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:

The homeobox transcription factor DUXBL controls exit from totipotency

Abstract

In mice, exit from the totipotent two-cell (2C) stage embryo requires silencing of the 2C-associated transcriptional program. However, the molecular mechanisms involved in this process remain poorly understood. Here we demonstrate that the 2C-specific transcription factor double homeobox protein (DUX) mediates an essential negative feedback loop by inducing the expression of DUXBL to promote this silencing. We show that DUXBL gains accessibility to DUX-bound regions specifically upon DUX expression. Furthermore, we determine that DUXBL interacts with TRIM24 and TRIM33, members of the TRIM superfamily involved in gene silencing, and colocalizes with them in nuclear foci upon DUX expression. Importantly, DUXBL overexpression impairs 2C-associated transcription, whereas Duxbl inactivation in mouse embryonic stem cells increases DUX-dependent induction of the 2C-transcriptional program. Consequently, DUXBL deficiency in embryos results in sustained expression of 2C-associated transcripts leading to early developmental arrest. Our study identifies DUXBL as an essential regulator of totipotency exit enabling the first divergence of cell fates.

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: The Duxbl gene generates two protein products upon DUX expression.
Fig. 2: DUXBL counteracts 2CLC conversion.
Fig. 3: DUXBL overexpression in zygotes halts development.
Fig. 4: DUXBL is essential for mouse embryo development.
Fig. 5: DUX-mediated chromatin opening facilitates DUXBL binding to DUX-bound regions.
Fig. 6: DUXBL interacts with the TRIM33/TRIM24 complex.
Fig. 7: DUXBL and TRIM24/TRIM33 colocalize with H3K9me3 deposition.

Similar content being viewed by others

Data availability

The proteomic data and search results associated with this study have been deposited to the ProteomeXchange Consortium via the MassIVE partner repository with the dataset identifier PXD047807, project doi:10.25345/C5XW4867R. The sequencing data generated in this study have been deposited in the Gene Expression Omnibus database under accession codes GSE210892 and GSE213703. Datasets obtained from publicly available sources include GSE98149, GSE95517, GSE45719, GSE85627, GSE66582 and GSE97304 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc = GSE98149, GSE95517, GSE45719, GSE85627, GSE66582 and GSE97304, respectively). Additional data and/or reagents that support the findings of this study are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

Code availability

The code used in this manuscript to analyze Cut&Run data and expression of TEs is available on Zenodo via the following links:

Cut&Run pipeline: https://doi.org/10.5281/zenodo.10658411 (ref. 61).

Gene/TE quantification pipeline: https://doi.org/10.5281/zenodo.10658421 (ref. 62).

References

  1. Riveiro, A. R. & Brickman, J. M. From pluripotency to totipotency: an experimentalist’s guide to cellular potency. Development 147, dev189845 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Sha, Q. Q., Zhang, J. & Fan, H. Y. A story of birth and death: mRNA translation and clearance at the onset of maternal-to-zygotic transition in mammals. Biol. Reprod. 101, 579–590 (2019).

    Article  PubMed  Google Scholar 

  3. Schultz, K. N. & Harrison, M. M. Mechanisms regulating zygotic genome activation. Nat. Rev. Genet. 20, 221–234 (2019).

    Article  Google Scholar 

  4. Jukamm, D., Shariati, S. A. M. & Skotheim, J. M. Zygotic genome activation in vertebrates. Dev. Cell 42, 316–332 (2017).

    Article  Google Scholar 

  5. 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 

  6. De Iaco, A. et al. DUX-family transcription factors regulate zygotic genome activation in placental mammals. Nat. Genet. 49, 941–945 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. 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 

