PUBLICATION

Intronic Cis-Regulatory Modules Mediate Tissue-Specific and Microbial Control of angptl4/fiaf Transcription

Authors
Camp, J.G., Jazwa, A.L., Trent, C.M., and Rawls, J.F.
ID
ZDB-PUB-120409-13
Date
2012
Source
PLoS Genetics   8(3): e1002585 (Journal)
Registered Authors
Camp, Gray, Rawls, John F., Trent, Chad
Keywords
Gastrointestinal tract, Zebrafish, Sequence alignment, Introns, Sequence motif analysis, DNA transcription, Epithelium, Gene regulation
MeSH Terms
  • Angiopoietins*/genetics
  • Angiopoietins*/metabolism
  • Animals
  • Conserved Sequence
  • Evolution, Molecular
  • Gene Expression Regulation, Developmental
  • Humans
  • Intestinal Mucosa/embryology
  • Intestinal Mucosa/metabolism
  • Intestinal Mucosa/microbiology*
  • Introns
  • Mammals
  • Mutagenesis, Site-Directed
  • Organ Specificity
  • Regulatory Sequences, Nucleic Acid*/genetics
  • Transcription, Genetic
  • Zebrafish*/embryology
  • Zebrafish*/genetics
  • Zebrafish*/microbiology
  • Zebrafish Proteins/genetics
  • Zebrafish Proteins/metabolism
PubMed
22479192 Full text @ PLoS Genet.
Abstract

The intestinal microbiota enhances dietary energy harvest leading to increased fat storage in adipose tissues. This effect is caused in part by the microbial suppression of intestinal epithelial expression of a circulating inhibitor of lipoprotein lipase called Angiopoietin-like 4 (Angptl4/Fiaf). To define the cis-regulatory mechanisms underlying intestine-specific and microbial control of Angptl4 transcription, we utilized the zebrafish system in which host regulatory DNA can be rapidly analyzed in a live, transparent, and gnotobiotic vertebrate. We found that zebrafish angptl4 is transcribed in multiple tissues including the liver, pancreatic islet, and intestinal epithelium, which is similar to its mammalian homologs. Zebrafish angptl4 is also specifically suppressed in the intestinal epithelium upon colonization with a microbiota. In vivo transgenic reporter assays identified discrete tissue-specific regulatory modules within angptl4 intron 3 sufficient to drive expression in the liver, pancreatic islet β-cells, or intestinal enterocytes. Comparative sequence analyses and heterologous functional assays of angptl4 intron 3 sequences from 12 teleost fish species revealed differential evolution of the islet and intestinal regulatory modules. High-resolution functional mapping and site-directed mutagenesis defined the minimal set of regulatory sequences required for intestinal activity. Strikingly, the microbiota suppressed the transcriptional activity of the intestine-specific regulatory module similar to the endogenous angptl4 gene. These results suggest that the microbiota might regulate host intestinal Angptl4 protein expression and peripheral fat storage by suppressing the activity of an intestine-specific transcriptional enhancer. This study provides a useful paradigm for understanding how microbial signals interact with tissue-specific regulatory networks to control the activity and evolution of host gene transcription.

Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping