1. Academic Validation
  2. Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation

Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation

  • Cell Death Dis. 2019 Jun 11;10(6):456. doi: 10.1038/s41419-019-1653-7.
Haitao Pan 1 Yujie Wang 1 Kun Na 1 Ying Wang 1 Lu Wang 1 Zhenhao Li 2 Chengjie Guo 1 Dandan Guo 1 Xingya Wang 3
Affiliations

Affiliations

  • 1 Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China.
  • 2 Zhejiang Shouxiangu Institute of Rare Medicine Plant, 12, Huanglong 3rd Road, 321200, Wuyi, Zhejiang, China.
  • 3 Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China. [email protected].
Abstract

Targeting Autophagy may serve as a promising strategy for Cancer therapy. Ganoderma lucidum polysaccharide (GLP) has been shown to exert promising anti-cancer effects. However, the underlying mechanisms remain elusive. Whether GLP regulates Autophagy in Cancer has never been reported. In this study, GLP induced the initiation of Autophagy in colorectal Cancer (CRC) HT-29 and HCT116 cells, as evidenced by enhanced level of LC3-II protein, GFP-LC3 puncta, and increased formation of double membrane vacuoles. However, GLP treatment caused marked increase of p62 expression. Addition of late stage Autophagy Inhibitor, chloroquine (CQ), further enhanced LC3-II and p62 level, as well as increased autophagosome accumulation, suggesting a blockage of autophagic flux by GLP in CRC cells. We then found GLP blocked autophagosome and lysosome fusion as determined by mRFP-GFP-LC3 colocalization analysis. Mechanistic study revealed that GLP-induced disruption of autophagosome-lysosome fusion is due to reduced lysosome acidification and lysosomal Cathepsin activities. Cell viability and flow cytometry assays revealed that GLP-induced autophagosome accumulation is responsible for GLP-induced Apoptosis in CRC cells. In line with this, inhibition of Autophagy initiation by 3-methyladenine (3-MA), an early stage Autophagy Inhibitor, attenuated GLP-induced Apoptosis. In contrast, suppression of Autophagy at late stage by CQ enhanced the anti-cancer effect of GLP. Furthermore, we demonstrated that GLP-induced autophagosome accumulation and Apoptosis is mediated via MAPK/ERK activation. Finally, GLP inhibited tumor growth and also inhibited autophagic flux in vivo. These results unveil new molecular mechanism underlying anti-cancer effects of GLP, suggesting that GLP is a potent Autophagy Inhibitor and might be useful in Anticancer therapy.

Figures
Products