A novel small molecule ZYZ384 targeting SMYD3 for hepatocellular carcinoma via reducing H3K4 trimethylation of the Rac1 promoter

Qian Ding , Jianghong Cai , Li Jin , Wei Hu , Wu Song , Peter Rose , Zhiyuan Tang , Yangyang Zhan , Leilei Bao , Wei Lei , Yi Zhun Zhu

MedComm ›› 2024, Vol. 5 ›› Issue (10) : e711

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (10) : e711 DOI: 10.1002/mco2.711
ORIGINAL ARTICLE

A novel small molecule ZYZ384 targeting SMYD3 for hepatocellular carcinoma via reducing H3K4 trimethylation of the Rac1 promoter

Author information +
History +
PDF

Abstract

SMYD3 (SET and MYND domain-containing 3) is a histone lysine methyltransferase highly expressed in different types of cancer(s) and is a promising epigenetic target for developing novel antitumor therapeutics. No selective inhibitors for this protein have been developed for cancer treatment. Therefore, the current study describes developing and characterizing a novel small molecule ZYZ384 screened and synthesized based on SMYD3 structure. Virtual screening was initially used to identify a lead compound and followed up by modification to get the novel molecules. Several technologies were used to facilitate compound screening about these novel molecules’ binding affinities and inhibition activities with SMYD3 protein; the antitumor activity has been assessed in vitro using various cancer cell lines. In addition, a tumor-bearing nude mice model was established, and the activity of the selected molecule was determined in vivo. Both RNA-seq and chip-seq were performed to explore the antitumor mechanism. This work identified a novel small molecule ZYZ384 targeting SMYD3 with antitumor activity and impaired hepatocellular carcinoma tumor growth by reducing H3K4 trimethylation of the Rac1 promoter triggering the tumor cell cycle arrest through the AKT pathway.

Keywords

antitumor / H3K4me3 / Rac1 promoter / small molecule / SMYD3 / ZYZ384

Cite this article

Download citation ▾
Qian Ding, Jianghong Cai, Li Jin, Wei Hu, Wu Song, Peter Rose, Zhiyuan Tang, Yangyang Zhan, Leilei Bao, Wei Lei, Yi Zhun Zhu. A novel small molecule ZYZ384 targeting SMYD3 for hepatocellular carcinoma via reducing H3K4 trimethylation of the Rac1 promoter. MedComm, 2024, 5(10): e711 DOI:10.1002/mco2.711

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Chen ZQ, Xie H, Hu MM, et al. Recent progress in treatment of hepatocellular carcinoma. Am J Cancer Res. 2020; 10(9): 2993-3036.

[2]

Torre LA, Siegel RL, Ward EM, Jemal A. Global cancer incidence and mortality rates and trends—an update. Cancer Epidemiol Biomark. 2016; 25(1): 16-27.

[3]

Costa FF. Non-coding RNAs, epigenetics and complexity. Gene. 2008; 410(1): 9-17.

[4]

Hamamoto R, Furukawa Y, Morita M, et al. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol. 2004; 6(8): 731-740.

[5]

Mazur PK, Reynoird N, Khatri P, et al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature. 2014; 510(7504): 283-287.

[6]

Sarris ME, Moulos P, Haroniti A, Giakountis A, Talianidis I. Smyd3 is a transcriptional potentiator of multiple cancer-promoting genes and required for liver and colon cancer development. Cancer Cell. 2016; 29(3): 354-366.

[7]

Hamamoto R, Silva FP, Tsuge M, et al. Enhanced SMYD3 expression is essential for the growth of breast cancer cells. Cancer Sci. 2006; 97(2): 113-118.

[8]

Paladino D, Yue PB, Furuya H, Acoba J, Rosser CJ, Turkson J. A novel nuclear Src and p300 signaling axis controls migratory and invasive behavior in pancreatic cancer. Oncotarget. 2016; 7(6): 7253-7267.

[9]

Vieira FQ, Costa-Pinheiro P, Ramalho-Carvalho J, et al. Deregulated expression of selected histone methylases and demethylases in prostate carcinoma. Endocr-Relat Cancer. 2014; 21(1): 51-61.

[10]

Shen B, Tan MY, Mu XY, et al. Upregulated SMYD3 promotes bladder cancer progression by targeting BCLAF1 and activating autophagy. Tumor Biol. 2016; 37(6): 7371-7381.

[11]

Liu Y, Liu HG, Luo XG, Deng JY, Pan Y, Liang H. Overexpression of SMYD3 and matrix metalloproteinase-9 are associated with poor prognosis of patients with gastric cancer. Tumor Biol. 2015; 36(6): 4377-4386.

[12]

Dai B, Wan WQ, Zhang P, et al. SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity. Oncol Rep. 2015; 34(5): 2722-2730.

[13]

Zhu Y, Zhu MX, Zhang XD, et al. SMYD3 stimulates EZR and LOXL2 transcription to enhance proliferation, migration, and invasion in esophageal squamous cell carcinoma. Hum Pathol. 2016; 52: 153-163.

[14]

Oliveira-Santos W, Rabello DA, Lucena-Araujo AR, et al. Residual expression of SMYD2 and SMYD3 is associated with the acquisition of complex karyotype in chronic lymphocytic leukemia. Tumor Biol. 2016; 37(7): 9473-9481.

[15]

Wang SZ, Luo XG, Shen J, Zou JN, Lu YH, Xi T. Knockdown of SMYD3 by RNA interference inhibits cervical carcinoma cell growth and invasion in vitro. BMB Rep. 2008; 41(4): 294-299.

[16]

Peserico A, Germani A, Sanese P, et al. A SMYD3 small-molecule inhibitor impairing cancer cell growth. J Cell Physiol. 2015; 230(10): 2447-2460.

[17]

Van Aller GS, Graves AP, Elkins PA, et al. Structure-based design of a novel SMYD3 inhibitor that bridges the SAM-and MEKK2-binding pockets. Structure. 2016; 24(5): 774-781.

[18]

Mitchell LH, Boriack-Sjodin PA, Smith S, et al. Novel oxindole sulfonamides and sulfamides: EPZ031686, the first orally bioavailable small molecule SMYD3 inhibitor. ACS Med Chem Lett. 2016; 7(2): 134-138.

[19]

Zhu HP, Chai J, Qin R, et al. Discovery of tetrahydrofuranyl spirooxindole-based SMYD3 inhibitors against gastric cancer via inducing lethal autophagy. Eur J Med Chem. 2023; 246: 115009.

[20]

Parenti MD, Naldi M, Manoni E, et al. Discovery of the 4-aminopiperidine-based compound EM127 for the site-specific covalent inhibition of SMYD3. Eur J Med Chem. 2022; 243: 114683.

[21]

Maia EHB, Assis LC, de Oliveira TA, da Silva AM, Taranto AG. Structure-based virtual screening: from classical to artificial intelligence. Front Chem. 2020; 8: 343.

[22]

Ma HC, Zou T, Li H, Cheng HM. The interaction of sodium dodecyl sulfate with trypsin: multi-spectroscopic analysis, molecular docking, and molecular dynamics simulation. Int J Biol Macromol. 2020; 162: 1546-1554.

[23]

Zhong Z, Vong CT, Chen F, et al. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: helping to fight against cancer via multiple targets. Med Res Rev. 2022; 42(3): 1246-1279.

[24]

Chan KI, Zhang S, Li G, et al. MYC oncogene: a druggable target for treating cancers with natural products. Aging Dis. 2024; 15(2): 640-697.

[25]

Fei X, Ma Y, Liu XW, Meng ZX. Overexpression of SMYD3 is predictive of unfavorable prognosis in hepatocellular carcinoma. Tohoku J Exp Med. 2017; 243(3): 219-226.

[26]

Rajajeyabalachandran G, Kumar S, Murugesan T, et al. Therapeutical potential of deregulated lysine methyltransferase SMYD3 as a safe target for novel anticancer agents. Expert Opin Ther Targets. 2017; 21(2): 145-157.

[27]

Gradl S, Steuber H, Weiske J, et al. Discovery of the SMYD3 inhibitor BAY-6035 using thermal shift assay (TSA)-based high-throughput screening. Slas Discov. 2021; 26(8): 947-960.

[28]

Alnabulsi SM, Al-shar’i NA. Hit identification of SMYD3 enzyme inhibitors using structure-based pharmacophore modeling. Future Med Chem. 2019; 11(10): 1107-1117.

[29]

Sun JX, Li ZB, Yang N. Mechanism of the conformational change of the protein methyltransferase SMYD3: a molecular dynamics simulation study. Int J Mol Sci. 2021; 22(13): 7185.

[30]

Peña PV, Davrazou F, Shi X, et al. Molecular mechanism of histone H3K4me3 recognition by plant homeodomain of ING2. Nature. 2006; 442: 100-103.

[31]

Feng W, Yonezawa M, Ye J, Jenuwein T, Grummt I. PHF8 activates transcription of rRNA genes through H3K4me3 binding and H3K9me1/2 demethylation. Nat Struct Mol Biol. 2010; 17(4): 445-450.

[32]

Cheung I, Shulha HP, Jiang Y, et al. Developmental regulation and individual differences of neuronal H3K4me3 epigenomes in the prefrontal cortex. Proc Natl Acad Sci USA. 2010; 107(19): 8824-8829.

[33]

Rathinam R, Berrier A, Alahari SK. Role of Rho GTPases and their regulators in cancer progression. Front Biosci (Landmark Ed). 2011; 16(7): 2561-2571.

[34]

Ehrlich JS, Hansen MD, Nelson WJ. Spatio-temporal regulation of Rac1 localization and lamellipodia dynamics during epithelial cell-cell adhesion. Dev Cell. 2002; 3(2): 259-270.

[35]

Saci A, Cantley LC, Carpenter CL. Rac1 regulates the activity of mTORC1 and mTORC2 and controls cellular size. Mol Cell. 2011; 42(1): 50-61.

[36]

Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 2016; 67: 11-28.

[37]

Huang HB, Zhang GG, Zhou YQ, et al. Reverse screening methods to search for the protein targets of chemopreventive compounds. Front Chem. 2018; 6: 138.

[38]

Khan S, Alhumaydhi FA, Ibrahim MM, et al. Recent advances and therapeutic journey of Schiff base complexes with selected metals (Pt, Pd, Ag, Au) as potent anticancer agents: a review. Anticancer Agents Med Chem. 2022; 22(18): 3086-3096.

[39]

Matela G. Schiff bases and complexes: a review on anti-cancer activity. Anticancer Agents Med Chem. 2020; 20(16): 1908-1917.

[40]

Siraj FM, Natarajan S, Huq MA, Kim YJ, Yang DC. Structural investigation of ginsenoside Rf with PPARgamma major transcriptional factor of adipogenesis and its impact on adipocyte. J Ginseng Res. 2015; 39(2): 141-147.

[41]

Xu T, Tian WY, Zhang Q, et al. Novel 1, 3, 4-thiadiazole/oxadiazole-linked honokiol derivatives suppress cancer via inducing PI3K/Akt/mTOR-dependent autophagy. Bioorg Chem. 2021; 115: 105257.

[42]

Messias A, Santos DES, Pontes FJS, Lima FS, Soares TA. Out of sight, out of mind: the effect of the equilibration protocol on the structural ensembles of charged glycolipid bilayers. Molecules. 2020; 25(21): 5120.

[43]

Zhu SD, Khan MA, Wang FY, Bano Z, Xia MZ. Exploration of adsorption mechanism of 2-phosphonobutane-1, 2, 4-tricarboxylic acid onto kaolinite and montmorillonite via batch experiment and theoretical studies. J Hazard Mater. 2021; 403: 123810.

[44]

Loschwitz J, Jackering A, Keutmann M, Olagunju M, Olubiyi OO, Strodel B. Dataset of AMBER force field parameters of drugs, natural products and steroids for simulations using GROMACS. Data Brief. 2021; 35: 106948.

[45]

Boateng HA. Periodic Coulomb tree method: an alternative to parallel particle mesh Ewald. J Chem Theory Comput. 2020; 16(1): 7-17.

[46]

Sinha S, Wang SM. Classification of VUS and unclassified variants in BRCA1 BRCT repeats by molecular dynamics simulation. Comput Struct Biotechnol. 2020; 18: 723-736.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

153

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/