[HTML][HTML] Loss of hepatocyte-nuclear-factor-4α affects colonic ion transport and causes chronic inflammation resembling inflammatory bowel disease in mice

M Darsigny, JP Babeu, AA Dupuis, EE Furth… - PloS one, 2009 - journals.plos.org
M Darsigny, JP Babeu, AA Dupuis, EE Furth, EG Seidman, É Lévy, EF Verdu, FP Gendron
PloS one, 2009journals.plos.org
Background Hnf4α, an epithelial specific transcriptional regulator, is decreased in
inflammatory bowel disease and protects against chemically-induced colitis in mice.
However, the precise role of this factor in maintaining normal inflammatory homeostasis of
the intestine remains unclear. The aim of this study was to evaluate the sole role of epithelial
Hnf4α in the maintenance of gut inflammatory homeostasis in mice. Methodology/Principal
Findings We show here that specific epithelial deletion of Hnf4α in mice causes …
Background
Hnf4α, an epithelial specific transcriptional regulator, is decreased in inflammatory bowel disease and protects against chemically-induced colitis in mice. However, the precise role of this factor in maintaining normal inflammatory homeostasis of the intestine remains unclear. The aim of this study was to evaluate the sole role of epithelial Hnf4α in the maintenance of gut inflammatory homeostasis in mice.
Methodology/Principal Findings
We show here that specific epithelial deletion of Hnf4α in mice causes spontaneous chronic intestinal inflammation leading to focal areas of crypt dropout, increased cytokines and chemokines secretion, immune cell infiltrates and crypt hyperplasia. A gene profiling analysis in diseased Hnf4α null colon confirms profound genetic changes in cell death and proliferative behaviour related to cancer. Among the genes involved in the immune protection through epithelial barrier function, we identify the ion transporter claudin-15 to be down-modulated early in the colon of Hnf4α mutants. This coincides with a significant decrease of mucosal ion transport but not of barrier permeability in young animals prior to the manifestation of the disease. We confirm that claudin-15 is a direct Hnf4α gene target in the intestinal epithelial context and is down-modulated in mouse experimental colitis and inflammatory bowel disease.
Conclusion
Our results highlight the critical role of Hnf4α to maintain intestinal inflammatory homeostasis during mouse adult life and uncover a novel function for Hnf4α in the regulation of claudin-15 expression. This establishes Hnf4α as a mediator of ion epithelial transport, an important process for the maintenance of gut inflammatory homeostasis.
PLOS