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:

Enhancer invasion shapes MYCN-dependent transcriptional amplification in neuroblastoma

Abstract

Amplification of the locus encoding the oncogenic transcription factor MYCN is a defining feature of high-risk neuroblastoma. Here we present the first dynamic chromatin and transcriptional landscape of MYCN perturbation in neuroblastoma. At oncogenic levels, MYCN associates with E-box binding motifs in an affinity-dependent manner, binding to strong canonical E-boxes at promoters and invading abundant weaker non-canonical E-boxes clustered at enhancers. Loss of MYCN leads to a global reduction in transcription, which is most pronounced at MYCN target genes with the greatest enhancer occupancy. These highly occupied MYCN target genes show tissue-specific expression and are linked to poor patient survival. The activity of genes with MYCN-occupied enhancers is dependent on the tissue-specific transcription factor TWIST1, which co-occupies enhancers with MYCN and is required for MYCN-dependent proliferation. These data implicate tissue-specific enhancers in defining often highly tumor-specific ‘MYC target gene signatures’ and identify disruption of the MYCN enhancer regulatory axis as a promising therapeutic strategy in neuroblastoma.

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

Fig. 1: Deregulated MYCN binds active chromatin and amplifies transcription in neuroblastoma.
Fig. 2: Enhancer invasion shapes MYCN transcriptional response in neuroblastoma.
Fig. 3: Enhancer invasion accounts for tumor-specific MYC/MYCN signatures.
Fig. 4: TWIST1 co-occupies enhancers with MYCN and is required for expression of the MYCN enhancer axis.

Similar content being viewed by others

References

  1. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Nesbit, C. E., Tersak, J. M. & Prochownik, E. V. MYC oncogenes and human neoplastic disease. Oncogene 18, 3004–3016 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Matthay, K. K., George, R. E. & Yu, A. L. Promising therapeutic targets in neuroblastoma. Clin. Cancer Res. 18, 2740–2753 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Seeger, R. C. et al. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N. Engl. J. Med. 313, 1111–1116 (1985).

    Article  CAS  PubMed  Google Scholar 

  5. Weiss, W. A., Aldape, K., Mohapatra, G., Feuerstein, B. G. & Bishop, J. M. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 16, 2985–2995 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wakamatsu, Y., Watanabe, Y., Nakamura, H. & Kondoh, H. Regulation of the neural crest cell fate by N-myc: promotion of ventral migration and neuronal differentiation. Development 124, 1953–1962 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Kang, J. H. et al. MYCN silencing induces differentiation and apoptosis in human neuroblastoma cells. Biochem. Biophys. Res. Commun. 351, 192–197 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tweddle, D. A., Malcolm, A. J., Cole, M., Pearson, A. D. & Lunec, J. p53 cellular localization and function in neuroblastoma: evidence for defective G1 arrest despite WAF1 induction in MYCN-amplified cells. Am. J. Pathol. 158, 2067–2077 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Muth, D. et al. Transcriptional repression of SKP2 is impaired in MYCN-amplified neuroblastoma. Cancer Res. 70, 3791–3802 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Bell, E., Lunec, J. & Tweddle, D. A. Cell cycle regulation targets of MYCN identified by gene expression microarrays. Cell Cycle 6, 1249–1256 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Yaari, S. et al. Disruption of cooperation between Ras and MycN in human neuroblastoma cells promotes growth arrest. Clin. Cancer Res. 11, 4321–4330 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Tonelli, R. et al. Anti-gene peptide nucleic acid specifically inhibits MYCN expression in human neuroblastoma cells leading to cell growth inhibition and apoptosis. Mol. Cancer Ther. 4, 779–786 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Negroni, A. et al. Decrease of proliferation rate and induction of differentiation by a MYCN antisense DNA oligomer in a human neuroblastoma cell line. Cell Growth Diff. 2, 511–518 (1991).

    CAS  PubMed  Google Scholar 

  14. Burkhart, C. A. et al. Effects of MYCN antisense oligonucleotide administration on tumorigenesis in a murine model of neuroblastoma. J. Natl. Cancer Inst. 95, 1394–1403 (2003).

    Article  PubMed  Google Scholar 

  15. Gustafson, W. C. et al. Drugging MYCN through an allosteric transition in Aurora kinase A. Cancer Cell 26, 414–427 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Puissant, A. et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov. 3, 308–323 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nie, Z. et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell 151, 68–79 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lin, C. Y. et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell 151, 56–67 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Walz, S. et al. Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature 511, 483–487 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sabò, A. et al. Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature 511, 488–492 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Guccione, E. et al. Myc-binding-site recognition in the human genome is determined by chromatin context. Nat. Cell Biol. 8, 764–770 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Wolf, E., Lin, C. Y., Eilers, M. & Levens, D. L. Taming of the beast: shaping Myc-dependent amplification. Trends Cell Biol. 25, 241–248 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Lutz, W. et al. Conditional expression of N-myc in human neuroblastoma cells increases expression of α-prothymosin and ornithine decarboxylase and accelerates progression into S-phase early after mitogenic stimulation of quiescent cells. Oncogene 13, 803–812 (1996).

    CAS  PubMed  Google Scholar 

  25. Lorenzin, F. et al. Different promoter affinities account for specificity in MYC-dependent gene regulation. eLife 5, e15161 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Orlando, D. A. et al. Quantitative ChIP–Seq normalization reveals global modulation of the epigenome. Cell Rep. 9, 1163–1170 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Kieffer-Kwon, K. R. et al. Myc regulates chromatin decompaction and nuclear architecture during B cell activation. Mol. Cell 67, 566–578 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Powers, J. T. et al. Multiple mechanisms disrupt the let-7 microRNA family in neuroblastoma. Nature 535, 246–251 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Modak, S. & Cheung, N. K. Neuroblastoma: therapeutic strategies for a clinical enigma. Cancer Treat. Rev. 36, 307–317 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Maniatis, T., Goodbourn, S. & Fischer, J. A. Regulation of inducible and tissue-specific gene expression. Science 236, 1237–1245 (1987).

    Article  CAS  PubMed  Google Scholar 

  31. Visel, A. et al. ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457, 854–858 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Shaffer, A. L. et al. IRF4 addiction in multiple myeloma. Nature 454, 226–231 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dang, C. V. et al. The c-Myc target gene network. Semin. Cancer Biol. 16, 253–264 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Ji, H. et al. Cell-type independent MYC target genes reveal a primordial signature involved in biomass accumulation. PLoS One 6, e26057 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kim, Y. H. et al. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene 25, 130–138 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Schlosser, I. et al. Dissection of transcriptional programmes in response to serum and c-Myc in a human B-cell line. Oncogene 24, 520–524 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Schuhmacher, M. et al. The transcriptional program of a human B cell line in response to Myc. Nucleic Acids Res. 29, 397–406 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zeller, K. I., Jegga, A. G., Aronow, B. J., O’Donnell, K. A. & Dang, C. V. An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol. 4, R69 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Matthay, K. K. et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. N. Engl. J. Med. 341, 1165–1173 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Wong, M. P. et al. Chromosomal aberrations of primary lung adenocarcinomas in nonsmokers. Cancer 97, 1263–1270 (2003).

    Article  PubMed  Google Scholar 

  41. Aggarwal, R., Ghobrial, I. M. & Roodman, G. D. Chemokines in multiple myeloma. Exp. Hematol. 34, 1289–1295 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pellat-Deceunynck, C. et al. Expression of CD28 and CD40 in human myeloma cells: a comparative study with normal plasma cells. Blood 84, 2597–2603 (1994).

    Article  CAS  PubMed  Google Scholar 

  43. Tong, A. W. et al. CD40 ligand-induced apoptosis is Fas-independent in human multiple myeloma cells. Leuk. Lymphoma 36, 543–558 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Westendorf, J. J. et al. CD40 expression in malignant plasma cells. Role in stimulation of autocrine IL-6 secretion by a human myeloma cell line. J. Immunol. 152, 117–128 (1994).

    CAS  PubMed  Google Scholar 

  45. Staege, M. S. et al. MYC overexpression imposes a nonimmunogenic phenotype on Epstein–Barr virus–infected B cells. Proc. Natl Acad. Sci. USA 99, 4550–4555 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Masui, K. et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 18, 726–739 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Suvà, M. L. et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res. 69, 9211–9218 (2009).

    Article  PubMed  CAS  Google Scholar 

  48. Wang, C. et al. EZH2 mediates epigenetic silencing of neuroblastoma suppressor genes CASZ1, CLU, RUNX3, and NGFR. Cancer Res. 72, 315–324 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. 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 

  51. Lin, C. Y. et al. Active medulloblastoma enhancers reveal subgroup-specific cellular origins. Nature 530, 57–62 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Saint-André, V. et al. Models of human core transcriptional regulatory circuitries. Genome Res. 26, 385–396 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Geerts, D., Schilderink, N., Jorritsma, G. & Versteeg, R. The role of the MEIS homeobox genes in neuroblastoma. Cancer Lett. 197, 87–92 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Dowen, J. M. et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Reiff, T. et al. Neuroblastoma Phox2b variants stimulate proliferation and dedifferentiation of immature sympathetic neurons. J. Neurosci. 30, 905–915 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Boeva, V. et al. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat. Genet. 49, 1408–1413 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. van Groningen, T. et al. Neuroblastoma is composed of two super-enhancer-associated differentiation states. Nat. Genet. 49, 1261–1266 (2017).

    Article  PubMed  CAS  Google Scholar 

  58. Entz‐Werlé, N. et al. Frequent genomic abnormalities at TWIST in human pediatric osteosarcomas. Int. J. Cancer 117, 349–355 (2005).

    Article  PubMed  CAS  Google Scholar 

  59. Kwok, W. K. et al. Up-regulation of TWIST in prostate cancer and its implication as a therapeutic target. Cancer Res. 65, 5153–5162 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Kyo, S. et al. High Twist expression is involved in infiltrative endometrial cancer and affects patient survival. Hum. Pathol. 37, 431–438 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Valsesia-Wittmann, S. et al. Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell 6, 625–630 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Littlewood, T. D., Kreuzaler, P. & Evan, G. I. All things to all people. Cell 151, 11–13 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Workman, P. et al. Guidelines for the welfare and use of animals in cancer research. Br. J. Cancer 102, 1555–1577 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oberthuer, A. et al. Comparison of performance of one-color and two-color gene-expression analyses in predicting clinical endpoints of neuroblastoma patients. Pharmacogenomics J. 10, 258–266 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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  Google Scholar 

  66. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

Download references

Acknowledgements

We thank Z. Herbert for his expertise and guidance with next-generation sequencing. We thank R. Young and D. Hnisz for assistance with reagents. We thank P. Rahl, C. Ott, and J. Perry for helpful comments on the manuscript. C.Y.L. is supported by the Cancer Prevention Research Institute of Texas (RR150093) and by the NCI (1R01CA215452-01), and is a Pew-Stewart Scholar for Cancer Research (Alexander and Margaret Stewart Trust). R.Z. and J.E.B. are supported by the V Foundation for Cancer Research Translational Grant.

Author information

Authors and Affiliations

Authors

Contributions

R.Z., C.Y.L., and J.E.B. designed this study. R.Z. designed and performed biology experiments. C.Y.L., J.M.R., D.R.P., and R.A.H. performed data analysis. M.A. Lawlor assisted in ChIP–seq experiments. E.P. performed mouse experiments. M.F. and M.A. Lopez performed experiments in multiple myeloma. T.G.S., Z.J., and K.L.K. assisted in cellular assays. B.N. assisted in RNA-seq profiling. M.A.E. and G.E.W. assisted in exon-scanning CRISPR–Cas9. N.P.M., T.F.W., and L.C. supervised experiments. R.Z., C.Y.L., and J.E.B. analyzed results and wrote the manuscript with comments from all authors.

Corresponding authors

Correspondence to Charles Y. Lin or James E. Bradner.

