Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Epigenetic events in mammalian germ-cell development: reprogramming and beyond

Key Points

  • Accumulating evidence indicates that epigenetic modifiers have key roles in genome-wide reprogramming and activation and repression of specific genes during mammalian germ-cell development.

  • Germ-cell specification and suppression of the somatic gene expression programme might involve epigenetic modifications. Furthermore, differentiating primordial germ cells (PGCs) show global reduction in repressive epigenetic marks, and activation and repression of post-migratory PGC-specific genes involves DNA methylation and histone-arginine methylation.

  • In post-migratory PGCs, epigenetic imprints that are specific to the parent of origin are erased. Subsequently, sex-specific imprints are established during male and female gametogenesis, and the de novo DNA methylation machinery has a central role in this process.

  • DNA methylation is important for silencing retrotransposons in germ cells and therefore contributes to the maintenance of genomic integrity. Recent studies have revealed a link between DNA methylation and small-RNA-mediated silencing mechanisms.

  • A number of histone methyltransferases are essential for establishing a chromosome structure that is appropriate for events that occur during meiosis and for activation and repression of genes that are necessary or unnecessary for meiosis. Meiotic sex-chromosome inactivation in male germ cells involves histone variants and epigenetic modifications.

  • Global nuclear remodelling including histone–protamine exchange occurs in haploid spermatids. Expression of some haploid-specific genes relies on histone demethylation.

  • A better understanding of the epigenetic regulation of germ-cell development will be of great importance for improvement of reproductive engineering technologies and human health.

Abstract

The epigenetic profile of germ cells, which is defined by modifications of DNA and chromatin, changes dynamically during their development. Many of the changes are associated with the acquisition of the capacity to support post-fertilization development. Our knowledge of this aspect has greatly increased— for example, insights into how the re-establishment of parental imprints is regulated. In addition, an emerging theme from recent studies is that epigenetic modifiers have key roles in germ-cell development itself — for example, epigenetics contributes to the gene-expression programme that is required for germ-cell development, regulation of meiosis and genomic integrity. Understanding epigenetic regulation in germ cells has implications for reproductive engineering technologies and human health.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Germ cell development and associated epigenetic events in mice.
Figure 2: Epigenetic reprogramming in primordial germ cells (PGCs).
Figure 3: Cross-talk between DNA methylation, histone marks and the piRNA pathway in male germ cells.
Figure 4: Epigenetic changes that occur during meiosis in male and female gametogenesis.

Similar content being viewed by others

References

  1. Goldberg, A. D., Allis, C. D. & Bernstein, E. Epigenetics: a landscape takes shape. Cell 128, 635–638 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Surani, M. A., Hayashi, K. & Hajkova, P. Genetic and epigenetic regulators of pluripotency. Cell 128, 747–762 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Morgan, H. D., Santos, F., Green, K., Dean, W. & Reik, W. Epigenetic reprogramming in mammals. Hum. Mol. Genet. 14, R47–R58 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Allegrucci, C., Thurston, A., Lucas, E. & Young, L. Epigenetics and the germline. Reproduction 129, 137–149 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Kimmins, S. & Sassone-Corsi, P. Chromatin remodelling and epigenetic features of germ cells. Nature 434, 583–589 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425–432 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Ginsburg, M., Snow, M. H. & McLaren A. Primordial germ cells in the mouse embryo during gastrulation. Development 110, 521–528 (1990).

    Article  CAS  PubMed  Google Scholar 

  8. Sato, M. et al. Identification of PGC7, a new gene expressed specifically in preimplantation embryos and germ cells. Mech. Dev. 113, 91–94 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Saitou, M., Barton, S. C. & Surani, M. A. A molecular programme for the specification of germ cell fate in mice. Nature 418, 293–300 (2002).

    Article  CAS  PubMed  Google Scholar 

  10. Lawson, K. A. & Hage, W. J. Clonal analysis of the origin of primordial germ cells in the mouse. CIBA Found. Symp. 182, 68–84 (1994).

    CAS  PubMed  Google Scholar 

  11. Tam, P. P. & Zhou, S. X. The allocation of epiblast cells to ectodermal and germ-line lineages is influenced by the position of the cells in the gastrulating mouse embryo. Dev. Biol. 178, 124–132 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Yoshimizu, T., Obinata, M. & Matsui M. Stage-specific tissue and cell interactions play key roles in mouse germ cell specification. Development 128, 481–490 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Lawson, K. A. et al. Bmp4 is required for the generation of primordial germ cells in the mouse embryo. Genes Dev. 13, 424–436 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ying, Y., Liu, X. M., Marble, A., Lawson, K. A. & Zhao, G. Q. Requirement of Bmp8b for the generation of primordial germ cells in the mouse. Mol. Endocrinol. 14, 1053–1063 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Ying, Y., Qi, X. & Zhao, G. Induction of primordial germ cell from murine epiblasts by synergistic action of BMP4 and BMP8b signaling pathway. Proc. Natl Acad. Sci. USA 98, 7858–7862 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ohinata, Y. et al. Blimp1 is a critical determinant of the germ cell lineage in mice. Nature 436, 207–213 (2005). This paper describes the first identification of a gene that directly regulates germ-cell specification in early mouse embryos.

    Article  CAS  PubMed  Google Scholar 

  17. Mello, C. C. et al. The PIE-1 protein and germline specification in C. elegans embryos. Nature 382, 710–712 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Seydoux, G. et al. Repression of gene expression in the embryonic germ lineage of C. elegans. Nature 382, 713–716 (1996).

    Article  CAS  PubMed  Google Scholar 

  19. Seydoux, G. & Dunn, M. A. Transcriptionally repressed germ cells lack a subpopulation of phosphorylated RNA polymerase II in early embryos of Caenorhabditis elegans and Drosophila melanogaster. Development 124, 2191–2201 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Schaner, C. E., Deshpande, G., Schedl, P. D. & Kelly, W. G. A conserved choromatin architecture marks and maintains the restricted germ cell lineage in worms and flies. Dev. Cell 5, 747–757 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jongens, T. A., Hay, B., Jan, L. Y. & Jan Y. N. The germ cell-less gene product: a posteriorly localized component necessary for germ cell development in Drosophila. Cell 70, 569–584 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Leatherman J. L., Levin, L., Boero, J. & Jongene, T. A. Germ cell-less act to repress transcription during the establishment of the Drosophila germ cell lineage. Curr. Biol. 12, 1681–1685 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Nakamura, A. et al. Requirement for a noncoding RNA in Drosophila polar granules for germ cell establishment. Science 274, 2075–2079 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Martinho, R. G., Kunwar P. S., Casanova, J. C. & Lehmann, R. A noncoding RNA is required for the repression of RNApolII-dependent transcription in primordial germ cells. Curr. Biol. 14, 159–165 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Deshpande, G., Calhoun, G., Yanowitz, J. L. & Schedl, P. D. Novel functions of nanos in downregulating mitosis and transcription during the development of the Drosophila germline. Cell 99, 271–281 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Ancelin, K. et al. Blimp1 associates with Prmt5 and directs histone agrinine methylation in mouse germ cells. Nature Cell Biol. 8, 623–630 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Seki, Y. et al. Extensive and orderly reprogramming of genome-wide chromatin modifications associated with specification and early development of germ cells in mice. Dev. Biol. 278, 440–58 (2006).

    Article  CAS  Google Scholar 

  28. Seki, Y. et al. Cellular dynamics associated with the genome-wide epigenetic reprogramming in migrating germ cell in mice. Development 134, 2627–2638 (2007). The first study to describe the genome-wide epigenetic changes in differentiating PGCs in detail.

    Article  CAS  PubMed  Google Scholar 

  29. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Spivakov, M. & Fisher, A. G. Epigenetic signatures of stem-cell identity. Nature Rev. Genet. 8, 263–271 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Maatouk, D. M. et al. DNA methylation is a primary mechanisms for silencing postmigratory primordial germ cell genes in both germ cell and somatic cell lineages. Development 133, 3411–3418 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Hajkova, P. et al. Epigenetc reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15–23 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Graham, P. L. & Kimble, J. The mog-1 gene is required for the switch from spermatogenesis to oogenesis in Caenorhabditis elegans. Genetics 133, 919–931 (1993).

    Article  CAS  PubMed  Google Scholar 

  34. Strahl, B. D. et al. Methylation of histone H4 at arginine 3 occurs in vivo and is mediated by the nuclear receptor coactivator PRMT1. Curr. Biol. 11, 996–1000 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, H. et al. Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor. Science 293, 853–857 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Pal, S., Vishwanath, S. N., Erdjument-Bromage, H., Tempest, P. & Sif, S. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol. Cell. Biol. 24, 9630–9645 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lee, J. et al. Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development 129, 1807–1817 (2002). The work is a beautiful example of the use of nuclear-transfer technology to study the epigenetic profile of single PGCs.

    Article  CAS  PubMed  Google Scholar 

  38. Monk, M. & McLaren, A. X-chromosome activity in foetal germ cells of the mouse. J. Embryol. Exp. Morphol. 63, 75–84 (1981).

    CAS  PubMed  Google Scholar 

  39. Tam P. P., Zhou, S. X. & Tan, S. S. X-chromosome activity of the mouse primordial germ cells revealed by the expression of an X-linked lacZ transgene. Development 120, 2925–2932 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. de Napoles, M., Nesterova, T. & Brockdorff, N. Early loss of Xist RNA expression and inactive X chromosome associated chromatin modification in developing primordial germ cells. PLoS ONE 2, e860 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Sugimoto, M. & Abe, K. X chromosome reactivation initiates in nascent primordial germ cells in mice. PLoS Genet. 3, 1309–1317 (2007).

    Article  CAS  Google Scholar 

  42. Lane, N. et al. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 35, 88–93 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Chong, S & Whitelaw, E. Epigenetic germline inheritance. Curr. Opin. Genet. Dev. 14, 692–696 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Richards, E. J. Inherited epigenetic variation — revisiting soft inheritance. Nature Rev. Genet. 7, 395–401 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Jirtle, R. L. & Skinner, M. K. Environmental epigenomics and disease susceptibility. Nature Rev. Genet. 8, 253–262 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Bowles, J. et al. Retinoid signaling determines germ cell fate in mice. Science 312, 596–600 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Davis, T. L., Yang, G. J., McCarrey, J. R. & Bartolomei, M. S. The H19 methylation imprint is erased and re-established differentially on the parental alleles during male germ cell development. Hum. Mol. Genet. 9, 2885–2894 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Ueda, T. et al. The paternal methylation imprint of the mouse H19 locus is acquired in the gonocyte stage during foetal testis development. Genes Cells. 5, 649–659 (2000).

    Article  CAS  PubMed  Google Scholar 

  49. Li, J.-Y., Lees-Murdock, D. J., Xu, G.-L. & Walsh, C. P. Timing of establishment of paternal methylation imprints in the mouse. Genomics 84, 952–960 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Kato, Y. et al. Role of Dnmt3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse. Hum. Mol. Genet. 16, 2272–2280 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Davis, T. L., Trasler, J. M., Moss, S. B., Yang, G. J. & Bartolomei, M. S. Acquisition of the H19 methylation imprint occurs differentially on the parental alleles during spermatogenesis. Genomics 58, 18–28 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Kaneda, M. et al. Essential role for de novo DNA methyltransferase Dnmt3a in paternal and maternal imprinting. Nature 429, 900–903 (2004). Using a germline-specific gene-knockout strategy, the authors showed that DNMT3A, but not DNMT3B, is essential for de novo DNA methylation of the imprinted loci in both male and female germ cells.

    Article  CAS  PubMed  Google Scholar 

  53. Bourc'his, D. & Bestor, T. H. Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking Dnmt3L. Nature 431, 96–99 (2004). This work was the first to reveal that disruption of a regulator of de novo DNA methylation causes reactivation of retrotransposons and male infertility.

    Article  CAS  PubMed  Google Scholar 

  54. Webster, K. et al. Meiotic and epigenetic defects in Dnmt3L-knockout mouse spermatogenesis. Proc. Natl Acad. Sci. USA 102, 4068–4073 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kanatsu-Shinohara, M. et al. Generation of pluripotent stem cells from neonatal mouse testis. Cell 119, 1001–1012 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Lucifero, D., Mann, M. R. W., Bartolomei, M. S. & Trasler, J. M. Gene-specific timing and epigenetic memory in oocyte imprinting. Hum. Mol. Genet. 13, 839–849 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Hiura, H., Obata, Y., Komiyama, J., Shirai, M. & Kono, T. Oocyte growth-dependent progression of maternal imprinting in mice. Genes Cells, 11, 353–361 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Bourc'his, D., Xu, G.-L., Lin, C. S., Bollman, B. & Bestor, T. H. Dnmt3L and the establishment of maternal genomic imprints. Science 294, 2536–2539 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Hata, K., Okano, M., Lei, H. & Li, E. Dnmt3L cooperates with the Dnmt3 family of de novo DNA methyltransferases to establish maternal imprints in mice. Development 129, 1983–1993 (2002).

    Article  CAS  PubMed  Google Scholar 

  60. Jia, D., Jurkowska, R. Z., Zhang, X., Jeltsch, A. & Cheng, X. Structure of Dnmt3a bound Dnmt3L suggests a model for de novo DNA methylation. Nature 449, 248–251 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ooi, S. K. T. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Fedoriw, A. M., Stein, P., Svoboda, P., Schultz, R. M. & Bartolomei, M. S. Transgenic RNAi reveals essential function for CTCT in H19 gene imprinting. Science 303, 238–240 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Tada, T. et al. imprint switching for non-random X-chromosome inactivation during mouse oocyte growth. Development 127, 3101–3103 (2000).

    Article  CAS  PubMed  Google Scholar 

  64. Kaneda, M. et al. Role of de novo DNA methyltransferases in initiation of genomic imprinting and X-chromosome inactivation. Cold Spring Harbor Symp. Quant. Biol. 69, 125–129 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Kono, T. et al. Birth of parthenogenetic mice that can develop to adulthood. Nature 428, 860–864 (2004). The surprising outcome of this work clearly showed that genomic imprinting is the main and perhaps only barrier to parthenogenesis in mammals.

    Article  CAS  PubMed  Google Scholar 

  66. Kuwahara, M. et al. High-frequency generation of viable mice from engineered bi-maternal embryos. Nature Biotechnol. 25, 1045–1050 (2007).

    Article  CAS  Google Scholar 

  67. Slotkin, R. K. & Martienssen, R. Transposable elements and the epigenetic regulation of the genome. Nature Rev. Genet. 8, 272–285 (2007).

    Article  CAS  PubMed  Google Scholar 

  68. Hata, K., Kusumi, M., Yokomine, T., Li, E. & Sasaki, H. Meiotic and epigenetic aberrations in Dnmt3L-deficient male germ cells. Mol. Reprod. Dev. 73, 116–122 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Aravin, A. A., Sachidanandam, R., Girard, A., Fejes-Toth, K. & Hannon, G. J. Developmentally regulated piRNA clusters implicate MILI in transposon control. Science 316, 744–747 (2007). This work provided evidence that a component of the piRNA regulatory pathway influences DNA methylation in male germ cells.

    Article  CAS  PubMed  Google Scholar 

  70. Kuramochi-Miyagawa, S. et al. Two mouse piwi-related genes: miwi and mili. Mech. Dev. 108, 121–133 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. Aravin, A. et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature 442, 203–207 (2006).

    Article  CAS  PubMed  Google Scholar 

  72. Lau, N. C. et al. Characterization of the piRNA complex from rat testes. Science 313, 363–367 (2006).

    Article  CAS  PubMed  Google Scholar 

  73. Kuramochi-Miyagawa, S. et al. Mili, a mammalian member of piwi family gene, is essential for spermatogenesis. Development 131, 839–849 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Brower-Toland, B. et al. Drosophila PIWI associates with chromatin and interacts directly with HP1a. Genes Dev. 21, 2300–2311 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Klenov, M. S. et al. Repeat-associated siRNAs cause chromatin silencing of retrotransposons in the Drosophila melanogaster germline. Nucl. Acids Res. 35, 5430–5438 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. De La Fuente, R. et al. Lsh is required for meiotic chromosome synapsis and retrotransposon silencing in female germ cells. Nature Cell Biol. 8, 1448–1454 (2006).

