Journal Home > Volume 12 , Issue 4

To explore the function of licochalcone A as an anticancer phytochemical on HepG2 cells and investigate its potential mechanisms, we analyzed the microRNAs (miRNAs) expression profile of HepG2 cells in response to licochalcone A (70 μmol/L) in vitro. 102 dysregulated miRNAs were detected, and SP1 was expected as the transcription factor that regulates the functions of most screened miRNAs. A sum of 431 targets, the overlap of predicted mRNAs from TargetScan, miRDB, and miRtarbase were detected as the targets for these dysregulated miRNAs. FoxO signaling pathway was the hub pathway for the targets. A protein-protein interaction network was structured on the STRING platform to discover the hub genes. Among them, PIK3R1, CDC42, ESR1, SMAD4, SUMO1, KRAS, AGO1, etc. were screened out. Afterwards, the miRNA-target networks were established to screen key dysregulated miRNAs. Two key miRNAs (hsa-miR-133b and hsa-miR-145-5p) were filtered. Finally, the miRNA-target-transcription factor networks were constructed for these key miRNAs. The networks for these key miRNAs included three and two transcription factors, respectively. These identified miRNAs, transcription factors, targets, and regulatory networks may offer hints to understand the molecular mechanism of licochalcone A as a natural anticarcinogen.


menu
Abstract
Full text
Outline
About this article

The anti-cancerous mechanism of licochalcone A on human hepatoma cell HepG2 based on the miRNA omics

Show Author's information Jun Wanga,b,1Xiuxiu Zhangb,1Zhijing NicElnur ElamcKiran Thakurb,cKexin LiaChuyan WangaJianguo Zhangb,c( )Zhaojun Weib,c( )
School of Biological Food and Environment, Hefei University, Hefei 230601, China
School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan 750021, China

1 Co-fi rst author.Peer review under responsibility of KeAi Communications Co., Ltd.]]>

Abstract

To explore the function of licochalcone A as an anticancer phytochemical on HepG2 cells and investigate its potential mechanisms, we analyzed the microRNAs (miRNAs) expression profile of HepG2 cells in response to licochalcone A (70 μmol/L) in vitro. 102 dysregulated miRNAs were detected, and SP1 was expected as the transcription factor that regulates the functions of most screened miRNAs. A sum of 431 targets, the overlap of predicted mRNAs from TargetScan, miRDB, and miRtarbase were detected as the targets for these dysregulated miRNAs. FoxO signaling pathway was the hub pathway for the targets. A protein-protein interaction network was structured on the STRING platform to discover the hub genes. Among them, PIK3R1, CDC42, ESR1, SMAD4, SUMO1, KRAS, AGO1, etc. were screened out. Afterwards, the miRNA-target networks were established to screen key dysregulated miRNAs. Two key miRNAs (hsa-miR-133b and hsa-miR-145-5p) were filtered. Finally, the miRNA-target-transcription factor networks were constructed for these key miRNAs. The networks for these key miRNAs included three and two transcription factors, respectively. These identified miRNAs, transcription factors, targets, and regulatory networks may offer hints to understand the molecular mechanism of licochalcone A as a natural anticarcinogen.

Keywords: HepG2 cells, Licochalcone A, Dysregulated miRNAs, Transcription factors, Targets, Regulatory networks

References(67)

[1]

C. Allemani, H.K. Weir, H. Carreira, et al., Global surveillance of cancer survival 1995–2009: analysis of individual data for 25676887 patients from 279 population-based registries in 67 countries (CONCORD-2), Lancet 385 (2015) 977-1010. https://doi.org/10.1016/S0140-6736(14)62038-9.

[2]

A. Ely, K. Bloom, M.B. Maepa, et al., Recent update on the role of circular RNAs in hepatocellular carcinoma, J. Hepatocell. Carcinoma. 8 (2021) 1-17. https://doi.org/10.2147/JHC.S268291.

