Journal Home > Volume 13 , Issue 3

3-Epi-betulinic acid 3-O-β-D-glucopyranoside (eBAG) is a pentacyclic triterpene mainly distributed in food and medicinal plants, which exhibits various pharmacological properties. However, whether these functions are attributed to eBAG or additional components in these plants remain unknown. Herein, we report that eBAG exerted an inhibitory activity against hepatocellular carcinoma and esophageal cancer cells. EBAG induced non-apoptotic cell death in hepatocellular carcinoma cells. The eBAG-induced cell death was inhibited by knock-down of autophagy related gene (ATG) 5 and ATG7, by administration of 3-methyladenine, a selective autophagy inhibitor that suppresses phosphoinositide 3-kinase (PI3K), and by chloroquine, a classic autophagy f lux inhibitor. We demonstrated that eBAG induced an autophagy-mediated cell death. Application of eBAG mimicked cellular bioenergetics depletion leading to the reduction of intracellular ATP, activation of AMP-activated protein kinase (AMPK), and inhibition of mTOR. Co-treatment with compound C, an AMPK inhibitor, abrogated cell death induced by eBAG. We further validated the anti-tumor effect of eBAG in the murine xenograft model of hepatocellular carcinoma and found that eBAG treatment promoted the induction of autophagy and reduction of tumor growth in mice. As a functional food ingredient, eBAG is a potential therapeutic agent for the treatment of hepatocellular carcinoma and esophageal cancer.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

3-Epi-betulinic acid 3-O-β-D-glucopyranoside (eBAG) induces autophagy by activation of AMP-activated protein kinase in hepatocellular carcinoma

Show Author's information Mengjie Liua,1,Xuqiang Liub,1( )Kaiyue HeaYongping JianaYujia LiaJinrong GuoaJingyu YangaZhixiang Xua( )Wenyi Kangb,c,d( )
School of Life Sciences, Henan University, Kaifeng 475004, China
National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China
Shenzhen Research Institute of Henan University, Shenzhen 518000, China

1 These authors contributed equally to this work.

Peer review under responsibility of Tsinghua University Press.

Abstract

3-Epi-betulinic acid 3-O-β-D-glucopyranoside (eBAG) is a pentacyclic triterpene mainly distributed in food and medicinal plants, which exhibits various pharmacological properties. However, whether these functions are attributed to eBAG or additional components in these plants remain unknown. Herein, we report that eBAG exerted an inhibitory activity against hepatocellular carcinoma and esophageal cancer cells. EBAG induced non-apoptotic cell death in hepatocellular carcinoma cells. The eBAG-induced cell death was inhibited by knock-down of autophagy related gene (ATG) 5 and ATG7, by administration of 3-methyladenine, a selective autophagy inhibitor that suppresses phosphoinositide 3-kinase (PI3K), and by chloroquine, a classic autophagy f lux inhibitor. We demonstrated that eBAG induced an autophagy-mediated cell death. Application of eBAG mimicked cellular bioenergetics depletion leading to the reduction of intracellular ATP, activation of AMP-activated protein kinase (AMPK), and inhibition of mTOR. Co-treatment with compound C, an AMPK inhibitor, abrogated cell death induced by eBAG. We further validated the anti-tumor effect of eBAG in the murine xenograft model of hepatocellular carcinoma and found that eBAG treatment promoted the induction of autophagy and reduction of tumor growth in mice. As a functional food ingredient, eBAG is a potential therapeutic agent for the treatment of hepatocellular carcinoma and esophageal cancer.

Keywords: Autophagy, Hepatocellular carcinoma, ATP, AMPK, eBAG

References(55)

[1]

A. Villanueva, Hepatocellular carcinoma, N. Engl. J. Med. 15 (2019) 1450-1462. https://doi.org/10.1056/NEJMra1713263.

[2]

A. L. Cheng, Y.K. Kang, D.Y. Lin, et al., sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase Ⅲ trial, J. Clin. Oncol. 32 (2013) 4067-4075. https://doi.org/10.1200/jco.2012.45.8372.

[3]

P.J. Johnson, S. Qin, J.W. Park, et al., Brivanib versus sorafenib as first-line therapy in patients with unresectable, advanced hepatocellular carcinoma:results from the randomized phase III BRISK-FL study, J. Clin. Oncol. 28 (2013) 3517-3524. https://doi.org/10.1200/jco.2012.48.4410.

[4]

A.X. Zhu, M. Ancukiewicz, J.G. Supko, et al., Efficacy, safety, pharmacokinetics, and biomarkers of cediranib monotherapy in advanced hepatocellular carcinoma: a phase II study, Clin. Cancer Res. 6 (2013) 1557-1566. https://doi.org/10.1158/1078-0432.Ccr-12-3041.

[5]

A. Cucchetti, F. Piscaglia, A.D. Pinna, et al., Efficacy and safety of systemic therapies for advanced hepatocellular carcinoma: a network metaanalysis of phase III trials, Liver Cancer 4 (2017) 337-348. https://doi.org/10.1159/000481314.

[6]

A.L. Cheng, S. Thongprasert, H.Y. Lim, et al., Randomized, open-label phase 2 study comparing frontline dovitinib versus sorafenib in patients with advanced hepatocellular carcinoma, Hepatology 3 (2016) 774-784. https://doi.org/10.1002/hep.28600.

[7]

F.H. Kong, Q.F. Ye, X.Y. Miao, et al., Current status of sorafenib nanoparticle delivery systems in the treatment of hepatocellular carcinoma, Theranostics 11 (2021) 5464-5490. https://doi.org/10.7150/thno.54822.

[8]

Y. Tian, X. Zhang, M. Du, et al., Synergistic antioxidant effects of araloside a and L-ascorbic acid on H2O2-induced Hek293 cells: regulation of cellular antioxidant status, Oxid. Med. Cell Longev. 2021 (2021) 9996040. https://doi.org/10.1155/2021/9996040.

[9]

Z. Zhang, M. Jiang, X. Xie, et al., Oleanolic acid ameliorates high glucoseinduced endothelial dysfunction via PPARδ activation, Sci. Rep. 7 (2017) 40237. https://doi.org/10.1038/srep40237.

[10]

X. Zhu, K. Wang, K. Zhang, et al., Ziyuglycoside II induces cell cycle arrest and apoptosis through activation of ROS/JNK pathway in human breast cancer cells, Toxicol. Lett. 1 (2014) 65-73. https://doi.org/10.1016/j.toxlet.2014.03.015.

[11]

Y. Kong, F. Li, Y. Nian, et al., Khf16 is a leading structure from cimicifuga foetida that suppresses breast cancer partially by inhibiting the NF-κB signaling pathway, Theranostics 6 (2016) 875-886. https://doi.org/10.7150/thno.14694.

[12]

Z. Özdemir, Z. Wimmer, Selected plant triterpenoids and their amide derivatives in cancer treatment: a review, Phytochemistry 203 (2022) 113340. https://doi.org/10.1016/j.phytochem.2022.113340.

[13]

Y. Kuang, B. Li, Z. Wang, et al., Terpenoids from the medicinal mushroom antrodia camphorata: chemistry and medicinal potential, Nat. Prod. Rep. 1 (2021) 83-102. https://doi.org/10.1039/d0np00023j.

[14]

J.I. Kitajima, Y. Tanaka, Two new triterpenoid glycosides from the leaves of Schefflera octophylla, Chem. Pharm. Bull. 37 (1989) 2727-2730.

[15]

Y.L. Wu, F. Han, S.S. Luan, et al., Triterpenoids from Ganoderma Lucidum and their potential anti-inflammatory effects, J. Agric. Food Chem. 18 (2019) 5147-5158. https://doi.org/10.1021/acs.jafc.9b01195.

[16]

S. Patel, A. Rauf, Adaptogenic herb ginseng (Panax) as medical food: status quo and future prospects, Biomed. Pharmacother. 85 (2017) 120-127. https://doi.org/10.1016/j.biopha.2016.11.112.

[17]

J. Chen, K.W.K. Tsim, A review of edible jujube, the ziziphus jujuba fruit:a heath food supplement for anemia prevalence, Front. Pharmacol. 11 (2020) 593655. https://doi.org/10.3389/fphar.2020.593655.

[18]

C. Mancuso, R. Santangelo, Panax ginseng and panax quinquefolius: from pharmacology to toxicology, Food Chem. Toxicol. 107 (2017) 362-372. https://doi.org/10.1016/j.fct.2017.07.019.

[19]

G. Kuttan, P. Pratheeshkumar, K.A. Manu, et al., Inhibition of tumor progression by naturally occurring terpenoids, Pharm. Biol. 10 (2011) 995-1007. https://doi.org/10.3109/13880209.2011.559476.

[20]

C. El-Baba, A. Baassiri, G. Kiriako, et al., Terpenoids’ anti-cancer effects:focus on autophagy, Apoptosis 9/10 (2021) 491-511. https://doi.org/10.1007/s10495-021-01684-y.

[21]

M. Allaire, P.E. Rautou, P. Codogno, et al., Autophagy in liver diseases:time for translation, J. Hepatol. 5 (2019) 985-998. https://doi.org/10.1016/j.jhep.2019.01.026.

[22]

L.S. Hou, Y.W. Zhang, H. Li, et al, The regulatory role and mechanism of autophagy in energy metabolism-related hepatic fibrosis, Pharmacol. Ther.234 (2022) 108117. https://doi.org/10.1016/j.pharmthera.2022.108117.

[23]

K.E. King, T.T. Losier, R.C. Russell, Regulation of autophagy enzymes by nutrient signaling, Trends Biochem. Sci. 8 (2021) 687-700. https://doi.org/10.1016/j.tibs.2021.01.006.

[24]

K.R. Parzych, D.J. Klionsky, An overview of autophagy: morphology, mechanism, and regulation, Antioxid. Redox. Signal. 3 (2014) 460-473. https://doi.org/10.1089/ars.2013.5371.

[25]

J. Kaur, J. Debnath, Autophagy at the crossroads of catabolism and anabolism, Nat. Rev. Mol. Cell. Biol. 8 (2015) 461-472. https://doi.org/10.1038/nrm4024.

[26]

C. Lu, J. Chen, H.G. Xu, et al., MIR106B and MIR93 prevent removal of bacteria from epithelial cells by disrupting ATG16L1-mediated autophagy, Gastroenterology 1 (2014) 188-199. https://doi.org/10.1053/j.gastro.2013.09.006.

[27]

E. White, Deconvoluting the context-dependent role for autophagy in cancer, Nat. Rev. Cancer 6 (2012) 401-410. https://doi.org/10.1038/nrc3262.

[28]

J.Y. Guo, B. Xia, E. White, Autophagy-mediated tumor promotion, Cell 6 (2013) 1216-1219. https://doi.org/10.1016/j.cell.2013.11.019.

[29]

J. Moscat, M.T. Diaz-Meco, P62 at the crossroads of autophagy, apoptosis, and cancer, Cell 6 (2009) 1001-1004. https://doi.org/10.1016/j.cell.2009.05.023.

[30]

M. Komatsu, H. Kurokawa, S. Waguri, et al., The selective autophagy substrate P62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1, Nat. Cell. Biol. 3 (2010) 213-223. https://doi.org/10.1038/ncb2021.

[31]

M. López, Hypothalamic AMPK as a possible target for energy balancerelated diseases, Trends Pharmacol. Sci. 7 (2022) 546-556. https://doi.org/10.1016/j.tips.2022.04.007.

[32]

S.M. Jeon, Regulation and function of AMPK in physiology and diseases, Exp. Mol. Med. 7 (2016) e245. https://doi.org/10.1038/emm.2016.81.

[33]

D.M. Gwinn, D.B. Shackelford, D.F. Egan, et al., AMPK phosphorylation of raptor mediates a metabolic checkpoint, Mol. Cell. 2 (2008) 214-226. https://doi.org/10.1016/j.molcel.2008.03.003.

[34]

K. Inoki, T. Zhu, K.L. Guan, TSC2 Mediates cellular energy response to control cell growth and survival, Cell 5 (2003) 577-590. https://doi.org/10.1016/s0092-8674(03)00929-2.

[35]

J. Kim, M. Kundu, B. Viollet, et al., AMPK and mtor regulate autophagy through direct phosphorylation of ULK1, Nat. Cell. Biol. 2 (2011) 132-141.https://doi.org/10.1038/ncb2152.

[36]

K.K. Wang, K.Y. He, J.Y. Yang, et al, Lactobacillus suppresses tumorigenesis of oropharyngeal cancer via enhancing anti-tumor immune response, Front. Cell. Dev. Biol. 10 (2022) 842153. https://doi.org/10.3389/fcell.2022.842153.

[37]

X. Liu, N. Jiang, X. Xu, et al., Anti-hepatoma compound determination by the method of spectrum effect relationship, component knock-out, and UPLC-MS2 in Scheflera Heptaphylla (L.) Frodin harms and its mechanism, Front. Pharmacol. 11 (2020) 1342. https://doi.org/10.3389/fphar.2020.01342.

[38]

T. Franco-Ávila, R. Moreno-González, M.E. Juan, et al., Table olive elicits antihypertensive activity in spontaneously hypertensive rats, J. Sci. Food Agric. (2022). https://doi.org/10.1002/jsfa.12112.

[39]

C. Li, W. Chen, L. Zheng, et al., Ameliorative effect of ursolic acid on ochratoxin A-induced renal cytotoxicity mediated by Lonp1/Aco2/Hsp75, Toxicon. 168 (2019) 141-146. https://doi.org/10.1016/j.toxicon.2019.07.014.

[40]

A. Fatima, T.S. Malick, I. Khan, et al., Effect of glycyrrhizic acid and 18β-glycyrrhetinic acid on the differentiation of human umbilical cordmesenchymal stem cells into hepatocytes, World J. Stem. Cells 10 (2021) 1580-1594. https://doi.org/10.4252/wjsc.v13.i10.1580.

[41]

S.W. Xu, B.Y.K. Law, S.L.Q. Qu, et al., Serca and p-glycoprotein inhibition and ATP depletion are necessary for celastrol-induced autophagic cell death and collateral sensitivity in multidrug-resistant tumor cells, Pharmacol. Res.153 (2020) 104660. https://doi.org/10.1016/j.phrs.2020.104660.

[42]

Y. Li, X. Ye, X. Zheng, et al., Transcription factor Eb (Tfeb)-mediated autophagy protects against ethyl carbamate-induced cytotoxicity, J. Hazard Mater. 364 (2019) 281-292. https://doi.org/10.1016/j.jhazmat.2018.10.037.

[43]

G. Kallifatidis, J.J. Hoy, B.L. Lokeshwar, Bioactive natural products for chemoprevention and treatment of castration-resistant prostate cancer, Semin Cancer Biol. 40/41 (2016) 160-169. https://doi.org/10.1016/j.semcancer.2016.06.003.

[44]

G. Deng, C. Ma, H. Zhao, et al., Anti-edema and antioxidant combination therapy for ischemic stroke via glyburide-loaded betulinic acid nanoparticles, Theranostics 23 (2019) 6991-7002. https://doi.org/10.7150/thno.35791.

[45]

G.M. Burkart, F. Brandizzi, A tour of tor complex signaling in plants, Trends Biochem. Sci. 5 (2021) 417-428. https://doi.org/10.1016/j.tibs.2020.11.004.

[46]

W. Li, P. He, Y. Huang, et al., Selective autophagy of intracellular organelles: recent research advances, Theranostics 1 (2021) 222-256. https://doi.org/10.7150/thno.49860.

[47]

P.R. Davis, S.G. Miller, N.A. Verhoeven, et al., Increased AMP deaminase activity decreases ATP content and slows protein degradation in cultured skeletal muscle, Metabolism 108 (2020) 154257. https://doi.org/10.1016/j.metabol.2020.154257.

[48]

H. Kikuchi, E. Sasaki, N. Nomura, et al., Failure to sense energy depletion may be a novel therapeutic target in chronic kidney disease, Kidney Int. 1 (2019) 123-137. https://doi.org/10.1016/j.kint.2018.08.030.

[49]

I. Kim, M. Kim, M.K. Park, et al., The disubstituted adamantyl derivative LW1564 inhibits the growth of cancer cells by targeting mitochondrial respiration and reducing hypoxia-inducible factor (HIF)-1α accumulation, Exp. Mol. Med. 11 (2020) 1845-1856. https://doi.org/10.1038/s12276-020-00523-5.

[50]

K.K. Brown, AMPK Ca(R)Sts a new light on amino acid sensing, Embo. J. 21 (2021) e109575. https://doi.org/10.15252/embj.2021109575.

[51]

Q. Zeng, X. Ma, Y. Song, et al., Targeting regulated cell death in tumor nanomedicines, Theranostics 2 (2022) 817-841. https://doi.org/10.7150/thno.67932.

[52]

W. Gao, X. Wang, Y. Zhou, et al., Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy, Signal. Transduct. Target Ther. 1 (2022) 196. https://doi.org/10.1038/s41392-022-01046-3.

[53]

D.P. Del Re, D. Amgalan, A. Linkermann, et al., Fundamental mechanisms of regulated cell death and implications for heart disease, Physiol. Rev. 4 (2019) 1765-1817. https://doi.org/10.1152/physrev.00022.2018.

[54]

C. Zhang, X. Liu, S. Jin, et al., Ferroptosis in cancer therapy: a novel approach to reversing drug resistance, Mol. Cancer 1 (2022) 47. https://doi.org/10.1186/s12943-022-01530-y.

[55]

B. Zhou, J. Liu, R. Kang, et al., Ferroptosis is a type of autophagy-dependent cell death, Semin Cancer Biol. 66 (2020) 89-100. https://doi.org/10.1016/j.semcancer.2019.03.002.

File
fshw-13-3-1453_ESM1.doc (343.5 KB)
fshw-13-3-1453_ESM2.docx (816 KB)
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 25 September 2022
Revised: 11 October 2022
Accepted: 14 November 2022
Published: 08 February 2024
Issue date: May 2024

Copyright

© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

Acknowledgements

Acknowledgment

This work was supported by Henan Provincial Science and Technology Research Project (212102310355) and the National Natural Science Foundation of China (82020108024 and 32161143021).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return