Notch Signaling Mediates Differentiation in Barrett's Esophagus and Promotes Progression to Adenocarcinoma

Authors

Bettina Kunze, Technical University of Munich
Frederik Wein, Technical University of Munich
Hsin Yu Fang, Technical University of Munich
Akanksha Anand, Technical University of Munich
Theresa Baumeister, Technical University of Munich
Julia Strangmann, Technical University of Munich
Sophie Gerland, Technical University of Munich
Jonas Ingermann, Technical University of Munich
Natasha Stephens Münch, Technical University of Munich
Maria Wiethaler, Technical University of Munich
Vincenz Sahm, Technical University of Munich
Ana Hidalgo-Sastre, Technical University of Munich
Sebastian Lange, Technical University of Munich
Charles J. Lightdale, Columbia University Irving Medical Center
Aqiba Bokhari, Yosemite Pathology Medical Group
Gary W. Falk, University of Pennsylvania Perelman School of Medicine
Richard A. Friedman, Columbia University Irving Medical Center
Gregory G. Ginsberg, University of Pennsylvania Perelman School of Medicine
Prasad G. Iyer, Mayo Clinic
Zhezhen Jin, Columbia University Irving Medical Center
Hiroshi Nakagawa, Columbia University Irving Medical Center
Carrie J. Shawber, Columbia University Irving Medical Center
The Anh Nguyen, Oregon Health & Science University
William J. Raab, Columbia University Irving Medical Center
Piero Dalerba, Columbia University Irving Medical Center
Anil K. Rustgi, Columbia University Irving Medical Center
Antonia R. Sepulveda, Columbia University Irving Medical Center
Kenneth K. Wang, Mayo Clinic
Roland M. Schmid, Technical University of Munich
Timothy C. Wang, Columbia University Irving Medical Center
Julian A. Abrams, Columbia University Irving Medical Center
Michael Quante, Technical University of Munich

Document Type

Journal Article

Publication Date

8-1-2020

Journal

Gastroenterology

Volume

159

Issue

2

DOI

10.1053/j.gastro.2020.04.033

Keywords

Barrett's Esophagus; Carcinogenesis; Esophageal Adenocarcinoma; Goblet Cells

Abstract

© 2020 AGA Institute Background & Aims: Studies are needed to determine the mechanism by which Barrett's esophagus (BE) progresses to esophageal adenocarcinoma (EAC). Notch signaling maintains stem cells in the gastrointestinal tract and is dysregulated during carcinogenesis. We explored the relationship between Notch signaling and goblet cell maturation, a feature of BE, during EAC pathogenesis. Methods: We measured goblet cell density and levels of Notch messenger RNAs in BE tissues from 164 patients, with and without dysplasia or EAC, enrolled in a multicenter study. We analyzed the effects of conditional expression of an activated form of NOTCH2 (pL2.Lgr5.N2IC), conditional deletion of NOTCH2 (pL2.Lgr5.N2fl/fl), or loss of nuclear factor κB (NF-κB) (pL2.Lgr5.p65fl/fl), in Lgr5+ (progenitor) cells in L2-IL1B mice (which overexpress interleukin 1 beta in esophagus and squamous forestomach and are used as a model of BE). We collected esophageal and stomach tissues and performed histology, immunohistochemistry, flow cytometry, transcriptome, and real-time polymerase chain reaction analyses. Cardia and forestomach tissues from mice were cultured as organoids and incubated with inhibitors of Notch or NF-kB. Results: Progression of BE to EAC was associated with a significant reduction in goblet cell density comparing nondysplastic regions of tissues from patients; there was an inverse correlation between goblet cell density and levels of NOTCH3 and JAG2 messenger RNA. In mice, expression of the activated intracellular form of NOTCH2 in Lgr5+ cells reduced goblet-like cell maturation, increased crypt fission, and accelerated the development of tumors in the squamocolumnar junction. Mice with deletion of NOTCH2 from Lgr5+ cells had increased maturation of goblet-like cells, reduced crypt fission, and developed fewer tumors. Esophageal tissues from in pL2.Lgr5.N2IC mice had increased levels of RelA (which encodes the p65 unit of NF-κB) compared to tissues from L2-IL1B mice, and we found evidence of increased NF-κB activity in Lgr5+ cells. Esophageal tissues from pL2.Lgr5.p65fl/fl mice had lower inflammation and metaplasia scores than pL2.Lgr5.N2IC mice. In organoids derived from pL2-IL1B mice, the NF-κB inhibitor JSH-23 reduced cell survival and proliferation. Conclusions: Notch signaling contributes to activation of NF-κB and regulates differentiation of gastric cardia progenitor cells in a mouse model of BE. In human esophageal tissues, progression of BE to EAC was associated with reduced goblet cell density and increased levels of Notch expression. Strategies to block this pathway might be developed to prevent EAC in patients with BE.

This document is currently not available here.

Share

COinS