Natural Products and Bioprospecting    2023, Vol. 13 Issue (6) : 54-54     DOI: 10.1007/s13659-023-00421-z
ORIGINAL ARTICLES |
Natural product rhynchophylline prevents stress-induced hair graying by preserving melanocyte stem cells via the β2 adrenergic pathway suppression
Xinxin Li1,2,3, Runlu Shi4, Lingchen Yan1, Weiwei Chu1,2, Ruishuang Sun5, Binkai Zheng1, Shuai Wang1,6, Hui Tan3, Xusheng Wang1, Ying Gao1,2,7
1. School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China;
2. Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China;
3. Center for Child Care and Mental Health, Shenzhen Children's Hospital Affiliated to Shantou University Medical College, Shenzhen, 518026, China;
4. Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China;
5. Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China;
6. The Yonghe Medical Beauty Clinic Department, Guangzhou, 510630, China;
7. Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, 528000, China
Download: PDF(12896 KB)   HTML ()  
Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks    
Abstract  Norepinephrine (NA), a stress hormone, can accelerate hair graying by binding to β2 adrenergic receptors (β2AR) on melanocyte stem cells (McSCs). From this, NA-β2AR axis could be a potential target for preventing the stress effect. However, identifying selective blockers for β2AR has been a key challenge. Therefore, in this study, advanced computer-aided drug design (CADD) techniques were harnessed to screen natural molecules, leading to the discovery of rhynchophylline as a promising compound. Rhynchophylline exhibited strong and stable binding within the active site of β2AR, as verified by molecular docking and dynamic simulation assays. When administered to cells, rhynchophylline effectively inhibited NA-β2AR signaling. This intervention resulted in a significant reduction of hair graying in a stress-induced mouse model, from 28.5% to 8.2%. To gain a deeper understanding of the underlying mechanisms, transcriptome sequencing was employed, which revealed that NA might disrupt melanogenesis by affecting intracellular calcium balance and promoting cell apoptosis. Importantly, rhynchophylline acted as a potent inhibitor of these downstream pathways. In conclusion, the study demonstrated that rhynchophylline has the potential to mitigate the negative impact of NA on melanogenesis by targeting β2AR, thus offering a promising solution for preventing stress-induced hair graying.
Keywords Natural product      Rhynchophylline      Stress      Hair graying      β2 adrenergic signaling     
Fund:This work was supported by the Shenzhen Science and Technology Innovation Committee (grant numbers JCYJ20200109142444449, JCYJ20210324120007021), the National Natural Science Foundation of China (grant numbers 31801196), Basic and Applied Basic Research Foundation of Guangdong Province (grant numbers 2022A1515110645).
Corresponding Authors: Hui Tan,E-mail:huitan@email.szu.edu.cn;Xusheng Wang,E-mail:wangxsh27@mail.sysu.edu.cn;Ying Gao,E-mail:gying1008@126.com     E-mail: huitan@email.szu.edu.cn;wangxsh27@mail.sysu.edu.cn;gying1008@126.com
Issue Date: 26 December 2023
Service
E-mail this article
E-mail Alert
RSS
Articles by authors
Xinxin Li
Runlu Shi
Lingchen Yan
Weiwei Chu
Ruishuang Sun
Binkai Zheng
Shuai Wang
Hui Tan
Xusheng Wang
Ying Gao
Trendmd:   
Cite this article:   
Xinxin Li,Runlu Shi,Lingchen Yan, et al. Natural product rhynchophylline prevents stress-induced hair graying by preserving melanocyte stem cells via the β2 adrenergic pathway suppression[J]. Natural Products and Bioprospecting, 2023, 13(6): 54-54.
URL:  
http://npb.kib.ac.cn/EN/10.1007/s13659-023-00421-z     OR     http://npb.kib.ac.cn/EN/Y2023/V13/I6/54
[1] O’Sullivan JDB, Nicu C, Picard M, Chéret J, et al. The biology of human hair greying. Biol Rev Camb Philos Soc. 2021. https://doi.org/10.1111/brv.12648.
