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.

DUX-family transcription factors regulate zygotic genome activation in placental mammals

Abstract

In animal embryos, transcription is mostly silent for several cell divisions, until the release of the first major wave of embryonic transcripts through so-called zygotic genome activation (ZGA)1. Maternally provided ZGA-triggering factors have been identified in Drosophila melanogaster and Danio rerio2,3, but their mammalian homologs are still undefined. Here, we provide evidence that the DUX family of transcription factors4,5 is essential to this process in mice and potentially in humans. First, human DUX4 and mouse Dux are both expressed before ZGA in their respective species. Second, both orthologous proteins bind the promoters of ZGA-associated genes and activate their transcription. Third, Dux knockout in mouse embryonic stem cells (mESCs) prevents the cells from cycling through a 2-cell-like state. Finally, zygotic depletion of Dux leads to impaired early embryonic development and defective ZGA. We conclude that DUX-family proteins are key inducers of zygotic genome activation in placental mammals.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: DUX4 promotes transcription of genes expressed during early ZGA.
Figure 2: DUX4 binds TSSs of genes expressed during early ZGA and activates their expression in hESCs.
Figure 3: Dux is necessary for formation of 2C-like mESCs.
Figure 4: TRIM28 regulates formation of 2C-like mESCs by repressing Dux expression.
Figure 5: Dux is necessary for mouse early embryonic development.

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

ArrayExpress

Gene Expression Omnibus

References

  1. Lee, M.T., Bonneau, A.R. & Giraldez, A.J. Zygotic genome activation during the maternal-to-zygotic transition. Annu. Rev. Cell Dev. Biol. 30, 581–613 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lee, M.T. et al. Nanog, Pou5f1 and SoxB1 activate zygotic gene expression during the maternal-to-zygotic transition. Nature 503, 360–364 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Leidenroth, A. et al. Evolution of DUX gene macrosatellites in placental mammals. Chromosoma 121, 489–497 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. 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 

  6. Hewitt, J.E. et al. Analysis of the tandem repeat locus D4Z4 associated with facioscapulohumeral muscular dystrophy. Hum. Mol. Genet. 3, 1287–1295 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Wijmenga, C. et al. Chromosome 4q DNA rearrangements associated with facioscapulohumeral muscular dystrophy. Nat. Genet. 2, 26–30 (1992).

    Article  CAS  PubMed  Google Scholar 

  8. Gabriëls, J. et al. Nucleotide sequence of the partially deleted D4Z4 locus in a patient with FSHD identifies a putative gene within each 3.3 kb element. Gene 236, 25–32 (1999).

    Article  PubMed  Google Scholar 

  9. 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 

  10. 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 

  11. 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  Google Scholar 

  12. Vassena, R. et al. Waves of early transcriptional activation and pluripotency program initiation during human preimplantation development. Development 138, 3699–3709 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Töhönen, V. et al. Novel PRD-like homeodomain transcription factors and retrotransposon elements in early human development. Nat. Commun. 6, 8207 (2015).

    Article  PubMed  Google Scholar 

  14. 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 

  15. 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 

  16. 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 

  17. 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 

  18. Eckersley-Maslin, M.A. et al. MERVL/Zscan4 network activation results in transient genome-wide DNA demethylation of mESCs. Cell Rep. 17, 179–192 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sun, Y. et al. Zelda overcomes the high intrinsic nucleosome barrier at enhancers during Drosophila zygotic genome activation. Genome Res. 25, 1703–1714 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. 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 

  22. Perrod, S. & Gasser, S.M. Long-range silencing and position effects at telomeres and centromeres: parallels and differences. Cell. Mol. Life Sci. 60, 2303–2318 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. van der Maarel, S.M., Tawil, R. & Tapscott, S.J. Facioscapulohumeral muscular dystrophy and DUX4: breaking the silence. Trends Mol. Med. 17, 252–258 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. 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 

  26. Lu, F., Liu, Y., Jiang, L., Yamaguchi, S. & Zhang, Y. Role of Tet proteins in enhancer activity and telomere elongation. Genes Dev. 28, 2103–2119 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schoorlemmer, J., Pérez-Palacios, R., Climent, M., Guallar, D. & Muniesa, P. Regulation of mouse retroelement MuERV-L/MERVL expression by REX1 and epigenetic control of stem cell potency. Front. Oncol. 4, 14 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Walter, M., Teissandier, A., Pérez-Palacios, R. & Bourc'his, D. An epigenetic switch ensures transposon repression upon dynamic loss of DNA methylation in embryonic stem cells. eLife 5, e11418 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Macfarlan, T.S. et al. Endogenous retroviruses and neighboring genes are coordinately repressed by LSD1/KDM1A. Genes Dev. 25, 594–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ecco, G. et al. Transposable elements and their KRAB-ZFP controllers regulate gene expression in adult tissues. Dev. Cell 36, 611–623 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. De Iaco, A. et al. TNPO3 protects HIV-1 replication from CPSF6-mediated capsid stabilization in the host cell cytoplasm. Retrovirology 10, 20 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. De Iaco, A. & Luban, J. Cyclophilin A promotes HIV-1 reverse transcription but its effect on transduction correlates best with its effect on nuclear entry of viral cDNA. Retrovirology 11, 11 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ran, F.A . et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Edgar, R., Domrachev, M. & Lash, A.E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kolesnikov, N. et al. ArrayExpress update: simplifying data submissions. Nucleic Acids Res. 43, D1113–D1116 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  37. Law, C.W., Chen, Y., Shi, W. & Smyth, G.K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Gentleman, R.C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zang, C. et al. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25, 1952–1958 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  43. Thomas-Chollier, M. et al. RSAT: regulatory sequence analysis tools. Nucleic Acids Res. 36, W119–W127 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Macfarlan (NIH) and M.E. Torres-Padilla (Helmholtz Zentrum Munchen) for sharing reagents and for helpful discussions, and we thank the Transgenic and Gene Expression Core Facility (EPFL) and S. Offner for technical assistance. This work was financed through grants from the Swiss National Science Foundation, the Gebert-Rüf Foundation, an INGENIUM grant (FP7 MC-ITN INGENIUM 290123), and the European Research Council (ERC 268721 and ERC 694658) to D.T.

Author information

Authors and Affiliations

Authors

Contributions

A.D.I. and D.T. conceived the project, designed the experiments, analyzed the data, and wrote the manuscript; A.D.I., A.C., and S.V. carried out the experiments; E.P. and J.D. performed bioinformatic and statistical analyses.

Corresponding author

Correspondence to Didier Trono.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Identification of genes specifically expressed at each stage of early embryo development.

(A) Pattern of genes with similar expression behavior during human embryo development. The blue line represents the average expression of the genes in each subset, while the gray lines represent each single gene. (B) Fraction of genes belonging to each pattern in (A) whose expression is not changing (gray), upregulated (cyan) or downregulated (orange) when DUX4 is ectopically expressed in human primary myoblast. (C) Fraction of genes belonging to each cluster illustrated in Figure 1B, the expression of which is unchanged (gray), increased (cyan) or decreased (orange) when DUX4 is ectopically expressed in human primary myoblasts. In B and C a fold change bigger than 2 and a p-value lower than 0.05 after Benjamini-Hochberg correction were selected.

Supplementary Figure 2 DUX4 ectopically expressed in hESCs and human primary myoblasts binds similar genomic sequences.

DUX4 was ectopically expressed in hESCs and ChIP-seq for the transcription factor was performed. (A) The Venn diagram represents the ChIP-seq peak overlap between our analysis in hESCs and the publicly available one in human primary myoblasts. (B) Percentage of individual integrants from families of transposable elements bound by DUX4. (C) Comparative expression during early human embryonic development of DUX4 (red), its putative target genes (blue, average), and the transposable elements (TEs) bound by DUX4 in hESCs and displaying increased expression at the peak of DUX4 transcription (green, average of all the TE integrants from each family). Oo, oocyte; Zy, zygote; 2C, 4C, 8C, corresponding n-cell stage; Mo, morula; Bl, blastocyst. (D) DUX4 binding motif identified with Homer. (E) Comparison of the fraction of all DUX4 peaks and the ones selected for being in a 5 kb window around the TSS of ZGA genes (from figure 2A) containing the DUX4 binding motif identified. (F) Comparison of the frequency of DUX4 binding motif identified in all DUX4 peaks or in the ones selected for being in a 5 kb window around the TSS of ZGA genes (from figure 2A).

Supplementary Figure 3 DUX4 binds TSSs of genes expressed during early ZGA when it is overexpressed in human primary myoblasts.

(A) Average coverage of ChIP-seq signal of DUX4 (blue) when overexpressed in human primary myoblasts in a window of 5 kb from the annotated TSS of genes belonging to the 2-4C and 2-8C clusters in figure 1B. Total input is represented in gray (line, average; shade, standard error of the mean). (B) Fraction of genes belonging to each cluster in figure 1B that have a DUX4 peak within a 5 kb window from their annotated TSS. Fisher’s exact test was performed to compare maternal vs 2-4C or 2-8C (p= 1.27e-47 and p= 1.49e-09 respectively). (C) Average coverage of ChIP-seq signal of DUX4 (blue) when overexpressed in human primary myoblasts in a window of 5 kb from TFE of RNAs found specifically upregulated at oocyte-to-4C and 4C-to-8C transitions. Total input is represented in gray (line, average; shade, standard error of the mean). (D) Fraction of TFE from oocyte-to-4C and 4C-to-8C transitions that have a DUX4 peak overlapping with their 5'. Fisher’s exact test was performed to compare 4C-to-8C vs. oocyte-to-4C TFEs (p= 4.48e-17).

Supplementary Figure 4 DUX4 binds TSSs of genes expressed during early ZGA.

UCSC genome browser screenshots on genes belonging to the 2-4C (TRIM43 and TRIM49) and the 2-8C (ZSCAN4 and MBD3L2) subset of genes. Starting from the top are displayed tracks for: RNA expression in early embryos (2C, 4C and 8C); DUX4 interaction with the locus in hESCs and human primary myoblasts; RefSeq track for gene annotation.

Supplementary Figure 5 Genomic and protein sequence organization of mDux.

(A) UCSC genome browser screenshot representing the genomic location of the D4Z4 repeats where the mouse orthologues of DUX4 are located. Tracks for gene annotation from two different Ensembl releases (84 and 75) and RefSeq are displayed. (B) Alignment of DUX4, Dux and Gm4981 protein sequences. Residues are colored according to their physicochemical properties (Clustal Omega code). The black boxes define conserved putative homeodomains and C-terminus.

Supplementary Figure 6 Genes highly expressed at the middle 2C stage are significantly represented in 2C-like mESCs.

(A) Pattern of genes with similar expression behavior during murine embryo development. The blue line represents the average expression of the genes in each subset, while the gray lines represent each single gene. (B) Fraction of genes belonging to mid 2C pattern (A) or rest of the genes, the expression of which is unchanged (gray), increased (cyan) or decreased (orange) in 2C-like cells compared to the rest of mESCs (selected from figure 3B). (C) Comparative expression of genes (Zscan4, Cml2, Dux, Zfp352) and transposable elements (MERVL) activated at ZGA, and two control housekeeping gene (Actb and Gapdh) in mESCs carrying an integrated MERVL-GFP reporter and sorted for GFP expression. Expression was normalized to Actb.

Supplementary Figure 7 CRISPR–Cas9 deletion of the Dux macrosatellite repeat in mESCs.

(A) UCSC genome browser screenshot representing the locus depleted in mESCs carrying a MERVL-GFP reporter, using a combination of two different sgRNAs cutting upstream and downstream of the murine D4Z4 repeat (2 vertical black lines). Starting from the top are displayed tracks for: gene annotation from RefSeq, mobile elements from repeat masker, and mappability of the locus by alignability of 100mers. Lower arrows show the primers used for the genotyping of the WT and Dux KO alleles. Dashed lines represent the PCR product amplified by the primers. (B) PCR products using the WT (upper blot) or KO (lower blot) primer combinations on three different WT (1-3) or Dux KO (4-6) clones, the bulk mESC population before (7) and after (8) Cas9/sgRNA transfection, and water (9). (C) Fraction of genes overexpressed in 2C-like cells compared to the remaining population of mESCs (from figure 3B), the expression of which is unchanged (gray), decreased (orange) or increased (cyan) upon Dux KO in mESCs (fold change bigger than 2 and p-value lower than 0.05 after Benjamini-Hochberg correction).

Supplementary Figure 8 Ectopic expression of Dux rescues the formation of 2C-like cells.

(A) Fraction of genes overexpressed in 2C-like cells compared to the remaining population of mESCs (from figure 3B), the expression of which is unchanged (gray), increased (orange) or decreased (cyan) upon Dux overexpression in Dux KO mESCs (fold change bigger than 2 and p-value lower than 0.05 after Benjamini-Hochberg correction). Comparative expression of indicated RNAs in (B) WT or (C) Dux KO MERVL-GFP mESCs transfected with plasmids encoding for LacZ, DUX4, Dux or Gm4981. RNA expression data is normalized to Actb. Shown are the mean ± SD for three independent clones. *** p ≤ 0.001, t-test. Immunofluorescence staining against (D) NANOG or (E) SOX2 in Dux KO mESC clones ectopically expressing Dux.

Supplementary Figure 9 RNA sequencing analyses show similar patterns of gene expression when Dux is expressed in different contexts.

(A) Principal component analysis and (B) heatmap showing the clustering of all the RNA-sequencing analysis of WT and Dux KO mESCs used in this manuscript. Pearson distances and complete agglomeration method were used to compute the heatmap’s dendrogram.

Supplementary Figure 10 DUX4 and Dux have partial interspecies activity on ZGA genes.

(A) GFP expression of 293T carrying several copies of integrated MT2/gag sequence regulating a GFP reporter and transfected with plasmids encoding for LacZ, DUX4, Dux or Gm4981. (B) Comparative expression in 293T of four genes activated at ZGA (ZSCAN4, ZSCAN5B, MBD3L2 and DUXA) and two control housekeeping gene (ACTB and TBP) 24 hours after transfection with plasmids expressing LacZ, DUX4, Dux or Gm4981. Expression was normalized to ACTB. (C) Graphical representation of the MERVL regulatory sequence and truncated mutants used in luciferase reporter assays, with canonical (in gag) and altered (♦; in the MT2_mm LTR) DUX4 binding sequences indicated. Luciferase activity was quantified 24 hours after transfection of 293T (D) and mESCs (E) with pGL4 plasmids containing a promoterless reporter driven by MT2/gag, the truncated mutants or a human PGK promoter together with plasmids expressing LacZ, DUX4, Dux or Gm4981. Ratios between the pGL4 plasmids containing sequences upstream of the luciferase reporter and the empty pGL4 plasmid are represented. Shown are the mean ± standard deviation for three experiments performed on different days. ** p ≤ 0.01, *** p ≤ 0.001, unpaired t-test.

Supplementary Figure 11 TRIM28 regulates formation of 2C-like mESCs by repressing Dux expression.

(A) Fraction of genes overexpressed in 2C-like cells compared to the remaining population of mESCs (from figure 3B), the expression of which is unchanged (gray), increased (orange) or decreased (cyan) upon Trim28 KD in WT (first two bars) or Dux KO (last two bars) in mESCs (fold change bigger than 2 and p-value lower than 0.05 after Benjamini-Hochberg correction). (B) Comparative expression of indicated RNAs in control or Dux KO MERVL-GFP mESCs transduced with lentiviral vectors targeting a control or Trim28. RNA expression data is normalized to Actb. (C) RNA sequencing of control and Trim28 KD on WT mESC clones. The dot plot represents the average gene expression of three independent clones in the two different conditions. The blue dot represents Trim28, pink dots different Dux transcripts, green dots the genes specifically expressed in 2C-like mESCs (selected from figure 3B), grey dots the rest of the genes. (D) Comparative expression of indicated RNAs in control or Trim28 KO mESCs transduced with control or Dux KD (sh2 and sh5) lentiviral vectors targeting a control or Dux. RNA expression data is normalized to Actb. (E) Fold enrichment of Trim28 ChIP-qPCR in mESCs of IAPEz (positive control) and Dux over MERVL (negative control). (F) Fold enrichment of H3K9me3 ChIP-qPCR in mESCs transduced with control or Trim28 KD lentiviral vector of IAPEz (positive control) and Dux over MERVL (negative control). Shown in (B) are the mean ± SD for three independent clones. Shown in (D) and (F) are the mean ± standard deviation for three experiments performed on different days. Shown in (E) are the mean ± standard deviation for two experiments performed on different days. * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, t-test.

Supplementary Figure 12 Ex vivo analysis of Dux KO in preimplantation embryos.

(A) Average percent of embryos reaching the different embryonic stages or failing differentiation (dead) over 4 days. Shown in the bar plot are the mean ± standard deviation for three experiments following between 16 and 23 embryos for each condition performed on different days. * p ≤ 0.05, ** p ≤ 0.01, two-way ANOVA. Statistical tests were made between control and Dux sgRNA-injected embryos. (B) Brightfield images of day 4 embryos injected with control or Dux-specific sgRNAs (* healthy blastocyst). (C) PCR products using the Dux KO specific primer combinations (Supplementary figure 7) on: a pool of day 4 embryos from Figure 5B injected with the control (1) or Dux-specific (2) sgRNAs, WT (3) and Dux KO (4) mESC clones, and water (5).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 (PDF 2735 kb)

Supplementary Table 1

Genes differentially expressed in human primary myoblasts upon ectopic expression of DUX4 (XLSX 38 kb)

Supplementary Table 2

Primer list (XLSX 28 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

De Iaco, A., Planet, E., Coluccio, A. et al. DUX-family transcription factors regulate zygotic genome activation in placental mammals. Nat Genet 49, 941–945 (2017). https://doi.org/10.1038/ng.3858

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3858

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research