Ethics declarations

Competing interests

R.Z. is now an employee of C4 Therapeutics. C.Y.L. is a consultant of Jnana Therapeutics and is a shareholder and inventor of intellectual property licensed to Syros Pharmaceuticals. J.E.B. is a Scientific Founder of Syros Pharmaceuticals, SHAPE Pharmaceuticals, Acetylon Pharmaceuticals, Tensha Therapeutics (now Roche), and C4 Therapeutics and is the inventor on intellectual property licensed to these entities. J.E.B. is now an executive and shareholder in Novartis AG.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Deregulated MYCN binding at active promoters and enhancers in neuroblastoma.

a, Cell-normalized steady-state mRNA transcript levels of MYCN in human neuroblastoma cell lines. b, Line plots of quantified western blot bands from increasing cell-number-normalized cropped western blots of MYCN protein levels. Error bars denote ±s.d. for four replicate blots. c, Scatterplots of average ranked MYCN occupancy across four cell lines (x axis) versus ranked MYCN occupancy in each respective cell line (y axis). Contour lines illustrate the density of correlation of MYCN occupancy and are color-coded from high density (red) to low density (yellow). d, Meta track representation of MYCN and H3K27ac ChIP–seq signal (RPM/bp) across four neuroblastoma cell lines at the RPL22 locus. e, Meta track representation of MYCN and H3K27ac ChIP–seq signal (RPM/bp) across four neuroblastoma cell lines at an upstream ID2 enhancer. f, Heatmap of MYCN (red) signal at promoters (left) and enhancers (right) in each respective neuroblastoma cell line. Each row shows the ±5-kb region centered on the TSS ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. g, Heatmap of H3K27ac (blue) signal at promoters (left) and enhancers (right) in each respective neuroblastoma cell line. Each row shows the ±5-kb region centered on the enhancer ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. h, Clustering of the indicated ChIP–seq signal at promoters in the BE(2)-C cell line. Color scaled intensities reflect pairwise similarity via Pearson correlation. i, Top, MYCN signal contribution in the BE(2)-C cell line at the top 20,000 promoters. Bottom, line plots of signal contribution at the top 20,000 promoters of the indicated marks. Error bars represent 95% confidence intervals of the mean. j, Line plots showing the correlation of ranked MYCN-enriched regions (x axis) versus average ChIP–seq signal (top), percentage overlap with a given genomic feature (middle), or E-box density (bottom) on the y axis. Error bars represent 95% confidence intervals of the mean.

Supplementary Figure 2 Dynamic chromatin consequences of direct MYCN shutdown.

a, ChIP-Rx signal (RPM/bp, before scaling) upon MYCN shutdown in the tet-off MYCN SHEP-21N cell line at the NPM1 locus. b, ChIP-Rx signal (RPM/bp) upon MYCN shutdown in the tet-off MYCN SHEP-21 cell line at the NPM1 locus. c, Heatmap and meta plot of MYCN signal at promoters (left) and enhancers (right) upon MYCN shutdown in the tet-off MYCN SHEP-21N cell line. Each row shows the ±5-kb region centered on the TSS or enhancer ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. d, Heatmap and meta plot of H3K27ac signal at promoters and enhancers upon MYCN shutdown in the tet-off MYCN SHEP-21N cell line. Each row shows the ±5-kb region centered on the TSS or enhancer ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. e, Box plots of MYCN and H3K27ac ChIP–seq signal at active promoters. MYCN and H3K27ac signal at 0, 2, and 24 h after MYCN shutdown for standard ChIP–seq; ChIP-Rx before/after scaling. Significant differences are denoted (Welch’s two-tailed t test): ***P < 1 × 10–9, **P < 1 × 10–6. f, Box plots of MYCN and H3K27ac ChIP–seq signal at active enhancers. MYCN and H3K27ac signal at 0, 2, and 24 h after MYCN shutdown for standard ChIP–seq; ChIP-Rx before/after scaling. Significant differences are denoted (Welch’s two-tailed t test): ***P < 1 × 10–9, **P < 1 × 10–6. g, Heatmap and meta plot of H3K4me3, RNA Pol II, and CTCF signal at promoters upon MYCN shutdown in the tet-off MYCN SHEP-21N cell line. Each row shows the ±5-kb region centered on the TSS ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. h, Cropped western blot of total histone H3 and H3K27ac levels at 0, 2, and 24 h after MYCN shutdown. The percentage of H3K27ac remaining versus 0 h is indicated. i, Distribution plots of RNA Pol II traveling ratios (TR) for all active genes in traditional normalized ChIP–seq data. Differences in the TR distribution at 0 h versus 2 h and 0 h versus 24 h are significant; Welch’s two-tailed t test: ***P < 1 × 10–9, **P < 1 × 10–6. j, Box plots of log2 fold changes in active gene expression (traditional normalization) at the indicated time points versus 0 h after MYCN shutdown. Significant differences are denoted (Welch’s two-tailed t test): **P < 1 × 10–6.

