Journal Home > Volume 9 , Issue 2

Inhibition of beta-site APP cleaving enzyme1 (BACE1) is one of the most promising therapeutic approaches for Alzheimer's disease. To find natural products for the treatment of Alzheimer's disease, absorption, distribution, metabolism, excretion and toxicity (ADMET) properties and in vitro BACE1 inhibitory activity of the peptides isolated from egg albumin were evaluated. Then, molecular docking and molecular dynamics simulation were used to explain the molecular mechanism of the interactions between BACE1 and peptides. The IC50 value of peptide KLPGF, with satisfactory ADMET properties, against BACE1 was (8.30 ± 0.56) mmol/L. Molecular docking revealed that KLPGF contacted with the residues of BACE1's active sites through twelve hydrogen bonds interactions, two hydrophobic interactions, one electrostatic interaction, and two Pi-cation interactions. The 5 ns molecular dynamics simulations confirmed that the structure of KLPGF with BACE1 was stable. Peptide KLPGF contacted the residues Lys321, Asp228, and Asn233 with stable hydrogen bonds. KLPGF may be a potential anti-BACE1 candidate.


menu
Abstract
Full text
Outline
About this article

Biological evaluation and interaction mechanism of beta-site APP cleaving enzyme 1 inhibitory pentapeptide from egg albumin

Show Author's information Zhipeng Yua,bSijia WuaWenzhu Zhaoa( )Long DingcDavid ShiuanaFuping Zhengb( )Jianrong Lia( )Jingbo Liud( )
College of Food Science and Engineering, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Bohai University, Jinzhou, 121013, PR China
Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, 102488, PR China
College of Food Science and Engineering, Northwest A & F University, Yangling, 712100, PR China
Lab of Nutrition and Functional Food, Jilin University, Changchun, 130062, PR China

Peer review under responsibility of KeAi Communications Co., Ltd

Abstract

Inhibition of beta-site APP cleaving enzyme1 (BACE1) is one of the most promising therapeutic approaches for Alzheimer's disease. To find natural products for the treatment of Alzheimer's disease, absorption, distribution, metabolism, excretion and toxicity (ADMET) properties and in vitro BACE1 inhibitory activity of the peptides isolated from egg albumin were evaluated. Then, molecular docking and molecular dynamics simulation were used to explain the molecular mechanism of the interactions between BACE1 and peptides. The IC50 value of peptide KLPGF, with satisfactory ADMET properties, against BACE1 was (8.30 ± 0.56) mmol/L. Molecular docking revealed that KLPGF contacted with the residues of BACE1's active sites through twelve hydrogen bonds interactions, two hydrophobic interactions, one electrostatic interaction, and two Pi-cation interactions. The 5 ns molecular dynamics simulations confirmed that the structure of KLPGF with BACE1 was stable. Peptide KLPGF contacted the residues Lys321, Asp228, and Asn233 with stable hydrogen bonds. KLPGF may be a potential anti-BACE1 candidate.

Keywords: Molecular dynamics, Molecular docking, Alzheimer's disease, BACE1, ADMET prediction, KLPGF

References(34)

[1]

J. Xu, S. Patassini, N. Rustogi, et al., Regional protein expression in human Alzheimer's brain correlates with disease severity, Commun. Biol. 2 (2019) 43, http://dx.doi.org/10.1038/s42003-018-0254-9.

[2]

H.A. Jung, M.Y. Ali, R.J. Choi, et al., Kinetics and molecular docking studies of fucosterol and fucoxanthin, BACE1 inhibitors from brown algae Undaria pinnatifida and Ecklonia stolonifera, Food Chem. Toxicol. 89 (2016) 104–111, http://dx.doi.org/10.1016/j.fct.2016.01.014.

[3]

G.F. Chen, T.H. Xu, Y. Yan, et al., Amyloid beta: structure, biology and structure-based therapeutic development, Acta Pharmacol. Sin. 38 (9) (2017) 1205–1235, http://dx.doi.org/10.1038/aps.2017.28.

[4]

D.J. Selkoe, J. Hardy, The amyloid hypothesis of Alzheimer's disease at 25 years, EMBO Mol. Med. 8 (6) (2016) 595–608, http://dx.doi.org/10.15252/emmm.201606210.

[5]

K.A. Jellinger, C. Bancher, Neuropathology of Alzheimer's disease: a critical update, J. Neural Transm. Suppl. 54 (1998) 77–95, http://dx.doi.org/10.1007/978-3-7091-7508-8_8.

[6]

N. Pradeep, M. Munikumar, S. Swargam, et al., Combination of e-pharmacophore modeling, multiple docking strategies and molecular dynamic simulations to discover of novel antagonists of BACE1, J. Biomol. Struct. Dyn. 33 (2015) 129–130, http://dx.doi.org/10.1080/07391102.2015.1032834.

[7]

J. Cui, X. Wang, X. Li, et al., Targeting the gamma-/beta-secretase interaction reduces beta-amyloid generation and ameliorates Alzheimer's disease-related pathogenesis, Cell Discov. 1 (2015) 15021, http://dx.doi.org/10.1038/celldisc.2015.21.

[8]

Q. Wu, X. Li, Q. Gao, et al., Interaction mechanism exploration of HEA derivatives as BACE1 inhibitors by in silico analysis, Mol. Biosyst. 12 (4) (2016) 1151–1165, http://dx.doi.org/10.1039/C5MB00859J.

[9]

M.P. Gleeson, A. Hersey, D. Montanari, et al., Probing the links between in vitro potency, ADMET and physicochemical parameters, Nat. Rev. Drug Discov. 10 (3) (2011) 197–208, http://dx.doi.org/10.1038/nrd3367.

[10]

Z. Zhang, O. Obianyo, E. Dall, et al., Inhibition of delta-secretase improves cognitive functions in mouse models of Alzheimer's disease, Nat. Commun. 8 (2017) 14740, http://dx.doi.org/10.1038/ncomms14740.

[11]

J. Shen, F.X. Cheng, Y. Xu, et al., Estimation of ADME properties with substructure pattern recognition, J. Chem. Inf. Model. 50 (6) (2010) 1034–1041, http://dx.doi.org/10.1021/ci100104j.

[12]

O.A. Joseph, K. Babatomiwa, A. Niyi, et al., Molecular docking and 3D qsar studies of C000000956 as a potent inhibitor of Bace-1, Drug Res. 69 (8) (2019) 451–457.

[13]

A. Kumar, A. Tiwari, A. Sharma, Changing paradigm from one target one ligand towards multi-target directed ligand design for key drug targets of Alzheimer disease: an important role of in silico methods in multi-target directed ligands design, Curr. Neuropharmacol. 16 (6) (2018) 726–739, http://dx.doi.org/10.2174/1570159X16666180315141643.

[14]

C.R. Butler, M.A. Brodney, E.M. Beck, et al., Discovery of a series of efficient, centrally efficacious BACE1inhibitors through structure-based drug design, J. Med. Chem. 58 (6) (2015) 2678–2702, http://dx.doi.org/10.1021/jm501833t.

[15]

Z. Yu, S. Wu, W. Zhao, et al., Anti-Alzheimers activity and molecular mechanism of albumin-derived peptides against AChE and BChE, Food Funct. 9 (2) (2018) 1173–1178, http://dx.doi.org/10.1039/C7FO01462G.

[16]

D. Lagorce, O. Sperandio, J.B. Baell, et al., FAF-Drugs3: a web server for compound property calculation and chemical library design, Nucleic Acids Res. 43 (W1) (2015) W200–W207, http://dx.doi.org/10.1093/nar/gkv353.

[17]

F. Cheng, W. Li, Y. Zhou, et al., admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties, J. Chem. Inf. Model. 52 (11) (2012) 3099–3105, http://dx.doi.org/10.1021/ci300367a.

[18]

S.M. Rafiquzzaman, E.Y. Kim, J.M. Lee, et al., Anti-Alzheimers and anti-inflammatory activities of a glycoprotein purified from the edible brown alga Undaria pinnatifida, Food Res. Int. 77 (2015) 118–124, http://dx.doi.org/10.1016/j.foodres.2015.08.021.

[19]

C.A. Coburn, S.J. Stachel, Y.M. Li, et al., Identification of a small molecule nonpeptide active site beta-secretase inhibitor that displays a nontraditional binding mode for aspartyl proteases, J. Med. Chem. 47 (25) (2004) 6117–6119, http://dx.doi.org/10.1021/jm049388p.

[20]

S. Patel, P. Modi, M. Chhabria, Rational approach to identify newer caspase-1 inhibitors using pharmacophore based virtual screening, docking and molecular dynamic simulation studies, J. Mol. Graph. 81 (2018) 106–115, http://dx.doi.org/10.1016/j.jmgm.2018.02.017.

