Abstract

Berberine (BBR), a natural alkaloid derived from Coptis, has anticancer activity. Some researchers have found that it could restrain epithelial-mesenchymal transition (EMT) of melanoma, neuroblastoma, and other tumor cells. However, it is unclear whether BBR can reverse EMT in hepatocellular carcinoma (HCC) and gastric carcinoma (GC). In our study, BBR inhibited the migration and invasion of HepG2, MGC803, and SGC7901 cells in a dose-dependent manner. Transcription sequencing assays showed that Vimentin, MMP, and Smad3 were downregulated, but Smad2, Smad6, TAB2, ZO-1, and claudin 7 were upregulated when treated with BBR. GO Enrichment analysis of KEGG pathway showed that BBR significantly inhibited TGF-β/Smad at 12 h, then, PI3K/Akt and Wnt/β-catenin signaling pathways at 24 h, which were closely related to the proliferation, migration, and EMT. The results of the transcriptome sequencing analysis were verified by Western Blot. It showed that the expression of epithelial marker E-cadherin and ZO-1 remarkably augmented with BBR treatment, as well as declined mesenchymal markers, including N-cadherin and Vimentin, decreased transcription factor Snail and Slug. The effects of BBR were similar to those of the PI3K inhibitor LY294002 and TGF-β receptor inhibitor SB431542. Furthermore, β-catenin and phosphorylation of AKT, Smad2, and Smad3 were changed dose-dependently by BBR treatment, which upregulated p-Smad2 and downregulated the others. Combined with LY or SB, respectively, BBR could enhance the effects of the two inhibitors. Simultaneously, IGF-1 and TGF-β, which is the activator of PI3K/AKT and TGF-β/Smad, respectively, could reverse the anti-EMT effect of BBR. The Molecular Docking results showed BBR had a high affinity with the TGF-β receptor I (TGFβR1), and the binding energy was -7.5 kcal/mol, which is better than the original ligand of TGFβR1. Although the affinity of BBR with TGF-β receptor II (TGFβR2) was lower than the original ligand of TGFβR2, the more considerable negative binding energy (−8.54 kcal/mol) was obtained. BBR upregulated p-Smad2, which was different from other reports, indicating that the function of Smad2 was relatively complex. Combination BBR with SB could enhance the effect of the inhibitor on EMT, and the results indicated that BBR binding to TGFβR was not competitive with SB to TGFβR since different binding amino acid sites. Our experiments demonstrated BBR increased p-Smad2 and decreased p-Smad3 by binding to TGFβR1 and TGβFR2 inhibiting TGF-β/Smad, then, PI3K/AKT and other signaling pathways to restrain EMT, metastasis, and invasion in tumor cells. The effect of BBR was similar on the three tumor cells.

Highlights

  • hepatocellular carcinoma (HCC) and gastric carcinoma (GC) are the most life-threatening tumor [1,2,3]

  • To select the benefiting concentration in the following assays, the effects on cell viability of the candidate concentrations of BBR (10, 20, 40, 80, 160, and 320 μM) in HepG2, MGC803, and SGC7901 cells were detected by MTT [3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazoliumbromide] experiment

  • Cells were preincubated with LY (PI3K/Akt inhibitor), or SB (TGFβR inhibitor), or IGF-1(PI3K/Akt agonist), or TGF β (TGFβR agonist) for 12 h, and treated alone or cotreated with BBR (40 μM) for 24 h, the proteins of epithelial-mesenchymal transition (EMT) was similar to the result of 2.4 when treated with BBR alone

Read more

Summary

Introduction

HCC and GC are the most life-threatening tumor [1,2,3]. Advanced HCC and GC remain poor prognosis of patients, mainly due to cancer metastasis, of which the mechanism is unclear. It is demonstrated that tumor metastasis could be inhibited by reversing the EMT process and restraining EMT activation to improve the prognosis of cancer patients [9,10,11,12,13,14]. Our manuscript is aimed at exploring whether BBR has the effect of reversing EMT, antimetastasis in HCC and GC, and the molecular mechanism of TGF-β/Smad pathway. TGFβ/Smad pathway has been widely divergent and puzzled It was described as inhibiting tumor in early phase and promoting cancer in advanced phase [34]. We selected liver and gastric cancer to study the effects of BBR on migration, invasion, EMT, and TGF-β/Smad signaling pathway, so as to provide a reference for clinical application

Materials and Methods
Results
Discussion
Conflicts of Interest
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.