Your browser doesn't support javascript.
loading
miR­199a­3p suppresses cervical epithelial cell inflammation by inhibiting the HMGB1/TLR4/NF­κB pathway in preterm birth.
Peng, Juan; Jiang, Jiang; Wang, Huizi; Feng, Xinzi; Dong, Xudong.
Afiliação
  • Peng J; Department of Obstetrics, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650031, P.R. China.
  • Jiang J; Department of Obstetrics, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650031, P.R. China.
  • Wang H; Department of Obstetrics, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650031, P.R. China.
  • Feng X; Department of Obstetrics, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650031, P.R. China.
  • Dong X; Department of Obstetrics, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650031, P.R. China.
Mol Med Rep ; 22(2): 926-938, 2020 08.
Article em En | MEDLINE | ID: mdl-32468045
ABSTRACT
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)­199a­3p/high­mobility group box 1 protein (HMGB1) signaling in cervical epithelial inflammation in PTB. The results of this study revealed that miR­199a­3p 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 LPS­induced ectocervical and endocervical cells. Whereas, the expression of HMGB1 and toll­like receptor 4 (TLR4) was significantly increased, which was associated with the upregulation of interleukin (IL)­1ß and tumor necrosis factor (TNF)­α expression. Furthermore, overexpression of miR­199a­3p significantly suppressed the expression and activation of HMGB1 and TLR4/NF­κB signaling, and decreased the levels of IL­1ß and TNF­α in vitro and in vivo. Additionally, overexpression of HMGB1 and/or TLR4 reversed the anti­inflammatory effects of miR­199a­3p mimics in vitro and in vivo. These results indicate that miR­199a­3p acts as a negative inflammatory regulator in PTB by targeting HMGB1 to regulate the TLR4/NF­κB pathway.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Colo do Útero / MicroRNAs / Nascimento Prematuro Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Revista: Mol Med Rep Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Colo do Útero / MicroRNAs / Nascimento Prematuro Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Revista: Mol Med Rep Ano de publicação: 2020 Tipo de documento: Article