Ligustroside derived from Ligustrum japonicum inhibits melanogenesis via blocking the MAPK and PKA/CREB signaling pathways

Xian-Rong Zhou , Fatih Karadeniz , Jung Hwan Oh , Youngwan Seo , Chang-Suk Kong

Asian Pacific Journal of Tropical Biomedicine ›› 2025, Vol. 15 ›› Issue (2) : 65 -74.

PDF (1336KB)
Asian Pacific Journal of Tropical Biomedicine ›› 2025, Vol. 15 ›› Issue (2) : 65 -74. DOI: 10.4103/apjtb.apjtb_568_24
Original Article

Ligustroside derived from Ligustrum japonicum inhibits melanogenesis via blocking the MAPK and PKA/CREB signaling pathways

Author information +
History +
PDF (1336KB)

Abstract

Objective: To investigate the anti-melanogenic potential of ligustroside isolated from Ligustrum japonicum.

Methods: The cytotoxicity of ligustroside was tested via MTT assay. Furthermore, the effects of ligustroside on the expression of critical melanogenic markers such as tyrosinase, tyrosinase related proteins (TRPs), and microphthalmia-associated transcription factor (MITF) were analyzed at both mRNA and protein levels via RT-qPCR and Western blot, respectively, in α-melanocyte stimulating hormone-induced B16F10 cells. In addition, phosphorylation of p38, ERK and JNK proteins was investigated. Immunofluorescence analysis of MITF was also conducted.

Results: Ligustroside significantly reduced intracellular tyrosinase activity and melanin content by 37.11% and 29.12%, respectively, compared to untreated cells. Moreover, it downregulated the expression of MITF, tyrosinase, TRP-1, and TRP-2 at the mRNA and protein levels by regulating both the mitogen-activated protein kinase (MAPK) and protein kinase A (PKA)/cAMP response element-binding protein (CREB) signaling pathways. Ligustroside also suppressed the nuclear protein expression of MITF, β-catenin, and p-CREB, and decreased immunofluorescence intensity of nuclear MITF.

Conclusions: Ligustroside derived from Ligustrum japonicum shows a significant anti-melanogenesis effect via suppression of the MAPK and PKA/CREB signaling pathways.

Keywords

Ligustrum japonicum / Ligustroside / MAPK / Melanogenesis / CREB / PKA

Cite this article

Download citation ▾
Xian-Rong Zhou, Fatih Karadeniz, Jung Hwan Oh, Youngwan Seo, Chang-Suk Kong. Ligustroside derived from Ligustrum japonicum inhibits melanogenesis via blocking the MAPK and PKA/CREB signaling pathways. Asian Pacific Journal of Tropical Biomedicine, 2025, 15(2): 65-74 DOI:10.4103/apjtb.apjtb_568_24

登录浏览全文

4963

注册一个新账户 忘记密码

Conflict of interest statement

The authors declare that they have no conflict of interest.

Funding

This research was supported by the BB21plus funded by Busan Metropolitan City and Busan Techno Park, and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. NRF-2023R1A2C1006268 and RS-2023-00212560).

Data availability statement

The data supporting the findings of this study are available from the corresponding authors upon request.

Authors’ contributions

XRZ and CSK conceived the idea and designed the study. XRZ, and JHO performed the experiments, acquired the data and validated the results. XRZ and FK drafted the original manuscript and XRZ, FK and YS contributed to the revisions. JHO and YS performed the extraction and fractionation. JHO, FK, and CSK performed the statistical analysis, data curation, and validation and visualization of the results. YS and CSK supervised the project.

Publisher’s note

The Publisher of the Journal remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

[1]

Langat MK, Dlova NC, Mulcahy-Ryan LE, Schwikkard SL, Opara EI, Crouch NR, et al. The effect of isolates from Cassipourea flanaganii (Schinz) alston, a plant used as a skin lightning agent, on melanin production and tyrosinase inhibition. J Ethnopharmacol 2021; 264. doi: 10.1016/j.jep.2020.113272.

[2]

Bin BH, Joo Y, Lee AY, Shin S, Cho EG, Lee T. Novel inhibitory effect of N-(2-Hydroxycyclohexyl)valiolamine on melanin production in a human skin model. Int J Mol Sci 2014; 15(7): 12188-12195.

[3]

Zonios G, Dimou A, Bassukas I, Galaris D, Tsolakidis A, Kaxiras E. Melanin absorption spectroscopy: New method for noninvasive skin investigation and melanoma detection. J Biomed Opt 2008; 13(1). doi: 10.1117/1.2844710.

[4]

Song EC, Park C, Shin Y, Kim WK, Kim SB, Cho S. Neurog1-derived peptides RMNE1 and DualPep-Shine penetrate the skin and inhibit melanin synthesis by regulating MITF transcription. Int J Mol Sci 2023; 4(7). doi: 10.3390/ijms24076158.

[5]

Lee JY, Cho YR, Park JH, Ahn EK, Jeong W, Shin HS, et al. Anti-melanogenic and anti-oxidant activities of ethanol extract of Kummerowia striata: Kummerowia striata regulate anti-melanogenic activity through down-regulation of TRP-1, TRP-2 and MITF expression. Toxicol Rep 2018; 6: 10-17.

[6]

Kim SS, Kim MJ, Choi YH, Kim BK, Kim KS, Park KJ, et al. Down-regulation of tyrosinase, TRP-1, TRP-2 and MITF expressions by citrus press-cakes in murine B16 F10 melanoma. Asian Pac J Trop Biomed 2013; 3(8): 617-622.

[7]

Pathria G, Garg B, Borgdorff V, Garg K, Wagner C, Superti-Furga G, et al. Overcoming MITF-conferred drug resistance through dual AURKA/ MAPK targeting in human melanoma cells. Cell Death Dis 2016; 7(3): e2135. doi: 10.1038/cddis.2015.369.

[8]

Kim T, Hyun CG. Imperatorin positively regulates melanogenesis through signaling pathways involving PKA/CREB, ERK, AKT, and GSK3β/β-catenin. Molecules 2022; 27(19). doi: 10.3390/molecules27196512.

[9]

Merecz-Sadowska A, Sitarek P, Kowalczyk T, Zajdel K, Kucharska E, Zajdel R. The modulation of melanogenesis in B16 cells upon treatment with plant extracts and isolated plant compounds. Molecules 2022; 27(14). doi: 10.3390/molecules27144360.

[10]

Choi MH, Yang SH, Park WK, Shin HJ. Bamboo lignin fractions with in vitro tyrosinase inhibition activity downregulate melanogenesis in B16F10 cells via PKA/CREB signaling pathway. Int J Mol Sci 2022; 23(13). doi: 10.3390/ijms23137462.

[11]

Bu J, Ma PC, Chen ZQ, Zhou WQ, Fu YJ, Li LJ, et al. Inhibition of MITF and tyrosinase by paeonol-stimulated JNK/SAPK to reduction of phosphorylated CREB. Am J Chin Med 2008; 36(2): 245-263.

[12]

Kim K, Jung KE, Shin Y, Kim C, Yoon T. Sorafenib induces pigmentation via the regulation of β-catenin signalling pathway in melanoma cells. Exp Dermatol 2022; 31(1): 57-63.

[13]

Zhai Y, Xu J, Feng L, Liu Q, Yao W, Li H, et al. Broad range metabolomics coupled with network analysis for explaining possible mechanisms of Er-Zhi-Wan in treating liver-kidney Yin deficiency syndrome of Traditional Chinese medicine. J Ethnopharmacol 2019; 234: 57-66.

[14]

Kim JE, Kim SH, Kim MA, Ko MS, Shin CS, Lee NH. Antiinflammatory and anti-oxidative activities for extract of fermented Ligustrum japonicum fruits. J Soc Cosmet Sci Korea 2023; 49(2): 117-125.

[15]

Oh JH, Karadeniz F, Lee JI, Seo Y, Kong CS. Ligustrum japonicum Thunb. fruits exert antiosteoporotic properties in bone marrow-derived mesenchymal stromal cells via regulation of adipocyte and osteoblast differentiation. Stem Cells Int 2021; 2021(1). doi: 10.1155/2021/8851884.

[16]

Li M, Wang X, Han L, Jia L, Liu E, Li Z, et al. Integration of multicomponent characterization, untargeted metabolomics and mass spectrometry imaging to unveil the holistic chemical transformations and key markers associated with wine steaming of Ligustri lucidi Fructus. J Chromatogr A 2020; 1624. doi: 10.1016/j.chroma.2020.461228.

[17]

Grewal R, Reutzel M, Dilberger B, Hein H, Zotzel J, Marx S, et al. Purified oleocanthal and ligstroside protect against mitochondrial dysfunction in models of early Alzheimer’s disease and brain ageing. Exp Neurol 2020; 328. doi: 10.1016/j.expneurol.2020.113248.