[3]

S. Li, A. Saviano, D.J. Erstad, et al., Risk factors, pathogenesis, and strategies for hepatocellular carcinoma prevention: emphasis on secondary prevention and its translational challenges, J. Clin. Med. 9 (2020) 3817. https://doi.org/10.3390/jcm9123817.

[4]

M. Abdul-Latif, K. Townsend, C. Dearman, et al., Immunotherapy in gastrointestinal cancer: the current scenario and future perspectives, Cancer Treat. Rev. 88 (2020) 102030. https://doi.org/10.1016/j.ctrv.2020.102030.

[5]

Z. Cheng, J. Lin, N. Gao, et al., Blueberry malvidin-3-galactoside modulated gut microbial dysbiosis and microbial TCA cycle KEGG pathway disrupted in a liver cancer model induced by HepG2 cells, Food Sci. Hum. Wellness 9 (2020) 245-255. https://doi.org/10.1016/j.fshw.2020.04.006.

[6]

C. Wang, L. Chen, C. Xu, et al., A comprehensive review for phytochemical, pharmacological, and biosynthesis studies on Glycyrrhiza spp., Am. J. Chin. Med. 48 (2020) 17-45. https://doi.org/10.1142/S0192415X20500020.

[7]

S. Li, G. Chen, C. Zhang, et al., Research progress of natural antioxidants in foods for the treatment of diseases, Food Sci. Hum. Wellness 3 (2014) 110-116. https://doi.org/10.1142/S0192415X20500020.

[8]

W.C. Huang, C.Y. Liu, S.C. Shen, et al., Protective effects of licochalcone A improve airway hyper-responsiveness and oxidative stress in a mouse model of asthma, Cells 8 (2019) 617. https://doi.org/10.3390/cells8060617.

[9]

C.J. Liou, Y.K. Lee, N.C. Ting, et al., Protective effects of licochalcone A ameliorates obesity and non-alcoholic fatty liver disease via promotion of the Sirt-1/AMPK pathway in mice fed a high-fat diet, Cells 8 (2019) 447. https://doi.org/10.3390/cells8050447.

[10]

K.S. de Freitas, I.S. Squarisi, N.O. Acésio, et al., Licochalcone A, a licorice flavonoid: antioxidant, cytotoxic, genotoxic, and chemopreventive potential, J. Toxicol. Environ. Health A 83 (2020) 673-686. https://doi.org/10.1080/15287394.2020.1813228.

[11]

Z.F. Wang, J. Liu, Y.A. Yang, et al., A review: the anti-inflammatory, anticancer and antibacterial properties of four kinds of licorice flavonoids isolated from licorice, Curr. Med. Chem. 27 (2020) 1997-2011. https://doi.org/10.2174/0929867325666181001104550.

[12]

Y. Fu, T.C. Hsieh, J. Guo, et al., Licochalcone-A, a novel flavonoid isolated from licorice root (Glycyrrhiza glabra), causes G2 and late-G1 arrests in androgen-independent PC-3 prostate cancer cells, Biochem. Biophys. Res. Commun. 322 (2004) 263-270. https://doi.org/10.1016/j.bbrc.2004.07.094.

[13]

R.C. Lin, S.F. Yang, H.L. Chiou, et al., Licochalcone A-induced apoptosis through the activation of p38 MAPK pathway mediated mitochondrial pathways of apoptosis in human osteosarcoma cells in vitro and in vivo, Cells 8 (2019) 1441. https://doi.org/10.3390/cells8111441.

[14]

C. Qiu, T. Zhang, W. Zhang, et al., Licochalcone A inhibits the proliferation of human lung cancer cell lines A549 and H460 by inducing G2/M cell cycle arrest and ER stress, Int. J. Mol. Sci. 18 (2017) 1761. https://doi.org/10.3390/ijms18081761.

[15]

W.C. Huang, H.H. Su, L.W. Fang, et al., Licochalcone A inhibits cellular motility by suppressing E-cadherin and MAPK signaling in breast cancer, Cells 8 (2019) 218. https://doi.org/10.3390/cells8030218.

