145
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

The Effect of Osthole on Transient Receptor Potential Channels: A Possible Alternative Therapy for Atopic Dermatitis

ORCID Icon &
Pages 881-898 | Received 06 Sep 2023, Accepted 18 Jan 2024, Published online: 09 Feb 2024

References

  • Cevikbas F, Lerner EA. Physiology and pathophysiology of itch. Physiol Rev. 2020;100(3):945–982. doi:10.1152/PHYSREV.00017.2019
  • Weidinger S, Novak N. Atopic dermatitis. Lancet. 2016;387(10023):1109–1122. doi:10.1016/S0140-6736(15)00149-X
  • Weidinger S, Beck LA, Bieber T, Kabashima K, Irvine AD. Atopic dermatitis. Nat Rev Dis Primers. 2018;4(1). doi:10.1038/S41572-018-0001-Z
  • Napolitano M, Fabbrocini G, Genco L, Martora F, Potestio L, Patruno C. Rapid improvement in pruritus in atopic dermatitis patients treated with upadacitinib: a real‐life experience. J Eur Acad Dermatol Venereol. 2022;36(9):1497–1498. doi:10.1111/jdv.18137
  • Paolino A, Alexander H, Broderick C, Flohr C. Non‐biologic systemic treatments for atopic dermatitis: current state of the art and future directions. Clin Exp Immunol. 2023;53(5):495–510. doi:10.1111/cea.14301
  • Twycross R, Greaves M, Handwerker H, Qjm EJ. Itch: scratching more than the surface. academic.oup.com; 2003. Available from: https://academic.oup.com/qjmed/article-abstract/96/1/7/1526242. Accessed April 23, 2023.
  • Parsons M. Histamine and its receptors. Wiley Online Lib. 2006;147(SUPPL. 1):127–135. doi:10.1038/sj.bjp.0706440
  • Rossbach K, Nassenstein C, Neuroscience MG. Histamine H1, H3 and H4 receptors are involved in pruritus. Elsevier; 2011. Available from: https://www.sciencedirect.com/science/article/pii/S0306452211006609. Accessed April 23, 2023.
  • Medzhitov R. Inflammation 2010: new adventures of an old flame. Cell. 2010;140(6):771–776. doi:10.1016/j.cell.2010.03.006
  • Steinhoff M, Schmelz M, Szabó IL, Oaklander AL. Clinical presentation, management, and pathophysiology of neuropathic itch. Lancet Neurol. 2018;17(8):709–720. doi:10.1016/S1474-4422(18)30217-5
  • Gouin O, L’Herondelle K, Lebonvallet N, et al. TRPV1 and TRPA1 in cutaneous neurogenic and chronic inflammation: pro-inflammatory response induced by their activation and their sensitization. Protein Cell. 2017;8(9):644–661. doi:10.1007/S13238-017-0395-5
  • Meng J, Wang J, Steinhoff M, Dolly JO. TNFα induces co-trafficking of TRPV1/TRPA1 in VAMP1-containing vesicles to the plasmalemma via Munc18-1/syntaxin1/SNAP-25 mediated fusion. Sci Rep. 2016;6. doi:10.1038/SREP21226
  • Meng J, Moriyama M, Feld M, et al. New mechanism underlying IL-31-induced atopic dermatitis. J Allergy Clin Immunol. 2018;141(5):1677–1689.e8. doi:10.1016/J.JACI.2017.12.1002
  • Cevikbas F, Wang X, Akiyama T, et al. A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: involvement of TRPV1 and TRPA1. J Allergy Clin Immunol. 2014;133(2). doi:10.1016/J.JACI.2013.10.048
  • Larkin C, Chen W, Szabó IL, et al. Novel insights into the TRPV3-mediated itch in atopic dermatitis. J Allergy Clin Immunol. 2021;147(3):1110–1114.e5. doi:10.1016/J.JACI.2020.09.028
  • Wilson SR, Thé L, Batia LM, et al. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell. 2013;155(2):285–295. doi:10.1016/J.CELL.2013.08.057
  • Choi JE, Di Nardo A. Skin neurogenic inflammation. Semin Immunopathol. 2018;40(3):249–259. doi:10.1007/S00281-018-0675-Z
  • Gibson RA, Robertson J, Mistry H, et al. A randomised trial evaluating the effects of the TRPV1 antagonist SB705498 on pruritus induced by histamine, and cowhage challenge in healthy volunteers. PLoS One. 2014;9(7):e100610. doi:10.1371/JOURNAL.PONE.0100610
  • Kim B, Lee S, Shim W. Histamine-induced Ca2+ influx via the PLA2/lipoxygenase/TRPV1 pathway in rat sensory neurons. Elsevier; 2004. Available from: https://www.sciencedirect.com/science/article/pii/S0304394004000370?casa_token=WWJD5UpHlD8AAAAA:mdPx9Ob0ZKz_29E4yMe_W-3kIPluJyZl02YmHPtmKRoVtkm5eEgsXLXxiGS0ZpCPqRFyX2ezZfXc. Accessed April 23, 2023.
  • Costa R, Marotta DM, Manjavachi MN, et al. Evidence for the role of neurogenic inflammation components in trypsin‐elicited scratching behaviour in mice. Wiley Online Lib. 2008;154(5):1094–1103. doi:10.1038/bjp.2008.172
  • Sokabe T, Fukumi-Tominaga T. The TRPV4 channel contributes to intercellular junction formation in keratinocytes*♦. ASBMB; 2010. Available from: https://www.jbc.org/article/S0021-9258(19)57581-0/abstract. Accessed April 23, 2023.
  • Denda M, Sokabe T. Effects of skin surface temperature on epidermal permeability barrier homeostasis. Elsevier; 2007. Available from: https://www.sciencedirect.com/science/article/pii/S0022202X15333030. Accessed April 23, 2023.
  • Bromma B, Scharein E, Darsow U. Effects of menthol and cold on histamine-induced itch and skin reactions in man. Elsevier; 1995. Available from: https://www.sciencedirect.com/science/article/pii/030439409511362Z. Accessed April 23, 2023.
  • Wang L, Peng Y, Shi K, et al. Osthole inhibits proliferation of human breast cancer cells by inducing cell cycle arrest and apoptosis. J Biomed Res. 2015;30:132. doi:10.7555/JBR.27.20120115
  • Che Y, Li J, Li Z, et al. Osthole enhances antitumor activity and irradiation sensitivity of cervical cancer cells by suppressing ATM/NF-κB signaling. Oncol Rep. 2018. doi:10.3892/or.2018.6514
  • Xu X, Liu X, Zhang Y, Pizzo SV. Osthole inhibits gastric cancer cell proliferation through regulation of PI3K/AKT. Pizzo S V., ed. PLoS One. 2018;13(3):e0193449. doi:10.1371/journal.pone.0193449
  • Kordulewska NK, Kostyra E, Cieślińska A, Fiedorowicz E, Jarmołowska B. Cytokine production by PBMC and serum from allergic and non-allergic subjects following in vitro histamine stimulation to test fexofenadine and osthole anti-allergic properties. Eur J Pharmacol. 2016;791:763–772. doi:10.1016/j.ejphar.2016.10.020
  • Kordulewska N, Topa J, Cieślińska A, Jarmołowska B. Osthole regulates secretion of pro-inflammatory cytokines and expression of TLR2 and NF-κB in normal human keratinocytes and fibroblasts. J Inflamm Res. 2022;15:1501. doi:10.2147/JIR.S349216
  • Kordulewska NK, Topa J, Stryiński R, Jarmołowska B. Osthole inhibits expression of genes associated with toll-like receptor 2 signaling pathway in an organotypic 3D skin model of human epidermis with atopic dermatitis. Cells. 2021;11(1):88. doi:10.3390/CELLS11010088
  • Kordulewska NK, Cieślińska A, Fiedorowicz E, Jarmołowska B, Kostyra E. High expression of IL-1RI and EP2 receptors in the IL-1β/COX-2 pathway, and a new alternative to non-steroidal drugs—osthole in inhibition COX-2. Int J Mol Sci. 2019;20(1):186. doi:10.3390/ijms20010186
  • Zafar S, Sarfraz I, Rasul A, et al. Osthole: a multifunctional natural compound with potential anticancer, antioxidant and anti-inflammatory activities. Mini-Rev Med Chem. 2020;21(18):2747–2763. doi:10.2174/1389557520666200709175948
  • Zhang QY, Tao SY, Lu C, et al. Osthole: a traditional Chinese medicine for ocular anti-angiogenic therapy. Ophthalmic Res. 2020;63(5):483–490. doi:10.1159/000505976
  • Chiang CY, Lee CC, Fan CK, Huang HM, Chiang BL, Lee YL. Osthole treatment ameliorates Th2-mediated allergic asthma and exerts immunomodulatory effects on dendritic cell maturation and function. Cell Mol Immunol. 2017;14(11):935–947. doi:10.1038/cmi.2017.71
  • Wang J, Fu Y, Wei Z, et al. Anti-asthmatic activity of osthole in an ovalbumin-induced asthma murine model. Respir Physiol Neurobiol. 2017;239:64–69. doi:10.1016/J.RESP.2017.01.011
  • Kordulewska NK, Topa J, Rozmus D, et al. Effects of osthole on inflammatory gene expression and cytokine secretion in histamine-induced inflammation in the Caco-2 cell line. Int J Mol Sci. 2021;22(24):13634. doi:10.3390/IJMS222413634
  • Huang R, Chen C, Huang Y, et al. Osthole increases glycosylation of hepatitis B surface antigen and suppresses the secretion of hepatitis B virus in vitro. Hepatology. 1996;24(3):508–515. doi:10.1002/hep.510240307
  • Song F. Experimental study of osthole on treatment of hyperlipidemic and alcoholic fatty liver in animals. World J Gastroenterol. 2006;12(27):4359. doi:10.3748/wjg.v12.i27.4359
  • Zhang J, Xue J, Wang H, Zhang Y, Xie M. Osthole improves alcohol-induced fatty liver in mice by reduction of hepatic oxidative stress. Phytother Res. 2011;25(5):638–643. doi:10.1002/ptr.3315
  • Kordulewska NK, Kostyra E, Matysiewicz M, Cieślińska A, Jarmołowska B. Impact of fexofenadine, osthole and histamine on peripheral blood mononuclear cell proliferation and cytokine secretion. Eur J Pharmacol. 2015;761. doi:10.1016/j.ejphar.2015.05.065
  • Kordulewska NK, Kostyra E, Cieślińska A, Matysiewicz M, Fiedorowicz E, Sienkiewicz-Szłapka E. Changes in gene expression induced by histamine, fexofenadine and osthole: expression of histamine H1 receptor, COX-2, NF-κB, CCR1, chemokine CCL5/RANTES and interleukin-1β in PBMC allergic and non-allergic patients. Immunobiology. 2017;222(3):571–581. doi:10.1016/j.imbio.2016.11.004
  • Kordulewska NK, Cieślińska A, Fiedorowicz E, Jarmołowska B, Piskorz-Ogórek K, Kostyra E. Cytokines concentrations in serum samples from allergic children—multiple analysis to define biomarkers for better diagnosis of allergic inflammatory process. Immunobiology. 2018;223(11):648–657. doi:10.1016/j.imbio.2018.07.010
  • Kordulewska NK, Kostyra E, Piskorz-Ogórek K, et al. Serum cytokine levels in children with spectrum autism disorder: differences in pro- and anti-inflammatory balance. J Neuroimmunol. 2019;337:577066. doi:10.1016/j.jneuroim.2019.577066
  • Kordulewska NK, Kostyra E, Chwała B, et al. A novel concept of immunological and allergy interactions in autism spectrum disorders: molecular, anti-inflammatory effect of osthole. Int Immunopharmacol. 2019;72:1–11. doi:10.1016/j.intimp.2019.01.058
  • Patel HK, Barot BS, Parejiya PB, Shelat PK, Shukla A. Topical delivery of clobetasol propionate loaded microemulsion based gel for effective treatment of vitiligo – part II: rheological characterization and in vivo assessment through dermatopharmacokinetic and pilot clinical studies. Colloids Surf B Biointerfaces. 2014;119:145–153. doi:10.1016/j.colsurfb.2014.02.005
  • Yuan N, Chen Z, Li L. Narrow-band UVB combined with compound clobetasol propionate can improve the therapeutic effect in hand eczema patients. Am J Transl Res. 2021;13(7):8193–8199.
  • Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29(9):45e–445. doi:10.1093/nar/29.9.e45
  • Gao X, Wang Z, Du L. Glial cells and itch: possible targets for novel antipruritic therapies. ACS Chem Neurosci. 2023;14(3):331–339. doi:10.1021/ACSCHEMNEURO.2C00638/ASSET/IMAGES/MEDIUM/CN2C00638_0003.GIF
  • Fang Y, Yang L, He J. Plantanone C attenuates LPS-stimulated inflammation by inhibiting NF-κB/iNOS/COX-2/MAPKs/Akt pathways in RAW 264.7 macrophages. Biomed Pharmacother. 2021;143:112104. doi:10.1016/j.biopha.2021.112104
  • Radha KM, Puri S. Evaluation of nutritional, phytochemical, and mineral composition of selected medicinal plants for therapeutic uses from cold desert of western himalaya. Plants. 2021;10(7):1429. doi:10.3390/plants10071429
  • Bitton A, Avlas S, Reichman H, et al. A key role for IL-13 signaling via the type 2 IL-4 receptor in experimental atopic dermatitis. Sci Immunol. 2020;5(44). doi:10.1126/SCIIMMUNOL.AAW2938/SUPPL_FILE/AAW2938_SM.PDF
  • Fu X, Hong C. Osthole attenuates mouse atopic dermatitis by inhibiting thymic stromal lymphopoietin production from keratinocytes. Exp Dermatol. 2019;28(5):561–567. doi:10.1111/EXD.13910
  • Yang Q, Kong L, Huang W, et al. Osthole attenuates ovalbumin-induced lung inflammation via the inhibition of IL-33/ST2 signaling in asthmatic mice. Int J Mol Med. 2020;46(4):1389–1398. doi:10.3892/IJMM.2020.4682/HTML
  • Shen W, Shi S, Zhang H. Osthole inhibits ovalbumin (OVA) induced asthma through regulating TLR9/JNK pathway. IJEB. 2020;58:235–241.
  • Ferreira S, Torres T. Dupilumab for the treatment of atopic dermatitis. Actas Dermo-Sifiliográficas. 2018;109(3):230–240. doi:10.1016/J.ADENGL.2018.02.019
  • Freitas E, Guttman-Yassky E, Torres T. Tralokinumab for the treatment of atopic dermatitis. Am J Clin Dermatol. 2021;22(5):625–638. doi:10.1007/S40257-021-00613-8
  • Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156(4):411–420. doi:10.1001/JAMADERMATOL.2020.0079
  • Tsianakas A, Luger TA. The anti-IL-4 receptor alpha antibody dupilumab: facing a new era in treating atopic dermatitis. Expert Opin Biol Ther. 2015;15(11):1661–1664. doi:10.1517/14712598.2015.1095887
  • Akdis CA, Arkwright PD, Brüggen MC, et al. Type 2 immunity in the skin and lungs. Wiley Online Lib. 2020;10(7):9. doi:10.1111/all.14318
  • Yang N, Ju Y, Huang D, et al. Desensitization of TRPV1 involved in the antipruritic effect of osthole on histamine-induced scratching behavior in mice. Evid Based Complement Alternat Med. 2021;2021:1–10. doi:10.1155/2021/4012812
  • Jian T, Yang N, Yang Y, et al. TRPV1 and PLC participate in histamine H4 receptor-induced itch. hindawi.com. Available from: https://www.hindawi.com/journals/np/2016/1682972/. Accessed April 26, 2023.
  • Shim WS, Tak MH, Lee MH, et al. TRPV1 mediates histamine-induced itching via the activation of phospholipase A2 and 12-lipoxygenase. J Neurosci. 2007;27(9):2331–2337. doi:10.1523/JNEUROSCI.4643-06.2007
  • Han SK, Mancino V, Simon MI. Phospholipase Cβ 3 Mediates the scratching response activated by the histamine H1 receptor on C-fiber nociceptive neurons. Neuron. 2006;52(4):691–703. doi:10.1016/j.neuron.2006.09.036
  • Gutzmer R, Mommert S, Gschwandtner M, Zwingmann K, Stark H, Werfel T. The histamine H4 receptor is functionally expressed on TH2 cells. J Allergy Clin Immunol. 2009;123(3):619–625. doi:10.1016/J.JACI.2008.12.1110
  • Glatzer F, Gschwandtner M, Ehling S, et al. Histamine induces proliferation in keratinocytes from patients with atopic dermatitis through the histamine 4 receptor. J Allergy Clin Immunol. 2013;132(6):1358–1367. doi:10.1016/J.JACI.2013.06.023
  • Glatzer F, Gschwandtner M, Ehling S, et al. Histamine induces proliferation in keratinocytes from atopic dermatitis patients. J Allergy Clin Immunol. 2013;132(6):1358. doi:10.1016/J.JACI.2013.06.023
  • Yu B, Shao Y, Zhang J, et al. Polymorphisms in human histamine receptor H4 gene are associated with atopic dermatitis. Br J Dermatol. 2010;162(5):1038–1043. doi:10.1111/j.1365-2133.2010.09675.x
  • Mahmoud O, Soares GB, Yosipovitch G. Transient receptor potential channels and itch. Int J Mol Sci. 2023;24(1). doi:10.3390/IJMS24010420
  • Cevikbas F, Wang X, Akiyama T. CKJ of allergy and, 2014 undefined. A sensory neuron–expressed IL-31 receptor mediates T helper cell–dependent itch: involvement of TRPV1 and TRPA1. Elsevier; 2014. Available from: https://www.sciencedirect.com/science/article/pii/S0091674913017107?casa_token=WM47qT_DrlkAAAAA:Y3_M05j1zYDJ_b-Vh_uWLMO-Jc8FiotlQvCfKnjx7Yl1Gy7iICBcDEAlrjB43J3YXRL0PUHDIzIX. Accessed April 26, 2023.
  • Fernandes ES, Vong CT, Quek S, et al. Superoxide generation and leukocyte accumulation: key elements in the mediation of leukotriene B4-induced itch by transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1. THE FASEB Journal. 2013;27(4):1664–1673. doi:10.1096/FJ.12-221218
  • Torres KV, Pantke S, Rudolf D, Eberhardt MM, Leffler A. The coumarin osthole is a non-electrophilic agonist of TRPA1. Neurosci Lett. 2022;789:136878. doi:10.1016/J.NEULET.2022.136878
  • Duchatelet S, Hovnanian A. Olmsted syndrome: clinical, molecular and therapeutic aspects. Orphanet J Rare Dis. 2015;10(1):33. doi:10.1186/S13023-015-0246-5
  • Yun J, Seo J, Jeong Y, Bae I. TRPV1 antagonist can suppress the atopic dermatitis-like symptoms by accelerating skin barrier recovery. Elsevier; 2011. Available from: https://www.sciencedirect.com/science/article/pii/S0923181110003300?casa_token=3-YhlgP7YAMAAAAA:xzvx2kPNd8s3trMIX6g757gth6yHdK01PWko2FX7pvkwB71-UbOco5YKll1kVgAT1lpvPns16iaE. Accessed April 27, 2023.
  • Yun J, Seo J, Jang W, Koh H, of IBTJ. Antipruritic effects of TRPV1 antagonist in murine atopic dermatitis and itching models. europepmc.org; 2011. Available from: https://europepmc.org/article/med/21471988. Accessed April 27, 2023.
  • Yun J, Seo J, Jeong Y, Bae I. TRPV1 antagonist can suppress the atopic dermatitis-like symptoms by accelerating skin barrier recovery. Elsevier; 2011. Available from: https://www.sciencedirect.com/science/article/pii/S0923181110003300?casa_token=5Qk1KVysWUQAAAAA:FueEdq_q0PqJ-IwmywcNTFhr6KLMVUxl9HYfffFlcFHZM70qHwkXE6mMyJWkjMXtELzn1wdByhiR. Accessed April 27, 2023.
  • Jian T, Yang N, Yang Y, et al. TRPV1 and PLC participate in histamine H4 receptor-induced itch. hindawi.com. Available from: https://www.hindawi.com/journals/np/2016/1682972/. Accessed April 27, 2023.
  • Oh M, Oh S, Lu J, Lou H. AMTJ of, 2013 undefined. TRPA1-dependent pruritus in IL-13–induced chronic atopic dermatitis. journals.aai.org; 2013. Available from: https://journals.aai.org/jimmunol/article/191/11/5371/39018. Accessed April 27, 2023.
  • Xiao R, Tian J, Tang J, The TRPV3 mutation associated with the hairless phenotype in rodents is constitutively active. Elsevier; 2008. Available from: https://www.sciencedirect.com/science/article/pii/S0143416007001303?casa_token=EL8UtlCGwtsAAAAA:M7LNabS_6B6EAviYRegwiaroP0dtNdq48kI_IDrOZDEVoXmKju-xX5WEhGbIROQUSyNJOgYr3a9G. Accessed April 27, 2023.
  • Song Z, Chen X, Zhao Q, et al. Hair loss caused by gain-of-function mutant TRPV3 is associated with premature differentiation of follicular keratinocytes. Elsevier. Available from: https://www.sciencedirect.com/science/article/pii/S0022202X21001585. Accessed April 27, 2023.
  • Larkin C, Chen W, Szabó I. CSJ of A and, 2021 undefined. Novel insights into the TRPV3-mediated itch in atopic dermatitis. jacionline.org; 2021. Available from: https://www.jacionline.org/article/S0091-6749(20)31394-4/abstract. Accessed April 27, 2023.
  • Buddenkotte J, Stroh C, Engels I. Agonists of proteinase-activated receptor-2 stimulate upregulation of intercellular cell adhesion molecule-1 in primary human keratinocytes via activation of. Elsevier; 2005. Available from: https://www.sciencedirect.com/science/article/pii/S0022202X15321321. Accessed April 27, 2023.
  • Szöllősi A, Vasas N, Angyal Á, Activation of TRPV3 regulates inflammatory actions of human epidermal keratinocytes. Elsevier; 2018. Available from: https://www.sciencedirect.com/science/article/pii/S0022202X17330221. Accessed April 27, 2023.
  • Xu H, Delling M, Jun J, Clapham DE. Oregano, thyme and clove-derived flavors and skin sensitizers activate specific TRP channels. Nat Neurosci. 2006;9(5):628–635. doi:10.1038/nn1692
  • Doerner J, Hatt H, Voltage- and temperature-dependent activation of TRPV3 channels is potentiated by receptor-mediated PI(4,5)P2 hydrolysis. rupress.org; 2011. Available from: https://rupress.org/jgp/article-abstract/137/3/271/42851. Accessed April 27, 2023.
  • Du S, Araki I, Kobayashi H, Zakoji H, Sawada N, Takeda M. Differential Expression Profile of Cold (TRPA1) and cool (TRPM8) receptors in human urogenital organs. Urology. 2008;72(2):450–455. doi:10.1016/J.UROLOGY.2007.11.127
  • Liu Y, Mikrani R, He Y, et al. TRPM8 channels: a review of distribution and clinical role. Eur J Pharmacol. 2020:882. doi:10.1016/J.EJPHAR.2020.173312
  • Liu B, Jordt SE. Cooling the itch via TRPM8. J Invest Dermatol. 2018;138(6):1254–1256. doi:10.1016/J.JID.2018.01.020
  • Palkar R, Ongun S, Catich E, et al. Cooling relief of acute and chronic itch requires TRPM8 channels and neurons. J Invest Dermatol. 2018;138(6):1391–1399. doi:10.1016/J.JID.2017.12.025
  • Kardon AP, Polgár E, Hachisuka J, et al. Dynorphin acts as a neuromodulator to inhibit itch in the dorsal horn of the spinal cord. Neuron. 2014;82(3):573–586. doi:10.1016/J.NEURON.2014.02.046
  • Kim JH, Jang YS, Park JY, et al. The activation of transient receptor potential melastatin 8 (TRPM8) receptors induces airway inflammation in bronchial asthma. J Allergy Clin Immunol. 2017;139(2):AB2. doi:10.1016/j.jaci.2016.12.060
  • Sun M, Sun M, Zhang J. Osthole: an overview of its sources, biological activities, and modification development. Med Chem Res. 2021;30(10):1767–1794. doi:10.1007/s00044-021-02775-w
  • Fan H, Gao Z, Ji K, et al. The in vitro and in vivo anti-inflammatory effect of osthole, the major natural coumarin from Cnidium monnieri (L.) Cuss, via the blocking of the activation of the NF-κB and MAPK/p38 pathways. Phytomedicine. 2019;58:152864. doi:10.1016/j.phymed.2019.152864
  • Wu SJ. Osthole attenuates inflammatory responses and regulates the expression of inflammatory mediators in HepG2 cells grown in differentiated medium from 3T3-L1 preadipocytes. J Med Food. 2015;18(9):972–979. doi:10.1089/jmf.2014.3314
  • Joshi P, Singh S, Wani A, et al. Osthol and curcumin as inhibitors of human Pgp and multidrug efflux pumps of Staphylococcus aureus: reversing the resistance against frontline antibacterial drugs. Med Chem Commun. 2014;5(10):1540–1547. doi:10.1039/C4MD00196F
  • Zheng H, Chen Y, Guo Q, et al. Inhibitory effect of osthole from Cnidium monnieri (L.) Cusson on fusarium oxysporum, a common fungal pathogen of potato. Molecules. 2021;26(13):3818. doi:10.3390/molecules26133818
  • Peng LL, Juan WX, Zhang JY, et al. Antifungal activity of osthol in vitro and enhancement in vivo through Eudragit S100 nanocarriers. Virulence. 2018;9(1):555–562. doi:10.1080/21505594.2017.1356503
  • Rashidpour S, Zahedipour F, Karimi G, Jamialahmadi K. Protective effects of osthole against free radical-induced hemolysis of erythrocytes. Pharm Sci. 2020;27(1):56–62. doi:10.34172/PS.2020.65
  • Huang WC, Liao PC, Huang CH, Hu S, Huang SC, Wu SJ. Osthole attenuates lipid accumulation, regulates the expression of inflammatory mediators, and increases antioxidants in FL83B cells. Biomed Pharmacother. 2017;91:78–87. doi:10.1016/j.biopha.2017.04.051
  • Gao L, Wang F, Chen Y, Li F, Han B, Liu D. The antithrombotic activity of natural and synthetic coumarins. Fitoterapia. 2021;154:104947. doi:10.1016/j.fitote.2021.104947
  • Liang HJ, Suk FM, Wang CK, et al. Osthole, a potential antidiabetic agent, alleviates hyperglycemia in db/db mice. Chem Biol Interact. 2009;181(3):309–315. doi:10.1016/j.cbi.2009.08.003
  • Luszczki JJ, Wojda E, Andres-Mach M, et al. Anticonvulsant and acute neurotoxic effects of imperatorin, osthole and valproate in the maximal electroshock seizure and chimney tests in mice: a comparative study. Epilepsy Res. 2009;85(2–3):293–299. doi:10.1016/j.eplepsyres.2009.03.027
  • Li Y, Sun Z, Xu H, Zhang Q, Zeng C. Osthole inhibits proliferation of kainic acid-activated BV-2 cells by modulating the Notch signaling pathway. Mol Med Rep. 2020. doi:10.3892/mmr.2020.11455
  • Zhang L, Jiang G, Yao F, et al. Growth inhibition and apoptosis induced by osthole, a natural coumarin, in hepatocellular carcinoma. Cox D, ed. PLoS One. 2012;7(5):e37865. doi:10.1371/journal.pone.0037865
  • Mei J, Wang T, Zhao S, Zhang Y. Osthole inhibits breast cancer progression through upregulating tumor suppressor GNG7. Hou Y, ed. J Oncol. 2021;2021:1–12. doi:10.1155/2021/6610511
  • Mark R, Lyu X, Lee JJL, Parra-Saldívar R, Chen WN. Sustainable production of natural phenolics for functional food applications. J Funct Foods. 2019;57:233–254. doi:10.1016/j.jff.2019.04.008