DOI QR코드

DOI QR Code

Vorinostat-induced acetylation of RUNX3 reshapes transcriptional profile through long-range enhancer-promoter interactions in natural killer cells

  • Eun-Chong Lee (Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine) ;
  • Kyungwoo Kim (Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine) ;
  • Woong-Jae Jung (Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine) ;
  • Hyoung-Pyo Kim (Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine)
  • Received : 2023.03.23
  • Accepted : 2023.05.09
  • Published : 2023.07.31

Abstract

Natural killer (NK) cells are an essential part of the innate immune system that helps control infections and tumors. Recent studies have shown that Vorinostat, a histone deacetylase (HDAC) inhibitor, can cause significant changes in gene expression and signaling pathways in NK cells. Since gene expression in eukaryotic cells is closely linked to the complex three-dimensional (3D) chromatin architecture, an integrative analysis of the transcriptome, histone profiling, chromatin accessibility, and 3D genome organization is needed to gain a more comprehensive understanding of how Vorinostat impacts transcription regulation of NK cells from a chromatin-based perspective. The results demonstrate that Vorinostat treatment reprograms the enhancer landscapes of the human NK-92 NK cell line while overall 3D genome organization remains largely stable. Moreover, we identified that the Vorinostat-induced RUNX3 acetylation is linked to the increased enhancer activity, leading to elevated expression of immune response-related genes via long-range enhancer-promoter chromatin interactions. In summary, these findings have important implications in the development of new therapies for cancer and immune-related diseases by shedding light on the mechanisms underlying Vorinostat's impact on transcriptional regulation in NK cells within the context of 3D enhancer network.

Keywords

Acknowledgement

This work was supported by the National Research Foundation of Korea (NRF) grants funded by the Korean government (MSIP) (2018M3A9D3079290, 2020R1A2C2013258, and 2022M3A9B6017424 to H.-P. Kim).

References

  1. Vivier E, Tomasello E, Baratin M, Walzer T and Ugolini S (2008) Functions of natural killer cells. Nat Immunol 9, 503-510  https://doi.org/10.1038/ni1582
  2. Caligiuri MA (2008) Human natural killer cells. Blood 112, 461-469  https://doi.org/10.1182/blood-2007-09-077438
  3. Kee BL, Morman RE and Sun M (2020) Transcriptional regulation of natural killer cell development and maturation. Adv Immunol 146, 1-28  https://doi.org/10.1016/bs.ai.2020.01.001
  4. Moretta A, Marcenaro E, Parolini S, Ferlazzo G and Moretta L (2008) NK cells at the interface between innate and adaptive immunity. Cell Death Differ 15, 226-233  https://doi.org/10.1038/sj.cdd.4402170
  5. Paul S and Lal G (2017) The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol 8, 1124 
  6. Chen Y, Lu D, Churov A and Fu R (2020) Research Progress on NK cell receptors and their signaling pathways. Mediators Inflamm 2020, 6437057 
  7. Meza Guzman LG, Keating N and Nicholson SE (2020) Natural killer cells: tumor surveillance and signaling. Cancers (Basel) 12, 952 
  8. Wang D and Malarkannan S (2020) Transcriptional regulation of natural killer cell development and functions. Cancers (Basel) 12, 1591 
  9. Bi J and Wang X (2020) Molecular regulation of NK cell maturation. Front Immunol 11, 1945 
  10. Spitz F and Furlong EE (2012) Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 13, 613-626  https://doi.org/10.1038/nrg3207
  11. Levine M and Tjian R (2003) Transcription regulation and animal diversity. Nature 424, 147-151  https://doi.org/10.1038/nature01763
  12. Holliday R (2006) Epigenetics: a historical overview. Epigenetics 1, 76-80  https://doi.org/10.4161/epi.1.2.2762
  13. Mumbach MR, Satpathy AT, Boyle EA et al (2017) Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nat Genet 49, 1602-1612  https://doi.org/10.1038/ng.3963
  14. Santosa EK, Lau CM, Sahin M, Leslie CS and Sun JC (2023) 3D chromatin dynamics during innate and adaptive immune memory acquisition. bioRxiv 
  15. Grunstein M (1997) Histone acetylation in chromatin structure and transcription. Nature 389, 349-352  https://doi.org/10.1038/38664
  16. Gujral P, Mahajan V, Lissaman AC and Ponnampalam AP (2020) Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod Biol Endocrinol 18, 84 
  17. Bondarev AD, Attwood MM, Jonsson J, Chubarev VN, Tarasov VV and Schioth HB (2021) Recent developments of HDAC inhibitors: emerging indications and novel molecules. Br J Clin Pharmacol 87, 4577-4597  https://doi.org/10.1111/bcp.14889
  18. Siegel D, Hussein M, Belani C et al (2009) Vorinostat in solid and hematologic malignancies. J Hematol Oncol 2, 31 
  19. Berghuis D, Schilham MW, Vos HI et al (2012) Histone deacetylase inhibitors enhance expression of NKG2D ligands in Ewing sarcoma and sensitize for natural killer cell-mediated cytolysis. Clin Sarcoma Res 2, 8 
  20. Desimio MG, Giuliani E and Doria M (2017) The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity. Virology 510, 9-21  https://doi.org/10.1016/j.virol.2017.06.033
  21. Ogbomo H, Michaelis M, Kreuter J, Doerr HW and Cinatl J Jr (2007) Histone deacetylase inhibitors suppress natural killer cell cytolytic activity. FEBS Lett 581, 1317-1322  https://doi.org/10.1016/j.febslet.2007.02.045
  22. Pace M, Williams J, Kurioka A et al (2016) Histone Deacetylase Inhibitors Enhance CD4 T Cell Susceptibility to NK Cell Killing but Reduce NK Cell Function. PLoS Pathog 12, e1005782 
  23. Kotzur R, Duev-Cohen A, Kol I, Reches A, Mandelboim O and Stein N (2022) NK-92 cells retain vitality and functionality when grown in standard cell culture conditions. PLoS One 17, e0264897 
  24. Misteli T (2020) The self-organizing genome: principles of genome architecture and function. Cell 183, 28-45  https://doi.org/10.1016/j.cell.2020.09.014
  25. Fortin JP and Hansen KD (2015) Reconstructing A/B compartments as revealed by Hi-C using long-range correlations in epigenetic data. Genome Biol 16, 180 
  26. Dixon JR, Selvaraj S, Yue F et al (2012) Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376-380  https://doi.org/10.1038/nature11082
  27. Rao SS, Huntley MH, Durand NC et al (2014) A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665-1680  https://doi.org/10.1016/j.cell.2014.11.021
  28. Boto P, Csuth TI and Szatmari I (2018) RUNX3-mediated immune cell development and maturation. Crit Rev Immunol 38, 63-78  https://doi.org/10.1615/CritRevImmunol.2018025488
  29. North TE, Stacy T, Matheny CJ, Speck NA and de Bruijn MF (2004) Runx1 is expressed in adult mouse hematopoietic stem cells and differentiating myeloid and lymphoid cells, but not in maturing erythroid cells. Stem Cells 22, 158-168  https://doi.org/10.1634/stemcells.22-2-158
  30. Lai CB and Mager DL (2012) Role of runt-related transcription factor 3 (RUNX3) in transcription regulation of natural cytotoxicity receptor 1 (NCR1/NKp46), an activating natural killer (NK) cell receptor. J Biol Chem 287, 7324-7334  https://doi.org/10.1074/jbc.M111.306936
  31. Kawane T, Qin X, Jiang Q et al (2018) RUNX2 is required for the proliferation of osteoblast progenitors and induces proliferation by regulating Fgfr2 and Fgfr3. Sci Rep 8, 13551 
  32. Jin YH, Jeon EJ, Li QL et al (2004) Transforming growth factor-beta stimulates p300-dependent RUNX3 acetylation, which inhibits ubiquitination-mediated degradation. J Biol Chem 279, 29409-29417  https://doi.org/10.1074/jbc.M313120200
  33. Chuang LS, Ito K and Ito Y (2013) RUNX family: regulation and diversification of roles through interacting proteins. Int J Cancer 132, 1260-1271  https://doi.org/10.1002/ijc.27964
  34. Lee JW, Kim DM, Jang JW et al (2019) RUNX3 regulates cell cycle-dependent chromatin dynamics by functioning as a pioneer factor of the restriction-point. Nat Commun 10, 1897 
  35. Lotem J, Levanon D, Negreanu V, Leshkowitz D, Friedlander G and Groner Y (2013) RUNX3-mediated transcriptional program in cytotoxic lymphocytes. PLoS One 8, e80467 
  36. Sciume G, Mikami Y, Jankovic D et al (2020) Rapid enhancer remodeling and transcription factor repurposing enable high magnitude gene induction upon acute activation of NK cells. Immunity 53, 745-758 e744 
  37. Zhao X, Peng T, Cao X et al (2022) In vivo G-CSF treatment activates the GR-SOCS1 axis to suppress IFN-gamma secretion by natural killer cells. Cell Rep 40, 111342 
  38. Mevel R, Draper JE, Lie ALM, Kouskoff V and Lacaud G (2019) RUNX transcription factors: orchestrators of development. Development 146, dev148296 
  39. Brillantes M and Beaulieu AM (2019) Transcriptional control of natural killer cell differentiation. Immunology 156, 111-119  https://doi.org/10.1111/imm.13017
  40. Vogel C and Marcotte EM (2012) Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet 13, 227-232 https://doi.org/10.1038/nrg3185