[18]

Castejón ML, Montoya T, Alarcón-De-La-Lastra C, Sánchez-Hidalgo M. Potential protective role exerted by secoiridoids from Olea europaea L. in cancer, cardiovascular, neurodegenerative, aging-related, and immunoinflammatory diseases. Antioxidants 2020; 9. doi: 10.3390/antiox9020149.

[19]

Peng Z, He J, Cheng Y, Xu J, Zhang W. Biologically active secoiridoids: A comprehensive update. Med Res Rev 2023; 43(4): 1201-1252.

[20]

Angelis A, Mavros P, Nikolaou PE, Mitakou S, Halabalaki M, Skaltsounis L. Phytochemical analysis of olive flowers’ hydroalcoholic extract and in vitro evaluation of tyrosinase, elastase and collagenase inhibition activity. Fitoterapia 2020; 143. doi: 10.1016/j.fitote.2020.104602.

[21]

Kumar A, Kaur S, Sangwan PL, Tasduq SA. Therapeutic and cosmeceutical role of glycosylated natural products in dermatology. Phytother Res 2023; 37(4): 1574-1589.

[22]

Chen YM, Su WC, Li C, Shi Y, Chen QX, Zheng J, et al. Anti-melanogenesis of novel kojic acid derivatives in B16F10 cells and zebrafish. Int J Biol Macromol 2019; 123: 723-731.

[23]

Kim H, Kong CS, Seo Y. Salidroside, 8(E)-Nuezhenide, and ligustroside from Ligustrum japonicum Fructus inhibit expressions of MMP-2 and -9 in HT 1080 fibrosarcoma. Int J Mol Sci 2022; 23. doi: 10.3390/ijms23052660.

[24]

Thu NT, Linh LH, Hoang DA, Oanh NT, Thao VM, Hang NT, et al. Neolignan glycoside and other constituents from the leaves of Ligustrum sinense and their anti-inflammatory activity. Nat Prod Commun 2024; 19(1). doi: 10.1177/1934578x241226825.

[25]

Papakonstantinou A, Koumarianou P, Diamantakos P, Melliou E, Magiatis P, Boleti H. A systematic ex-vivo study of the anti-proliferative/cytotoxic bioactivity of major olive secoiridoids’ double combinations and of total olive oil phenolic extracts on multiple cell-culture based cancer models highlights synergistic effects. Nutrients 2023; 15. doi: 10.3390/nu15112538.

[26]

Gu J, Wang B, Wang T, Zhang N, Liu H, Gui J, et al. Effects of cartilage progenitor cells, bone marrow mesenchymal stem cells and chondrocytes on cartilage repair as seed cells: An in vitro study. Drug Des Devel Ther 2022; 16: 1217-1230.

[27]

Zhou XR, Oh JH, Karadeniz F, Lee H, Kim HE, Jo M, et al. Inhibitory effect of Cyrtomium falcatum on melanogenesis in α-MSH-stimulated B16F 10 murine melanoma cells. Asian Pac J Trop Biomed 2023; 13(9): 403-410.

[28]

Kim CS, Noh SG, Park Y, Kang D, Chun P, Chung HY, et al. A potent tyrosinase inhibitor, (E)-3-(2,4-dihydroxyphenyl)-1-(thiophen-2-yl)prop-2-en-1-one, with anti-melanogenesis properties in α-MSH and IBMX-induced B16F10 melanoma cells. Molecules 2018; 23. doi: 10.3390/molecules23102725.

[29]

Ha SY, Jung JY, Yang JK. Camellia japonica essential oil inhibits α-MSH-induced melanin production and tyrosinase activity in B16F10 melanoma cells. Evid Based Complement Alternat Med 2021; 2021. doi: 10.1155/2021/6328767.

[30]

Goenka S, Johnson F, Simon SR. Novel chemically modified curcumin (CMC) derivatives inhibit tyrosinase activity and melanin synthesis in B16F10 mouse melanoma cells. Biomolecules 2021; 11. doi: 10.3390/biom11050674.

[31]

Zhu YJ, Qiu LJ, Zhou JJ, Guo HY, Hu YH, Li ZC, et al. Inhibitory effects of hinokitiol on tyrosinase activity and melanin biosynthesis and its antimicrobial activities. J Enzyme Inhib Med Chem 2010; 25: 798-803.

[32]

Hushcha Y, Blo I, Oton-Gonzalez L, Di Mauro G, Martini F, Tognon M, et al. microRNAs in the regulation of melanogenesis. Int J Mol Sci 2021; 22. doi: 10.3390/ijms22116104.

[33]

Chen YC, Liu YY, Chen L, Tang DM, Zhao Y, Luo XD. Antimelanogenic effect of isoquinoline alkaloids from Plumula Nelumbinis. J Agric Food Chem 2023; 71: 16090-16101.

[34]

Serre C, Busuttil V, Botto JM. Intrinsic and extrinsic regulation of human skin melanogenesis and pigmentation. Int J Cosmet Sci 2018; 40: 328347.

[35]

Kang M, Park SH, Park SJ, Oh SW, Yoo JA, Kwon K, et al. p44/ 42 MAPK signaling is a prime target activated by phenylethyl resorcinol in its anti-melanogenic action. Phytomedicine 2019; 58. doi: 10.1016/j.phymed.2019.152877.

[36]

Choi H, Yoon JH, Youn K, Jun M. Decursin prevents melanogenesis by suppressing MITF expression through the regulation of PKA/CREB, MAPKs, and PI3K/Akt/GSK-3β cascades. Biomed Pharmacother 2022; 147. doi: 10.1016/j.biopha.2022.112651.

[37]

Ngeow KC, Friedrichsen HJ, Li L, Zeng Z, Andrews S, Volpon L, et al. BRAF/MAPK and GSK3 signaling converges to control MITF nuclear export. Proc Natl Acad Sci U S A 2018; 115: E8668-E8677.

[38]

Kim EH, Jeong HS, Yun HY, Baek KJ, Kwon NS, Park KC, et al. Geranylgeranylacetone inhibits melanin synthesis via ERK activation in Mel-Ab cells. Life Sci 2013; 93: 226-232.

[39]

Karunarathne WAHM, Molagoda IMN, Kim MS, Choi YH, Oren M, Park EK, et al. Flumequine-mediated upregulation of p38 MAPK and JNK results in melanogenesis in B16F10 cells and zebrafish larvae. Biomolecules 2019; 9. doi: 10.3390/biom9100596.

[40]

Liu J, Xu X, Zhou J, Sun G, Li Z, Zhai L, et al. Phenolic acids in Panax ginseng inhibit melanin production through bidirectional regulation of melanin synthase transcription via different signaling pathways. J Ginseng Res 2023; 47: 714-725.

[41]

Gao R, Zhang X, Zou K, Meng D, Lv J. Cryptochrome 1 activation inhibits melanogenesis and melanosome transport through negative regulation of cAMP/PKA/CREB signaling pathway. Front Pharmacol 2023; 14. doi: 10.3389/fphar.2023.1081030.

[42]

Castellano-Pellicena I, Morrison CG, Bell M, O’Connor C, Tobin DJ. Melanin distribution in human skin: Influence of cytoskeletal, polarity, and centrosome-related machinery of stratum basale keratinocytes. Int J Mol Sci 2021; 22. doi: 10.3390/ijms22063143.

[43]

Su TR, Lin JJ, Tsai CC, Huang TK, Yang ZY, Wu MO, et al. Inhibition of melanogenesis by gallic acid: Possible involvement of the PI3K/Akt, MEK/ERK and Wnt/β-catenin signaling pathways in B16F10 cells. Int J Mol Sci 2013; 14(10): 20443-20458.

[44]

Mamat N, Dou J, Lu X, Eblimit A, Akber AH. Isochlorogenic acid A promotes melanin synthesis in B16 cell through the β-catenin signal pathway. Acta Biochim Biophys Sin 2017; 49(9): 800-807.

[45]

Taira N, Katsuyama Y, Yoshioka M, Muraoka O, Morikawa T. Structural requirements of alkylglyceryl-L-ascorbic acid derivatives for melanogenesis inhibitory activity. Int J Mol Sci 2018; 19(4). doi: 10.3390/ijms19041144.

[46]

Gryn-Rynko A, Sperkowska B, Majewski MS. Screening and structure-activity relationship for selective and potent anti-melanogenesis agents derived from species of mulberry (Genus Moms). Molecules 2022; 27(24). doi: 10.3390/molecules27249011.

AI Summary AI Mindmap
PDF (1336KB)

380

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/