Biliary atresia (BA) is the most common cause of obstructive jaundice in infants and is lethal if uncorrected. BA affects 1/10,000 infants and is the leading indication for liver transplantation in children. The etiology of this fibroinflammatory disorder remains elusive, but recent work has implicated the cytokine interferon-gamma (IFN3) as having a crucial role, although the mechanism of IFN3 activation is unclear. IFN3 levels are elevated in patients with BA, and targeted deletion of IFNg inhibits development of the rhesus rotavirus (RRV)-injected mouse model of BA. The exclusive occurrence of BA in the developing liver suggests that developmental pathways play an important role in the pathogenesis of BA. Epigenetic modifications, such as DNA methylation, function as stable gene silencing mechanisms that repress gene expression in differentiated cells. Inhibition of DNA methylation, resulting in derepression of gene expression, has been shown to have a profound effect on development, and also increases the expression of IFN3 pathway genes. We have evidence that broad chemical (azacytidine [azaC]) or genetic (dnmt1, ahcy) inhibition of DNA methylation leads to defects in hepatobiliary development associated with a significant increase in IFN3 pathway genes, and that patients with BA demonstrate bile duct cell-specific DNA hypomethylation. These findings led us to hypothesize that bile duct cell DNA hypomethylation plays a pathogenic role in BA. We believe this to be mediated through TGF2, which plays a key role in biliary development and is inhibited by IFN3 activation. The goals of this proposal are to further understand the mechanisms by which inhibition of DNA methylation leads to developmental biliary defects, and to determine the importance of DNA methylation in the pathogenesis of BA. Our mechanistic studies will utilize zebrafish, in which we have considerable experience examining hepatobiliary development. Zebrafish are a facile model organism for studying development, as they develop rapidly ex vivo and there are considerable tools available for genetic manipulation and analysis. We will also utilize mouse models and take advantage of the high volume of patients with infantile biliary disease at The Children's Hospital of Philadelphia. Using these resources, we propose to test the hypothesis that DNA hypomethylation leads to biliary defects in zebrafish and mouse models and in patients with BA, and that activation of IFN3 plays a critical role.

Public Health Relevance

Diseases such as biliary atresia (BA) that affect the liver of infants have a devastating effect on patients and their families, as BA is the leading indication for liver transplantation in children and has a sizable impact on healthcare in the U.S. We use the zebrafish to model infantile liver diseases such as BA, and we have found similarities between several of our models and BA that suggest a novel mechanism for BA involving epigenetic changes to DNA. We propose studies in both zebrafish and mouse to determine the mechanisms by which these epigenetic changes lead to defects in liver development, and to more fully characterize such changes in infants with liver diseases such as BA.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK090260-02
Application #
8317546
Study Section
Hepatobiliary Pathophysiology Study Section (HBPP)
Program Officer
Sherker, Averell H
Project Start
2011-08-15
Project End
2016-05-31
Budget Start
2012-06-01
Budget End
2013-05-31
Support Year
2
Fiscal Year
2012
Total Cost
$364,313
Indirect Cost
$146,813
Name
Children's Hospital of Philadelphia
Department
Type
DUNS #
073757627
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Cofer, Zenobia C; Cui, Shuang; EauClaire, Steven F et al. (2016) Methylation Microarray Studies Highlight PDGFA Expression as a Factor in Biliary Atresia. PLoS One 11:e0151521
Cui, Shuang; Eauclaire, Steven F; Matthews, Randolph P (2013) Interferon-gamma directly mediates developmental biliary defects. Zebrafish 10:177-83