Generic placeholder image

Letters in Drug Design & Discovery

Editor-in-Chief

ISSN (Print): 1570-1808
ISSN (Online): 1875-628X

Letter Article

Computational Study of Lactucine and its Derivatives to Investigate its Anti-cancerous Properties Targeting Apoptosis-inducing Protein

Author(s): Mamta Arya, Apoorv Tiwari, Dev Bukhsh Singh and Gohar Taj*

Volume 21, Issue 7, 2024

Published on: 16 March, 2023

Page: [1137 - 1147] Pages: 11

DOI: 10.2174/1570180820666230224143303

Price: $65

Abstract

Background: Lactucine is related to the sesquiterpene lactone group of naturally occurring compounds and has a variety of pharmacological effects including anticancer properties found in Chicory, Wormwood, Laurus nobilis, Pyrethrum, Chamomile, etc. Lactucine has an anticancer effect which may induce apoptosis in cancerous cells and protect other cells from getting infected.

Objective: In this study, Lactucine and its derivatives were screened, and performed their in silico docking study with the proteins involved in the apoptosis-inducing effect in human leukemia cancer.

Methods: The three-dimensional structure of lactucine and its derivatives were retrieved in the SDF format. Active sites of protein structures were determined by Sitemap. LigPrep module was used for geometrical refining of chemical structures of lactucine and its derivatives. The protein preparation wizard of Maestro (Schrodinger) was used for protein preparation. From the receptor-complex structure, the cocrystallized ligands were removed from the active site position of the receptor chain. All ligands were docked using default Glide settings for a grid centered on the ligand and structure. Flexible docking was performed using the extra precision (XP) feature of Glide module. The best docking poses for the lactucine and their derivatives were selected based on their docking score. The ADMET properties of lactucine 15- oxalate have been predicted by admetSAR software.

Results: Proteins and ligands three-dimensional structures were retrieved from PDB and Pubchem databases, respectively. All lactucine derivatives suitably docked on the apoptosis-inducing proteins with ample Glide scores Lactucin 15-oxalate interacts with proteins which are responsible for apoptosis with a maximum of six H-bonds. Other types of interactions are also involved, like Pi-cation, Pi-Pi stacking, salt bridges, and halogen bonds. Protein CDK-4 has shown the highest number of H-bond (LYS142 salt bridges), ALA16, VAL14, ASP99, LYS35, TYR17, and ASN145) with the Lactucin 15-oxalate. ADMET properties of lactucin 15-oxalate met with the criteria of being eligible to be a novel drug for the treatment of human leukemia cancer. The Dock score of both the Dasatinib drug and the lactucine-15-oxalate with the apoptosis-inducing protein stipulates that the selected ligand has equitable interaction with the target proteins.

Conclusion: In this study, lactucine derivatives were docked with apoptosis-inducing proteins for the prediction of its anticancer effect. Lactucin15-oxalate has shown the highest binding affinity for the CDK-4 target and can be used as a lead compound for cancer treatment. Glide and Dock score for docking of lactucin 15-oxalate with CDK-4, well as the number of hydrogen bonding, is in agreement to use this ligand for study. These in silico results are valuable to proceed with the in vitro and in vivo studies related to the anti-cancer role of lactucin 15-oxalate.

Keywords: Molecular docking, apoptosis, cancer, lactucine, Cichorium intybus, therapeutics, CDK-4, leukemia.

Next »
Graphical Abstract
[1]
Vardiman, J.W.; Thiele, J.; Arber, D.A.; Brunning, R.D.; Borowitz, M.J.; Dinh, P.; Arefi, A.G. Antimalarial activity of lactucine and lactucopicrin: Sesquiterpene isolated from Cichorium intybus L. J. Ethnopharmacol., 2004, 95, 455-457.
[http://dx.doi.org/10.1016/j.jep.2004.06.031] [PMID: 15507374]
[2]
Kerr, J F R.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer, 1972, 26(4), 239-257.
[http://dx.doi.org/10.1038/bjc.1972.33] [PMID: 4561027]
[3]
Kiraz, Y.; Adan, A.; Kartal Yandim, M.; Baran, Y. Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol., 2016, 37(7), 8471-8486.
[http://dx.doi.org/10.1007/s13277-016-5035-9] [PMID: 27059734]
[4]
Ellis, H.; Horvitz, H.R. Genetic control of programmed cell death in the nematode C. elegans. Cell, 1986, 44(6), 817-829.
[http://dx.doi.org/10.1016/0092-8674(86)90004-8] [PMID: 3955651]
[5]
Formigli, L.; Papucci, L.; Tani, A.; Schiavone, N.; Tempestini, A.; Orlandini, G.E.; Capaccioli, S.; Zecchi Orlandini, S. Aponecrosis: Morphological and biochemical exploration of a syncretic process of cell death sharing apoptosis and necrosis. J. Cell. Physiol., 2000, 182(1), 41-49.
[http://dx.doi.org/10.1002/(SICI)1097-4652(200001)182:1<41::AID-JCP5>3.0.CO;2-7] [PMID: 10567915]
[6]
Sperandio, S.; de Belle, I.; Bredesen, D.E. An alternative, nonapoptotic form of programmed cell death. Proc. Natl. Acad. Sci. USA, 2000, 97(26), 14376-14381.
[http://dx.doi.org/10.1073/pnas.97.26.14376] [PMID: 11121041]
[7]
Debnath, J.; Brugge, J.S. Modelling glandular epithelial cancers in three-dimensional cultures. Nat. Rev. Cancer, 2005, 5(9), 675-688.
[http://dx.doi.org/10.1038/nrc1695] [PMID: 16148884]
[8]
Cooper, G.M. The Cell: A Molecular Approach; 2nd edition. Sunderland (MA): Sinauer Associates, 2000. The Development and Causes of Cancer. Available from: https://www.ncbi.nlm.nih.gov/books/NBK9963/
[9]
Savill, J.; Fadok, V. Corpse clearance defines the meaning of cell death. Nature, 2000, 407(6805), 784-788.
[http://dx.doi.org/10.1038/35037722] [PMID: 11048729]
[10]
Hengartner, M.O. The biochemistry of apoptosis. Nature, 2000, 407(6805), 770-776.
[http://dx.doi.org/10.1038/35037710] [PMID: 11048727]
[11]
D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int., 2019, 43(6), 582-592.
[http://dx.doi.org/10.1002/cbin.11137] [PMID: 30958602]
[12]
Taylor, R.C.; Cullen, S.P.; Martin, S.J. Apoptosis: Controlled demolition at the cellular level. Nat. Rev. Mol. Cell Biol., 2008, 9(3), 231-241.
[http://dx.doi.org/10.1038/nrm2312] [PMID: 18073771]
[13]
Budihardjo, I.; Oliver, H.; Lutter, M.; Luo, X.; Wang, X. Biochemical pathways of caspase activation during apoptosis. Annu. Rev. Cell Dev. Biol., 1999, 15(1), 269-290.
[http://dx.doi.org/10.1146/annurev.cellbio.15.1.269] [PMID: 10611963]
[14]
Devarajan, E.; Sahin, A.A.; Chen, J.S.; Krishnamurthy, R.R.; Aggarwal, N.; Brun, A.M.; Sapino, A.; Zhang, F.; Sharma, D.; Yang, X.H.; Tora, A.D.; Mehta, K. Down-regulation of caspase 3 in breast cancer: A possible mechanism for chemoresistance. Oncogene, 2002, 21(57), 8843-8851.
[http://dx.doi.org/10.1038/sj.onc.1206044] [PMID: 12483536]
[15]
Still, W.C.; Tempczyk, A.; Hawley, R.C.; Hendrickson, T. Semianalytical treatment of solvation for molecular mechanics and dynamics. J. Am. Chem. Soc., 1990, 112(16), 6127-6129.
[http://dx.doi.org/10.1021/ja00172a038]
[16]
Kaushik, U.; Sharma, V.; Kumar, V. Computation of pharmacophore models for the prediction of mitogen-activated protein kinase activated protein kinase-2 inhibitory activity of pyrrolopyridines. Med. Chem. Res., 2012, 21(11), 3777-3784.
[http://dx.doi.org/10.1007/s00044-011-9910-z]
[17]
Glide; Schrodinger, LLC: New York, NY, 2021.
[18]
Guan, L.; Yang, H.; Cai, Y.; Sun, L.; Di, P.; Li, W.; Liu, G.; Tang, Y. ADMET-score-a comprehensive scoring function for evaluation of chemical drug-likeness. Med. Chem. Comm., 2019, 10(1), 148-157.
[http://dx.doi.org/10.1039/C8MD00472B] [PMID: 30774861]
[19]
Siramshetty, V.B.; Nickel, J.; Omieczynski, C.; Gohlke, B.O.; Drwal, M.N.; Preissner, R. WITHDRAWN—a resource for withdrawn and discontinued drugs. Nucleic Acids Res., 2016, 44(D1), D1080-D1086.
[http://dx.doi.org/10.1093/nar/gkv1192] [PMID: 26553801]
[20]
Cheng, F.; Li, W.; Zhou, Y.; Shen, J.; Wu, Z.; Liu, G. admetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. J. Chem. Inf. Model., 2012, 52(11), 3099-3105.
[21]
Tabassum, S.; Zaki, M.; Afzal, M.; Arjmand, F. Synthesis and characterization of Cu(II)-based anticancer chemotherapeutic agent targeting topoisomerase Iα: In vitro DNA binding, pBR322 cleavage, molecular docking studies and cytotoxicity against human cancer cell lines. Eur. J. Med. Chem., 2014, 74, 509-523.
[http://dx.doi.org/10.1016/j.ejmech.2013.12.046] [PMID: 24508781]
[22]
e Zahra S.N.; Khattak, N.A.; Mir, A. Comparative modeling and docking studies of p16ink4/Cyclin D1/Rb pathway genes in lung cancer revealed functionally interactive residue of RB1 and its functional partner E2F1. Theor. Biol. Med. Model., 2013, 10(1), 1-9.
[http://dx.doi.org/10.1186/1742-4682-10-1] [PMID: 23276293]
[23]
Zhang, F.H.; Yan, Y.L.; Wang, Y.; Liu, Z. Lactucin induces potent anti-cancer effects in HL-60 human leukemia cancer cells by inducing apoptosis and sub-G1 cell cycle arrest. Bangladesh J. Pharmacol., 2016, 11(2), 478-484.
[http://dx.doi.org/10.3329/bjp.v11i2.26729]
[24]
Dougherty, D.A. Cation-π interactions in chemistry and biology: A new view of benzene, Phe, Tyr, and Trp. Science, 1996, 271(5246), 163-168.
[http://dx.doi.org/10.1126/science.271.5246.163] [PMID: 8539615]
[25]
Bissantz, C.; Kuhn, B.; Stahl, M. A medicinal chemist’s guide to molecular interactions. J. Med. Chem., 2010, 53(14), 5061-5084.
[http://dx.doi.org/10.1021/jm100112j] [PMID: 20345171]
[26]
Boehr, D.D.; Farley, A.R.; Wright, G.D.; Cox, J.R. Analysis of the π-π stacking interactions between the aminoglycoside antibiotic kinase APH(3′)-IIIa and its nucleotide ligands. Chem. Biol., 2002, 9(11), 1209-1217.
[http://dx.doi.org/10.1016/S1074-5521(02)00245-4] [PMID: 12445771]
[27]
Wintjens, R.; Liévin, J.; Rooman, M.; Buisine, E. Contribution of cation-π interactions to the stability of protein-DNA complexes. J. Mol. Biol., 2000, 302(2), 393-408.
[http://dx.doi.org/10.1006/jmbi.2000.4040] [PMID: 10970741]
[28]
Gallivan, J.P.; Dougherty, D.A. Cation-π interactions in structural biology. Proc. Natl. Acad. Sci. USA, 1999, 96(17), 9459-9464.
[http://dx.doi.org/10.1073/pnas.96.17.9459] [PMID: 10449714]
[29]
Zacharias, N.; Dougherty, D.A. Cation–π interactions in ligand recognition and catalysis. Trends Pharmacol. Sci., 2002, 23(6), 281-287.
[http://dx.doi.org/10.1016/S0165-6147(02)02027-8] [PMID: 12084634]
[30]
Khanna, V.; Ranganathan, S. Physiochemical property space distribution among human metabolites, drugs and toxins. BMC Bioinformatics, 2009, 10(Suppl. 15), S10.
[http://dx.doi.org/10.1186/1471-2105-10-S15-S10]
[31]
Ntie-Kang, F.; Zofou, D.; Babiaka, S.B.; Meudom, R.; Scharfe, M.; Lifongo, L.L.; Mbah, J.A.; Mbaze, L.M.; Sippl, W.; Efange, S.M.N. AfroDb: A select highly potent and diverse natural product library from African medicinal plants. PLoS One, 2013, 8(10), e78085.
[http://dx.doi.org/10.1371/journal.pone.0078085] [PMID: 24205103]
[32]
Guruprasad, K.; Reddy, B.V.B.; Pandit, M.W. Correlation between stability of a protein and its dipeptide composition: a novel approach for predicting in vivo stability of a protein from its primary sequence. Protein Eng. Des. Sel., 1990, 4(2), 155-161.
[http://dx.doi.org/10.1093/protein/4.2.155] [PMID: 2075190]
[33]
Sankar, P.R.; Sailu, A.B.; Eswarudu, M.M.; Satya, M.N.; Sreeja, P.; Roja, P.; Rijwana, S. Analytical methods for determination of different members Of FDA approved tyrosine kinase inhibitors like dasatinib, lapatinib, imatinib, sorafenib, nintedanib, sunitinib and pazopanib: A review. J. Pharm. Sci. Res., 2021, 13(6), 313-318.

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