Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin

Abstract Background Previous studies reported that emodin extracted from Rheum palmatum L. exerts antiproliferation and antimetastatic effects in a variety of human cancer types. However, the role of emodin in hepatocellular carcinoma (HCC) remain unknown. Methods EdU and colony formation assays wer...

Full description

Bibliographic Details
Main Authors: Binyu Qin, Zhili Zeng, Jianliang Xu, Jing Shangwen, Zeng Jie Ye, Shutang Wang, Yanheng Wu, Gongfeng Peng, Qi Wang, Wenyi Gu, Ying Tang
Format: Article
Language:English
Published: BMC 2022-06-01
Series:BMC Cancer
Subjects:
Online Access:https://doi.org/10.1186/s12885-022-09684-0
_version_ 1811343626570039296
author Binyu Qin
Zhili Zeng
Jianliang Xu
Jing Shangwen
Zeng Jie Ye
Shutang Wang
Yanheng Wu
Gongfeng Peng
Qi Wang
Wenyi Gu
Ying Tang
author_facet Binyu Qin
Zhili Zeng
Jianliang Xu
Jing Shangwen
Zeng Jie Ye
Shutang Wang
Yanheng Wu
Gongfeng Peng
Qi Wang
Wenyi Gu
Ying Tang
author_sort Binyu Qin
collection DOAJ
description Abstract Background Previous studies reported that emodin extracted from Rheum palmatum L. exerts antiproliferation and antimetastatic effects in a variety of human cancer types. However, the role of emodin in hepatocellular carcinoma (HCC) remain unknown. Methods EdU and colony formation assays were performed to evaluate the effects of emodin on proliferation. The mobility capacities of HCC treated with emodin were evaluated using wound healing assay. Transwell invasion and migration assays were performed to evaluate anti-migratory and anti-invasive effects of emodin on HCC. Annexin V-FITC/PI was performed to analyze the apoptosis. PI stain was performed to analyze cell cycle. RNA sequencing technology was used to identify the differentially expressed genes (DEGs) induced by emodin in HCC. The impact of emodin on autophagic flux in HepG2 cells was examined by mCherry-GFP-LC3 analysis. Western blot was used to assess the protein expressions of epithelial-mesenchymal transition (EMT), autophagy, PI3K/AKT/mTOR and Wnt/β-catenin signaling pathway. Results We found that emodin inhibited the growth of HepG2 cells in a dose- and time-dependent manner. In addition, emodin inhibited cell proliferation, induced S and G2/M phases arrest, and promoted apoptosis in HepG2 cells. The migration and invasion of HepG2 cells were also suppressed by emodin. Enrichment analysis revealed that DEGs involved in cell adhesion, cancer metastasis and cell cycle arrest. Moreover, western bolt results show that emodin-induced autophagy promotes Snail and β-catenin degradation. We also found that blocking autophagic flux after emodin treatment caused EMT reversal. Furthermore, the PI3K agonist Y-P 740 significantly reversed the phosphorylation levels of GSK3β and mTOR. These results indicated that emodin induced autophagy and inhibited the EMT in part through suppression of the PI3K/AKT/mTOR and Wnt/β-catenin pathways. Conclusion Our study indicated that emodin inhibited cell metastasis in HCC via the crosstalk between autophagy and EMT.
first_indexed 2024-04-13T19:32:15Z
format Article
id doaj.art-44a99bfcf56d477db881d2be575980a8
institution Directory Open Access Journal
issn 1471-2407
language English
last_indexed 2024-04-13T19:32:15Z
publishDate 2022-06-01
publisher BMC
record_format Article
series BMC Cancer
spelling doaj.art-44a99bfcf56d477db881d2be575980a82022-12-22T02:33:08ZengBMCBMC Cancer1471-24072022-06-0122111710.1186/s12885-022-09684-0Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-cateninBinyu Qin0Zhili Zeng1Jianliang Xu2Jing Shangwen3Zeng Jie Ye4Shutang Wang5Yanheng Wu6Gongfeng Peng7Qi Wang8Wenyi Gu9Ying Tang10Institute of Tumor, Guangzhou University of Chinese MedicineInstitute of Tumor, Guangzhou University of Chinese MedicineHepatobilliary Surgery Department, The Third affiliated Hospital of Su Yat-sen UniversityScience and Technology Innovation Center, Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineDepartment of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese MedicineGillion ITM Research Institute, Guangzhou HongkeyuanGillion ITM Research