Abstract

Inflammation inhibits normal lung morphogenesis in preterm infants. Soluble inflammatory mediators present in the lungs of patients developing bronchopulmonary dysplasia disrupt expression of multiple genes critical for development. However, the mechanisms linking innate immune signaling and developmental programs are not clear. NF-κB activation inhibits expression of the critical morphogen FGF-10. Here, we show that interactions between the RELA subunit of NF-κB and SP3 suppress SP1-mediated FGF-10 expression. SP3 co-expression reduced SP1-mediated Fgf-10 promoter activity, suggesting antagonistic interactions between SP1 and SP3. Chromatin immunoprecipitation of LPS-treated primary mouse fetal lung mesenchymal cells detected increased interactions between SP3, RELA, and the Fgf-10 promoter. Expression of a constitutively active IκB kinase β mutant not only decreased Fgf-10 promoter activity but also increased RELA-SP3 nuclear interactions. Expression of a dominant-negative IκB, which blocks NF-κB nuclear translocation, prevented inhibition of FGF-10 by SP3. The inhibitory functions of SP3 required sequences located in the N-terminal region of the protein. These data suggested that inhibition of FGF-10 by inflammatory signaling involves the NF-κB-dependent interactions between RELA, SP3, and the Fgf-10 promoter. NF-κB activation may therefore lead to reduced gene expression by recruiting inhibitory factors to specific gene promoters following exposure to inflammatory stimuli.

Highlights

  • Inflammation and NF-␬B activation inhibit FGF-10 expression and lung development by an unknown mechanism

  • Because inflammation plays a major role in the pathogenesis of bronchopulmonary dysplasia, we previously investigated how innate immunity and NF-␬B activation in the fetal lung affects expression of genes important for normal development

  • SP3 Is Expressed throughout Development in the Fetal Lung— We hypothesized that NF-␬B inhibits FGF-10 transcription by interfering with other transcriptional activators

Read more

Summary

Background

Inflammation and NF-␬B activation inhibit FGF-10 expression and lung development by an unknown mechanism. The inhibitory functions of SP3 required sequences located in the N-terminal region of the protein These data suggested that inhibition of FGF-10 by inflammatory signaling involves the NF-␬B-dependent interactions between RELA, SP3, and the Fgf-10 promoter. Because inflammation plays a major role in the pathogenesis of bronchopulmonary dysplasia, we previously investigated how innate immunity and NF-␬B activation in the fetal lung affects expression of genes important for normal development. NF-␬B activation by microbial products or inflammatory mediators inhibits FGF-10 expression in the fetal mouse lung, leading to alterations in normal airway formation [13]. We show that NF-␬B activation recruits Sp3 to the Fgf-10 promoter, where it functions as a transcription inhibitor This novel mechanism may provide new insight into how inflammation can alter expression of developmentally important genes

EXPERIMENTAL PROCEDURES
The abbreviations used are
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call