リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Elucidation of Hair Growth and Hair Pigmentation Promotion Effect of 3,4,5-Tri-O-Caffeoylquinic Acid」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Elucidation of Hair Growth and Hair Pigmentation Promotion Effect of 3,4,5-Tri-O-Caffeoylquinic Acid

メリエム, ベジャウィ Meriem, Bejaoui 筑波大学

2020.07.27

概要

Hair follicle (HF) undergoes a regular cycle composed of three phases: anagen, catagen, and telogen. During anagen phase or the growth phase, dermal papilla cells (DPs) located at the proximal end of the HF, regulates the induction of the hair cycle and the formation of the hair shaft. Moreover, during this phase, the bulbar melanocytes are active and produce the pigment melanin. The HF enters then a regression phase (catagen) followed by a resting phase (telogen) where the hairs are normally shed. Various signaling pathways regulate the hair growth cycle, such as the canonical Wnt/β-catenin signaling as it is involved in the regulation of the development, growth, and proliferation of the HF and the regulation of the melanocytes and the pigmentation. Wnt/β- catenin activation in the HF initiates the anagen phase initiation by inducing the proliferation and differentiation of matrix cells producing the pigmented hair shaft. An alteration in the pathways regulating the HF, can lead to a shorten length of anagen phase and an increase rate of hair loss (alopecia) and hair graying (canities).

 As hair plays an important role in individual's general appearance, hair problems are a major concern in the modern society. Hair loss and hair graying are common disorders that appear both in men and women undependably of the age or the origin, yet the mechanism by which they occur is not well understood. The current available therapies rely on synthetic chemicals ingredients and have showed many severe side effects and temporary results. The Food and Drug Administration (FDA) have approved two treatment against hair loss: Minoxidil and Finasteride but unwanted sides effect were observed including: impotence, dizziness, unwanted grown hair, weakness, headache, skin rash, and others. Developing a safe drug with a lasting effect has become a necessity to treat hair disorders and to have an alternative therapy for promoting hair growth and pigmentation.

 In this context, caffeoylquinic acid (CQA) is a phenylpropanoid compound exhibiting several beneficial properties including anti-oxidant, anti-allergic, neuroprotective, and melanogenesis-regulating effects. TCQA or 3,4,5-tri-O-caffeoylquinic acid is a derivative of CQA and chlorogenic acid (CGA) family that has a stable albumin affinity and is composed of multi- esters formed between quinic acid and one-to-four residues of trans-caffeic acids. TCQA has been found to induce a powerful inhibitory activities against aldose reductase, hypertension, hyperglycemia, and Alzheimer's disease without unwanted secondary effects. Moreover, TCQA induces neurogenesis, improves learning and memory in aged mice, and promotes the differentiation of human neural stem cells, however its effect on hair growth and hair pigmentation promotion has not yet been assessed. In the present study, the effect of TCQA on the induction of hair pigmentation and growth through exploring the possible common pathways such as Wnt/β- catenin was investigated In this purpose, the dorsal back of eight-weeks-old C3H male mice was shaved and TCQA was applied topically for a month. Results showed that TCQA stimulated the induction and the acceleration of the anagen phase of the hair growth cycle compared to the untreated mice. TCQA significantly enhanced the hair growth by approximately 120% by the end of treatment. In contrast, for the control group, only 37% hair regrowth area was observed. In addition, the hair plucked from the treated area of TCQA-treated mice displayed a darker color compared with the control and melanin assay showed that the melanin content increased up to 175% after TCQA application. To uncover the mechanism behind the effect of TCQA on hair cycle, microarray analysis was conducted. An upregulation in hair growth- and pigmentation-associated genes was observed. Genes relevant in β-catenin binding, pigmentation, neural cells differentiation and migration, transcription regulation, and Wnt signaling were upregulated. On the other hand, genes involved in Wnt repression, melanin degradation, and β-catenin degradation complex were downregulated. Immunohistochemistry results from skin collected from the treated area, revealed an accumulation of β-catenin expression in the epidermis and the hair bulb where the DPs and the active melanocytes are located. These results demonstrated also an activation of Tyrosinase (TYR) involved in melanin synthesis, in the hair bulb, showing that the melanocyte are active and producing the pigment melanin. A staining of CD34 an anagen marker in the skin, showed the stimulation of this protein in TCQA-treated skin compared with the control. These immunohistochemistry results revealed that the HF from TCQA-treated mice are in the anagen phase of the hair cycle, which means that the hair matrix cells are differentiating to form the hair shaft and the melanocytes are active and produce the melanin.

 Furthermore, TCQA was used on human and mouse cell lines to further validate its effect on hair growth and melanogenesis. Human hair follicle dermal papilla cells (HFDPC) was cultured in the presence of the sample and the proliferation along with the ATP content were significantly enhanced upon the treatment. In addition, the protein and the gene expression of β-catenin was upregulated in TCQA-treated cells, even after inhibition with XAV939. This results prove further the effect of TCQA on the stimulation of hair growth via the activation of β-catenin in dermal papilla cells.

 Human melanocytes (HEM), melanin-producing cells and B16 murine melanoma cells (B16F10), were used to further validate the effect of TCQA on pigmentation. Melanin content along with the gene and protein expression of the melanogenesis enzymes TYR, TYRP1 (tyrosinase-related protein 1), and DCT (dopachrome tautomerase) were upregulated upon treatment. This effect is attributed to the activation of microphthalmia-associated transcription factor (MITF) the transcription factor of the melanogenesis enzymes caused by the enhancement of the expression of β-catenin known to activate Mitf.

 Taken together, the finding of this thesis showed that TCQA triggered the activation of Wnt/β-catenin pathway leading to the transition from telogen to anagen phase, anagen phase initiation and elongation, hair matrix differentiation, and hair shaft development and pigmentation.

 Furthermore, the observed stimulation of hair growth cycle was supported by the downregulation of telogen- and aging-associated genes, Wnt signal inhibitors, and melanin degradation-associated genes which contributed to further enhance the effect of β-catenin and Wnt proteins activation. TCQA successfully activated the proliferation of dermal papilla cells often considered the key regulator of hair growth cycle and the melanocytes, melanin-producing cells.

 In this context, the potential effect of TCQA on the activation of melanocytes through a communication with dermal papilla cells via Wnt/β-catenin will be looked at. Clinical studies would be however necessary to introduce TCQA as a safer drug to initiate the hair growth cycle and to enhance melanogenesis.

この論文で使われている画像

参考文献

1. Jaks V, Kasper M, Toftgård R. The hair follicle-a stem cell zoo. Exp Cell Res. Elsevier Inc.; 2010; 316: 1422–8.

2. Schneider MR, Schmidt-Ullrich R, Paus R. The Hair Follicle as a Dynamic Miniorgan. Curr Biol. Elsevier Ltd; 2009; 19: R132–42.

3. Hardy MH. The secret life of the hair follicle. Trends Genet. 1992; 8: 55–61.

4. Yang CC, Cotsarelis G. Review of hair follicle dermal cells. J Dermatol Sci. 2010; 57: 2– 11.

5. Lin MH, Leimeister C, Gessler M, Kopan R. Activation of the Notch pathway in the hair cortex leads to aberrant differentiation of the adjacent hair-shaft layers. Development. 2000; 127: 2421–32.

6. Millar SE. WNT signaling in the control of hair growth and structure. Dev Biol [Internet]. 1999; 207: 133–49. Available from: http://dx.doi.org/10.1006/dbio.1998.9140

7. Nishimura EK, Yoshida H, Kunisada T, Nishikawa SI. Regulation of E- and P-cadherin expression correlated with melanocyte migration and diversification. Dev Biol. 1999; 215: 155–66.

8. Tobin DJ, Slominski A, Botchkarev V, Paus R. The fate of hair follicle melanocytes during the hair growth cycle. J Investig Dermatology Symp Proc [Internet]. Elsevier Masson SAS; 1999; 4: 323–32. Available from: http://dx.doi.org/10.1038/sj.jidsp.5640239

9. Stenn KS, Paus R. Controls of hair follicle cycling. Physiol Rev. 2001; 81: 449–94.

10. Driskell RR, Clavel C, Rendl M, Watt FM. Hair follicle dermal papilla cells at a glance. J Cell Sci. 2011; 124: 1179–82.

11. Lavker RM, Sun T, Oshima H, Barrandon Y, Akiyama M, Ferraris C, Chevalier G, Favier B, Jahoda CAB, Dhouailly D, Panteleyev AA, Christiano AM. Hair Follicle Stem Cells. J Investig Dermatology Symp Proc. Elsevier Masson SAS; 2003; 8: 28–38.

12. Lin JY, Fisher DE. Melanocyte biology and skin pigmentation. Nature [Internet]. Nature Publishing Group; 2007; 445: 843. Available from: https://doi.org/10.1038/nature05660

13. Nishimura EK, Jordan SA, Oshima H, Yoshida H, Osawa M, Moriyama M, Jackson IJ, Barrandon Y, Miyachi Y, Nishikawa S-I. Dominant role of the niche in melanocyte stem- cell fate determination. Nature [Internet]. 2002; 416: 854–60. Available from: https://doi.org/10.1038/416854a

14. Elliott K, Stephenson TJ, Messenger AG. Differences in hair follicle dermal papilla volume are due to extracellular matrix volume and cell number: Implications for the control of hair follicle size and androgen responses. J Invest Dermatol [Internet]. Elsevier Masson SAS; 1999; 113: 873–7. Available from: http://dx.doi.org/10.1046/j.1523- 1747.1999.00797.x

15. Müller-Röver S, Handjiski B, Van Der Veen C, Eichmüller S, Foitzik K, McKay IA, Stenn KS, Paus R. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J Invest Dermatol. 2001; 117: 3–15.

16. Lindner G, Botchkarev VA, Botchkareva N V., Ling G, Van Der Veen C, Paus R. Analysis of apoptosis during hair follicle regression (catagen). Am J Pathol. 1997; 151: 1601–17.

17. Ma L, Liu J, Wu T, Plikus M, Jiang TX, Bi Q, Liu YH, Müller-Röver S, Peters H, Sundberg JP, Maxson R, Maas RL, Chuong CM. “Cyclic alopecia” in Msx2 mutants: Defects in hair cycling and hair shaft differentiation. Development. 2003; 130: 379–89.

18. Messenger a G. The control of hair growth: an overview. J Invest Dermatol. 1993; 101: 4S-9S.

19. Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell. 2004; 118: 635–48.

20. McNeill H, Woodgett JR. When pathways collide: Collaboration and connivance among signalling proteins in development. Nat Rev Mol Cell Biol [Internet]. Nature Publishing Group; 2010; 11: 404–13. Available from: http://dx.doi.org/10.1038/nrm2902

21. Guo L, Yu Q, Fuchs E. Targeting expression of keratinocyte growth factor to keratinocytes elicits striking changes in epithelial differentiation in transgenic mice. 1993; 12: 973–86.

22. Kuwahara A, Sakai H, Xu Y, Itoh Y, Hirabayashi Y, Gotoh Y. Tcf3 represses Wnt-β- catenin signaling and maintains neural stem cell population during neocortical development. PLoS One. 2014; 9: 1–12.

23. Andl T, Reddy ST, Gaddapara T, Millar SE. WNT signals are required for the initiation of hair follicle development. Dev Cell. 2002; 2: 643–53.

24. Merrill BJ, Gat U, DasGupta R, Fuchs E. Tcf3 and Lef1 regulate lineage differentiation of multipotent stem cells in skin. Genes Dev. 2001; 15: 1688–705.

25. Gat U, DasGupta R, Degenstein L, Fuchs E. De novo hair follicle morphogenesis and hair tumors in mice expressing a truncated β-catenin in skin. Cell. 1998; 95: 605–14.

26. Van Genderen C, Okamura RM, Farinas I, Quo RG, Parslow TG, Bruhn L, Grosschedl R. Development of several organs that require inductive epithelial- mesenchymal interactions is impaired in LEF-1-deficient mice. Genes Dev. 1994; 8: 2691–703.

27. Widlund HR, Horstmann MA, Roydon Price E, Cui J, Lessnick SL, Wu M, He X, Fisher DE. β-Catenin-induced melanoma growth requires the downstream target Microphthalmia- associated transcription factor. J Cell Biol. 2002; 158: 1079–87.

28. Huelsken J, Vogel R, Erdmann B, Cotsarelis G, Birchmeier W. β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001; 105: 533–45.

29. Legrand JMD, Roy E, Ellis JJ, Francois M, Brooks AJ, Khosrotehrani K. STAT5 Activation in the Dermal Papilla Is Important for Hair Follicle Growth Phase Induction. J Invest Dermatol [Internet]. The Authors; 2016; 136: 1781–91. Available from: http://dx.doi.org/10.1016/j.jid.2016.04.014

30. St-Jacques B, Dassule HR, Karavanova I, Botchkarev VA, Li J, Danielian PS, McMahon JA, Lewis PM, Paus R, McMahon AP. Sonic hedgehog signaling is essential for hair development. Curr Biol. 1998; 8: 1058–69.

31. Wang LC, Liu ZY, Gambardella L, Delacour A, Shapiro R, Yang J, Sizing I, Rayhorn P, Garber EA, Benjamin CD, Williams KP, Taylor FR, Barrandon Y, et al. Conditional disruption of hedgehog signaling pathway defines its critical role in hair development and regeneration. J Invest Dermatol. Elsevier Masson SAS; 2000; 114: 901–8.

32. Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, Toftgård R. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet. 2008; 40: 1291–9.

33. Huh SH, Närhi K, Lindfors PH, Häärä O, Yang L, Ornitz DM, Mikkola ML. Fgf20 governs formation of primary and secondary dermal condensations in developing hair follicles. Genes Dev. 2013; 27: 450–8.

34. Coutu DL, Galipeau J. Roles of FGF signaling in stem cell self-renewal, senescence and aging. Aging (Albany NY). 2011; 3: 920–33.

35. Lin WH, Xiang LJ, Shi HX, Zhang J, Jiang LP, Cai PT, Lin ZL, Lin BB, Huang Y, Zhang HL, Fu XB, Guo DJ, Li XK, et al. Fibroblast growth factors stimulate hair growth through β -Catenin and shh expression in C57BL/6 mice. Biomed Res Int. Hindawi Publishing Corporation; 2015; 2015.

36. Botchkarev VA, Kishimoto J. Molecular control of epithelial-mesenchymal interactions during hair follicle cycling. J Investig Dermatology Symp Proc. Elsevier Masson SAS; 2003; 8: 46–55.

37. Ten Dijke P, Hill CS. New insights into TGF-β-Smad signalling. Trends Biochem Sci. 2004; 29: 265–73.

38. Rishikaysh P, Dev K, Diaz D, Shaikh Qureshi WM, Filip S, Mokry J. Signaling involved in hair follicle morphogenesis and development. Int J Mol Sci. 2014; 15: 1647–70.

39. Kobielak K, Pasolli HA, Alonso L, Polak L, Fuchs E. Defining BMP functions in the hair follicle by conditional ablation of BMP receptor IA. J Cell Biol. 2003; 163: 609–23.

40. Kobayashi T, Urabe K, Winder A, Jiménez-Cervantes C, Imokawa G, Brewington T, Solano F, García-Borrón JC, Hearing VJ. Tyrosinase related protein 1 (TRP1) functions as a DHICA oxidase in melanin biosynthesis. EMBO J [Internet]. 1994; 13: 5818–25. Available from: http://www.ncbi.nlm.nih.gov/pubmed/7813420%0Ahttp://www.pubmedcentral.nih.gov/art iclerender.fcgi?artid=PMC395555

41. Hou L, Panthier JJ, Arnheiter H. Signaling and transcriptional regulation in the neural crest-derived melanocyte lineage: interactions between KIT and MITF. Development [Internet]. 2000; 127: 5379–89. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11076759

42. D’Mello SAN, Finlay GJ, Baguley BC, Askarian-Amiri ME. Signaling pathways in melanogenesis. Int J Mol Sci. 2016; 17: 1–18.

43. Ngeow KC, Friedrichsen HJ, Li L, Zeng Z, Andrews S, Volpon L, Brunsdon H, Berridge G, Picaud S, Fischer R, Lisle R, Knapp S, Filippakopoulos P, et al. BRAF/MAPK and GSK3 signaling converges to control MITF nuclear export. Proc Natl Acad Sci. 2018; 115: E8668–77.

44. Passeron T, Valencia JC, Bertolotto C, Hoashi T, Le Pape E, Takahashi K, Ballotti R, Hearing VJ. SOX9 is a key player in ultraviolet B-induced melanocyte differentiation and pigmentation. Proc Natl Acad Sci. 2007; 104: 13984–9.

45. Levy C, Khaled M, Fisher DE. MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol Med. 2006; 12: 406–14.

46. Ouji Y, Yoshikawa M, Moriya K, Nishiofuku M, Matsuda R, Ishizaka S. Wnt-10b, uniquely among Wnts, promotes epithelial differentiation and shaft growth. Biochem Biophys Res Commun. 2008; 367: 299–304.

47. Zhou L, Xu M, Yang Y, Yang K, Wickett RR, Andl T, Millar SE, Zhang Y. Activation of β-Catenin Signaling in CD133-Positive Dermal Papilla Cells Drives Postnatal Hair Growth. PLoS One. 2016; 11: e0160425.

48. Almohanna HM, Ahmed AA, Tsatalis JP, Tosti A. The Role of Vitamins and Minerals in Hair Loss: A Review. Dermatol Ther (Heidelb) [Internet]. Springer Healthcare; 2019; 9: 51–70. Available from: https://doi.org/10.1007/s13555-018-0278-6

49. Tru RM. Molecular mechanisms of androgenetic alopecia. Exp Gerontol. 2002; 37: 981–90.

50. Hordinsky M, Junqueira AL. Alopecia areata update. Semin Cutan Med Surg. 2015; 34: 72–5.

51. Harrison S, Sinclair R. Telogen effluvium. 2002; : 389–95.

52. Schnohr P, Lange P, Nyboe J, Appleyard M, Jensen G. Gray hair, baldness, and wrinkles in relation to myocardial infarction: The Copenhagen City Heart Study. Am Heart J. 1995; 130: 1003–10.

53. ElFaramawy AAA, Hanna IS, Darweesh RM, Ismail AS, Kandil HI. The degree of hair graying as an independent risk marker for coronary artery disease, a CT coronary angiography study. Egypt Hear J [Internet]. Egyptian Society of Cardiology; 2018; 70: 15–9. Available from: https://doi.org/10.1016/j.ehj.2017.07.001

54. Shin H, Ryu HH, Yoon J, Jo S, Jang S, Choi M, Kwon O, Jo SJ. Association of premature hair graying with family history, smoking, and obesity: A cross-sectional study. J Am Acad Dermatol [Internet]. Elsevier Inc; 2015; 72: 321–7. Available from: http://dx.doi.org/10.1016/j.jaad.2014.11.008

55. Doubaj Y, De Sandre-Giovannoli A, Vera EV, Navarro CL, Elalaoui SC, Tajir M, Lévy N, Sefiani A. An inherited LMNA gene mutation in atypical Progeria syndrome. Am J Med Genet Part A. 2012; 158 A: 2881–7.

56. Seiberg M. Age-induced hair greying - The multiple effects of oxidative stress. Int J Cosmet Sci. 2013; 35: 532–8.

57. Tobin DJ, Paus R. Graying: Gerontobiology of the hair follicle pigmentary unit. Exp Gerontol. 2001; 36: 29–54.

58. Arck PC, Overall R, Spatz K, Liezman C, Handjiski B, Klapp BF, Birch-Machin MA, Peters EMJ. Towards a “free radical theory of graying”: Melanocyte apoptosis in the aging human hair follicle is an indicator of oxidative stress induced tissue damage. FASEB J. 2006; 20.

59. Commo S, Gaillard O, Bernard BA. Human hair greying is linked to a specific depletion of hair follicle melanocytes affecting both the bulb and the outer root sheath. Br J Dermatol. 2004; 150: 435–43.

60. Slominski A, Wortsman J, Plonka PM, Schallreuter KU, Paus R, Tobin DJ. Hair follicle pigmentation. J Invest Dermatol [Internet]. Elsevier Masson SAS; 2005; 124: 13–21. Available from: http://dx.doi.org/10.1111/j.0022-202X.2004.23528.x

61. Nishimura EK, Granter SR, Fisher DE. Mechanisms of hair graying: Incomplete melanocyte stem cell maintenance in the niche. Science (80- ). 2005; 307: 720–4.

62. Nishimura EK, Suzuki M, Igras V, Du J, Lonning S, Miyachi Y, Roes J, Beermann F, Fisher DE. Key Roles for Transforming Growth Factor β in Melanocyte Stem Cell Maintenance. Cell Stem Cell. 2010; 6: 130–40.

63. Nishimura EK. Melanocyte stem cells: A melanocyte reservoir in hair follicles for hair and skin pigmentation. Pigment Cell Melanoma Res. 2011; 24: 401–10.

64. Li M, Marubayashi A, Nakaya Y, Fukui K, Arase S. Minoxidil-induced hair growth is mediated by adenosine in cultured dermal papilla cells: Possible involvement of sulfonylurea receptor 2B as a target of minoxidil. J Invest Dermatol [Internet]. Elsevier Masson SAS; 2001; 117: 1594–600. Available from: http://dx.doi.org/10.1046/j.0022- 202x.2001.01570.x

65. Kaufman KD, Olsen EA, Whiting D, Savin R, DeVillez R, Bergfeld W, Price VH, Van Neste D, Roberts JL, Hordinsky M, Shapiro J, Binkowitz B, Gormley GJ, et al. Finasteride in the treatment of men with androgenetic alopecia. J Am Acad Dermatol. 1998; 39: 578– 89.

66. Van Neste D, Fuh V, Sanchez-Pedreno P, Lopez-Bran E, Wolff H, Whiting D, Roberts J, Kopera D, Stene JJ, Calvieri S, Tosti A, Prens E, Guarrera M, et al. Finasteride increases anagen hair in men with androgenetic alopecia. Br J Dermatol. 2000; 143: 804–10.

67. Kwack MH, Kang BM, Kim MK, Kim JC, Sung YK. Minoxidil activates b -catenin pathway in human dermal papilla cells : A possible explanation for its anagen prolongation effect. J Dermatol Sci [Internet]. Japanese Society for Investigative Dermatology; 2011; 62: 154–9. Available from: http://dx.doi.org/10.1016/j.jdermsci.2011.01.013

68. Rathi V, Rathi JC, Tamizharasi S, Kumar A. Phcog Rev .: Short Review Plants used for hair growth promotion : A review. Rev Lit Arts Am. 2008; 2: 185–7.

69. Miyamae Y, Han J, Sasaki K, Terakawa M, Isoda H, Shigemori H. 3,4,5-tri-O- caffeoylquinic acid inhibits amyloid β-mediated cellular toxicity on SH-SY5Y cells through the upregulation of PGAM1 and G3PDH. Cytotechnology. 2011; 63: 191–200.

70. Han J, Miyamae Y, Shigemori H, Isoda H. Neuroprotective effect of 3,5-di-O- caffeoylquinic acid on SH-SY5Y cells and senescence-accelerated-prone mice 8 through the up-regulation of phosphoglycerate kinase-1. Neuroscience [Internet]. Elsevier Inc.; 2010; 169: 1039–45. Available from: http://dx.doi.org/10.1016/j.neuroscience.2010.05.049

71. KIMURA Y, OKUDA H, OKUDA T, HATANO T, AGATA I, ARICHI S. Studies on the activities of tannins and related compounds from medicinal plants and drugs. VI. Inhibitory effects of caffeoylquinic acids on histamine release from rat peritoneal mast cells. Chem Pharm Bull (Tokyo) [Internet]. 1985; 33: 690–6. Available from: http://joi.jlc.jst.go.jp/JST.Journalarchive/cpb1958/33.690?from=CrossRef

72. Kim HJ, Kim JS, Woo JT, Lee IS, Cha BY. Hyperpigmentation mechanism of methyl 3,5- di-caffeoylquinate through activation of p38 and MITF induction of tyrosinase. Acta Biochim Biophys Sin (Shanghai). 2015; 47: 548–56.