Institute, Guangzhou HongkeyuanScience and Technology Innovation Center, Guangzhou University of Chinese MedicineGillion ITM Research Institute, Guangzhou HongkeyuanInstitute of Tumor, Guangzhou University of Chinese MedicineAbstract Background Previous studies reported that emodin extracted from Rheum palmatum L. exerts antiproliferation and antimetastatic effects in a variety of human cancer types. However, the role of emodin in hepatocellular carcinoma (HCC) remain unknown. Methods EdU and colony formation assays were performed to evaluate the effects of emodin on proliferation. The mobility capacities of HCC treated with emodin were evaluated using wound healing assay. Transwell invasion and migration assays were performed to evaluate anti-migratory and anti-invasive effects of emodin on HCC. Annexin V-FITC/PI was performed to analyze the apoptosis. PI stain was performed to analyze cell cycle. RNA sequencing technology was used to identify the differentially expressed genes (DEGs) induced by emodin in HCC. The impact of emodin on autophagic flux in HepG2 cells was examined by mCherry-GFP-LC3 analysis. Western blot was used to assess the protein expressions of epithelial-mesenchymal transition (EMT), autophagy, PI3K/AKT/mTOR and Wnt/β-catenin signaling pathway. Results We found that emodin inhibited the growth of HepG2 cells in a dose- and time-dependent manner. In addition, emodin inhibited cell proliferation, induced S and G2/M phases arrest, and promoted apoptosis in HepG2 cells. The migration and invasion of HepG2 cells were also suppressed by emodin. Enrichment analysis revealed that DEGs involved in cell adhesion, cancer metastasis and cell cycle arrest. Moreover, western bolt results show that emodin-induced autophagy promotes Snail and β-catenin degradation. We also found that blocking autophagic flux after emodin treatment caused EMT reversal. Furthermore, the PI3K agonist Y-P 740 significantly reversed the phosphorylation levels of GSK3β and mTOR. These results indicated that emodin induced autophagy and inhibited the EMT in part through suppression of the PI3K/AKT/mTOR and Wnt/β-catenin pathways. Conclusion Our study indicated that emodin inhibited cell metastasis in HCC via the crosstalk between autophagy and EMT.https://doi.org/10.1186/s12885-022-09684-0EmodinHepatocellular carcinomaAutophagyEpithelial-mesenchymal transitionPI3K/AKT/mTORWnt/β-catenin
spellingShingle Binyu Qin
Zhili Zeng
Jianliang Xu
Jing Shangwen
Zeng Jie Ye
Shutang Wang
Yanheng Wu
Gongfeng Peng
Qi Wang
Wenyi Gu
Ying Tang
Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
BMC Cancer
Emodin
Hepatocellular carcinoma
Autophagy
Epithelial-mesenchymal transition
PI3K/AKT/mTOR
Wnt/β-catenin
title Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
title_full Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
title_fullStr Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
title_full_unstemmed Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
title_short Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin
title_sort emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy mediated degradation of snail and β catenin
topic Emodin
Hepatocellular carcinoma
Autophagy
Epithelial-mesenchymal transition
PI3K/AKT/mTOR
Wnt/β-catenin
url https://doi.org/10.1186/s12885-022-09684-0
work_keys_str_mv AT binyuqin emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT zhilizeng emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT jianliangxu emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT jingshangwen emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT zengjieye emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT shutangwang emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT yanhengwu emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT gongfengpeng emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT qiwang emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT wenyigu emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin
AT yingtang emodininhibitsinvasionandmigrationofhepatocellularcarcinomacellsviaregulatingautophagymediateddegradationofsnailandbcatenin