Abstract

SETD3 is a member of the protein lysine methyltransferase (PKMT) family, which catalyzes the addition of methyl group to lysine residues. However, the protein network and the signaling pathways in which SETD3 is involved remain largely unexplored. In the current study, we show that SETD3 is a positive regulator of DNA-damage-induced apoptosis in colon cancer cells. Our data indicate that depletion of SETD3 from HCT-116 cells results in a significant inhibition of apoptosis after doxorubicin treatment. Our results imply that the positive regulation is sustained by methylation, though the substrate remains unknown. We present a functional cross-talk between SETD3 and the tumor suppressor p53. SETD3 binds p53 in cells in response to doxorubicin treatment and positively regulates p53 target genes activation under these conditions. Mechanistically, we provide evidence that the presence of SETD3 and its catalytic activity is required for the recruitment of p53 to its target genes. Finally, Kaplan–Meier survival analysis, of two-independent cohorts of colon cancer patients, revealed that low expression of SETD3 is a reliable predictor of poor survival in these patients, which correlates with our findings. Together, our data uncover a new role of the PKMT SETD3 in the regulation of p53-dependent activation of apoptosis in response to DNA damage.

Highlights

  • Apoptosis is a conserved and essential cellular process of programmed cell death which allows damaged cells removal, maintaining and regulating homeostasis in multicellular organisms[1]

  • SETD3 is a potential regulator of DNA damage We utilized the R2 Genomics Analysis and Visualization

  • To study the cellular role of SETD3 in colorectal carcinoma cells (HCT-116), we immunoprecipitated overexpressed FLAG-SETD3 from the nuclear fraction followed by mass spectrometry (Fig. 1b)

Read more

Summary

Introduction

Apoptosis is a conserved and essential cellular process of programmed cell death which allows damaged cells removal, maintaining and regulating homeostasis in multicellular organisms[1]. DNA-damage-induced agents such as chemotherapeutic drugs and irradiation can lead to apoptotic death through a p53-dependent pathway[2]. The transcription factor p53 is an established tumor suppressor, which is activated upon DNA damage. P53 induces the transcription of many pro-apoptotic genes, such as BAX3, PUMA4, and NoxA5. The activation of these target genes results in a cascade of downstream events, which involves mitochondrial outer membrane permeabilization, cytochrome c release followed by the activation of caspases that eventually causes. Little is known about the PKMT SETD3, though it is abundantly expressed in many tissues, including muscle, where it promotes myocyte-differentiation by regulating the transcription of muscle-related genes[9]. We have recently shown that SETD3 methylates the transcription factor FoxM1 to regulate

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.