    Article  CAS  PubMed  Google Scholar 

  77. Watanabe, T. et al. Identification and characterization of two novel classes of small RNAs in the mouse germline:retrotransposon-derived siRNAs in oocytes and germline small RNAs in testis. Genes Dev. 20, 1732–1743 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Payne, C. & Braun, R. E. Histone lysine trimethylation exhibits a distinct perinuclear distribution in Plzf-expressing spermatogonia. Dev. Biol. 293, 461–472 (2006).

    Article  CAS  PubMed  Google Scholar 

  79. Peters, A. H. F. M. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001). This paper first reported that histone H3K9 methyltransferases have a key role in early meiotic progression.

    Article  CAS  PubMed  Google Scholar 

  80. Tachibana, M., Nozaki, M., Takeda, N. & Shinkai, Y. Functional dynamics of H3K9 methylation during meiotic prophase progression. EMBO J. 26, 3346–3359 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Hayashi, K., Yoshida, K. & Matsui, Y. A histone H3 methyltransferase controls epigenetic events required for meiotic prophase. Nature 438, 374–378 (2005). The authors first indicated that a histone H3K4 methyltransferase (PRDM9) controls meiotic prophase progression by transcriptional regulation.

    Article  CAS  PubMed  Google Scholar 

  82. Turner, J. M. A. Meiotic sex chromosome inactivation. Development 134, 1823–1831 (2007).

    CAS  PubMed  Google Scholar 

  83. Fernandez-Capetillo, O. et al. H2AX is required for chromatin remodeling and inactivation of sex chromosomes in male mouse meiosis. Dev. Cell 4, 497–508 (2003).

    Article  CAS  PubMed  Google Scholar 

  84. Turner, J. M. A. et al. BRCA1, histone H2AX phosphorylation, and male meiotic sex chromosome inactivation. Curr. Biol. 14, 2135–2142 (2004). This study revealed the mechanisms of histone H2AX phosphorylation that are crucial for meiotic sex-chromosome inactivation.

    Article  CAS  PubMed  Google Scholar 

  85. Khalil, A. M., Boyar, F. Z. & Driscoll, D. J. Dynamic histone modifications mark sex chromosome inactivation and reactivation during mammalian spermatogenesis. Proc. Natl Acad. Sci. USA 101, 16583–16587 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Takada, Y. et al. Mammalian Polycomb Scmh1 mediates exclusion of Polycomb complexes from the XY body in the pachytene spermatocytes. Development 134, 579–590 (2007).

    Article  CAS  PubMed  Google Scholar 

  87. Huynh, K. D. & Lee, J. T. Inheritance of a pre-inactivated paternal X chromosome in early mouse embryos. Nature 426, 857–862 (2003).

    Article  CAS  PubMed  Google Scholar 

  88. Namekawa, S. H. et al. Postmeiotic sex chromatin in the male germ line of mice. Curr. Biol. 16, 660–607 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Turner, J. M. A., Mahadevaiah, S. K., Ellis, P. J. I., Mitchell, M. J. & Burgoyne, P. S. Pachytene asynapsis drives meiotic sex chromosome inactivation and leads to substantial postmeiotic repression in spermatids. Dev. Cell 10, 521–529 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Okamoto, I. et al. Evidence for de novo imprinted X-chromosome inactivation independent of meiotic inactivation in mice. Nature 438, 369–373 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Kim, J.-M., Liu, H., Tazaki, M., Nagata, M. & Aoki, F. Changes in histone acetylation during mouse oocytes meiosis. J. Cell Biol. 162, 37–46 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Akiyama, T., Nagata, M & Aoki, F. Inadequate histone deacetylation during oocyte meiosis causes aneuploidy and embryo death in mice. Proc. Natl Acad. Sci. USA 103, 7339–7344 (2006). This paper reported the importance of histone deacetylation for proper segregation of chromosomes during oocyte meiosis, which has implications for aneuploidy in pregnancies in older women.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Martianov, I. et al. Polar nuclear localization of H1T2, a histone H1 variant, required for spermatid elongation and DNA condensation during spermiogenesis. Proc. Natl Acad. Sci. USA 102, 2808–2813 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Rousseaux, S. et al. Establishment of male-specific epigenetic information. Gene 345, 139–153 (2005).

