Abstract

Cancer cells rewire their metabolism to satisfy the demands of uncontrolled proliferation and survival. The reprogramming of lipid metabolism supports tumor growth, metastasis, and therapy-resistance. Therefore, targeting lipid metabolic reprogramming is a potential cancer treatment strategy. We recently isolated the novel natural triterpene GL22 from Ganoderma leucocontextum, a traditional Chinese medicine. Here, we show that GL22 significantly inhibits the growth of the liver cancer cell line Huh7.5 in vitro and of Huh7.5-derived tumor xenografts in vivo. We further find that GL22 induces mitochondrial dysfunction and cell death in Huh7.5 cells, in part due to fatty acid immobilization and loss of the mitochondrial lipid cardiolipin, which has vital structural and metabolic functions. Importantly, we demonstrate that GL22 treatment decreases the expression of fatty acid-binding proteins (FABPs), which likely underlies the loss of cardiolipin, mitochondrial dysfunction, and cell death. The over-expressions of FABPs prevented the GL22-induced cell death, loss of cardiolipin, decrease of ATP production, and reduction of oxygen consumption rate in Huh7.5 cells. Our results support targeting lipid metabolism via manipulating FABPs as a cancer treatment strategy, and promote Chinese medicine as an important source of novel anticancer drugs.

Highlights

  • Despite recent improvements in treatment strategies, cancer remains one of the leading causes of death worldwide

  • To further evaluate the bioactivity of GL22, a triterpenefarnesyl hydroquinone hybrid isolated from G. leucocontextum (Fig. 1a), we determined if it would affect the growth of the liver cancer cell line Huh7.5

  • We further evaluated the therapeutic potential of GL22 on Huh7.5 cell xenografts in BALB/c-nu mice

Read more

Summary

Introduction

Despite recent improvements in treatment strategies, cancer remains one of the leading causes of death worldwide. We show that GL22 treatment potently inhibits liver cancer growth in vitro and in vivo by suppressing the expression of FABPs, which leads to FA immobilization and loss of cardiolipin, mitochondrial dysfunction and cell death.

Results
Conclusion
Full Text
Published version (Free)

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