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:

lncRNA DIGIT and BRD3 protein form phase-separated condensates to regulate endoderm differentiation

Abstract

Cooperation between DNA, RNA and protein regulates gene expression and controls differentiation through interactions that connect regions of nucleic acids and protein domains and through the assembly of biomolecular condensates. Here, we report that endoderm differentiation is regulated by the interaction between the long non-coding RNA (lncRNA) DIGIT and the bromodomain and extraterminal domain protein BRD3. BRD3 forms phase-separated condensates of which the formation is promoted by DIGIT, occupies enhancers of endoderm transcription factors and is required for endoderm differentiation. BRD3 binds to histone H3 acetylated at lysine 18 (H3K18ac) in vitro and co-occupies the genome with H3K18ac. DIGIT is also enriched in regions of H3K18ac, and the depletion of DIGIT results in decreased recruitment of BRD3 to these regions. Our findings show that cooperation between DIGIT and BRD3 at regions of H3K18ac regulates the transcription factors that drive endoderm differentiation and suggest that protein–lncRNA phase-separated condensates have a broader role as regulators of transcription.

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: DIGIT interacts with BRD3.
Fig. 2: DIGIT interacts with recombinant BRD3 in vitro, and bromodomains mediate the interaction between BRD3 and DIGIT.
Fig. 3: BRD3 forms protein–RNA phase-separated condensates.
Fig. 4: DIGIT promotes the formation of BRD3 phase-separated condensates.
Fig. 5: BRD3 regulates endoderm genes.
Fig. 6: BRD3 and DIGIT interact with acetylated H3K18.
Fig. 7: BRD3 interacts with DIGIT-regulated genes on H3K18ac-marked chromatin.
Fig. 8: Loss of DIGIT inhibits the recruitment of BRD3.

Similar content being viewed by others

Data availability

ChIP–seq and RIP–seq data generated by this study have been deposited in the GEO under the accession codes GSE126661 (BRD3 ChIP–seq) and GSE149032 (BRD3 RIP–seq). Previously published RNA-seq data that were reanalysed can be found at GSE75297. Previously published ChIP–seq data that were reanalysed can be found at GSE16256. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Flynn, R. A. & Chang, H. Y. Long noncoding RNAs in cell-fate programming and reprogramming. Cell Stem Cell 14, 752–761 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Perry, R. B.-T. & Ulitsky, I. The functions of long noncoding RNAs in development and stem cells. Development 143, 3882–3894 (2016).

    Article  PubMed  Google Scholar 

  3. Jiang, W., Liu, Y., Liu, R., Zhang, K. & Zhang, Y. The lncRNA DEANR1 facilitates human endoderm differentiation by activating FOXA2 expression. Cell Rep. 11, 137–148 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Daneshvar, K. et al. DIGIT Is a conserved long noncoding RNA that regulates GSC expression to control definitive endoderm differentiation of embryonic stem cells. Cell Rep. 17, 353–365 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bell, C. C. et al. The Evx1/Evx1as gene locus regulates anterior-posterior patterning during gastrulation. Sci. Rep. 6, 26657 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Luo, S. et al. Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell 18, 637–652 (2016).

  7. Kanno, T. et al. Selective recognition of acetylated histones by bromodomain proteins visualized in living cells. Mol. Cell 13, 33–43 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Filippakopoulos, P. et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 149, 214–231 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Paillisson, A. et al. Bromodomain testis-specific protein is expressed in mouse oocyte and evolves faster than its ubiquitously expressed paralogs BRD2, -3, and -4. Genomics 89, 215–223 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Wai, D. C. C. et al. The BRD3 ET domain recognizes a short peptide motif through a mechanism that is conserved across chromatin remodelers and transcriptional regulators. J. Biol. Chem. 293, 7160–7175 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Deeney, J. T., Belkina, A. C., Shirihai, O. S., Corkey, B. E. & Denis, G. V. BET bromodomain proteins Brd2, Brd3 and Brd4 selectively regulate metabolic pathways in the pancreatic β-cell. PLoS ONE 11, e0151329 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Jang, M. K. et al. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol. Cell 19, 523–534 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Houzelstein, D. et al. Growth and early postimplantation defects in mice deficient for the bromodomain-containing protein Brd4. Mol. Cell. Biol. 22, 3794–3802 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shang, E., Wang, X., Wen, D., Greenberg, D. A. & Wolgemuth, D. J. Double bromodomain-containing gene Brd2 is essential for embryonic development in mouse. Dev. Dyn. 238, 908–917 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Di Micco, R. et al. Control of embryonic stem cell identity by BRD4-dependent transcriptional elongation of super-enhancer-associated pluripotency genes. Cell Rep. 9, 234–247 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Fernandez-Alonso, R. et al. Brd4-Brd2 isoform switching coordinates pluripotent exit and Smad2-dependent lineage specification. EMBO Rep. 18, 1108–1122 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Leppek, K. & Stoecklin, G. An optimized streptavidin-binding RNA aptamer for purification of ribonucleoprotein complexes identifies novel ARE-binding proteins. Nucleic Acids Res. 42, e13 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Srisawat, C. & Engelke, D. R. Streptavidin aptamers: affinity tags for the study of RNAs and ribonucleoproteins. RNA 7, 632–641 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Neumann, P. et al. The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2. Nat. Commun. 9, 237 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Oh, J. et al. BAF60a interacts with p53 to recruit the SWI/SNF complex. J. Biol. Chem. 283, 11924–11934 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Alajem, A. et al. Differential association of chromatin proteins identifies BAF60a/SMARCD1 as a regulator of embryonic stem cell differentiation. Cell Rep. 10, 2019–2031 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Maori, E. et al. A Secreted RNA binding protein forms RNA-stabilizing granules in the honeybee royal jelly. Mol. Cell 74, 598–608 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Queiroz, R. M. L. et al. Comprehensive identification of RNA-protein interactions in any organism using orthogonal organic phase separation (OOPS). Nat. Biotechnol. 37, 169–178 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kato, M. et al. Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell 149, 753–767 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, P. et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 483, 336–340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lin, Y., Protter, D. S. W., Rosen, M. K. & Parker, R. Formation and maturation of phase-separated liquid droplets by RNA-binding proteins. Mol. Cell 60, 208–219 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lu, H. et al. Phase-separation mechanism for C-terminal hyperphosphorylation of RNA polymerase II. Nature 558, 318–323 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Boija, A. et al. Transcription factors activate genes through the phase-separation capacity of their activation domains. Cell 175, 1842–1855 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Banani, S. F., Lee, H. O., Hyman, A. A. & Rosen, M. K. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18, 285–298 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kowarz, E., Löscher, D. & Marschalek, R. Optimized sleeping beauty transposons rapidly generate stable transgenic cell lines. Biotechnol. J. 10, 647–653 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Deglincerti, A., Etoc, F., Ozair, M. Z. & Brivanlou, A. H. Self-organization of spatial patterning in human embryonic stem cells. Curr. Top. Dev. Biol. 116, 99–113 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Warmflash, A., Sorre, B., Etoc, F., Siggia, E. D. & Brivanlou, A. H. A method to recapitulate early embryonic spatial patterning in human embryonic stem cells. Nat. Methods 11, 847–854 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jin, Q. et al. Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation. EMBO J. 30, 249–262 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Lasko, L. M. et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 550, 128–132 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Raisner, R. et al. Enhancer activity requires CBP/P300 bromodomain-dependent histone H3K27 acetylation. Cell Rep. 24, 1722–1729 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Tada, S. et al. Characterization of mesendoderm: a diverging point of the definitive endoderm and mesoderm in embryonic stem cell differentiation culture. Development 132, 4363–4374 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Vallier, L. et al. Early cell fate decisions of human embryonic stem cells and mouse epiblast stem cells are controlled by the same signalling pathways. PLoS ONE 4, e6082 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Cuatrecasas, P., Fuchs, S. & Anfinsen, C. B. Catalytic properties and specificity of the extracellular nuclease of Staphylococcus aureus. J. Biol. Chem. 242, 1541–1547 (1967).

    Article  CAS  PubMed  Google Scholar 

  41. West, J. A. et al. The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol. Cell 55, 791–802 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mondal, T. et al. MEG3 long noncoding RNA regulates the TGF-β pathway genes through formation of RNA–DNA triplex structures. Nat. Commun. 6, 7743 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kopp, F. & Mendell, J. T. Functional classification and experimental dissection of long noncoding RNAs. Cell 172, 393–407 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xi, Q. et al. A poised chromatin platform for TGF-β access to master regulators. Cell 147, 1511–1524 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Luo, M. et al. H3K18ac primes mesendodermal differentiation upon nodal signaling. Stem Cell Rep. 13, 642–656 (2019).

    Article  CAS  Google Scholar 

  47. Pouponnot, C., Jayaraman, L. & Massagué, J. Physical and functional interaction of SMADs and p300/CBP. J. Biol. Chem. 273, 22865–22868 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Janknecht, R., Wells, N. J. & Hunter, T. TGF-β-stimulated cooperation of Smad proteins with the coactivators CBP/p300. Genes Dev. 12, 2114–2119 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rahnamoun, H. et al. RNAs interact with BRD4 to promote enhanced chromatin engagement and transcription activation. Nat. Struct. Mol. Biol. 25, 687–697 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Molliex, A. et al. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell 163, 123–133 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Yamazaki, T. et al. Functional domains of NEAT1 architectural lncRNA induce paraspeckle assembly through phase separation. Mol. Cell 70, 1038–1053 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Hutchinson, J. N. et al. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC Genom. 8, 39 (2007).

    Article  Google Scholar 

  53. Ito, T., Ikehara, T., Nakagawa, T., Kraus, W. L. & Muramatsu, M. p300-mediated acetylation facilitates the transfer of histone H2A-H2B dimers from nucleosomes to a histone chaperone. Genes Dev. 14, 1899–1907 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Loh, K. M. et al. Efficient endoderm induction from human pluripotent stem cells by logically directing signals controlling lineage bifurcations. Cell Stem Cell 14, 237–252 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Deglincerti, A. et al. Self-organization of human embryonic stem cells on micropatterns. Nat. Protoc. 11, 2223–2232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Mellacheruvu, D. et al. The CRAPome: a contaminant repository for affinity purification–mass spectrometry data. Nat. Methods 10, 730–736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Skene, P. J., Henikoff, J. G. & Henikoff, S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat. Protoc. 13, 1006–1019 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Putnam, F. W. & Neurath, H. The precipitation of proteins by synthetic detergents 1a. J. Am. Chem. Soc. 66, 692–697 (1944).

    Article  CAS  Google Scholar 

  59. Rinn, J. L. et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chicaybam, L., Sodre, A. L., Curzio, B. A. & Bonamino, M. H. An efficient low cost method for gene transfer to T lymphocytes. PLoS ONE 8, e60298 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Grau, D. J. et al. Compaction of chromatin by diverse Polycomb group proteins requires localized regions of high charge. Genes Dev. 25, 2210–2221 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Abmayr, S. M., Yao, T., Parmely, T. & Workman, J. L. Preparation of nuclear and cytoplasmic extracts from mammalian cells. Curr. Protoc. Mol. Biol. 75, 12.1.1–12.1.10 (2006).

    Article  Google Scholar 

  63. Saltzman, A. L., Soo, M. W., Aram, R. & Lee, J. T. Multiple histone methyl-lysine readers ensure robust development and germline immortality in Caenorhabditis elegans. Genetics https://doi.org/10.1534/genetics.118.301518 (2018).

  64. Aranda, P. S., LaJoie, D. M. & Jorcyk, C. L. Bleach gel: a simple agarose gel for analyzing RNA quality. Electrophoresis 33, 366–369 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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 

  66. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  68. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  70. Kent, W. J., Zweig, A. S., Barber, G., Hinrichs, A. S. & Karolchik, D. BigWig and BigBed: enabling browsing of large distributed datasets. Bioinformatics 26, 2204–2207 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Tomaino and staff at the Taplin Mass Spectrometry Core at Harvard Medical School, the MGH nextGen Sequencing Core, the MGH PMB Microscopy Core Facility and the HSCI-CRM Flow Cytometry Core Facility at MGH; T. Kitao-Ando (Gifu University) for providing consultation in the optimization of the RNA pull-down protocol; and M. Beal, J. Ryu and R. Cook of Optical Biosystems for their continuous help and collaboration with RNA detection and high-throughput microscopy. A.C.M. was supported by NIH/NICHD grant R01HD09277302 and a Pew Biomedical Scholars Award. R.E.K. was supported by NIH grants R01GM043901, R37GM048405 and R35GM131743. R.A.Y. was supported by NIH grant R01GM123511.

Author information

Authors and Affiliations

Authors

Contributions

K.D. and A.C.M. conceived the study. K.D. and A.C.M. designed the experiments with M.B.A., I.A.K. and A.M.; M.B.A. made a conceptual contribution to the functional characterization of BRD3. Computational analyses were performed by C.Z. and A.M. with help from K.D., A.J.K. and S.P.M. Bench experiments were performed by K.D., M.B.A., I.A.K, F.-K.H., A.J.K., S.O.L.C., A.M., W.L., B.M.C., S.K.G. and J.V.P. The manuscript was written by K.D. and A.C.M. with input from R.A.Y., R.E.K. and all of the other authors.

Corresponding author

Correspondence to Alan C. Mullen.

Ethics declarations

Competing interests

A.C.M. receives research funding from Boehringer Ingelheim, Bristol-Myers Squibb, Roche Pharmaceuticals and Takeda Pharmaceuticals for unrelated projects. R.A.Y. is a founder and shareholder of Syros Pharmaceuticals, Camp4 Therapeutics, Dewpoint Therapeutics and Omega Therapeutics. I.A.K. is a consultant and a shareholder of Dewpoint Therapeutics.

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 Optimization of RNA pull-down, endoderm differentiation, and evaluation of GATA6-AS in endoderm.

a, Recovery of DIGIT-4xS1m versus endogenous GAPDH mRNA with increasing concentrations of KCl. RNA recovered with 250 mM KCl is set to 100 for each RNA species. Error bars represent the standard deviation of n = 3 independent experiments. b, Enrichment of SCRM-4xS1m after streptavidin pull-down as quantified by RT-qPCR. The transcript level in input was set to 1.0. RNA levels are normalized to GAPDH. **** indicates p < 0.0001 (Student’s t-test, n = 3 independent experiments). Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). c, Immunostaining shows the differentiation of hESCs to definitive endoderm (DE). FOXA2 (red) and SOX17 (green) are used as markers of definitive endoderm. SOX2 (yellow) is used as a marker of pluripotency. Hoechst staining (blue) marks nuclei. Scale bar represents 100 μm. This experiment was performed twice with similar results. d, Flow cytometry plots show the population of cells that are CXCR4 and c-KIT double-positive in hESCs and DE cells generated using the high-efficiency DE differentiation protocol54. This experiment was performed three times with similar results. e, The diagram shows the gene encoding lncRNA GATA6-AS, which is divergently transcribed from the gene encoding GATA6. Annotated GATA6-AS transcripts are shown in red and the transcript cloned in DE cells is shown in black. f, qRT-PCR shows levels of GATA6-AS in hESCs and on the indicated days of endoderm differentiation. Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). **** indicates p < 0.0001 compared to day 0 (One-way ANOVA, n = 4 independent experiments). Statistical source data are provided in Source data extended data Fig. 1.

Source data

Extended Data Fig. 2 RIP-seq, expression of BET proteins in endoderm, and interactions with SMARCD1.

a, RIP shows the levels of DIGIT following SMARCD1 and IgG immunoprecipitation. *** indicates p < 0.001 n = 3 independent experiments. Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). b, Co-immunoprecipitation followed by immunoblotting shows the interaction of BRD3 and SMARCD1. This experiment was performed twice with similar results. c, Volcano plots show transcripts enriched with BRD3 RIP. Transcripts from BRD3 RIP were normalized to IgG RIP for endoderm cells (DE, Fig. 1g) and normalized to total nuclear RNA (input), left. BRD3 RIP normalized to IgG (center) and input (right) are shown for hESCs. The 10 most enriched transcripts in common between IgG and input for hESCs are labeled on the volcano plots. Points are colored to indicate different thresholds as labeled on the far right. RIP-seq was performed in two independent samples and pooled for analysis. d, Volcano plots show lncRNA transcripts enriched with BRD3 RIP when normalized to IgG or input in DE (left) and hESCs (right). lncRNA transcripts in common between IgG and input controls are labeled (purple). Points are shaded to indicate different thresholds as in (c). RIP-seq was performed in two independent samples and pooled for analysis. e, The table (left) shows normalized expression values (RPKM) in endoderm differentiation4. Immunoblot (right) shows the protein levels of BET proteins with differentiation. Beta-Actin (ACTB) is used as a loading control. This experiment was performed twice with similar results. f, Immunoprecipitation was performed using antibodies specific to each BET protein. IgG serves as a control. This experiment was performed twice with similar results. Unprocessed blots and statistical source data are provided in Source data extended data Fig. 2.

Source data

Extended Data Fig. 3 Generation of recombinant BRD3 and GFP, production of Cy-5 labeled RNAs, EMSA replicate, localization of BRD3 domains, and immunoprecipitation of BRD3 domains.

a, Immunoblot was performed to detect recombinant FLAG-mEGFP-BRD3 (long isoform) and FLAG-mEGFP using an anti-GFP antibody (left) and an anti-BRD3 antibody (right). b, Fluorescent detection of the Cy5-labeled DIGIT and SCRM on a 1% bleach-agarose gel. Experiments in A and B were performed twice with similar results. c, Replicate electrophoretic mobility shift assay (EMSA) of in vitro transcribed DIGIT (left, top) and SCRM (left, bottom), which are labeled with Cy5 and incubated with recombinant BRD3. Migration was visualized on the right by quantifying the signal for DIGIT (top) or >SCRM (bottom) across the gel. Migration of 0 on the x-axis was defined by the peak of DIGIT and SCRM with 0 nM BRD3 protein, and migration of 100 was defined by the peak of DIGIT and SCRM with 2048 nM BRD3 protein. This experiment shows a second replicate of Fig. 2c. d, The dissociation constants (Kd) were calculated for (c). e, Fluorescence microscopy shows the nuclear localization of ectopically expressed GFP, GFP fused to full-length BRD3, and GFP fused to the indicated bromodomains (BD) or extra terminal (ET) domains of BRD3 in HEK 293 cells. Scale bar represents 20 μm. This experiment was performed twice with similar results. f, Immunoprecipitation followed by immunoblotting using an antibody against GFP shows the precipitation of the proteins described in (e). This experiment was performed twice with similar results. Unprocessed blots are provided in Source data extended data Fig. 3.

Source data

Extended Data Fig. 4 Prediction of ordered/disordered protein structures, strategy for tagging endogenous BRD3 with mEGFP, formation of BRD3 puncta in hESCs, and in vitro droplet formation.

a, PONDR VSL2 plots showing the ordered and disordered regions of the long (left) and short (middle) isoforms of BRD3, and BRD2 (right). b, Creation of mEGFP-BRD3 fusion protein. The cDNA encoding mEGFP was inserted at the N-terminus of BRD3. This terminus is shared by the long and short isoforms of BRD3. The site targeted by gRNA is indicated with an arrow. The start of the coding sequence (CDS) is indicated, and the mEGFP cDNA is inserted in frame after the start codon. c, Design of the homology vector for the insertion of mEGFP. The genomic locations of homology arms are indicated. Arrow indicates that mEGFP was inserted after the start codon for BRD3. d, Immunoblot using anti-BRD3 antibody confirms the generation of mEGFP-BRD3 fused protein. hESCs were lysed and IP was performed using an IgG isotype control and an antibody recognizing GFP. Total cell lysates (input) and the IPs were then probed with an anti-BRD3 antibody. This experiment was performed twice with similar results. e, Immunofluorescence of an hESC showing BRD3 puncta (green) in the nucleus (blue). Scale bar represents 10 ÎĽm. This experiment was performed twice with similar results. f, Droplet assay shows the formation of mEGFP-BRD3 droplets in the presence of three different molecular crowding agents. Scale bar represents 2 ÎĽm. This experiment was performed three times with similar results. g, Fluorescent detection of the Cy3-labeled DIGIT on a 1% bleach-agarose gel. This experiment was performed twice with similar results.

Source data

Extended Data Fig. 5 Homology construct for targeting BRD3, and analysis of long and short isoforms of BRD3.

a, Map of the homology construct for insertion of a puromycin resistance cassette and a stop codon cassette downstream of the sixth codon of the gene encoding BRD3. b, hESCs were differentiated towards DE for three days prior to qRT-PCR. Primers that uniquely recognize either the long (left) or short (right) isoforms of BRD3 were used for amplification. Cells were also transfected with plasmids expressing the long (left) and short (right) isoforms of BRD3 as positive (+) controls for the primers. The long isoform of BRD3 is detected with DE differentiation while the short isoform is not detected. ACTB was used as a control. This experiment was performed twice with similar results. c, Wildtype and BRD3-/- hESCs were transfected with plasmids expressing GFP (green), the short isoform of BRD3 (yellow), and the long isoform of BRD3 (orange). Expression of CXCR4 and SOX17 was quantified by qRT-PCR after three days of DE differentiation. Ectopic expression of both the long and short isoform of BRD3 rescued expression of CXCR4 and SOX17. * indicates p < 0.05 (n = 3 independent experiments). Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). Statistical source data are provided in Source data extended data Fig. 5.

Source data

Extended Data Fig. 6 Binding of BRD3 to modified histones, effects of inhibition of P300 on the enrichment of BRD3, the activity of MNase in presence of Ba+2 and Sr+2 cations, and enrichment of lncRNAs in specific histone modifications.

a, Peptide arrays show the binding of recombinant BRD3 to histone modifications. b, Spot intensity of BRD3 (long and short isoforms, top and bottom respectively) binding to the top ten histone modifications as quantified by image processing software (see Materials and Methods). c, Immunoblots to detect the levels of acetylation on H3K18 and H3K27, as well as levels of BRD3 and total H3 in hESCs treated with DMSO, A485, or GNE049. This experiment was performed twice with similar results. d, CUT&RUN followed by qPCR shows the enrichment of BRD3 at the HEXIM2 and ZFP36L1 loci in hESCs. Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). *** indicates p < 0.001 (Student’s t-test, n = 3 independent experiments). e, RNase activity of MNase in the presence of Ba2+ and Sr2+ cations. This experiment was performed twice with similar results. f, DNase activity of MNase in the presence of Ba2+ and Sr2+ cations. This experiment was performed twice with similar results. g, CUT&RUNER followed by qPCR shows the enrichment of MALAT1 and MEG3 transcripts in IgG control, markers of gene activation (H3K4me3 and H3K18ac) and repression (H3K27me3). Box plots show the first and third quartiles, median (horizontal line), and minimum and maximum values (whiskers). * indicates p < 0.05, *** indicates p < 0.001, and **** indicates p < 0.0001 compared to IgG (Student’s t-test, n = 3 independent experiments). Statistical source data are provided in Source data extended data Fig. 6.

Source data

Extended Data Fig. 7 BRD3 occupies endoderm genes and enhancers and interaction between BRD3 and DIGIT is not dependent on location of transcription.

a, The number of BRD3 regions (y-axis) that contain promoters and enhancers are shown for hESCs (white) and endoderm cells (black). BRD3 regions are defined as containing promoters if the region is located within 2 kb of a transcription start site (TSS). BRD3 regions are defined as containing enhancers if the region overlaps with enhancers defined by H3K27ac. Regions that contain both promoters and enhancers are counted in both categories. In Fig. 7a, BRD3 regions are first evaluated for association with promoters. Regions that are not associated with promoters are then evaluated for association with enhancers (See Supplementary Methods). Data from two independent BRD3 ChIPs were pooled for analysis of hESCs and DE. b, Box plots show the length of regions occupied by BRD3 for the categories described in (a). The bold horizontal line represents the median for each box plot. The lower edge of the box represents the first quartile (Q1) and the upper edge of the box represents the third quartile (Q3). The lower whiskers represent Q1–1.5*(Q3-Q1) and the upper whisker represents Q3 + 1.5*(Q3-Q1). The width of each box is proportional to the number of BRD3 regions in each category. Data from two independent BRD3 ChIPs were pooled for analysis of hESCs and DE. The width of each box is proportional to the number of BRD3 regions in each category. Data from two independent BRD3 ChIPs were pooled for analysis of hESCs and DE. c, Transcript enrichment from BRD3 RIP in DE vs IgG control (Fig. 1g, y-axis) is shown for each gene occupied by BRD3 in DE (x-axis). Endoderm genes are labeled in red. Data from two independent BRD3 ChIPs and two independent BRD3 RIPs were pooled for analysis.

Extended Data Fig. 8 BRD3 occupancy at genes not regulated by DIGIT.

ChIP-seq data shows H3K18ac (top, green) and BRD3 occupancy (bottom, black) at HEXIM2, PITX2/PANCR and ZAP70 in hESCs and endoderm/mesendoderm cells. BRD3 occupies HEXIM2 in hESCs and mesendoderm/endoderm. BRD3 occupies PITX2/PANCR and ZAP70 in mesendoderm/endoderm but not in hESCs. Data from two independent BRD3 ChIPs were pooled for analysis. H3K18ac was analyzed from GSE16256.

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Table 1

MS and RIP–seq analysis, related to Fig. 1.

Supplementary Table 2

Histone array data, related to Fig. 6.

Supplementary Table 3

Regions occupied by BRD3, H3K18ac and H3K27ac, related to Figs. 6 and 7.

Supplementary Table 4

Genes, transcripts and regions analysed in Fig. 7.

Supplementary Table 5

Sequences of oligonucleotides, gRNAs and RNA probes.

Supplementary Table 6

Antibody catalogue numbers and dilutions.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Daneshvar, K., Ardehali, M.B., Klein, I.A. et al. lncRNA DIGIT and BRD3 protein form phase-separated condensates to regulate endoderm differentiation. Nat Cell Biol 22, 1211–1222 (2020). https://doi.org/10.1038/s41556-020-0572-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0572-2

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