Skip to main content
Log in

New Insight into the Pharmacological Importance of Atropine as the Potential Inhibitor of AKR1B1 via Detailed Computational Investigations: DFTs, ADMET, Molecular Docking, and Molecular Dynamics Studies

  • Original Article
  • Published:
Applied Biochemistry and Biotechnology Aims and scope Submit manuscript

Abstract

The aim of this research is to investigate the quantum geometric properties and chemical reactivity of atropine, a pharmaceutically active tropane alkaloid. Using density functional theory (DFT) computations with the B3LYP/SVP functional theory basis set, the most stable geometry of atropine was determined. Additionally, a variety of energetic molecular parameters were calculated, such as the optimized energy, atomic charges, dipole moment, frontier molecular orbital energies, HOMO–LUMO energy gap, molecular electrostatic potential, chemical reactivity descriptors, and molecular polarizability. To determine atropine’s inhibitory potential, molecular docking was used to analyze ligand interactions within the active pockets of aldo–keto reductase (AKR1B1 and AKR1B10). The results of these studies showed that atropine has greater inhibitory action against AKR1B1 than AKR1B10, which was further validated through molecular dynamic simulations by analyzing root mean square deviation (RMSD) and root mean square fluctuations (RMSF). The results of the molecular docking simulation were supplemented with simulation data, and the ADMET characteristics were also determined to predict the drug likeness of a potential compound. In conclusion, the research suggests that atropine has potential as an inhibitor of AKR1B1 and could be used as a parent compound for the synthesis of more potent leads for the treatment of colon cancer associated with the sudden expression of AKR1B1.

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
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data Availability

All data generated or analyzed during this study are included in this article (and its supplementary information file).

References

  1. Chen, J., & Huang, X.-F. (2009). The signal pathways in azoxymethane-induced colon cancer and preventive implications. Cancer biology & therapy, 8(14), 1313–1317.

    Article  CAS  Google Scholar 

  2. Bauer, B., et al. (2020). Quantum algorithms for quantum chemistry and quantum materials science. Chemical Reviews, 120(22), 12685–12717.

    Article  CAS  PubMed  Google Scholar 

  3. Chen, J., & Huang, X.-F. (2009). The signal pathways in azoxymethane-induced colon cancer and preventive implications. Cancer biology therapy, 8(14), 1313–1317.

    Article  CAS  PubMed  Google Scholar 

  4. Matsunaga, T., et al. (2012). Aldo–keto reductase 1B10 and its role in proliferation capacity of drug-resistant cancers. Frontiers in pharmacology, 3, 5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ruiz, F. X., et al. (2011). Human and rodent aldo–keto reductases from the AKR1B subfamily and their specificity with retinaldehyde. Chemico-biological interactions, 191(1–3), 199–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Demirkol Canlı, S., et al. (2020). Evaluation of an aldo-keto reductase gene signature with prognostic significance in colon cancer via activation of epithelial to mesenchymal transition and the p70S6K pathway. Carcinogenesis, 41(9), 1219–1228.

    Article  PubMed  Google Scholar 

  7. Alam, M., et al. (2018). DFT/TD-DFT calculations, spectroscopic characterizations (FTIR, NMR, UV–vis), molecular docking and enzyme inhibition study of 7-benzoyloxycoumarin. Computational Biology Chemistry, 73, 65–78.

    Article  CAS  PubMed  Google Scholar 

  8. Kaur, R., & Arora, S. (2015). Alkaloids-important therapeutic secondary metabolites of plant origin. Critical Review, 2(3), 1–8.

    Google Scholar 

  9. Devi, R., et al. (2020). Fungal secondary metabolites and their biotechnological applications for human health. New and future developments in microbial biotechnology and bioengineering (pp. 147–161). Elsevier.

    Chapter  Google Scholar 

  10. Kaur, R., & Arora, S. (2015). Alkaloids-important therapeutic secondary metabolites of plant origin. J Crit Rev, 2(3), 1–8.

    Google Scholar 

  11. Grynkiewicz, G., & Gadzikowska, M. (2008). Tropane alkaloids as medicinally useful natural products and their synthetic derivatives as new drugs. Pharmacological Reports, 60(4), 439.

    CAS  PubMed  Google Scholar 

  12. Kohnen-Johannsen, K. L., & Kayser, O. (2019). Tropane alkaloids: Chemistry, pharmacology, biosynthesis and production. Molecules, 24(4), 796.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Hallcher, L. M., & Sherman, W. R. (1980). The effects of lithium ion and other agents on the activity of myo-inositol-1-phosphatase from bovine brain. Journal of Biological Chemistry, 255(22), 10896–10901.

    Article  CAS  PubMed  Google Scholar 

  14. Mohanlall, V. & Ally, F. (2020). An overview of tropane alkaloids from Datura stramonium L. Journal of Pharmacognosy Phytochemistry, 9(3),5–13

  15. Prieto-Martínez, F. D., Arciniega, M., & Medina-Franco, J. L. (2018). Molecular docking: Current advances and challenges. TIP. Revista especializada en ciencias químico-biológicas, 21(1), 65–87.

  16. Rasheed, M. A., et al. (2021). Identification of lead compounds against Scm (fms10) in Enterococcus faecium using computer aided drug designing. Life, 11(2), 77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Alam, M., et al. (2018). DFT/TD-DFT calculations, spectroscopic characterizations (FTIR, NMR, UV–vis), molecular docking and enzyme inhibition study of 7-benzoyloxycoumarin. Computational Biology and Chemistry, 73, 65–78.

    Article  CAS  PubMed  Google Scholar 

  18. Frisch, M., et al. (2003). Revision B. Gaussian 03. Gaussian. Inc., Pittsburgh (PA).

  19. Goerigk, L., & Reimers, J. R. (2013). Efficient methods for the quantum chemical treatment of protein structures: The effects of London-dispersion and basis-set incompleteness on peptide and water-cluster geometries. Journal of Chemical Theory Computation, 9(7), 3240–3251.

    Article  CAS  PubMed  Google Scholar 

  20. Tirado-Rives, J., & Jorgensen, W. L. (2008). Performance of B3LYP density functional methods for a large set of organic molecules. Journal of chemical theory computation, 4(2), 297–306.

    Article  CAS  PubMed  Google Scholar 

  21. Dennington, R., Keith, T. A. & Millam, J. M. (2016). GaussView, version 6.0. 16. Semichem Inc Shawnee Mission KS, USA.

  22. Torres, J. A. G., et al. (2019). Low-scaling algorithm for nudged elastic band calculations using a surrogate machine learning model. Physical review letters, 122(15), 156001.

    Article  Google Scholar 

  23. Dennington, R., Keith, T.A. & Millam, J. M. (2016). GaussView 6.0. 16. Semichem Inc.

  24. Channar S. A., et al., (2022). Exploring thiazole-linked thioureas using alkaline phosphatase assay, biochemical evaluation, computational analysis and structure–activity relationship (SAR) studies. Medicinal Chemistry Research, 31(10), 1792–802.

  25. Aziz, M., et al. (2022). Identification of potent inhibitors of NEK7 protein using a comprehensive computational approach. Scientific reports, 12(1), 1–17.

    Article  CAS  Google Scholar 

  26. Berman, H., Henrick, K., & Nakamura, H. (2003). Announcing the worldwide protein data bank. Nature Structural Molecular Biology, 10(12), 980–980.

    Article  CAS  Google Scholar 

  27. Morris, G. M., et al. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of computational chemistry, 30(16), 2785–2791.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Trott, O., & Olson, A. J. (2010). AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of computational chemistry, 31(2), 455–461.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Belhassan, A., et al. (2021). Camphor, artemisinin and sumac phytochemicals as inhibitors against COVID-19: Computational approach. Computers in Biology Medicine, 136, 104758.

    Article  CAS  PubMed  Google Scholar 

  30. Ouassaf, M., et al. (2021). Combined docking methods and molecular dynamics to identify effective antiviral 2, 5-diaminobenzophenonederivatives against SARS-CoV-2. Journal of King Saud University-Science, 33(2), 101352.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Morris, G. M., et al. (1998). Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. Journal of computational chemistry, 19(14), 1639–1662.

    Article  CAS  Google Scholar 

  32. Heinzerling, L., Klein, R., & Rarey, M. (2012). Fast force field-based optimization of protein–ligand complexes with graphics processor. Journal of Computational Chemistry, 33(32), 2554–2565.

    Article  CAS  PubMed  Google Scholar 

  33. Visualizer, Discovery Studio. Accelrys software inc. Discovery studio visualizer 2.

  34. Wang, Z., et al. (2016). Comprehensive evaluation of ten docking programs on a diverse set of protein–ligand complexes: The prediction accuracy of sampling power and scoring power. Physical Chemistry Chemical Physics, 18(18), 12964–12975.

    Article  CAS  PubMed  Google Scholar 

  35. Aziz, M., et al. (2022). Identification of NEK7 inhibitors: Structure based virtual screening, molecular docking, density functional theory calculations and molecular dynamics simulations. Journal of Biomolecular Structure Dynamics, 1–15.

  36. Bowers, K. J., et al. (2006). Scalable algorithms for molecular dynamics simulations on commodity clusters. In SC'06: Proceedings of the 2006 ACM/IEEE Conference on Supercomputing. 84.

  37. Ferreira, L. G., et al. (2015). Molecular docking and structure-based drug design strategies. Molecules, 20(7), 13384–13421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Belhassan, A., et al. (2022). In silico detection of potential inhibitors from vitamins and their derivatives compounds against SARS-CoV-2 main protease by using molecular docking, molecular dynamic simulation and ADMET profiling. Journal of molecular structure, 1258, 132652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Channar, P. A., et al. (2021). Structural and functional insight into thiazolidinone derivatives as novel candidates for anticancer drug design: In vitro biological and in-silico strategies. Journal of Biomolecular Structure Dynamics, 1–12.

  40. Xiong, G., et al. (2021). ADMETlab 2.0: An integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Research, 49(W1), W5–W14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was supported by Taif University Researchers supporting project (number TURSP-2020/38), Taif University, Taif, Saudi Arabia, for research resources.

Author information

Authors and Affiliations

Authors

Contributions

S.A.E. and G.S.B. devised and supervised the study plan. S.A.E., S.S.A., M.A., and F.S. carried out the molecular docking investigations and DFTs. The MD simulations were performed by S.A.E and M.A. The manuscript write up was carried out by S.A.E., G.S.B., A.A., and S.M.A. All authors read and approved the manuscript for publication.

Corresponding author

Correspondence to Syeda Abida Ejaz.

Ethics declarations

Ethics Approval

Not applicable

Consent to Participate

All authors were agreed to participate.

Consent for Publication

All authors have approved the last version of the manuscript for its submission.

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.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 2333 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ejaz, S.A., Aziz, M., Ahmed, A. et al. New Insight into the Pharmacological Importance of Atropine as the Potential Inhibitor of AKR1B1 via Detailed Computational Investigations: DFTs, ADMET, Molecular Docking, and Molecular Dynamics Studies. Appl Biochem Biotechnol 195, 5136–5157 (2023). https://doi.org/10.1007/s12010-023-04411-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12010-023-04411-2

Keywords

Navigation