73. Tang B, Huang Y, Yang H, Tang P, Li H. Molecular mechanism of the binding of 3,4,5- tri-O-caffeoylquinic acid to human serum albumin: Saturation transfer difference NMR, multi-spectroscopy, and docking studies. J Photochem Photobiol B Biol [Internet]. Elsevier B.V.; 2016; 165: 24–33. Available from: http://dx.doi.org/10.1016/j.jphotobiol.2016.10.017

74. Sasaki K, Davies J, Doldán NG, Arao S, Ferdousi F, Szele FG, Isoda H. 3,4,5- Tricaffeoylquinic acid induces adult neurogenesis and improves deficit of learning and memory in aging model senescence-accelerated prone 8 mice. Aging (Albany NY) [Internet]. 2019; 11: 1–22. Available from: http://www.aging-us.com/article/101748/text

75. Plikus M V., Chuong CM. Complex hair cycle domain patterns and regenerative hair waves in living rodents. J Invest Dermatol. 2008; 128: 1071–80.

76. Alonso L, Fuchs E. The hair cycle. J Cell Sci. 2006; 119: 391–3.

77. Milner Y, Sudnik J, Filippi M, Kizoulis M, Kashgarian M, Stenn K. Exogen, shedding phase of the hair growth cycle: Characterization of a mouse model. J Invest Dermatol. Elsevier Masson SAS; 2002; 119: 639–44.

78. Kishimoto J, Burgeson RE, Morgan BA. Wnt signaling maintains the hair-inducing activity of the dermal papilla service Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 2000; 14: 1181–5.

79. Paus R. Principles of hair cycle control. J Dermatol. 1998; 25: 793–802.

80. Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, Pervaiz S, Dharmarajan A. Multi-lineage differentiation of mesenchymal stem cells - To Wnt, or not Wnt. Int J Biochem Cell Biol [Internet]. Elsevier Ltd; 2015; 68: 139–47. Available from: http://dx.doi.org/10.1016/j.biocel.2015.09.008

81. Choi YS, Zhang Y, Xu M, Yang Y, Ito M, Peng T, Cui Z, Nagy A, Hadjantonakis AK, Lang RA, Cotsarelis G, Andl T, Morrisey EE, et al. Distinct functions for Wnt/β-Catenin in hair follicle stem cell proliferation and survival and interfollicular epidermal homeostasis. Cell Stem Cell [Internet]. Elsevier Inc.; 2013; 13: 720–33. Available from: http://dx.doi.org/10.1016/j.stem.2013.10.003

82. Guo L, Yu Q-C, Fuchs1 E. Targeting expression of keratinocyte growth factor to keratinocytes elicits striking changes in epithelial differentiation in transgenic mice. EMBO J [Internet]. 1993; 12: 973–86. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC413298/pdf/emboj00075-0168.pdf

83. Pan Y, Lin MH, Tian X, Cheng HT, Gridley T, Shen J, Kopan R. γ-Secretase functions through Notch signaling to maintain skin appendages but is not required for their patterning or initial morphogenesis. Dev Cell. 2004; 7: 731–43.

84. Habas R, Dawid IB, He X. Coactivation of Rac and Rho by Wnt/Frizzled signaling is required for vertebrate gastrulation. Genes Dev. 2003; 17: 295–309.

85. Bejaoui M, Villareal MO, Isoda H. β-catenin-mediated hair growth induction effect of 3,4,5-tri-O-caffeoylquinic acid. Aging (Albany NY) [Internet]. Impact Journals, LLC; 2019; 11: 4216–37. Available from: https://doi.org/10.18632/aging.102048

86. Le Pape E, Wakamatsu K, Ito S, Wolber R, Hearing VJ. Regulation of eumelanin/pheomelanin synthesis and visible pigmentation in melanocytes by ligands of the melanocortin 1 receptor. Pigment Cell Melanoma Res. 2008; 21: 477–86.

87. Huang DW, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009; 37: 1– 13.

88. Zacariotti RL, do Valle R del R. Observation of mating in the Calico Snake Oxyrhopus petola Linnaeus, 1758. Herpetol Notes. 2010; 3: 139–40.

89. Millar SE. Molecular mechanisms regulating hair follicle development [Internet]. Journal of Investigative Dermatology. Elsevier Masson SAS; 2002. p. 216–25. Available from: http://dx.doi.org/10.1046/j.0022-202x.2001.01670.x

90. Lee S-H, Yoon J, Shin SH, Zahoor M, Kim HJ, Park PJ, Park W-S, Min DS, Kim H-Y, Choi K-Y. Valproic Acid Induces Hair Regeneration in Murine Model and Activates Alkaline Phosphatase Activity in Human Dermal Papilla Cells. PLoS One [Internet]. 2012; 7: e34152. Available from: http://dx.plos.org/10.1371/journal.pone.0034152

91. Sinclair R, Jolley D, Mallari R, Magee J, Tosti A, Piracinni BM, Vincenzi C, Happle R, Ferrando J, Grimalt R, Thérèse L, Van Neste D, Zlotogorski A, et al. Morphological approach to hair disorders. J Investig Dermatology Symp Proc. 2003; 8: 56–64.

92. Steingrímsson E, Copeland NG, Jenkins NA. Melanocyte stem cell maintenance and hair graying. Cell. 2005; 121: 9–12.

93. Driskell RR, Clavel C, Rendl M, Watt FM. Hair follicle dermal papilla cells at a glance. J Cell Sci [Internet]. 2011; 124: 1179–82. Available from: http://jcs.biologists.org/cgi/doi/10.1242/jcs.082446

94. Lin W, Xiang L-J, Shi H-X, Zhang J, Jiang L, Cai P, Lin Z-L, Lin B-B, Huang Y, Zhang H-L, Fu X-B, Guo D-J, Li X-K, et al. Fibroblast Growth Factors Stimulate Hair Growth throughβ-Catenin and Shh Expression in C57BL/6 Mice. Biomed Res Int. 2015; 2015: 1– 9.