[21]

K. Muthusamy, K.D. Singh, S. Chinnasamy, et al., High throughput virtual screening and E-pharmacophore filtering in the discovery of new BACE-1 inhibitors, Interdiscip. Sci. 5 (2) (2013) 119–126, http://dx.doi.org/10.1007/s12539-013-0157-x.

[22]

M.A. Miteva, S. Violas, M. Montes, et al., FAF-Drugs: free ADME/tox filtering of compound collections, Nucleic Acids Res. 34 (2006) W738–W744, http://dx.doi.org/10.1093/nar/gkl065.

[23]

S. Das, M.A. Laskar, S.D. Sarker, et al., Prediction of anti-Alzheimer's activity of flavonoids targeting acetylcholinesterase in silico, Phytochem. Anal. 28 (4) (2017) 324–331, http://dx.doi.org/10.1002/pca.2679.

[24]

S. Kauthale, S. Tekale, M. Damale, et al., Synthesis, antioxidant, antifungal, molecular docking and ADMET studies of some thiazolyl hydrazones, Bioorg. Med. Chem. Lett. 27 (16) (2017) 3891–3896, http://dx.doi.org/10.1016/j.bmcl.2017.06.043.

[25]

C. Giromini, F. Cheli, R. Rebucci, et al., Invited review: dairy proteins and bioactive peptides: modeling digestion and the intestinal barrier, J. Dairy Sci. 102 (2) (2019) 929–942, http://dx.doi.org/10.3168/jds.2018-15163.

[26]

N. Kumar, N. Goel, T.C. Yadav, et al., Quantum chemical, ADMET and molecular docking studies of ferulic acid amide derivatives with a novel anticancer drug target, Med. Chem. Res. 26 (8) (2017) 1822–1834, http://dx.doi.org/10.1007/s00044-017-1893-y.

[27]

Y.M. Liu, Y.D. Feng, X. Lu, et al., Isosteroidal alkaloids as potent dual-binding site inhibitors of both acetylcholinesterase and butyrylcholinesterase from the bulbs of Fritillaria walujewii, Eur. J. Med. Chem. 137 (2017) 280–291, http://dx.doi.org/10.1016/j.ejmech.2017.06.007.

[28]

J. Han, Y. Ji, K. Youn, et al., Baicalein as a potential inhibitor against BACE1 and AChE: mechanistic comprehension through in vitro and computational approaches, Nutrients 11 (11) (2019) 2694, http://dx.doi.org/10.3390/nu11112694.

[29]

R.G. Coleman, K.A. Sharp, Protein pockets: inventory, shape, and comparison, J. Chem. Inf. Model. 50 (4) (2010) 589–603.

[30]

J. Yuan, S. Venkatraman, Y. Zheng, et al., Structure-based design of beta-site APP cleaving enzyme 1 (BACE1) inhibitors for the treatment of Alzheimer's disease, J. Med. Chem. 56 (11) (2013) 4156–4180, http://dx.doi.org/10.1021/jm301659n.

[31]

M. Hernandez-Rodriguez, J. Correa-Basurto, A. Gutierrez, et al., Asp32 and Asp228 determine the selective inhibition of BACE1 as shown by docking and molecular dynamics simulations, Eur. J. Med. Chem. 124 (2016) 1142–1154, http://dx.doi.org/10.1016/j.ejmech.2016.08.028.

[32]

J.L. Dominguez, F. Fernandez-Nieto, M. Castro, et al., Computer-aided structure-based design of multitarget leads for Alzheimer's disease, J. Chem. Inf. Model. 55 (1) (2015) 135–148, http://dx.doi.org/10.1021/ci500555g.

[33]

N. Nagasundaram, H.L. Zhu, J.M. Liu, et al., Analysing the effect of mutation on protein function and discovering potential inhibitors of CDK4: molecular modelling and dynamics studies, PLoS One 10 (8) (2015), e0133969, http://dx.doi.org/10.1371/journal.pone.0133969.

[34]

S. Saxena, M. Abdullah, D. Sriram, et al., Discovery of novel inhibitors of Mycobacterium tuberculosis MurG: homology modelling, structure based pharmacophore, molecular docking, and molecular dynamics simulations, J. Biomol. Struct. Dyn. 36 (12) (2018) 3184–3198, http://dx.doi.org/10.1080/07391102.2017.1384398.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 12 October 2019
Revised: 21 November 2019
Accepted: 15 January 2020
Published: 24 January 2020
Issue date: June 2020

Copyright

© 2020 "Society information". Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

Acknowledgements

This work was supported by the National Natural Science Funds of China (No.31901635) and the National Key Research and Development Program of China (2018YFD0400301).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return