BMB Reports 2023; 56(7): 398-403  https://doi.org/10.5483/BMBRep.2023-0044
Vorinostat-induced acetylation of RUNX3 reshapes transcriptional profile through long-range enhancer-promoter interactions in natural killer cells
Eun-Chong Lee1 , Kyungwoo Kim1,2 , Woong-Jae Jung1 & Hyoung-Pyo Kim1,2,3,4,*
1Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 03722, 2Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, 3Yonsei Genome Center, Yonsei University College of Medicine, Seoul 03722, 4Division of Biology, Pohang University of Science and Technology, Pohang 37673, Korea
Correspondence to: Tel: +82-2-2228-1842; Fax: +82-2-363-8676; E-mail: kimhp@yuhs.ac
Received: March 23, 2023; Revised: April 7, 2023; Accepted: May 9, 2023; Published online: May 24, 2023.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
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: Chromatin, Enhancer, Natural killer cells, RUNX3, Vorinostat
INTRODUCTION

Natural killer (NK) cells are highly specialized cytotoxic lymphocytes of the innate immune system that play a critical role in the control of microbial infections and several types of tumors (1). Unlike T or B cells, NK cells exert cytolytic functions without prior sensitization by utilizing germline-encoded, non-rearranged activating receptors (2, 3). They also modulate the function of other innate and adaptive immune cells by secreting several cytokines after activation (4, 5). Recently, key molecules involved in multiple signaling cascades downstream of major activating receptors were identified (6, 7). Moreover, the development and function of NK cells rely on the precise regulation of gene expression mediated by a complex matrix of transcription factors in response to the environmental cues (8, 9). Therefore, a better understanding of the molecular mechanisms underlying the transcriptional regulation of NK cells is important for developing new treatment strategies to combat infectious diseases and cancers.

Gene expression of eukaryotic cells is controlled by transcription factors (TFs) that bind to regulatory elements such as promoters and enhancers (10, 11). Moreover, epigenetic features, such as histone modifications and DNA methylation, are thought to play a crucial role in shaping chromatin structures and accessibilities of these regulatory elements (12). Enhancers are often located far from their target genes, and three-dimensional (3D) genome structure provides a mechanism to precisely regulate gene expression. The action range of enhancers is restricted by topologically associating domain (TAD) boundaries; intra-TAD chromatin looping facilitates the interaction of distal enhancers with their target promoters (13, 14).

Histone acetylation is a post-translational modification that can neutralize the positive charge of lysine residue, thereby reducing the interaction between the histone and negatively charged DNA, making DNA more accessible to the transcriptional machinery, and promoting gene expression (15). Histone acetylation is strictly regulated by the concerted activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs) (16). Thus far, quite a few HDAC inhibitors have been reported, which have been shown to increase the acetylation of core histones, resulting in altered chromatin structure and gene expression (17).

Recent studies have demonstrated that the drug Vorinostat, an inhibitor of class I/II HDAC, could be used in a promising novel approach for treating leukemias and other malignancies (18). Moreover, Vorinostat has shown potential as an immunotherapeutic enhancer, improving the ability of NK cells to recognize tumor cells that have evaded immune surveillance (19, 20). However, it may also inhibit activating receptors’ expression and NK cell lytic activity against leukemic cells (21, 22). Therefore, further extensive studies are required to optimize the immunotherapeutic application of Vorinostat for the treatment of cancers and other diseases. Particular focus should be made on understanding how Vorinostat regulates the complex transcriptional network of NK cells in the 3D chromatin architecture. We analyzed the global transcriptional, epigenetic, and topological changes in NK cells in response to HDAC inhibitor and demonstrated the regulatory role that Vorinostat exerts in 3D enhancer network via acetylation of RUNX3 protein.

RESULTS

Genome-wide changes in gene expression and chromatin state of NK-92 cells in response to Vorinostat

Human NK cell line NK-92 cells display characteristics of activated NK cells in vivo and are currently in clinical trials as an off-the-shelf cellular immunotherapy for various cancers due to its prolonged activation, non-toxicity, low immunogenicity, and can be easily and rapidly expanded in culture (23). Exposing NK-92 cells to up to 1 μM Vorinostat for 24 hours did not significantly inhibit cell viability (Fig. 1A) or induce apoptosis (Fig. 1B). To understand how HDAC affects gene expression in NK cells, NK-92 cells were cultured for 24 hours in the absence/presence of 1 μM Vorinostat, and the global transcriptional, epigenetic, and topological changes occurring in NK-92 cells were analyzed (Fig. 1C). Upon Vorinostat treatment, differential RNA expression analysis revealed 1,829 upregulated and 1,991 downregulated genes (Fig. 1D). Gene ontology analysis indicated that the genes upregulated by Vorinostat were primarily enriched for immune-related pathways, while the downregulated genes were mostly associated with ribosome biogenesis and protein folding (Fig. 1E).

H3K27 acetylation (H3K27ac) is closely linked to the epigenetic control of gene expression and is a marker for active enhancers and promoters. To investigate the impact of Vorinostat treatment on H3K27ac levels, immunoblot assays were performed on total cell lysates, revealing higher levels of H3K27ac in the cells treated with Vorinostat (Fig. 1F). To further explore the effects of Vorinostat on enhancer activities, chromatin immunoprecipitation sequencing (ChIP-seq) was conducted to compare genome-wide H3K27ac occupancies (n = 2 per group) (Fig. 1G). Differential H3K27ac regions were identified: 7,061 were hyperacetylated and 785 were hypoacetylated by Vorinostat (Fig. 1G). We next performed assay for transposase-accessible chromatin with sequencing (ATAC-seq) to map the location and accessibility of regulatory elements genome-wide. Compared to the dynamic changes in H3K27ac levels, the degree of chromatin accessibility was marginally affected by Vorinostat in NK-92 cells (Fig. 1H). Representative genomic regions where Vorinostat induced changes in gene expression and chromatin state were shown in Fig. 1I.

Limited impact of Vorinostat on the 3D chromatin architecture in NK-92 cells

Mammalian genomes are packaged into intricate 3D structures consisting of hierarchical components such as compartments, TADs, and chromatin loops (24). Chromosomes separate into either active “A” or inactive “B” compartments, which can occur due to homotypic interactions between regions of the genome that share similar transcription and chromatin characteristics (25). TADs, the structural basis for organizing chromatin at a scale of hundreds of kilobases or below, are defined by preferential interactions between their chromatin (26, 27). We then investigated the extent to which Vorinostat affects higher-level genome organization of NK-92 cells by performing in situ Hi-C. Contact maps (Fig. 2A, Supplementary Table 1), the first eigenvector values from principal component analysis (Fig. 2B), and compartmentalization strength represented by saddle plots (Fig. 2C) indicated that Vorinostat had a negligible effect on segmentation of active and inactive chromosome domains into A and B compartments. Later, we defined TADs using insulation scores and found that the number of TADs (Fig. 2D) and the insulation capacity at TAD boundaries (Fig. 2E) were comparable between control and Vorinostat-treated cells. Moreover, genome-wide intra-TAD chromatin interactions (Fig. 2F) showed minimal changes upon Vorinostat treatment. We further explored intra-TAD chromatin looping by performing H3K27ac HiChIP in NK-92 cells. When statistically significant H3K27ac-based chromatin interactions were called using FitHiChIP, we identified a comparable number of high-confidence H3K27ac HiChIP loops (Fig. 2G, Supplementary Table 2). The major fraction (∼60%) of H3K27ac HiChIP loops represented chromatin interactions between promoter pairs, enhancer pairs, and promoter-enhancer pairs (Fig. 2G). Differential analysis for H3K27ac HiChIP loops demonstrated that the spatial proximity of nearly all H3K27ac HiChIP loops was preserved following Vorinostat treatment (Fig. 2H). Based on these topological data, it appears that Vorinostat has a limited impact on the three-dimensional chromatin architecture at the resolution of compartments, TADs, and enhancer-centric chromatin looping.

Vorinostat-induced RUNX3 acetylation reprograms enhancer landscape in NK-92 cells

To investigate the molecular mechanism underlying the effects of Vorinostat on gene expression, we analyzed TF binding motifs on ATAC-seq peaks that overlapped with differential H3K27ac peaks. Runt family motifs were enriched at hyperacetylated regions, while ERG and ETV family motifs were enriched at hypoacetylated regions in Vorinostat-treated cells (Fig. 3A). The Runt-related TF family comprises RUNX1, RUNX2, and RUNX3, which shares the evolutionarily conserved Runt domain. Tissue-specific and overlapping expression patterns indicate both exclusive and redundant roles for these three RUNX genes (28). Of these, RUNX3 is crucial in NK cell development and effector function, and considering its higher expression level compared to RUNX1 and RUNX2 in NK-92 cells (Fig. 3B) (28-31), we prioritized the analysis of RUNX3 and conducted ChIP-seq to examine their genome-wide binding pattern (Fig. 3C-F). The ChIP-seq analysis uncovered the presence of approximately 1,500 distinct genomic regions enriched with RUNX3 peaks, distributed across diverse genomic loci such as promoters, introns, and intergenic regions (Fig. 3D). While the levels of RUNX3 occupancy were similar in NK-92 cells cultured with or without Vorinostat (Fig. 3E, F), there was a notable increase in H3K27ac enrichment at the sites where RUNX3 was bound in Vorinostat-treated cells compared to control cells (Fig. 3G, H). Specifically, we identified a subset of RUNX3 binding sites (n = 604) with significantly elevated H3K27ac levels upon Vorinostat treatment, while only 36 RUNX3 peaks showed reduced H3K27ac levels (Fig. 3H). We next examined whether the increase in H3K27ac levels upon Vorinostat treatment was associated with posttranslational modifications of RUNX3. Previous studies have shown that acetylation plays a crucial role in enhancing RUNX3-dependent transactivation activity (32). The acetylation of RUNX3 by the histone acetyltransferase p300 is critical for the formation of a complex with bromodomain-containing protein 2 (BRD2), which in turn facilitates the recruitment of SWI/SNF and TFIID (33, 34). To test this hypothesis, we immunoprecipitated lysates from NK-92 cells using anti-acetyl-lysine antibodies and performed Western blotting with an anti-RUNX3 antibody. Our results indicate that Vorinostat treatment increases the acetylation level of RUNX3 protein, as shown in Fig. 3I.

RUNX3 acetylation enhances immune response-related gene expression via long-range enhancer-promoter chromatin interactions

Next, we sought to identify candidate genes whose expression could be regulated by Vorinostat-induced RUNX3 acetylation (i.e., activated RUNX3). To do so, we analyzed RUNX3 binding sites with enhanced H3K27ac levels and used H3K27ac HiChIP loops to link the activated enhancers with their target genes (Fig. 4A). Remarkably, only a few genes were directly bound by activated RUNX3 at their promoter regions, while most RUNX3 target genes were regulated by distal enhancers through long-range chromatin interactions (Fig. 4A). Analysis of the differential RNA expression of genes associated with activated RUNX3 found that 154 genes were deregulated by Vorinostat treatment (FDR < 0.05, FC > 1.3). Among these, 111 and 43 genes were upregulated and downregulated, respectively (Fig. 4B, Supplementary Table 3). Notably, gene ontology (GO) analysis revealed that the upregulated RUNX3 target genes (n = 111) were primarily enriched in immune response and cytokine production pathways (Fig. 4C). Specifically, Vorinostat treatment upregulated genes involved in transcription and signaling activity, such as NFKBIZ and MXI1, mediated by long-range enhancer-promoter chromatin interactions connecting their promoters with activated RUNX3-dependent distal enhancers (Fig. 4D, E).

DISCUSSION

The essential role of enhancers in the control of gene transcription during NK cell differentiation, activation, and function has been extensively studied in recent years (35, 36). However, there is limited research on the 3D chromatin architecture in NK cells and how it relates to enhancer activity (37). In this study, we systematically mapped the landscape of enhancer histone acetylation, chromatin accessibilities, higher-order chromatin structure, and enhancer-promoter regulatory interactions in human natural killer cells (Fig. 1C) to characterize the underlying mechanisms of the effect of Vorinostat on the gene expression profile. The ChIP-seq and ATAC-seq analysis revealed that HDAC inhibition by Vorinostat dramatically reprogrammed enhancer activities through changes in histone acetylation rather than chromatin accessibility in NK-92 cells (Fig. 1F-H). In contrast, in situ Hi-C analysis demonstrated that Vorinostat treatment did not induce changes in global genome organization at the compartment and TAD levels (Fig. 2A-F). Interestingly, Vorinostat-induced enhancer activation did not change spatial proximity between enhancer and promoter, given that most H3K27ac HiChIP loops remain stable after Vorinostat treatment (Fig. 2G, H). Taken together, gene expression changes following Vorinostat-mediated HDAC inhibition are controlled mainly at the level of enhancer histone acetylation rather than 3D chromatin architecture or chromatin accessibility.

Our analysis also revealed an overrepresentation of binding motifs for RUNX family transcription factors in Vorinostat-induced activated enhancer regions, with RUNX3 having the highest expression level in Vorinostat-induced NK-92 cells (Fig. 3A, B). Previous studies have demonstrated that expression of RUNX3 increases with NK maturation, and defects in its expression or activity can impede NK cell development (28, 38). Additionally, RUNX3-deficiency alters expression of genes involved in proliferation, survival, migration, and effector function of NK cells (39). The post-translational regulation of RUNX3’s transcriptional activity involves acetylation by p300 acetyltransferase, which protects it from degradation by the ubiquitin ligase Smurf and facilitates its interaction with BRD2 (32-34). The level of RUNX3 acetylation is influenced by environmental cues and is downregulated by HDAC activity. Therefore, RUNX3 may function as a sensor of HDAC inhibitor Vorinostat, and its acetylation potentially induces transcriptional reprogramming by elevating enhancer activities (Fig. 4F). However, it cannot be ruled out that other RUNX family proteins may exhibit compensatory effects on Vorinostat-induced enhancer reprogramming. To regulate its target genes, activated RUNX3 primarily depends on long-range chromatin interactions (Fig. 4A). The target genes upregulated by RUNX3 were significantly enriched in pathways related to immune response (Fig. 4B, C). Thus, RUNX3 acetylation induced by Vorinostat elevates the expression of immune response-related genes of NK cells through a spatial enhancer network.

It is important to note that mRNA expression levels are often used as a proxy for protein abundance. However, they do not always reflect the true abundance of the corresponding protein due to various post-transcriptional regulatory mechanisms that can affect mRNA stability, splicing, and translation (40). Given that Vorinostat downregulates the expression of genes involved in protein folding (Fig. 1G), further studies are warranted to investigate the regulatory effect of Vorinostat on the actual effector function of NK cells.

In conclusion, our study provides a comprehensive analysis of the molecular mechanisms underlying the effect of HDAC inhibitors on the transcriptional profile of NK cells. Our findings shed light on the intricate interplay between 3D chromatin structure, histone modifications, and gene expression in NK cells and offer insights into regulatory mechanisms that control NK cell function. This understanding may facilitate the development of novel therapeutic approaches to treat diseases resulting from dysregulated NK cell function.

MATERIALS AND METHODS

Materials and methods are available in supplementary material.

ACKNOWLEDGEMENTS

This work was supported by the National Research Foundation of Korea (NRF) grants funded by the Korean government (MSIP) (2018M3A9D3079290, 2020R1A2C2013258, and 2022M3A9 B6017424 to H.-P. Kim).

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Genome-wide changes in gene expression and chromatin state of NK-92 in response to Vorinostat treatment. (A) Viability of NK-92 cells measured 6, 24, and 48 hours after treatment with various concentrations of Vorinostat (n = 3 per group). (B) Representative flow cytometric plots of the frequency of live cells measured 24 hours after treatment with 1 μM Vorinostat (n = 2 per group). (C) Scheme of NK-92 cells cultured with and without 1 μM Vorinostat for 24 hours. (D) RNA-seq MA plot of control versus Vorinostat. The number of genes exhibiting > 1.3-fold increases in control (orange) or Vorinostat (purple) with a false discovery rate (FDR) < 0.05 are indicated. (E) Gene ontology terms associated with upregulated (left) or downregulated (right) genes by Vorinostat treatment. (F) Immunoblot assay for H3K27ac levels in total cell lysates. (G) Volcano plot showing significant Vorinostat-induced changes on the H3K27ac level. The number of peaks exhibiting > 1.5-fold increases in control (orange) or Vorinostat (purple) with an FDR < 0.05 are indicated. (H) ATAC-seq scatter plot of control versus Vorinostat. The number of peaks exhibiting > 1.5-fold increases in control (orange) or Vorinostat (purple) with an FDR < 0.05 are indicated. (I) Snapshot of RNA-seq, ChIP-seq, and ATAC-seq signal tracks in the representative genomic region.
Fig. 2. Global 3D chromatin structure of NK-92 cells is largely unaffected by Vorinostat treatment. (A) Hi-C contact maps generated by Juicebox at 250 kb, 100 kb, and 50 kb resolutions. (B) Distributions of first eigenvector values across the entirety of chromosome 1. (C) Saddle plots of compartmentalization strength with and without Vorinostat treatment. (D) Number of TAD boundaries obtained with Hi-C data. (E) Genome-wide averaged insulation scores plotted against distance around insulation center at TAD boundaries of control NK-92 cells. (F) Heatmaps show the average observed/expected Hi-C interactions in the TAD regions. (G) Distribution of regulatory elements at the anchors of H3K27ac HiChIP loops. (H) Volcano plot showing significant Vorinostat-induced changes in H3K27ac HiChIP loop strength. The number of loops exhibiting > 1.5-fold increases in control (orange) or Vorinostat (purple) with an FDR < 0.05 has been indicated.
Fig. 3. Vorinostat-induced RUNX3 acetylation increases enhancer activity of NK-92 cells. (A) Transcription factor motif identification from ATAC-seq peaks in differential H3K27ac peaks. (B) Expression of RUNX1, RUNX2, and RUNX3 genes determined by RNA-seq. (C) Snapshot of signal tracks for RUNX3 ChIP-seq, H3K27ac ChIP-seq, and ATAC-seq in the representative genomic region. (D) Box plot presenting the distribution of RUNX3 peaks among different gene features. (E) Scatterplot of ChIP-Seq signals called for RUNX3. (F) Heatmaps of ChIP-Seq signals called for RUNX3 (left) and H3K27ac (right). (G) Box plot of H3K27ac enrichment at RUNX3 binding sites (n = 1,807). (H) Scatter plot of relative enrichment in RUNX3 and H3K27ac ChIP-Seq signals at RUNX3 binding sites (n = 1,807). (I) acetylation of RUNX3 by Vorinostat treatment. The cell lysates were analyzed by immunoprecipitation using an antibody against acetylated lysine, followed by Western blotting using an anti-RUNX3 antibody (upper panel). To examine the level of RUNX3 expression, the same lysates were analyzed by Western blotting using anti-RUNX3 and anti-α-Tubulin antibodies (middle and lower panels).
Fig. 4. RUNX3 acetylation-mediated enhancer activation increases immune function-related gene expression via long-range chromatin interactions. (A) Activated RUNX3 target genes identified by integrated analysis of RUNX3 ChIP-seq, H3K27ac ChIP-seq, and H3K27ac HiChIP. (B) RNA-seq MA plot for the activated RUNX3 target genes. The number of genes exhibiting > 1.3-fold increases in control (orange) or Vorinostat (purple) with an FDR < 0.05 are indicated. (C) Enrichment of biological process gene ontology terms on the activated RUNX3 target genes whose RNA expression was upregulated in Vorinostat. (D, E) Snapshots (left panels) showing RUNX3 and H3K27ac ChIP-seq signal tracks and significant H3K27ac HiChIP loops at the typical activated RUNX3 target genes, including NFKBIZ (D) MXI1 (E). Arcs showing significant interactions with −Log10(Q) ≥ 5. Gray and blue vertical bars highlight the location of promoters of activated RUNX3 target genes and enhancers bound by activated RUNX3, respectively. Only activated RUNX3-mediated loops interacting with the viewpoint are displayed. Expression of RUNX3 target genes was measured by qRT-PCR (right panels). (F) Scheme showing the action mechanism of Vorinostat where RUNX3 acetylation-mediated enhancer activation increases immune function-related gene expression via long-range chromatin interactions.
REFERENCES
  1. Vivier E, Tomasello E, Baratin M, Walzer T and Ugolini S (2008) Functions of natural killer cells. Nat Immunol 9, 503-510
    Pubmed CrossRef
  2. Caligiuri MA (2008) Human natural killer cells. Blood 112, 461-469
    Pubmed KoreaMed CrossRef
  3. Kee BL, Morman RE and Sun M (2020) Transcriptional regulation of natural killer cell development and maturation. Adv Immunol 146, 1-28
    Pubmed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  7. Meza Guzman LG, Keating N and Nicholson SE (2020) Natural killer cells: tumor surveillance and signaling. Cancers (Basel) 12, 952
    Pubmed KoreaMed CrossRef
  8. Wang D and Malarkannan S (2020) Transcriptional regulation of natural killer cell development and functions. Cancers (Basel) 12, 1591
    Pubmed KoreaMed CrossRef
  9. Bi J and Wang X (2020) Molecular regulation of NK cell maturation. Front Immunol 11, 1945
    Pubmed KoreaMed CrossRef
  10. Spitz F and Furlong EE (2012) Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 13, 613-626
    Pubmed CrossRef
  11. Levine M and Tjian R (2003) Transcription regulation and animal diversity. Nature 424, 147-151
    Pubmed CrossRef
  12. Holliday R (2006) Epigenetics: a historical overview. Epigenetics 1, 76-80
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  14. Santosa EK, Lau CM, Sahin M, Leslie CS and Sun JC (2023) 3D chromatin dynamics during innate and adaptive immune memory acquisition. bioRxiv
    Pubmed KoreaMed CrossRef
  15. Grunstein M (1997) Histone acetylation in chromatin structure and transcription. Nature 389, 349-352
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed CrossRef
  18. Siegel D, Hussein M, Belani C et al (2009) Vorinostat in solid and hematologic malignancies. J Hematol Oncol 2, 31
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  24. Misteli T (2020) The self-organizing genome: principles of genome architecture and function. Cell 183, 28-45
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  28. Boto P, Csuth TI and Szatmari I (2018) RUNX3-mediated immune cell development and maturation. Crit Rev Immunol 38, 63-78
    Pubmed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef
  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
    Pubmed CrossRef
  38. Mevel R, Draper JE, Lie ALM, Kouskoff V and Lacaud G (2019) RUNX transcription factors: orchestrators of development. Development 146, dev148296
    Pubmed CrossRef
  39. Brillantes M and Beaulieu AM (2019) Transcriptional control of natural killer cell differentiation. Immunology 156, 111-119
    Pubmed KoreaMed CrossRef
  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
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles
  • CrossRef (0)

Author ORCID Information

Funding Information
  • National Research Foundation of Korea
      10.13039/501100003725
      2018M3A9D3079290, 2020R1A2C2013258, 2022M3A9B6017424
  • Ministry of Science, ICT and Future Planning
      10.13039/501100003621
      2018M3A9D3079290, 2020R1A2C2013258, 2022M3A9B6017424

Collections

Services
Social Network Service

e-submission

Archives