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:

Mechanisms regulating zygotic genome activation

Abstract

Following fertilization, the two specified gametes must unite to create an entirely new organism. The genome is initially transcriptionally quiescent, allowing the zygote to be reprogrammed into a totipotent state. Gradually, the genome is activated through a process known as the maternal-to-zygotic transition, which enables zygotic gene products to replace the maternal supply that initiated development. This essential transition has been broadly characterized through decades of research in several model organisms. However, we still lack a full mechanistic understanding of how genome activation is executed and how this activation relates to the reprogramming of the zygotic chromatin architecture. Recent work highlights the central role of transcriptional activators and suggests that these factors may coordinate transcriptional activation with other developmental changes.

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

Fig. 1: New technologies enable precise detection of ZGA.
Fig. 2: ZGA is conserved across animals.
Fig. 3: Several mechanisms contribute to the timing of ZGA.
Fig. 4: Chromatin is reprogrammed during ZGA.

Similar content being viewed by others

References

  1. Gurdon, J. B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. Development 10, 622–640 (1962).

    CAS  Google Scholar 

  2. Campbell, K. H. S., McWhir, J., Ritchie, W. A. & Wilmut, I. Sheep cloned by nuclear transfer from a cultured cell line. Nature 380, 64–66 (1996).

    Article  CAS  PubMed  Google Scholar 

  3. Newport, J. & Kirschner, M. A major developmental transition in early Xenopus embryos: II. Control of the onset of transcription. Cell 30, 687–696 (1982).

    Article  CAS  PubMed  Google Scholar 

  4. Tadros, W. & Lipshitz, H. D. The maternal-to-zygotic transition: a play in two acts. Development 136, 3033–3042 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Yartseva, V. & Giraldez, A. J. The maternal-to-zygotic transition during vertebrate development: a model for reprogramming. Curr. Top. Dev. Biol. 113, 191–232 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Yuan, K., Seller, C. A., Shermoen, A. W. & O’Farrell, P. H. Timing the Drosophila mid-blastula transition: a cell cycle-centered view. Trends Genet. 32, 496–507 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bertrand, E. et al. Localization of ASH1 mRNA particles in living yeast. Mol. Cell 2, 437–445 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Nelles, D. A. et al. Programmable RNA tracking in live cells with CRISPR/Cas9. Cell 165, 488–496 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, Z., Gerstein, M. & Snyder, M. RNA-Seq: a revolutionary tool for transcriptomics. Nat. Rev. Genet. 10, 57–63 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Furey, T. S. ChIP–seq and beyond: new and improved methodologies to detect and characterize protein–DNA interactions. Nat. Rev. Genet. 13, 840–852 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Giresi, P. G., Kim, J., McDaniell, R. M., Iyer, V. R. & Lieb, J. D. FAIRE (Formaldehyde-Assisted Isolation of Regulatory Elements) isolates active regulatory elements from human chromatin. Genome Res. 17, 877–885 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  13. Belton, J.-M. et al. Hi–C: a comprehensive technique to capture the conformation of genomes. Methods 58, 268–276 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Gao, L. et al. Chromatin accessibility landscape in human early embryos and its association with evolution. Cell 173, 248–259 (2018). This study profiles chromatin accessibility across early human embryogenesis by sequencing DNase I hypersensitive sites and implicates OCT4 as an activator of ZGA.

    Article  CAS  PubMed  Google Scholar 

  15. Mezger, A. et al. High-throughput chromatin accessibility profiling at single-cell resolution. Nat. Commun. 9, 3647 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Dahl, J. A. et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhang, B. et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Liu, X. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Eckersley-Maslin, M. A., Alda-Catalinas, C. & Reik, W. Dynamics of the epigenetic landscape during the maternal-to-zygotic transition. Nat. Rev. Mol. Cell. Biol. 19, 436–450 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Xu, Q. & Xie, W. Epigenome in early mammalian development: inheritance, reprogramming and establishment. Trends Cell Biol. 28, 237–253 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Collart, C. et al. High-resolution analysis of gene activity during the Xenopus mid-blastula transition. Development 141, 1927–1939 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Harvey, S. A. et al. Identification of the zebrafish maternal and paternal transcriptomes. Development 140, 2703–2710 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lott, S. E. et al. Noncanonical compensation of the zygotic X transcription in early Drosophila melanogaster development revealed through single-embryo RNA-Seq. PLOS Biol. 9, e1000590 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Newport, J. & Kirschner, M. A major developmental transition in early Xenopus embryos: I. characterization and timing of cellular changes at the midblastula stage. Cell 30, 675–686 (1982).

    Article  CAS  PubMed  Google Scholar 

  25. Prioleau, M. N., Huet, J., Sentenac, A. & Mechali, M. Competition between chromatin and transcription complex assembly regulates gene expression during early development. Cell 77, 439–449 (1994).

    Article  CAS  PubMed  Google Scholar 

  26. Dekens, M. P. S., Pelegri, F. J., Maischein, H.-M. & Nusslein-Volhard, C. The maternal-effect gene futile cycle is essential for pronuclear congression and mitotic spindle assembly in the zebrafish zygote. Development 130, 3907–3916 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Edgar, B. A., Kiehle, C. P. & Schubiger, G. Cell cycle control by the nucleo-cytoplasmic ratio in early Drosophila development. Cell 44, 365–372 (1986).

    Article  CAS  PubMed  Google Scholar 

  28. Lu, X., Li, J. M., Elemento, O., Tavazoie, S. & Wieschaus, E. F. Coupling of zygotic transcription to mitotic control at the Drosophila mid-blastula transition. Development 136, 2101–2110 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lee, D. R., Lee, J. E., Yoon, H. S., Roh, S. I. & Kim, M. K. Compaction in preimplantation mouse embryos is regulated by a cytoplasmic regulatory factor that alters between 1- and 2-cell stages in a concentration-dependent manner. J. Exp. Zool. 290, 61–71 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Guven-Ozkan, T., Nishi, Y., Robertson, S. M. & Lin, R. Global transcriptional repression in C. elegans germline precursors by regulated sequestration of TAF-4. Cell 135, 149–160 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Veenstra, G. J., Destree, O. H. & Wolffe, A. P. Translation of maternal TATA-binding protein mRNA potentiates basal but not activated transcription in Xenopus embryos at the midblastula transition. Mol. Cell. Biol. 19, 7972–7982 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Benoit, B. et al. An essential role for the RNA-binding protein Smaug during the Drosophila maternal-to-zygotic transition. Development 136, 923–932 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tadros, W. et al. SMAUG is a major regulator of maternal mRNA destabilization in Drosophila and its translation is activated by the PAN GU kinase. Dev. Cell 12, 143–155 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Lee, M. T. et al. Nanog, Pou5f1 and SoxB1 activate zygotic gene expression during the maternal-to-zygotic transition. Nature 503, 360–364 (2013). Using ribosome profiling, the authors identify Pou5f3 (previously Pou5f1), Nanog and Sox2 as the most highly translated transcription factors in the early zebrafish embryo and then show that depletion of these factors together results in a failure to initiate ZGA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chan, S. H. et al. Brd4 and P300 regulate zygotic genome activation through histone acetylation. Preprint at bioRxiv https://doi.org/10.1101/369231 (2018).

  36. Harrison, M. M., Botchan, M. R. & Cline, T. W. Grainyhead and Zelda compete for binding to the promoters of the earliest-expressed Drosophila genes. Dev. Biol. 345, 248–255 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Stancheva, I. & Meehan, R. R. Transient depletion of xDnmt1 leads to premature gene activation in Xenopus embryos. Genes Dev. 14, 313–327 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Ruzov, A. et al. Kaiso is a genome-wide repressor of transcription that is essential for amphibian development. Development 131, 6185–6194 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Pritchard, D. K. & Schubiger, G. Activation of transcription in Drosophila embryos is a gradual process mediated chardby the nucleocytoplasmic ratio. Genes Dev. 10, 1131–1142 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Luo, R. X. & Dean, D. C. Chromatin remodeling and transcriptional regulation. J. Natl Cancer Inst. 91, 1288–1294 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Amodeo, A. A., Jukam, D., Straight, A. F. & Skotheim, J. M. Histone titration against the genome sets the DNA-to-cytoplasm threshold for the Xenopus midblastula transition. Proc. Natl Acad. Sci. USA 112, E1086–E1095 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Joseph, S. R. et al. Competition between histone and transcription factor binding regulates the onset of transcription in zebrafish embryos. eL ife 6, e23326 (2017). This study shows a decrease in the concentration of unbound histones in the nuclei of zebrafish embryos that corresponds with ZGA and presents evidence that this decrease permits transcription factors to compete for DNA binding and activate transcription.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Jevtic´, P. & Levy, D. L. Nuclear size scaling during Xenopus early development contributes to midblastula transition timing. Curr. Biol. 25, 45–52 (2015).

    Article  PubMed  CAS  Google Scholar 

  44. Jevtic´, P. & Levy, D. L. Both nuclear size and DNA amount contribute to midblastula transition timing in Xenopus laevis. Sci. Rep. 7, 7908 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Hahn, S. Structure and mechanism of the RNA polymerase II transcription machinery. Nat. Struct. Mol. Biol. 11, 394–403 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gottesfeld, J. M. & Forbes, D. J. Mitotic repression of the transcriptional machinery. Trends Biochem. Sci. 22, 197–202 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Rothe, M., Pehl, M., Taubert, H. & Jackle, H. Loss of gene function through rapid mitotic cycles in the Drosophila embryo. Nature 359, 156–159 (1992).

    Article  CAS  PubMed  Google Scholar 

  48. Shermoen, A. W. & O’Farrell, P. H. Progression of the cell cycle through mitosis leads to abortion of nascent transcripts. Cell 67, 303–310 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Heyn, P. et al. The earliest transcribed zygotic genes are short, newly evolved, and different across species. Cell Rep. 6, 285–292 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. De Renzis, S., Elemento, O., Tavazoie, S. & Wieschaus, E. F. Unmasking activation of the zygotic genome using chromosomal deletions in the Drosophila embryo. PLOS Biol. 5, e117 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Swinburne, I. A. & Silver, P. A. Intron delays and transcriptional timing during development. Dev. Cell 14, 324–330 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Collart, C., Allen, G. E., Bradshaw, C. R., Smith, J. C. & Zegerman, P. Titration of four replication factors is essential for the Xenopus laevis midblastula transition. Science 341, 893–896 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kimelman, D., Kirschner, M. & Scherson, T. The events of the midblastula transition in Xenopus are regulated by changes in the cell cycle. Cell 48, 399–407 (1987).

    Article  CAS  PubMed  Google Scholar 

  54. Farrell, J. A. & O’Farrell, P. H. Mechanism and regulation of Cdc25/Twine protein destruction in embryonic cell-cycle remodeling. Curr. Biol. 23, 118–126 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang, M., Kothari, P., Mullins, M. & Lampson, M. A. Regulation of zygotic genome activation and DNA damage checkpoint acquisition at the mid-blastula transition. Cell Cycle 13, 3828–3838 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. McCleland, M. L. & O’Farrell, P. H. RNAi of mitotic cyclins in Drosophila uncouples the nuclear and centrosome cycle. Curr. Biol. 18, 245–254 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sung, H., Spangenberg, S., Vogt, N. & Großhans, J. Number of nuclear divisions in the Drosophila blastoderm controlled by onset of zygotic transcription. Curr. Biol. 23, 133–138 (2017).

    Article  CAS  Google Scholar 

  58. Jiang, C. & Pugh, B. F. Nucleosome positioning and gene regulation: advances through genomics. Nat. Rev. Genet. 10, 161–172 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bannister, A. J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Steger, K. & Balhorn, R. Sperm nuclear protamines: a checkpoint to control sperm chromatin quality. Anat. Histol. Embryol. 47, 273–279 (2018).

    Article  PubMed  Google Scholar 

  61. Zhou, L. & Dean, J. Reprogramming the genome to totipotency in mouse embryos. Trends Cell Biol. 25, 82–91 (2015).

    Article  CAS  PubMed  Google Scholar 

  62. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–365 (1993).

    Article  CAS  PubMed  Google Scholar 

  63. Panning, B. X-chromosome inactivation: the molecular basis of silencing. J. Biol. 7, 30 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Guo, H. et al. The DNA methylation landscape of human early embryos. Nature 511, 606–610 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Shen, L. et al. Tet3 and DNA replication mediate demethylation of both the maternal and paternal genomes in mouse zygotes. Cell Stem Cell 15, 459–471 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Messerschmidt, D. M., Knowles, B. B. & Solter, D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev. 28, 812–828 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Veenstra, G. J. C. & Wolffe, A. P. Constitutive genomic methylation during embryonic development of Xenopus. Biochim. Biophys. Acta 1521, 39–44 (2001).

    Article  CAS  PubMed  Google Scholar 

  68. Bogdanovic, O. et al. Temporal uncoupling of the DNA methylome and transcriptional repression during embryogenesis. Genome Res. 21, 1313–1327 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Potok, M. E., Nix, D. A., Parnell, T. J. & Cairns, B. R. Reprogramming the maternal zebrafish genome after fertilization to match the paternal methylation pattern. Cell 153, 759–772 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Jiang, L. et al. Sperm, but not oocyte, DNA methylome is inherited by zebrafish early embryos. Cell 153, 773–784 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Liu, G., Wang, W., Hu, S., Wang, X. & Zhang, Y. Inherited DNA methylation primes the establishment of accessible chromatin during genome activation. Genome Res. 28, 998–1007 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Yin, Y. et al. Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science 356, eaaj2239 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  73. Takayama, S. et al. Genome methylation in D. melanogaster is found at specific short motifs and is independent of DNMT2 activity. Genome Res. 24, 821–830 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kelly, W. G. Transgenerational epigenetics in the germline cycle of Caenorhabditis elegans. Epigenetics Chromatin 7, 6 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Li, X.-Y., Harrison, M. M., Villalta, J. E., Kaplan, T. & Eisen, M. B. Establishment of regions of genomic activity during the Drosophila maternal to zygotic transition. eLife 3, e03737 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  76. Bogdanovic, O. et al. Dynamics of enhancer chromatin signatures mark the transition from pluripotency to cell specification during embryogenesis. Genome Res. 22, 2043–2053 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Eissenberg, J. C. & Shilatifard, A. Histone H3 lysine 4 (H3K4) methylation in development and differentiation. Dev. Biol. 339, 240–249 (2010).

    Article  CAS  PubMed  Google Scholar 

  78. Vastenhouw, N. L. et al. Chromatin signature of embryonic pluripotency is established during genome activation. Nature 464, 922–926 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lindeman, L. C. et al. Prepatterning of developmental gene expression by modified histones before zygotic genome activation. Dev. Cell 21, 993–1004 (2011).

    Article  CAS  PubMed  Google Scholar 

  80. Akkers, R. C. et al. A hierarchy of H3K4me3 and H3K27me3 acquisition in spatial gene regulation in Xenopus embryos. Dev. Cell 17, 425–434 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Hontelez, S. et al. Embryonic transcription is controlled by maternally defined chromatin state. Nat. Commun. 6, 10148 (2015).

    Article  CAS  PubMed  Google Scholar 

  82. Chen, K. et al. A global change in RNA polymerase II pausing during the Drosophila midblastula transition. eLife 2, e00861 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Zenk, F. et al. Germ line–inherited H3K27me3 restricts enhancer function during maternal-to-zygotic transition. Science 357, 212–216 (2017).

    Article  CAS  PubMed  Google Scholar 

  84. Gaydos, L. J., Wang, W. & Strome, S. H3K27me and PRC2 transmit a memory of repression across generations and during development. Science 345, 1515–1518 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. 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 

  86. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Zheng, H. et al. Resetting epigenetic memory by reprogramming of histone modifications in mammals. Mol. Cell 63, 1066–1079 (2016).

    Article  CAS  PubMed  Google Scholar 

  88. Weber, C. M. & Henikoff, S. Histone variants: dynamic punctuation in transcription. Genes Dev. 28, 672–682 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Whittle, C. M. et al. The genomic distribution and function of histone variant HTZ-1 during C. elegans embryogenesis. PLOS Genet. 4, e1000187 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Lin, C.-J., Conti, M. & Ramalho-Santos, M. Histone variant H3.3 maintains a decondensed chromatin state essential for mouse preimplantation development. Development 140, 3624–3634 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Yang, P., Wu, W. & Macfarlan, T. S. Maternal histone variants and their chaperones promote paternal genome activation and boost somatic cell reprogramming. Bioessays 37, 52–59 (2015).

    Article  CAS  PubMed  Google Scholar 

  92. Gaume, X. & Torres-Padilla, M.-E. Regulation of reprogramming and cellular plasticity through histone exchange and histone variant incorporation. Cold Spring Harb. Symp. Quant. Biol. 80, 165–175 (2015).

    Article  PubMed  Google Scholar 

  93. Perez-Montero, S., Carbonell, A., Moran, T., Vaquero, A. & Azorin, F. The embryonic linker histone H1 variant of Drosophila, dBigH1, regulates zygotic genome activation. Dev. Cell 26, 578–590 (2013).

    Article  CAS  PubMed  Google Scholar 

  94. Smith, R. C., Dworkin-Rastl, E. & Dworkin, M. B. Expression of a histone H1-like protein is restricted to early Xenopus development. Genes Dev. 2, 1284–1295 (1988).

    Article  CAS  PubMed  Google Scholar 

  95. Fu, G. et al. Mouse oocytes and early embryos express multiple histone H1 subtypes1. Biol. Reprod. 68, 1569–1576 (2003).

    Article  CAS  PubMed  Google Scholar 

  96. Murphy, P. J., Wu, S. F., James, C. R., Wike, C. L. & Cairns, B. R. Placeholder nucleosomes underlie germline-to-embryo DNA methylation reprogramming. Cell 172, 993–1006 (2018).

    Article  CAS  PubMed  Google Scholar 

  97. Zhang, Y. et al. Canonical nucleosome organization at promoters forms during genome activation. Genome Res. 24, 260–266 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Blythe, S. A. & Wieschaus, E. F. Establishment and maintenance of heritable chromatin structure during early Drosophila embryogenesis. eLife 5, e20148 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Lu, F. et al. Establishing chromatin regulatory landscape during mouse preimplantation development. Cell 165, 1375–1388 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Wu, J. et al. The landscape of accessible chromatin in mammalian preimplantation embryos. Nature 534, 652–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  101. Li, L. et al. Single-cell multi-omics sequencing of human early embryos. Nat. Cell Biol. 20, 847–858 (2018).

    Article  CAS  PubMed  Google Scholar 

  102. Wu, J. et al. Chromatin analysis in human early development reveals epigenetic transition during ZGA. Nature 557, 256–260 (2018).

    Article  CAS  PubMed  Google Scholar 

  103. Svoboda, P. et al. RNAi and expression of retrotransposons MuERV-L and IAP in preimplantation mouse embryos. Dev. Biol. 269, 276–285 (2004).

    Article  CAS  PubMed  Google Scholar 

  104. Jachowicz, J. W. et al. LINE-1 activation after fertilization regulates global chromatin accessibility in the early mouse embryo. Nat. Genet. 49, 1502–1510 (2017).

    Article  CAS  PubMed  Google Scholar 

  105. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Ishiuchi, T. et al. Early embryonic-like cells are induced by downregulating replication-dependent chromatin assembly. Nat. Struct. Mol. Biol. 22, 662–671 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. Dixon, J. R., Gorkin, D. U. & Ren, B. Chromatin domains: the unit of chromosome organization. Mol. Cell 62, 668–680 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bonev, B. & Cavalli, G. Organization and function of the 3D genome. Nat. Rev. Genet. 17, 661–678 (2016).

    Article  CAS  PubMed  Google Scholar 

  109. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hug, C. B. & Vaquerizas, J. M. The birth of the 3D genome during early embryonic development. Trends Genet. 34, P903–P914 (2018).

    Article  CAS  Google Scholar 

  111. Hug, C. B., Grimaldi, A. G., Kruse, K. & Vaquerizas, J. M. Chromatin architecture emerges during zygotic genome activation independent of transcription. Cell 169, 216–228 (2017). This paper details the first profile of chromatin conformation changes in the early embryo during the MZT and demonstrates that TAD boundaries are established in concert with ZGA in Drosophila melanogaster.

    Article  CAS  PubMed  Google Scholar 

  112. Ogiyama, Y., Schuettengruber, B., Papadopoulos, G. L., Chang, J.-M. & Cavalli, G. Polycomb-dependent chromatin looping contributes to gene silencing during Drosophila development. Mol. Cell 71, 73–88 (2018).

    Article  CAS  PubMed  Google Scholar 

  113. Ke, Y. et al. 3D chromatin structures of mature gametes and structural reprogramming during mammalian embryogenesis. Cell 170, 367–381 (2017).

    Article  CAS  PubMed  Google Scholar 

  114. Du, Z. et al. Allelic reprogramming of 3D chromatin architecture during early mammalian development. Nature 547, 232–235 (2017).

    Article  CAS  PubMed  Google Scholar 

  115. Kaaij, L. J. T., van der Weide, R. H., Ketting, R. F. & de Wit, E. Systemic loss and gain of chromatin architecture throughout zebrafish development. Cell Rep. 24, 1–10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Liang, H.-L. et al. The zinc-finger protein Zelda is a key activator of the early zygotic genome in Drosophila. Nature 456, 400–403 (2008). This paper shows that Zelda is an essential factor for ZGA in Drosophila melanogaster and in so doing identifies the first major activator of the zygotic genome of any species.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Harrison, M. M., Li, X.-Y., Kaplan, T., Botchan, M. R. & Eisen, M. B. Zelda binding in the early Drosophila melanogaster embryo marks regions subsequently activated at the maternal-to-zygotic transition. PLOS Genet. 7, e1002266 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nien, C. Y. et al. Temporal coordination of gene networks by Zelda in the early Drosophila embryo. PLOS Genet. 7, e1002339 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ribeiro, L. et al. Evolution and multiple roles of the Pancrustacea specific transcription factor zelda in insects. PLOS Genet. 13, e1006868 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Leichsenring, M., Maes, J., Mossner, R., Driever, W. & Onichtchouk, D. Pou5f1 transcription factor controls zygotic gene activation in vertebrates. Science 341, 1005–1009 (2013). The authors demonstrate that Pou5f3 is bound to chromatin before the onset of zygotic transcription and is instrumental in activating early gene expression.

    Article  CAS  PubMed  Google Scholar 

  121. Takahashi, K. & Yamanaka, S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell. Biol. 17, 183–193 (2016).

    Article  CAS  PubMed  Google Scholar 

  122. Le Bin, G. C. et al. Oct4 is required for lineage priming in the developing inner cell mass of the mouse blastocyst. Development 141, 1001–1010 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Hendrickson, P. G. et al. Conserved roles of mouse DUX and human DUX4 in activating cleavage-stage genes and MERVL/HERVL retrotransposons. Nat. Genet. 49, 925–934 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. De Iaco, A. et al. DUX-family transcription factors regulate zygotic genome activation in placental mammals. Nat. Genet. 49, 941–945 (2017). The studies in references 123 and 124 identify the DUX transcription factors as regulators of mammalian ZGA and support this conclusion by manipulating DUX4 and/or DUX levels in mouse embryos and cell lines.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Whiddon, J. L., Langford, A. T., Wong, C.-J., Zhong, J. W. & Tapscott, S. J. Conservation and innovation in the DUX4-family gene network. Nat. Genet. 49, 935–940 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Schulz, K. N. et al. Zelda is differentially required for chromatin accessibility, transcription factor binding, and gene expression in the early Drosophila embryo. Genome Res. 25, 1715–1726 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Sun, Y. et al. Zelda overcomes the high intrinsic nucleosome barrier at enhancers during Drosophila zygotic genome activation. Genome Res. 25, 1703–1714 (2015). The studies in references 126 and 127 demonstrate that Zelda possesses an essential characteristic of pioneer transcription factors, the ability to establish or maintain regions of accessible chromatin.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Veil, M., Yampolsky, L., Gruening, B. & Onichtchouk, D. Pou5f3, SoxB1 and Nanog remodel chromatin on high nucleosome affinity regions at zygotic genome activation. Preprint at bioRxiv https://doi.org/10.1101/344168 (2018).

    Article  Google Scholar 

  129. Oldfield, A. J. et al. Histone-fold domain protein NF-Y promotes chromatin accessibility for cell type-specific master transcription factors. Mol. Cell 55, 708–722 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Nardini, M. et al. Sequence-specific transcription factor NF-Y displays histone-like DNA binding and H2B-like ubiquitination. Cell 152, 132–143 (2013).

    Article  CAS  PubMed  Google Scholar 

  131. Coustry, F., Hu, Q., de Crombrugghe, B. & Maity, S. N. CBF/NF-Y functions both in nucleosomal disruption and transcription activation of the chromatin-assembled topoisomerase IIα promoter. J. Biol. Chem. 276, 40621–40630 (2001).

    Article  CAS  PubMed  Google Scholar 

  132. Choi, S. H. et al. DUX4 recruits p300/CBP through its C-terminus and induces global H3K27 acetylation changes. Nucleic Acids Res. 44, 5161–5173 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Soufi, A., Donahue, G. & Zaret, K. S. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell 151, 994–1004 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zaret, K. S. & Carroll, J. S. Pioneer transcription factors: establishing competence for gene expression. Genes Dev. 25, 2227–2241 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. King, H. W. & Klose, R. J. The pioneer factor OCT4 requires the chromatin remodeller BRG1 to support gene regulatory element function in mouse embryonic stem cells. eL ife 6, e22631 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Stadhouders, R. et al. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nat. Genet. 50, 238–249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Meier, M. et al. Cohesin facilitates zygotic genome activation in zebrafish. Development 145, dev156521 (2018).

    Article  PubMed  CAS  Google Scholar 

  139. Mir, M. et al. Dense Bicoid hubs accentuate binding along the morphogen gradient. Genes Dev. 31, 1784–1794 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Liu, Z. et al. 3D imaging of Sox2 enhancer clusters in embryonic stem cells. eL ife 3, e04236 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  141. Hamm, D. C., Bondra, E. R. & Harrison, M. M. Transcriptional activation is a conserved feature of the early embryonic factor Zelda that requires a cluster of four zinc fingers for DNA binding and a low-complexity activation domain. J. Biol. Chem. 290, 3508–3518 (2015).

    Article  CAS  PubMed  Google Scholar 

  142. Mir, M. et al. Dynamic multifactor hubs interact transiently with sites of active transcription in Drosophila embryos. eLife (in the press).

    Article  Google Scholar 

  143. Dufourt, J. et al. Temporal control of gene expression by the pioneer factor Zelda through transient interactions in hubs. Nat Commun. 9, 5194 (2018).

    Article  Google Scholar 

  144. Staudt, N., Fellert, S., Chung, H.-R., Jäckle, H. & Vorbrüggen, G. Mutations of the Drosophila zinc finger-encoding gene vielfältig impair mitotic cell divisions and cause improper chromosome segregation. Mol. Biol. Cell 17, 2356–2365 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Hamm, D. C. et al. A conserved maternal-specific repressive domain in Zelda revealed by Cas9-mediated mutagenesis in Drosophila melanogaster. PLOS Genet. 13, e1007120 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Onichtchouk, D. & Driever, W. Zygotic genome activators, developmental timing, and pluripotency. Curr. Top. Dev. Biol. 116, 273–297 (2016).

    Article  PubMed  Google Scholar 

  147. Lunde, K., Belting, H.-G. & Driever, W. Zebrafish pou5f1/pou2, homolog of mammalian Oct4, functions in the endoderm specification cascade. Curr. Biol. 14, 48–55 (2004).

    Article  CAS  PubMed  Google Scholar 

  148. Onichtchouk, D. et al. Oct4/Pou5f1 controls tissue-specific repressors in early zebrafish embryo. J. Stem Cells Regen. Med. 6, 82 (2010).

    CAS  PubMed  Google Scholar 

  149. Veil, M. et al. Maternal Nanog is required for zebrafish embryo architecture and for cell viability during gastrulation. Development 145, dev155366 (2018).

    Article  PubMed  CAS  Google Scholar 

  150. Gagnon, J. A., Obbad, K. & Schier, A. F. The primary role of zebrafish nanog is in extra-embryonic tissue. Development 145, dev147793 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Vanderplanck, C. et al. The FSHD atrophic myotube phenotype is caused by DUX4 expression. PLOS ONE 6, e26820 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Lemmers, R. J. L. F. et al. A unifying genetic model for facioscapulohumeral muscular dystrophy. Science 329, 1650–1653 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Geng, L. N. et al. DUX4 activates germline genes, retroelements, and immune mediators: implications for facioscapulohumeral dystrophy. Dev. Cell 22, 38–51 (2012).

    Article  CAS  PubMed  Google Scholar 

  154. Young, J. M. et al. DUX4 binding to retroelements creates promoters that are active in FSHD muscle and testis. PLOS Genet. 9, e1003947 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Iglesias, J. M., Gumuzio, J. & Martin, A. G. Linking pluripotency reprogramming and cancer. Stem Cells Transl Med. 6, 335–339 (2017).

    Article  PubMed  Google Scholar 

  156. Liu, A., Yu, X. & Liu, S. Pluripotency transcription factors and cancer stem cells: small genes make a big difference. Chin. J. Cancer 32, 483–487 (2013).

    PubMed  PubMed Central  Google Scholar 

  157. Adamson, E. D. & Woodland, H. R. Histone synthesis in early amphibian development: histone and DNA syntheses are not co-ordinated. J. Mol. Biol. 88, 263–285 (1974).

    Article  CAS  PubMed  Google Scholar 

  158. Wang, Y. et al. Unique molecular events during reprogramming of human somatic cells to induced pluripotent stem cells (iPSCs) at naïve state. eLife 7, e29518 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  159. Deng, W. et al. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158, 849–860 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Henikoff, S. & Shilatifard, A. Histone modification: cause or cog? Trends Genet. 27, 389–396 (2011).

    Article  CAS  PubMed  Google Scholar 

  161. Bazzini, A. A. et al. Codon identity regulates mRNA stability and translation efficiency during the maternal-to-zygotic transition. EMBO J. 35, 2087–2103 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Mishima, Y. & Tomari, Y. Codon usage and 3′ UTR length determine maternal mRNA stability in zebrafish. Mol. Cell 61, 874–885 (2016).

    Article  CAS  PubMed  Google Scholar 

  163. Ivanova, I. et al. The RNA m6A reader YTHDF2 is essential for the post-transcriptional regulation of the maternal transcriptome and oocyte competence. Mol. Cell 67, 1059–1067 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Zhao, B. S. et al. m6A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature 542, 475–478 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Edgar, B. A. & Datar, S. A. Zygotic degradation of two maternal Cdc25 mRNAs terminates Drosophila’s early cell cycle program. Genes Dev. 10, 1966–1977 (1996).

    Article  CAS  PubMed  Google Scholar 

  166. Bashirullah, A. et al. Joint action of two RNA degradation pathways controls the timing of maternal transcript elimination at the midblastula transition in Drosophila melanogaster. EMBO J. 18, 2610–2620 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Hamatani, T., Carter, M. G., Sharov, A. A. & Ko, M. S. H. Dynamics of global gene expression changes during mouse preimplantation development. Dev. Cell 6, 117–131 (2004).

    Article  CAS  PubMed  Google Scholar 

  168. Giraldez, A. J. et al. Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science 312, 75–79 (2006).

    Article  CAS  PubMed  Google Scholar 

  169. Lund, E., Liu, M., Hartley, R. S., Sheets, M. D. & Dahlberg, J. E. Deadenylation of maternal mRNAs mediated by miR-427 in Xenopus laevis embryos. RNA 15, 2351–2363 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Bushati, N., Stark, A., Brennecke, J. & Cohen, S. M. Temporal reciprocity of mi-RNAs and their targets during the maternal-to-zygotic transition in Drosophila. Curr. Biol. 18, 501–506 (2008).

    Article  CAS  PubMed  Google Scholar 

  171. Yan, L. et al. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1131–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  172. Briggs, J. A. et al. The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution. Science 360, eaar5780 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Farrell, J. A. et al. Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis. Science 360, eaar3131 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Wagner, D. E. et al. Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo. Science 360, 981–987 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Sawant, A. A. et al. A versatile toolbox for posttranscriptional chemical labeling and imaging of RNA. Nucleic Acids Res. 44, e16 (2016).

    Article  PubMed  CAS  Google Scholar 

  176. Strom, A. R. et al. Phase separation drives heterochromatin domain formation. Nature 547, 241–245 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Mir, M. et al. Single molecule imaging in live embryos using lattice light-sheet microscopy. Methods Mol. Biol. 1814, 541–559 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  178. Bothma, J. P., Norstad, M. R., Alamos, S. & Garcia, H. G. LlamaTags: a versatile tool to image transcription factor dynamics in live embryos. Cell 173, 1810–1822 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Campbell, P. D., Chao, J. A., Singer, R. H. & Marlow, F. L. Dynamic visualization of transcription and RNA subcellular localization in zebrafish. Development 142, 1368–1374 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Ferraro, T. et al. Transcriptional memory in the Drosophila embryo. Curr. Biol. 26, 212–218 (2016).

    Article  CAS  PubMed  Google Scholar 

  181. Bothma, J. P. et al. Dynamic regulation of eve stripe 2 expression reveals transcriptional bursts in living Drosophila embryos. Proc. Natl Acad. Sci. USA 111, 10598–10603 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Senecal, A. et al. Transcription factors modulate c-Fos transcriptional bursts. Cell Rep. 8, 75–83 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Garcia, H. G., Tikhonov, M., Lin, A. & Gregor, T. Quantitative imaging of transcription in living Drosophila embryos links polymerase activity to patterning. Curr. Biol. 23, 2140–2145 (2013).

    Article  CAS  PubMed  Google Scholar 

  184. Deng, W., Shi, X., Tjian, R., Lionnet, T. & Singer, R. H. CASFISH: CRISPR/Cas9-mediated in situ labeling of genomic loci in fixed cells. Proc. Natl Acad. Sci. USA 112, 11870–11875 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Qin, P. et al. Live cell imaging of low- and non-repetitive chromosome loci using CRISPR-Cas9. Nat. Commun. 8, 14725 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. O’Connell, M. R. et al. Programmable RNA recognition and cleavage by CRISPR/Cas9. Nature 516, 263–266 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Gualdi, R. et al. Hepatic specification of the gut endoderm in vitro: cell signaling and transcriptional control. Genes Dev. 10, 1670–1682 (1996).

    Article  CAS  PubMed  Google Scholar 

  188. Lee, C. S., Friedman, J. R., Fulmer, J. T. & Kaestner, K. H. The initiation of liver development is dependent on Foxa transcription factors. Nature 435, 944–947 (2005).

    Article  CAS  PubMed  Google Scholar 

  189. Zaret, K. S. & Mango, S. E. Pioneer transcription factors, chromatin dynamics, and cell fate control. Curr. Opin. Genet. Dev. 37, 76–81 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Blythe, S. A., Cha, S.-W., Tadjuidje, E., Heasman, J. & Klein, P. S. β-Catenin primes organizer gene expression by recruiting a histone H3 arginine 8 methyltransferase, Prmt2. Dev. Cell 19, 220–231 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Cirillo, L. A. et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 (2002).

    Article  CAS  PubMed  Google Scholar 

  192. Clark, K. L., Halay, E. D., Lai, E. & Burley, S. K. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 (1993).

    Article  CAS  PubMed  Google Scholar 

  193. Taube, J. H., Allton, K., Duncan, S. A., Shen, L. & Barton, M. C. Foxa1 functions as a pioneer transcription factor at transposable elements to activate Afp during differentiation of embryonic stem cells. J. Biol. Chem. 285, 16135–16144 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Lachnit, M., Kur, E. & Driever, W. Alterations of the cytoskeleton in all three embryonic lineages contribute to the epiboly defect of Pou5f1/Oct4 deficient MZspg zebrafish embryos. Dev. Biol. 315, 1–17 (2008).

    Article  CAS  PubMed  Google Scholar 

  195. Reim, G., Mizoguchi, T., Stainier, D. Y., Kikuchi, Y. & Brand, M. The POU domain protein spg (pou2/Oct4) is essential for endoderm formation in cooperation with the HMG domain protein casanova. Dev. Cell 6, 91–101 (2004).

    Article  CAS  PubMed  Google Scholar 

  196. Reim, G. & Brand, M. Maternal control of vertebrate dorsoventral axis formation and epiboly by the POU domain protein Spg/Pou2/Oct4. Development 133, 2757–2770 (2006).

    Article  CAS  PubMed  Google Scholar 

  197. Okuda, Y., Ogura, E., Kondoh, H. & Kamachi, Y. B1 SOX coordinate cell specification with patterning and morphogenesis in the early zebrafish embryo. PLOS Genet. 6, e1000936 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Xu, C. et al. Nanog-like regulates endoderm formation through the Mxtx2-Nodal pathway. Dev. Cell 22, 625–638 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Fogarty, N. M. E. et al. Genome editing reveals a role for OCT4 in human embryogenesis. Nature 550, 67–73 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank members of the Harrison laboratory and the reviewers for helpful feedback on the manuscript. K.N.S. was supported in part by the National Institutes of Health (NIH) National Research Service award T32 GM007215. M.M.H. was supported by grant R01GM11694 from the National Institute of General Medical Sciences and a Vallee Scholar Award.

Reviewer information

Nature Reviews Genetics thanks B. Cairns, K. Kuznetsova, N. Vastenhouw and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Melissa M. Harrison.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Totipotent

The property of a cell with the capacity to form all the cells of an organism, including extra-embryonic tissues.

Zygotic

Relating to the diploid, fertilized egg cell (zygote) that results from the fusion of an egg and a sperm.

Germ layers

The three layers of cells (ectoderm, mesoderm and endoderm) that are formed during gastrulation in the early embryo and differentiate to give rise to all the organs and tissues of the body.

Chromatin

The complex of DNA, RNA and protein that makes up the chromosomes of eukaryotes.

Cleavage divisions

The rapid, modified cell cycles of the early embryo that consist of only M (mitosis) and S (replication) phases and omit the G1 and G2 gap phases. These cycles occur in the absence of cell growth and therefore result in no change in the size of the embryo.

Nucleocytoplasmic ratio

(N:C ratio). The ratio of the nuclear content to the cytoplasmic content in a cell or embryo.

Polyspermic

Refers to an egg that has been fertilized by more than one sperm and thus contains three or more copies of each chromosome.

Haploid

Characterized by a single set of chromosomes. Most animals have diploid somatic cells (with two paired sets of chromosomes) but produce haploid gametes.

Compound chromosomes

Chromosomes formed by the attachment of two homologues through a single centromere that are therefore inherited together through mitosis and meiosis. They can be used to generate embryos deficient for an entire chromosome.

Protamines

Small, basic proteins that are used in the place of histones to help package DNA in the sperm of some species.

Demethylation

The process by which a demethylase enzyme removes a methyl group from a molecule.

Transposable elements

DNA sequences that can move from one position within the genome to another.

Topologically associating domains

(TADs). 3D chromosome structures within which DNA regions physically interact with each other more frequently than with regions outside.

MERVL

A family of endogenous retroviruses expressed in mouse embryos during zygotic genome activation. The human version is known as HERVL.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schulz, K.N., Harrison, M.M. Mechanisms regulating zygotic genome activation. Nat Rev Genet 20, 221–234 (2019). https://doi.org/10.1038/s41576-018-0087-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-018-0087-x

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