  9. Huang, Y. et al. Stella modulates transcriptional and endogenous retrovirus programs during maternal-to-zygotic transition. eLife 6, e22345 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sakashita, A. et al. Transcription of MERVL retrotransposons is required for preimplantation embryo development. Nat. Genet. 55, 484–495 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Guo, M. et al. Precise temporal regulation of Dux is important for embryo development. Cell Res. 29, 956–959 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zuo, F. et al. A TRIM66/DAX1/Dux axis suppresses the totipotent 2-cell-like state in murine embryonic stem cells. Cell Stem Cell 29, 948–961 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Yan, Y. L. et al. DPPA2/4 and SUMO E3 ligase PIAS4 opposingly regulate zygotic transcriptional program. PLoS Biol. 17, e3000324 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Xie, S. Q. et al. Nucleolar-based Dux repression is essential for embryonic two-cell stage exit. Genes Dev. 36, 331–347 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ruebel, M. L., Vincent, K. A., Schall, P. Z., Wang, K. & Latham, K. E. SMCHD1 terminates the first embryonic genome activation event in mouse two-cell embryos and contributes to a transcriptionally repressive state. Am. J. Physiol. Cell Physiol. 317, C655–C664 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Yang, F. et al. DUX-miR-344-ZMYM2-mediated activation of MERVL LTRs induces a totipotent 2C-like state. Cell Stem Cell 26, 234–250 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Garland, W. et al. Chromatin modifier HUSH co-operates with RNA decay factor NEXT to restrict transposable element expression. Mol. Cell 82, 1691–1707 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Clapp, J. et al. Evolutionary conservation of a coding function for D4Z4, the tandem DNA repeat mutated in facioscapulohumeral muscular dystrophy. Am. J. Hum. Genet. 81, 264–279 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Olbrich, T. et al. CTCF is a barrier for 2C-like reprogramming. Nat. Commun. 12, 4856 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Leidenroth, A. & Hewitt, J. E. A family history of DUX4: phylogenetic analysis of DUXA, B, C and Duxbl reveals the ancestral DUX gene. BMC Evol. Biol. 10, 364 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu, S. L. et al. Characterization of genomic structures and expression profiles of three tandem repeats of a mouse double homeobox gene: Duxbl. Dev. Dyn. 239, 927–940 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Klein, F. et al. The transcription factor Duxbl mediates elimination of pre-T cells that fail β-selection. J. Exp. Med. 216, 638–655 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Preussner, J. et al. Oncogenic amplification of zygotic Dux factors in regenerating p53-deficient muscle stem cells defines a molecular cancer subtype. Cell Stem Cell 23, 794–805 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Deng, Q., Ramsköld, D., Reinius, B. & Sandberg, R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343, 193–196 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  28. Iturbide, A. et al. Retinoic acid signaling is critical during the totipotency window in early mammalian development. Nat. Struct. Mol. Biol. 28, 521–532 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shen, H. et al. Mouse totipotent stem cells captured and maintained through spliceosomal repression. Cell 184, 2843–2859 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Raccaud, M. et al. Mitotic chromosome binding predicts transcription factor properties in interphase. Nat. Commun. 10, 487 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xie, L. et al. A dynamic interplay of enhancer elements regulates Klf4 expression in naïve pluripotency. Genes Dev. 31, 1795–1808 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Herquel, B., Ouararhni, K. & Davidson, I. The TIF1α-related TRIM cofactors couple chromatin modifications to transcriptional regulation, signaling and tumor suppression. Transcription 2, 231–236 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Herquel, B. et al. Transcription cofactors TRIM24, TRIM28, and TRIM33 associate to form regulatory complexes that suppress murine hepatocellular carcinoma. Proc. Natl Acad. Sci. USA 108, 8212–8217 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14, 431–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Maksakova, I. A. et al. Distinct roles of KAP1, HP1 and G9a/GLP in silencing of the two-cell-specific retrotransposon MERVL in mouse ES cells. Epigenetics Chromatin 6, 15 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rowe, H. M. et al. TRIM28 repression of retrotransposon-based enhancers is necessary to preserve transcriptional dynamics in embryonic stem cells. Genome Res. 23, 452–461 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dan, J., Yang, J., Liu, Y., Xiao, A. & Liu, L. Roles for histone acetylation in regulation of telomere elongation and two-cell state in mouse ES cells. J. Cell. Physiol. 230, 2337–2344 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang, C. et al. Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development. Nat. Cell Biol. 20, 620–631 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Modzelewski, A. J. et al. A mouse-specific retrotransposon drives a conserved Cdk2ap1 isoform essential for development. Cell 184, 5541–5558 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Xu, R. et al. Stage-specific H3K9me3 occupancy ensures retrotransposon silencing in human pre-implantation embryos. Cell Stem Cell 29, 1051–1066 (2022).

