PCGF6-PRC1 suppresses premature differentiation of mouse embryonic stem cells by regulating germ cell-related genes

  1. Mitsuhiro Endoh  Is a corresponding author
  2. Takaho A Endo
  3. Jun Shinga
  4. Katsuhiko Hayashi
  5. Anca Farcas
  6. Kit-Wan Ma
  7. Shinsuke Ito
  8. Jafar Sharif
  9. Tamie Endoh
  10. Naoko Onaga
  11. Manabu Nakayama
  12. Tomoyuki Ishikura
  13. Osamu Masui
  14. Benedikt M Kessler
  15. Toshio Suda
  16. Osamu Ohara
  17. Akihiko Okuda
  18. Robert J Klose
  19. Haruhiko Koseki  Is a corresponding author
  1. RIKEN Center for Integrative Medical Sciences, Japan
  2. Faculty of Medical Sciences, Kyushu University, Japan
  3. Oxford University, United Kingdom
  4. Kazusa DNA Research Institute, Japan
  5. University of Oxford, United Kingdom
  6. National University of Singapore, Singapore
  7. Saitama Medical University, Japan
  8. Faculty of Medical Sciences, Kyushu University, United Kingdom

Abstract

The ring finger protein PCGF6 (polycomb group ring finger 6) interacts with RING1A/B and E2F6 associated factors to form a non-canonical PRC1 (polycomb repressive complex 1) known as PCGF6-PRC1. Here, we demonstrate that PCGF6-PRC1 plays a role in repressing a subset of PRC1 target genes by recruiting RING1B and mediating downstream mono-ubiquitination of histone H2A. PCGF6-PRC1 bound loci are highly enriched for promoters of germ cell-related genes in mouse embryonic stem cells (ESCs). Conditional ablation of Pcgf6 in ESCs leads to robust de-repression of such germ cell-related genes, in turn affecting cell growth and viability. We also find a role for PCGF6 in pre- and peri-implantation mouse embryonic development. We further show that a heterodimer of the transcription factors MAX and MGA recruits PCGF6 to target loci. PCGF6 thus links sequence specific target recognition by the MAX/MGA complex to PRC1-dependent transcriptional silencing of germ cell-specific genes in pluripotent stem cells.

Data availability

The following data sets were generated
The following previously published data sets were used
    1. Luciano Di Croce
    (2013) Cbx7_ChIPSeq
    Publicly available at the NCBI Gene Expression Omnibus (accession no: GSM1041373).
    1. Francesco Neri
    (2014) Max_ChIPSeq
    Publicly available at the NCBI Gene Expression Omnibus (accession no: GSM1171650).
    1. Francesco Neri
    (2014) BioMyc_ChIPSeq
    Publicly available at the NCBI Gene Expression Omnibus (accession no: GSM1171648).
    1. Blackledge NP
    (2014) KDM2Bfl/fl_RING1B_ChIPSeq
    Publicly available at the NCBI Gene Expression Omnibus (accession no: GSE55698).

Article and author information

Author details

  1. Mitsuhiro Endoh

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    For correspondence
    csime@nus.edu.sg
    Competing interests
    The authors declare that no competing interests exist.
  2. Takaho A Endo

    Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  3. Jun Shinga

    Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  4. Katsuhiko Hayashi

    Department of Developmental Stem Cell Biology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
    Competing interests
    The authors declare that no competing interests exist.
  5. Anca Farcas

    Department of Biochemistry, Oxford University, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  6. Kit-Wan Ma

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  7. Shinsuke Ito

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  8. Jafar Sharif

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  9. Tamie Endoh

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  10. Naoko Onaga

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  11. Manabu Nakayama

    Chromosome Engineering Team, Department of Technology Development, Kazusa DNA Research Institute, Kisarazu, Japan
    Competing interests
    The authors declare that no competing interests exist.
  12. Tomoyuki Ishikura

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  13. Osamu Masui

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  14. Benedikt M Kessler

    TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  15. Toshio Suda

    Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
    Competing interests
    The authors declare that no competing interests exist.
  16. Osamu Ohara

    Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    Competing interests
    The authors declare that no competing interests exist.
  17. Akihiko Okuda

    Division of Developmental Biology, Saitama Medical University, Hidaka, Japan
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-5298-5564
  18. Robert J Klose

    Department of Developmental Stem Cell Biology, Faculty of Medical Sciences, Kyushu University, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8726-7888
  19. Haruhiko Koseki

    Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
    For correspondence
    haruhiko.koseki@riken.jp
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-8424-5854

Funding

RIKEN

  • Haruhiko Koseki

Ministry of Education, Culture, Sports, Science, and Technology

  • Haruhiko Koseki

Japan Science and Technology Agency (Strategic Basic Research Programs)

  • Haruhiko Koseki

Ministry of Education, Culture, Sports, Science, and Technology (Grant-in-Aid for Scientific Research on Innovative Areas (#26112516))

  • Mitsuhiro Endoh

Ministry of Education, Culture, Sports, Science, and Technology (Grant-in-Aid for Young Scientist (B) (#25871129))

  • Mitsuhiro Endoh

Ministry of Education, Culture, Sports, Science, and Technology (Grant-in-Aid for Scientific Research (C) (#16K07372))

  • Mitsuhiro Endoh

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Robb Krumlauf, Stowers Institute for Medical Research, United States

Ethics

Animal experimentation: All animal experiments were carried out according to the in-house guidelines for the care and use of laboratory animals of the RIKEN Center for Integrative Medical Sciences, Yokohama, Japan [Approval number: Kei-27-001(7)].

Version history

  1. Received: September 1, 2016
  2. Accepted: March 15, 2017
  3. Accepted Manuscript published: March 17, 2017 (version 1)
  4. Version of Record published: March 31, 2017 (version 2)
  5. Version of Record updated: April 25, 2017 (version 3)

Copyright

© 2017, Endoh et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 4,711
    views
  • 1,068
    downloads
  • 104
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Mitsuhiro Endoh
  2. Takaho A Endo
  3. Jun Shinga
  4. Katsuhiko Hayashi
  5. Anca Farcas
  6. Kit-Wan Ma
  7. Shinsuke Ito
  8. Jafar Sharif
  9. Tamie Endoh
  10. Naoko Onaga
  11. Manabu Nakayama
  12. Tomoyuki Ishikura
  13. Osamu Masui
  14. Benedikt M Kessler
  15. Toshio Suda
  16. Osamu Ohara
  17. Akihiko Okuda
  18. Robert J Klose
  19. Haruhiko Koseki
(2017)
PCGF6-PRC1 suppresses premature differentiation of mouse embryonic stem cells by regulating germ cell-related genes
eLife 6:e21064.
https://doi.org/10.7554/eLife.21064

Share this article

https://doi.org/10.7554/eLife.21064

Further reading

    1. Cell Biology
    2. Neuroscience
    Marcos Moreno-Aguilera, Alba M Neher ... Carme Gallego
    Research Article Updated

    Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.