Oncotarget

Research Papers:

Telomere shortening accelerates tumor initiation in the L2-IL1B mouse model of Barrett esophagus and emerges as a possible biomarker

Vincenz Sahm, Carlo Maurer, Theresa Baumeister, Akanksha Anand, Julia Strangmann, Roland M. Schmid, Timothy C. Wang and Michael Quante _

PDF  |  Full Text  |  How to cite

Oncotarget. 2022; 13:347-359. https://doi.org/10.18632/oncotarget.28198

Metrics: PDF 1372 views  |   Full Text 2581 views  |   ?  


Abstract

Vincenz Sahm1, Carlo Maurer1, Theresa Baumeister1,3, Akanksha Anand1, Julia Strangmann1,3, Roland M. Schmid1, Timothy C. Wang2 and Michael Quante1,3

1 II Medizinische Klinik, Technische Universität München, Munich, Germany

2 Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA

3 Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany

Correspondence to:

Michael Quante, email: [email protected]

Keywords: Barrett's esophagus; telomere shortening; esophageal cancer; risk factor; TERT/TERC

Received: October 05, 2021     Accepted: February 07, 2022     Published: February 14, 2022

Copyright: © 2022 Sahm et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

ABSTRACT

Barrett’s esophagus (BE) is a precursor of the esophageal adenocarcinoma (EAC). BE- development and its progression to cancer is associated with gastroesophageal reflux disease. However, there is currently no molecular risk prediction model that accurately identifies patients at high risk for EAC. Here, we investigated the impact of shortened telomeres in a mouse model for Barrett esophagus (L2-IL1B). The L2-IL1B mouse model is characterized by IL-1β-mediated inflammation, which leads to a Barrett-like metaplasia in the transition zone between the squamous forestomach and glandular cardia/stomach. Telomere shortening was achieved by mTERC knockout. In the second generation (G2) of mTERC knockout L2-IL1B.mTERC−/− G2 mice exhibited telomere dysfunction with significantly shorter telomeres as measured by qFISH compared to L2-IL1B mice, correlating with stronger DNA damage in the form of phosphorylation of H2AX (γH2AX). Macroscopically, tumor area along the squamocolumnar junction (SCJ) was increased in L2-IL1B.mTERC−/− G2 mice, along with increased histopathological dysplasia. In vitro studies indicated increased organoid formation capacity in BE tissue from L2-IL1B.mTERC−/− G2 mice. In addition, pilot studies of human BE-, dysplasia- and EAC tissue samples confirmed that BE epithelial cells with or without dysplasia (LGD) had shorter telomeres compared to gastric cardia tissue. Of note, differentiated goblet cells retained longer telomeres than columnar lined BE epithelium. In conclusion, our studies suggest that shortened telomeres are functionally important for tumor development in a mouse model of BE and are associated with proliferating columnar epithelium in human BE. We propose that shortened telomeres should be evaluated further as a possible biomarker of cancer risk in BE patients.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 28198