[2] Tobin DJ. Age-related hair pigment loss. Curr Probl Dermatol. 2015. https://doi.org/10.1159/000369413.
[3] Paus R. A neuroendocrinological perspective on human hair follicle pigmentation. Pigment Cell Melanoma Res. 2011. https://doi.org/10.1111/j.1755-148X.
[4] Yale K, Juhasz M, Atanaskova MN. Medication-induced repigmentation of gray hair: a systematic review. Skin Appendage Disord. 2020. https://doi.org/10.1159/000504414.
[5] Li RQ, Zhao XH, Zhu Q, Liu T, Hondermarck H, Thorne RF, et al. Exploring neurotransmitters and their receptors for breast cancer prevention and treatment. Theranostics. 2023. https://doi.org/10.7150/thno.81403.
[6] Zhang B, Ma S, Rachmin I, He M, Baral P, Choi S, et al. Hyperactivation of sympathetic nerves drives depletion of melanocyte stem cells. Nature. 2020. https://doi.org/10.1038/s41586-020-1935-3.
[7] Ma X, Hu Y, Batebi H, Heng J, et al. Analysis of β2AR-Gs and β2AR-Gi complex formation by NMR spectroscopy. Proc Natl Acad Sci U S A. 2020. https://doi.org/10.1073/pnas.2009786117.
[8] Bai C, Wang J, Mondal D, Du Y, et al. Exploring the activation process of the β2AR-Gs complex. J Am Chem Soc. 2021. https://doi.org/10.1021/jacs.1c03696.
[9] Ferrara N, Komici K, Corbi G, Pagano G, Furgi G, Rengo C, et al. β-adrenergic receptor responsiveness in aging heart and clinical implications. Front Physiol. 2014. https://doi.org/10.3389/fphys.2013.00396.
[10] Kamiar A, Yousefi K, Dunkley JC, Webster KA, Shehadeh LA. β2-Adrenergic receptor agonism as a therapeutic strategy for kidney disease. Am J Physiol Regul Integr Comp Physiol. 2021. https://doi.org/10.1152/ajpregu.00287.2020.
[11] Rambacher KM, Moniri NH. The β2-adrenergic receptor-ROS signaling axis: an overlooked component of β2AR function? Biochem Pharmacol. 2020. https://doi.org/10.1016/j.bcp.2019.113690.
[12] Abosamak NR, Shahin MH. Beta 2 Receptor Agonists/Antagonists. StatPearls. Treasure Island (FL): StatPearls Publishing. 2023.
[13] Léauté-Labrèze C, Hoeger P, Mazereeuw-Hautier J, Guibaud L, Baselga E, Posiunas G, et al. A randomized, controlled trial of oral propranolol in infantile hemangioma. N engl J Med. 2015. https://doi.org/10.1056/NEJMoa1404710.
[14] Popp DA, Tse TF, Shah SD, Clutter WE, Cryer PE. Oral propranolol and metoprolol both impair glucose recovery from insulin-induced hypoglycemia in insulin-dependent diabetes mellitus. Diabetes Care. 1984. https://doi.org/10.2337/diacare.7.3.243.
[15] Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des. 2011. https://doi.org/10.2174/157340911795677602.
[16] Khan S, Farooq U, Kurnikova M. Exploring protein stability by comparative molecular dynamics simulations of homologous hyperthermophilic, mesophilic, and psychrophilic proteins. J Chem Inf Model. 2016. https://doi.org/10.1021/acs.jcim.6b00305.
[17] Liu K, Watanabe E, Kokubo H. Exploring the stability of ligand binding modes to proteins by molecular dynamics simulations. J Comput Aided Mol Des. 2017. https://doi.org/10.1007/s10822-016-0005-2.
[18] Salmaso V, Moro S. Bridging molecular docking to molecular dynamics in exploring ligand-protein recognition process: an overview. Front Pharmacol. 2018. https://doi.org/10.3389/fphar.2018.00923.
[19] Zhang D, Lazim R. Application of conventional molecular dynamics simulation in evaluating the stability of apomyoglobin in urea solution. Sci Rep. 2017. https://doi.org/10.1038/srep44651.
