scholarly journals Echinacoside exerts anti-tumor activity via the miR-503-3p/TGF-β1/Smad aixs in liver cancer

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Wen Li ◽  
Jing Zhou ◽  
Yajie Zhang ◽  
Jing Zhang ◽  
Xue Li ◽  
...  

Abstract Background Echinacoside (ECH) is the main active ingredient of Cistanches Herba, which is known to have therapeutic effects on metastatic tumors. However, the effects of ECH on liver cancer are still unclear. This study was to investigate the effects of ECH on the aggression of liver cancer cells. Methods Two types of liver cancer cells Huh7 and HepG2 were treated with different doses of ECH at different times and gradients. MTT and colony formation assays were used to determine the effects of ECH on the viability of Huh7 and HepG2 cells. Transwell assays and flow cytometry assays were used to detect the effects of ECH treatment on the invasion, migration, apoptosis and cell cycle of Huh7 and HepG2 cells. Western blot analysis was used to detect the effects of ECH on the expression levels of TGF-β1, smad3, smad7, apoptosis-related proteins (Caspase-3, Caspase-8), and Cyto C in liver cancer cells. The relationship between miR-503-3p and TGF-β1 was detected using bioinformatics analysis and Luciferase reporter assay. Results The results showed that ECH inhibited the proliferation, invasion and migration of Huh7 and HepG2 cells in a dose- and time-dependent manner. Moreover, we found that ECH caused Huh7 and HepG2 cell apoptosis by blocking cells in S phase. Furthermore, the expression of miR-503-3p was found to be reduced in liver tumor tissues, but ECH treatment increased the expression of miR-503-3p in Huh7 and HepG2 cells. In addition, we found that TGF-β1 was identified as a potential target of miR-503-3p. ECH promoted the activation of the TGF-β1/Smad signaling pathway and increased the expression levels of Bax/Bcl-2. Moreover, ECH could trigger the release of mitochondrial Cyto C, and cause the reaction Caspases grade. Conclusions This study demonstrates that ECH exerts anti-tumor activity via the miR-503-3p/TGF-β1/Smad aixs in liver cancer, and provides a safe and effective anti-tumor agent for liver cancer.

2021 ◽  
Vol 18 (3) ◽  
pp. 505-511
Author(s):  
Heze Yu ◽  
Geao Liang ◽  
Bo Dou ◽  
Jiangdong Ni

Purpose: To investigate the effect of cyclopentadione-aniline conjugate (CAC) on proliferation of liver cancer cells. Methods: MTT assay and flow cytometry were used for the determination of the effect of CAC on cell proliferation and apoptosis. Western blotting was used to measure the influence of CAC on the expressions of various proteins, while Matrigel-coated Transwell assay was used for assessment of cell invasion. Results: CAC inhibited proliferation of liver cancer cells in a concentration-dependent manner. The degree of proliferation of HepG2 cells was 98, 89, 76, 66, 51, 42 or 36 %, on treatment with 0.25, 0.5, 1.0, 1.5, 2.0, 2.5 or 3.0 µM CAC, respectively. In H4TG cells, treatment with 0.25, 0.5, 1.0, 1.5, 2.0, 2.5 and 3.0 µM CAC decreased proliferation of cells to 99, 91, 79, 70, 54, 46 and 40 %, respectively. Apoptosis was induced in 34.56, 37.37 and 52.98 % cells, on treatment with 2.0, 2.5 and 3.0 µM CAC, respectively. The invasive potential of HepG2 cells was significantly decreased by CAC (p < 0.05). Marked decreases were observed in Bcl-2, MMP-2, MMP-9, c-ERK1/2 and phospho-Akt levels in CACtreated HepG2 cells. CAC treatment markedly upregulated Bax and phospho ERK1/2, but significantly downregulated phospho PI3K, phospho mTOR and phospho Akt in HepG2 cells (p < 0.05). However, the level of phospho p38 was decreased in CAC-treated cells. Conclusion: These results demonstrate that CAC inhibits proliferation of liver cancer cells via apoptosis induction. Thus, CAC can potentially be used for the treatment of liver cancer in humans


Molecules ◽  
2020 ◽  
Vol 25 (12) ◽  
pp. 2883 ◽  
Author(s):  
Keiko Takagi ◽  
Yutaka Midorikawa ◽  
Tadatoshi Takayama ◽  
Hayato Abe ◽  
Kyoko Fujiwara ◽  
...  

Synthetic pyrrole-imidazole (PI) polyamides bind to the minor groove of double-helical DNA with high affinity and specificity, and inhibit the transcription of corresponding genes. In liver cancer, transforming growth factor (TGF)-β expression is correlated with tumor grade, and high-grade liver cancer tissues express epithelial-mesenchymal transition markers. TGF-β1 was reported to be involved in cancer development by transforming precancer cells to cancer stem cells (CSCs). This study aimed to evaluate the effects of TGF-β1-targeting PI polyamide on the growth of liver cancer cells and CSCs and their TGF-β1 expression. We analyzed TGF-β1 expression level after the administration of GB1101, a PI polyamide that targets human TGF-β1 promoter, and examined its effects on cell proliferation, invasiveness, and TGF-β1 mRNA expression level. GB1101 treatment dose-dependently decreased TGF-β1 mRNA levels in HepG2 and HLF cells, and inhibited HepG2 colony formation associated with downregulation of TGF-β1 mRNA. Although GB1101 did not substantially inhibit the proliferation of HepG2 cells compared to untreated control cells, GB1101 significantly suppressed the invasion of HLF cells, which displayed high expression of CD44, a marker for CSCs. Furthermore, GB1101 significantly inhibited HLF cell sphere formation by inhibiting TGF-β1 expression, in addition to suppressing the proliferation of HLE and HLF cells. Taken together, GB1101 reduced TGF-β1 expression in liver cancer cells and suppressed cell invasion; therefore, GB1101 is a novel candidate drug for the treatment of liver cancer.


Open Medicine ◽  
2019 ◽  
Vol 14 (1) ◽  
pp. 264-270 ◽  
Author(s):  
Jing Zhou ◽  
Yajun Li ◽  
Danhua Li ◽  
Zhi Liu ◽  
Jie Zhang

AbstractObjectiveAccumulating reports reveal that serving as an oncogenic factor LAMTOR5 is involved in the progression of many specific cancers. Glucose transporter 1 (GLUT1) is frequently identified in many cancers. However, it remains unexplored whether GLUT1 plays a role in LAMTOR5-enhanced liver cancer. Here, we aim to decipher the function of LAMTOR5 in the regulation of GLUT1 in liver cancer.MethodsThe effect of LAMTOR5 on GLUT1 was analyzed using Western blotting and RT-PCR assay. Dose-increased over-expression or silencing of LAMTOR5 was performed through transient transfection. LAMTOR5-activated GLUT1 promoter was revealed by luciferase reporter assay. The regulation of GLUT1 by LAMTOR5/NF-κB was examined via Western blotting and luciferase reporter assays.ResultsThe data showed that in liver cancer cells under the administration with dose-increased LAMTOR5, the level of mRNA and protein of GLUT1 was obviously raised. Our data revealed that the activities of GLUT1 promoter were induced by LAMTOR5. Then, we found that the elevation of GLUT 1 mediated by LAMTOR5 slowed when the inhibitor or siRNAs of NF-κB was introduced into the liver cancer cells. Conclusion. LAMTOR5 is responsible for the activation of GLUT1 via transcription factor NF-κB in liver cancer.


2020 ◽  
Vol 11 ◽  
Author(s):  
Silin Zhang ◽  
Meilan Zhang ◽  
Jiaxin Chen ◽  
Jiaqi Zhao ◽  
Jielin Su ◽  
...  

This study aimed to demonstrate that ginsenoside compound K (20 (S)-ginsenoside CK; CK) downregulates Bcl-2-associated transcription factor 1 (Bclaf1), which inhibits the hypoxia-inducible factor-1α (HIF-1α)-mediated glycolysis pathway to inhibit the proliferation of liver cancer cells. Treatment of hepatoma cells (Bel-7404 and Huh7) under hypoxic conditions with different concentrations of CK showed that CK inhibited the proliferation of hepatoma cells in a time- and concentration-dependent manner; furthermore, the ability of the cells to form colonies was reduced, and cell growth was blocked in the G0/G1 phase. CK promoted the degradation of HIF-1α ubiquitination in liver cancer cells by regulating the expression of HIF-1α and related ubiquitination proteins; moreover, it reduced the activity of key enzymes involved in glycolysis, the pressure of cellular glycolysis, and the rate of real-time ATP production, thereby inhibiting the glycolysis pathway. It also decreased the expression of Bclaf1 in hypoxic liver cancer cells and thus reduced the ability of Bclaf1 to bind to HIF-1α. CK treatment of Bel-7404 and Huh7 cells with CRISPR/Cas9-engineered knock out of Bclaf1 gene under hypoxic conditions further suppressed the expression of HIF-1α, promoted HIF-1α ubiquitination, and inhibited the glycolysis pathway. In a rat model of primary liver cancer induced by diethylnitrosamine, positron emission tomography and computed tomography scans showed that after CK administration, tumor tissue volumes were reduced and glucose uptake capacity decreased. Increased Bclaf1 and HIF-1α expression promoted the ubiquitination of HIF-1α and inhibited the glycolysis pathway, thereby inhibiting the proliferation of liver cancer cells. In summary, this study confirmed by in vitro and in vivo experiments that in hypoxic liver cancer cells CK downregulates the expression of Bclaf1, inhibits the HIF-1α-mediated glycolysis pathway, and inhibits cell proliferation, suggesting that the CK-mediated effects on Bclaf1 may represent a novel therapeutic approach for the treatment of liver cancer patients.


2016 ◽  
Vol 38 (5) ◽  
pp. 1897-1905 ◽  
Author(s):  
Chuanjie Yang ◽  
Quanxu Wang ◽  
Xiaodan Liu ◽  
Xiulian Cheng ◽  
Xiaoyu Jiang ◽  
...  

Objective: Radiation therapy, one of the major treatments for liver cancer, causes DNA damage and cell death. Since the liver cancer cells have a strong capacity to repair irradiative injury, new medicines to enhance this treatment are urgently required. In this study, we investigated the effect of NU7441, a synthetic small-molecule compound, as a specific inhibitor of DNA-dependent protein kinase (DNA-PK) in radiosensitization of hepatocellular carcinoma HepG2 cells. Methods: Cell Counting Kit-8 (CCK-8) was first used to evaluate the proliferation of HepG2 cells under NU7441 treatment. SDS-PAGE and Western blot were then performed to study the protein expression leading to the DNA damage repair. Further, neutral single cell gel electrophoresis and immunofluorescence assay were carried out to assess DNA repair. Finally, flow cytometry was implemented to examine the changes in cell cycle. Results: NU7441 reduced the CCK-8 counts in the HepG2 culture, further enhanced 60Coγ radiation injury to HepG2 cells, which was manifested by decreasing the DNA-PKcs (S2056) protein expression, increasing γH2AX foci number, prolonging the tail moment of the comet cells, and inducing cell cycle arrest at G2/M phase. Conclusion: NU7441 inhibited the growth of liver cancer cells, enhanced the radiosensitization of these cancer cells by interfering with the DNA repair and cell cycle checkpoint. These data implicate NU7441 as a potential radiotherapy sensitizer for the treatment of liver cancer.


2021 ◽  
Vol 13 (8) ◽  
pp. 1547-1556
Author(s):  
Juan He ◽  
Song Li ◽  
Junli Liu

The onset of the disease is secretive, and the accuracy of early diagnosis is not high. Patients will lose the opportunity of surgical treatment because of severe liver cirrhosis and cancer cell metastasis. Vanib nano-drug was prepared in this study by a polymer, which was used for in vitro research of liver cancer cells. First, polyethylene glycol (PEG)-poly lactic acid (PLA) and Vanib were dissolved in acetone to form a mixed solution, and then, water-soluble nano-particles were obtained by ultrasonic and deionized water. Then, the nano-particles were modified by iRGD and characterized by electron microscopy, DLS, and in vitro release. Besides, CCK-8, flow cytometry apoptosis test, flow cytometry cell cycle test, Transwell cell invasion and metastasis test, and angiogenesis test were applied to detect the effect of the nano-drug on the proliferation, cell metastasis, and angiogenesis of liver cancer cells. The results showed that Vanib nano-particles inhibited the proliferation of liver cancer cells in a dose-dependent manner, and induced apoptosis to G1 block. What’s more, Vanib nano-particles reduced cell viability in a Caspase-dependent manner, thereby inducing cell death. The Vanib nano-particles up-regulated the expression of P21 and also inhibited the cyclins Cyclin E2 and Cycllin B, thereby blocking the cell cycle process. In addition, Vanib nano-particles and free Vanib could inhibit the activity of human umbilical vein endothelial cells (HUVECs) and angiogenesis, which proved that the prepared Vanib nano-particles could effectively curb the growth of liver cancer cells.


2011 ◽  
Vol 110 (5) ◽  
pp. 326-335 ◽  
Author(s):  
Baoxiong Zhuang ◽  
Yangde Zhang ◽  
Jian Peng ◽  
Haowei Zhang ◽  
Tiehui Hu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document