Skip to main content

Advertisement

Log in

An arginine-rich peptide inhibits AChE: template-based design, molecular modeling, synthesis, and biological evaluation

  • Original Paper
  • Published:
Journal of Molecular Modeling Aims and scope Submit manuscript

Abstract

Life expectancy is growing especially in developed countries. In this regard, aging-associated diseases such as Alzheimer’s disease (AD) are more common. Multi interconnected pathological factors involved in AD demand multi-target therapeutics. AChE, as a well-known target in AD, decreases the acetylcholine (ACh) in cholinergic synapse and, besides, increases the rate of amyloid-beta (Aβ) aggregation. To block the destructive effects of AChE on cholinergic neurons in AD, we designed a peptidic inhibitor of the peripheral anionic site (PAS). The PAS plays a crucial role to attract and direct the ACh to the enzyme active site and increase the rate of Aβ aggregation by changing the folding state. We utilized the template-based approach in combination with molecular docking, molecular dynamic simulation, and data mining to design a peptide library. Scoring was performed according to binding energy and the interaction profile of AChE inhibitors. The best candidate (p8, RMLRTTRY) was synthesized using solid-phase peptide synthesis, purified by RP-HPLC, and identified by ESI–MS. The inhibitory effect of p8 on AChE was 102.2 ± 15.2 μM. The kinetic and molecular modeling studies indicated the mixed inhibition mechanism for p8. The Arg residues in p8 had an essential role in binding to PAS.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

Data were reported in the manuscript as much as possible, and other materials will be available via request to the corresponding author.

Code availability

As detailed for each software application in the “Material and methods” section.

References

  1. Koehn FE, Carter GT (2005) The evolving role of natural products in drug discovery. Nat Rev Drug Discov 4:206–220

    Article  CAS  Google Scholar 

  2. Sachdeva S (2017) Peptides as ‘drugs’: the journey so far. Int J Pept Res Ther 23:49–60

    Article  CAS  Google Scholar 

  3. Henninot A, Collins JC, Nuss JM (2018) The current state of peptide drug discovery: back to the future? J Med Chem 61:1382–1414

    Article  CAS  Google Scholar 

  4. Hollenstein K, de Graaf C, Bortolato A et al (2014) Insights into the structure of class B GPCRs. Trends Pharmacol Sci 35:12–22

    Article  CAS  Google Scholar 

  5. Lau JL, Dunn MK (2018) Therapeutic peptides: historical perspectives, current development trends, and future directions. Bioorg Med Chem 26:2700–2707

    Article  CAS  Google Scholar 

  6. Saroj Devi N, Shanmugam R, Ghorai J et al (2018) Ligand-based modeling for the prediction of pharmacophore features for multi-targeted inhibition of the arachidonic acid cascade. Mol Inform 37:1700073

    Article  Google Scholar 

  7. Bolognesi ML, Cavalli A (2016) Multitarget drug discovery and polypharmacology. ChemMedChem 11:1190–1192

    Article  CAS  Google Scholar 

  8. Wang N, Qiu P, Cui W et al (2019) Recent advances in multi-target anti-Alzheimer disease compounds (2013 up to the present). Curr Med Chem 26:5684–5710

    Article  CAS  Google Scholar 

  9. (2020) 2020 Alzheimer’s disease facts and figures. Alzheimer’s Dement 16:391–460. https://doi.org/10.1002/alz.12068

  10. Hampel H, Mesulam M-M, Cuello AC et al (2019) Revisiting the cholinergic hypothesis in Alzheimer’s disease: emerging evidence from translational and clinical research. J Prev Alzheimer’s Dis 6:2–15

    CAS  Google Scholar 

  11. Maraković N, Knežević A, Vinković V et al (2016) Design and synthesis of N-substituted-2-hydroxyiminoacetamides and interactions with cholinesterases. Chem Biol Interact 259:122–132

    Article  Google Scholar 

  12. Johnson JL, Thomas JL, Emani S et al (2005) Measuring carbamoylation and decarbamoylation rate constants by continuous assay of AChE. Chem Biol Interact 157:384–385

    Article  Google Scholar 

  13. Inestrosa NC, Dinamarca MC, Alvarez A (2008) Amyloid–cholinesterase interactions: implications for Alzheimer’s disease. FEBS J 275:625–632

    Article  CAS  Google Scholar 

  14. Saxena AK, Saini R (2018) The structural hybrids of acetylcholines-terase inhibitors in the treatment of Alzheimer’s disease: a review. J Alzheimers Neurodegener Dis 4:1–25

    Article  Google Scholar 

  15. Cheung J, Rudolph MJ, Burshteyn F et al (2012) Structures of human acetylcholinesterase in complex with pharmacologically important ligands. J Med Chem 55:10282–10286

    Article  CAS  Google Scholar 

  16. Su G, Zhao T, Zhao Y et al (2016) Effect of anchovy (Coilia mystus) protein hydrolysate and its Maillard reaction product on combating memory-impairment in mice. Food Res Int 82:112–120