[20] Fage CD, Lathouwers T, Vanmeert M, Gao LJ, Vrancken K, Lammens EM, et al. The kalimantacin polyketide antibiotics inhibit fatty acid biosynthesis in staphylococcus aureus by targeting the enoyl-acyl carrier protein binding site of FabI. Angew Chem Int Ed Engl. 2020. https://doi.org/10.1002/anie.201915407.
[21] Horiguchi T, Hayashi K, Tsubotani S, Iinuma S, Harada S, Tanida S. New naphthacenecarboxamide antibiotics, TAN-1518 A and B, have inhibitory activity against mammalian DNA topoisomerase I. J Antibiot (Tokyo). 1994. https://doi.org/10.7164/antibiotics.47.545.
[22] Silvers MA, Pakhomova S, Neau DB, Silvers WC, Anzalone N, Taylor CM, et al. Crystal structure of carboxyltransferase from staphylococcus aureus bound to the antibacterial agent Moiramide B. Biochemistry. 2016. https://doi.org/10.1021/acs.biochem.6b00641.
[23] Nakajima H, Hori Y, Terano H, Okuhara M, Manda T, Matsumoto S, et al. New antitumor substances, FR901463, FR901464 and FR901465. II. Activities against experimental tumors in mice and mechanism of action. J Antibiot (Tokyo). 1996. https://doi.org/10.7164/antibiotics.49.1204.
[24] Cao W, Wang Y, Lv X, Yu X, Li X, Li H, et al. Rhynchophylline prevents cardiac dysfunction and improves survival in lipopolysaccharide-challenged mice via suppressing macrophage I-κBα phosphorylation. Int Immunopharmacol. 2012. https://doi.org/10.1016/j.intimp.2012.07.010.
[25] Fu WY, Hung KW, Lau SF, Butt B, Yuen VW, Fu G, et al. Rhynchophylline administration ameliorates amyloid-β pathology and inflammation in an Alzheimer’s disease transgenic mouse model. ACS Chem Neurosci. 2021. https://doi.org/10.1021/acschemneuro.1c00600.
[26] Yao C-L, Lin Y-M, Mohamed MS, Chen J-H. Inhibitory effect of ectoine on melanogenesis in B16-F0 and A2058 melanoma cell lines. Biochem Eng J. 2013. https://doi.org/10.1016/j.bej.2013.01.005.
[27] Bartosova L, Bajgar J. Transdermal drug delivery in vitro using diffusion cells. Curr Med Chem. 2012. https://doi.org/10.2174/092986712803306358.
[28] Wu JY, Li YJ, Liu TT, Ou G, Hu XB, Tang TT, et al. Microemulsions vs chitosan derivative-coated microemulsions for dermal delivery of 8-methoxypsoralen. Int J Nanomedicine. 2019. https://doi.org/10.2147/ijn.S191940.
[29] Feske S, Wulff H, Skolnik EY. Ion channels in innate and adaptive immunity. Annu Rev Immunol. 2015. https://doi.org/10.1146/annurev-immunol-032414-112212.
[30] Yu Y, Chang L, Hu Q, Zhu J, Zhang J, Xia Q, et al. P2rx1 deficiency alleviates acetaminophen-induced acute liver failure by regulating the STING signaling pathway. Cell Biol Toxicol. 2023. https://doi.org/10.1007/s10565-023-09800-1.
[31] Zhou J, Zhou S. Antihypertensive and neuroprotective activities of rhynchophylline: the role of rhynchophylline in neurotransmission and ion channel activity. J Ethnopharmacol. 2010. https://doi.org/10.1016/j.jep.2010.08.041.
[32] Ostojic J, Yoon YS, Sonntag T, Nguyen B, Vaughan JM, Shokhirev M, et al. Transcriptional co-activator regulates melanocyte differentiation and oncogenesis by integrating cAMP and MAPK/ERK pathways. Cell Rep. 2021. https://doi.org/10.1016/j.celrep.2021.109136.
[33] 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. https://doi.org/10.1016/j.lfs.2016.08.015.
[34] Arora N, Siddiqui EM, Mehan S. Involvement of adenylate cyclase/cAMP/CREB and SOX9/MITF in melanogenesis to prevent vitiligo. Mol Cell Biochem. 2021. https://doi.org/10.1007/s11010-020-04000-5.
[35] Wang J, Gong J, Wang Q, Tang T, Li W. VDAC1 negatively regulates melanogenesis through the Ca2+-calcineurin-CRTC1-MITF pathway. Life Sci Alliance. 2022. https://doi.org/10.26508/lsa.202101350.
[36] Motiani RK, Tanwar J, Raja DA, Vashisht A, Khanna S, et al. Stim1 activation of adenylyl cyclase 6 connects Ca2+ and camp signaling during melanogenesis. EMBO J. 2018. https://doi.org/10.15252/embj.201797597.
[37] Pinton P, Giorgi C, Siviero R, Zecchini E, Rizzuto R. Calcium and apoptosis: ER-mitochondria Ca2+ transfer in the control of apoptosis. Oncogene. 2008. https://doi.org/10.1038/onc.2008.308.
[38] Danese A, Leo S, Rimessi A, Wieckowski MR, Fiorica F, Giorgi C, et al. Cell death as a result of calcium signaling modulation: a cancer-centric prospective. Biochim Biophys Acta Mol Cell Res. 2021. https://doi.org/10.1016/j.bbamcr.2021.119061.
[39] Macalino SJ, Gosu V, Hong S, Choi S. Role of computer-aided drug design in modern drug discovery. Arch Pharm Res. 2015. https://doi.org/10.1007/s12272-015-0640-5.
[40] Heishima K, Sugito N, Soga T, Nishikawa M, Ito Y, Honda R, et al. Petasin potently inhibits mitochondrial complex I-based metabolism that supports tumor growth and metastasis. J Clin Invest. 2021. https://doi.org/10.1172/jci139933.
[41] Zhong W, Myers JS, Wang F, Wang K, Lucas J, Rosfjord E, et al. Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumors. BMC Genomics. 2020. https://doi.org/10.1186/s12864-019-6344-3.
[42] Joost S, Annusver K, Jacob T, Sun X, Dalessandri T, Sivan U, et al. The molecular anatomy of mouse skin during hair growth and rest. Cell Stem Cell. 2020. https://doi.org/10.1016/j.stem.2020.01.012.
[43] Takahashi R, Grzenda A, Allison TF, Rawnsley J, Balin SJ, Sabri S, et al. Defining transcriptional signatures of human hair follicle cell states. J Invest Dermatol. 2020. https://doi.org/10.1016/j.jid.2019.07.726.
[44] Liao Y, Smyth GK, Shi W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014. https://doi.org/10.1093/bioinformatics/btt656.
[45] Chen Y, Lun AT, Smyth GK. From reads to genes to pathways: differential expression analysis of RNA-Seq experiments using Rsubread and the edgeR quasi-likelihood pipeline. F1000Res. 2016. https://doi.org/10.12688/f1000research.8987.2.
[46] McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 2012. https://doi.org/10.1093/nar/gks042.
[47] Robinson MD, McCarthy DJ, Smyth GK. EdgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010. https://doi.org/10.1093/bioinformatics/btp616.
[48] Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. ClusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb). 2021. https://doi.org/10.1016/j.xinn.2021.
[49] Yu G, Wang LG, Han Y, He QY. ClusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012. https://doi.org/10.1089/omi.2011.0118.
[1] Daniel W. Armstrong, Alain Berthod. Occurrence of D-amino acids in natural products[J]. Natural Products and Bioprospecting, 2023, 13(6): 47-47.
[2] Lingli Ding, Zhao Gao, Siluo Wu, Chen Chen, Yamei Liu, Min Wang, Yage Zhang, Ling Li, Hong Zou, Guoping Zhao, Shengnan Qin, Liangliang Xu. Ginsenoside compound-K attenuates OVX-induced osteoporosis via the suppression of RANKL-induced osteoclastogenesis and oxidative stress[J]. Natural Products and Bioprospecting, 2023, 13(6): 49-49.
