Your browser doesn't support javascript.
loading
H3K9me3 maintenance on a human artificial chromosome is required for segregation but not centromere epigenetic memory.
Martins, Nuno M C; Cisneros-Soberanis, Fernanda; Pesenti, Elisa; Kochanova, Natalia Y; Shang, Wei-Hao; Hori, Tetsuya; Nagase, Takahiro; Kimura, Hiroshi; Larionov, Vladimir; Masumoto, Hiroshi; Fukagawa, Tatsuo; Earnshaw, William C.
Affiliation
  • Martins NMC; Wellcome Trust Centre for Cell Biology, Edinburgh, UK.
  • Cisneros-Soberanis F; Wellcome Trust Centre for Cell Biology, Edinburgh, UK.
  • Pesenti E; Wellcome Trust Centre for Cell Biology, Edinburgh, UK.
  • Kochanova NY; Wellcome Trust Centre for Cell Biology, Edinburgh, UK.
  • Shang WH; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
  • Hori T; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
  • Nagase T; Kazusa DNA Research Institute, Kisarazu, Japan.
  • Kimura H; Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.
  • Larionov V; National Cancer Institute, National Institutes of Health, Bethesda, USA.
  • Masumoto H; Kazusa DNA Research Institute, Kisarazu, Japan.
  • Fukagawa T; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
  • Earnshaw WC; Wellcome Trust Centre for Cell Biology, Edinburgh, UK bill.earnshaw@ed.ac.uk.
J Cell Sci ; 133(14)2020 07 24.
Article de En | MEDLINE | ID: mdl-32576667
ABSTRACT
Most eukaryotic centromeres are located within heterochromatic regions. Paradoxically, heterochromatin can also antagonize de novo centromere formation, and some centromeres lack it altogether. In order to investigate the importance of heterochromatin at centromeres, we used epigenetic engineering of a synthetic alphoidtetO human artificial chromosome (HAC), to which chimeric proteins can be targeted. By tethering the JMJD2D demethylase (also known as KDM4D), we removed heterochromatin mark H3K9me3 (histone 3 lysine 9 trimethylation) specifically from the HAC centromere. This caused no short-term defects, but long-term tethering reduced HAC centromere protein levels and triggered HAC mis-segregation. However, centromeric CENP-A was maintained at a reduced level. Furthermore, HAC centromere function was compatible with an alternative low-H3K9me3, high-H3K27me3 chromatin signature, as long as residual levels of H3K9me3 remained. When JMJD2D was released from the HAC, H3K9me3 levels recovered over several days back to initial levels along with CENP-A and CENP-C centromere levels, and mitotic segregation fidelity. Our results suggest that a minimal level of heterochromatin is required to stabilize mitotic centromere function but not for maintaining centromere epigenetic memory, and that a homeostatic pathway maintains heterochromatin at centromeres.This article has an associated First Person interview with the first authors of the paper.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Chromosomes artificiels humains Limites: Humans Langue: En Journal: J Cell Sci Année: 2020 Type de document: Article Pays d'affiliation: Royaume-Uni

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Chromosomes artificiels humains Limites: Humans Langue: En Journal: J Cell Sci Année: 2020 Type de document: Article Pays d'affiliation: Royaume-Uni
...