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.

  • Article
  • Published:

BTG4 is a meiotic cell cycle–coupled maternal-zygotic-transition licensing factor in oocytes

Abstract

The mRNAs stored in oocytes undergo general decay during the maternal-zygotic transition (MZT), and their stability is tightly interconnected with meiotic cell-cycle progression. However, the factors that trigger decay of maternal mRNA and couple this event to oocyte meiotic maturation remain elusive. Here, we identified B-cell translocation gene-4 (BTG4) as an MZT licensing factor in mice. BTG4 bridged CNOT7, a catalytic subunit of the CCR4–NOT deadenylase, to eIF4E, a key translation initiation factor, and facilitated decay of maternal mRNA. Btg4-null females produced morphologically normal oocytes but were infertile, owing to early developmental arrest. The intrinsic MAP kinase cascade in oocytes triggered translation of Btg4 mRNA stored in fully grown oocytes by targeting the 3′ untranslated region, thereby coupling CCR4–NOT deadenylase–mediated decay of maternal mRNA with oocyte maturation and fertilization. This is a key step in oocyte cytoplasmic maturation that determines the developmental potential of mammalian embryos.

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: Btg4 is a maternal-effect gene essential for MZT.
Figure 2: BTG4 is required for decay of maternal mRNA.
Figure 3: BTG4 interacts with CCR4–NOT transcription-complex subunits CNOT7 and CNOT8 and is required for 3′-poly(A)-tail shortening of maternal transcripts.
Figure 4: Interaction with CNOT7 and CNOT8 is essential for BTG4 function in vivo.
Figure 5: BTG4 is an adaptor protein that mediates the interaction between CNOT7 and eIF4E.
Figure 6: Btg4-mRNA translation is coupled to the meiotic cell cycle by its 3′ UTR.
Figure 7: ERK1/2 triggers Btg4-mRNA translation by targeting its 3′ UTR.
Figure 8: Summary of BTG4 function and regulation during MZT.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Schier, A.F. The maternal-zygotic transition: death and birth of RNAs. Science 316, 406–407 (2007).

    CAS  PubMed  Google Scholar 

  2. Li, L., Lu, X. & Dean, J. The maternal to zygotic transition in mammals. Mol. Aspects Med. 34, 919–938 (2013).

    PubMed Central  PubMed  Google Scholar 

  3. Schultz, R.M. From egg to embryo: a peripatetic journey. Reproduction 130, 825–828 (2005).

    CAS  PubMed  Google Scholar 

  4. Li, L., Baibakov, B. & Dean, J. A subcortical maternal complex essential for preimplantation mouse embryogenesis. Dev. Cell 15, 416–425 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  5. Christians, E., Davis, A.A., Thomas, S.D. & Benjamin, I.J. Maternal effect of Hsf1 on reproductive success. Nature 407, 693–694 (2000).

    CAS  PubMed  Google Scholar 

  6. Burns, K.H. et al. Roles of NPM2 in chromatin and nucleolar organization in oocytes and embryos. Science 300, 633–636 (2003).

    CAS  PubMed  Google Scholar 

  7. Wu, X. et al. Zygote arrest 1 (Zar1) is a novel maternal-effect gene critical for the oocyte-to-embryo transition. Nat. Genet. 33, 187–191 (2003).

    CAS  PubMed  Google Scholar 

  8. Walser, C.B. & Lipshitz, H.D. Transcript clearance during the maternal-to-zygotic transition. Curr. Opin. Genet. Dev. 21, 431–443 (2011).

    CAS  PubMed  Google Scholar 

  9. Schellander, K., Hoelker, M. & Tesfaye, D. Selective degradation of transcripts in mammalian oocytes and embryos. Theriogenology 68 (Suppl. 1), S107–S115 (2007).

    CAS  PubMed  Google Scholar 

  10. Hou, Y. et al. Genome analyses of single human oocytes. Cell 155, 1492–1506 (2013).

    CAS  PubMed  Google Scholar 

  11. Gallardo, T.D. et al. Genomewide discovery and classification of candidate ovarian fertility genes in the mouse. Genetics 177, 179–194 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  12. Qiu, Z. et al. High-efficiency and heritable gene targeting in mouse by transcription activator-like effector nucleases. Nucleic Acids Res. 41, e120 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  13. Huang, P. et al. Heritable gene targeting in zebrafish using customized TALENs. Nat. Biotechnol. 29, 699–700 (2011).

