Volltext-Downloads (blau) und Frontdoor-Views (grau)

Bitte verwenden Sie diesen Link, wenn Sie dieses Dokument zitieren oder verlinken wollen: https://nbn-resolving.org/urn:nbn:de:gbv:9-opus-59138

HELLS Is Negatively Regulated by Wild-Type P53 in Liver Cancer by a Mechanism Involving P21 and FOXM1

  • Simple Summary The tumor suppressor protein P53 is a major player in preventing liver cancer development and progression. In this study we could show that P53 negatively regulates the expression of Helicase, lymphoid specific (HELLS), previously described as an important pro-tumorigenic epigenetic regulator in hepatocarcinogenesis. The regulatory mechanism included induction of the P53 target gene P21 (CDKN1A) resulting in repression of HELLS via downregulation of the transcription factor Forkhead Box Protein M1 (FOXM1). Our in vitro and in vivo findings indicate an important additional aspect of the tumor suppressive function of P53 in liver cancer linked to epigenetic regulation. Abstract The major tumor suppressor P53 (TP53) acts primarily as a transcription factor by activating or repressing subsets of its numerous target genes, resulting in different cellular outcomes (e.g., cell cycle arrest, apoptosis and senescence). P53-dependent gene regulation is linked to several aspects of chromatin remodeling; however, regulation of chromatin-modifying enzymes by P53 is poorly understood in hepatocarcinogenesis. Herein, we identified Helicase, lymphoid specific (HELLS), a major epigenetic regulator in liver cancer, as a strong and selective P53 repression target within the SNF2-like helicase family. The underlying regulatory mechanism involved P53-dependent induction of P21 (CDKN1A), leading to repression of Forkhead Box Protein M1 (FOXM1) that in turn resulted in downregulation of HELLS expression. Supporting our in vitro data, we found higher expression of HELLS in murine HCCs arising in a Trp53−/− background compared to Trp53+/+ HCCs as well as a strong and highly significant correlation between HELLS and FOXM1 expression in different HCC patient cohorts. Our data suggest that functional or mutational inactivation of P53 substantially contributes to overexpression of HELLS in HCC patients and indicates a previously unstudied aspect of P53′s ability to suppress liver cancer formation.

Download full text files

Export metadata

Additional Services

Search Google Scholar

Statistics

frontdoor_oas
Metadaten
Author: Stefanie Schuller, Jan Sieker, Philip Riemenschneider, Bianca Köhler, Elisabeth Drucker, Sofia M. E. Weiler, Daniel Dauch, Carsten Sticht, Benjamin Goeppert, Stephanie Roessler, Silvia Ribback, Kai Breuhahn, Falko Fend, Frank Dombrowski, Kerstin Singer, Stephan Singer
URN:urn:nbn:de:gbv:9-opus-59138
DOI:https://doi.org/10.3390/cancers14020459
ISSN:2072-6694
Parent Title (English):Cancers
Publisher:MDPI
Editor: Vangelis Kondylis, Mathias Heikenwälder
Document Type:Article
Language:English
Date of first Publication:2022/01/17
Release Date:2022/07/05
Tag:HCC; P53 network; chromatin remodeling; gene repression
GND Keyword:-
Volume:14
Article Number:459
Page Number:20
Faculties:Universitätsmedizin / Institut für Pathologie
Licence (German):License LogoCreative Commons - Namensnennung