Abstract

While the over-expression of tumor suppressor programmed cell death 4 (PDCD4) induces apoptosis, it was recently shown that PDCD4 knockdown also induced apoptosis. In this study, we examined the cell cycle regulators whose activation is affected by PDCD4 knockdown to investigate the contribution of PDCD4 to cell cycle regulation in three types of hepatoma cells: HepG2, Huh7 (mutant p53 and p16-deficient), and Hep3B (p53- and Rb-deficient). PDCD4 knockdown suppressed cell growth in all three cell lines by inhibiting Rb phosphorylation via down-regulating the expression of Rb itself and CDKs, which phosphorylate Rb, and up-regulating the expression of the CDK inhibitor p21 through a p53-independent pathway. We also found that apoptosis was induced in a p53-dependent manner in PDCD4 knockdown HepG2 cells (p53+), although the mechanism of cell death in PDCD4 knockdown Hep3B cells (p53-) was different. Furthermore, PDCD4 knockdown induced cellular senescence characterized by β-galactosidase staining, and p21 knockdown rescued the senescence and cell death as well as the inhibition of Rb phosphorylation induced by PDCD4 knockdown. Thus, PDCD4 is an important cell cycle regulator of hepatoma cells and may be a promising therapeutic target for the treatment of hepatocellular carcinoma.

Highlights

  • Genetic alterations, including the activation of oncogenes and inactivation of tumor suppressor genes, are critical events in tumorigenesis [1, 2]

  • It contains two MA-3 domains that are homologous to the M1 domain in the eukaryotic translation initiation factor 4G, which interacts with eukaryotic translation initiation factor 4A to form the cap-dependent translation initiation factor complex eukaryotic translation initiation factor 4 F and is thought to Induction of Cellular Senescence by Programmed cell death 4 (PDCD4) Knockdown be involved in protein-protein interactions [14, 15]

  • The results indicated that PDCD4 knockdown suppressed hepatoma cell growth

Read more

Summary

Introduction

Genetic alterations, including the activation of oncogenes and inactivation of tumor suppressor genes, are critical events in tumorigenesis [1, 2]. PDCD4 was first isolated from a human glioma complementary DNA (cDNA) library and localized to chromosome 10q24 [12, 13]. It contains two MA-3 domains that are homologous to the M1 domain in the eukaryotic translation initiation factor 4G (eIF4G), which interacts with eukaryotic translation initiation factor 4A (eIF4A) to form the cap-dependent translation initiation factor complex eukaryotic translation initiation factor 4 F (eIF4F) and is thought to Induction of Cellular Senescence by PDCD4 Knockdown be involved in protein-protein interactions [14, 15]. Previous studies have shown that PDCD4 inhibits the interaction between eIF4A and eIF4G as well as eIF4A’s RNAhelicase activity [16]. It has been reported that PDCD4 expression is increased during apoptosis [20, 21] and decreased by treatment with interleukin-2 or interleukin-15 [22] as well as mitogens, such as a serum [23], epidermal growth factor (EGF) and 12-O-tetradecanoylphorbor13-acetate (TPA) [24]

Methods
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