[3] Dalila Carbone, Carmela Gallo, Genoveffa Nuzzo, Giusi Barra, Mario Dell'Isola, Mario Affuso, Olimpia Follero, Federica Albiani, Clementina Sansone, Emiliano Manzo, Giuliana d'Ippolito, Angelo Fontana. Marine natural product lepadin A as a novel inducer of immunogenic cell death via CD91-dependent pathway[J]. Natural Products and Bioprospecting, 2023, 13(5): 34-34.
[4] Phanankosi Moyo, Luke Invernizzi, Sephora M. Mianda, Wiehan Rudolph, Andrew W. Andayi, Mingxun Wang, Neil R. Crouch, Vinesh J. Maharaj. Prioritised identification of structural classes of natural products from higher plants in the expedition of antimalarial drug discovery[J]. Natural Products and Bioprospecting, 2023, 13(5): 37-37.
[5] Phanankosi Moyo, Luke Invernizzi, Sephora M. Mianda, Wiehan Rudolph, Warren A. Andayi, Mingxun Wang, Neil R. Crouch, Vinesh J. Maharaj. Leveraging off higher plant phylogenetic insights for antiplasmodial drug discovery[J]. Natural Products and Bioprospecting, 2023, 13(5): 35-35.
[6] Ji-Kai Liu. Natural products in cosmetics[J]. Natural Products and Bioprospecting, 2022, 12(6): 40-40.
[7] Si-Yuan Luo, Jun-Yu Zhu, Ming-Feng Zou, Sheng Yin, Gui-Hua Tang. Mulberry Diels–Alder-type adducts: isolation, structure, bioactivity, and synthesis[J]. Natural Products and Bioprospecting, 2022, 12(5): 31-31.
[8] Ji-Kai Liu. Antiaging agents: safe interventions to slow aging and healthy life span extension[J]. Natural Products and Bioprospecting, 2022, 12(3): 18-18.
[9] Yulian Lv, Tian Tian, Yong-Jiang Wang, Jian-Ping Huang, Sheng-Xiong Huang. Advances in chemistry and bioactivity of the genus Erythroxylum[J]. Natural Products and Bioprospecting, 2022, 12(3): 15-15.
[10] Ding-kang Chen, Hui-yan Shao, Liu Yang, Jiang-miao Hu. The bibenzyl derivatives of Dendrobium officinale prevent UV-B irradiation induced photoaging via SIRT3[J]. Natural Products and Bioprospecting, 2022, 12(1): 1-11.
[11] Ghodsi Mohammadi Ziarani, Negar Jamasbi, Fatemeh Mohajer. Recent advances on the synthesis of natural pyrrolizidine alkaloids: alexine, and its stereoisomers[J]. Natural Products and Bioprospecting, 2022, 12(1): 1-15.
[12] Oyere Tanyi Ebob, Smith B. Babiaka, Fidele Ntie-Kang. Natural Products as Potential Lead Compounds for Drug Discovery Against SARS-CoV-2[J]. Natural Products and Bioprospecting, 2021, 11(6): 611-628.
[13] Christian Bailly, Gérard Vergoten. Anticancer Properties and Mechanism of Action of Oblongifolin C, Guttiferone K and Related Polyprenylated Acylphloroglucinols[J]. Natural Products and Bioprospecting, 2021, 11(6): 629-641.
[14] Patrick O. Sakyi, Richard K. Amewu, Robert N. O. A. Devine, Emahi Ismaila, Whelton A. Miller, Samuel K. Kwofie. The Search for Putative Hits in Combating Leishmaniasis: The Contributions of Natural Products Over the Last Decade[J]. Natural Products and Bioprospecting, 2021, 11(5): 489-544.
[15] Jean-François Goossens, Laurence Goossens, Christian Bailly. Hinokiflavone and Related C-O-C-Type Biflavonoids as Anti-cancer Compounds: Properties and Mechanism of Action[J]. Natural Products and Bioprospecting, 2021, 11(4): 365-377.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed