4.5 Article

Tanshinone I induces cell apoptosis by reactive oxygen species-mediated endoplasmic reticulum stress and by suppressing p53/DRAM-mediated autophagy in human hepatocellular carcinoma

Journal

ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY
Volume 48, Issue 1, Pages 488-497

Publisher

TAYLOR & FRANCIS LTD
DOI: 10.1080/21691401.2019.1709862

Keywords

Hepatocellular carcinoma (HCC); apoptosis; cell cycle arrest; reactive oxygen species (ROS); autophagy; p53; DRAM; ER stress

Funding

  1. Sanming Project of Medicine in Shenzhen [SZSM201612021]
  2. Special Foundation for Science and Technology Development of Guangdong Province [2017B090904010]
  3. Scientific Research Project of Health and Family Planning Commission of Shenzhen Municipality [SZXJ2018086]

Ask authors/readers for more resources

Human hepatocellular carcinoma (HCC) is the most common type of liver cancer, and it has a high mortality rate. Despite surgical treatments, radiotherapy, and chemotherapy, the median survival of patients with advanced HCC is low. Evidence has shown that tanshinone (TA) I exhibits anti-proliferative activity against numerous cancers. However, the role of TA I and its mechanism in HCC remain unknown. Here, we determined the anti-cancer potential of TA I against HCC cell lines HepG2 and Huh7. Cell viability was analyzed using a Cell Counting Kit-8 assay. Flow cytometry was used to analyze cell cycles and apoptosis. Western blotting was used to detect protein expression and phosphorylation levels. TA I was found to inhibit cell proliferation, induce G0/G1 phase arrest, and trigger apoptosis in HepG2 and Huh7 cells. We further explored the molecular mechanism of TA I-mediated apoptosis. Our results showed that TA I induced G0/G1 phase arrest through downregulation of cyclin D1 expression and upregulation of p21 expression. TA I induced cell apoptosis via reactive oxygen species-mediated endoplasmic reticulum stress and by inhibiting p53/damage-regulated autophagy modulator (DRAM)-mediated autophagy in HepG2 and Huh7 cells. Therefore, TA I may be an anti-cancer drug candidate in the treatment of HCC.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available