breast cancer patients [seecomments], Journal of Pathology, vol.190, issue.1, pp.15-19, 2000.

M. Hsu, F. E. Meier, and M. Nesbit, E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors, American Journal of Pathology, vol.156, issue.5, pp.1515-1525, 2000.

J. E. Kim, E. Leung, B. C. Baguley, and G. J. Finlay, Heterogeneity of expression of epithelial-mesenchymal transition markers in melanocytes and melanoma cell lines, Frontiers in Genetics, vol.4, p.97, 2013.

Y. Ogawara, S. Kishishita, and T. Obata, Akt enhances Mdm2-mediated ubiquitination and degradation of p53, Journal of Biological Chemistry, vol.277, issue.24, pp.21843-21850, 2002.

C. Chang, C. Chao, and W. Xia, P53 regulates epithelialmesenchymal transition and stem cell properties through modulating miRNAs, Nature Cell Biology, vol.13, issue.3, pp.317-323, 2011.

J. I. Yook, X. Li, and I. Ota, A Wnt-Axin2-GSK3 cascade regulates Snail1 activity in breast cancer cells, Nature Cell Biology, vol.8, issue.12, pp.1398-1406, 2006.

D. C. Radisky, D. D. Levy, and L. E. Littlepage, Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability, Nature, vol.436, issue.7047, pp.123-127, 2005.

B. P. Zhou, J. Deng, and W. Xia, Dual regulation of Snail by GSK-3 -mediated phosphorylation in control of epithelialmesenchymal transition, Nature Cell Biology, vol.6, issue.10, pp.931-940, 2004.

D. Javelaud, V. Delmas, and M. Möller, Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo, Oncogene, vol.24, issue.51, pp.7624-7629, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00147458

J. Massague, TGFbeta in cancer, Cell, vol.134, issue.2, pp.215-230, 2008.

S. Dennler, J. André, and I. Alexaki, Induction of sonic hedgehog mediators by transforming growth factor-: smad3-dependent activation of Gli2 and Gli1 expression in vitro and in vivo, Cancer Research, vol.67, issue.14, pp.6981-6986, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00194393

C. Y. Perrot, C. Gilbert, V. Marsaud, A. Postigo, D. Javelaud et al., GLI2 cooperates with ZEB1 for transcriptional repression of CDH1 expression in human melanoma cells, Pigment Cell & Melanoma Research, vol.26, issue.6, pp.861-873, 2013.

C. Y. Perrot, D. Javelaud, and A. Mauviel, Insights into the transforming growth factor-beta signaling pathway in cutaneous melanoma, Annals of Dermatology, vol.25, issue.2, pp.135-144, 2013.

C. Y. Logan and R. Nusse, The Wnt signaling pathway in development and disease, Annual Review of Cell and Developmental Biology, vol.20, pp.781-810, 2004.

J. Goodall, S. Martinozzi, and T. J. Dexter, Brn-2 expression controls melanoma proliferation and is directly regulated bycatenin, Molecular and Cellular Biology, vol.24, issue.7, pp.2915-2922, 2004.

T. He, A. B. Sparks, and C. Rago, Identification of c-MYC as a target of the APC pathway, Science, vol.281, issue.5382, pp.1509-1512, 1998.

M. Shtutman, J. Zhurinsky, and I. Simcha, The cyclin D1 gene is a target of the -catenin/LEF-1 pathway, Proceedings of the National Academy of Sciences of the United States of America, vol.96, pp.5522-5527, 1999.

D. L. Rimm, K. Caca, G. Hu, F. B. Harrison, and E. R. Fearon, Frequent nuclear/cytoplasmic localization of -catenin without exon 3 mutations in malignant melanoma, American Journal of Pathology, vol.154, issue.2, pp.325-329, 1999.

J. Reifenberger, C. B. Knobbe, and M. Wolter, Molecular genetic analysis of malignant melanomas for aberrations of the wnt signaling pathway genes CTNNB1, APC, ICAT and BTRC, International Journal of Cancer, vol.100, issue.5, pp.549-556, 2002.

V. Delmas, F. Beermann, and S. Martinozzi, Catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development, Genes and Development, vol.21, issue.22, pp.2923-2935, 2007.

C. Haqq, M. Nosrati, and D. Sudilovsky, The gene expression signatures of melanoma progression, Proceedings of the National Academy of Sciences of the United States of America, vol.102, pp.6092-6097, 2005.

S. Kuphal, S. Lodermeyer, F. Bataille, M. Schuierer, B. H. Hoang et al., Expression of Dickkopf genes is strongly reduced in malignant melanoma, Oncogene, vol.25, issue.36, pp.5027-5036, 2006.

Y. Lin, L. You, and Z. Xu, Wnt inhibitory factor-1 gene transfer inhibits melanoma cell growth, Human Gene Therapy, vol.18, issue.4, pp.379-386, 2007.

A. M. Mikheev, S. A. Mikheeva, R. Rostomily, and H. Zarbl, Dickkopf-1 activates cell death in MDA-MB435 melanoma cells, Biochemical and Biophysical Research Communications, vol.352, issue.3, pp.675-680, 2007.

A. J. Chien, E. C. Moore, and A. S. Lonsdorf, Activated Wnt/ -catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model, Proceedings of the National Academy of Sciences of the United States of America, vol.106, issue.4, pp.1193-1198, 2009.

K. S. Hoek, N. C. Schlegel, and P. Brafford, Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature, Pigment Cell Research, vol.19, issue.4, pp.290-302, 2006.

T. L. Biechele, R. M. Kulikauskas, and R. A. Toroni, Wnt/ -catenin signaling and AXIN1 regulate apoptosis triggered by inhibition of the mutant kinase BRAFV600E in human melanoma, Science Signaling, vol.5, issue.206, p.3, 2012.

S. K. Dissanayake, M. Wade, and C. E. Johnson, The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition, Journal of Biological Chemistry, vol.282, issue.23, pp.17259-17271, 2007.

A. T. Weeraratna, Y. Jiang, and G. Hostetter, Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma, Cancer Cell, vol.1, issue.3, pp.279-288, 2002.

K. Burridge and K. Wennerberg, Rho and RAC take center stage, Cell, vol.116, issue.2, pp.167-179, 2004.

X. R. Bustelo, V. Sauzeau, and I. M. Berenjeno, GTP-binding proteins of the Rho/Rac family: regulation, effectors and functions in vivo, BioEssays, vol.29, issue.4, pp.356-370, 2007.

L. M. Machesky and O. J. Sansom, Rac1 in the driver's seat for melanoma, Pigment Cell & Melanoma Research, vol.25, issue.6, pp.762-764, 2012.

M. J. Davis, B. H. Ha, E. C. Holman, R. Halaban, J. Schlessinger et al., RAC1P29S is a spontaneously activating cancer-associated GTPase, Proceedings of the National Academy of Sciences of the United States of Amrica, vol.110, pp.912-917, 2013.

C. R. Lindsay, S. Lawn, and A. D. Campbell, P-Rex1 is required for efficient melanoblast migration and melanoma metastasis, Nature Communications, vol.2, p.555, 2011.

A. Li, Y. Ma, and M. Jin, Activated mutant NRas(Q61K) drives aberrant melanocyte signaling, survival, and invasiveness via a Rac1-dependent mechanism, Journal of Investigative Dermatology, vol.132, issue.11, pp.2610-2621, 2012.

S. Wilhelm, C. Carter, and M. Lynch, Discovery and development of sorafenib: a multikinase inhibitor for treating cancer, Nature Reviews Drug Discovery, vol.5, issue.10, pp.835-844, 2006.

P. B. Chapman, A. Hauschild, and C. Robert, Improved survival with vemurafenib in melanoma with BRAF V600E mutation, New England Journal of Medicine, vol.364, issue.26, pp.2507-2516, 2011.

R. J. Sullivan and K. T. Flaherty, Resistance to BRAF-targeted therapy in melanoma, European Journal of Cancer, vol.49, issue.6, pp.1297-1304, 2013.

R. Anforth, P. Fernandez-penas, and G. V. Long, Cutaneous toxicities of RAF inhibitors, The Lancet Oncology, vol.14, issue.1, pp.11-18, 2013.

P. I. Poulikakos, C. Zhang, G. Bollag, K. M. Shokat, and N. Rosen, RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF, Nature, vol.464, issue.7287, pp.427-430, 2010.

A. Hauschild, J. J. Grob, and L. V. Demidov, Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, openlabel, phase 3 randomised controlled trial, The Lancet, vol.380, issue.9839, pp.358-365, 2012.

P. A. Ascierto, D. Schadendorf, and C. Berking, MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study, The Lancet Oncology, vol.14, issue.3, pp.249-256, 2013.

K. T. Flaherty, C. Robert, and P. Hersey, Improved survival with MEK inhibition in BRAF-mutated melanoma, New England Journal of Medicine, vol.367, issue.2, pp.107-114, 2012.

J. M. Kirkwood, L. Bastholt, and C. Robert, Phase II, openlabel, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanoma, Clinical Cancer Research, vol.18, issue.2, pp.555-567, 2012.

J. R. Todd, T. M. Becker, R. F. Kefford, and H. Rizos, Secondary c-Kit mutations confer acquired resistance to RTK inhibitors in c-Kit mutant melanoma cells, Pigment Cell & Melanoma Research, vol.26, issue.4, pp.518-526, 2013.

X. Jiang, J. Zhou, and N. K. Yuen, Imatinib targeting of KITmutant oncoprotein in melanoma, Clinical Cancer Research, vol.14, issue.23, pp.7726-7732, 2008.

S. Ugurel, R. Hildenbrand, and A. Zimpfer, Lack of clinical efficacy of imatinib in metastatic melanoma, British Journal of Cancer, vol.92, issue.8, pp.1398-1405, 2005.

K. Wyman, M. B. Atkins, and V. Prieto, Multicenter phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy, Cancer, vol.106, issue.9, 2005.

F. S. Hodi, P. Friedlander, and C. L. Corless, Major response to imatinib mesylate in KIT-mutated melanoma, Journal of Clinical Oncology, vol.26, issue.12, pp.2046-2051, 2008.

J. Lutzky, J. Bauer, and B. C. Bastian, Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation, Pigment Cell & Melanoma Research, vol.21, issue.4, pp.492-493, 2008.

F. S. Hodi, C. L. Corless, and A. Giobbie-hurder, Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin, Journal of Clinical Oncology, vol.31, issue.26, pp.3182-3190, 2013.

D. A. Oble, R. Loewe, P. Yu, and M. C. Mihm, Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human melanoma, Cancer Immunity, vol.9, 2009.

D. T. Alexandrescu, T. E. Ichim, and N. H. Riordan, Immunotherapy for melanoma: current status and perspectives, Journal of Immunotherapy, vol.33, issue.6, pp.570-590, 2010.

T. Iida, H. Ohno, and C. Nakaseko, Regulation of cell surface expression of CTLA-4 by secretion of CTLA-4-containing lysosomes upon activation of CD4+ T cells, Journal of Immunology, vol.165, issue.9, pp.5062-5068, 2000.

V. D. Van-den and . Eertwegh, Improved survival with ipilimumab in patients with metastatic melanoma, New England Journal of Medicine, vol.363, issue.8, pp.711-723, 2010.

C. Robert, L. Thomas, and I. Bondarenko, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma, New England Journal of Medicine, vol.364, issue.26, pp.2517-2526, 2011.

O. Hamid, Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma, New England Journal of Medicine, vol.369, issue.2, pp.134-144, 2013.

J. D. Wolchok, Nivolumab plus ipilimumab in advanced melanoma, New England Journal of Medicine, vol.369, issue.2, pp.122-133, 2013.

E. K. Nishimura, S. R. Granter, and D. E. Fisher, Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche, Science, vol.307, issue.5710, pp.720-724, 2005.

M. L. Harris, A dual role for SOX10 in the maintenance of the postnatal melanocyte lineage and the differentiation of melanocyte stem cell progenitors, PLOS Genetics, vol.9, issue.7, 2013.

N. Li, M. Fukunaga-kalabis, and H. Yu, Human dermal stem cells differentiate into functional epidermal melanocytes, Journal of Cell Science, vol.123, issue.6, pp.853-860, 2010.

D. Fang, T. K. Nguyen, and K. Leishear, A tumorigenic subpopulation with stem cell properties in melanomas, Cancer Research, vol.65, issue.20, pp.9328-9337, 2005.

E. Monzani, F. Facchetti, and E. Galmozzi, Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential, European Journal of Cancer, vol.43, issue.5, pp.935-946, 2007.

J. Dou, M. Pan, and P. Wen, Isolation and identification of cancer stem-like cells from murine melanoma cell lines, Cellular & Molecular Immunology, vol.4, issue.6, pp.467-472, 2007.

A. D. Boiko, O. V. Razorenova, and M. Van-de-rijn, Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271, Nature, vol.466, issue.7302, pp.133-137, 2010.

T. Schatton, G. F. Murphy, and N. Y. Frank, Identification of cells initiating human melanomas, Nature, vol.451, issue.7176, pp.345-349, 2008.

Y. Luo, K. Dallaglio, and Y. Chen, ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets, Stem Cells, vol.30, issue.10, pp.2100-2113, 2012.

A. Roesch, M. Fukunaga-kalabis, and E. C. Schmidt, A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth, Cell, vol.141, issue.4, pp.583-594, 2010.

A. Roesch, A. Vultur, and I. Bogeski, Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells, Cancer Cell, vol.23, issue.6, pp.811-825, 2013.

I. Vsquez-moctezuma, M. A. Meraz-ros, and C. G. Villanueva-lpez, ATP-binding cassette transporter ABCB5 gene is expressed with variability in malignant melanoma, Actas Dermo-Sifiliograficas, vol.101, issue.4, pp.341-348, 2010.

N. Y. Frank, A. Margaryan, and Y. Huang, ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma, Cancer Research, vol.65, issue.10, pp.4320-4333, 2005.

H. T. Khong, Q. J. Wang, and S. A. Rosenberg, Identification of multiple antigens recognized by tumor-infiltrating lymphocytes from a single patient: tumor escape by antigen loss and loss of MHC expression, Journal of Immunotherapy, vol.27, issue.3, pp.184-190, 2004.

J. Du, A. J. Miller, H. R. Widlund, M. A. Horstmann, S. Ramaswamy et al., MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma, American Journal of Pathology, vol.163, issue.1, pp.333-343, 2003.

T. A. Manolio, F. S. Collins, and N. J. Cox, Finding the missing heritability of complex diseases, Nature, vol.461, issue.7265, pp.747-753, 2009.