95. Lee J, Tumbar T. Hairy tale of signaling in hair follicle development and cycling. Semin Cell Dev Biol [Internet]. Elsevier Ltd; 2012; 23: 906–16. Available from: http://dx.doi.org/10.1016/j.semcdb.2012.08.003

96. Yamamoto N, Tanigaki K, Han H, Hiai H, Honjo T. Notch/RBP-J signaling regulates epidermis/hair fate determination of hair follicular stem cells. Curr Biol. 2003; 13: 333–8.

97. Blanpain C, Lowry WE, Pasolli HA, Fuchs E. Canonical notch signaling functions as a commitment switch in the epidermal lineage. Genes Dev. 2006; 20: 3022–35.

98. Honjo T. The shortest path from the surface to the nucleus: RBP-Jκ/Su(H) transcription factor. Genes to Cells. 1996; 1: 1–9.

99. Dornier E, Coumailleau F, Ottavi JF, Moretti J, Boucheix C, Mauduit P, Schweisguth F, Rubinstein E. Tspanc8 tetraspanins regulate ADAM10/Kuzbanian trafficking and promote Notch activation in flies and mammals. J Cell Biol. 2012; 199: 481–96.

100. Lowry WE, Blanpain C, Nowak JA, Guasch G, Lewis L, Fuchs E. Defining the impact of β-catenin/Tcf transactivation on epithelial stem cells. Genes Dev. 2005; 19: 1596–611.

101. Li J, Baxter RM, Weiner L, Goetinck PF, Calautti E, Brissette JL. Foxn1 promotes keratinocyte differentiation by regulating the activity of protein kinase C. Differentiation [Internet]. International Society of Differentiation; 2007; 75: 694–701. Available from: http://dx.doi.org/10.1111/j.1432-0436.2007.00176.x

102. Takabayashi Y, Nambu M, Ishihara M, Kuwabara M, Fukuda K, Nakamura S, Hattori H, Kiyosawa T. Enhanced effect of fibroblast growth factor‑2‑containing dalteparin/protamine nanoparticles on hair growth. Clin Cosmet Investig Dermatol. 2016; 9: 127–34.

103. Tsai SY, Sennett R, Rezza A, Clavel C, Grisanti L, Zemla R, Najam S, Rendl M. Wnt/β- catenin signaling in dermal condensates is required for hair follicle formation. Dev Biol [Internet]. Elsevier; 2014; 385: 179–88. Available from: http://dx.doi.org/10.1016/j.ydbio.2013.11.023

104. Maretto S, Cordenonsi M, Dupont S, Braghetta P, Broccoli V, Hassan AB, Volpin D, Bressan GM, Piccolo S. Mapping Wnt/ -catenin signaling during mouse development and in colorectal tumors. Proc Natl Acad Sci [Internet]. 2003; 100: 3299–304. Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.0434590100

105. Enshell-Seijffers D, Lindon C, Kashiwagi M, Morgan BA. β-catenin Activity in the Dermal Papilla Regulates Morphogenesis and Regeneration of Hair. Dev Cell [Internet]. Elsevier Ltd; 2010; 18: 633–42. Available from: http://dx.doi.org/10.1016/j.devcel.2010.01.016

106. Yamada T, Hasegawa S, Inoue Y, Date Y, Yamamoto N, Mizutani H, Nakata S, Matsunaga K, Akamatsu H. Wnt/β-Catenin and kit signaling sequentially regulate melanocyte stem cell differentiation in uvb-induced epidermal pigmentation. J Invest Dermatol. Elsevier Masson SAS; 2013; 133: 2753–62.

107. Dorsky RI, Raible DW, Moon RT. Direct regulation of nacre , a zebrafish MITF homolog required for pigment cell formation , by the Wnt pathway service Direct regulation of nacre , a zebrafish MITF homolog required for pigment cell formation , by the Wnt pathway. 2000; : 158–62.

108. Jouneau A, Yu YQ, Pasdar M, Larue L. Plasticity of cadherin-catenin expression in the melanocyte lineage. Pigment Cell Res. 2000; 13: 260–72.

109. Goding CR. Mitf from neural crest to melanoma: Signal transduction and transcription in the melanocyte lineage. Genes Dev. 2000; 14: 1712–28.

110. Villareal MO, Kume S, Neffati M, Isoda H. Upregulation of Mitf by Phenolic Compounds- Rich Cymbopogon schoenanthus Treatment Promotes Melanogenesis in B16 Melanoma Cells and Human Epidermal Melanocytes. Biomed Res Int [Internet]. 2017; 2017: 1–11. Available from: https://www.hindawi.com/journals/bmri/2017/8303671/

111. Moore KJ. Insight into the microphthalmia gene. Trends Genet. 1995; 11: 442–8.

112. Bonaventure J, Domingues MJ, Larue L. Cellular and molecular mechanisms controlling the migration of melanocytes and melanoma cells. Pigment Cell Melanoma Res. 2013; 26: 316–25.

113. Flaherty KT, Hodi FS, Fisher DE. From genes to drugs: targeted strategies for melanoma. Nat Rev Cancer [Internet]. Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.; 2012; 12: 349. Available from: https://doi.org/10.1038/nrc3218

114. Kawakami A, Fisher DE. The master role of microphthalmia-associated transcription factor in melanocyte and melanoma biology. Lab Investig [Internet]. United States & Canadian Academy of Pathology USCAP, Inc; 2017; 97: 649. Available from: https://doi.org/10.1038/labinvest.2017.9

115. Gaggioli C, Buscà R, Abbe P, Ortonne JP, Ballotti R. Microphthalmia-associated transcription factor (MITF) is required but is not sufficient to induce the expression of melanogenic genes. Pigment Cell Res. 2003; 16: 374–82.

116. Shin SY, Choi JH, Jung E, Gil H-N, Lim Y, Lee YH. The EGR1–STAT3 Transcription Factor Axis Regulates α-Melanocyte–Stimulating Hormone–Induced Tyrosinase Gene Transcription in Melanocytes. J Invest Dermatol. 2019; 139: 1616–9.

117. Pingault V, Ente D, Dastot-Le Moal F, Goossens M, Marlin S, Bondurand N. Review and update of mutations causing Waardenburg syndrome. Hum Mutat. 2010; 31: 391–406.

118. Dong L, Li Y, Cao J, Liu F, Pier E, Chen J, Xu Z, Chen C, Wang RA, Cui R. FGF2 regulates melanocytes viability through the STAT3-transactivated PAX3 transcription. Cell Death Differ. 2012; 19: 616–22.

119. Riley PA. Melanin. Int J Biochem Cell Biol [Internet]. 1997; 29: 1235–9. Available from: http://www.sciencedirect.com/science/article/pii/S1357272597000137

120. Fuller BB, Drake MA, Spaulding DT, Chaudhry F. Downregulation of tyrosinase activity in human melanocyte cell cultures by yohimbine. J Invest Dermatol [Internet]. Elsevier Masson SAS; 2000; 114: 268–76. Available from: http://dx.doi.org/10.1046/j.1523- 1747.2000.00860.x

121. Murtas D, Pilloni L, Diana A, Casula L, Tomei S, Piras F, Ferreli C, Maxia C, Perra MT. Tyrosinase and nestin immunohistochemical expression in melanocytic nevi as a histopathologic pattern to trace melanocyte differentiation and nevogenesis. Histochem Cell Biol [Internet]. 2019; 151: 175–85. Available from: https://doi.org/10.1007/s00418-018-1730-5

122. Trempus CS, Morris RJ, Ehinger M, Elmore A, Bortner CD, Ito M, Cotsarelis G, Nijhof JGW, Peckham J, Flagler N, Kissling G, Humble MM, King LC, et al. CD34 expression by hair follicle stem cells is required for skin tumor development in mice. Cancer Res. 2007; 67: 4173–81.

123. Joshi SS, Tandukar B, Pan L, Huang JM, Livak F, Smith BJ, Hodges T, Mahurkar AA, Hornyak TJ. CD34 defines melanocyte stem cell subpopulations with distinct regenerative properties. PLoS Genet. 2019; 15: e1008034.

124. Wu G, Huang H, Abreu JG, He X. Inhibition of GSK3 phosphorylation of β-catenin via phosphorylated PPPSPXS motifs of Wnt coreceptor LRP6. PLoS One. 2009; 4.

125. Zhu W, Shiojima I, Ito Y, Li Z, Ikeda H, Yoshida M, Naito AT, Nishi JI, Ueno H, Umezawa A, Minamino T, Nagai T, Kikuchi A, et al. IGFBP-4 is an inhibitor of canonical Wnt signalling required for cardiogenesis. Nature. 2008; 454: 345–9.

126. Geyfman M, Gordon W, Paus R, Andersen B. Identification of telogen markers underscores that telogen is far from a quiescent hair cycle phase. J Invest Dermatol. 2012; 132: 721–4.

127. Horsley V, Aliprantis AO, Polak L, Glimcher LH, Fuchs E. NFATc1 Balances Quiescence and Proliferation of Skin Stem Cells. Cell. 2008; 132: 299–310.

128. Keyes BE, Segal JP, Heller E, Lien W-H, Chang C-Y, Guo X, Oristian DS, Zheng D, Fuchs E. Nfatc1 orchestrates aging in hair follicle stem cells. Proc Natl Acad Sci [Internet]. 2013; 110: E4950–9. Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.1320301110

129. Avigad Laron E, Aamar E, Enshell-Seijffers D. The Mesenchymal Niche of the Hair Follicle Induces Regeneration by Releasing Primed Progenitors from Inhibitory Effects of Quiescent Stem Cells. Cell Rep [Internet]. ElsevierCompany.; 2018; 24: 909-921.e3. Available from: https://doi.org/10.1016/j.celrep.2018.06.084

130. Chen H, Weng QY, Fisher DE. UV signaling pathways within the skin. J Invest Dermatol [Internet]. Elsevier Masson SAS; 2014; 134: 2080–5. Available from: http://dx.doi.org/10.1038/jid.2014.161

131. Yang Y, Jang G, Yang X, Wang Q, He S, Li S, Quach C, Zhao S, Li F, Yuan Z, Lee H-R, Zhong H, Liang C. Central role of autophagic UVRAG in melanogenesis and the suntan response. Proc Natl Acad Sci. 2018; 115: E7728–37.

132. Stout R, Birch-Machin M. Mitochondria’s Role in Skin Ageing. Biology (Basel). 2019; 8: 29.

133. Tobin DJ. The cell biology of human hair follicle pigmentation. Pigment Cell Melanoma Res [Internet]. 2011; 24: 75–88. Available from: http://doi.wiley.com/10.1111/j.1755- 148X.2010.00803.x

134. Paus R, Cotsarelis G. T He B Iology of H Air F Ollicles. New Engl Jouranal Med. 1999; 341: 491–7.

135. Rosenquist TA, Martin GR. Fibroblast growth factor signalling in the hair growth cycle: expression of the fibroblast growth factor receptor and ligand genes in the murine hair follicle. Dev Dyn [Internet]. 1996; 205: 379–86. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8901049

136. Cotsarelis G, Sun TT, Lavker RM. Label-retaining cells reside in the bulge area of pilosebaceous unit: Implications for follicular stem cells, hair cycle, and skin carcinogenesis. Cell. 1990; 61: 1329–37.

137. Sano S, Kira M, Takagi S, Yoshikawa K, Takeda J, Itami S. Correction for Sano et al., Two distinct signaling pathways in hair cycle induction: Stat3-dependent and -independent pathways. Proc Natl Acad Sci [Internet]. 2015; 112: 201506638. Available from: http://www.pnas.org/lookup/doi/10.1073/pnas.1506638112

138. Jahoda CAB, Horne KA, Oliver RF. Induction of hair growth by implantation of cultured dermal papilla cells. Nature. 1984; 311: 560–2.

139. Kulak O, Chen H, Holohan B, Wu X, He H, Borek D, Otwinowski Z, Yamaguchi K, Garofalo LA, Ma Z, Wright W, Chen C, Shay JW, et al. Disruption of Wnt/β-Catenin Signaling and Telomeric Shortening Are Inextricable Consequences of Tankyrase Inhibition in Human Cells. Mol Cell Biol. 2015; 35: 2425–35.

140. Fuchs E, Merrill BJ, Jamora C, Dasgupta R. At the Roots of a Never-Ending Cycle. Dev Cell. 2001; 1: 13–25.

141. Adachi K, Watanabe Y, Inouye K. Activity of glucose-6-phosphate 1-dehydrogenase in hair follicles with male-pattern alopecia. Biosci Biotechnol Biochem. 1999; 63.

142. Zhang Y, Andl T, Yang SH, Teta M, Liu F, Seykora JT, Tobias JW, Piccolo S, Schmidt- Ullrich R, Nagy A, Taketo MM, Dlugosz AA, Millar SE. Activation of β-catenin signaling programs embryonic epidermis to hair follicle fate. Development. 2008; 135: 2161–72.

143. McGonigle S, Chen Z, Wu J, Chang P, Kolber-Simonds D, Ackermann K, Twine NC, Shie J, Miu JT, Huang K-C, Moniz GA, Nomoto K. E7449: A dual inhibitor of PARP1/2 and tankyrase1/2 inhibits growth of DNA repair deficient tumors and antagonizes Wnt signaling. Oncotarget. 2015; 6.

144. Li C, Zheng X, Han Y, Lv Y, Lan F, Zhao J. XAV939 inhibits the proliferation and migration of lung adenocarcinoma A549 cells through the WNT pathway. Oncol Lett. 2018; 15: 8973–82.

145. Slominski a, Paus R. Melanogenesis is coupled to murine anagen: toward new concepts for the role of melanocytes and the regulation of melanogenesis in hair growth. J Invest Dermatol. 1993; 101: 90S-97S.

146. Tobin DJ, Hordinsky M, Bernard BA. Hair pigmentation: a research update. J Investig Dermatol Symp Proc [Internet]. Elsevier Masson SAS; 2005; 10: 275–9. Available from: http://dx.doi.org/10.1111/j.1087-0024.2005.10117.x

147. Slominski A. Melanin Pigmentation in Mammalian Skin and Its Hormonal Regulation. Physiol Rev [Internet]. 2004; 84: 1155–228. Available from: http://physrev.physiology.org/cgi/doi/10.1152/physrev.00044.2003

148. Seiberg M, Marthinuss J, Stenn KS. Changes in expression of apoptosis-associated genes in skin mark early catagen. J Invest Dermatol [Internet]. Elsevier Masson SAS; 1995; 104: 78–82. Available from: http://dx.doi.org/10.1111/1523-1747.ep12613555

149. Park HY, Kosmadaki M, Yaar M, Gilchrest BA. Cellular mechanisms regulating human melanogenesis. Cell Mol Life Sci. 2009; 66: 1493–506.

150. Schiaffino MV. Signaling pathways in melanosome biogenesis and pathology. Int J Biochem Cell Biol [Internet]. Elsevier Ltd; 2010; 42: 1094–104. Available from: http://dx.doi.org/10.1016/j.biocel.2010.03.023

151. Rouzaud F, Kadekaro AL, Abdel-Malek ZA, Hearing VJ. MC1R and the response of melanocytes to ultraviolet radiation. Mutat Res - Fundam Mol Mech Mutagen. 2005; 571: 133–52.

152. Tsatmali M, Ancans J, Thody AJ. Melanocyte function and its control by melanocortin peptides. J Histochem Cytochem. 2002; 50: 125–33.

153. Hosoi J, Abe E, Suda T, Kuroki T. Regulation of Melanin Synthesis of B16 Mouse Melanoma Cells by 1α, 25-Dihydroxyvitamin D3 and Retinoic Acid. Cancer Res. 1985; 45: 1474–8.

154. Costin GE, Hearing VJ. Human skin pigmentation: Melanocytes modulate skin color in response to stress. FASEB J. 2007; 21: 976–94.

155. Slominski A, Zmijewski MA, Pawelek J. L-tyrosine and L-dihydroxyphenylalanine as hormone-like regulators of melanocyte functions. Pigment Cell Melanoma Res. 2012; 25: 14–27.

156. Slominski A, Moellmann G, Kuklinska E, Bomirski A, Pawelek J. Positive regulation of melanin pigmentation by two key substrates of the melanogenic pathway, L-tyrosine and L-dopa. J Cell Sci. 1988; 89: 287–96.

157. Dunn KJ, Brady M, Ochsenbauer-Jambor C, Snyder S, Incao A, Pavan WJ. WNT1 and WNT3a promote expansion of melanocytes through distinct modes of action. Pigment Cell Res. 2005; 18: 167–80.

158. Jin EJ, Erickson CA, Takada S, Burrus LW. Wnt and BMP signaling govern lineage segregation of melanocytes in the avian embryo. Dev Biol. 2001; 233: 22–37.

159. Takeda K, Yasumoto KI, Takada R, Takada S, Watanabe KI, Udono T, Saito H, Takahashi K, Shibahara S. Induction of melanocyte-specific microphthalmia-associated transcription factor by Wnt-3a. J Biol Chem. 2000; 275: 14013–6.

160. Kang J Il, Kim MK, Lee JH, Jeon YJ, Hwang EK, Koh YS, Hyun JW, Kwon SY, Yoo ES, Kang HK. Undariopsis peterseniana promotes hair growth by the activation of Wnt/β- catenin and ERK pathways. Mar Drugs. 2017; 15.

161. Habas R, He X. Activation of Rho and Rac by Wnt/Frizzled signaling. Methods Enzymol. 2006; 406: 500–11.

162. Kollmar R. Who does the hair cell’s ’do? Rho GTPases and hair-bundle morphogenesis. Current Opinion in Neurobiology. 1999. p. 394–8.

163. Kalinec F, Zhang M, Urrutia R, Kalinec G. Rho GTPases mediate the regulation of cochlear outer hair cell motility by acetylcholine. J Biol Chem. 2000; 275: 28000–5.

164. Sutera M, De Caro V, Giannola L. Small endogenous molecules as moiety to improve targeting of CNS drugs motility. Exp. Op. Drug Delivery 2017; 14(1), 93-107

参考文献をもっと見る