Transcription factor-mediated intestinal metaplasia and the role of a shadow enhancer

  1. Ramesh A. Shivdasani1,2,3,8
  1. 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA;
  2. 2Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA;
  3. 3Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
  4. 4Division of Hematology Oncology, Boston Children's Hospital, Boston, Massachusetts 02215, USA;
  5. 5Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA;
  6. 6Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA;
  7. 7Hubretch Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, Netherlands;
  8. 8Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA;
  9. 9Howard Hughes Medical Institute, Boston, Massachusetts 02215, USA;
  10. 10Departments of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
  1. Corresponding author: ramesh_shivdasani{at}dfci.harvard.edu

Abstract

Barrett's esophagus (BE) and gastric intestinal metaplasia are related premalignant conditions in which areas of human stomach epithelium express mixed gastric and intestinal features. Intestinal transcription factors (TFs) are expressed in both conditions, with unclear causal roles and cis-regulatory mechanisms. Ectopic CDX2 reprogrammed isogenic mouse stomach organoid lines to a hybrid stomach–intestinal state transcriptionally similar to clinical metaplasia; squamous esophageal organoids resisted this CDX2-mediated effect. Reprogramming was associated with induced activity at thousands of previously inaccessible intestine-restricted enhancers, where CDX2 occupied DNA directly. HNF4A, a TF recently implicated in BE pathogenesis, induced weaker intestinalization by binding a novel shadow Cdx2 enhancer and hence activating Cdx2 expression. CRISPR/Cas9-mediated germline deletion of that cis-element demonstrated its requirement in Cdx2 induction and in the resulting activation of intestinal genes in stomach cells. dCas9-conjugated KRAB repression mapped this activity to the shadow enhancer's HNF4A binding site. Altogether, we show extensive but selective recruitment of intestinal enhancers by CDX2 in gastric cells and that HNF4A-mediated ectopic CDX2 expression in the stomach occurs through a conserved shadow cis-element. These findings identify mechanisms for TF-driven intestinal metaplasia and a likely pathogenic TF hierarchy.

Keywords

Footnotes

  • Received August 30, 2021.
  • Accepted December 13, 2021.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance