Open Access

MAPK4 silencing together with a PARP1 inhibitor as a combination therapy in triple‑negative breast cancer cells

  • Authors:
    • Xiaoqi Zeng
    • Shanshan Jiang
    • Simin Ruan
    • Zhaoze Guo
    • Jingyun Guo
    • Minfeng Liu
    • Changsheng Ye
    • Jianyu Dong
  • View Affiliations

  • Published online on: June 2, 2021     https://doi.org/10.3892/mmr.2021.12187
  • Article Number: 548
  • Copyright: © Zeng 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

Triple‑negative breast cancer (TNBC) is the most common type of cancer among females worldwide and is associated with poor prognosis. Poly ADP‑ribose polymerase‑1 (PARP1) inhibitors are effective against TNBC with mutations in the breast cancer type 1 susceptibility protein (BRCA1) and/or BRCA2 genes; however, the development of resistance to PARP1 inhibitors limits their use. Thus, identifying strategies to overcome this resistance is urgently required. The aim of the present study was to investigate the potential function and mechanism of small interfering (si)RNA‑MAPK4 (siMAPK4) in enhancing the efficacy of a PARP1 inhibitor and reducing the resistance. In the present study, data on the mRNA expression level of MAPK4 in normal breast tissues and TNBC tissues were obtained from The Cancer Genome Atlas database. The mRNA and protein expression levels of MAPK4 in normal breast cells and TNBC cells were analyzed using reverse transcription‑quantitative PCR and western blotting, respectively. The phosphorylated (p) histone H2AX (γH2AX) protein expression was assessed via immunofluorescence. Cell Counting Kit‑8, wound healing and TUNEL assays were used to determine the proliferative, migratory and apoptotic abilities of HCC1937 cells. MAPK4 was highly expressed in TNBC patient tissues and cell lines. Moreover, overexpression of MAPK4 could promote HCC1937 cell proliferation. Treatment of HCC1937 cells with the combination of siMAPK4 and a PARP1 inhibitor olaparib decreased their proliferation and migration and increased their apoptosis. The protein expression levels of the DNA repair‑related proteins p‑DNA‑dependent protein kinase catalytic subunit (DNA‑PK) and RAD51 recombinase (RAD51) were inhibited in the siMAPK4 and siMAPK4 + olaparib groups. However, the marker of a double‑stranded break γH2AX showed increased protein expression in the siMAPK4 + olaparib group. As MAPK4 could phosphorylate AKT at threonine 308 (AKTT308), the current study restored p‑AKTT308 using a constitutively active AKT plasmid (AKT‑CA). p‑DNA‑PK and RAD51 showed high expression and γH2AX exhibited lower protein expression in the AKT‑CA group. The present findings suggested that siMAPK4 can enhance the sensitivity of TNBC cells to PARP1 inhibitors.
View Figures
View References

Related Articles

Journal Cover

August-2021
Volume 24 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng X, Jiang S, Ruan S, Guo Z, Guo J, Liu M, Ye C and Dong J: MAPK4 silencing together with a PARP1 inhibitor as a combination therapy in triple‑negative breast cancer cells. Mol Med Rep 24: 548, 2021
APA
Zeng, X., Jiang, S., Ruan, S., Guo, Z., Guo, J., Liu, M. ... Dong, J. (2021). MAPK4 silencing together with a PARP1 inhibitor as a combination therapy in triple‑negative breast cancer cells. Molecular Medicine Reports, 24, 548. https://doi.org/10.3892/mmr.2021.12187
MLA
Zeng, X., Jiang, S., Ruan, S., Guo, Z., Guo, J., Liu, M., Ye, C., Dong, J."MAPK4 silencing together with a PARP1 inhibitor as a combination therapy in triple‑negative breast cancer cells". Molecular Medicine Reports 24.2 (2021): 548.
Chicago
Zeng, X., Jiang, S., Ruan, S., Guo, Z., Guo, J., Liu, M., Ye, C., Dong, J."MAPK4 silencing together with a PARP1 inhibitor as a combination therapy in triple‑negative breast cancer cells". Molecular Medicine Reports 24, no. 2 (2021): 548. https://doi.org/10.3892/mmr.2021.12187