    Article  CAS  Google Scholar 

  17. Zare-Zardini H, Tolueinia B, Hashemi A et al (2013) Antioxidant and cholinesterase inhibitory activity of a new peptide from Ziziphus jujuba fruits. Am J Alzheimer’s Dis Other Dementias® 28:702–709

    Article  Google Scholar 

  18. Malomo SA, Aluko RE (2016) In vitro acetylcholinesterase-inhibitory properties of enzymatic hemp seed protein hydrolysates. J Am Oil Chem Soc 93:411–420

    Article  CAS  Google Scholar 

  19. Perumal P, Vasudevan M, Sridevi C, Manikandan S (2017) Anticholinesterase activity of octa peptides related to human histatin 8: in-silico drug design and in-vitro. Asian J Pharm Clin Res 10:115–117

    Article  CAS  Google Scholar 

  20. Mondal P, Gupta V, Das G et al (2018) Peptide-based acetylcholinesterase inhibitor crosses the blood-brain barrier and promotes neuroprotection. ACS Chem Neurosci 9:2838–2848

    Article  CAS  Google Scholar 

  21. Tahmasebi E, Dastan D, Ebadi A (2020) Design, synthesis and biological evaluation of anticholinesterase peptides: fragment-based vs. template-based peptide design. Bioorg Chem 105:104351

    Article  CAS  Google Scholar 

  22. Nachon F, Carletti E, Ronco C et al (2013) Crystal structures of human cholinesterases in complex with huprine W and tacrine: elements of specificity for anti-Alzheimer’s drugs targeting acetyl-and butyryl-cholinesterase. Biochem J 453:393–399

    Article  CAS  Google Scholar 

  23. Hess B, Kutzner C, Van Der Spoel D, Lindahl E (2008) GROMACS 4: algorithms for highly efficient, load-balanced, and scalable molecular simulation. J Chem Theory Comput 4:435–447

    Article  CAS  Google Scholar 

  24. Ebadi A, Olyaie SS, Dastan D (2020) To be ionized or not to be ionized: the vital role of physicochemical properties of galbanic acid derivatives in AChE assay. J Biomol Struct Dyn. https://doi.org/10.1080/07391102.2020.1764391

    Article  PubMed  PubMed Central  Google Scholar 

  25. Curtin RR, Edel M, Lozhnikov M et al (2018) mlpack 3: a fast, flexible machine learning library. J Open Source Softw 3:726

    Article  Google Scholar 

  26. Kortemme T, Kim DE, Baker D (2004) Computational alanine scanning of protein-protein interfaces. Sci STKE 2004:pl2–pl2

  27. Laskowski RA, Jabłońska J, Pravda L et al (2018) PDBsum: structural summaries of PDB entries. Protein Sci 27:129–134

    Article  CAS  Google Scholar 

  28. Dominguez C, Boelens R, Bonvin AMJJ (2003) HADDOCK: a protein− protein docking approach based on biochemical or biophysical information. J Am Chem Soc 125:1731–1737

    Article  CAS  Google Scholar 

  29. Dastan D, Fasihi K, Ebadi A (2020) From venom to AChE inhibitor: design, molecular modeling, and synthesis of a peptidic inhibitor of AChE. Int J Pept Res Ther. https://doi.org/10.1007/s10989-020-10103-w

    Article  Google Scholar 

  30. Waldrop GL (2009) A qualitative approach to enzyme inhibition. Biochem Mol Biol Educ 37:11–15

    Article  CAS  Google Scholar 

  31. Axelsen PH, Harel M, Silman I, Sussman JL (1994) Structure and dynamics of the active site gorge of acetylcholinesterase: synergistic use of molecular dynamics simulation and X-ray crystallography. Protein Sci 3:188–197

    Article  CAS  Google Scholar 

  32. Fehrentz J-A, Martinez J, Subra G, Amblard M (2006) Methods and protocols of modern solid phase peptide synthesis. Mol Biotechnol 33:239–254

    Article  Google Scholar 

Download references

Acknowledgements

We would like to thank the Research and Technology Vice-chancellor of Hamadan University of Medical Sciences for financial support.

Funding

This research was funded by Vice-chancellor for Research and Technology, Hamadan University of Medical Sciences (No. 9604132260).

Author information

Authors and Affiliations

Authors

Contributions

The initial idea about a peptidic inhibitors of PAS in AChE was conceived by AE. Accordingly, AE and DD hypothesized the problem and designed the study. AE, RZ and KF performed all the calculations. RZ and KF synthesized the peptide and evaluated the AChE inhibitory effect. AE and DD contributed with results, discussions, and especially during the revision. RZ and KF drafted most of the basic structure of the introduction and methods. AE wrote the main draft of the paper with active help primarily from DD and rigorously revised the paper until satisfactory. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Ahmad Ebadi.

Ethics declarations

Ethics approval

N/A

Consent to participate

N/A

Consent for publication

N/A

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dastan, D., Zhiyani, R., Fasihi, K. et al. An arginine-rich peptide inhibits AChE: template-based design, molecular modeling, synthesis, and biological evaluation. J Mol Model 28, 86 (2022). https://doi.org/10.1007/s00894-022-05058-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00894-022-05058-2

Keywords

Navigation