We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

The epigenetics orchestra of Notch signaling: a symphony for cancer therapy

    Alireza Paniri

    Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745,Iran

    Zoonoses Research Center, Pasteur Institute of Iran, 4619332976, Amol, Iran

    ,
    Mohammad Mahdi Hosseini

    Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745, Iran

    ,
    Fatemeh Amjadi-Moheb

    Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745,Iran

    ,
    Reza Tabaripour

    Department of Cellular and Molecular Biology, Babol Branch, Islamic Azad University, Babol, 4747137381, Iran

    ,
    Elnaz Soleimani

    Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745,Iran

    ,
    Maryam Pilehchian Langroudi

    Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745,Iran

    ,
    Parisa Zafari

    Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, 4691786953, Iran

    &
    Haleh Akhavan-Niaki

    *Author for correspondence: Tel.: +98 911 125 5920;

    E-mail Address: halehakhavan@yahoo.com

    Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, 4717647745,Iran

    Zoonoses Research Center, Pasteur Institute of Iran, 4619332976, Amol, Iran

    Published Online:https://doi.org/10.2217/epi-2023-0270

    The aberrant regulation of the Notch signaling pathway, which is a fundamental developmental pathway, has been implicated in a wide range of human cancers. The Notch pathway can be activated by both canonical and noncanonical Notch ligands, and its role can switch between acting as an oncogene or a tumor suppressor depending on the context. Epigenetic modifications have the potential to modulate Notch and its ligands, thereby influencing Notch signal transduction. Consequently, the utilization of epigenetic regulatory mechanisms may present novel therapeutic opportunities for both single and combined therapeutics targeted at the Notch signaling pathway. This review offers insights into the mechanisms governing the regulation of Notch signaling and explores their therapeutic potential.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Sato C, Zhao G, Ilagan MX. An overview of Notch signaling in adult tissue renewal and maintenance. Curr. Alzheimer Res. 9(2), 227–240 (2012).
    • 2. Andersen P, Uosaki H, Shenje LT, Kwon C. Non-canonical Notch signaling: emerging role and mechanism. Trends Cell Biol. 22(5), 257–265 (2012).
    • 3. Turkoz M, Townsend RR, Kopan R. The Notch intracellular domain has an RBPj-independent role during mouse hair follicular development. J. Invest. Dermatol. 136(6), 1106–1115 (2016).
    • 4. Aster JC, Pear WS, Blacklow SC. The varied roles of Notch in cancer. Annu. Rev. Pathol. 12, 245–275 (2017).
    • 5. Hibdon ES, Razumilava N, Keeley TM et al. Notch and mTOR signaling pathways promote human gastric cancer cell proliferation. Neoplasia 21(7), 702–712 (2019).
    • 6. Ghoshal P, Nganga AJ, Moran-Giuati J et al. Loss of the SMRT/NCoR2 corepressor correlates with JAG2 overexpression in multiple myeloma. Cancer Res. 69(10), 4380–4387 (2009).
    • 7. Jin L, Vu T, Yuan G, Datta PK. STRAP promotes stemness of human colorectal cancer via epigenetic regulation of the NOTCH pathway. Cancer Res. 77(20), 5464–5478 (2017). •• Reveals that the signaling scaffold protein STRAP plays an oncogenic role in maintaining cancer stem-like cell subpopulations in colorectal cancer by antagonizing the formation of the chromatin modifier PRC2. STRAP, NOTCH1 and HES1 axis can be considered for improving treatment strategies.
    • 8. Zhang TH, Liang LZ, Liu XL et al. Long non-coding RNA MALAT1 interacts with miR-124 and modulates tongue cancer growth by targeting JAG1. Oncol. Rep. 37(4), 2087–2094 (2017).
    • 9. Wang L, Long H, Zheng Q, Bo X, Xiao X, Li B. Circular RNA circRHOT1 promotes hepatocellular carcinoma progression by initiation of NR2F6 expression. Mol. Cancer 18(1), 119 (2019).
    • 10. Kuwabara S, Yamaki M, Yu H, Itoh M. Notch signaling regulates the expression of glycolysis-related genes in a context-dependent manner during embryonic development. Biochem. Biophys. Res. Commun. 503(2), 803–808 (2018).
    • 11. Weidenbusch M, Rodler S, Song S et al. Gene expression profiling of the Notch–AhR–IL22 axis at homeostasis and in response to tissue injury. Biosci. Rep. 37(6), 1–14 (2017).
    • 12. Magee CN, Murakami N, Borges TJ et al. Notch-1 inhibition promotes immune regulation in transplantation via regulatory T cell-dependent mechanisms. Circulation 140(10), 846–863 (2019).
    • 13. Ibrahim SA, Gadalla R, El-Ghonaimy EA et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol. Cancer 16(1), 57 (2017).
    • 14. Brahmi M, Bally O, Eberst L, Cassier P. Therapeutic targeting of Notch signaling in cancer. Bull. Cancer 104(10), 883–891 (2017).
    • 15. Kushwah R, Guezguez B, Lee JB, Hopkins CI, Bhatia M. Pleiotropic roles of Notch signaling in normal, malignant, and developmental hematopoiesis in the human. EMBO Rep. 15(11), 1128–1138 (2014).
    • 16. Yan B, Liu L, Zhao Y et al. Xiaotan Sanjie decoction attenuates tumor angiogenesis by manipulating Notch-1-regulated proliferation of gastric cancer stem-like cells. World J. Gastroenterol. 20(36), 13105–13118 (2014).
    • 17. Huang Z, Lin S, Long C et al. Notch signaling pathway mediates doxorubicin-driven apoptosis in cancers. Cancer Manag. Res. 10, 1439–1448 (2018).
    • 18. Bigas A, Espinosa L. Notch signaling in cell–cell communication pathways. Curr. Stem Cell Rep. 2(4), 349–355 (2016).
    • 19. Li Z, Fischer M, Satkunarajah M, Zhou D, Withers SG, Rini JM. Structural basis of Notch O-glucosylation and O-xylosylation by mammalian protein-O-glucosyltransferase 1 (POGLUT1). Nat. Commun. 8(1), 185 (2017).
    • 20. D'souza B, Meloty-Kapella L, Weinmaster G. Canonical and non-canonical Notch ligands. In: Current Topics in Developmental Biology. Elsevier, 73–129 (2010).
    • 21. Arnett KL, Seegar TC, Blacklow SC. Structural biology of Notch signaling. In: Targeting Notch in Cancer. Springer, 1–33 (2018).
    • 22. Bray SJ. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 17(11), 722–735 (2016). • The Notch signaling pathway, known for its high conservation, plays a crucial role in various developmental and homeostatic processes. Despite its direct route from the membrane to the nucleus, which provides fewer opportunities for regulation compared to other signaling pathways, Notch generates intricate patterns in structures such as sensory hair cells and arterial networks. The pathway's activity can paradoxically promote tissue growth and cancers in some situations while inducing cell death and tumor suppression in others. The diverse outcomes are attributed to multiple regulatory mechanisms, including the receptor–ligand landscape, tissue topology, nuclear environment and the connectivity of regulatory networks, which collectively contribute to shaping the Notch pathway's activity based on contextual requirements.
    • 23. Lovendahl KN, Blacklow SC, Gordon WR. The molecular mechanism of Notch activation. In: Molecular Mechanisms of Notch Signaling. Springer, 47–58 (2018). •• In recent years, research has highlighted that Notch activation is intricately regulated by the conformational control of Notch proteolysis, particularly by the extracellular juxtamembrane NRR of Notch. This NRR sterically occludes the S2 protease site until ligand binding occurs. Understanding how the conformational exposure of the protease site is achieved during physiological activation, and how this normal process is bypassed in disease pathogenesis, has been a focus of intense study. This chapter provides a summary of the structural features of the NRR domains of Notch receptors that establish the autoinhibited state. Additionally, it reviews recent studies that employ force spectroscopy and molecular tension sensors to test the mechanotransduction model for Notch signaling.
    • 24. Seegar TCM, Killingsworth LB, Saha N et al. Structural basis for regulated proteolysis by the alpha-secretase ADAM10. Cell 171(7), 1638–1648e7 (2017).
    • 25. Sosa Iglesias V, Theys J, Groot AJ et al. Synergistic effects of NOTCH/gamma-secretase inhibition and standard of care treatment modalities in non-small cell lung cancer cells. Front. Oncol. 8(460), 460 (2018).
    • 26. Jang J, Byun SH, Han D et al. Notch intracellular domain deficiency in nuclear localization activity retains the ability to enhance neural stem cell character and block neurogenesis in mammalian brain development. Stem Cells Dev. 23(23), 2841–2850 (2014).
    • 27. Andersen P, Uosaki H, Shenje LT, Kwon C. Non-canonical Notch signaling: emerging role and mechanism. Trends Cell Biol. 22(5), 257–265 (2012).
    • 28. Krishna BM, Jana S, Singhal J et al. Notch signaling in breast cancer: from pathway analysis to therapy. Cancer Lett. 461, 123–131 (2019).
    • 29. Huang Q, Li J, Zheng J, Wei A. The carcinogenic role of the Notch signaling pathway in the development of hepatocellular carcinoma. J. Cancer 10(6), 1570–1579 (2019).
    • 30. Lim JS, Ibaseta A, Fischer MM et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 545(7654), 360–364 (2017).
    • 31. Hu YY, Zheng MH, Zhang R, Liang YM, Han H. Notch signaling pathway and cancer metastasis. Adv. Exp. Med. Biol. 727, 186–198 (2012).
    • 32. Landor SK, Lendahl U. The interplay between the cellular hypoxic response and Notch signaling. Exp. Cell Res. 356(2), 146–151 (2017).
    • 33. Ayaz F, Osborne BA. Non-canonical notch signaling in cancer and immunity. Front. Oncol. 4, 345 (2014).
    • 34. Osipo C, Golde TE, Osborne BA, Miele LA. Off the beaten pathway: the complex cross talk between Notch and NF-κB. Lab. Invest. 88(1), 11 (2008).
    • 35. Perumalsamy LR, Nagala M, Banerjee P, Sarin A. A hierarchical cascade activated by non-canonical Notch signaling and the mTOR–Rictor complex regulates neglect-induced death in mammalian cells. Cell Death Differ. 16(6), 879–889 (2009).
    • 36. Ang HL, Tergaonkar V. Notch and NFκB signaling pathways: do they collaborate in normal vertebrate brain development and function? BioEssays 29(10), 1039–1047 (2007).
    • 37. Traustadottir GA, Jensen CH, Thomassen M et al. Evidence of non-canonical NOTCH signaling: delta-like 1 homolog (DLK1) directly interacts with the NOTCH1 receptor in mammals. Cell. Signal. 28(4), 246–254 (2016).
    • 38. Wang MM. Notch signaling and Notch signaling modifiers. Int. J. Biochem. Cell Biol. 43(11), 1550–1562 (2011).
    • 39. Du J, Wang X, Zhang X, Zhang X, Jiang H. DNER modulates the length, polarity and synaptogenesis of spiral ganglion neurons via the Notch signaling pathway. Mol. Med. Rep. 17(2), 2357–2365 (2018).
    • 40. Ballester-Lopez C, Conlon TM, Ertuz Z et al. The Notch ligand DNER regulates macrophage IFNγ release in chronic obstructive pulmonary disease. EBioMedicine 43, 562–575 (2019).
    • 41. Pan W, Song XY, Hu QB, Zhang M, Xu XH. TSP2 acts as a suppresser of cell invasion, migration and angiogenesis in medulloblastoma by inhibiting the Notch signaling pathway. Brain Res. 1718, 223–230 (2019).
    • 42. Rauen T, Raffetseder U, Frye BC et al. YB-1 acts as a ligand for Notch-3 receptors and modulates receptor activation. J. Biol. Chem. 284(39), 26928–26940 (2009).
    • 43. Tang H, Xiao WR, Liao YY et al. EGFL7 silencing inactivates the Notch signaling pathway; enhancing cell apoptosis and suppressing cell proliferation in human cutaneous melanoma. Neoplasma 66(2), 187–196 (2019).
    • 44. Gupta R, Hong D, Iborra F, Sarno S, Enver T. NOV (CCN3) functions as a regulator of human hematopoietic stem or progenitor cells. Science 316(5824), 590–593 (2007).
    • 45. Vatanmakanian M, Tavallaie M, Ghadami S. Imatinib independent aberrant methylation of NOV/CCN3 in chronic myelogenous leukemia patients: a mechanism upstream of BCR-ABL1 function? Cell Commun. Signal. 17(1), 38 (2019).
    • 46. Meng H, Zhang X, Hankenson KD, Wang MM. Thrombospondin 2 potentiates notch3/jagged1 signaling. J. Biol. Chem. 284(12), 7866–7874 (2009).
    • 47. Shi J, Li P, Zou L, Chen P, Zhang LP. Extracellular Y-box binding protein-1 promotes proliferation and metastasis of HepG2 cells through Notch3 receptor. Zhonghua Gan Zang Bing Za Zhi 24(3), 202–206 (2016).
    • 48. Lindquist JA, Mertens PR. Cold shock proteins: from cellular mechanisms to pathophysiology and disease. Cell Commun. Signal. 16(1), 63 (2018).
    • 49. Ke B, Fan C, Tu W, Fang X. The role of Y-box binding protein 1 in kidney injury: friend or foe? Cell. Physiol. Biochem. 46(1), 314–321 (2018).
    • 50. Hong G, Kuek V, Shi J et al. EGFL7: master regulator of cancer pathogenesis, angiogenesis and an emerging mediator of bone homeostasis. J. Cell. Physiol. 233(11), 8526–8537 (2018).
    • 51. Palmer WH, Deng WM. Ligand-independent mechanisms of Notch activity. Trends Cell Biol. 25(11), 697–707 (2015).
    • 52. Sakamoto K, Yamaguchi S, Ando R et al. The nephroblastoma overexpressed gene (NOV/ccn3) protein associates with Notch1 extracellular domain and inhibits myoblast differentiation via Notch signaling pathway. J. Biol. Chem. 277(33), 29399–29405 (2002).
    • 53. Thibout H, Martinerie C, Creminon C et al. Characterization of human NOV in biological fluids: an enzyme immunoassay for the quantification of human NOV in sera from patients with diseases of the adrenal gland and of the nervous system. J. Clin. Endocrinol. Metab. 88(1), 327–336 (2003).
    • 54. Miyamoto A, Lau R, Hein PW, Shipley JM, Weinmaster G. Microfibrillar proteins MAGP-1 and MAGP-2 induce Notch1 extracellular domain dissociation and receptor activation. J. Biol. Chem. 281(15), 10089–10097 (2006).
    • 55. Aithal MG, Rajeswari N. Role of Notch signalling pathway in cancer and its association with DNA methylation. J. Genet. 92(3), 667–675 (2013).
    • 56. Wang J, Sullenger BA, Rich JN. Notch signaling in cancer stem cells. (0065–2598 [Print]).
    • 57. Capaccione KM, Pine SR. The Notch signaling pathway as a mediator of tumor survival. (1460–2180 [Electronic]).
    • 58. Xiao W, Gao Z, Duan Y, Yuan W, Ke Y. Notch signaling plays a crucial role in cancer stem-like cells maintaining stemness and mediating chemotaxis in renal cell carcinoma. J. Exp. Clin. Cancer Res. 36(1), 41 (2017).
    • 59. Natsuizaka M, Whelan KA, Kagawa S et al. Interplay between Notch1 and Notch3 promotes EMT and tumor initiation in squamous cell carcinoma. Nat. Commun. 8(1), 1758 (2017).
    • 60. De Francesco EM, Maggiolini M, Musti AM. Crosstalk between Notch, HIF-1α and GPER in breast cancer EMT. Int. J. Mol. Sci. 19(7), 2011 (2018).
    • 61. Pandya Thakkar N, Pereira BMV, Katakia YT et al. Elevated H3K4me3 through MLL2-WDR82 upon hyperglycemia causes jagged ligand dependent notch activation to interplay with differentiation state of endothelial cells. Font. Cell Dev. Biol. 10, 839109 (2022).
    • 62. Gottlieb TM, Leal JF, Seger R, Taya Y, Oren M. Cross-talk between Akt, p53 and Mdm2: possible implications for the regulation of apoptosis. (0950–9232 [Print]).
    • 63. Dotto GP. Crosstalk of Notch with p53 and p63 in cancer growth control. Nat. Rev. Cancer 9(8), 587–595 (2009).
    • 64. Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat. Rev. Cancer 3(10), 756–767 (2003).
    • 65. South AP, Cho RJ, Aster JC. The double-edged sword of Notch signaling in cancer. Semin. Cell Dev. Biol. 23(4), 458–464 (2012).
    • 66. Roy G, Yang T, Liu S, Luo Y-L, Liu Y, Zhong Q. Epigenetic regulation of MAP3K8 in EBV-associated gastric carcinoma. Int. J. Mol. Sci. 24(3), 1964 (2023).
    • 67. Ellisen LW, Bird J, West DC et al. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. (0092–8674 [Print]).
    • 68. Reynolds TC, Smith SD, Sklar J. Analysis of DNA surrounding the breakpoints of chromosomal translocations involving the beta T cell receptor gene in human lymphoblastic neoplasms. (0092–8674 [Print]).
    • 69. Girard L, Hanna Z, Beaulieu N et al. Frequent provirus insertional mutagenesis of Notch1 in thymomas of MMTVD/myc transgenic mice suggests a collaboration of c-myc and Notch1 for oncogenesis. Genes Dev. 10(15), 1930–1944 (1996).
    • 70. Tohda S. NOTCH signaling roles in acute myeloid leukemia cell growth and interaction with other stemness-related signals. Anticancer Res. 34(11), 6259–6264 (2014).
    • 71. Gallahan D, Kozak C, Callahan R. A new common integration region (int-3) for mouse mammary tumor virus on mouse chromosome 17. J. Virol. 61(1), 218–220 (1987).
    • 72. Reedijk M, Odorcic S, Chang L et al. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res. 65(18), 8530 (2005).
    • 73. Zagouras P, Stifani S, Blaumueller CM, Carcangiu ML, Artavanis-Tsakonas S. Alterations in Notch signaling in neoplastic lesions of the human cervix. Proc. Natl Acad. Sci. USA 92(14), 6414–6418 (1995).
    • 74. Zhang Y, Li B, Ji ZZ, Zheng PS. Notch1 regulates the growth of human colon cancers. Cancer 116(22), 5207–5218 (2010).
    • 75. Westhoff B, Colaluca IN, D'Ario G et al. Alterations of the Notch pathway in lung cancer. Proc. Natl Acad. Sci. USA 106(52), 22293–22298 (2009).
    • 76. Klinakis A, Lobry C, Abdel-Wahab O et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature 473(7346), 230–233 (2011).
    • 77. Lowell S, Watt FM. Delta regulates keratinocyte spreading and motility independently of differentiation. Mech. Dev. 107(1–2), 133–140 (2001).
    • 78. Rampias T, Vgenopoulou P, Avgeris M et al. A new tumor suppressor role for the Notch pathway in bladder cancer. Nat. Med. 20(10), 1199–1205 (2014).
    • 79. Zhu B, Sun L, Luo W et al. Activated Notch signaling augments cell growth in hepatocellular carcinoma via up-regulating the nuclear receptor NR4A2. Oncotarget 8(14), 23289–23302 (2017).
    • 80. Thélu J, Rossio P, Favier B. Notch signalling is linked to epidermal cell differentiation level in basal cell carcinoma, psoriasis and wound healing. BMC Dermatol. 2, 7–7 (2002).
    • 81. Lo Muzio L, Pannone G, Staibano S et al. WNT-1 expression in basal cell carcinoma of head and neck. An immunohistochemical and confocal study with regard to the intracellular distribution of beta-catenin. Anticancer Res. 22(2A), 565–576 (2002).
    • 82. Sriuranpong V, Borges MW, Ravi RK et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res. 61(7), 3200–3205 (2001).
    • 83. Zhang Y, Li D, Feng F et al. Progressive and prognosis value of Notch receptors and ligands in hepatocellular carcinoma: a systematic review and meta-analysis. Sci. Rep. 7(1), 14809 (2017).
    • 84. Wang M, Xue L, Cao Q, Lin Y et al. Expression of Notch1, Jagged1 and beta-catenin and their clinicopathological significance in hepatocellular carcinoma. (0028–2685 [Print]).
    • 85. Sun L, Liu M, Sun GC et al. Notch signaling activation in cervical cancer cells induces cell growth arrest with the involvement of the nuclear receptor NR4A2. J. Cancer 7(11), 1388–1395 (2016).
    • 86. Yoshikawa A, Gotanda Y, Suzuki Y et al. Age- and gender-specific distributions of hepatitis B virus (HBV) genotypes in Japanese HBV-positive blood donors. Transfusion 49(7), 1314–1320 (2009).
    • 87. Wang L, Qin H, Chen B, Xin X, Li J, Han H. Overexpressed active Notch1 induces cell growth arrest of HeLa cervical carcinoma cells. Int. J. Gynecol. Cancer 17(6), 1283–1292 (2007).
    • 88. Suman S, Das TP, Ankem MK, Damodaran C. Targeting Notch signaling in colorectal cancer. Curr. Colorectal Cancer Rep. 10(4), 411–416 (2014).
    • 89. Galluzzo P, Bocchetta M. Notch signaling in lung cancer. Expert Rev. Anticancer Ther. 11(4), 533–540 (2011).
    • 90. Motooka Y, Fujino K, Sato Y, Kudoh S, Suzuki M, Ito T. Pathobiology of Notch2 in lung cancer. Pathology 49(5), 486–493 (2017).
    • 91. Di Ianni M, Baldoni S, Rosati E et al. A new genetic lesion in B-CLL: a NOTCH1 PEST domain mutation. (1365–2141 [Electronic]).
    • 92. Puente XS, Pinyol M, Quesada V et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475(7354), 101–105 (2011).
    • 93. Agrawal N, Frederick MJ, Pickering CR et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333(6046), 1154–1157 (2011).
    • 94. Pickering CR, Zhou JH, Lee JJ et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin. Cancer Res. 20(24), 6582–6592 (2014).
    • 95. Song Y, Li L, Ou Y et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature 509(7498), 91–95 (2014).
    • 96. George J, Lim JS, Jang SJ et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524(7563), 47–53 (2015).
    • 97. Rekhtman N, Pietanza MC, Hellmann MD et al. Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma-like and non-small cell carcinoma-like subsets. Clin. Cancer Res. 22(14), 3618–3629 (2016).
    • 98. Cancer Genome Atlas Research Network, Brat DJ, Verhaak RG et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 372(26), 2481–2498 (2015).
    • 99. Biswas S, Rao CM. Epigenetics in cancer: fundamentals and beyond. Pharmacol. Ther. 173, 118–134 (2017).
    • 100. Kagohara LT, Stein-O'Brien GL, Kelley D et al. Epigenetic regulation of gene expression in cancer: techniques, resources and analysis. Brief Funct. Genomics 17(1), 49–63 (2017).
    • 101. Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell 128(4), 669–681 (2007).
    • 102. Du Q, Luu PL, Stirzaker C, Clark SJ. Methyl-CpG-binding domain proteins: readers of the epigenome. Epigenomics 7(6), 1051–1073 (2015).
    • 103. Chen QW, Zhu XY, Li YY, Meng ZQ. Epigenetic regulation and cancer (review). Oncol. Rep. 31(2), 523–532 (2014).
    • 104. Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11(5), 384–400 (2012).
    • 105. Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat. Rev. Drug Discov. 13(9), 673 (2014).
    • 106. Pande V. Understanding the complexity of epigenetic target space: miniperspective. J. Med. Chem. 59(4), 1299–1307 (2016).
    • 107. Brownell JE, Allis CD. Special HATs for special occasions: linking histone acetylation to chromatin assembly and gene activation. Curr. Opin. Genet. Dev. 6(2), 176–184 (1996).
    • 108. Ruiz-Carrillo A, Wangh LJ, Allfrey VG. Processing of newly synthesized histone molecules. Science 190(4210), 117–128 (1975).
    • 109. Baxter E, Windloch K, Gannon F, Lee JS. Epigenetic regulation in cancer progression. Cell Biosci. 4(1), 45 (2014).
    • 110. Huang Y, Greene E, Stewart TM et al. Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc. Natl Acad. Sci. USA 104(19), 8023–8028 (2007).
    • 111. Shi Y, Lan F, Matson C et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7), 941–953 (2004).
    • 112. Banerjee T, Chakravarti D. A peek into the complex realm of histone phosphorylation. Mol. Cell. Biol. 05631–05611 (2011).
    • 113. Kschonsak M, Haering CH. Shaping mitotic chromosomes: from classical concepts to molecular mechanisms. BioEssays 37(7), 755–766 (2015).
    • 114. Nowak SJ, Corces VG. Phosphorylation of histone H3: a balancing act between chromosome condensation and transcriptional activation. Trends Genet. 20(4), 214–220 (2004).
    • 115. Rossetto D, Avvakumov N, Cote J. Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics 7(10), 1098–1108 (2012).
    • 116. Aihara H, Nakagawa T, Yasui K et al. Nucleosomal histone kinase-1 phosphorylates H2A Thr 119 during mitosis in the early Drosophila embryo. Genes Dev. 18(8), 877–888 (2004).
    • 117. Lowndes NF, Toh GW. DNA repair: the importance of phosphorylating histone H2AX. Curr. Biol. 15(3), R99–R102 (2005).
    • 118. Pinto DMS, Flaus A. Structure and function of histone H2AX. In: Genome Stability and Human Diseases. Springer, 55–78 (2010).
    • 119. Cole AJ, Clifton-Bligh R, Marsh DJ. Histone H2B monoubiquitination: roles to play in human malignancy. Endocr. Relat. Cancer 22(1), T19–T33 (2015).
    • 120. Wang Z, Li Y, Kong D, Ahmad A, Banerjee S, Sarkar FH. Cross-talk between miRNA and Notch signaling pathways in tumor development and progression. Cancer Lett. 292(2), 141–148 (2010).
    • 121. Geisler S, Coller J. RNA in unexpected places: long non-coding RNA functions in diverse cellular contexts. Nat. Rev. Mol. Cell Biol. 14(11), 699–712 (2013).
    • 122. Reicher A, Foßelteder J, Kwong LN, Pichler M. Crosstalk between the Notch signaling pathway and long non-coding RNAs. Cancer Lett. 420, 91–96 (2018).
    • 123. Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol. Cell 43(6), 904–914 (2011).
    • 124. Pan Y, Mao Y, Jin R, Jiang L. Crosstalk between the Notch signaling pathway and non-coding RNAs in gastrointestinal cancers. Oncol. Lett. 15(1), 31–40 (2018).
    • 125. Piazzi G, Bazzoli F, Ricciardiello L. Epigenetic silencing of Notch signaling in gastrointestinal cancers. Cell Cycle 11(23), 4323–4327 (2012).
    • 126. Piazzi G, Fini L, Selgrad M et al. Epigenetic regulation of delta-like1 controls Notch1 activation in gastric cancer. Oncotarget 2(12), 1291–1301 (2011).
    • 127. Gaykalova DA, Zizkova V, Guo T et al. Integrative computational analysis of transcriptional and epigenetic alterations implicates DTX1 as a putative tumor suppressor gene in HNSCC. Oncotarget 8(9), 15349 (2017).
    • 128. Hernandez-Vargas H, Lambert M-P, Le Calvez-Kelm F et al. Hepatocellular carcinoma displays distinct DNA methylation signatures with potential as clinical predictors. PLOS ONE 5(3), e9749 (2010).
    • 129. Zhu Y, Ye M, Xu H et al. Methylation status of CpG sites in the NOTCH4 promoter region regulates NOTCH4 expression in patients with tetralogy of Fallot. Mol. Med. Rep. 22(5), 4412–4422 (2020).
    • 130. Bhagat TD, Zou Y, Huang S et al. Notch pathway is activated via genetic and epigenetic alterations and is a therapeutic target in clear cell renal cancer. J. Biol. Chem. M116, 745208 (2016).
    • 131. Kuang SQ, Fang Z, Zweidler-Mckay PA et al. Epigenetic inactivation of Notch–Hes pathway in human B-cell acute lymphoblastic leukemia. PLOS ONE 8(4), e61807 (2013).
    • 132. Scourzic L, Couronne L, Pedersen MT et al. DNMT3A(R882H) mutant and Tet2 inactivation cooperate in the deregulation of DNA methylation control to induce lymphoid malignancies in mice. Leukemia 30(6), 1388–1398 (2016).
    • 133. Arzate-Mejia RG, Valle-Garcia D, Recillas-Targa F. Signaling epigenetics: novel insights on cell signaling and epigenetic regulation. IUBMB Life 63(10), 881–895 (2011).
    • 134. Cierpicki T, Risner LE, Grembecka J et al. Structure of the MLL CXXC domain-DNA complex and its functional role in MLL-AF9 leukemia. Nat. Struct. Mol. Biol. 17(1), 62–68 (2010).
    • 135. Huang YC, Lin SJ, Shih HY et al. Epigenetic regulation of NOTCH1 and NOTCH3 by KMT2A inhibits glioma proliferation. Oncotarget 8(38), 63110–63120 (2017).
    • 136. Rauen T, Grammatikos AP, Hedrich CM et al. cAMP-responsive element modulator alpha (CREMalpha) contributes to decreased Notch-1 expression in T cells from patients with active systemic lupus erythematosus (SLE). J. Biol. Chem. 287(51), 42525–42532 (2012).
    • 137. Xia Y, Cao X, Xue X et al. Development of hair cells in inner ear is associated with expression and promoter methylation of Notch-1 in postnatal mice. Int. J. Clin. Exp. Med. 8(9), 15542 (2015).
    • 138. Moshkin YM, Kan TW, Goodfellow H et al. Histone chaperones ASF1 and NAP1 differentially modulate removal of active histone marks by LID-RPD3 complexes during NOTCH silencing. Mol. Cell 35(6), 782–793 (2009).
    • 139. Sawarkar R, Paro R. Interpretation of developmental signaling at chromatin: the polycomb perspective. Dev. Cell 19(5), 651–661 (2010).
    • 140. De Koning L, Corpet A, Haber JE, Almouzni G. Histone chaperones: an escort network regulating histone traffic. Nat. Struct. Mol. Biol. 14(11), 997–1007 (2007).
    • 141. Eitoku M, Sato L, Senda T, Horikoshi M. Histone chaperones: 30 years from isolation to elucidation of the mechanisms of nucleosome assembly and disassembly. Cell. Mol. Life Sci. 65(3), 414–444 (2008).
    • 142. Pile LA, Schlag EM, Wassarman DA. The SIN3/RPD3 deacetylase complex is essential for G2 phase cell cycle progression and regulation of SMRTER corepressor levels. Mol. Cell. Biol. 22(14), 4965–4976 (2002).
    • 143. Vaquero A, Sternglanz R, Reinberg D. NAD+-dependent deacetylation of H4 lysine 16 by class III HDACs. Oncogene 26(37), 5505–5520 (2007).
    • 144. Majidinia M, Darband SG, Kaviani M, Nabavi SM, Jahanban-Esfahlan R, Yousefi B. Cross-regulation between Notch signaling pathway and miRNA machinery in cancer. DNA Repair 66–67, 30–41 (2018).
    • 145. Kefas B, Comeau L, Floyd DH et al. The neuronal microRNA miR-326 acts in a feedback loop with notch and has therapeutic potential against brain tumors. J. Neurosci. 29(48), 15161–15168 (2009).
    • 146. Xing CG, Zhu BS, Fan XQ et al. Effects of LY294002 on the invasiveness of human gastric cancer in vivo in nude mice. World J. Gastroenterol. 15(40), 5044–5052 (2009).
    • 147. Song G, Zhang Y, Wang L. MicroRNA-206 targets notch3, activates apoptosis, and inhibits tumor cell migration and focus formation. J. Biol. Chem. 284(46), 31921–31927 (2009).
    • 148. Cai H, Yao J, An Y et al. lncRNA HOTAIR acts as competing endogenous RNA to control the expression of Notch3 via sponging miR-613 in pancreatic cancer. Oncotarget 8(20), 32905 (2017).
    • 149. Raveh E, Matouk IJ, Gilon M, Hochberg A. The H19 long non-coding RNA in cancer initiation, progression and metastasis – a proposed unifying theory. Mol. Cancer 14(1), 184 (2015).
    • 150. Zhang K, Han X, Zhang Z et al. The liver-enriched lnc-LFAR1 promotes liver fibrosis by activating TGFβ and Notch pathways. Nat. Commun. 8(1), 144 (2017).
    • 151. Kong D, Wang Y. Retracted: knockdown of lncRNA HULC inhibits proliferation, migration, invasion, and promotes apoptosis by sponging miR-122 in osteosarcoma. J. Cell. Biochem. 119(1), 1050–1061 (2018).
    • 152. Lee M, Kim HJ, Kim SW et al. The long non-coding RNA HOTAIR increases tumour growth and invasion in cervical cancer by targeting the Notch pathway. Oncotarget 7(28), 44558–44571 (2016).
    • 153. Rani N, Nowakowski TJ, Zhou H et al. A primate lncRNA mediates notch signaling during neuronal development by sequestering miRNA. Neuron 90(6), 1174–1188 (2016).
    • 154. Ohtsuka M, Ling H, Ivan C et al. H19 noncoding RNA, an independent prognostic factor, regulates essential Rb-E2F and CDK8-beta-catenin signaling in colorectal cancer. EBioMedicine 13, 113–124 (2016).
    • 155. Durinck K, Wallaert A, Van De Walle I et al. The Notch driven long non-coding RNA repertoire in T-cell acute lymphoblastic leukemia. Haematologica 99(12), 1808–1816 (2014).
    • 156. Katsushima K, Natsume A, Ohka F et al. Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment. Nat. Commun. 7, 13616 (2016).
    • 157. Ounzain S, Micheletti R, Arnan C et al. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis. J. Mol. Cell. Cardiol. 89(Pt A), 98–112 (2015). • The study investigates the role of lncRNAs in human cardiac precursor cells (CPCs), revealing 570 modulated lncRNAs during cardiac differentiation in CPCs from the human fetal heart. These lncRNAs are associated with active cardiac enhancers and super-enhancers, correlating with the expression of nearby cardiac genes. A prominently upregulated lncRNA, named CARMEN, exhibits RNA-dependent enhancing activity and is positioned upstream of the cardiac mesoderm-specifying gene regulatory network. CARMEN interacts with SUZ12 and EZH2, components of PRC2. Knockdown of CARMEN impedes cardiac specification and differentiation in CPCs, independently of MIR-143 and -145 expression. Significantly, CARMEN is activated during pathological remodeling in mouse and human hearts and is crucial for maintaining cardiac identity in differentiated cardiomyocytes. This study establishes CARMEN as a critical regulator of cardiac cell differentiation and homeostasis.
    • 158. Ebbesen KK, Kjems J, Hansen TB. Circular RNAs: identification, biogenesis and function. Biochim. Biophys. Acta 1859(1), 163–168 (2016).
    • 159. Ntziachristos P, Lim JS, Sage J, Aifantis I. From fly wings to targeted cancer therapies: a centennial for notch signaling. Cancer Cell 25(3), 318–334 (2014).
    • 160. Olsauskas-Kuprys R, Zlobin A, Osipo C. Gamma secretase inhibitors of Notch signaling. Onco Targets Ther. 6, 943–955 (2013).
    • 161. Clarke EE, Churcher I, Ellis S et al. Intra- or intercomplex binding to the gamma-secretase enzyme. A model to differentiate inhibitor classes. J. Biol. Chem. 281(42), 31279–31289 (2006).
    • 162. Kummar S, O'Sullivan Coyne G, Do KT et al. Clinical activity of the gamma-secretase inhibitor PF-03084014 in adults with desmoid tumors (aggressive fibromatosis). J. Clin. Oncol. 35(14), 1561–1569 (2017).
    • 163. Wei P, Walls M, Qiu M et al. Evaluation of selective γ-secretase inhibitor PF-03084014 for its antitumor efficacy and gastrointestinal safety to guide optimal clinical trial design. Mol. Cancer Ther. 9(6), 1618–1628 (2010). •• The paper describes the potent antitumor effects of PF-03084014, a selective γ-secretase inhibitor targeting Notch signaling, demonstrated by reduced Notch intracellular domain levels and downregulation of Notch target genes in T-cell acute lymphoblastic leukemia (T-ALL) cell lines. PF-03084014 shows broad antitumor efficacy in Notch-dependent models, and its intermittent dosing schedule minimizes gastrointestinal toxicity, presenting a promising approach for developing therapies for Notch receptor-dependent cancers, currently under investigation in phase I clinical trials for T-ALL and advanced solid tumors.
    • 164. Takebe N, Nguyen D, Yang SX. Targeting notch signaling pathway in cancer: clinical development advances and challenges. Pharmacol. Ther. 141(2), 140–149 (2014).
    • 165. Huynh C, Poliseno L, Segura MF et al. The novel gamma secretase inhibitor RO4929097 reduces the tumor initiating potential of melanoma. PLOS ONE 6(9), e25264 (2011).
    • 166. Zhang C, Qin S, Xie H et al. RO4929097, a selective γ-secretase inhibitor, inhibits subretinal fibrosis via suppressing Notch and ERK1/2 signaling in laser-induced mouse model. Invest. Ophthalmol. Vis. Sci. 63(10), 14 (2022).
    • 167. Ryeom SW. The cautionary tale of side effects of chronic Notch1 inhibition. J. Clin. Invest. 121(2), 508–509 (2011).
    • 168. Wu Y, Cain-Hom C, Choy L et al. Therapeutic antibody targeting of individual Notch receptors. Nature 464(7291), 1052–1057 (2010).
    • 169. Zheng X, Pang B, Gu G et al. Melatonin inhibits glioblastoma stem-like cells through suppression of EZH2-NOTCH1 signaling axis. Int. J. Biol. Sci. 13(2), 245–253 (2017).
    • 170. Palermo R, Checquolo S, Giovenco A et al. Acetylation controls Notch3 stability and function in T-cell leukemia. Oncogene 31(33), 3807–3817 (2012). • The study reveals a novel property of Notch3, demonstrating that acetylation at lysines 1692 and 1731 by p300 and HDAC1, respectively, regulates its ubiquitination and proteasomal-mediated degradation. Histone deacetylase inhibitors disrupt this balance, favoring hyperacetylation and leading to decreased Notch3 expression and transcriptional activity, impairing downstream signaling. The findings suggest that targeting the acetylation/deacetylation process of Notch3 could be a promising approach for therapeutic intervention in T-ALL.
    • 171. Mohammed TA, Holen KD, Jaskula-Sztul R et al. A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. Oncologist 16(6), 835–843 (2011).
    • 172. Stockhausen MT, Sjolund J, Manetopoulos C, Axelson H. Effects of the histone deacetylase inhibitor valproic acid on Notch signalling in human neuroblastoma cells. Br. J. Cancer 92(4), 751–759 (2005).
    • 173. A Sun L, He Q, Tsai C et al. HDAC inhibitors suppressed small cell lung cancer cell growth and enhanced the suppressive effects of receptor-targeting cytotoxins via upregulating somatostatin receptor II. Am. J. Transl. Res. 10(2), 545–553 (2018). • The study investigates the potential therapeutic approach for small-cell lung cancer (SCLC), noting that Notch signaling activation suppresses SCLC cell growth. Histone deacetylase inhibitors, particularly valproic acid (VPA), also exhibit suppressive effects on SCLC cell growth, inducing cell cycle arrest at G1 phase and activating Notch signaling. VPA treatment increases the expression of SSTR2, a target for anticancer drug development. Combining VPA with SSTR2-targeted cytotoxins (CPT-SST and COL-SST) leads to stronger suppression of SCLC cell growth, suggesting a potential novel clinical strategy for enhanced anticancer therapy in SCLC treatment.
    • 174. Tsai C, Leslie JS, Franko-Tobin LG et al. Valproic acid suppresses cervical cancer tumor progression possibly via activating Notch1 signaling and enhances receptor-targeted cancer chemotherapeutic via activating somatostatin receptor type II. Arch. Gynecol. Obstet. 288(2), 393–400 (2013).
    • 175. Franko-Tobin LG, Mackey LV, Huang W et al. Notch1-mediated tumor suppression in cervical cancer with the involvement of SST signaling and its application in enhanced SSTR-targeted therapeutics. Oncologist 17(2), 220–232 (2012).
    • 176. Sen P, Ghosh SS. γ-Secretase inhibitor potentiates the activity of suberoylanilide hydroxamic acid by inhibiting its ability to induce epithelial to mesenchymal transition and stemness via Notch pathway activation in triple-negative breast cancer cells. ACS Pharmacol. Transl. Sci. 6(10), 1396–1415 (2023).
    • 177. Butler LM, Agus DB, Scher HI et al. Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res. 60(18), 5165–5170 (2000).
    • 178. Vigushin DM, Ali S, Pace PE et al. Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin. Cancer Res. 7(4), 971–976 (2001).
    • 179. Hu W, Lu C, Dong HH et al. Biological roles of the Delta family Notch ligand Dll4 in tumor and endothelial cells in ovarian cancer. Cancer Res. 71(18), 6030–6039 (2011). •• The study explores the clinical and biological significance of DLL4 in ovarian cancer, finding that DLL4 is overexpressed in 72% of tumors and is an independent predictor of poor survival. Patients responding to anti-VEGF therapy had lower DLL4 levels than those with stable or progressive disease. Under hypoxic conditions, VEGF increased DLL4 expression in tumor vasculature. DLL4 downregulated VEGFR2 expression in endothelial cells through methylation of the VEGFR2 promoter. Silencing DLL4 inhibited cell growth and angiogenesis in ovarian tumor cells and tumor-associated endothelial cells, inducing hypoxia in the tumor microenvironment. Combining DLL4-targeted siRNA with bevacizumab showed greater inhibition of tumor growth, suggesting that targeting DLL4 in conjunction with anti-VEGF treatment could improve outcomes in ovarian cancer treatment.
    • 180. Liu Q, Zheng JM, Chen JK et al. Histone deacetylase 5 promotes the proliferation of glioma cells by upregulation of Notch 1. Mol. Med. Rep. 10(4), 2045–2050 (2014).
    • 181. Pandya K, Meeke K, Clementz AG et al. Targeting both Notch and ErbB-2 signalling pathways is required for prevention of ErbB-2-positive breast tumour recurrence. Br. J. Cancer 105(6), 796–806 (2011).
    • 182. Farnie G, Willan PM, Clarke RB, Bundred NJ. Combined inhibition of ErbB1/2 and Notch receptors effectively targets breast ductal carcinoma in situ (DCIS) stem/progenitor cell activity regardless of ErbB2 status. PLOS ONE 8(2), e56840 (2013).
    • 183. Ma Y, Ren Y, Han EQ et al. Inhibition of the Wnt–beta-catenin and Notch signaling pathways sensitizes osteosarcoma cells to chemotherapy. Biochem. Biophys. Res. Commun. 431(2), 274–279 (2013).
    • 184. Jin R, Nakada M, Teng L et al. Combination therapy using Notch and Akt inhibitors is effective for suppressing invasion but not proliferation in glioma cells. Neurosci. Lett. 534, 316–321 (2013).
    • 185. Shepherd C, Banerjee L, Cheung CW et al. PI3K/mTOR inhibition upregulates NOTCH-MYC signalling leading to an impaired cytotoxic response. Leukemia 27(3), 650–660 (2013).
    • 186. Yao J, Qian C, Shu T, Zhang X, Zhao Z, Liang Y. Combination treatment of PD98059 and DAPT in gastric cancer through induction of apoptosis and downregulation of WNT/beta-catenin. Cancer Biol. Ther. 14(9), 833–839 (2013).
    • 187. Doody RS, Raman R, Farlow M et al. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N. Engl. J. Med. 369(4), 341–350 (2013).
    • 188. Siemers ER, Quinn JF, Kaye J et al. Effects of a gamma-secretase inhibitor in a randomized study of patients with Alzheimer disease. Neurology 66(4), 602–604 (2006).
    • 189. Fleisher AS, Raman R, Siemers ER et al. Phase 2 safety trial targeting amyloid beta production with a gamma-secretase inhibitor in Alzheimer disease. Arch. Neurol. 65(8), 1031–1038 (2008).
    • 190. Mandasari M, Sawangarun W, Katsube K, Kayamori K, Yamaguchi A, Sakamoto K. A facile one-step strategy for the generation of conditional knockout mice to explore the role of Notch1 in oroesophageal tumorigenesis. Biochem. Biophys. Res. Commun. 469(3), 761–767 (2016).
    • 191. Han N, Hu G, Shi L et al. Notch1 ablation radiosensitizes glioblastoma cells. Oncotarget 8(50), 88059–88068 (2017).
    • 192. Huo L, Wei W, Wu S et al. Effect of dihydroarteminin combined with siRNA targeting Notch1 on Notch1/c-Myc signaling in T-cell lymphoma cells. Exp. Ther. Med. 15(3), 3059–3065 (2018).
    • 193. Yamaguchi N, Oyama T, Ito E et al. NOTCH3 signaling pathway plays crucial roles in the proliferation of ErbB2-negative human breast cancer cells. Cancer Res. 68(6), 1881–1888 (2008).
    • 194. Lewis NA, Klein RH, Kelly C, Yee J, Knoepfler PS. Histone H3.3 K27M chromatin functions implicate a network of neurodevelopmental factors including ASCL1 and NEUROD1 in DIPG. Epigenetics Chromatin 15(1), 18 (2022). • Enhanced gene expression in H3.3K27M cells is linked to accessible enhancers and super-enhancers, particularly involving neurogenesis and NOTCH signaling. Motif analysis implicates ASCL1 and NEUROD1 transcription factors in the neuro-oncogenic K27M signaling pathway.