Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(21): 15318-25, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23558680

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 ß 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.


Subject(s)
Cell Nucleus/metabolism , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation , Response Elements , Sp3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , CHO Cells , Cell Nucleus/genetics , Cell Nucleus/immunology , Cell Nucleus/pathology , Cricetinae , Fetus/immunology , Fetus/metabolism , Fetus/pathology , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
2.
J Immunol ; 185(8): 4896-903, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20861353

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a frequent complication of preterm birth. This chronic lung disease results from arrested saccular airway development and is most common in infants exposed to inflammatory stimuli. In experimental models, inflammation inhibits expression of fibroblast growth factor-10 (FGF-10) and impairs epithelial-mesenchymal interactions during lung development; however, the mechanisms connecting inflammatory signaling with reduced growth factor expression are not yet understood. In this study we found that soluble inflammatory mediators present in tracheal fluid from preterm infants can prevent saccular airway branching. In addition, LPS treatment led to local production of mediators that inhibited airway branching and FGF-10 expression in LPS-resistant C.C3-Tlr4(Lpsd)/J fetal mouse lung explants. Both direct NF-κB activation and inflammatory cytokines (IL-1ß and TNF-α) that activate NF-κB reduced FGF-10 expression, whereas chemokines that signal via other inflammatory pathways had no effect. Mutational analysis of the FGF-10 promoter failed to identify genetic elements required for direct NF-κB-mediated FGF-10 inhibition. Instead, NF-κB activation appeared to interfere with the normal stimulation of FGF-10 expression by Sp1. Chromatin immunoprecipitation and nuclear coimmunoprecipitation studies demonstrated that the RelA subunit of NF-κB and Sp1 physically interact at the FGF-10 promoter. These findings indicate that inflammatory signaling through NF-κB disrupts the normal expression of FGF-10 in fetal lung mesenchyme by interfering with the transcriptional machinery critical for lung morphogenesis.


Subject(s)
Fibroblast Growth Factor 10/biosynthesis , Lung/embryology , NF-kappa B/metabolism , Protein Kinases/metabolism , Animals , Chorioamnionitis/metabolism , Chromatin Immunoprecipitation , Female , Gene Expression , Gene Expression Regulation , Humans , Immunohistochemistry , Immunoprecipitation , Infant, Newborn , Lung/metabolism , Mice , Mice, Inbred BALB C , Pregnancy , Premature Birth , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
3.
Microbiology (Reading) ; 156(Pt 2): 452-462, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19875437

ABSTRACT

Inositol is essential in eukaryotes, and must be imported or synthesized. Inositol biosynthesis in Saccharomyces cerevisiae is controlled by three non-essential genes that make up the inositol regulon: ScINO2 and ScINO4, which together encode a heterodimeric transcriptional activator, and ScOPI1, which encodes a transcriptional repressor. ScOpi1p inhibits the ScIno2-ScIno4p activator in response to extracellular inositol levels. An important gene controlled by the inositol regulon is ScINO1, which encodes inositol-3-phosphate synthase, a key enzyme in inositol biosynthesis. In the pathogenic yeast Candida albicans, homologues of the S. cerevisiae inositol regulon genes are 'transcriptionally rewired'. Instead of regulating the CaINO1 gene, CaINO2 and CaINO4 regulate ribosomal genes. Another Candida species that is a prevalent cause of infections is Candida glabrata; however, C. glabrata is phylogenetically more closely related to S. cerevisiae than C. albicans. Experiments were designed to determine if C. glabrata homologues of the inositol regulon genes function similarly to S. cerevisiae or are transcriptionally rewired. CgINO2, CgINO4 and CgOPI1 regulate CgINO1 in a manner similar to that observed in S. cerevisiae. However, unlike in S. cerevisiae, CgOPI1 is essential. Genetic data indicate that CgOPI1 is a repressor that affects viability by regulating activation of a target of the inositol regulon.


Subject(s)
Candida glabrata/physiology , Inositol/genetics , Regulon , Candida glabrata/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Genes, Essential , Genes, Fungal , Inositol/physiology , Intramolecular Lyases/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...