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:

Hierarchy within the mammary STAT5-driven Wap super-enhancer

Abstract

Super-enhancers comprise dense transcription factor platforms highly enriched for active chromatin marks. A paucity of functional data led us to investigate the role of super-enhancers in the mammary gland, an organ characterized by exceptional gene regulatory dynamics during pregnancy. ChIP-seq analysis for the master regulator STAT5A, the glucocorticoid receptor, H3K27ac and MED1 identified 440 mammary-specific super-enhancers, half of which were associated with genes activated during pregnancy. We interrogated the Wap super-enhancer, generating mice carrying mutations in STAT5-binding sites within its constituent enhancers. Individually, the most distal site displayed the greatest enhancer activity. However, combinatorial mutation analysis showed that the 1,000-fold induction in gene expression during pregnancy relied on all enhancers. Disabling the binding sites of STAT5, NFIB and ELF5 in the proximal enhancer incapacitated the entire super-enhancer. Altogether, these data suggest a temporal and functional enhancer hierarchy. The identification of mammary-specific super-enhancers and the mechanistic exploration of the Wap locus provide insights into the regulation of cell-type-specific expression of hormone-sensing genes.

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

Access options

Buy this article

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

Figure 1: Identification of mammary-specific super-enhancers.
Figure 2: Transcription factor binding signatures in mammary-specific super-enhancers.
Figure 3: Assembly at constituent enhancers in mammary-specific super-enhancers during pregnancy.
Figure 4: In vivo functions of individual enhancers in the mammary-specific Wap super-enhancer.
Figure 5: Consequences of individual and combined loss of STAT5-binding sites at enhancers in the mammary-specific Wap super-enhancer.
Figure 6: In vivo function of the E1 epicenter in the Wap super-enhancer.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Hennighausen, L. & Robinson, G.W. Information networks in the mammary gland. Nat. Rev. Mol. Cell Biol. 6, 715–725 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Lydon, J.P. et al. Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities. Genes Dev. 9, 2266–2278 (1995).

    Article  CAS  PubMed  Google Scholar 

  3. Ormandy, C.J. et al. Null mutation of the prolactin receptor gene produces multiple reproductive defects in the mouse. Genes Dev. 11, 167–178 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Horseman, N.D. et al. Defective mammopoiesis, but normal hematopoiesis, in mice with a targeted disruption of the prolactin gene. EMBO J. 16, 6926–6935 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu, X. et al. Stat5a is mandatory for adult mammary gland development and lactogenesis. Genes Dev. 11, 179–186 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Cui, Y. et al. Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation. Mol. Cell. Biol. 24, 8037–8047 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhou, J. et al. Elf5 is essential for early embryogenesis and mammary gland development during pregnancy and lactation. EMBO J. 24, 635–644 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wakao, H., Gouilleux, F. & Groner, B. Mammary gland factor (MGF) is a novel member of the cytokine regulated transcription factor gene family and confers the prolactin response. EMBO J. 13, 2182–2191 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu, X., Robinson, G.W., Gouilleux, F., Groner, B. & Hennighausen, L. Cloning and expression of Stat5 and an additional homologue (Stat5b) involved in prolactin signal transduction in mouse mammary tissue. Proc. Natl. Acad. Sci. USA 92, 8831–8835 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yamaji, D., Kang, K., Robinson, G.W. & Hennighausen, L. Sequential activation of genetic programs in mouse mammary epithelium during pregnancy depends on STAT5A/B concentration. Nucleic Acids Res. 41, 1622–1636 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Hennighausen, L.G. & Sippel, A.E. Characterization and cloning of the mRNAs specific for the lactating mouse mammary gland. Eur. J. Biochem. 125, 131–141 (1982).

    Article  CAS  PubMed  Google Scholar 

  12. Richards, D.A., Rodgers, J.R., Supowit, S.C. & Rosen, J.M. Construction and preliminary characterization of the rat casein and α-lactalbumin cDNA clones. J. Biol. Chem. 256, 526–532 (1981).

    CAS  PubMed  Google Scholar 

  13. Pittius, C.W., Sankaran, L., Topper, Y.J. & Hennighausen, L. Comparison of the regulation of the whey acidic protein gene with that of a hybrid gene containing the whey acidic protein gene promoter in transgenic mice. Mol. Endocrinol. 2, 1027–1032 (1988).

    Article  CAS  PubMed  Google Scholar 

  14. Li, S. & Rosen, J.M. Distal regulatory elements required for rat whey acidic protein gene expression in transgenic mice. J. Biol. Chem. 269, 14235–14243 (1994).

    CAS  PubMed  Google Scholar 

  15. Li, S. & Rosen, J.M. Nuclear factor I and mammary gland factor (STAT5) play a critical role in regulating rat whey acidic protein gene expression in transgenic mice. Mol. Cell. Biol. 15, 2063–2070 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McKnight, R.A., Wall, R.J. & Hennighausen, L. Expression of genomic and cDNA transgenes after co-integration in transgenic mice. Transgenic Res. 4, 39–43 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Burdon, T.G., Maitland, K.A., Clark, A.J., Wallace, R. & Watson, C.J. Regulation of the sheep β-lactoglobulin gene by lactogenic hormones is mediated by a transcription factor that binds an interferon-γ activation site–related element. Mol. Endocrinol. 8, 1528–1536 (1994).

    CAS  PubMed  Google Scholar 

  18. Greenberg, N.M., Reding, T.V., Duffy, T. & Rosen, J.M. A heterologous hormone response element enhances expression of rat β-casein promoter-driven chloramphenicol acetyltransferase fusion genes in the mammary gland of transgenic mice. Mol. Endocrinol. 5, 1504–1512 (1991).

    Article  CAS  PubMed  Google Scholar 

  19. Gordon, K. et al. Production of human tissue plasminogen activator in transgenic mouse milk. 1987. Biotechnology 24, 425–428 (1992).

    CAS  PubMed  Google Scholar 

  20. Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272–286 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Ong, C.T. & Corces, V.G. Enhancer function: new insights into the regulation of tissue-specific gene expression. Nat. Rev. Genet. 12, 283–293 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ong, C.T. & Corces, V.G. Enhancers: emerging roles in cell fate specification. EMBO Rep. 13, 423–430 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Natoli, G. & Andrau, J.C. Noncoding transcription at enhancers: general principles and functional models. Annu. Rev. Genet. 46, 1–19 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Heinz, S., Romanoski, C.E., Benner, C. & Glass, C.K. The selection and function of cell type–specific enhancers. Nat. Rev. Mol. Cell Biol. 16, 144–154 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Pott, S. & Lieb, J.D. What are super-enhancers? Nat. Genet. 47, 8–12 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Adam, R.C. et al. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 521, 366–370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Brown, J.D. et al. NF-κB directs dynamic super enhancer formation in inflammation and atherogenesis. Mol. Cell 56, 219–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chapuy, B. et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell 24, 777–790 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chipumuro, E. et al. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159, 1126–1139 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fang, Z. et al. Transcription factor co-occupied regions in the murine genome constitute T-helper-cell subtype-specific enhancers. Eur. J. Immunol. 45, 3150–3157 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. Hnisz, D. et al. Convergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers. Mol. Cell 58, 362–370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Huang, J. et al. Dynamic control of enhancer repertoires drives lineage and stage-specific transcription during hematopoiesis. Dev. Cell 36, 9–23 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, Y. et al. CRISPR reveals a distal super-enhancer required for Sox2 expression in mouse embryonic stem cells. PLoS One 9, e114485 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, C.F. & Lefebvre, V. The transcription factors SOX9 and SOX5/SOX6 cooperate genome-wide through super-enhancers to drive chondrogenesis. Nucleic Acids Res. 43, 8183–8203 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mansour, M.R. et al. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346, 1373–1377 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ohba, S., He, X., Hojo, H. & McMahon, A.P. Distinct transcriptional programs underlie Sox9 regulation of the mammalian chondrocyte. Cell Rep. 12, 229–243 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Parker, S.C. et al. Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc. Natl. Acad. Sci. USA 110, 17921–17926 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Pelish, H.E. et al. Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature 526, 273–276 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pinz, S., Unser, S. & Rascle, A. Signal transducer and activator of transcription STAT5 is recruited to c-Myc super-enhancer. BMC Mol. Biol. 17, 10 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Siersbæk, R. et al. Transcription factor cooperativity in early adipogenic hotspots and super-enhancers. Cell Rep. 7, 1443–1455 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Thakurela, S., Sahu, S.K., Garding, A. & Tiwari, V.K. Dynamics and function of distal regulatory elements during neurogenesis and neuroplasticity. Genome Res. 25, 1309–1324 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Vahedi, G. et al. Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Hennighausen, L.G. & Sippel, A.E. Mouse whey acidic protein is a novel member of the family of 'four-disulfide core' proteins. Nucleic Acids Res. 10, 2677–2684 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Burdon, T., Sankaran, L., Wall, R.J., Spencer, M. & Hennighausen, L. Expression of a whey acidic protein transgene during mammary development. Evidence for different mechanisms of regulation during pregnancy and lactation. J. Biol. Chem. 266, 6909–6914 (1991).

    CAS  PubMed  Google Scholar 

  49. Siersbæk, R. et al. Extensive chromatin remodelling and establishment of transcription factor 'hotspots' during early adipogenesis. EMBO J. 30, 1459–1472 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Robinson, G.W. et al. Coregulation of genetic programs by the transcription factors NFIB and STAT5. Mol. Endocrinol. 28, 758–767 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bayna, E.M. & Rosen, J.M. Tissue-specific, high level expression of the rat whey acidic protein gene in transgenic mice. Nucleic Acids Res. 18, 2977–2985 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Miyoshi, K. et al. Signal transducer and activator of transcription (Stat) 5 controls the proliferation and differentiation of mammary alveolar epithelium. J. Cell Biol. 155, 531–542 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li, S. & Rosen, J.M. Glucocorticoid regulation of rat whey acidic protein gene expression involves hormone-induced alterations of chromatin structure in the distal promoter region. Mol. Endocrinol. 8, 1328–1335 (1994).

    CAS  PubMed  Google Scholar 

  54. McKnight, R.A. et al. An Ets site in the whey acidic protein gene promoter mediates transcriptional activation in the mammary gland of pregnant mice but is dispensable during lactation. Mol. Endocrinol. 9, 717–724 (1995).

    CAS  PubMed  Google Scholar 

  55. McKnight, R.A., Spencer, M., Wall, R.J. & Hennighausen, L. Severe position effects imposed on a 1 kb mouse whey acidic protein gene promoter are overcome by heterologous matrix attachment regions. Mol. Reprod. Dev. 44, 179–184 (1996).

    Article  CAS  PubMed  Google Scholar 

  56. Witte, S., O'Shea, J.J. & Vahedi, G. Super-enhancers: asset management in immune cell genomes. Trends Immunol. 36, 519–526 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. González, A.J., Setty, M. & Leslie, C.S. Early enhancer establishment and regulatory locus complexity shape transcriptional programs in hematopoietic differentiation. Nat. Genet. 47, 1249–1259 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhou, H. et al. Epstein–Barr virus oncoprotein super-enhancers control B cell growth. Cell Host Microbe 17, 205–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Vahedi, G. et al. STATs shape the active enhancer landscape of T cell populations. Cell 151, 981–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yang, X.P. et al. Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat. Immunol. 12, 247–254 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kang, K., Yamaji, D., Yoo, K.H., Robinson, G.W. & Hennighausen, L. Mammary-specific gene activation is defined by progressive recruitment of STAT5 during pregnancy and the establishment of H3K4me3 marks. Mol. Cell. Biol. 34, 464–473 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Li, P., Spolski, R., Liao, W. & Leonard, W.J. Complex interactions of transcription factors in mediating cytokine biology in T cells. Immunol. Rev. 261, 141–156 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Yao, Z. et al. Stat5a/b are essential for normal lymphoid development and differentiation. Proc. Natl. Acad. Sci. USA 103, 1000–1005 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yao, Z. et al. Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109, 4368–4375 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Laurence, A. et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26, 371–381 (2007).

    Article  CAS  PubMed  Google Scholar 

  66. Wei, L., Laurence, A., Elias, K.M. & O'Shea, J.J. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J. Biol. Chem. 282, 34605–34610 (2007).

    Article  CAS  PubMed  Google Scholar 

  67. Metser, G. et al. An autoregulatory enhancer controls mammary-specific STAT5 functions. Nucleic Acids Res. 44, 1052–1063 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. 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 

  69. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ramírez, F., Dündar, F., Diehl, S., Grüning, B.A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Neph, S. et al. BEDOPS: high-performance genomic feature operations. Bioinformatics 28, 1919–1920 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  75. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Liao, Y., Smyth, G.K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. 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  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wikham, H. Ggplot2: Elegant Graphics for Data Analysis (2009).

Download references

Acknowledgements

We thank H. Smith from the NIDDK genomics core for never-ending help with next-generation sequencing and C. Liu from the NHLBI transgenic core for generating the CRISPR/Cas9-based mouse mutants. M.W. is a graduate student of the Individual Graduate Partnership Program (GPP) between NIH/NIDDK and the Medical University of Innsbruck. This work was performed in partial fulfillment of the graduation requirements for M.W. We thank K. Kang and S. Oh for discussions in the early stage of this project and Z. Trajanoski for advising M.W. during her graduate studies. This research was funded by the IPR of the NIDDK/NIH.

Author information

Authors and Affiliations

Authors

Contributions

H.Y.S. designed, executed and supervised genotyping, identified and validated all CRISPR/Cas9-based mutant founders, performed ChIP-seq, gene expression experiments and histological analysis, analyzed data and wrote the manuscript. M.W. designed experiments, analyzed ChIP-seq and RNA-seq data, performed computational and statistical analyses, and wrote the manuscript. K.H.Y. designed and conducted ChIP-seq, performed gene expression experiments and histological analysis, and analyzed data. X.Z. conducted genotyping, and performed and analyzed gene expression experiments for mutant mice. C.W. performed DNase-seq and ChIP-seq experiments and analyzed data. G.M. genotyped mutant mice, performed and analyzed gene expression experiments, and identified founder mice carrying mutations in combination. L.H. conceived and supervised the study, analyzed data and wrote the manuscript. H.Y.S., M.W. and L.H. wrote and finalized the manuscript, and all authors reviewed and approved the submitted version.

Corresponding author

Correspondence to Lothar Hennighausen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Representative scatterplots validating the reproducibility of ChIP-seq replicates.

(a) Reproducibility of STAT5A ChIP-seq peaks at L1. r is Spearman’s correlation coefficient. (b) Reproducibility of H3K27ac ChIP-seq peaks at L1. (c) Reproducibility of MED1 ChIP-seq peaks at L1. (d) Reproducibility of NFIB ChIP-seq peaks at L1. (e) Reproducibility of ELF5 ChIP-seq peaks at L1. (f) Reproducibility of STAT5A ChIP-seq peaks at p13.

Supplementary Figure 2 Identification of mammary-specific super-enhancers.

ChIP-seq peak calling was conducted for two STAT5A replicates. Of a total of 15,790 verified peaks, 10,953 coincided with H3K27ac marks (±500 bp). 549 of these peaks coincided with known promoter regions and 10,404 were located in non-promoter regions. 5,503 of these coincided with a GAS motif (TTCNNNGAA) (±250 bp). The non-promoter STAT5 peaks served as the basis for the super-enhancer analysis calculation applying the ROSE algorithm. As the size of mammary-specific super-enhancers might be different from that of super-enhancers identified in embryonic stem cells, the analysis was carried out using three stitched sizes: 12.5 kb, 25 kb and 35 kb. To obtain only mammary-specific super-enhancers, each stitched size was used as a parameter for the calculations based on STAT5A peaks and H3K27ac, GR and MED1. The basic workflow for the analysis was applied for the three stitching sizes. The first step comprised the application of the ROSE algorithm based on STAT5A peaks: (i) a stitching size of 12.5 kb for H3K27ac returned 504 super-enhancers, for GR returned 592 super-enhancers and for MED1 returned 347 super-enhancers; (ii) the stitching size of 25 kb resulted in 392 super-enhancers for H3K27ac, 359 super-enhancers for GR and 191 super-enhancers for MED1; and (iii) the third applied size of 35 kb specified 428 super-enhancers for H3K27ac, 424 super-enhancers for GR and 199 super-enhancers for MED1. Subsequently, these super-enhancers were overlapped, and only super-enhancers identified and reproduced for at least two factors (gray) were considered for following analysis. With regard to the three stitching sizes, this criterion was satisfied by 405 super-enhancers for 12.5 kb, 272 super-enhancers for 25 kb and 311 super-enhancers for 35 kb. Mammary-specific super-enhancers were identified from the subtraction of STAT5 super-enhancers shared with T cells and liver (12.5 kb), if at least 30% of the mammary super-enhancer overlapped with the T cell and liver ones. 347 mammary-specific super-enhancers were identified in the category with 12.5 kb, 233 mammary-specific enhancers were identified for the category with 25 kb and 270 mammary-specific super-enhancers were identified using a stitched size of 35 kb. These mammary-specific super-enhancers were annotated taking into account the two nearest genes, choosing the one with the higher FPKM value based on expression data obtained at day 1 of lactation. In the final step, nested super-enhancers, which had been annotated to the same gene, were removed, and a total of 440 high-confidence mammary-specific super-enhancers were obtained.

Supplementary Figure 3 Super-enhancers in mammary tissue.

(a) The Glycam1 locus is shown as an example of a mammary-specific super-enhancer. STAT5 binding and H3K27ac marks exist only in mammary tissue but not in T cells or liver. (b) The Aldoc locus is shown as an example of a highly expressed mammary gene with a lone enhancer.

Supplementary Figure 4 Transcription factor binding profile within a mammary-specific super-enhancer.

The Olah super-enhancer consists of three constituent enhancers (asterisk) that feature colocalization of STAT5A, GR, NFIB, ELF5, MED1 and H3K27ac.

Supplementary Figure 5 Gene expression of super-enhancers at day 13 of pregnancy.

Established super-enhancers (n = 22; mean of ~205 FPKM) show higher gene expression at day 13 of pregnancy than progressive super-enhancers from groups II and III (n = 386; mean of ~46 FPKM). Median, middle bar inside the box; IQR, 50% of the data; whiskers, 1.5 times the IQR.

Supplementary Figure 6 Transcription factor binding at a putative super-enhancer controlling the Csn locus.

(a) Transcription factor binding is shown at day 13 of pregnancy and day 1 of lactation. Transcription factor binding and H3K27ac marks are similar at these two developmental stages. (b) STAT5A binding and H3K27ac marks are shown at days 13, 14 and 16 of pregnancy and day 1 of lactation. STAT5A binding and H3K27ac marks are similar across the developmental stages.

Supplementary Figure 7 Alveolar development at days 13, 14 and 16 of pregnancy and day 1 of lactation.

(a) Sections of mammary tissue stained with hematoxylin and eosin at days 13, 14 and 16 of pregnancy and day 1 of lactation with low magnification (400×). The arrow indicates alveoli. At days 13 and 14 of pregnancy, mammary glands were sparsely filled with undeveloped alveoli. The degree of alveolar occupancy was similar at day 16 of pregnancy. At day 1 of lactation, mammary glands were filled with fully differentiated alveoli containing milk (asterisk). (b) Sections of mammary tissue stained with hematoxylin and eosin at days 13, 14 and 16 of pregnancy and day 1 of lactation with high magnification (800×).

Supplementary Figure 8 Scatterplots showing the relationship between mammary epithelial cell and mammary tissue ChIP-seq.

(a) The scatterplot depicts the correlation between mammary tissue and MECs at day 13 of pregnancy. r is Spearman’s correlation coefficient. (b) The scatterplot depicts the correlation between mammary tissue and MECs at day 1 of lactation.

Supplementary Figure 9 Characterization of mutant mice generated by CRISPR/Cas9, TALEN and homologous recombination to introduce point mutations.

ΔE1a carries an 11-bp deletion spanning the GAS motif in E1 (0.7 kb upstream of the TSS), and ΔE1b has a 27-bp deletion spanning the GAS motif and juxtaposed NFIB motif in E1. ΔE1c carries point mutations in the GAS, NFIB and ELF5 motifs in E1. The GAS motif is in red, the NFIB motif is in blue and the ELF5 motif is in green. Point mutations are underlined. ΔE2 has a 26-bp deletion spanning the GAS motif in E2 (1.4 kb upstream of the TSS), and ΔE3 has an 11-bp deletion spanning the GAS motif in E3 (5.6 kb upstream of the TSS). ΔE1a/ΔE2 has a 16-bp deletion spanning the GAS motif in E1 and a 6-bp deletion spanning the GAS motif in E2. ΔE2/ΔE3 has a 9-bp deletion spanning the GAS motif in E2 and an 11-bp deletion spanning the GAS motif in E3. ΔE1a/ΔE2/ΔE3 carries deletions in all three GAS sites.

Supplementary Figure 10 Comparison of transcription factor binding at E1 and E2 of the Wap locus.

(a) Binding of STAT5, NFIB and ELF5 does not coincide with the GAS motif at E1, whereas binding of NFIB and ELF5 colocalizes with STAT5 binding at E2. (b) ELF5 binding remains in ΔE1a, whereas ELF5 binding is absent in ΔE2, indicating that ELF5 binding is dependent on STAT5A at the E2 site.

Supplementary Figure 11 Transcription factor binding at the Wap super-enhancer in ΔE1b and ΔE1c mutant mice.

Binding of STAT5A and NFIB was reduced at the E1 site in ΔE1b mutant mice. Binding of STAT5A, NFIB and ELF5 was completely absent at the E1 site in ΔE1c mutant mice. The Lao1 locus is shown as a ChIP-seq control.

Supplementary Figure 12 Establishment of STAT5 enhancers at the Nfib and Elf5 loci.

(a) STAT5A binding and H3K27ac marks are enriched in a putative regulatory region of the Nfib locus. (b) STAT5A binding and H3K27ac marks are enriched in a putative regulatory region of the Elf5 locus.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Tables 4 and 5. (PDF 1827 kb)

Supplementary Table 1

Genes associated with mammary-specific super-enhancers. (XLSX 1702 kb)

Supplementary Table 2

Summary table for current super-enhancer studies. (XLSX 12 kb)

Supplementary Table 3

ChIP-seq mapping quality. (XLSX 20 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shin, H., Willi, M., Yoo, K. et al. Hierarchy within the mammary STAT5-driven Wap super-enhancer. Nat Genet 48, 904–911 (2016). https://doi.org/10.1038/ng.3606

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3606

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