2,136
Views
2
CrossRef citations to date
0
Altmetric
Research Articles

Identification of a potential DNA methyltransferase (DNMT) inhibitor

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 4730-4744 | Received 13 Jan 2023, Accepted 01 Jun 2023, Published online: 09 Jul 2023

References

  • Agrawal, K., Das, V., Vyas, P., & Hajdúch, M. (2018). Nucleosidic DNA demethylating epigenetic drugs – a comprehensive review from discovery to clinic. Pharmacology & Therapeutics, 188, 45–79. https://doi.org/10.1016/j.pharmthera.2018.02.006
  • Ahmed, W. S., Philip, A. M., & Biswas, K. H. (2021). Decreased interfacial dynamics caused by the N501Y mutation in the SARS-CoV-2 S1 spike: ACE2 complex. Front Mol Biosci., 9, 846996. https://doi.org/10.1101/2021.01.07.425307
  • Altamash, T., Ahmed, W., Rasool, S., & Biswas, K. H. (2021). Intracellular ionic strength sensing using NanoLuc. International Journal of Molecular Sciences, 22(2), 677. https://doi.org/10.3390/ijms22020677
  • Anwar, S. L., & Lehmann, U. (2014). DNA methylation, microRNAs, and their crosstalk as potential biomarkers in hepatocellular carcinoma. World Journal of Gastroenterology, 20(24), 7894–7913. https://doi.org/10.3748/wjg.v20.i24.7894
  • Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports, 7, 42717. https://doi.org/10.1038/srep42717
  • Essmann, U., Perera, L., Berkowitz, M. L., Darden, T., Lee, H., & Pedersen, L. G. (1995). A smooth particle mesh Ewald method. The Journal of Chemical Physics, 103(19), 8577–8593. https://doi.org/10.1063/1.470117
  • Esteller, M. (2008). Epigenetics in cancer. The New England Journal of Medicine, 358(11), 1148–1159. https://doi.org/10.1056/NEJMra072067
  • Feller, S. E., Zhang, Y., Pastor, R. W., & Brooks, B. R. (1995). Constant pressure molecular dynamics simulation: The Langevin piston method. The Journal of Chemical Physics, 103(11), 4613–4621. https://doi.org/10.1063/1.470648
  • Gao, F., & Das, S. K. (2014). Epigenetic regulations through DNA methylation and hydroxymethylation: Clues for early pregnancy in decidualization. Biomolecular Concepts, 5(2), 95–107. https://doi.org/10.1515/bmc-2013-0036
  • Gao, L., Emperle, M., Guo, Y., Grimm, S. A., Ren, W., Adam, S., Uryu, H., Zhang, Z.-M., Chen, D., Yin, J., Dukatz, M., Anteneh, H., Jurkowska, R. Z., Lu, J., Wang, Y., Bashtrykov, P., Wade, P. A., Wang, G. G., Jeltsch, A., & Song, J. (2020). Comprehensive structure-function characterization of DNMT3B and DNMT3A reveals distinctive de novo DNA methylation mechanisms. Nature Communications, 11(1), 3355. https://doi.org/10.1038/s41467-020-17109-4
  • Geethakumari, A. M., Ahmed, W. S., Rasool, S., Fatima, A., Nasir Uddin, S. M., Aouida, M., & Biswas, K. H. (2022). A genetically encoded BRET-based SARS-CoV-2 Mpro protease activity sensor. Communications Chemistry, 5(1), 117. https://doi.org/10.1038/s42004-022-00731-2
  • Grant, B. J., Rodrigues, A. P. C., ElSawy, K. M., McCammon, J. A., & Caves, L. S. D. (2006). Bio3d: An R package for the comparative analysis of protein structures. Bioinformatics, 22(21), 2695–2696. https://doi.org/10.1093/bioinformatics/btl461
  • Horton, J. R., Pathuri, S., Wong, K., Ren, R., Rueda, L., Fosbenner, D. T., Heerding, D. A., McCabe, M. T., Pappalardi, M. B., Zhang, X., King, B. W., & Cheng, X. (2022). Structural characterization of dicyanopyridine containing DNMT1-selective, non-nucleoside inhibitors. Structure, 30(6), 793–802.e5. https://doi.org/10.1016/j.str.2022.03.009
  • Huang, J., Rauscher, S., Nawrocki, G., Ran, T., Feig, M., de Groot, B. L., Grubmüller, H., & MacKerell, A. D. (2017). CHARMM36m: An improved force field for folded and intrinsically disordered proteins. Nature Methods, 14(1), 71–73. https://doi.org/10.1038/nmeth.4067
  • Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular dynamics. Journal of Molecular Graphics, 14(1), 33–38. https://doi.org/10.1016/0263-7855(96)00018-5
  • Irwin, J. J., & Shoichet, B. K. (2005). ZINC − a free database of commercially available compounds for virtual screening. Journal of Chemical Information and Modeling, 45(1), 177–182. https://doi.org/10.1021/ci049714+
  • Jinawath, A., Miyake, S., Yanagisawa, Y., Akiyama, Y., & Yuasa, Y. (2005). Transcriptional regulation of the human DNA methyltransferase 3A and 3B genes by Sp3 and Sp1 zinc finger proteins. The Biochemical Journal, 385(Pt 2), 557–564. https://doi.org/10.1042/BJ20040684
  • Jithesh, P. V., Risk, J. M., Schache, A. G., Dhanda, J., Lane, B., Liloglou, T., & Shaw, R. J. (2013). The epigenetic landscape of oral squamous cell carcinoma. British Journal of Cancer, 108(2), 370–379. https://doi.org/10.1038/bjc.2012.568
  • Jo, S., Kim, T., Iyer, V. G., & Im, W. (2008). CHARMM-GUI: A web-based graphical user interface for CHARMM. Journal of Computational Chemistry, 29(11), 1859–1865. https://doi.org/10.1002/jcc.20945
  • Jones, P. A., & Taylor, S. M. (1980). Cellular differentiation, cytidine analogs and DNA methylation. Cell, 20(1), 85–93. https://doi.org/10.1016/0092-8674(80)90237-8
  • Jorgensen, W. L., Chandrasekhar, J., Madura, J. D., Impey, R. W., & Klein, M. L. (1983). Comparison of simple potential functions for simulating liquid water. Journal of Chemical Physics. 79(2), 926–935. https://doi.org/10.1063/1.445869
  • Kumar, S., Chinnusamy, V., & Mohapatra, T. (2018). Epigenetics of modified DNA bases: 5-Methylcytosine and beyond. Frontiers in Genetics, 9, 640. https://doi.org/10.3389/fgene.2018.00640
  • Kuna, L., Bozic, I., Kizivat, T., Bojanic, K., Mrso, M., Kralj, E., Smolic, R., Wu, G. Y., & Smolic, M. (2018). Models of drug induced liver injury (DILI) – current issues and future perspectives. Current Drug Metabolism, 19(10), 830–838. https://doi.org/10.2174/1389200219666180523095355
  • Lopez, M., Gilbert, J., Contreras, J., Halby, L., & Arimondo, P. B. (2022). Inhibitors of DNA Methylation. Advances in Experimental Medicine and Biology, 1389, 471–513. https://doi.org/10.1007/978-3-031-11454-0_17
  • Lyko, F. (2018). The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nature Reviews-Genetics, 19(2), 81–92. https://doi.org/10.1038/nrg.2017.80
  • Moore, L. D., Le, T., & Fan, G. (2013). DNA methylation and its basic function. Neuropsychopharmacology, 38(1), 23–38. https://doi.org/10.1038/npp.2012.112
  • Pappalardi, M. B., Keenan, K., Cockerill, M., Kellner, W. A., Stowell, A., Sherk, C., Wong, K., Pathuri, S., Briand, J., Steidel, M., Chapman, P., Groy, A., Wiseman, A. K., McHugh, C. F., Campobasso, N., Graves, A. P., Fairweather, E., Werner, T., Raoof, A., … McCabe, M. T. (2021). Discovery of a first-in-class reversible DNMT1-selective inhibitor with improved tolerability and efficacy in acute myeloid leukemia. Nature Cancer, 2(10), 1002–1017. https://doi.org/10.1038/s43018-021-00249-x
  • Phillips, J. C., Braun, R., Wang, W., Gumbart, J., Tajkhorshid, E., Villa, E., Chipot, C., Skeel, R. D., Kalé, L., & Schulten, K. (2005). Scalable molecular dynamics with NAMD. Journal of Computational Chemistry, 26(16), 1781–1802. https://doi.org/10.1002/jcc.20289
  • Pires, D. E. V., Blundell, T. L., & Ascher, D. B. (2015). pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. Journal of Medicinal Chemistry, 58(9), 4066–4072. https://doi.org/10.1021/acs.jmedchem.5b00104
  • Reddy, A. S., & Zhang, S. (2013). Polypharmacology: Drug discovery for the future. Expert Review of Clinical Pharmacology, 6(1), 41–47. https://doi.org/10.1586/ecp.12.74]
  • Roll, J. D., Rivenbark, A. G., Jones, W. D., & Coleman, W. B. (2008). DNMT3b overexpression contributes to a hypermethylator phenotype in human breast cancer cell lines. Molecular Cancer, 7, 15. https://doi.org/10.1186/1476-4598-7-15
  • Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch, T., Preibisch, S., Rueden, C., Saalfeld, S., Schmid, B., Tinevez, J.-Y., White, D. J., Hartenstein, V., Eliceiri, K., Tomancak, P., & Cardona, A. (2012). Fiji: An open-source platform for biological-image analysis. Nature Methods, 9(7), 676–682. https://doi.org/10.1038/nmeth.2019
  • Scholz, C., Knorr, S., Hamacher, K., & Schmidt, B. (2015). DOCKTITE-a highly versatile step-by-step workflow for covalent docking and virtual screening in the molecular operating environment. Journal of Chemical Information and Modeling, 55(2), 398–406. https://doi.org/10.1021/ci500681r
  • Sheikh, A., Al-Taher, A., Al-Nazawi, M., Al-Mubarak, A. I., & Kandeel, M. (2020). Analysis of preferred codon usage in the coronavirus N genes and their implications for genome evolution and vaccine design. Journal of Virological Methods, 277, 113806. https://doi.org/10.1016/j.jviromet.2019.113806
  • Skjærven, L., Yao, X.-Q., Scarabelli, G., & Grant, B. J. (2014). Integrating protein structural dynamics and evolutionary analysis with Bio3D. BMC Bioinformatics, 15(1), 399. https://doi.org/10.1186/s12859-014-0399-6
  • Steinbach, P. J., & Brooks, B. R. (1994). New spherical-cutoff methods for long-range forces in macromolecular simulation. Journal of Computational Chemistry, 15(7), 667–683. https://doi.org/10.1002/jcc.540150702
  • Sterling, T., & Irwin, J. J. (2015). ZINC 15 – Ligand discovery for everyone. Journal of Chemical Information and Modeling, 55(11), 2324–2337. https://doi.org/10.1021/acs.jcim.5b00559
  • Stresemann, C., & Lyko, F. (2008). Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. International Journal of Cancer, 123(1), 8–13. https://doi.org/10.1002/ijc.23607
  • Vilar, S., Cozza, G., & Moro, S. (2008). Medicinal chemistry and the molecular operating environment (MOE): Application of QSAR and molecular docking to drug discovery. Current Topics in Medicinal Chemistry, 8(18), 1555–1572. https://doi.org/10.2174/156802608786786624
  • Wong, K. K. (2021). DNMT1: A key drug target in triple-negative breast cancer. Seminars in Cancer Biology, 72, 198–213. https://doi.org/10.1016/j.semcancer.2020.05.010
  • Wong, K. K., Lawrie, C. H., & Green, T. M. (2019). Oncogenic roles and inhibitors of DNMT1, DNMT3A, and DNMT3B in acute myeloid leukaemia. Biomarker Insights, 14, 1177271919846454. https://doi.org/10.1177/1177271919846454
  • Xie, T., Yu, J., Fu, W., Wang, Z., Xu, L., Chang, S., Wang, E., Zhu, F., Zeng, S., Kang, Y., & Hou, T. (2019). Insight into the selective binding mechanism of DNMT1 and DNMT3A inhibitors: A molecular simulation study. Physical Chemistry Chemical Physics, 21(24), 12931–12947. https://doi.org/10.1039/C9CP02024A
  • Yang, H., Sun, L., Wang, Z., Li, W., Liu, G., & Tang, Y. (2018). ADMETopt: A web server for ADMET optimization in drug design via scaffold hopping. Journal of Chemical Information and Modeling, 58(10), 2051–2056. https://doi.org/10.1021/acs.jcim.8b00532
  • Yoshikawa, N., & Hutchison, G. R. (2019). Fast, efficient fragment-based coordinate generation for Open Babel. Journal of Cheminformatics, 11(1), 49. https://doi.org/10.1186/s13321-019-0372-5
  • Zhang, W., & Xu, J. (2017). DNA methyltransferases and their roles in tumorigenesis. Biomarker Research, 5, 1. https://doi.org/10.1186/s40364-017-0081-z
  • Zhang, Z.-M., Liu, S., Lin, K., Luo, Y., Perry, J. J., Wang, Y., & Song, J. (2015). Crystal structure of human DNA methyltransferase 1. Journal of Molecular Biology, 427(15), 2520–2531. https://doi.org/10.1016/j.jmb.2015.06.001
  • Zhang, Z.-M., Lu, R., Wang, P., Yu, Y., Chen, D., Gao, L., Liu, S., Ji, D., Rothbart, S. B., Wang, Y., Wang, G. G., & Song, J. (2018). Structural basis for DNMT3A-mediated de novo DNA methylation. Nature, 554(7692), 387–391. https://doi.org/10.1038/nature25477