Neat1 is a p53-inducible lincRNA essential for transformation suppression

  1. Laura D. Attardi1,3
  1. 1Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA;
  2. 2Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA;
  3. 3Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA;
  4. 4Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA;
  5. 5Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA;
  6. 6Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan;
  7. 7RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan;
  8. 8Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
  1. Corresponding author: attardi{at}stanford.edu

Abstract

The p53 gene is mutated in over half of all cancers, reflecting its critical role as a tumor suppressor. Although p53 is a transcriptional activator that induces myriad target genes, those p53-inducible genes most critical for tumor suppression remain elusive. Here, we leveraged p53 ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) and RNA-seq (RNA sequencing) data sets to identify new p53 target genes, focusing on the noncoding genome. We identify Neat1, a noncoding RNA (ncRNA) constituent of paraspeckles, as a p53 target gene broadly induced by mouse and human p53 in different cell types and by diverse stress signals. Using fibroblasts derived from Neat1−/− mice, we examined the functional role of Neat1 in the p53 pathway. We found that Neat1 is dispensable for cell cycle arrest and apoptosis in response to genotoxic stress. In sharp contrast, Neat1 plays a crucial role in suppressing transformation in response to oncogenic signals. Neat1 deficiency enhances transformation in oncogene-expressing fibroblasts and promotes the development of premalignant pancreatic intraepithelial neoplasias (PanINs) and cystic lesions in KrasG12D-expressing mice. Neat1 loss provokes global changes in gene expression, suggesting a mechanism by which its deficiency promotes neoplasia. Collectively, these findings identify Neat1 as a p53-regulated large intergenic ncRNA (lincRNA) with a key role in suppressing transformation and cancer initiation, providing fundamental new insight into p53-mediated tumor suppression.

Keywords

Footnotes

  • Received May 25, 2016.
  • Accepted May 26, 2017.

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