Supplementary Figure 3 Replicative analysis of the SHEP-21N system and the dynamics of the tet-on MYCN SHEP system.

a, Left, differential analysis volcano plot of two ChIP–seq replicates comparing the log2 fold change at 2 h versus log10 P value. Blue and red circles denote distal and TSS MYCN sites, respectively. Right, box plot representation of differential regions (P < 0.1, fold change > log2 0.5) showing the log2 fold change at MYCN TSSs and distal sites. b, Left, differential analysis volcano plot of two ChIP–seq replicates comparing the log2 fold change at 24 h versus log10 P value. Blue and red circles denote distal and TSS MYCN sites, respectively. Right, box plot representation of differential regions (P < 0.1, fold change > log2 0.5) showing the log2 fold change at MYCN TSSs and distal sites. c, Scatter of MYCN peak AUC between MYCN ChIP–seq replicates at 0, 2, and 24 h after MYCN shutdown. Pearson correlation is noted. d, Box plots of the log2 fold change of MYCN load at the TSSs and distal enhancers of the top 5,000 genes ranked by proximal MYCN signal. Left, ChIP-Rx before scaling. Right, ChIP-Rx after scaling. e, Heatmap and meta plot of MYCN signal at promoters (left) and enhancers (right) upon MYCN induction in the tet-on MYCN SHEP cell line. Each row shows the ±5-kb region centered on the TSS or enhancer ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. f, Heatmap and meta plot of H3K27ac signal at promoters (left) and enhancers (right) upon MYCN induction in the tet-on MYCN SHEP cell line. Each row shows the ±5-kb region centered on the TSS or enhancer ranked by average H3K27ac signal. Color scaled intensities are in units of RPM/bp. g, Box plots of MYCN (red) and H3K27ac (blue) ChIP–seq signal (RPM/bp) at active promoters and enhancers at 0, 2, and 6 h after MYNC induction. Significant differences are denoted (Welch’s two-tailed t test): ***P < 1 × 10–9.

Supplementary Figure 4 Deregulated MYCN binding at promoters and enhancers in the TH-MYCN genetically engineered neuroblastoma mouse model.

a, Immunohistochemistry staining of MYCN and a corresponding no-stain control in celiac ganglia, TH-MYCN tumors, and spleen. b, Meta track representation of H3K27ac ChIP–seq signal (RPM/bp) in ganglia (celiac and superior cervical) and TH-MYCN tumors as well as MYCN ChIP–seq signal in TH-MYCN tumors at the indicated loci. c, Meta track representation of H3K27ac ChIP–seq signal (RPM/bp) in ganglia (celiac and superior cervical) and TH-MYCN tumors as well as MYCN ChIP–seq signal in TH-MYCN tumors at an upstream Id2 enhancer. d, Meta track representation of H3K27ac ChIP–seq signal (RPM/bp) in ganglia (celiac and superior cervical) and TH-MYCN tumors as well as MYCN ChIP–seq signal in TH-MYCN tumors at the indicated loci. e, Meta track representation of MYCN and H3K27ac ChIP–seq signal (RPM/bp) across four neuroblastoma cell lines at the indicated loci. f, Pie chart showing the percentage of MYCN binding sites in human neuroblastoma cell lines that exhibit H3K27ac signal in TH-MYCN tumors and/or ganglia at promoters (left) and enhancers (right).

Supplementary Figure 5 Enhancer invasion shapes transcriptional sensitivity to MYCN perturbation in neuroblastoma.

a, Top, plot showing the top 5,000 genes in SHEP-21N ranked by total proximal MYCN signal. Bottom, dot plot of the percentage of enhancer contribution, the size of contributing MYCN binding, the density of contributing MYCN, and the area under the curve of contributing MYCN binding with a best fit line superimposed (loess correlation). b, Top, plot showing the top 5,000 genes across four neuroblastoma cell lines ranked by total proximal MYCN signal. Bottom, dot plot of the percentage of enhancer contribution, the size of contributing MYCN binding, the density of contributing MYCN, and the area under the curve of contributing MYCN binding with a best fit line superimposed (loess correlation). c, Standard (left) and cell count (right) normalized levels of the RPL22, HAND2, and ID2 transcripts during MYCN shutdown. Units are FPKM and cell-count-normalized FPKM, respectively, for triplicate biological replicates. Error bars represent s.d. d, Standard normalized log2 fold change (versus 0 h) of gene expression changes during MYCN shutdown in the SHEP-21N system. Genes are grouped according to rank-ordered MYCN proximal load (promoters and enhancers). Error bars represent the 95% confidence interval (CI) of the mean. e, Box plots of the log2 fold change (versus 0 h) of the amount of RNA Pol II (ChIP-Rx, before scaling) at the TSS (left) and gene body (right) of genes grouped according to rank-ordered MYCN proximal load. Significance is denoted by Welch’s two-tailed t test: ***P < 1 × 10–9, **P < 1 × 10–6, *P < 1 × 10–3. f, Box plots of the log2 fold change (versus 0 h) of the amount of RNA Pol II (ChIP–seq) at the TSS (left) and gene body (right) of genes grouped according to rank-ordered MYCN proximal load. Significance is denoted by Welch’s two-tailed t test: ***P < 1 × 10–9, **P < 1 × 10–6, *P < 1 × 10–3.

Supplementary Figure 6 BET bromodomain inhibition in MYCN-driven neuroblastoma.

a, Cropped western blot of vinculin and MYCN levels at 0, 1, 2, 4, 8, and 24 h of treatment with the BET inhibitor JQ1 (1 μM). The percentage of MYCN remaining versus 0 h is indicated. b, Box plots of the log2 mRNA fold change (versus 0 h) at active genes (defined by H3K27ac ChIP–seq signal at TSSs and expressed) with JQ1 (1 μM). Cell-normalized and traditional normalized log2 mRNA fold change levels are shown. Significance is denoted by Welch’s two-tailed t test: ***P < 1 × 10–9, **P < 1 × 10–6, *P < 1 × 10–3. c, Cell-normalized (left) and standard normalized (right) levels of the RPL22, HAND2, and ID2 transcripts during MYCN shutdown. Units are cell FPKM for triplicate biological replicates. Error bars represent s.d. d, Cell-normalized and standard normalized log2 fold change of gene expression changes ranked by total MYCN load during JQ1 treatment. Genes are grouped according to rank-ordered MYCN proximal load (promoters and enhancers). Error bars represent the 95% confidence interval (CI) of the mean. e, log2 fold change of gene expression changes of the top 5,000 genes ranked by total MYCN load during JQ1 treatment. Genes are grouped according to rank-ordered MYCN proximal load at promoters (left) or enhancers (right). Error bars represent the 95% confidence interval (CI) of the mean.

Supplementary Figure 7 Enhancer invasion accounts for tumor-specific MYCN signatures in neuroblastoma.

a, Differential MYCN signal contribution in the BE(2)-C cell line for promoters (red) and enhancers (blue) of associated genes (y axis) of the top 5,000 proximal MYCN-bound regions are shown ranked by difference in MYCN enhancer to promoter contribution (x axis). b, GSEA plots of MYCN-bound promoter- (red) versus enhancer- (blue) dominant gene sets defined by leading edge analysis. c, Normalized enrichment scores (NESs) of target gene signatures (Molecular Signature Database) are plotted on the x axis versus the FDR (false discovery rate) on the y axis. d, Highly significant gene signatures from promoter (red) and enhancer (blue) bias gene sets are highlighted and tabulated. e, Differential H3K27ac signal contribution in the BE(2)-C cell line for promoters (red) and enhancers (blue) of associated genes (y axis) of the top 5,000 proximal H3K27ac-bound regions are shown ranked by difference in H3K27ac enhancer to promoter contribution (x axis). f, GSEA plots of H3K27ac-bound promoter- (red) versus enhancer- (blue) dominant gene sets defined by leading edge analysis. g, Normalized enrichment scores (NESs) of target gene signatures (Molecular Signature Database) are plotted on the x axis versus the FDR (false discovery rate) on the y axis. h, Highly significant gene signatures from promoter (red) and enhancer (blue) bias gene sets are highlighted and tabulated. i, Overall survival of patients ranked by expression of the top 25 genes defined by total MYCN load at promoter-dominant genes (left) or enhancer-dominant genes (right) for all patients with neuroblastoma. Significance is denoted by a chi-squared test, and P values are shown. j, Overall survival of patients ranked by expression of the top 25 genes defined by total MYCN load at promoter-dominant genes (left) or enhancer-dominant genes (right) for patients with neuroblastoma without MYCN amplification. Significance is denoted by a chi-squared test, and P values are shown. k, Heatmap of gene signatures enriched in promoter- (red) or enhancer- (blue) dominant pathways as defined by MYCN or H3K27ac in the BE(2)-C cell line. Selected signatures are annotated at an FDR < 0.01 and NES > 2 cutoff. l, Normalized enrichment scores (NESs) of target gene signatures (Molecular Signature Database) are plotted on the x axis versus the FDR (false discovery rate) on the y axis at 0, 2, and 6 h after MYCN induction.

Supplementary Figure 8 MYC-bound enhancer axes reflect tumor-specific pathways in deregulated-MYC-driven cancers.

a, Top, plot showing the top 5,000 genes (x axis) in SHEP-21N ranked by total proximal MYCN signal. Bottom, dot plot of the percentage of enhancer contribution (enhancer/total MYC signal) sampled across bins (100 genes/bin) with a best fit line superimposed (loess correlation). b, Differential MYC signal contribution in the H2171 cell line for promoters (red) and enhancers (blue) of associated genes (y axis) of the top 5,000 proximal MYC-bound regions are shown ranked by difference in MYC enhancer to promoter contribution (x axis). c, GSEA plots of MYC-bound promoter- (red) versus enhancer- (blue) dominant gene sets defined by leading edge analysis. d, Normalized enrichment scores (NESs) of target gene signatures (Molecular Signature Database) are plotted on the x axis versus the FDR (false discovery rate) on the y axis. e, Highly significant gene signatures from promoter (red) and enhancer (blue) bias gene sets are highlighted and tabulated. fi, Same as in ae (respectively) in the U87 cell line. ko, Same as in ae (respectively) in the MM1.S cell line. pt, Same as in ae (respectively) in the P493-6 cell line.

Supplementary Figure 9 TWIST1 co-occupies enhancers with MYCN and is a MYCN-specific dependency in neuroblastoma.

a, Schematic of the computational approach to identifying a core regulatory circuit based on TF enhancer binding by identifying transcription factor motifs within ATAC–seq-defined nucleosome-free regions. The ‘in’ and ‘out’ degrees of transcription factor binding are defined. b, Network depiction of the conserved enhancer regulatory transcription factor network in the indicated neuroblastoma cell lines. TF nodes are denoted, and predicted binding interactions with super-enhancers driving other TFs are shown as lines (edges). Enhancer-regulated bHLH TFs are highlighted in orange. c, Heatmap of enhancer-regulated TFs in neuroblastoma cell lines (rows) clustered by similarity of regulatory degree. The bHLH TFs HAND2 and TWIST1 are highlighted in orange. d, Network depiction of the conserved enhancer regulatory transcription factor network across the four neuroblastoma cell lines. TF nodes are denoted, and predicted binding interactions with super-enhancers driving other TFs are shown as lines (edges). e, Top, waterfall plot of log2 MYCN fold change at 2 versus 0 h after MYCN shutdown. Bottom, line plot of log2 TWIST1 fold change at 2 h after MYCN shutdown. Regions ranked by change in MYCN levels 2 h after MYCN shutdown. f, CRISPR scan of the NSD1 locus. Top, Illumina sequencing readout of log2 fold enrichment/depletion (early versus late time point) of 3,351 sgRNAs. Bottom, simple moving average of log2 fold enrichment/depletion is shown. g, ChIP–seq signal of H3K27ac (blue), ATAC–seq (green), MYCN (red), and TWIST1 (orange) at the NSD1 locus with respect to CRISPR sgRNAs. h, Left, cropped western blot of TWIST1, MYCN, and vinculin protein levels upon siRNA-mediated knockdown of TWIST1 in the indicated cell lines. Right, corresponding viable cell counts 72 h after siRNA transfection. Error bars denote ±s.d. i, Left, cropped western blot of TWIST1, MYCN, and vinculin protein levels upon siRNA-mediated knockdown of TWIST1 in the MYCN ‘on’ and MYCN ‘off’ states in the SHEP21-N cell line. Right, corresponding viable cell counts at 72 h after siRNA transfection. Error bars denote ±s.d. j, log2 fold change of gene expression changes upon MYCN shutdown of genes ranked by MYCN promoter load (top) or MYCN distal enhancer load (bottom). Genes are grouped according to rank-ordered MYCN proximal load (promoters and enhancers). Error bars represent the 95% CI.

Supplementary Figure 10 TCF3 is a TF dependency in IgH/MYC-translocated multiple myeloma.

a, Network depiction of the conserved enhancer regulatory transcription factor network in the MM1.S cell line. TF nodes are denoted, and predicted binding interactions with super-enhancers driving other TFs are shown as lines (edges). Enhancer-regulated bHLH TFs are highlighted in orange. b, Histogram plot ranking core regulatory circuit TFs based on motif co-occupancy with MYC. c, Left axis, quantified mRNA levels of TCF3 upon shRNA-mediated knockdown in the MM.1S cell line. Right axis, corresponding viability defect observed upon TCF3 knockdown as measured by ATP levels. Shown is the average of three experiments performed in triplicate. d, Line plot of 3H thymidine uptake at days 1 and 3 after shRNA cell lines expressing selection for each respective shRNA in the MM.1S cell line. Thymidine uptake is shown normalized to day 1 levels. e, Bar plot of the fraction of live shRNA-expressing cells as measured by GFP positivity. Data are represented as fold change as compared to day 1 after infection. f, Percentage of early (Annexin V) and late (Annexin V and DAPI) apoptotic and live (unstained) cells four days after shRNA infection in the MM.1S cell line. The MM1.S cell line (left) and U266 cell line (right) are shown. gj, Same as in cf (respectively) in the U266 cell line.

Supplementary Figure 11 Full scan (uncropped) western blots from main figures

Uncropped full scans of western blots from the corresponding cropped western blots shown within the main text. Molecular weight markers are indicated. Figure subpanel is indicated for each respective blot.

Supplementary Figure 12 Full scan (uncropped) western blots from supplementary figures

Uncropped full scans of western blots from the corresponding cropped western blots shown within the supplementary figures. Molecular weight markers are indicated. Figure subpanel is indicated for each respective blot.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Note.

Life Sciences Reporting Summary

Supplementary Table 1

Quantified levels of MYCN in neuroblastoma cell lines.

Supplementary Table 2

Overall patient survival stratification.

Supplementary Table 3

CRISPR scan of TWIST1 and inducible knockdown of TWIST1.

Supplementary Table 4

CRISPR scan of NSD1 and siRNA-mediated knockdown of TWIST1.

Supplementary Table 5

Characterization of TCF3 in multiple myeloma.

Supplementary Table 6

Microarray sample detail summary.

Supplementary Table 7

ChIP–seq, ChIP-Rx, and ATAC–seq sample detail summary.

Supplementary Table 8

RNA-seq sample detail summary.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zeid, R., Lawlor, M.A., Poon, E. et al. Enhancer invasion shapes MYCN-dependent transcriptional amplification in neuroblastoma. Nat Genet 50, 515–523 (2018). https://doi.org/10.1038/s41588-018-0044-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0044-9

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer