Open Access

PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis

Corrigendum in: /10.3892/or.2022.8433

  • Authors:
    • Jun Pu
    • Ting Li
    • Nanjing Liu
    • Chunli Luo
    • Zhen Quan
    • Luo Li
    • Xiaohou Wu
  • View Affiliations

  • Published online on: February 26, 2020     https://doi.org/10.3892/or.2020.7520
  • Pages: 1397-1412
  • Copyright: © Pu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Radiotherapy (RT) has been used as a therapeutic option for treatment of prostate cancer (PCa) for a number of years; however, patients frequently develop RT resistance, particularly in castration‑resistant PCa (CRPC), although the underlying mechanisms remain unknown. Understanding the underlying mechanism of RT resistance in CRPC may potentially highlight novel targets to improve therapeutic options for patients with PCa. In the present study, the expression levels of phospholipase Cε (PLCε), androgen receptor (AR) and DNA‑dependent protein kinase catalytic subunit (PKcs) were examined in PCa tissue samples and PCa cells, and the effects of PLCε knockdown on AR and DNA damage repair (DDR)‑related molecules were determined. The association between PLCε, AR and Poly (ADP‑ribose) polymerase 1 (PARP1), as well as their respective roles in radiation resistance, were assessed using gene knockdown and pharmaceutical inhibitors or activators. A chromatin immunoprecipitation assay was used to determine the epigenetic regulatory effects of PLCε on PARP1. Animal experiments were performed to assess whether the mechanisms observed in vitro could be replicated in vivo. The expression levels of PLCε, AR and DNA‑PKcs were significantly upregulated in PCa, particularly in CRPC. PLCε knockdown reduced the viability and increased apoptosis of cells subjected to radiation. Additionally, PLCε deficiency suppressed DDR progression by downregulating an AR and PARP1 positive feedback loop and the associated downstream molecules following radiation. PLCε depletion also increased the presence of histone H3 lysine 27 trimethylation in the PARP1 promoter region, suggesting increased methylation of the PARP1 gene and thus resulting in reduced expression of PARP1. In vivo, PLCε knockdown significantly potentiated the effects of radiation on tumor growth. Taken together, the results of the present study demonstrated that PLCε knockdown enhanced the radiosensitivity of CRPC by downregulating the AR/PARP1/DNA‑PKcs axis.
View Figures
View References

Related Articles

Journal Cover

May-2020
Volume 43 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pu J, Li T, Liu N, Luo C, Quan Z, Li L and Wu X: PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis Corrigendum in /10.3892/or.2022.8433. Oncol Rep 43: 1397-1412, 2020
APA
Pu, J., Li, T., Liu, N., Luo, C., Quan, Z., Li, L., & Wu, X. (2020). PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis Corrigendum in /10.3892/or.2022.8433. Oncology Reports, 43, 1397-1412. https://doi.org/10.3892/or.2020.7520
MLA
Pu, J., Li, T., Liu, N., Luo, C., Quan, Z., Li, L., Wu, X."PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis Corrigendum in /10.3892/or.2022.8433". Oncology Reports 43.5 (2020): 1397-1412.
Chicago
Pu, J., Li, T., Liu, N., Luo, C., Quan, Z., Li, L., Wu, X."PLCε knockdown enhances the radiosensitivity of castration‑resistant prostate cancer via the AR/PARP1/DNA‑PKcs axis Corrigendum in /10.3892/or.2022.8433". Oncology Reports 43, no. 5 (2020): 1397-1412. https://doi.org/10.3892/or.2020.7520