Impaired removal of H3K4 methylation affects cell fate determination and gene transcription

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Impaired removal of H3K4 methylation affects cell fate determination and gene transcription. / Lussi, Yvonne C; Mariani, Luca; Rundsten, Carsten Friis; Peltonen, Juhani; Myers, Toshia R; Krag, Claudia; Wong, Garry; Salcini, Anna Elisabetta.

In: Development (Cambridge, England), Vol. 143, No. 20, 18.10.2016, p. 3751-3762.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Lussi, YC, Mariani, L, Rundsten, CF, Peltonen, J, Myers, TR, Krag, C, Wong, G & Salcini, AE 2016, 'Impaired removal of H3K4 methylation affects cell fate determination and gene transcription', Development (Cambridge, England), vol. 143, no. 20, pp. 3751-3762. https://doi.org/10.1242/dev.139139

APA

Lussi, Y. C., Mariani, L., Rundsten, C. F., Peltonen, J., Myers, T. R., Krag, C., Wong, G., & Salcini, A. E. (2016). Impaired removal of H3K4 methylation affects cell fate determination and gene transcription. Development (Cambridge, England), 143(20), 3751-3762. https://doi.org/10.1242/dev.139139

Vancouver

Lussi YC, Mariani L, Rundsten CF, Peltonen J, Myers TR, Krag C et al. Impaired removal of H3K4 methylation affects cell fate determination and gene transcription. Development (Cambridge, England). 2016 Oct 18;143(20):3751-3762. https://doi.org/10.1242/dev.139139

Author

Lussi, Yvonne C ; Mariani, Luca ; Rundsten, Carsten Friis ; Peltonen, Juhani ; Myers, Toshia R ; Krag, Claudia ; Wong, Garry ; Salcini, Anna Elisabetta. / Impaired removal of H3K4 methylation affects cell fate determination and gene transcription. In: Development (Cambridge, England). 2016 ; Vol. 143, No. 20. pp. 3751-3762.

Bibtex

@article{b5a0b4eaa7be402aade8e3c84c67e818,
title = "Impaired removal of H3K4 methylation affects cell fate determination and gene transcription",
abstract = "Methylation of Histone 3 Lysine 4 (H3K4) is largely associated with promoters and enhancers of actively transcribed genes and it is finely regulated during development by the action of histone methyltransferases and demethylases. H3K4me3 demethylases of the KDM5 family have been previously implicated in development, but how the regulation of H3K4me3 level controls developmental processes is not fully established. Here, we show that the H3K4 demethylase RBR-2, the unique member of the KDM5 family in C. elegans, acts cell-autonomously and in a catalytic-dependent manner to control vulva precursor cells fate acquisition, by promoting the LIN-12/Notch pathway. Using genome-wide approaches, we show that RBR-2 reduces the H3K4me3 level at transcription start sites (TSSs) and in regions upstream the TSSs, and acts both as a transcription repressor and activator. The analysis of the lin-11 genetic locus, a direct RBR-2 target gene required for vulva precursor cell fate acquisition, shows that RBR-2 controls the epigenetic signature of the lin-11 vulva-specific enhancer and lin-11 expression, providing in vivo evidences that RBR-2 can positively regulate transcription and cell fate acquisition by controlling enhancer activity.",
author = "Lussi, {Yvonne C} and Luca Mariani and Rundsten, {Carsten Friis} and Juhani Peltonen and Myers, {Toshia R} and Claudia Krag and Garry Wong and Salcini, {Anna Elisabetta}",
note = "{\textcopyright} 2016. Published by The Company of Biologists Ltd.",
year = "2016",
month = oct,
day = "18",
doi = "10.1242/dev.139139",
language = "English",
volume = "143",
pages = "3751--3762",
journal = "Development",
issn = "0950-1991",
publisher = "The Company of Biologists",
number = "20",

}

RIS

TY - JOUR

T1 - Impaired removal of H3K4 methylation affects cell fate determination and gene transcription

AU - Lussi, Yvonne C

AU - Mariani, Luca

AU - Rundsten, Carsten Friis

AU - Peltonen, Juhani

AU - Myers, Toshia R

AU - Krag, Claudia

AU - Wong, Garry

AU - Salcini, Anna Elisabetta

N1 - © 2016. Published by The Company of Biologists Ltd.

PY - 2016/10/18

Y1 - 2016/10/18

N2 - Methylation of Histone 3 Lysine 4 (H3K4) is largely associated with promoters and enhancers of actively transcribed genes and it is finely regulated during development by the action of histone methyltransferases and demethylases. H3K4me3 demethylases of the KDM5 family have been previously implicated in development, but how the regulation of H3K4me3 level controls developmental processes is not fully established. Here, we show that the H3K4 demethylase RBR-2, the unique member of the KDM5 family in C. elegans, acts cell-autonomously and in a catalytic-dependent manner to control vulva precursor cells fate acquisition, by promoting the LIN-12/Notch pathway. Using genome-wide approaches, we show that RBR-2 reduces the H3K4me3 level at transcription start sites (TSSs) and in regions upstream the TSSs, and acts both as a transcription repressor and activator. The analysis of the lin-11 genetic locus, a direct RBR-2 target gene required for vulva precursor cell fate acquisition, shows that RBR-2 controls the epigenetic signature of the lin-11 vulva-specific enhancer and lin-11 expression, providing in vivo evidences that RBR-2 can positively regulate transcription and cell fate acquisition by controlling enhancer activity.

AB - Methylation of Histone 3 Lysine 4 (H3K4) is largely associated with promoters and enhancers of actively transcribed genes and it is finely regulated during development by the action of histone methyltransferases and demethylases. H3K4me3 demethylases of the KDM5 family have been previously implicated in development, but how the regulation of H3K4me3 level controls developmental processes is not fully established. Here, we show that the H3K4 demethylase RBR-2, the unique member of the KDM5 family in C. elegans, acts cell-autonomously and in a catalytic-dependent manner to control vulva precursor cells fate acquisition, by promoting the LIN-12/Notch pathway. Using genome-wide approaches, we show that RBR-2 reduces the H3K4me3 level at transcription start sites (TSSs) and in regions upstream the TSSs, and acts both as a transcription repressor and activator. The analysis of the lin-11 genetic locus, a direct RBR-2 target gene required for vulva precursor cell fate acquisition, shows that RBR-2 controls the epigenetic signature of the lin-11 vulva-specific enhancer and lin-11 expression, providing in vivo evidences that RBR-2 can positively regulate transcription and cell fate acquisition by controlling enhancer activity.

U2 - 10.1242/dev.139139

DO - 10.1242/dev.139139

M3 - Journal article

C2 - 27578789

VL - 143

SP - 3751

EP - 3762

JO - Development

JF - Development

SN - 0950-1991

IS - 20

ER -

ID: 178251465