    Article  CAS  PubMed  Google Scholar 

  43. Asimi, V. et al. Hijacking of transcriptional condensates by endogenous retroviruses. Nat. Genet. 54, 1238–1247 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wu, K. et al. SETDB1-mediated cell fate transition between 2C-like and pluripotent states. Cell Rep. 30, 25–36 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. De Iaco, A., Verp, S., Offner, S., Grun, D. & Trono, D. DUX is a non-essential synchronizer of zygotic genome activation. Development 147, dev.177725 (2019).

    Article  Google Scholar 

  46. Chen, Z. & Zhang, Y. Loss of DUX causes minor defects in zygotic genome activation and is compatible with mouse development. Nat. Genet. 51, 947–951 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Guo, Y. et al. Obox4 secures zygotic genome activation upon loss of Dux. Preprint at bioRxiv https://doi.org/10.1101/2022.07.04.498763 (2022).

  48. Ji, S. et al. OBOX regulates mouse zygotic genome activation and early development. Nature 620, 1047–1053 (2023).

    Article  CAS  PubMed  Google Scholar 

  49. Bosnakovski, D. et al. Antagonism among DUX family members evolved from an ancestral toxic single homeodomain protein. iScience 26, 107823 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gambini, A. et al. Developmentally programmed tankyrase activity upregulates β-catenin and licenses progression of embryonic genome activation. Dev. Cell 53, 545–560 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, 160–165 (2016).

    Article  Google Scholar 

  53. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, (2011).

  55. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Jin, Y., Tam, O. H., Paniagua, E. & Hammell, M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics 31, 3593–3599 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general-purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Xie, C. et al. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 39, W316–W322 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tillo, D. Code for Cut&Run analysis. Zenodo https://doi.org/10.5281/zenodo.10658411 (2024).

  62. Tillo, D. Code for Gene/TE quantification. Zenodo https://doi.org/10.5281/zenodo.10658421 (2024).

Download references

Acknowledgements

We thank principal investigators from the Laboratory of Genome Integrity for helpful comments and discussion on this work. We thank K. Tachibana and W. Kobayashi for their suggestions and critical reading of the manuscript. We also thank M. Malik, D. Goldstein and the CCR Genomics Core for sequencing support; F. Livak, S. Siddiqui and the CCR Flow Cytometry Core, G. Pegoraro and the High-Throughput Imaging Facility and M. Kruhlak and the Microscopy Core for experimental support at the NCI; the Epigenomics and DNA Sequencing, Fluorescence Microscopy and Imaging Core facilities at National Institute of Environmental Health Sciences (NIEHS) and to the core facilities in Max Planck Institute for Heart and Lung Research for support with mouse transgenics (S. Krüger, S. Kreutzer and D. Heil). Research in S.R. and C.J.W. laboratory is supported by the Intramural Research Program of the National Institutes of Health (NIH). T.O. is supported by a postdoctoral fellowship of the Helen Hay Whitney Foundation. Research in J.K. Laboratory was supported by the Max Planck Society and Deutsche Forschungsgemeinschaft (DFG, German Research Foundation): DFG project 496710327 and Excellence Cluster CPI (EXC2026). D.B. is supported by the R01 grant 1R01AR081228-01.

Author information

Authors and Affiliations

Authors

Contributions

M.V.-S., F.F.L., T.O., J.K. and S.R. conceived the study. T.O., M.V.-S. and F.F.L. designed, performed and analyzed most of in vitro experiments. Y.C., J.Z., T.L. and L.S. designed and performed experiments. K.-P.K. and H.S. provided vectors. F.F.L., P.S., V.S. and L.R. performed microinjection and in vivo experiments. C.N.D. provided technical support. S.G. and D.T. analyzed sequencing data. C.K. and M.B. analyzed data and performed bioinformatic analyses under the supervision of M.L. G.I.C. provided support with high-throughput microscopy imaging. B.S. analyzed live confocal microscopy data. T.K.M. and L.M.J. performed and analyzed MS experiments. D.B. and M.K. provided critical input. S.R., J.K. and C.J.W. supervised the study and wrote the manuscript with comments and help from all authors.

Corresponding authors

Correspondence to Johnny Kim or Sergio Ruiz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Haruhiko Siomi, Jianlong Wang, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 DUX induces two protein products from the Duxbl gene.

a) High-throughput imaging quantification of GFP mean nuclear intensity in untreated BR1-GFP reporter ESCDUX or treated with DOX for 16 h. Centerlines indicate mean values. n = 537. b) Graph plots showing quantified mean intensity levels of RFP and GFP obtained from untreated and DOX-treated LTR-RFP reporter BR2- or BR3-GFP ESCDUX. c) DNA sequences surrounding exon 2 of Duxbl showing the deletions detected in representative DUXBLKO ESCDUX clones analyzed. DNA sequence from exon 2 is highlighted in bold. The alternative ATG at the end of the second Duxbl exon is highlighted in red. d) Schematic representation of the DUXBL protein products. e) Protein sequence alignment of DUX, DUXBLLG (large DUXBL isoform) and DUXBLSM (small DUXBL isoform). Highlighted in blue are the two homeodomains (note that DUXBLSM lacks most of the first homeodomains). Highlighted in dark gray are conserved residues and in light gray are similar residues. Highlighted in green is the peptide sequence used to generate our custom DUXBL antibody. f) Western blot analysis of DUXBL performed in lysates from a pool of approximately 488 embryos at the indicated stages of development. Increasing amounts of DOX-treated WT ESCDUX, as well as DOX-treated DUXBLKO ESCDUX lysates, were included as controls. Ponceau staining is shown as a loading control. For a, b and f, two independent experiments were performed but one representative experiment is shown.

Source data

Extended Data Fig. 2 DUXBL does not function as a transcriptional activator.

a) Volcano plots showing differentially expressed genes between DUX- (left panel) or DUXBL-expressing ESC (right panel) compared to control ESC. b) Heatmap showing the top 50 differentially expressed genes between DUX- and DUXBL-expressing ESC. c) Western blot analysis of the indicated proteins performed with V5 immunoprecipitates obtained from 293T cells transfected with the corresponding expression vectors. This experiment was repeated multiple times, but one is shown. Data in a and b were generated using three replicates per condition.

Source data

Extended Data Fig. 3 Duxbl-deficient ESC showed increased 2CLC conversion.

a) Time-lapse microscopy experiment performed in LTR-RFP reporter WT and DUXBLKO ESCDUX. Two independent experiments with two ESC lines per condition were performed, but one representative is shown. Time since the addition of DOX is indicated. Scale bar, 10 μm. b) High-throughput imaging (HTI) quantification of RFP+ cells in LTR-RFP reporter WT and DUXBLKO ESC. Centerlines indicate mean values. Percentages of RFP+ cells above the threshold (dotted line) are indicated. n = 2000; p value is from one-tailed unpaired t-test. Three independent experiments were performed but one representative is shown. c) Real-time PCR analysis shown in a box and whisker plot showing the relative fold change expression of nine 2C-associated genes/repeats (Dux, Zscan4c, Zfp352, Tcstv3, Sp110, Tdpoz1, Dub1, Eif1ad8 and MERVLs) in RFP negative, and RFP positive cells sorted from LTR-RFP reporter WT and DUXBLKO ESCDUX cultures. Gapdh expression was used to normalize. Centerline indicates the median, box extends from the 25th to 75th percentiles and whiskers show min to max values. P values are shown from one-tailed unpaired t-tests. Two independent experiments were performed, but one representative is shown. d) Unidimensional PCA plot of RNA-seq data from untreated or DOX-treated WT and DUXBLKO ESCDUX (two replicates each) together with WT DUX-expressing ESC from ref. 5. DUX-expressing ESCs were sorted into GFP+ or GFP based on the activation of the LTR-GFP reporter. e) Box and whisker plot showing normalized fold change expression of the 244 genes downregulated as shown in Fig. 2b. RNA-seq data obtained from27. Centerline indicates the median, box extends from the 25th to 75th percentiles and whisker extends from the hinge to the largest or smallest value no further than 1.5-fold from the interquartile range. Data beyond whiskers were plotted individually. For ce, data were generated using two independent ESC lines per genotype and condition. f) Venn diagram showing the overlap between the 210 genes described in Fig. 2d and the upregulated genes in LTR+ sorted DUX-expressing ESC (data obtained from ref. 5). P value was obtained from a two-tailed Fisher’s exact test. g) Genome browser tracks from individual samples showing RNA-seq RPKM read count in untreated or DOX-treated WT and DUXBLKO ESCDUX.

Source data

Extended Data Fig. 4 DUXBL expression limits 2CLC conversion.

a) Western blot analysis of DUXBL (FLAG) performed in lysates from untreated or indole-3-acetic acid (IAA)/DOX-treated WT or ESCCTCF-AID expressing DUXBL (FLAG). Tubulin levels are shown as a loading control. b) High-throughput imaging (HTI) quantification of RFP+ cells in LTR-RFP reporter WT and DUXBLKO ESC incubated with 0.53 mM RA for 48 hours. c) HTI quantification of RFP+ cells in untreated or DOX-treated LTR-RFP reporter ESCDUXBL incubated with 0.53 mM RA for 48 hours. d) Western blot analysis using lysates from untreated or DOX-treated ESCDUXBL incubated with 2.5 mM PlaB for 24 hours or 0.53 mM RA for 48 hours. e) Box and whisker plot showing the relative fold change (log2) expression of eight 2C-associated genes (Dux, Zscan4c, Zfp352, Tcstv3, Sp110, Tdpoz1, Dub1 and Eif1ad8) in untreated or DOX-treated ESCDUXBL incubated with 2.5 mM PlaB for 24 hours or 0.53 mM RA for 48 hours. Two independent experiments were performed, but one representative is shown. P values are shown from one-tailed unpaired t-tests. Centerline indicates the median, box extends from the 25th to 75th percentiles and whiskers show min to max values. f) Western blot analysis of CTCF performed in lysates from untreated or IAA-treated ESCCTCF-AID. Tubulin levels are shown as a loading control. g) HTI quantification of RFP+ cells in untreated LTR-RFP reporter ESCCTCF-AID expressing DUXBL or IAA/DOX-treated for 48 hours. h) HTI quantification of RFP+ cells in untreated LTR-RFP reporter WT or DUXBLKO ESCCTCF-AID or IAA-treated for 24 and 48 hours. For a,d and f, two independent experiments were performed, but one representative is shown. For b,c,g and h, centerlines indicate mean values. Percentages of RFP+ cells above the threshold (dotted line) are indicated. n = 2000. P values are shown from one-tailed unpaired t-tests. In all cases, three independent experiments were performed, but only one representative is shown.

Source data

Extended Data Fig. 5 DUXBL expression in zygotes impairs proper ZGA.

a) Representative bright-field and fluorescence images of zygotes not injected or microinjected with mRNA encoding for mCherryNLS or DUXBLV5 plus mCherryNLS. Images were taken 6 hours post microinjection. Scale bar, 50 μm. Five independent experiments were performed, but one representative is shown. b) Representative immunofluorescence analysis of the V5 tag in zygotes not injected or microinjected with mRNA encoding DUXBLV5 plus mCherryNLS. DAPI was used to visualize the nuclei. Scale bars, 50 μm. At least 10 independent embryos per condition were stained, but one representative is shown. c) Heatmap and k-means clustering (3) showing the distribution of the indicated samples described in Fig. 3a. d,e) Volcano plots highlighting the differentially expressed genes observed by RNA-seq analysis between DUXBLV5 and mCherryNLS-overexpressing embryos (red: FC ≥2 DUXBL/mCherry; blue: FC ≤ −2 DUXBLV5/mCherry; padj < 0.05). f) Volcano plot showing differentially expressed genes between DUXBL-overexpressing embryos compared to mCherry-overexpressing embryos. Genes differentially induced by DUX are highlighted in red. ≤−1 log2 fold change ≥1; padj value < 0.05. For cf, data were generated by using a total of 4–6 pools of 5 embryos/pool per stage and condition.

Extended Data Fig. 6 Downregulation of Duxbl in zygotes compromises development.

a) Plot showing mean ± standard error of the mean (SEM) summarizing four independent experiments with a total of 25–45 microinjected zygotes per group (non-microinjected, control morpholino (MO), or a second morpholino (MO2)-injected zygotes) per experiment. The percentage of embryos reaching each embryo stage is shown. Mor: morula, Blast: blastocyst. P values are shown from one-tailed unpaired t-tests. b) Plot showing mean ± standard error of the mean (SEM) summarizing four independent experiments with a total of 25–45 microinjected zygotes per group (non-microinjected, control or Duxbl siRNAs injected GV oocytes) per experiment. The percentage of embryos reaching each embryo stage is shown. Mor: morula, Blast: blastocyst. P values are shown from one-tailed unpaired t-tests. c) Relative fold change (log2) expression of Duxbl in uninduced or DOX-induced in ESCDUX transfected 36 hours prior to induction with the corresponding siRNAs. Reactions were performed by duplicate in two different ESCDUX lines. Two independent experiments were performed, but one representative is shown. P value is shown from one-tailed unpaired t-test. d) Pie chart showing percentages of genotypes from Duxbl+/Δ crossings at P14 after F1 generation. e) Gel showing PCR results from genotyping all 8C-stage embryos obtained from Duxbl+/Δ crossings. The identity of the bands obtained by PCR was confirmed by genotyping on positive and negative controls at least two times. f) Gene set enrichment analysis (PANTHER gene sets) according to differentially expressed genes observed in Duxbl Δ/Δ embryos compared to Duxbl+/Δ/Duxbl+/+ embryos (Kobas FDR < 0.2).

Source data

Extended Data Fig. 7 DUXBL gains access at DUX-bound sites following DUX expression.

a) Predicted binding sites for DUX and DUXBL (Jaspar database). b) Venn diagrams showing the number of DUXBL peaks overlapping between those identified with DUXBL antibodies (blue) and FLAG antibodies (yellow) in ESCDUXBL. c) Genome browser tracks corresponding to individual samples from b showing DUXBL occupancy. Input (IgG) is shown as control. For b,c, two independent experiments with two independent ESC lines were performed. d) Pie chart showing the genomic distance of DUXBL peaks from gene TSS. e) Venn diagrams showing the number of DUXBL peaks overlapping with traditional ESC enhancers (left) and ESC SEs (right). Enhancer data obtained from31,32. P values shown are from two-tailed Fisher’s exact test. f) Gene ontology (GO) terms obtained from analyzing DUXBL peaks. P values were obtained from a binomial test. g) CUT&RUN read density plot (RPGC) showing DUXBL and DUX enrichment at MERVL-int elements in DOX-treated ESCDUXBL. Data from DUX-expressing ESC were obtained from5. Input (IgG) is shown as control. Data from one representative ESC line are shown. h) Venn diagrams showing the number of DUXBL (left panel, blue), and DUX peaks (right panel, blue) overlapping with accessible regions in ESC (yellow). ATAC-seq data were obtained from33. i) Genome browser tracks from individual samples showing DUXBL and DUX occupancy as well as ATAC-seq signal in DOX-treated WT ESC expressing DUX or DUXBL. Data from DUX-expressing ESC and ATAC-seq experiments were obtained from5,33. Input (IgG) is shown as control. j) Plot showing DUXBL enrichment over DUX-bound sites in the condition of DUX/DUXBL co-expression compared to only DUXBL expression. We used a log2(FC) cutoff of >1 and p-value < 0.01 to define differentially enriched sites. Representative genes in proximity to DUX-bound sites enriched for DUXBL binding are shown. Two independent experiments using one ESC line were performed, but data from one representative experiment are shown. k) Immunofluorescence analysis of DUXBL (FLAG) in untreated or DOX-treated LTR-RFP reporter ESCDUXBL/DUX. Scale bar, 20 μm. l) Immunofluorescence analysis of DUXBL in untreated or DOX-treated ESCDUXBL. In k and l, two independent experiments were performed, but one representative is shown. DAPI was used to visualize nuclei. Scale bar, 100 μm.

Extended Data Fig. 8 DUXBL interacts with the TRIM33/TRIM24 complex.

a, b) Western blot analysis of the indicated proteins performed with total protein extracts (input) and DUXBL immunoprecipitates (10% of total IP) obtained from untreated or DOX-treated ESCDUXBL (a) or untreated or DOX-treated ESCDUX (b). c) Volcano plot showing the enrichment of proteins obtained from endogenous DUXBL immunoprecipitation followed by mass spectrometry (IP-MS) analysis in two independent untreated or DOX-treated ESCDUX. d) Western blot analyses of the indicated proteins performed with total extracts (input) and DUXBL immunoprecipitates (IP) obtained from untreated and doxycycline treated ESCDUX. We also included the flowthrough (FT) following the immunoprecipitation to show the specific depletion of DUXBL. e) Schematic representation of the different TRIM24 mutants used in our immunoprecipitation assays. f, g) Western blot analysis of the indicated proteins performed with total protein extracts (input) and FLAG pulldowns obtained from DOX-treated ESC containing stably transfected Piggy-bac constructs encoding for HA-tagged DUXBL and/or each of the different FLAG-tagged TRIM24 mutants. h) Schematic representation of the different DUXBL protein forms used in our immunoprecipitation assays. i) Western blot analysis of the indicated proteins performed with total protein extracts (input) and FLAG pulldowns obtained from WT and DOX-treated DUXBLKO ESC containing stably transfected Piggy-bac constructs encoding for each of the different FLAG-tagged DUXBL protein forms. TRIM24 endogenous levels are shown. Two independent experiments were performed in a,b,d,f,g and i, but one representative is shown.

Source data

Extended Data Fig. 9 DUXBL/TRIM33/TRIM24 colocalize at DUX-induced nuclear foci.

a) Immunofluorescence analysis of HA (DUX) and endogenous TRIM24 in untreated or DOX-treated ESCDUX-2XHA. Scale bars, 20 μm. b) Immunofluorescence analysis of endogenous TRIM24 in endogenous 2CLC observed in LTR-RFP reporter WT ESC. Scale bars, 20 μm. For a,b, DAPI was used to visualize nuclei and two independent experiments were performed but one representative is shown. c) High-throughput imaging (HTI) quantification of the number, total intensity and area of TRIM24 foci (upper, middle and lower panels, respectively) per cell in untreated or DOX-treated LTR-RFP reporter WT or DUXBLKO ESCDUX. d) Western blot analysis of TRIM24 performed in WT and TRIM24KO ESCDUX. e) Western blot analysis of TRIM33 performed in WT and TRIM33KO ESCDUX lysates. f) HTI quantification of the number, total intensity and area of DUXBL foci (upper, middle and lower panels, respectively) per cell in untreated or DOX-treated LTR-RFP reporter WT or TRIM33KO ESCDUX. For c and f, percentages of cells above the threshold (dotted line) are indicated. Centerlines indicate mean values. n = 2000; p values are shown from one-tailed unpaired t-tests. Two independent experiments using at least two WT, DUXBLKO ESCDUX or TRIM33KO ESCDUX clones were performed but one representative is shown. g) Graph showing 2CLC residency time evaluated by using the LTR-RFP reporter in DOX-treated WT and TRIM24KO ESCDUX. P value is shown from a two-tailed unpaired t-test. Plot was generated by using data from two independent ESC lines per genotype. h) Western blot analysis of TRIM24 performed in WT and TRIM24FKBP ESCDUX. i) Western blot analysis of TRIM24 and DUXBL performed in untreated and DOX/dTAG-treated for 16 hours TRIM24FKBP and DUXBLKO; TRIM24FKBP ESCDUX. For d,e,h,i, tubulin levels were used as loading control and two independent experiments were performed but one representative is shown. j) Graph showing 2CLC residency time evaluated by using the LTR-RFP reporter in dTAG-treated WT and DUXBLKO; TRIM24FKBP ESCDUX. P values are shown from two-tailed unpaired t-tests. Plot was generated by using data from two independent ESC lines per genotype.

Source data

Extended Data Fig. 10 DUXBL/TRIM24/TRIM33 complex co-localizes with H3K9me3 at DUX-induced nuclear foci.

a) Immunofluorescence analysis of DUXBL in untreated or DOX-treated ESCDUX-FKBP. Treatments include DOX for 24 hours or DOX for 16 hours plus 8 hours with dTAG compounds. DAPI was used to visualize nuclei. Scale bar, 20 μm. Three independent experiments were performed but one representative is shown. b) High-throughput imaging (HTI) quantification of the number, total intensity and area of DUXBL foci (upper, middle and lower panels, respectively) per cell in ESCDUX-FKBP treated as in (a). c) HTI quantification of the number, total intensity and area of HA (DUX) foci (upper, middle and lower panels, respectively) per cell in untreated or DOX-treated ESCDUX-FKBP treated as in (a). d) HTI quantification of the number, total intensity and total area of TRIM24 foci (upper, middle and lower panels, respectively) per cell in ESCDUX-FKBP treated as in (a). For b,c, and d, centerlines indicate mean values; n = 2000. P values are shown from one-tailed unpaired t-tests. Percentages of cells above the threshold (dotted line) are indicated. At least, two independent experiments were performed but one representative experiment is shown. e,f) Plot showing the ratio for HDAC (e) and HP1 (f) between the fluorescence intensity found at TRIM24 foci and the mean nuclear intensity per cell detected in DOX-treated LTR-RFP reporter ESCDUX. Centerlines indicate mean values. n = 2000. Two independent experiments using two ESCDUX clones were performed but one representative is shown. g) HTI quantification of the number, total intensity and area of H3K9me3 foci (left, middle and right panels, respectively) per cell in untreated or DOX-treated LTR-RFP reporter ESCDUX. Centerlines indicate mean values. n = 2000; p values are shown from one-tailed unpaired t-tests. Three independent experiments using at least two ESCDUX clones were performed, but one representative is shown.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–7, Supplementary Methods and supporting data for Supplementary Fig. 4c.

Reporting Summary

Peer Review File

Supplementary Video 1

Live-cell imaging of DOX-treated LTR-RFP and BR2-EGFP reporter ESCDUX.

Supplementary Video 2

Live-cell imaging of DOX-treated LTR-RFP and BR3-EGFP reporter ESCDUX.

Supplementary Video 3

Live-cell imaging of LTR-RFP reporter DOX-induced ESCDUX endogenously tagged with a miRFP702 fluorescent protein at the Duxbl locus.

Supplementary Code

Custom code used to process Cut&Run data and analyze the expression of TEs.

Supplementary Tables

Supplementary Tables 1–14.

Supplementary Data

Statistical supporting for Supplementary Figs. 1 and 4.

Source data

Source Data Fig. 1

Uncropped western blots and/or gels.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Uncropped western blots and/or gels.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Uncropped western blots and/or gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Uncropped western blots and/or gels.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Uncropped western blots and/or gels.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 8

Uncropped western blots and/or gels.

Source Data Extended Data Fig. 9

Uncropped western blots and/or gels.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vega-Sendino, M., Lüttmann, F.F., Olbrich, T. et al. The homeobox transcription factor DUXBL controls exit from totipotency. Nat Genet 56, 697–709 (2024). https://doi.org/10.1038/s41588-024-01692-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-024-01692-z

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