Advertisement
No access
Report

Germline DNA Demethylation Dynamics and Imprint Erasure Through 5-Hydroxymethylcytosine

Science
6 Dec 2012
Vol 339, Issue 6118
pp. 448-452

Epigenetic Controls

Germ cells in mammals give rise to sperm and eggs. During their development, germ cells undergo extensive epigenetic reprogramming, including global DNA demethylation, which is vital for the totipotency of the developing embryo. Hackett et al. (p. 448) show that the enzymes Tet1 and Tet2 are involved in the demethylation of individual genes and in imprinted gametic differentially methylated regions. The enzymes were also responsible for the global conversion of CpG methylation to 5-hydroxymethylcytosine, which then progressively declines. The findings suggest that demethylation can occur by replication-coupled dilution, although active mechanisms cannot be excluded. A small number of loci escape demethylation, providing a possible mechanistic basis for transgenerational inheritance.

Abstract

Mouse primordial germ cells (PGCs) undergo sequential epigenetic changes and genome-wide DNA demethylation to reset the epigenome for totipotency. Here, we demonstrate that erasure of CpG methylation (5mC) in PGCs occurs via conversion to 5-hydroxymethylcytosine (5hmC), driven by high levels of TET1 and TET2. Global conversion to 5hmC initiates asynchronously among PGCs at embryonic day (E) 9.5 to E10.5 and accounts for the unique process of imprint erasure. Mechanistically, 5hmC enrichment is followed by its protracted decline thereafter at a rate consistent with replication-coupled dilution. The conversion to 5hmC is an important component of parallel redundant systems that drive comprehensive reprogramming in PGCs. Nonetheless, we identify rare regulatory elements that escape systematic DNA demethylation in PGCs, providing a potential mechanistic basis for transgenerational epigenetic inheritance.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Figs. S1 to S24
References (25, 26)

Resources

File (hackett.sm.pdf)
File (papv2.pdf)

References and Notes

1
Surani M. A., Hayashi K., Hajkova P., Genetic and epigenetic regulators of pluripotency. Cell 128, 747 (2007).
2
Hajkova P., et al., Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877 (2008).
3
Hackett J. A., Zylicz J. J., Surani M. A., Parallel mechanisms of epigenetic reprogramming in the germline. Trends Genet. 28, 164 (2012).
4
Tahiliani M., et al., Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930 (2009).
5
Ito S., et al., Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466, 1129 (2010).
6
Ficz G., et al., Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 473, 398 (2011).
7
Ito S., et al., Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 333, 1300 (2011); 10.1126/science.1210597.
8
Maatouk D. M., et al., DNA methylation is a primary mechanism for silencing postmigratory primordial germ cell genes in both germ cell and somatic cell lineages. Development 133, 3411 (2006).
9
Hackett J. A., et al., Promoter DNA methylation couples genome-defence mechanisms to epigenetic reprogramming in the mouse germline. Development 139, 3623 (2012).
10
Hayashi K., Ohta H., Kurimoto K., Aramaki S., Saitou M., Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519 (2011).
11
Hajkova P., et al., Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15 (2002).
12
Inoue A., Zhang Y., Replication-dependent loss of 5-hydroxymethylcytosine in mouse preimplantation embryos. Science 334, 194 (2011); 10.1126/science.1212483.
13
Tam P. P., Snow M. H., Proliferation and migration of primordial germ cells during compensatory growth in mouse embryos. J. Embryol. Exp. Morphol. 64, 133 (1981).
14
Popp C., et al., Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature 463, 1101 (2010).
15
Qin C., et al., Intracisternal A particle genes: Distribution in the mouse genome, active subtypes, and potential roles as species-specific mediators of susceptibility to cancer. Mol. Carcinog. 49, 54 (2010).
16
Smallwood S. A., et al., Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nat. Genet. 43, 811 (2011).
17
Borgel J., et al., Targets and dynamics of promoter DNA methylation during early mouse development. Nat. Genet. 42, 1093 (2010).
18
Kurimoto K., et al., Complex genome-wide transcription dynamics orchestrated by Blimp1 for the specification of the germ cell lineage in mice. Genes Dev. 22, 1617 (2008).
19
Gu T. P., et al., The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature 477, 606 (2011).
20
Wossidlo M., et al., 5-Hydroxymethylcytosine in the mammalian zygote is linked with epigenetic reprogramming. Nat. Commun. 2, 241 (2011).
21
Iqbal K., Jin S. G., Pfeifer G. P., Szabó P. E., Reprogramming of the paternal genome upon fertilization involves genome-wide oxidation of 5-methylcytosine. Proc. Natl. Acad. Sci. U.S.A. 108, 3642 (2011).
22
Hajkova P., et al., Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 329, 78 (2010).
23
Cortellino S., et al., Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell 146, 67 (2011).
24
Dawlaty M. M., et al., Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell 9, 166 (2011).
25
Guibert S., Forné T., Weber M., Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 22, 633 (2012).
26
Lao K. Q., et al., mRNA-sequencing whole transcriptome analysis of a single cell on the SOLiD system. J. Biomol. Tech. 20, 266 (2009).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 339 | Issue 6118
25 January 2013

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 24 August 2012
Accepted: 16 November 2012
Published in print: 25 January 2013

Permissions

Request permissions for this article.

Acknowledgments

We thank N. Miller for fluorescence-activated cell sorting analysis, F. Tang and W. Tang for experimental support, and G-L. Xu for reagents. This work was funded by the Wellcome Trust (RG49135, RG44593, and 083563) and the Human Frontier Science Program. Sequencing data have been deposited in the Sequence Read Archive (SRA060914).

Authors

Affiliations

Jamie A. Hackett
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Roopsha Sengupta*
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Jan J. Zylicz*
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
Kazuhiro Murakami*
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Caroline Lee
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Thomas A. Down
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
M. Azim Surani [email protected]
Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK.
Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.

Notes

*
These authors contributed equally to this work.
To whom correspondence should be addressed. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Epigenetic Modification of Cytosines in Hematopoietic Differentiation and Malignant Transformation, International Journal of Molecular Sciences, 24, 2, (1727), (2023).https://doi.org/10.3390/ijms24021727
    Crossref
  2. Transgenerational Epigenetic Inheritance of Traumatic Experience in Mammals, Genes, 14, 1, (120), (2023).https://doi.org/10.3390/genes14010120
    Crossref
  3. Epigenetic markers in the embryonal germ cell development and spermatogenesis, Basic and Clinical Andrology, 33, 1, (2023).https://doi.org/10.1186/s12610-022-00179-3
    Crossref
  4. Dynamic Switching of Active Promoter and Enhancer Domains Regulates Tet1 and Tet2 Expression during Cell State Transitions between Pluripotency and Differentiation , Molecular and Cellular Biology, 35, 6, (1026-1042), (2023).https://doi.org/10.1128/MCB.01172-14
    Crossref
  5. Tet-Mediated DNA Demethylation Is Required for SWI/SNF-Dependent Chromatin Remodeling and Histone-Modifying Activities That Trigger Expression of the Sp7 Osteoblast Master Gene during Mesenchymal Lineage Commitment, Molecular and Cellular Biology, 37, 20, (2023).https://doi.org/10.1128/MCB.00177-17
    Crossref
  6. Limb blastema formation: How much do we know at a genetic and epigenetic level?, Journal of Biological Chemistry, 299, 2, (102858), (2023).https://doi.org/10.1016/j.jbc.2022.102858
    Crossref
  7. Transgenerational inheritance of acquired epigenetic signatures at CpG islands in mice, Cell, 186, 4, (715-731.e19), (2023).https://doi.org/10.1016/j.cell.2022.12.047
    Crossref
  8. Endocannabinoid system and epigenetics in spermatogenesis and testicular cancer, Hormones and Epigenetics, (75-106), (2023).https://doi.org/10.1016/bs.vh.2023.01.002
    Crossref
  9. Cancer and meiotic gene expression: Two sides of the same coin?, Meiosis in Development and Disease, (43-68), (2023).https://doi.org/10.1016/bs.ctdb.2022.06.002
    Crossref
  10. Mechanisms of DNA Methylation and Demethylation During Mammalian Development, Handbook of Epigenetics, (11-26), (2023).https://doi.org/10.1016/B978-0-323-91909-8.00009-8
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media