    PubMed  Google Scholar 

  14. Winkler, G.S. The mammalian anti-proliferative BTG/Tob protein family. J. Cell. Physiol. 222, 66–72 (2010).

    CAS  PubMed  Google Scholar 

  15. Tirone, F. The gene PC3(TIS21/BTG2), prototype member of the PC3/BTG/TOB family: regulator in control of cell growth, differentiation, and DNA repair? J. Cell. Physiol. 187, 155–165 (2001).

    CAS  PubMed  Google Scholar 

  16. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  17. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Charlesworth, A., Meijer, H.A. & de Moor, C.H. Specificity factors in cytoplasmic polyadenylation. Wiley Interdiscip. Rev. RNA 4, 437–461 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  19. Buanne, P. et al. Cloning of PC3B, a novel member of the PC3/BTG/TOB family of growth inhibitory genes, highly expressed in the olfactory epithelium. Genomics 68, 253–263 (2000).

    CAS  PubMed  Google Scholar 

  20. Mauxion, F., Chen, C.Y., Séraphin, B. & Shyu, A.B. BTG/TOB factors impact deadenylases. Trends Biochem. Sci. 34, 640–647 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  21. Doidge, R., Mittal, S., Aslam, A. & Winkler, G.S. Deadenylation of cytoplasmic mRNA by the mammalian Ccr4-Not complex. Biochem. Soc. Trans. 40, 896–901 (2012).

    CAS  PubMed  Google Scholar 

  22. Doidge, R., Mittal, S., Aslam, A. & Winkler, G.S. The anti-proliferative activity of BTG/TOB proteins is mediated via the Caf1a (CNOT7) and Caf1b (CNOT8) deadenylase subunits of the Ccr4-not complex. PLoS One 7, e51331 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Miller, J.E. & Reese, J.C. Ccr4-Not complex: the control freak of eukaryotic cells. Crit. Rev. Biochem. Mol. Biol. 47, 315–333 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Horiuchi, M. et al. Structural basis for the antiproliferative activity of the Tob-hCaf1 complex. J. Biol. Chem. 284, 13244–13255 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  25. Winkler, G.S. & Balacco, D.L. Heterogeneity and complexity within the nuclease module of the Ccr4-Not complex. Front. Genet. 4, 296 (2013).

    PubMed Central  PubMed  Google Scholar 

  26. Gosselin, P. et al. Tracking a refined eIF4E-binding motif reveals Angel1 as a new partner of eIF4E. Nucleic Acids Res. 41, 7783–7792 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Matsuo, H. et al. Structure of translation factor eIF4E bound to m7GDP and interaction with 4E-binding protein. Nat. Struct. Biol. 4, 717–724 (1997).

    CAS  PubMed  Google Scholar 

  28. Marcotrigiano, J., Gingras, A.C., Sonenberg, N. & Burley, S.K. Cocrystal structure of the messenger RNA 5′ cap-binding protein (eIF4E) bound to 7-methyl-GDP. Cell 89, 951–961 (1997).

    CAS  PubMed  Google Scholar 

  29. Ptushkina, M. et al. Cooperative modulation by eIF4G of eIF4E-binding to the mRNA 5′ cap in yeast involves a site partially shared by p20. EMBO J. 17, 4798–4808 (1998).

    CAS  PubMed Central  PubMed  Google Scholar 

  30. Sarkissian, M., Mendez, R. & Richter, J.D. Progesterone and insulin stimulation of CPEB-dependent polyadenylation is regulated by Aurora A and glycogen synthase kinase-3. Genes Dev. 18, 48–61 (2004).

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Richter, J.D. Breaking the code of polyadenylation-induced translation. Cell 132, 335–337 (2008).

    CAS  PubMed  Google Scholar 

  32. Mendez, R. et al. Phosphorylation of CPE binding factor by Eg2 regulates translation of c-mos mRNA. Nature 404, 302–307 (2000).

    CAS  PubMed  Google Scholar 

  33. Groisman, I. et al. CPEB, maskin, and cyclin B1 mRNA at the mitotic apparatus: implications for local translational control of cell division. Cell 103, 435–447 (2000).

    CAS  PubMed  Google Scholar 

  34. Eliscovich, C., Peset, I., Vernos, I. & Méndez, R. Spindle-localized CPE-mediated translation controls meiotic chromosome segregation. Nat. Cell Biol. 10, 858–865 (2008).

    CAS  PubMed  Google Scholar 

  35. Ellis, R.E. & Kimble, J. The fog-3 gene and regulation of cell fate in the germ line of Caenorhabditis elegans. Genetics 139, 561–577 (1995).

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Chen, P.J., Cho, S., Jin, S.W. & Ellis, R.E. Specification of germ cell fates by FOG-3 has been conserved during nematode evolution. Genetics 158, 1513–1525 (2001).

    CAS  PubMed Central  PubMed  Google Scholar 

  37. Ezzeddine, N. et al. Human TOB, an antiproliferative transcription factor, is a poly(A)-binding protein-dependent positive regulator of cytoplasmic mRNA deadenylation. Mol. Cell. Biol. 27, 7791–7801 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Richter, J.D. & Lasko, P. Translational control in oocyte development. Cold Spring Harb. Perspect. Biol. 3, a002758 (2011).

    PubMed Central  PubMed  Google Scholar 

  39. Piqué, M., López, J.M., Foissac, S., Guigó, R. & Méndez, R. A combinatorial code for CPE-mediated translational control. Cell 132, 434–448 (2008).

    PubMed  Google Scholar 

  40. Chen, J. et al. Genome-wide analysis of translation reveals a critical role for deleted in azoospermia-like (Dazl) at the oocyte-to-zygote transition. Genes Dev. 25, 755–766 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  41. Wolf, J. et al. Structural basis for Pan3 binding to Pan2 and its function in mRNA recruitment and deadenylation. EMBO J. 33, 1514–1526 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  42. Zheng, D. et al. Deadenylation is prerequisite for P-body formation and mRNA decay in mammalian cells. J. Cell Biol. 182, 89–101 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  43. Fan, H.Y. et al. MAPK3/1 (ERK1/2) in ovarian granulosa cells are essential for female fertility. Science 324, 938–941 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Lan, Z.J., Xu, X. & Cooney, A.J. Differential oocyte-specific expression of Cre recombinase activity in GDF-9-iCre, Zp3cre, and Msx2Cre transgenic mice. Biol. Reprod. 71, 1469–1474 (2004).

    CAS  PubMed  Google Scholar 

  45. Shen, B. et al. Generation of gene-modified mice via Cas9/RNA-mediated gene targeting. Cell Res. 23, 720–723 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  46. Zhou, J. et al. One-step generation of different immunodeficient mice with multiple gene modifications by CRISPR/Cas9 mediated genome engineering. Int. J. Biochem. Cell Biol. 46, 49–55 (2014).

    CAS  PubMed  Google Scholar 

  47. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

This study was funded by the National Basic Research Program of China (2012CB944403) and the National Natural Science Foundation of China (31528016, 91519313, and 31371449) to H.-Y.F. We thank B. Zhang (Peking University) for vectors.

Author information

Authors and Affiliations

Authors

Contributions

H.-Y.F. and C.Y. conceived the project. H.-Y.F., C.Y., S.-Y.J., Q.-Q.S., Y.D., Y.-L.Z., and F.T. designed and analyzed experiments. C.Y., S.-Y.J., Q.-Q.S., Y.D., Y.-L.Z., J.-J.Z., Y.L., B.H., and S.-C.S. performed experiments. Z.-W.W. and Q.-Y.S. assisted in microinjection of mouse embryos. C.Y., Y.-L.Z., and H.-Y.F. wrote the paper. C.Y., S.-Y.J., Q.-Q.S., Y.D., J.-J.Z., and Y.-L.Z. contributed equally to this work.

Corresponding author

Correspondence to Heng-Yu Fan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Phenotypic analyses of Btg4-knockout mice.

(a) Quantitative RT-PCR results showing relative expression levels of mouse Btg4 in somatic tissues, oocytes (GV and MII), and early embryos (1-cell, 2-cell, 4-cell and blastula). Error bars indicate S.E.M. (b) PCR results of tail genomic DNAs. The WT allele (“+”) was amplified with WT-F and WT-R primers, and a 7-nucleotide-deleted null allele (“−”) was amplified with the primer pairs GT-F and GT-R. “+/–” and “−/−” refer to mice heterozygous and homozygous for the Btg4 mutant allele, respectively. (c) Average body weights of 3-week-old WT, Btg4+/−, and Btg4−/− females. n=9 for each genotype. Error bars indicate S.E.M. (d) Ovarian histology of WT and Btg4−/− females (3-week and 18-week old). Scale bar: 200 μm. (e) The average numbers of ovulated oocytes by 3-week-old WT (n=4) and Btg4−/− (n=5) females, as determined by a superovulation assay. Error bars indicate S.E.M. (f) Average in vitro GVBD and PB1 emission (PBE) rates of GV oocytes collected from WT and Btg4−/− females. The numbers of WT and Btg4−/− oocytes analyzed were 87 and 105, respectively. Error bars indicate S.E.M. (g) Representative images of early embryos collected from the oviducts of WT, Btg4−/− and Btg4 C-terminal truncated (Btg4ΔC/ΔC) females. n = 5 mice for each genotype at different developmental stages. Scale bar: 100 μm. (h) Pups born from Btg4−/− and WT foster mothers after embryo transfer. The genotypes of foster mothers and transferred embryos are indicated. (i) Genotyping results showing that the pups born by Btg4−/− foster mothers are Btg4 wild types.

Supplementary Figure 2 Immunofluorescence staining for markers of DNA damage and genome reprogramming in embryos derived from WT and Btg4−/− females.

(a) Immunofluorescent staining for DNA damage marker (γH2AX) in embryos derived from WT and Btg4−/− females. Scale bar; 10 μm. (b) BrdU incorporation assay showing DNA replication in zygotes derived from WT and Btg4−/− females mated with WT males. More than 20 embryos were analyzed for each genotype with different markers. Scale bar: 10 μm. (c) Immunofluorescent staining of 5‑methylcytosine (5mC) and 5‑hydroxymethylcytosine (5hmC) in zygotes derived from WT and Btg4−/− females mated with WT males. Scale bar: 10 μm. (d,e) Immunofluorescent staining of trimethylated histone H3 at lysine-4 (H3K4me3) (d) and lysine-9 (H3K9me3) (e) in zygotes derived from WT and Btg4−/− females mated with WT males. Scale bar: 10 μm.

Supplementary Figure 3 Immunofluorescence staining of genome transcriptional-activation markers in embryos derived from WT and Btg4−/− females.

(a) Immunofluorescent staining of phosphorylated RNA polymerase II CTD repeat YSPTSPS (pS2) in 1- or 2-cell embryos derived from WT and Btg4−/− females mated with WT males. Scale bar: 10 μm. (b-d) Immunofluorescent staining of acetylated histone H2B at lysine-5 (H2BK5-ace) (b), H3 at lysine-9 (H2BK9-ace) (c), and H3 at lysine-18 (H2BK18-ace) (d), in 1- or 2-cell embryos derived from WT and Btg4−/− females mated with WT males. Scale bar: 10 μm.

Supplementary Figure 4 Degradation of maternal mRNA during meiotic maturation and fertilization in WT and Btg4−/− oocytes.

(a) Numbers of gene transcripts detected by RNA-seq. (b) Down-regulated and up-regulated genes in WT and Btg4−/− oocytes/embryos at different stages. When comparing two samples, the up-regulation and down-regulation means the upper right one compared to the lower left one. (c) Heatmap illustration showing differentially expressed transcripts in WT oocytes/embryos. Cluster I, transcripts that are significantly degraded at GV to MII transition and not re-accumulate after that in WT oocytes. Cluster II, transcripts that are significantly degraded upon fertilization in WT oocytes. Cluster III, transcripts that are significantly up-regulated at zygote to 2-cell transition (or, zygotic genes). Cluster IV, transcripts that are significantly degraded at GV to MII transition but re-accumulated at zygote to 2-cell transition. (d) Heatmap illustration showing gene transcripts that are degraded during meiotic maturation in WT oocytes but failed to be degraded in Btg4−/− oocytes. (e) Heatmap illustration showing gene transcripts that are up-regulated during zygote to 2-cell transition in WT embryos but are not up-regulated in Btg4−/− 2-cell embryos.

Source data

Supplementary Figure 5 Btg4 knockout impairs zygotic genome activation.

(a) Overlapping results showing the numbers and percentages of genes up-regulated in Btg4-deleted oocytes at MII oocytes and zygotes. (b) Overlapping results showing the numbers and percentages of genes up-regulated and down-regulated in Btg4−/− at 2-cell stage when compared to WT 2-cell embryos. (c) RNA-seq results showing the relative expression levels of representative genes. The error bars indicate S.E.M. (d) qRT-PCR showing the relative levels of indicated transcripts in oocytes/zygotes of WT and Btg4−/− females. Error bars indicates S.E.M. (e) RNA-seq results showing the expression levels of Btg and Tob family genes in human oocytes and early embryos. FPKM numbers were extracted from previously published data1. (f) RNA-seq results showing mRNA expression levels of Btg/Tob family genes in mouse oocytes and early embryos. (g) RNA-seq results showing mRNA expression levels of Cnot7/8 in human oocytes and early embryos. FPKM numbers were extracted from previously published data. (h) Quantitative RT-PCR results showing the relative expression levels of mouse Cnot7/8 in somatic tissues, oocytes, and early embryos. Error bars indicate S.E.M.

Supplementary Figure 6 BTG proteins interact with eIF4E.

(a) Deadenylation assay demonstrating that BTG4 binding did not affect CNOT7 activity in vitro. (b) Co-IP results showing the interactions of BTG2, BTG3, and BTG4 with eIF4E. (c) Co-IP results showing the interactions of BTG2 and BTG3 mutant forms with eIF4E. (d-e) Co-IP results showing the interactions of eIF4E4 mutant forms with BTG4.

Supplementary Figure 7 Regulation of Btg4-mRNA translation by its 3′ UTR.

(a-b) Epifluorescence (a) and Western blot (b) results showing that injected mRNAs encoded for GFP-BTG4 were expressed in both GV and MII oocytes, but endogenous BTG4 was only detected in MII oocytes. Scale bar: 100 μm. (c) Fluorescent microscopy results showing meiotic maturation coupled activation of Btg4 3′-UTR. Scale bar: 100 μm. In vitro transcribed FLAG-GFP-(Btg4)3′-UTR mRNA and mCherry mRNA were co-injected into GV oocytes. The injected oocytes were then further cultured in M2 medium with or without milrinone for 12 h. (d) Polyadenylation enabled the expression of FLAG-GFP-(Btg4)3′-UTR mRNA in GV oocytes, even in the absence of CPE elements. Scale bar: 100 μm. In vitro transcribed WT and CPE-deleted FLAG-GFP-(Btg4)3′-UTR mRNAs were polyadenylated in vitro and injected into GV oocytes. The injected oocytes were than further cultured in M2 medium with or without milrinone for 12 h.

Supplementary Figure 8 Effects of Cpeb1 depletion and aurora A inhibition on mouse oocyte maturation.

(a) qRT-PCR results showing the knockdown efficiency of Cpeb1 in GV oocytes. Error bars indicate S.E.M. (b-c) Injection of Cpeb1 targeted siRNA (siCpeb1) impaired PB1 emission during in vitro mouse oocyte maturation. Error bars indicate S.E.M. Scale bar: 100 μm. (d) Injection of siCpeb1 impaired PB1 emission and disrupted chromosome alignment in the middle of the spindle. Scale bar: 10 μm. (e) MLN8273 did not block BTG4 expression and ERK1/2 phosphorylation in mature oocytes. (f-g) The aurora A inhibitor, MLN8273 blocked PB1 emission, and disrupted chromosome alignment in the middle of the spindle during in vitro mouse oocyte maturation. Scale bar: 10 μm. Error bars indicate S.E.M. (h) The morphology of MLN8237 treated oocytes showed that MLN8273 blocked PB1 emission during in vitro mouse oocyte maturation. Scale bar: 100 μm.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 2–4 (PDF 8657 kb)

Supplementary Table 1

FPKM of RNA-seq results (XLS 8170 kb)

Supplementary Data Set 1

Original images of gels and blots used in this study (PDF 5498 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, C., Ji, SY., Sha, QQ. et al. BTG4 is a meiotic cell cycle–coupled maternal-zygotic-transition licensing factor in oocytes. Nat Struct Mol Biol 23, 387–394 (2016). https://doi.org/10.1038/nsmb.3204

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3204

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