Nature - 2019.08.29

(Frankie) #1

RESEARCH LETTER


BRD4, which rendered the resistant cells more sensitive to BET inhi-
bition (Extended Data Fig. 9; Supplementary Note 2). Together, these
results indicate the establishment of an alternative transcription factor
regulatory network controlled by BORIS-induced chromatin remod-
elling to support the resistant cell state.
Thus, using a pair of isogenic ALK-inhibitor sensitive and resistant
neuroblastoma cell lines, we show that the CTCF paralogue BORIS
can promote regulatory DNA interactions that support a phenotypic
switch in the context of treatment resistance (Fig. 4e). This mechanism
appears relevant to different neuroblastoma cell lines and kinase inhib-
itors and may extend to other cancers. In Ewing sarcoma, in which
overexpression of BORIS is associated with metastasis and relapse
(Extended Data Fig. 1c), we observed increased BORIS occupancy at
regulatory regions in chemotherapy-resistant cell lines (Extended Data
Fig. 10; Supplementary Note 3). Further work will establish whether
BORIS-mediated alteration of chromatin looping is a general mecha-
nism by which tumour cells co-opt developmental networks to sustain
alternative cell states in response to targeted or conventional therapies.

Online content
Any methods, additional references, Nature Research reporting summaries,
source data, extended data, supplementary information, acknowledgements,
peer review information; details of author contributions and competing
interests; and statements of data and code availability are available at https://doi.org/
10.1038/s41586-019-1472-0.

Received: 21 July 2017; Accepted: 11 July 2019;
Published online 7 August 2019.


  1. Dixon, J. R. et al. Topological domains in mammalian genomes identified by
    analysis of chromatin interactions. Nature 485 , 376–380 (2012).

  2. Phillips-Cremins, J. E. et al. Architectural protein subclasses shape 3D
    organization of genomes during lineage commitment. Cell 153 , 1281–1295
    (2013).

  3. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome
    neighborhoods. Science 351 , 1454–1458 (2016).

  4. Katainen, R. et al. CTCF/cohesin-binding sites are frequently mutated in cancer.
    Nat. Genet. 47 , 818–821 (2015).

  5. Flavahan, W. A. et al. Insulator dysfunction and oncogene activation in IDH
    mutant gliomas. Nature 529 , 110–114 (2016).

  6. Loukinov, D. I. et al. BORIS, a novel male germ-line-specific protein associated
    with epigenetic reprogramming events, shares the same 11-zinc-finger domain
    with CTCF, the insulator protein involved in reading imprinting marks in the
    soma. Proc. Natl Acad. Sci. USA 99 , 6806–6811 (2002).

  7. Klenova, E. M., Morse, H. C. III, Ohlsson, R. & Lobanenkov, V. V. The novel BORIS

    • CTCF gene family is uniquely involved in the epigenetics of normal biology
      and cancer. Semin. Cancer Biol. 12 , 399–414 (2002).



  8. Martin-Kleiner, I. BORIS in human cancers — a review. Eur. J. Cancer 48 ,
    929–935 (2012).

  9. Garikapati, K. R. et al. Down-regulation of BORIS/CTCFL efficiently regulates
    cancer stemness and metastasis in MYCN amplified neuroblastoma cell line by


modulating Wnt/β-catenin signaling pathway. Biochem. Biophys. Res. Commun.
484 , 93–99 (2017).


  1. Cheung, N. K. & Dyer, M. A. Neuroblastoma: developmental biology,
    cancer genomics and immunotherapy. Nat. Rev. Cancer 13 , 397–411
    (2013).

  2. Monk, M., Hitchins, M. & Hawes, S. Differential expression of the embryo/cancer
    gene ECSA(DPPA2), the cancer/testis gene BORIS and the pluripotency
    structural gene OCT4, in human preimplantation development. Mol. Hum.
    Reprod. 14 , 347–355 (2008).

  3. Brodeur, G. M., Seeger, R. C., Schwab, M., Varmus, H. E. & Bishop, J. M.
    Amplification of N-myc in untreated human neuroblastomas correlates with
    advanced disease stage. Science 224 , 1121–1124 (1984).

  4. George, R. E. et al. Activating mutations in ALK provide a therapeutic target in
    neuroblastoma. Nature 455 , 975–978 (2008).

  5. Galkin, A. V. et al. Identification of NVP-TAE684, a potent, selective, and
    efficacious inhibitor of NPM-ALK. Proc. Natl Acad. Sci. USA 104 , 270–275
    (2007).

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

  7. Durbin, A. D. et al. Selective gene dependencies in MYCN-amplified
    neuroblastoma include the core transcriptional regulatory circuitry. Nat. Genet.
    50 , 1240–1246 (2018).

  8. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res.
    9 , 2579–2605 (2008).

  9. Zeid, R. et al. Enhancer invasion shapes MYCN-dependent transcriptional
    amplification in neuroblastoma. Nat. Genet. 50 , 515–523 (2018).

  10. Muhar, M. et al. SLAM-seq defines direct gene-regulatory functions of the
    BRD4-MYC axis. Science 360 , 800–805 (2018).

  11. Wegner, M. & Stolt, C. C. From stem cells to neurons and glia: a Soxist’s view of
    neural development. Tr e n d s N e u r o s c i. 28 , 583–588 (2005).

  12. Gao, Y. et al. Overcoming resistance to the THZ series of covalent transcriptional
    CDK inhibitors. Cell Chem. Biol. 25 , 135–142 (2018).

  13. Pugacheva, E. M. et al. Comparative analyses of CTCF and BORIS occupancies
    uncover two distinct classes of CTCF binding genomic regions. Genome Biol. 16 ,
    161 (2015).

  14. Bergmaier, P. et al. Choice of binding sites for CTCFL compared to CTCF is
    driven by chromatin and by sequence preference. Nucleic Acids Res. 46 ,
    7097–7107 (2018).

  15. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed
    genome architecture. Nat. Methods 13 , 919–922 (2016).

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

  17. Sanborn, A. L. et al. Chromatin extrusion explains key features of loop and
    domain formation in wild-type and engineered genomes. Proc. Natl Acad. Sci.
    USA 112 , E6456–E6465 (2015).

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

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

  20. Kumar, J. P. The sine oculis homeobox (SIX) family of transcription factors
    as regulators of development and disease. Cell. Mol. Life Sci. 66 , 565–583
    (2009).

  21. Dennis, D. J., Han, S. & Schuurmans, C. bHLH transcription factors in neural
    development, disease, and reprogramming. Brain Res. 1705 , 48–65 (2019).


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

© The Author(s), under exclusive licence to Springer Nature Limited 2019

680 | NAT URE | VOL 572 | 29 AUGUST 2019

Free download pdf