Generic placeholder image

Current Biotechnology

Editor-in-Chief

ISSN (Print): 2211-5501
ISSN (Online): 2211-551X

Research Article

The Klf6 Super-enhancer Determines Klf6 Sensitivity to BRD4 Inhibitors in Human Hepatoma (HepG2) Cells

Author(s): KumChol Ri*, MyongRyong Ri, YongJae Song, KyuHwan Kim and Chol Kim

Volume 9, Issue 3, 2020

Page: [209 - 218] Pages: 10

DOI: 10.2174/2211550109999200802154246

Price: $65

Abstract

Background: The Klf6 gene, belonging to Krüppel-like family of C2H2 zinc finger transcription factors, is strongly associated with tumor formation through high somatic mutations in carcinomas of the prostate, liver, colon, stomach, lung, neck, pituitary gland and nervous system. Recently, Klf6 super-enhancer which strongly regulates Klf6 gene expression has been identified, and the function of Klf6 super-enhancer which regulates cell growth was studied.

Objective: The development of inhibitors targeting BRD4-binding super-enhancers is a potential target therapeutic strategy for tumor therapy. However, the suppression of Klf6 super-enhancer function by BRD4 inhibitors is not known.

Methods: CRISPR-Cas9 editing technique was used for the Klf6 super-enhancer deletion experiment, and the expression levels of several genes for cell clones were detected by qRT-PCR analysis and Western blotting. Cell proliferation assay was applied to evaluate the functional role of Klf6 super-enhancer using several BRD4 inhibitors. The interaction of several BRD4 inhibitors against the target protein was analyzed by molecular docking simulation.

Results: JQ-1, a human BRD4 inhibitor, inhibited Klf6 gene expression and its activity in HepG2 cells in a time and dose-dependent manner while simultaneously inhibiting cell growth. Besides, BETd-246, a human BRD4 inhibitor, strongly inhibited Klf6 gene expression, significantly inhibited cell growth, and exhibited higher efficacy than JQ-1. Molecular docking studies revealed that some key residues were critical for ligand-receptor interactions by forming hydrogen bonds with ligands (JQ-1: ASN140, BETd-246: ASN140, TYR106, LYS65, GLN58, MET105, and MET53).

Conclusion: Our findings suggest that KLF6 is regulated by Klf6 super-enhancer and the targeting of Klf6 super-enhancer by BRD4 inhibitors may be an effective therapeutic strategy for liver cancer therapy.

Keywords: Super-enhancer, Klf6 gene, BRD4 inhibitors, CRISPR, JQ-1, BETd-246, HepG2.

Graphical Abstract
[1]
Narla G, DiFeo A, Yao S, et al. Targeted inhibition of the KLF6 splice variant, KLF6 SV1, suppresses prostate cancer cell growth and spread. Cancer Res 2005; 65(13): 5761-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-0217] [PMID: 15994951]
[2]
Banck MS, Beaven SW, Narla G, Walsh MJ, Friedman SL, Beutler AS. KLF6 degradation after apoptotic DNA damage. FEBS Lett 2006; 580(30): 6981-6.
[http://dx.doi.org/10.1016/j.febslet.2006.10.077] [PMID: 17113081]
[3]
Chen C, Hyytinen E-R, Sun X, et al. Deletion, mutation, and loss of expression of Klf6 in human prostate cancer. Am J Pathol 2003; 162: 1349-54.
[http://dx.doi.org/10.1016/S0002-9440(10)63930-2] [PMID: 12651626]
[4]
Reeves HL, Narla G, Ogunbiyi O, et al. Kruppel-like factor 6 (KLF6) is a tumor-suppressor gene frequently inactivated in colorectal cancer. Gastroenterology 2004; 126(4): 1090-103.
[http://dx.doi.org/10.1053/j.gastro.2004.01.005] [PMID: 15057748]
[5]
Benzeno S, Narla G, Allina J, et al. Cyclin-dependent kinase inhibition by the KLF6 tumor suppressor protein through interaction with cyclin D1. Cancer Res 2004; 64(11): 3885-91.
[http://dx.doi.org/10.1158/0008-5472.CAN-03-2818] [PMID: 15172998]
[6]
Ri K, Kim JS, Kim C. Identification of Klf6-related super enhancer in human hepatoma (HepG2) cells by CRISPR technique. Genet Mol Res 2017; 16(4): gmr16039841.
[http://dx.doi.org/10.4238/gmr16039841]
[7]
Marc R. Mansour, Brian J. Abraham, Lars Anders, Alla Berezovskaya. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 2014; 346: 6215.
[8]
Didych DA, Tyulkina DV, Pleshkan VV, Alekseenko IV, Sverdlov ED. Super-enhancers. Are they regulators of regulatory genes of development and cancer? Mol Biol (Mosk) 2015; 49(6): 915-22.
[PMID: 26710770]
[9]
Suzuki HI, Young RA, Sharp PA. Super-enhancer-mediated RNA processing revealed by integrative microRNA network analysis. Cell 2017; 168(6): 1000-1014.e15.
[http://dx.doi.org/10.1016/j.cell.2017.02.015] [PMID: 28283057]
[10]
Yang R, Wu Y, Ming Y, Xu Y. A super-enhancer maintains homeostatic expression of regnase-1. Cell 2018; 30(669): 35-41.
[11]
Stratton MS, Lin CY, Anand P, et al. Signal-dependent recruitment of BRD4 to cardiomyocyte super-enhancers is suppressed by a microRNA. Cell Rep 2016; 16(5): 1366-78.
[http://dx.doi.org/10.1016/j.celrep.2016.06.074] [PMID: 27425608]
[12]
Zou Q, Liang Y, Luo H, Yu W. MiRNA-mediated RNA by targeting enhancers. Adv Exp Med Biol 2017; 983: 113-25.
[http://dx.doi.org/10.1007/978-981-10-4310-9_8] [PMID: 28639195]
[13]
Lovén J, Hoke HA, Lin CY, et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 2013; 153(2): 320-34.
[http://dx.doi.org/10.1016/j.cell.2013.03.036] [PMID: 23582323]
[14]
Whyte WA, Orlando DA, Hnisz D, et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 2013; 153(2): 307-19.
[http://dx.doi.org/10.1016/j.cell.2013.03.035] [PMID: 23582322]
[15]
Sengupta S, George RE. Super-enhancer-driven transcriptional dependencies in cancer. Trends Cancer 2017; 3(4): 269-81.
[http://dx.doi.org/10.1016/j.trecan.2017.03.006] [PMID: 28718439]
[16]
Wang Y, Zhang T, Kwiatkowski N, et al. CDK7-dependent transcriptional addiction in triple-negative breast cancer. Cell 2015; 163(1): 174-86.
[http://dx.doi.org/10.1016/j.cell.2015.08.063] [PMID: 26406377]
[17]
Filippakopoulos P, Qi J, Picaud S, et al. Selective inhibition of BET bromodomains. Nature 2010; 468(7327): 1067-73.
[http://dx.doi.org/10.1038/nature09504] [PMID: 20871596]
[18]
Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011; 146(6): 904-17.
[http://dx.doi.org/10.1016/j.cell.2011.08.017] [PMID: 21889194]
[19]
Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci USA 2011; 108(40): 16669-74.
[http://dx.doi.org/10.1073/pnas.1108190108] [PMID: 21949397]
[20]
Shi J, Wang Y, Zeng L, et al. Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell 2014; 25(2): 210-25.
[http://dx.doi.org/10.1016/j.ccr.2014.01.028] [PMID: 24525235]
[21]
Liu Z, Wang P, Chen H, et al. Drug discovery targeting bromodomain-containing protein 4. J Med Chem 2017; 60(11): 4533-58.
[http://dx.doi.org/10.1021/acs.jmedchem.6b01761] [PMID: 28195723]
[22]
Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: A novel epigenetic approach. Ann Oncol 2017; 28(8): 1776-87.
[http://dx.doi.org/10.1093/annonc/mdx157] [PMID: 28838216]
[23]
Borck PC, Guo LW, Plutzky J. Lian-Wang Guo, Jorge Plutzky. BET epigenetic reader proteins in cardiovascular transcriptional programs. Circ Res 2020; 126(9): 1190-208.
[http://dx.doi.org/10.1161/CIRCRESAHA.120.315929] [PMID: 32324495]
[24]
da Motta LL, Ledaki I, Purshouse K, et al. The BET inhibitor JQ1 selectively impairs tumour response to hypoxia and downregulates CA9 and angiogenesis in triple negative breast cancer. Oncogene 2017; 36(1): 122-32.
[http://dx.doi.org/10.1038/onc.2016.184] [PMID: 27292261]
[25]
Shu S, Lin CY, He HH, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 2016; 529(7586): 413-7.
[http://dx.doi.org/10.1038/nature16508] [PMID: 26735014]
[26]
Vázquez R, Riveiro ME, Astorgues-Xerri L, et al. The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus. Oncotarget 2017; 8(5): 7598-613.
[http://dx.doi.org/10.18632/oncotarget.13814] [PMID: 27935867]
[27]
Bai L, Zhou B, Yang CY, et al. Chao-Yie Yang, Jiao Ji, Donna McEachern. Targeted degradation of BET proteins in triple-negative breast cancer. Cancer Res 2017; 77(9): 2476-87.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-2622] [PMID: 28209615]
[28]
Ri K, Kim K, Kong S, Ri J. The disruption of Klf6-related super- enhancer induces growth inhibition and apoptosis in human hepG2 cells. Genet Mol Res 2018; 17(1): gmr16039888.
[29]
Ri K, Kim C, Pak C, Ri P, Om H. The Klf6 super enhancer modulates cell proliferation via MiR-1301 in human hepatoma cells. MicroRNA 2020; 9: 64-9.
[30]
Ri K, Kim C, Choe SI, So JH. O SH. The Klf6-related super enhancer regulates Klf6-SV2 expression-mediated proliferation in human hepatoma (HepG2) cells. Beni-Suef Univ J of Appl Sci 2019; 8: 10.
[http://dx.doi.org/10.1186/s43088-019-0001-4]
[31]
Jain AN. Surflex-Dock 2.1: Robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comput Aided Mol Des 2007; 21(5): 281-306.
[http://dx.doi.org/10.1007/s10822-007-9114-2] [PMID: 17387436]
[32]
Schoch GA, D’Arcy B, Stihle M, et al. Molecular switch in the glucocorticoid receptor: Active and passive antagonist conformations. J Mol Biol 2010; 395(3): 568-77.
[http://dx.doi.org/10.1016/j.jmb.2009.11.011] [PMID: 19913032]
[33]
Clark M, Cramer RD, Vanopdenbosch N. Validation of the general-purpose Tripos 5.2 force field. J Comput Chem 1989; 10: 982-1012.
[http://dx.doi.org/10.1002/jcc.540100804]
[34]
Pott S, Lieb JD. What are super-enhancers? Nat Genet 2015; 47(1): 8-12.
[http://dx.doi.org/10.1038/ng.3167] [PMID: 25547603]
[35]
Chipumuro E, Marco E, Christensen CL, et al. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 2014; 159(5): 1126-39.
[http://dx.doi.org/10.1016/j.cell.2014.10.024] [PMID: 25416950]
[36]
Shin HY. Targeting super-enhancers for disease treatment and diagnosis. Mol Cells 2018; 41(6): 506-14.
[PMID: 29754476]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy