351
Views
42
CrossRef citations to date
0
Altmetric
Reviews

Design and discovery of mushroom tyrosinase inhibitors and their therapeutic applications

, &

Bibliography

  • Chang TS. An update review of tyrosinase inhibitors. Int J Mol Sci 2009;10:2440-75
  • Song YM, Ha YM, Kim JA, et al. Synthesis of novel azo-resveratrol, azo-oxyresveratrol and their derivatives as potent tyrosinase inhibitors. Bioorg Med Chem Lett 2012;22:7451-5
  • Liu ZJ, Herlyn M. Molecular biology of cutaneous melanoma. In: DeVita VT Jr, Hellman S, Rosenberg SA, editors, Cancer: principles and practice of oncology. 7th edition. Lippincott Williams & Wilkins, Philadelphia, PA; 2005
  • Cavalieri EL, Li KM, Balu N, et al. Catechol ortho-quinones: the electrophilic compounds that form depurinating DNA adducts and could initiate cancer and other diseases. Carcinogenesis 2002;23:1071-7
  • Vontzalidou A, Zoidis G, Chaita E, et al. Design, synthesis and molecular studies of dihydrostilbene derivatives as potentetyrosinase inhibitors. Bioorg Med Chem Lett 2012;22:5523-6
  • Hasegawa T. Tyrosinase-expressing neuronal cell line as in vitro model of Parkinson's disease. Int J Mol Sci 2010;11:1082-9
  • Tessari I, Bisaglia M, Valle F, et al. The reaction of-synuclein with tyrosinase. Possible implications for Parkinson disease. J Biol Chem 2008;283:16808-17
  • Greggio E, Bergantino E, Carter D. Tyrosinase exacerbates dopamine toxicity but is not genetically associated with Parkinson's disease. J Neurochem 2005;93:246-56
  • Nordlund J, Boissy R, Hearing V, et al. The pigmentary system: physiology and pathophysiology. Blackwell Publishing, Ltd, Oxford, UK; 2006
  • Walters AK, Roberts MS. Dermatologic, cosmeceutic, and cosmetic development: therapeutic and novel approaches. Informa Healthcare USA, Inc., New York; 2008
  • Raper HS. The tyrosinase-tyrosine reaction:production from tyrosine of 5: 6-dihydroxyindole and 5: 6- dihydroxyindole-2-carboxylic acid-the precursors of melanin. Biochem J 1927;21:89-96
  • Masom HS. The chemistry of melanin. III. Mechanism of the oxidation of dihydroxyphenyalanine by tyrosinase. J Biol Chem 1948;172:83-99
  • Schallreuter KU, Kothari S, Chavan B, Spencer JD. Regulation of melanogenesis – controversies and new concepts. Exp Dermatol 2008;17:395-404
  • Chang TS. Natural melanogenesis inhibitors acting through the down-regualtion of tyrosine activity. Materials 2012;5:1661-85
  • Tsukamoto K, Jackson IJ, Urabe K, et al. A second tyrosinase-related protein, TRP-2, is a melanogenic enzyme termed DOPAchrome tautomerase. EMBO J 1992;11:519-26
  • Jackson IJ, Chambers DM, Tsukamoto K, et al. A second tyrosinase related protein, TRP-2, maps to and is mutated at the mouse slaty locus. EMBO J 1992;11:527-35
  • Sarangarajan R, Zhao Y, Babcock G, et al. Mutant alleles at the brown locus encoding tyrosinase-related protein-1 (TRP-1) affect proliferation of mouse melanocytes in culture. Pigment Cell Res 2000;13:337-44
  • Ghanem G, Fabrice J. Tyrosinase related protein 1 (TYRP1/gp75) in human cutaneous melanoma. Mol Oncol 2011;5:150-5
  • Olivares C, Jimenez-Cervantes C, Lozano JA, et al. The 5,6-dihydroxyindole-2-carboxylic acid (DHICA) oxidase activity of human tyrosinase. Biochem J 2001;354:131-9
  • Wan P, Hu Y, He L. Regulation of melanocyte pivotal transcription factor MITF by some other transcription factors. Mol Cell Biochem 2011;354:241-6
  • Sun W, Wendt M, Klebe G, Röhm KH. On the interpretation of tyrosinase inhibition kinetics. J Enzyme Inhib Med Chem 2014;29(1):92-9
  • Rescigno A, Sollai F, Pisu B, et al. Tyrosinase inhibition: general and applied aspects. J Enzyme Inhib Med Chem 2002;17:207-18
  • Lim GGF, Imura Y, Yoshimura E. Substrate inhibition competes with halide inhibition in polyphenol oxidase. Protein J 2012;31:609-14
  • Kim YJ, Uyama H. Tyrosinase inhibitors from natural and synthetic sources: structure, inhibition mechanism and perspective for the future. Cell Mol Life Sci 2005;62:1707-23
  • Land EJ, Ramsden CA, Riley PA. The mechanism of suicide-inactivation of tyrosinase: a substrate structure investigation. Tohoku J Exp Med 2007;212:341-8
  • Land EJ, Ramsden CA, Riley PA, Stratford MRL. Evidence consistent with the requirement of cresolase activity for suicide-inactivation of tyrosinase. Tohoku J Exp Med 2008;216:231-8
  • Munoz-Munoz JL, Garcia-Molina F, Varon R, et al. Suicide inactivation of the diphenolase and monophenolase activities of tyrosinase. IUBMB Life 2010;62:539-47
  • Satooka H, Kubo I. Resveratrol as a Kcat inhibitor for tyrosinase: potentiated melanogenesis inhibitor. Bioorg Med Chem 2012;20:1090-9
  • Franco DCZ, Carvalho GSG, Rocha PR, et al. Inhibitory effects of resveratrol analogs on mushroom tyrosinase activity. Molecules 2012;17:11816-25
  • Bernard P, Berthon JY. Resveratrol: an original mechanism on tyrosinase inhibition. Int J Cosmet Sci 2000;22:219-26
  • Bae SJ, Ha YM, Park YJ, et al. Design, synthesis, and evaluation of (E)-N-substituted benzylidene-aniline derivatives as tyrosinase inhibitors. Eur J Med Chem 2012;57:383-90
  • Bae SJ, Ha YM, Kim JA, et al. A novel synthesized tyrosinase inhibitor: (E)-2-((2,4-dihydroxyphenyl)diazenyl)phenyl 4-methylbenzenesulfonate as an azo-resveratrol analog. Biosci Biotechnol Biochem 2013;77:65-72
  • El-Seedi HR, El-Said AMA, Khalifa SAM, et al. Biosynthesis, natural sources, dietary intake, pharmacokinetic properties, and biological activities of hydroxycinnamic acids. J Agric Food Chem 2012;60:10877-95
  • De P, Bedos-Belval F, Vanucci-Bacque C, et al. Cinnamic acid derivatives in tuberculosis, malaria and cardiovascular diseases - A Review. Curr Org Chem 2012;16:747-68
  • Sova M. Antioxidant and Antimicrobial activities of cinnamic acid derivatives. Mini Rev Med Chem 2012;12:749-67
  • De P, Baltas M, Bedos-Belval F. Cinnamic acid derivatives as anticancer agents-a review. Curr Med Chem 2011;18:1672-703
  • Shi Y, Chen QX, Wang Q, et al. Inhibitory effects of cinnamic acid and its derivatives on the diphenolase activity of mushroom (Agaricus bisporus) tyrosinase. Food Chem 2005;92:707-12
  • Fan Q, Jiang H, Yuan E, et al. Tyrosinase inhibitory effects and antioxidative activities of novel cinnamoyl amides with amino acid ester moiety. Food Chem 2005;134:1081-7
  • Kwak SY, Yang JK, Choi HR, et al. Synthesis and dual biological effects of hydroxycinnamoylphenylalanyl/prolylhydroxamic acid derivatives as tyrosinase inhibitor and antioxidant. Bioorg Med Chem Lett 2013;23:1136-42
  • Georgiev L, Chochkova M, Totseva I, et al. Anti-tyrosinase, antioxidant and antimicrobial activities of hydroxycinnamoylamides. Med Chem Res 2013;22:4173-82
  • Shi ZH, Li NG, Shi QP, et al. Design, synthesis, and preliminary evaluation of substituted cinnamic acid esters as selective matrix metalloproteinase inhibitors. Drug Dev Res 2012;73:317-24
  • Zhang Z, Liu J, Wu F, et al. Inhibitory effects of substituted cinnamic acid esters on mushroom tyrosinase. Lett Drug Des Discov 2013;10:529-34
  • Chochkova M, Stoykova B, Ivanova G, et al. N-Hydroxycinnamoyl amides of fluorinated amino acids: synthesis, anti-tyrosinase and DPPH scavenging activities. J Fluor Chem 2013;156:203-8
  • Hu YH, Liu X, Jia YL, et al. Inhibitory kinetics of chlorocinnamic acids on mushroom tyrosinase. J Biosci Bioeng 2014;117:142-6
  • Matoba Y, Kumagai T, Yamamoto A, et al. Crystallographic evidence that the dinuclear copper center of tyrosinase is flexible during catalysis. J Biol Chem 2006;281:8981-90
  • Klabunde T, Eicken C, Sacchettini JC, Krebs B. Crystal structure of a plant catechol oxidase containing a dicopper center. Nat Struct Biol 1998;5:1084-90
  • Jones K, Hughes J, Hong M, et al. Modulation of melanogenesis by aloesin: a competitive inhibitor of tyrosinase. Pigment Cell Res 2002;15:335-40
  • Fais A, Corda M, Era B, et al. Tyrosinase inhibitor activity of coumarin-resveratrol hybrids. Molecules 2009;14:2514-20
  • Matos MJ, Santana L, Uriarte E, et al. Tyrosine-like condensed derivatives as tyrosinase inhibitors. J Pharm Pharmacol 2012;64:742-6
  • Fang Y, Chen Y, Feng G, et al. Benzyl benzoates: new phlorizin analogs as mushroom tyrosinase inhibitors. Bioorg Med Chem 2011;19:1167-71
  • Roh JS, Han JY, Kim JH, et al. Inhibitory effects of active compounds isolated from safflower (Carthamustinctorius L.) seeds for melanogenesis. Biol Pharm Bull 2004;27:1976-8
  • Cho SJ, Roh JS, Sun WS, et al. N-benzylbenzamides: a new class of potent tyrosinase inhibitors. Bioorg Med Chem Lett 2006;16:2682-4
  • Baek HS, Hong YD, Lee CS, et al. Adamantyl N-benzylbenzamide: new series of depigmentation agents with tyrosinase inhibitory activity. Bioorg Med Chem Lett 2012;22:2110-13
  • Seo WD, Ryu YB, Curtis-Long MJ, et al. Evaluation of anti-pigmentary effect of synthetic sulfonylaminochalcone. Eur J Med Chem 2010;45:2010-17
  • Liu J, Chen C, Wu F, Zhao L. Microwave-assisted synthesis and tyrosinase inhibitory activity of chalcone derivatives. Chem Biol Drug Des 2013;82:39-47
  • Sonmez F, Sevmezler S, Atahan A, et al. Evaluation of new chalcone derivatives as polyphenol oxidase inhibitors. Bioorg Med Chem Lett 2011;21:7479-82
  • Bao K, Dai Y, Zhu ZB, et al. Design and synthesis of biphenyl derivatives as mushroom tyrosinase inhibitors. Bioorg Med Chem 2010;18:6708-14
  • Ha YM, Lee HJ, Park D, et al. Molecular docking studies of (1E,3E,5E)-1,6-Bis(substituted phenyl)hexa-1,3,5-triene and 1,4-Bis(substituted trans-styryl)benzene analogs as novel tyrosinase inhibitors. Biol Pharm Bull 2013;36:55-65
  • Noh JM, Kwak SY, Seo HS, et al. Kojic acid-amino acid conjugates as tyrosinase inhibitors. Bioorg Med Chem Lett 2009;19:5586-9
  • Rho HS, Ahn SM, Yoo DS, et al. Kojylthioether derivatives having both tyrosinase inhibitory and anti-inflammatory properties. Bioorg Med Chem Lett 2010;15:6569-71
  • Ahn SM, Rho HS, Baek HS, et al. Inhibitory activity of novel kojic acid derivative containing trolox moiety on melanogenesis. Bioorg Med Chem Lett 2011;21:7466-9
  • Criton M, Le Mellay-Hamon V. Analogues on N-hydroxy-N'-phenylthiourea and N-hydroxy-N'-phenylurea as inhibitors of tyrosinase and melanin formation. Bioorg Med Chem Lett 2008;18:3607-10
  • Thanigaimalai P, Lee KC, Sharma VK, et al. Structural requirement of phenylthiourea analogs for their inhibitory activity of melanogenesis and tyrosinase. Bioorg Med Chem Lett 2011;21:6824-8
  • Thanigaimalai P, Hoang TAL, Ki-Cheul L, et al. Structural requirement(s) of N-phenylthioureas and benzaldehydethiosemicarbazones as inhibitors of melanogenesis in melanoma B 16 cells. Bioorg Med Chem Lett 2010;20:2991-3
  • Gençer N, Demir D, Sonmez F, et al. New saccharin derivatives as tyrosinase inhibitors. Bioorg Med Chem 2012;20:2811-21
  • Lee KC, Thanigaimalai P, Jung SH, et al. Structural characteristics of thiosemicarbazones as inhibitors of melanogenesis. Bioorg Med Chem Lett 2010;20:6794-6
  • Yi W, Dubois W, Yahiaoui C, et al. Refinement of arylthiosemicarbazone pharmacophore in inhibition of mushroom tyrosinase. Eur J Med Chem 2011;46:4330-5
  • Yang MH, Chen CM, Hu YH, et al. Inhibitory kinetics of DABT and DABPT as novel tyrosinase inhibitors. J Biosci Bioeng 2013;115:514-17
  • Liu J, Cao R, Yi W, et al. A class of potent tyrosinase inhibitors: alkylidenethiosemicarbazide compounds. Eur J Med Chem 2009;44:1773-8
  • Liu J, Wu F, Chen L, et al. Biological evaluation of coumarin derivatives as mushroom tyrosinase inhibitors. Food Chem 2012;135:2872-8
  • Bandgar BP, Totre J V, Gawande SS, et al. Synthesis of novel 3,5-diaryl pyrazole derivatives using combinatorial chemistry as inhibitors of tyrosinase as well as potent anticancer, anti-inflammatory agents. Bioorg Med Chem 2010;18:6149-55
  • Tepper AW, Bubacco L, Canters GW. Structural basis and mechanism of the inhibition of the type-3 copper protein tyrosinase from Streptomyces antibioticus by halide ions. J Biol Chem 2002;277:30436-44
  • Zhou Z, Zhuo J, Yan S, Ma L. Design and synthesis of 3,5-diaryl-4,5-dihydro-1H-pyrazoles as new tyrosinase inhibitors. Bioorg Med Chem 2013;21:2156-62
  • Gawande SS, Warangkar SC, Bandgar BP, Khobragade CN. Synthesis of new heterocyclic hybrids based on pyrazole and thiazolidinone scaffolds as potent inhibitors of tyrosinase. Bioorg Med Chem 2013;2:2772-7
  • Ghani U, Ullah N. New potent inhibitors of tyrosinase: novel clues to binding of 1,3,4-thiadiazole-2(3H)-thiones, 1,3,4-oxadiazole-2(3H)-thiones, 4-amino-1,2,4-triazole-5(4H)-thiones, and substituted hydrazides to the dicopper active site. Bioorg Med Chem 2010;18:4042-8
  • Lam KW, Syahida A, Ul-Haq Z, et al. Synthesis and biological activity of oxadiazole and triazolothiadiazole derivatives as tyrosinase inhibitors. Bioorg Med Chem Lett 2010;20:3755-9
  • Ha YM, Park JY, Park YJ, et al. Synthesis and biological activity of hydroxy substituted phenyl-benzo[d]thiazole analogues for antityrosinase activity in B16 cells. Bioorg Med Chem Lett 2011;21:2445-9
  • Park JW, Ha YM, Moon K, et al. De novo tyrosinase inhibitor: 4-(6,7-Dihydro-5H-indeno[5,6-d]thiazol-2-yl) benzene-1,3-diol (MHY1556). Bioorg Med Chem Lett 2013;23:4172-6
  • Ha YM, Park YJ, Lee et al. Design, synthesis and biological evaluation of 2-(substituted phenyl)thiazolidine-4-carboxylic acid derivatives as novel tyrosinase inhibitors. Biochimie 2012;94:533-40
  • Han YK, Park YJ, Ha YM, et al. Characterization of a novel tyrosinase inhibitor, (2RS,4R)-2-(2,4-dihydroxyphenyl) thiazolidine-4-carboxylic acid (MHY384). Biochim Biophys Acta 2012;1820:542-9
  • Ha YM, Kim J-A, Park YJ, et al. Synthesis and biological activity of hydroxybenzylidenyl pyrrolidine-2,5-dione derivatives as new potent inhibitors of tyrosinase. Med Chem Commun 2011;2:542-9
  • Chung KW, Park YJ, Choi YJ, et al. Evaluation of in vitro and in vivo anti-melanogenic activity of a newly synthesized strong tyrosinase inhibitor (E)-3-(2,4 dihydroxybenzylidene)pyrrolidine-2,5-dione (3-DBP). Biochim Biophys Acta 2012;1820:962-9
  • Kim SH, Ha YM, Moon KM, et al. Anti-melanogenic effect of (Z)-5-(2,4-dihydroxybenzylidene)thiazolidine-2,4-dione, a novel tyrosinase inhibitor. Arch Pharm Res 2013;36:1189-97
  • Ha YM, Kim JA, Park YJ, et al. Analogs of 5-(substituted benzylidene)hydantoin as inhibitors of tyrosinase and melanin formation. Biochim Biophys Acta 2011;1810:612-19
  • Chung KW, Jeong HO, Jang EJ, et al. Characterization of a small molecule inhibitor of melanogenesis that inhibits tyrosinase activity and scavenges nitric oxide (NO). Biochim Biophys Acta 2013;1830:4752-61
  • Espín JC, Wichers HJ. Effect of captopril on mushroom tyrosinase activity in vitro. Biochim Biophys Acta 2001;1544:289-300
  • Chu HL, Wang BS, Chang LC, et al. Effects of captopril on melanin formation in B16 cells. J Food Drug Anal 2012;20:668-73
  • Kuo TC, Ho FM. Competitive inhibition of mushroom tyrosinase by Captopril. Res J Biotech 2013;8:26-9
  • Ubeid AA, Zhao L, Wang Y, Hantash BM. Short-sequence oligopeptides with inhibitory activity against mushroom and human tyrosinase. J Invest Dermatol 2009;129:2242-9
  • Reddy B, Jow T, Hantash BM. Bioactive oligopeptides in dermatology: part I. Exp Dermatol 2012;21:563-8
  • Kolbe L, Mann T, Gerwat W, et al. 4-n-Butylresorcinol, a highly effective tyrosinase inhibitor for the topical treatment of hyperpigmentation. J Eur Acad Dermatol Venereol 2013;27:19-23
  • Chandra M, Levitt J, Pensabene CA. Hydroquinone therapy for post-inflammatory hyperpigmentation secondary to acne: not just prescribable by dermatologists. Acta Derm Venereol 2012;92:232-5
  • Gillbro JM, Olsson MJ. The melanogenesis and mechanisms of skin-lightening agents - existing and new approaches. Int J Cosmet Sci 2011;33:210-21
  • An SM, Koh JS, Boo YC. p-Coumaric acid not only inhibits human tyrosinase activity in vitro but also melanogenesis in cells exposed to UVB. Phytother Res 2010;24:1175-80
  • Song K, An SM, Kim M, et al. Comparison of the antimelanogenic effects of p-coumaric acid and its methyl ester and their skin permeabilities. J Dermatol Sci 2011;63:17-22
  • Sugimoto K, Nishimura T, Nomura K, et al. Synthesis of arbutin-alpha-glycosides and a comparison of their inhibitory effects with those of alpha–arbutin and arbutin on human tyrosinase. Chem Pharm Bull (Tokyo) 2003;51:798-801
  • Shibahara S, Tomita Y, Tagami H, et al. Molecular basis for the heterogeneity of human tyrosinase. Tohoku J Exp Med 1988;156:403-14
  • Choi TY, Kim JH, Ko DH, et al. Zebra fish as a new model for phenotype-based screening of melanogenic regulatory compounds. Pigment Cell Res 2007;20:120-7
  • Ding HY, Chang TS, Chiang CM, et al. Melanogenesis inhibition by a crude extract of Magnolia officinalis. J Med Plants Res 2011;5:237-44

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.