Cudraflavone C Induces Tumor-Specific Apoptosis in Colorectal Cancer Cells through Inhibition of the Phosphoinositide 3-Kinase (PI3K)-AKT Pathway

PLoS One. 2017 Jan 20;12(1):e0170551. doi: 10.1371/journal.pone.0170551. eCollection 2017.

Abstract

Cudraflavone C (Cud C) is a naturally-occurring flavonol with reported anti-proliferative activities. However, the mechanisms by which Cud C induced cytotoxicity have yet to be fully elucidated. Here, we investigated the effects of Cud C on cell proliferation, caspase activation andapoptosis induction in colorectal cancer cells (CRC). We show that Cud C inhibits cell proliferation in KM12, Caco-2, HT29, HCC2998, HCT116 and SW48 CRC but not in the non-transformed colorectal epithelial cells, CCD CoN 841. Cud C induces tumor-selective apoptosis via mitochondrial depolarization and activation of the intrinsic caspase pathway. Gene expression profiling by microarray analyses revealed that tumor suppressor genes EGR1, HUWE1 and SMG1 were significantly up-regulated while oncogenes such as MYB1, CCNB1 and GPX2 were down-regulated following treatment with Cud C. Further analyses using Connectivity Map revealed that Cud C induced a gene signature highly similar to that of protein synthesis inhibitors and phosphoinositide 3-kinase (PI3K)-AKT inhibitors, suggesting that Cud C might inhibit PI3K-AKT signaling. A luminescent cell free PI3K lipid kinase assay revealed that Cud C significantly inhibited p110β/p85α PI3K activity, followed by p120γ, p110δ/p85α, and p110α/p85α PI3K activities. The inhibition by Cud C on p110β/p85α PI3K activity was comparable to LY-294002, a known PI3K inhibitor. Cud C also inhibited phosphorylation of AKT independent of NFκB activity in CRC cells, while ectopic expression of myristoylated AKT completely abrogated the anti-proliferative effects, and apoptosis induced by Cud C in CRC. These findings demonstrate that Cud C induces tumor-selective cytotoxicity by targeting the PI3K-AKT pathway. These findings provide novel insights into the mechanism of action of Cud C, and indicate that Cud C further development of Cud C derivatives as potential therapeutic agents is warranted.

MeSH terms

  • Apoptosis / drug effects*
  • Caco-2 Cells
  • Cell Line, Tumor
  • Colorectal Neoplasms / drug therapy*
  • Flavones / pharmacology*
  • Gene Expression Profiling
  • HCT116 Cells
  • HT29 Cells
  • Humans
  • Phosphatidylinositol 3-Kinases / drug effects*
  • Proto-Oncogene Proteins c-akt / drug effects*
  • Real-Time Polymerase Chain Reaction
  • Signal Transduction / drug effects*

Substances

  • Flavones
  • cudraflavone C
  • Phosphatidylinositol 3-Kinases
  • Proto-Oncogene Proteins c-akt

Grants and funding

We express sincere gratitude to researchers of the International Medical University (IMU) (http://www.imu.edu.my/imu/) Institute for Research, Development and Innovation (IRDI) especially to Center for Cancer and Stem Cell Research for the insightful scientific discussions. This work was funded by the IMU BMedSci Research Training Program (BMS I01/2015) for HCS. KHL was granted an Early Career Research and Knowledge Transfer Award provided by the University of Nottingham in 2011 (A2RHF1) (http://www.nottingham.edu.my/index.aspx), which supported the plant collection, extraction and isolation work. VAY was a recipient of the postgraduate scholarship from the Ministry of Higher Education, Malaysia from 2013-2015 (https://www.mohe.gov.my/en/).