    Article  CAS  PubMed  Google Scholar 

  95. Okada, Y., Scott, G., Ray, M. K., Mishina, Y. & Zhang, Y. Histone demethylase JHDM2A is critical for Tnp1 and Prm1 transcription and spermatogenesis. Nature 450, 119–123 (2007). This paper first reported that a histone demethylase is involved in activation of a set of haploid-specific genes and is essential for packaging of sperm chromatin.

    Article  CAS  PubMed  Google Scholar 

  96. Wykes, S. M. & Krawetz, S. A. The structural organization of sperm chromatin. J. Biol. Chem. 278, 29471–29477 (2003).

    Article  CAS  PubMed  Google Scholar 

  97. Oakes, C. C., La Salle, S., Smiraglia, D. J., Robaire, B. & Trasler, J. M. Developmental acquisition of genome-wide DNA methylation occurs prior to meiosis in male germ cells. Dev. Biol. 307, 368–379 (2007).

    Article  CAS  PubMed  Google Scholar 

  98. Ariel, M., Cedar, H. & McCarrey, J. Developmental changes in methylation of spermatogenesis-specific genes include reprogramming in the epididymis. Nature Genet. 7, 59–63 (1994).

    Article  CAS  PubMed  Google Scholar 

  99. Flanagan, J. M. et al. Intra- and interindividual epigenetic variation in human germ cells. Am. J. Hum. Genet. 79, 67–84 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Marques, C. J., Carvalho, F., Sousa, M. & Barros, A. Genomic imprinting in disruptive spermatogenesis. Lancet 363, 1700–1702 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. Kobayashi, H. et al. Aberrant DNA methylation of imprinted loci in sperm from oligospermic patients. Hum. Mol. Genet. 16, 2542–2551 (2007).

    Article  CAS  PubMed  Google Scholar 

  102. Egli, D., Rosains, J., Birkhoff, G. & Eggan, K. Developmental reprogramming after chromosome transfer into mitotic mouse zygotes. Nature 447, 679–685 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Kanatsu-Shinohara, M. et al. Production of knockout mice by random or targeted mutagenesis in spermatogonial stem cells. Proc. Natl Acad. Sci. USA 103, 8018–8023 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. O'Neill, L. P., VerMilyea, M. D. & Turner, B. M. Epigenetic characterization of the early embryos with a chromatin immunoprecipitation protocol applicable to small cell populations. Nature Genet. 38, 835–841 (2006).

    Article  CAS  PubMed  Google Scholar 

  105. Barker, D. L. et al. Two methods of whole-genome amplification enable accurate genotyping across a 2320-SNP linkage panel. Genome Res. 14, 901–907 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Okita, K., Ichisaka, T. & Yamanaka, S. Generation of germline-competent induced pluripotent stem cells. Nature 448, 313–317 (2007).

    Article  CAS  PubMed  Google Scholar 

  107. Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318–324 (2007).

    Article  CAS  PubMed  Google Scholar 

  108. Hubner, K. et al. Derivation of oocytes from mouse embryonic stem cells. Science 300, 1251–1256 (2003).

    Article  PubMed  CAS  Google Scholar 

  109. Toyooka, Y., Tsunekawa, N., Akutsu, R. & Noce, T. Embryonic stem cells can form germ cells in vitro. Proc. Natl Acad. Sci. USA. 100, 11457–11462 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Geijsen, N. et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 427, 148–154 (2004).

    Article  CAS  PubMed  Google Scholar 

  111. Nayernia, K. et al. In vitro-differentiated embryonic stem cells give rise to male gametes that can generate offspring mice. Dev. Cell 11, 125–132 (2006).

    Article  CAS  PubMed  Google Scholar 

  112. Anway, M. D., Cupp, A. S., Uzumcu, M. & Skinner, M. K. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005).

    Article  CAS  PubMed  Google Scholar 

  113. Cropley, J. E., Suter, C. M., Beckman, K. B. & Martin D. I. K. Germ-line epigenetic modification of the murine Avy allele by nutritional supplementation. Proc. Natl Acad. Sci. USA. 103, 17308–17312 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Limey, L. H. Decreased birthweights in infants after maternal in utero exposure to the Dutch famine of 1944–1945. Paediatric Perinatal Epidemiol. 6, 240–253 (1992).

    Article  Google Scholar 

  115. Kaati, G., Bygren, L. O. & Edvinsson, S. Cardiovascular and diabetes mortality determined by nutrition during parents' and grandparents' slow growth period. Eur. J. Hum. Genet. 10, 682–688 (2002).

    Article  CAS  PubMed  Google Scholar 

  116. Kono, T., Obata, Y., Yoshimzu, T., Nakahara, T. & Carroll, J. Epigenetic modifications during oocyte growth correlates with extended parthenogenetic development in the mouse. Nature Genet. 13, 91–94 (1996).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to S. Tomizawa, T. Horiike and K. Takada for their help in preparation of the manuscript. Research in our laboratories is supported in part by Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science and Ministry of Education, Culture, Sports, Science and Technology of Japan.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Hiroyuki Sasaki or Yasuhisa Matsui.

Related links

Related links

FURTHER INFORMATION

Hiroyuki Sasaki's homepage

Cell Resource Center for Biomedical Research

Glossary

DNA methylation

A covalent modification that occurs predominantly at CpG dinucelotides in the vertebrate genome. It is catalysed by DNA methyltransferases and converts cytosines to 5-methylcytosines. It represses transcription directly by inhibiting the binding of specific transcription factors, and indirectly by recruiting methyl-CpG-binding proteins and their associated repressive chromatin-remodelling activities.

Histone modifications

Histones undergo post-translational modifications that alter their interactions with DNA and nuclear proteins. In particular, the tails of histones H3 and H4 can be covalently modified at several residues. Modifications of the tail include methylation, acetylation, phosphorylation and ubiquitylation, and influence several biological processes, including gene expression, DNA repair and chromosome condensation.

Nucleosome

The basic structural subunit of chromatin, responsible in part for the compactness of a chromosome. Each nucleosome consists of a sequence of DNA wrapped around a histone core, which is a histone octamer containing two copies of each of the core histones: H2A, H2B, H3 and H4.

Pluripotent

Able to give rise to a wide range of, but not all, cell lineages (usually all fetal lineages and a subset of extraembryonic lineages).

Epiblast

An embryonic lineage that is derived from the inner cell mass of the blastocyst, which gives rise to the body of the fetus.

Gastrulation

A process of cell and tissue movements whereby the cells of the blastula are rearranged to form a three-layered body plan, which consists of the outer ectoderm, inner ectoderm and interstitial mesoderm.

Primitive streak

A transitory embryonic structure, which is present as a strip of cells, that pre-figures the anterior–posterior axis of the embryo. During gastrulation embryonic cells progress through the streak.

Embryonic stem cell

A type of pluripotent stem cell that is derived from the inner cell mass of the early embryo. Pluripotent cells are capable of generating virtually all cell types of the organism.

Blastocyst

An early stage of mammalian embryonic development at which the first cell lineages become established.

Embryoid body

Spherical structure formed by differentiating ES cells in culture, which resembles the early embryo.

X-chromosome inactivation

The process that occurs in female mammals by which gene expression from one of the pair of X chromosomes is downregulated to match the levels of gene expression from the single X chromosome that is present in males. The inactivation process involves a range of epigenetic mechanisms on the inactivated chromosome, including changes in DNA methylation and histone modifications.

Chromosome synapsis

The association or pairing of the two pairs of sister chromatids (representing homologous chromosomes) that occurs at the start of meiosis.

Argonaute proteins

Argonaute proteins are the central components of RNA-silencing mechanisms. They provide the platform for target-mRNA recognition by short guide RNA strands and the catalytic activity for mRNA cleavage.

Pericentric heterochromatin

This is the highly compacted chromatin region that is juxtaposed to centromeres and contains large blocks of tandem repeats. It is irreversibly silenced and remains so throughout the cell cycle.

Histone variants

Structurally distinct, non-typical versions of the histone proteins. They are encoded by independent genes and often subject to regulation that is distinct from that of the canonical histones.

Aneuploidy

Presence of an abnormal number of chromosomes. For example, in the case of trisomies, an extra copy of a chromosome is present.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sasaki, H., Matsui, Y. Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nat Rev Genet 9, 129–140 (2008). https://doi.org/10.1038/nrg2295

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg2295

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing