Title
Author
DOI
Article Type
Special Issue
Volume
Issue
Platycodin D induces apoptosis in human prostate carcinoma cells via ROS-dependent inactivation of the PI3K/AKT/mTOR signaling pathway
1Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-Eui University, 47227 Busan, Republic of Korea
2Department of Biochemistry, College of Korean Medicine, Dong-Eui University, 47227 Busan, Republic of Korea
DOI: 10.22514/jomh.2025.064 Vol.21,Issue 5,May 2025 pp.18-29
Submitted: 16 January 2025 Accepted: 24 February 2025
Published: 30 May 2025
*Corresponding Author(s): Yung Hyun Choi E-mail: choiyh@deu.ac.kr
Background: Platycodin D is a triterpene saponin present in Platycodi Radix, an herbal medicine used for treating various diseases. Recently, this saponin has been identified as a bioactive natural compound with health benefits, including anticancer properties and no side effects. However, studies on its anticancer effects in human prostate cancer (PCa) cells remain limited. In this study, we elucidated the mechanism of anticancer activity of platycodin D in human PCa cells. Methods: After measuring the cytotoxicity of platycodin D, the anticancer potential of this compound was determined by analyzing the induction of apoptotic cell death, DNA damage, reactive oxygen species (ROS) generation, glutathione content, caspase activity and mitochondrial membrane potential (MMP). Additionally, the underlying mechanism of platycodin D-induced apoptosis was determined through various analyses including protein expression analysis. Results: Platycodin D treatment induced apoptosis in PCa cells by activating the caspase cascade and reducing the protein expression levels of the inhibitor of the apoptosis family. In addition, platycodin D disrupted mitochondrial integrity by altering Bcl-2 family protein expression and MMP levels, leading to increased cytochrome c release into the cytosol. Moreover, platycodin D suppressed the pro-survival phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) (PAM) pathway. Pretreatment with a PI3K inhibitor significantly enhanced the cytotoxic effect of platycodin D on PC-3 cells. Furthermore, platycodin D promoted the generation of ROS, while ROS scavengers restored reduced cell viability by attenuating DNA damage and apoptosis through inhibition of platycodin D-mediated inactivation of the PAM pathway. Platycodin D induces apoptosis in PC-3 cells via ROS-dependent inactivation of the PAM pathway, suggesting that ROS generation plays a key role as an early mediator of platycodin D-mediated anticancer effects. Conclusions: Our findings indicate that platycodin D may serve as a potential candidate for inhibiting human PCa cell proliferation.
Platycodin D; Apoptosis; Prostate cancer cells; ROS; PI3K/AKT/mTOR
Yung Hyun Choi. Platycodin D induces apoptosis in human prostate carcinoma cells via ROS-dependent inactivation of the PI3K/AKT/mTOR signaling pathway. Journal of Men's Health. 2025. 21(5);18-29.
[1] Blanco Carcache PJ, Clinton SK, Kinghorn AD. Discovery of natural products for cancer prevention. Cancer Journal. 2024; 30: 313–319.
[2] Melfi F, Carradori S, Mencarelli N, Campestre C, Gallorini M, Di Giacomo S, et al. Natural products as a source of new anticancer chemotypes. Expert Opinion on Therapeutic Patents. 2023; 33: 721–744.
[3] Timilsena YP, Phosanam A, Stockmann R. Perspectives on saponins: food functionality and applications. International Journal of Molecular Sciences. 2023; 24: 13538.
[4] Shi L, Cui T, Wang X, Wu R, Wu J, Wang Y, et al. Biotransformation and pharmacological activities of platycosides from Platycodon grandiflorum roots. Chinese Herbal Medicines. 2024; 16: 392–400.
[5] Zhang S, Chai X, Hou G, Zhao F, Meng Q. Platycodon grandiflorum (Jacq.) A. DC.: a review of phytochemistry, pharmacology, toxicology and traditional use. Phytomedicine. 2022; 106: 154422.
[6] Xie L, Zhao YX, Zheng Y, Li XF. The pharmacology and mechanisms of platycodin D, an active triterpenoid saponin from Platycodon grandiflorus. Frontiers in Pharmacology. 2023; 14: 1148853.
[7] Li Q, Yang T, Zhao S, Zheng Q, Li Y, Zhang Z, et al. Distribution, biotransformation, pharmacological effects, metabolic mechanism and safety evaluation of platycodin D: a comprehensive review. Current Drug Metabolism. 2022; 23: 21–29.
[8] Zhang L, Wang Y, Yang D, Zhang C, Zhang N, Li M, et al. Platycodon grandiflorus—an ethnopharmacological, phytochemical and pharmacological review. Journal of Ethnopharmacology. 2015; 164: 147–161.
[9] Wang CH, Baskaran R, Ng SS, Wang TF, Li CC, Ho TJ, et al. Platycodin D confers oxaliplatin resistance in colorectal cancer by activating the LATS2/YAP1 axis of the hippo signaling pathway. Journal of Cancer. 2023; 14: 393–402.
[10] Li WH, Wang F, Song GY, Yu QH, Du RP, Xu P. PARP-1: a critical regulator in radioprotection and radiotherapy-mechanisms, challenges, and therapeutic opportunities. Frontiers in Pharmacology. 2023; 14: 1198948.
[11] Zhang X, Zhai T, Hei Z, Zhou D, Jin L, Han C, et al. Effects of platycodin D on apoptosis, migration, invasion and cell cycle arrest of gallbladder cancer cells. Oncology Letters. 2020; 20: 311.
[12] Wu D, Zhang W, Chen Y, Ma H, Wang M. Platycodin D inhibits proliferation, migration and induces chemosensitization through inactivation of the NF-κB and JAK2/STAT3 pathways in multiple myeloma cells. Clinical and Experimental Pharmacology and Physiology. 2019; 46: 1194–1200.
[13] Feng M, Wang XJ, Liu Y, Zhang W, Wang Y, Zhang C, et al. Transcriptomics reveals the mechanism of platycodin D targeting TGFβ for anti-lung cancer activity. Integrative Cancer Therapies. 2024; 23: 15347354241263041.
[14] Han SH, Lee JH, Woo JS, Jung GH, Jung SH, Han EJ, et al. Platycodin D induces apoptosis via regulating MAPK pathway and promotes autophagy in colon cancer cell. Biomedicine & Pharmacotherapy. 2024; 172: 116216.
[15] Jiang X, Lin Y, Zhao M, Li Y, Ye P, Pei R, et al. Platycodin D induces apoptotic cell death through PI3K/AKT and MAPK/ERK pathways and synergizes with venetoclax in acute myeloid leukemia. European Journal of Pharmacology. 2023; 956: 175957.
[16] Ni Z, Dawa Z, Suolang D, Pingcuo Q, Langga Z, Quzhen P, et al. Platycodin D inhibits the proliferation, invasion and migration of endometrial cancer cells by blocking the PI3K/Akt signaling pathway via ADRA2A upregulation. Oncology Letters. 2023; 25: 136.
[17] Park C, Cha HJ, Lee H, Jeong JW, Han M, Song KS, et al. Induction of apoptosis through inactivation of ROS-dependent PI3K/Akt signaling pathway by platycodin D in human bladder urothelial carcinoma cells. General Physiology and Biophysics. 2022; 41: 263–274.
[18] Seo YS, Kang OH, Kong R, Zhou T, Kim SA, Ryu S, et al. Polygalacin D induces apoptosis and cell cycle arrest via the PI3K/Akt pathway in non-small cell lung cancer. Oncology Reports. 2018; 39: 1702–1710.
[19] Lu Y, Xie L, Qi M, Ren S, Wang Y, Hu J, et al. Platycodin D ameliorates cognitive impairment in type 2 diabetes mellitus mice via regulating PI3K/Akt/GSK3β signaling pathway. Journal of Agricultural and Food Chemistry. 2024; 72: 12516–12528.
[20] Guo R, Meng Q, Wang B, Li F. Anti-inflammatory effects of Platycodin D on dextran sulfate sodium (DSS) induced colitis and E. coli Lipopolysaccharide (LPS) induced inflammation. International Immunopharmacology. 2021; 94: 107474.
[21] Hu JN, Leng J, Shen Q, Liu Y, Li XD, Wang SH, et al. Platycodin D suppresses cisplatin-induced cytotoxicity by suppressing ROS-mediated oxidative damage, apoptosis, and inflammation in HEK-293 cells. Journal of Biochemical and Molecular Toxicology. 2021; 35: e22624.
[22] Li W, Zhang Y, Cao Y, Zhao X, Xie J. Protective effects and regulatory mechanisms of platycodin D against LPS-induced inflammatory injury in BEAS-2B cells. International Immunopharmacology. 2024; 139: 112782.
[23] Song Y, Lv P, Yu J. Platycodin D inhibits diabetic retinopathy via suppressing TLR4/MyD88/NF-κB signaling pathway and activating Nrf2/HO-1 signaling pathway. Chemical Biology & Drug Design. 2024; 103: e14419.
[24] Zhang JT, Xie LY, Shen Q, Liu W, Li MH, Hu RY, et al. Platycodin D stimulates AMPK activity to inhibit the neurodegeneration caused by reactive oxygen species-induced inflammation and apoptosis. Journal of Ethnopharmacology. 2023; 308: 116294.
[25] Zeng CC, Zhang C, Yao JH, Lai SH, Han BJ, Li W, et al. Platycodin D induced apoptosis and autophagy in PC-12 cells through mitochondrial dysfunction pathway. Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy. 2016; 168: 199–205.
[26] Yu JS, Kim AK. Platycodin D induces reactive oxygen species-mediated apoptosis signal-regulating kinase 1 activation and endoplasmic reticulum stress response in human breast cancer cells. Journal of Medicinal Food. 2012; 15: 691–699.
[27] Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nature Reviews Cancer. 2020; 20: 74–88.
[28] Koundouros N, Poulogiannis G. Phosphoinositide 3-kinase/Akt signaling and redox metabolism in cancer. Frontiers in Oncology. 2018; 8: 160.
[29] Zhou R, Lu Z, Liu K, Guo J, Liu J, Zhou Y, et al. Platycodin D induces tumor growth arrest by activating FOXO3a expression in prostate cancer in vitro and in vivo. Current Cancer Drug Targets. 2015; 14: 860–871.
[30] Lu Z, Song W, Zhang Y, Wu C, Zhu M, Wang H, et al. Combined anti-cancer effects of platycodin D and sorafenib on androgen-independent and PTEN-deficient prostate cancer. Frontiers in Oncology. 2021; 11: 648985.
[31] Kang JB, Son HK, Park DJ, Jin YB, Koh PO. Chlorogenic acid regulates the expression of protein phosphatase 2A subunit B in the cerebral cortex of a rat stroke model and glutamate-exposed neurons. Laboratory Animal Research. 2024; 40: 8.
[32] Jeon SJ, Jung GH, Choi EY, Han EJ, Lee JH, Han SH, et al. Kaempferol induces apoptosis through the MAPK pathway and regulates JNK-mediated autophagy in MC-3 cells. Toxicological Research. 2024; 40: 45–55.
[33] Manigandan S, Yun JW. Sodium-potassium adenosine triphosphatase α2 subunit (ATP1A2) negatively regulates UCP1-dependent and UCP1-independent thermogenesis in 3T3-L1 adipocytes. Biotechnology and Bioprocess Engineering. 2023; 28: 644–657.
[34] Cao M, Fan B, Zhen T, Das A, Wang J. Ruthenium biochanin-A complex ameliorates lung carcinoma through the downregulation of the TGF-β/PPARγ/PI3K/TNF-α pathway in association with caspase-3-mediated apoptosis. Toxicological Research. 2023; 39: 455–475.
[35] Park C, Kim DH, Kim TH, Jeong SU, Yoon JH, Moon SK, et al. Improvement of oxidative stress-induced cytotoxicity of Angelica keiskei (Miq.) Koidz. leaves extract through activation of heme oxygenase-1 in C2C12 murine myoblasts. Biotechnology and Bioprocess Engineering. 2023; 28: 51–62.
[36] Park C, Cha HJ, Hwangbo H, Bang E, Kim HS, Yun SJ, et al. Activation of heme oxygenase-1 by mangiferin in human retinal pigment epithelial cells contributes to blocking oxidative damage. Biomolecules & Therapeutics. 2024; 32: 329–340.
[37] Hwangbo H, Park C, Bang E, Kim HS, Bae SJ, Kim E, et al. Morroniside protects C2C12 myoblasts from oxidative damage caused by ROS-mediated mitochondrial damage and induction of endoplasmic reticulum stress. Biomolecules & Therapeutics. 2024; 32: 349–360.
[38] Nisar S, Hashem S, Macha MA, Yadav SK, Muralitharan S, Therachiyil L, et al. Exploring dysregulated signaling pathways in cancer. Current Pharmaceutical Design. 2020; 26: 429–445.
[39] Sahoo G, Samal D, Khandayataray P, Murthy MK. A review on caspases: key regulators of biological activities and apoptosis. Molecular Neurobiology. 2023; 60: 5805–5837.
[40] Ai Y, Meng Y, Yan B, Zhou Q, Wang X. The biochemical pathways of apoptotic, necroptotic, pyroptotic, and ferroptotic cell death. Molecular Cell. 2024; 84: 170–179.
[41] Hanifeh M, Ataei F. XIAP as a multifaceted molecule in cellular signaling. Apoptosis. 2022; 27: 441–453.
[42] Dadsena S, Zollo C, García-Sáez AJ. Mechanisms of mitochondrial cell death. Biochemical Society Transactions. 2021; 49: 663–674.
[43] Glover HL, Schreiner A, Dewson G, Tait SWG. Mitochondria and cell death. Nature Cell Biology. 2024; 26: 1434–1446.
[44] Huang J, Chen G, Wang J, Liu S, Su J. Platycodin D regulates high glucose-induced ferroptosis of HK-2 cells through glutathione peroxidase 4 (GPX4). Biological Engineering. 2022; 13: 6627–6637.
[45] An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death and Disease. 2024; 15: 556.
[46] Lamontagne F, Paz-Trejo C, Zamorano Cuervo N, Grandvaux N. Redox signaling in cell fate: beyond damage. Biochimica et Biophysica Acta—Molecular Cell Research. 2024; 1871: 119722.
[47] Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, et al. The regulation of PTEN: novel insights into functions as cancer biomarkers and therapeutic targets. Journal of Cellular Physiology. 2023; 238: 1693–1715.
[48] Yan Y, Huang H. Interplay among PI3K/AKT, PTEN/FOXO and AR signaling in prostate cancer. Advances in Experimental Medicine and Biology. 2019; 1210: 319–331.
[49] Hosseini FS, Ahmadi A, Kesharwani P, Hosseini H, Sahebkar A. Regulatory effects of statins on akt signaling for prevention of cancers. Cellular Signalling. 2024; 120: 111213.
[50] Wang Y, Kreisberg JI, Ghosh PM. Cross-talk between the androgen receptor and the phosphatidylinositol 3-kinase/Akt pathway in prostate cancer. Current Cancer Drug Targets. 2007; 7: 591–604.
[51] Gargalionis AN, Papavassiliou KA, Basdra EK, Papavassiliou AG. mTOR signaling components in tumor mechanobiology. International Journal of Molecular Sciences. 2022; 23: 1825.
[52] Lien EC, Lyssiotis CA, Cantley LC. Metabolic reprogramming by the PI3K-Akt-mTOR pathway in cancer. Recent Results in Cancer Research. 2016; 207: 39–72.
[53] Li Y, Zhang H, Yu C, Dong X, Yang F, Wang M, et al. New insights into mitochondria in health and diseases. International Journal of Molecular Sciences. 2024; 25: 9975.
[54] Guan S, Zhao L, Peng R. Mitochondrial respiratory chain supercomplexes: From structure to function. International Journal of Molecular Sciences. 2022; 23: 13880.
[55] García-Sánchez A, Miranda-Díaz AG, Cardona-Muñoz EG. The role of oxidative stress in physiopathology and pharmacological treatment with pro- and antioxidant properties in chronic diseases. Oxidative Medicine and Cellular Longevity. 2020; 2020: 2082145.
[56] Adhikari S, Bhattacharya A, Adhikary S, Singh V, Gadad SS, Roy S, et al. The paradigm of drug resistance in cancer: an epigenetic perspective. Bioscience Reports. 2022; 42: BSR20211812.
[57] Chiu YC, Chen HH, Zhang T, Zhang S, Gorthi A, Wang LJ, et al. Predicting drug response of tumors from integrated genomic profiles by deep neural networks. BMC Medical Genomics. 2019; 12: 18.
[58] Gano CA, Fatima S, Failes TW, Arndt GM, Sajinovic M, Mahns D, et al. Anti-cancer potential of synergistic phytochemical combinations is influenced by the genetic profile of prostate cancer cell lines. Frontiers in Nutrition. 2023; 10: 1119274.
Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,200 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.
Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.
Directory of Open Access Journals (DOAJ) DOAJ is a unique and extensive index of diverse open access journals from around the world, driven by a growing community, committed to ensuring quality content is freely available online for everyone.
SCImago The SCImago Journal & Country Rank is a publicly available portal that includes the journals and country scientific indicators developed from the information contained in the Scopus® database (Elsevier B.V.)
Publication Forum - JUFO (Federation of Finnish Learned Societies) Publication Forum is a classification of publication channels created by the Finnish scientific community to support the quality assessment of academic research.
Scopus: CiteScore 0.9 (2023) Scopus is Elsevier's abstract and citation database launched in 2004. Scopus covers nearly 36,377 titles (22,794 active titles and 13,583 Inactive titles) from approximately 11,678 publishers, of which 34,346 are peer-reviewed journals in top-level subject fields: life sciences, social sciences, physical sciences and health sciences.
Norwegian Register for Scientific Journals, Series and Publishers Search for publication channels (journals, series and publishers) in the Norwegian Register for Scientific Journals, Series and Publishers to see if they are considered as scientific. (https://kanalregister.hkdir.no/publiseringskanaler/Forside).
Top