Abstract

The G/T transversion rs35705950, located approximately 3 kb upstream of the MUC5B start site, is the cardinal risk factor for idiopathic pulmonary fibrosis (IPF). Here, we investigate the function and chromatin structure of this –3 kb region and provide evidence that it functions as a classically defined enhancer subject to epigenetic programming. We use nascent transcript analysis to show that RNA polymerase II loads within 10 bp of the G/T transversion site, definitively establishing enhancer function for the region. By integrating Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) analysis of fresh and cultured human airway epithelial cells with nuclease sensitivity data, we demonstrate that this region is in accessible chromatin that affects the expression of MUC5B. Through applying paired single-nucleus RNA- and ATAC-seq to frozen tissue from IPF lungs, we extend these findings directly to disease, with results indicating that epigenetic programming of the –3 kb enhancer in IPF occurs in both MUC5B-expressing and nonexpressing lineages. In aggregate, our results indicate that the MUC5B-associated variant rs35705950 resides within an enhancer that is subject to epigenetic remodeling and contributes to pathologic misexpression in IPF.

Highlights

  • Idiopathic pulmonary fibrosis (IPF) affects 5 million people worldwide and is associated with a poor prognosis [1]

  • Our results indicate that the MUC5B-associated variant rs35705950 resides within an enhancer that is subject to epigenetic remodeling and contributes to pathologic misexpression in idiopathic pulmonary fibrosis (IPF)

  • We have characterized a functional interaction between FOXA2 and a highly conserved binding site located within 32 bp of the variant site [15], and we have observed methylation of this region associated with IPF, MUC5B expression, and the MUC5B promoter variant [15]

Read more

Summary

Introduction

Idiopathic pulmonary fibrosis (IPF) affects 5 million people worldwide and is associated with a poor prognosis [1]. Transcription factors have been reported to directly bind in the region of the MUC5B variant [12], increase MUC5B expression [12,13,14,15], and affect goblet cell differentiation [16]. Histone acetylation has been implicated in regulating MUC5B [18, 19], suggesting a dynamic role for epigenetic modifications in controlling MUC5B expression. Despite this growing understanding of MUC5B regulation and epigenetics, a comprehensive mechanistic understanding of MUC5B regulation and heterogeneous expression in association with IPF remains elusive

Methods
Results
Conclusion
Full Text
Paper version not known

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