[16]

J. Wang, Y.S. Zhang, K. Thakur, et al., Licochalcone A from licorice root, an inhibitor of human hepatoma cell growth via induction of cell apoptosis and cell cycle arrest, Food Chem. Toxicol. 120 (2018) 407-417. https://doi.org/10.1016/j.fct.2018.07.044.

[17]

S.M. Hammond, An overview of microRNAs, Adv. Drug Deliv. Rev. 87 (2015) 3-14. https://doi.org/10.1016/j.addr.2015.05.001.

[18]

S.L. Anwar, U. Lehmann, MicroRNAs: emerging novel clinical biomarkers for hepatocellular carcinomas, J. Clin. Med. 4 (2015) 1631-1650. https://doi.org/10.3390/jcm4081631.

[19]

B. Zhang, L. Tian, J. Xie, et al., Targeting miRNAs by natural products: a new way for cancer therapy, Biomed. Pharmacother. 130 (2020) 110546. https://doi.org/10.1016/j.biopha.2020.110546.

[20]

J. Wang, C.Y. Wang, Integrated miRNA and mRNA omics reveal the anti-cancerous mechanism of licochalcone B on human hepatoma cell HepG2, Food Chem. Toxicol. 120 (2021) 112097. https://doi.org/10.1016/j.fct.2021.112096.

[21]

D.H. Kim, H. Khan, H. Ullah, et al., MicroRNA targeting by quercetin in cancer treatment and chemoprotection, Pharmacol. Res. 147 (2019) 104346. https://doi.org/10.1016/j.phrs.2019.104346.

[22]

S.S. Hussain, F. Zhang, Y. Zhang, et al., Stevenleaf from Gynostemma Pentaphyllum inhibits human hepatoma cell (HepG2) through cell cycle arrest and apoptotic induction, Food Sci. Hum. Wellness 9 (2020) 295-303. https://doi.org/10.1016/j.fshw.2020.04.011.

[23]

F. Zhang, Z. Ni, L. Ye, et al., Asparanin A inhibits cell migration and invasion in human endometrial cancer via Ras/ERK/MAPK pathway, Food Chem. Toxicol. 150 (2021) 112036. https://doi.org/10.1016/j.fct.2021.112036.

[24]

M. Pathan, S. Keerthikumar, C.S. Ang, et al., FunRich: an open access standalone functional enrichment and interaction network analysis tool, Proteomics 15 (2015) 2597-2601. https://doi.org/10.1002/pmic.201400515.

[25]

G. Bindea, B. Mlecnik, H. Hackl, et al., ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks, Bioinformatics 25 (2009) 1091-1093. https://doi.org/10.1093/bioinformatics/btp101.

[26]

F. Zhang, Y. Zhang, R. Ma, et al., Multi-omics reveals the anticancer mechanism of asparagus saponin-asparanin A on endometrial cancer Ishikawa cells, Food Funct. 12 (2021) 614-632. https://doi.org/10.1039/D0FO02265A.

[27]

C.H. Chin, S.H. Chen, H.H. Wu, et al., cytoHubba: identifying hub objects and sub-networks from complex interactome, BMC Syst. Biol. 8 (2014) 511. https://doi.org/10.1186/1752-0509-8-S4-S11.

[28]

Y. Gao, S. Zhang, Y. Zhang, et al., Identification of microRNA-target gene-transcription factor regulatory networks in colorectal adenoma using microarray expression data, Front. Genet. 11 (2020) 434. https://doi.org/10.3389/fgene.2020.00463.

[29]

R.S. Janky, A. Verfaillie, H. Imrichová, et al., iRegulon: from a gene list to a gene regulatory network using large motif and track collections, PLoS Comput. Biol. 10 (2014) e1003731. https://doi.org/10.1371/journal.pcbi.1003731.

[30]

C. Dai, Y. Xie, X. Zhuang, et al., MiR-206 inhibits epithelial ovarian cancer cells growth and invasion via blocking c-Met/AKT/mTOR signaling pathway, Biomed. Pharmacother. 104 (2018) 763-770. https://doi.org/10.1016/j.biopha.2018.05.077.

[31]

P. Deng, K. Li, F. Gu, et al., LINC00242/miR-1-3p/G6PD axis regulates Warburg effect and affects gastric cancer proliferation and apoptosis, Mol. Med. 27 (2021) 9. https://doi.org/10.1186/s10020-020-00259-y.

[32]

H. Zhang, Z. Zhang, L. Gao, et al., miR-1-3p suppresses proliferation of hepatocellular carcinoma through targeting SOX9, OncoTargets Ther. 12 (2019) 2149-2157. https://doi.org/10.2147/OTT.S197326.

[33]

Y. Liao, C. Wang, Z. Yang, et al., Dysregulated Sp1/miR-130b-3p/HOXA5 axis contributes to tumor angiogenesis and progression of hepatocellular carcinoma, Theranostics 10 (2020) 5209-5224. https://doi.org/10.7150/thno.43640.

[34]

X. Xie, S. Li, Y. Zhu, et al., Egr-1 mediates leptin-induced PPARγ reduction and proliferation of pulmonary artery smooth muscle cells, Mol. Biol. Cell 29 (2018) 356-362. https://doi.org/10.1091/mbc.e17-03-0141.

[35]

C.Z. Zhang, S.L. Chen, C.H. Wang, et al., CBX8 exhibits oncogenic activity via AKT/β-Catenin activation in hepatocellular carcinoma, Cancer Res. 78 (2018) 51-63. https://doi.org/10.1158/0008-5472.CAN-17-0700.

[36]

Y. Li, P. Li, N. Wang, Effect of let-7c on the PI3K/Akt/FoxO signaling pathway in hepatocellular carcinoma, Oncol. Lett. 21 (2021) 96. https://doi.org/10.3892/ol.2020.12357.

[37]

H. Luo, E. Hao, D. Tan, et al., Apoptosis effect of Aegiceras corniculatum on human colorectal cancer via activation of FoxO signaling pathway, Food Chem. Toxicol. 134 (2019) 110861. https://doi.org/10.1016/j.fct.2019.110861.

[38]

S. Shukla, F. Rizvi, S. Raisuddin, et al., FoxO proteins' nuclear retention and BH3-only protein Bim induction evoke mitochondrial dysfunction-mediated apoptosis in berberine-treated HepG2 cells, Free Radicals Biol. Med. 76 (2014) 185-199. https://doi.org/10.1016/j.freeradbiomed.2014.07.039.

[39]

Q. Wang, W.N. Yu, X. Chen, et al., Spontaneous hepatocellular carcinoma after the combined deletion of Akt isoforms, Cancer Cell 29 (2016) 523-535. https://doi.org/10.1016/j.ccell.2016.02.008.

[40]

H. Ding, Y. Wang, H. Zhang, CCND1 silencing suppresses liver cancer stem cell differentiation and overcomes 5-Fluorouracil resistance in hepatocellular carcinoma, J. Pharmacol. Sci. 143 (2020) 219-225. https://doi.org/10.1016/j.jphs.2020.04.006.

[41]

A. Adamek, A. Kasprzak, Insulin-like growth factor (IGF) system in liver diseases, Int. J. Mol. Sci. 19 (2018) 1308. https://doi.org/10.3390/ijms19051308.

[42]

R. Urtasun, M. Elizalde, M. Azkona, et al., Splicing regulator SLU7 preserves survival of hepatocellular carcinoma cells and other solid tumors via oncogenic miR-17-92 cluster expression, Oncogene 35 (2016) 4719-4729. https://doi.org/10.1038/onc.2015.517.

[43]

Y. Shi, D.D. Zhang, J.B. Liu, et al., Comprehensive analysis to identify DLEU2L/TAOK1 axis as a prognostic biomarker in hepatocellular carcinoma, Mol. Ther. Nucleic Acids 23 (2021) 702-718. https://doi.org/10.1016/j.omtn.2020.12.016.

[44]

W. Li, J. Xiao, X. Zhou, et al., STK4 regulates TLR pathways and protects against chronic inflammation-related hepatocellular carcinoma, J. Clin. Invest. 125 (2015) 4239-4254. https://doi.org/10.1172/JCI81203.

[45]

H. Ye, C. Zhang, B.J. Wang, et al., Synergistic function of Kras mutation and HBx in initiation and progression of hepatocellular carcinoma in mice, Oncogene 33 (2014) 5133-5138. https://doi.org/10.1038/onc.2013.468.

[46]

B. Wang, T. Xu, J. Liu, et al., Overexpression of activated Cdc42-associated kinase1 (Ack1) predicts tumor recurrence and poor survival in human hepatocellular carcinoma, Pathol. Res. Pract. 210 (2014) 787-792. https://doi.org/10.1016/j.prp.2014.09.014.

[47]

M. Wang, L. Zhang, Z. Liu, et al., AGO1 may influence the prognosis of hepatocellular carcinoma through TGF-β pathway, Cell Death Dis. 9 (2018) 324. https://doi.org/10.1038/s41419-018-0338-y.

[48]

N. Honma, T. Genda, Y. Matsuda, et al., MEK/ERK signaling is a critical mediator for integrin-induced cell scattering in highly metastatic hepatocellular carcinoma cells, Lab. Invest. 86 (2006) 687-696. https://doi.org/10.1038/labinvest.3700427.

[49]

J. Li, Y. Zhou, Y. Liu, et al., Sorafenib inhibits caspase-1 expression through suppressing TLR4/stat3/SUMO1 pathway in hepatocellular carcinoma, Cancer Biol. Ther. 19 (2018) 1057-1064. https://doi.org/10.1080/15384047.2018.1480280.

[50]

L. Wang, M. Cui, D. Cheng, et al., miR-9-5p facilitates hepatocellular carcinoma cell proliferation, migration and invasion by targeting ESR1, Mol. Cell. Biochem. 476 (2021) 575-583. https://doi.org/10.1007/s11010-020-03927-z.

[51]

P. Seshacharyulu, M.P. Ponnusamy, D. Haridas, et al., Targeting the EGFR signaling pathway in cancer therapy, Expert Opin. Ther. Targets. 16 (2012) 15-31. https://doi.org/10.1517/14728222.2011.648617.

[52]

Y.H. He, M.F. Li, X.Y. Zhang, et al., NLRC5 promotes cell proliferation via regulating the AKT/VEGF-A signaling pathway in hepatocellular carcinoma, Toxicology 359/360 (2016) 47-57. https://doi.org/10.1016/j.tox.2016.06.012.

[53]

M. Ma, L. Li, F. Long, et al., MiR-133b inhibits colorectal cancer metastasis via lncRNA-LUCAT1, Future Oncol. 17 (2021) 1013-1023. https://doi.org/10.2217/fon-2020-0420.

[54]

Z. Tian, H. Jiang, Y. Liu, et al., MicroRNA-133b inhibits hepatocellular carcinoma cell progression by targeting Sirt1, Exp. Cell Res. 343 (2016) 135-147. https://doi.org/10.1016/j.yexcr.2016.03.027.

[55]

W. Zeng, J.F. Zhu, J.Y. Liu, et al., miR-133b inhibits cell proliferation, migration and invasion of esophageal squamous cell carcinoma by targeting EGFR, Biomed. Pharmacother. 111 (2019) 476-484. https://doi.org/10.1016/j.biopha.2018.12.057.

[56]

C. Lei, F. Du, L. Sun, et al., miR-143 and miR-145 inhibit gastric cancer cell migration and metastasis by suppressing MYO6, Cell Death Dis. 8 (2017) e3101. https://doi.org/10.1038/cddis.2017.493.

[57]

A. Pagliuca, C. Valvo, E. Fabrizi, et al., Analysis of the combined action of miR-143 and miR-145 on oncogenic pathways in colorectal cancer cells reveals a coordinate program of gene repression, Oncogene 32 (2013) 4806-4813. https://doi.org/10.1038/onc.2012.495.

[58]

X. Yan, X. Chen, H. Liang, et al., miR-143 and miR-145 synergistically regulate ERBB3 to suppress cell proliferation and invasion in breast cancer, Mol. Cancer 13 (2014) 220. https://doi.org/10.1186/1476-4598-13-220.

[59]

S.B. Villadsen, J.B. Bramsen, M.S. Ostenfeld, et al., The miR-143/-145 cluster regulates plasminogen activator inhibitor-1 in bladder cancer, Br. J. Cancer 106 (2012) 366-374. https://doi.org/10.1038/bjc.2011.520.

[60]

G. Dong, S. Zhang, S. Shen, et al., SPATS2, negatively regulated by miR-145-5p, promotes hepatocellular carcinoma progression through regulating cell cycle, Cell Death Dis. 11 (2020) 837. https://doi.org/10.1038/s41419-020-03039-y.

[61]

J.C. To, A.P. Chiu, B.R. Tschida, et al., ZBTB20 regulates WNT/CTNNB1 signalling pathway by suppressing PPARG during hepatocellular carcinoma tumourigenesis, JHEP Reports 3 (2021) 100223. https://doi.org/10.1016/j.jhepr.2020.100223.

[62]

M. Dai, J. Boudreault, N. Wang, et al., Differential regulation of cancer progression by CDK4/6 plays a central role in DNA replication and repair pathways, Cancer Res. 3 (2020) 2121. https://doi.org/10.1158/0008-5472.CAN-20-2121.

[63]

S. Han, Y. Wang, J. Ma, et al., Sulforaphene inhibits esophageal cancer progression via suppressing SCD and CDH3 expression, and activating the GADD45B-MAP2K3-p38-p53 feedback loop, Cell Death Dis. 11 (2020) 713. https://doi.org/10.1038/s41419-020-02859-2.

[64]

X.X. Kong, Y.R. Lv, L.P. Shao, et al., HBx-induced MiR-1269b in NF-κB dependent manner upregulates cell division cycle 40 homolog (CDC40) to promote proliferation and migration in hepatoma cells, J. Transl. Med. 14 (2016) 189. https://doi.org/10.1186/s12967-016-0949-y.

[65]

C. Zhuang, Y. Liu, S. Fu, et al., Silencing of lncRNA MIR497HG via CRISPR/Cas13d induces bladder cancer progression through promoting the crosstalk between Hippo/Yap and TGF-β/Smad signaling, Front. Mol. Biosci. 7 (2020) 616768. https://doi.org/10.3389/fmolb.2020.616768.

[66]

J.J. Zheng, Q.Y. Que, H.T. Xu, et al., Hypoxia activates SOX5/WNT/β-Catenin signaling by suppressing MiR-338-3p in gastric cancer, Technol. Cancer Res. Treat. 19 (2020) 1-9. https://doi.org/10.1177/1533033820905825.

[67]

J. Organista-Nava, Y. Gómez-Gómez, B. Illades-Aguiar, et al., Regulation of the miRNA expression by TEL/AML1, BCR/ABL, MLL/AF4 and TCF3/PBX1 oncoproteins in acute lymphoblastic leukemia (review, Oncol. Rep. 36 (2016) 1226-1232. https://doi.org/10.3892/or.2016.4948.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 21 March 2021
Revised: 27 March 2021
Accepted: 31 March 2021
Published: 18 November 2022
Issue date: July 2023

Copyright

© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

This study was supported by the Hefei University Scientific Research and Development Fund (20ZR09ZDB), the talent fund of Hefei University (20RC48), the University Natural Sciences Research Project of Anhui Province (KJ2021A1009), the Major Projects of Science and Technology in Anhui Province (201903a06020021, 202004a06020042, 202004a06020052, 201904a06020008), and the National Natural Science Foundation of China (31850410476).

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