4.5 Article

miR-199a-3p suppresses cervical epithelial cell inflammation by inhibiting the HMGB1/TLR4/NF-κB pathway in preterm birth

Journal

MOLECULAR MEDICINE REPORTS
Volume 22, Issue 2, Pages 926-938

Publisher

SPANDIDOS PUBL LTD
DOI: 10.3892/mmr.2020.11184

Keywords

microRNA-199a-3p; preterm birth; cervical epithelial cells; high-mobility group box 1 protein; inflammation; toll-like receptor 4; NF-kappa B pathway

Funding

  1. Yunnan Applied Basic Research Project-Kunming Medical University Union Foundation [2017FE468-117]
  2. Health Science and Technology Program of Yunnan Province [2016NS199]
  3. Science and Technology Talents and Platform Plan of Yunnan Province [2017HC008, S2016IC027]

Ask authors/readers for more resources

Preterm birth (PTB) is the primary cause of neonatal mortality worldwide. Infection and inflammation are considered to be the primary causes of PTB. Cervical remodeling is an important step in the process of preterm delivery, and the destruction of the cervical epithelial barrier and inflammation are important triggers of cervical remodeling. The aim of the present study was to determine the effect and underlying mechanism of microRNA (miR)-199a-3p/high-mobility group box 1 protein (HMGB1) signaling in cervical epithelial inflammation in PTB. The results of this study revealed that miR-199a-3p was significantly decreased in cervical epithelial tissue samples from patients in both the preterm labor and preterm premature rupture of membrane groups. This decrease was also observed in tissue samples from a lipopolysaccharide (LPS)-induced PTB mouse model and in LPS-induced ectocervical and endocervical cells. Whereas, the expression of HMGB1 and toll-like receptor 4 (TLR4) was significantly increased, which was associated with the upregulation of interleukin (IL)-1 beta and tumor necrosis factor (TNF)-alpha expression. Furthermore, overexpression of miR-199a-3p significantly suppressed the expression and activation of HMGB1 and TLR4/NF-kappa B signaling, and decreased the levels of IL-1 beta and TNF-alpha in vitro and in vivo. Additionally, overexpression of HMGB1 and/or TLR4 reversed the anti-inflammatory effects of miR-199a-3p mimics in vitro and in vivo. These results indicate that miR-199a-3p acts as a negative inflammatory regulator in PTB by targeting HMGB1 to regulate the TLR4/NF-kappa B pathway.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available