DOI QR코드

DOI QR Code

Whitening effect of novel peptide mixture by regulating melanosome biogenesis, transfer and degradation

  • Received : 2020.01.02
  • Accepted : 2020.10.23
  • Published : 2021.01.01

Abstract

Peptides are short chain of amino acids linked by peptide bonds. They are widely used as effective and biocompatible active ingredients in cosmetic industry. In this study, we developed novel peptide mixture and identified its anti-pigmentation effect on melanocytes and keratinocytes. Our results revealed that peptide mixture inhibited melanosome biogenesis through the regulation of microphthalmia-associated transcription factor, a key factor of melanogenesis in melanocytes. And we observed that peptide mixture inhibited melanosome uptake through the reduction of protease-activated receptor 2, a phagocytosis-related receptor in keratinocytes. Furthermore, peptide mixture activated autophagy system resulting in degradation of transferred melanosomes in keratinocytes. The anti-pigmentation effect of multi-targeting peptide mixture was assessed in a human skin equivalent model (MelanoDerm). Melanin contents in epidermal layer were significantly decreased by topical treatment of peptide mixture, suggesting that it can be applied as a novel cosmetics material having a whitening function.

Keywords

Acknowledgement

This study was conducted under the support of the World Class 300 R&D Project by the Ministry of Trade, Industry, and Energy (Project number: S2641452).

References

  1. Hirobe T. Keratinocytes regulate the function of melanocytes. Dermatol Sin. 2014;32:200-204. https://doi.org/10.1016/j.dsi.2014.05.002
  2. Hachiya A, Kobayashi A, Ohuchi A, Takema Y, Imokawa G. The paracrine role of stem cell factor/c-kit signaling in the activation of human melanocytes in ultraviolet-B-induced pigmentation. J Invest Dermatol. 2001;116:578-586. https://doi.org/10.1046/j.1523-1747.2001.01290.x
  3. Hirobe T, Hasegawa K, Furuya R, Fujiwara R, Sato K. Effects of fibroblast-derived factors on the proliferation and differentiation of human melanocytes in culture. J Dermatol Sci. 2013;71:45-57. https://doi.org/10.1016/j.jdermsci.2013.03.012
  4. Schauer E, Trautinger F, Kock A, Schwarz A, Bhardwaj R, Simon M, Ansel JC, Schwarz T, Luger TA. Proopiomelanocortin-derived peptides are synthesized and released by human keratinocytes. J Clin Invest. 1994;93:2258-2262. https://doi.org/10.1172/JCI117224
  5. Chakraborty AK, Funasaka Y, Slominski A, Ermak G, Hwang J, Pawelek JM, Ichihashi M. Production and release of proopiomelanocortin (POMC) derived peptides by human melanocytes and keratinocytes in culture: regulation by ultraviolet B. Biochim Biophys Acta. 1996;1313:130-138. https://doi.org/10.1016/0167-4889(96)00063-8
  6. Wakamatsu K, Graham A, Cook D, Thody AJ. Characterisation of ACTH peptides in human skin and their activation of the melanocortin-1 receptor. Pigment Cell Res. 1997;10:288-297. https://doi.org/10.1111/j.1600-0749.1997.tb00688.x
  7. Imokawa G. Autocrine and paracrine regulation of melanocytes in human skin and in pigmentary disorders. Pigment Cell Res. 2004;17:96-110. https://doi.org/10.1111/j.1600-0749.2003.00126.x
  8. Chakraborty A, Slominski A, Ermak G, Hwang J, Pawelek J. Ultraviolet B and melanocyte-stimulating hormone (MSH) stimulate mRNA production for alpha MSH receptors and proopiomelanocortin-derived peptides in mouse melanoma cells and transformed keratinocytes. J Invest Dermatol. 1995;105:655-659. https://doi.org/10.1111/1523-1747.ep12324134
  9. Khaled M, Larribere L, Bille K, Aberdam E, Ortonne JP, Ballotti R, Bertolotto C. Glycogen synthase kinase 3beta is activated by cAMP and plays an active role in the regulation of melanogenesis. J Biol Chem. 2002;277:33690-33697. https://doi.org/10.1074/jbc.M202939200
  10. Kim YM, Cho SE, Seo YK. The activation of melanogenesis by p-CREB and MITF signaling with extremely low-frequency electromagnetic fields on B16F10 melanoma. Life Sci. 2016;162:25-32. https://doi.org/10.1016/j.lfs.2016.08.015
  11. Kameyama K, Sakai C, Kuge S, Nishiyama S, Tomita Y, Ito S, Wakamatsu K, Hearing VJ. The expression of tyrosinase, tyrosinaserelated proteins 1 and 2 (TRP1 and TRP2), the silver protein, and a melanogenic inhibitor in human melanoma cells of differing melanogenic activities. Pigment Cell Res. 1995;8:97-104. https://doi.org/10.1111/j.1600-0749.1995.tb00648.x
  12. Van Gele M, Geusens B, Schmitt AM, Aguilar L, Lambert J. Knockdown of myosin Va isoforms by RNAi as a tool to block melanosome transport in primary human melanocytes. J Invest Dermatol. 2008;128:2474-2484. https://doi.org/10.1038/jid.2008.100
  13. Ohbayashi N, Fukuda M. Role of Rab family GTPases and their effectors in melanosomal logistics. J Biochem. 2012;151:343-351. https://doi.org/10.1093/jb/mvs009
  14. Park JI, Lee HY, Lee JE, Myung CH, Hwang JS. Inhibitory effect of 2-methyl-naphtho[1,2,3-de]quinolin-8-one on melanosome transport and skin pigmentation. Sci Rep. 2016;6:29189. https://doi.org/10.1038/srep29189
  15. Oberhofer A, Spieler P, Rosenfeld Y, Stepp WL, Cleetus A, Hume AN, Mueller-Planitz F, Okten Z. Myosin Va's adaptor protein melanophilin enforces track selection on the microtubule and actin networks in vitro. Proc Natl Acad Sci U S A. 2017;114:E4714-E4723. https://doi.org/10.1073/pnas.1619473114
  16. Provance DW, James TL, Mercer JA. Melanophilin, the product of the leaden locus, is required for targeting of myosin-Va to melanosomes. Traffic. 2002;3:124-132. https://doi.org/10.1034/j.1600-0854.2002.030205.x
  17. Strom M, Hume AN, Tarafder AK, Barkagianni E, Seabra MC. A family of Rab27-binding proteins. Melanophilin links Rab27a and myosin Va function in melanosome transport. J Biol Chem. 2002;277:25423-25430. https://doi.org/10.1074/jbc.M202574200
  18. Wu XS, Rao K, Zhang H, Wang F, Sellers JR, Matesic LE, Copeland NG, Jenkins NA, Hammer JA 3rd. Identification of an organelle receptor for myosin-Va. Nat Cell Biol. 2002;4:271-278. https://doi.org/10.1038/ncb760
  19. Kuroda TS, Ariga H, Fukuda M. The actin-binding domain of Slac2-a/melanophilin is required for melanosome distribution in melanocytes. Mol Cell Biol. 2003;23:5245-5255. https://doi.org/10.1128/MCB.23.15.5245-5255.2003
  20. Cardinali G, Ceccarelli S, Kovacs D, Aspite N, Lotti LV, Torrisi MR, Picardo M. Keratinocyte growth factor promotes melanosome transfer to keratinocytes. J Invest Dermatol. 2005;125:1190-1199. https://doi.org/10.1111/j.0022-202X.2005.23929.x
  21. Epstein JH. Photocarcinogenesis, skin cancer, and aging. J Am Acad Dermatol. 1983;9:487-502. https://doi.org/10.1016/S0190-9622(83)70160-X
  22. Speeckaert R, Van Gele M, Speeckaert MM, Lambert J, van Geel N. The biology of hyperpigmentation syndromes. Pigment Cell Melanoma Res. 2014;27:512-524. https://doi.org/10.1111/pcmr.12235
  23. Maymone MBC, Neamah HH, Secemsky EA, Vashi NA. Correlating the Dermatology Life Quality Index and Skin Discoloration Impact Evaluation Questionnaire tools in disorders of hyperpigmentation. J Dermatol. 2018;45:361-362. https://doi.org/10.1111/1346-8138.14172
  24. Pillaiyar T, Manickam M, Namasivayam V. Skin whitening agents: medicinal chemistry perspective of tyrosinase inhibitors. J Enzyme Inhib Med Chem. 2017;32:403-425. https://doi.org/10.1080/14756366.2016.1256882
  25. Juhasz MLW, Levin MK. The role of systemic treatments for skin lightening. J Cosmet Dermatol. 2018;17:1144-1157. https://doi.org/10.1111/jocd.12747
  26. Zhang L, Falla TJ. Cosmeceuticals and peptides. Clin Dermatol. 2009;27:485-494. https://doi.org/10.1016/j.clindermatol.2009.05.013
  27. Reddy B, Jow T, Hantash BM. Bioactive oligopeptides in dermatology: part I. Exp Dermatol. 2012;21:563-568. https://doi.org/10.1111/j.1600-0625.2012.01528.x
  28. Chiaverini C, Beuret L, Flori E, Busca R, Abbe P, Bille K, Bahadoran P, Ortonne JP, Bertolotto C, Ballotti R. Microphthalmia-associated transcription factor regulates RAB27A gene expression and controls melanosome transport. J Biol Chem. 2008;283:12635-12642. https://doi.org/10.1074/jbc.M800130200
  29. Alves CP, Yokoyama S, Goedert L, Pontes CLS, Sousa JF, Fisher DE, Espreafico EM. MYO5A gene is a target of MITF in melanocytes. J Invest Dermatol. 2017;137:985-989. https://doi.org/10.1016/j.jid.2016.11.026
  30. Hartman ML, Czyz M. MITF in melanoma: mechanisms behind its expression and activity. Cell Mol Life Sci. 2015;72:1249-1260. https://doi.org/10.1007/s00018-014-1791-0
  31. Lajis AFB, Ariff AB. Discovery of new depigmenting compounds and their efficacy to treat hyperpigmentation: evidence from in vitro study. J Cosmet Dermatol. 2019;18:703-727. https://doi.org/10.1111/jocd.12900
  32. Garcia-Jimenez A, Teruel-Puche JA, Berna J, Rodriguez-Lopez JN, Tudela J, Garcia-Canovas F. Action of tyrosinase on alpha and betaarbutin: a kinetic study. PLoS One. 2017;12:e0177330. https://doi.org/10.1371/journal.pone.0177330
  33. Yu JS, Kim AK. Effect of combination of taurine and azelaic acid on antimelanogenesis in murine melanoma cells. J Biomed Sci. 2010;17(Suppl 1):S45. https://doi.org/10.1186/1423-0127-17-S1-S45
  34. Lee CS, Jang WH, Park M, Jung K, Baek HS, Joo YH, Park YH, Lim KM. A novel adamantyl benzylbenzamide derivative, AP736, suppresses melanogenesis through the inhibition of cAMP-PKACREB-activated microphthalmia-associated transcription factor and tyrosinase expression. Exp Dermatol. 2013;22:762-764. https://doi.org/10.1111/exd.12248
  35. Huang HC, Chang SJ, Wu CY, Ke HJ, Chang TM. [6]-Shogaol inhibits α-MSH-induced melanogenesis through the acceleration of ERK and PI3K/Akt-mediated MITF degradation. Biomed Res Int. 2014;2014:842569. https://doi.org/10.1155/2014/842569
  36. Kim B, Lee JY, Lee HY, Nam KY, Park J, Lee SM, Kim JE, Lee JD, Hwang JS. Hesperidin suppresses melanosome transport by blocking the interaction of Rab27A-melanophilin. Biomol Ther (Seoul). 2013;21:343-348. https://doi.org/10.4062/biomolther.2013.032
  37. Chang H, Choi H, Joo KM, Kim D, Lee TR. Manassantin B inhibits melanosome transport in melanocytes by disrupting the melanophilin-myosin Va interaction. Pigment Cell Melanoma Res. 2012;25:765-772. https://doi.org/10.1111/pcmr.12002
  38. Makino-Okamura C, Niki Y, Takeuchi S, Nishigori C, Declercq L, Yaroch DB, Saito N. Heparin inhibits melanosome uptake and inflammatory response coupled with phagocytosis through blocking PI3k/Akt and MEK/ERK signaling pathways in human epidermal keratinocytes. Pigment Cell Melanoma Res. 2014;27:1063-1074. https://doi.org/10.1111/pcmr.12287
  39. Seiberg M, Paine C, Sharlow E, Andrade-Gordon P, Costanzo M, Eisinger M, Shapiro SS. The protease-activated receptor 2 regulates pigmentation via keratinocyte-melanocyte interactions. Exp Cell Res. 2000;254:25-32. https://doi.org/10.1006/excr.1999.4692
  40. Murase D, Hachiya A, Takano K, Hicks R, Visscher MO, Kitahara T, Hase T, Takema Y, Yoshimori T. Autophagy has a significant role in determining skin color by regulating melanosome degradation in keratinocytes. J Invest Dermatol. 2013;133:2416-2424. https://doi.org/10.1038/jid.2013.165
  41. Kim ES, Shin JH, Seok SH, Kim JB, Chang H, Park SJ, Jo YK, Choi ES, Park JS, Yeom MH, Lim CS, Cho DH. Autophagy mediates anti-melanogenic activity of 3'-ODI in B16F1 melanoma cells. Biochem Biophys Res Commun. 2013;442:165-170. https://doi.org/10.1016/j.bbrc.2013.11.048
  42. Li L, Chen X, Gu H. The signaling involved in autophagy machinery in keratinocytes and therapeutic approaches for skin diseases. Oncotarget. 2016;7:50682-50697. https://doi.org/10.18632/oncotarget.9330
  43. Schagen SK. Topical peptide treatments with effective anti-aging results. Cosmetics. 2017;4:16. https://doi.org/10.3390/cosmetics4020016
  44. Reddy BY, Jow T, Hantash BM. Bioactive oligopeptides in dermatology: part II. Exp Dermatol. 2012;21:569-575. https://doi.org/10.1111/j.1600-0625.2012.01527.x
  45. Stromstedt AA, Pasupuleti M, Schmidtchen A, Malmsten M. Evaluation of strategies for improving proteolytic resistance of antimicrobial peptides by using variants of EFK17, an internal segment of LL-37. Antimicrob Agents Chemother. 2009;53:593-602. https://doi.org/10.1128/AAC.00477-08
  46. Pai VV, Bhandari P, Shukla P. Topical peptides as cosmeceuticals. Indian J Dermatol Venereol Leprol. 2017;83:9-18. https://doi.org/10.4103/0378-6323.186500
  47. Marepally S, Boakye CH, Shah PP, Etukala JR, Vemuri A, Singh M. Design, synthesis of novel lipids as chemical permeation enhancers and development of nanoparticle system for transdermal drug delivery. PLoS One. 2013;8:e82581. https://doi.org/10.1371/journal.pone.0082581
  48. Kalluri H, Banga AK. Transdermal delivery of proteins. AAPS PharmSciTech. 2011;12:431-441. https://doi.org/10.1208/s12249-011-9601-6