Open Access

PAD4 silencing inhibits inflammation whilst promoting trophoblast cell invasion and migration by inactivating the NEMO/NF‑κB pathway

  • Authors:
    • Min Zeng
    • Minjuan Xu
    • Xiafang Li
    • Junying Li
    • Yuanyuan Liu
  • View Affiliations

  • Published online on: July 12, 2022     https://doi.org/10.3892/etm.2022.11505
  • Article Number: 568
  • 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

Preeclampsia (PE), presenting with onset hypertension and proteinuria, is a pregnancy‑specific disorder that can result in maternal and fetal morbidity and mortality. Insufficient trophoblast invasion and migration has been considered to be an important cause of this disease. The present study aimed to investigate the role of peptidyl arginine deiminase 4 (PAD4), whose knockdown has been previously indicated to reduce inflammation and susceptibility to pregnancy loss in mice, in the development of PE in vitro. Lipopolysaccharide (LPS) was used to treat a human trophoblast cell line (HTR8/SVneo). After PAD4 silencing via transfection with short hairpin RNA against PAD4, the concentrations of inflammatory factors IL‑6, IL‑12 and monocyte chemoattractant protein (MCP)‑1 were measured using ELISA. Cell viability was also measured using Cell Counting Kit‑8 assay. HTR8/SVneo cell invasion and migration were detected using Transwell and wound healing assays, respectively. Western blotting was used to measure the expression of citrullinated NF‑κB essential modulator (NEMO) and nuclear NF‑κB p65 protein levels. TNF‑α was applied for evaluating the potential regulatory effects of PAD4 on NF‑κB in LPS‑stimulated HTR8/SVneo cells. LPS increased the levels of IL‑6, IL‑12 and MCP‑1 and reduced the migration and invasion of HTR8/SVneo cells. PAD4‑knockdown was found to markedly reduce the levels of IL‑6, IL‑12 and MCP‑1 secretion. HTR8/SVneo cell invasion and migration was also significantly elevated after PAD4 silencing following LPS exposure. In addition, LPS stimulation notably upregulated the protein levels of citrullinated NEMO and nuclear NF‑κB p65, which was restored by PAD4 knockdown. Furthermore, TNF‑α treatment partially counteracted the effects of PAD4 knockdown on the secretion of IL‑6, MCP‑1 and IL‑12, which are markers of inflammation, and invasion and migration in LPS‑induced HTR8/SVneo cells. To conclude, these results suggest that PAD4 silencing can suppress inflammation whilst promoting invasion and migration by trophoblast cells through inhibiting the NEMO/NF‑κB pathway. These findings furthered the understanding in the complex molecular mechanism that can trigger PE and provide a promising target for the treatment of this disease.
View Figures
View References

Related Articles

Journal Cover

September-2022
Volume 24 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng M, Xu M, Li X, Li J and Liu Y: PAD4 silencing inhibits inflammation whilst promoting trophoblast cell invasion and migration by inactivating the NEMO/NF‑κB pathway. Exp Ther Med 24: 568, 2022
APA
Zeng, M., Xu, M., Li, X., Li, J., & Liu, Y. (2022). PAD4 silencing inhibits inflammation whilst promoting trophoblast cell invasion and migration by inactivating the NEMO/NF‑κB pathway. Experimental and Therapeutic Medicine, 24, 568. https://doi.org/10.3892/etm.2022.11505
MLA
Zeng, M., Xu, M., Li, X., Li, J., Liu, Y."PAD4 silencing inhibits inflammation whilst promoting trophoblast cell invasion and migration by inactivating the NEMO/NF‑κB pathway". Experimental and Therapeutic Medicine 24.3 (2022): 568.
Chicago
Zeng, M., Xu, M., Li, X., Li, J., Liu, Y."PAD4 silencing inhibits inflammation whilst promoting trophoblast cell invasion and migration by inactivating the NEMO/NF‑κB pathway". Experimental and Therapeutic Medicine 24, no. 3 (2022): 568. https://doi.org/10.3892/etm.2022.11505