Investigating the Anti-Leishmanial Potential of Phytoconstituents Derived from Anchusa arvensis: In-vitro and Network Pharmacology Study

Authors

  • Maryam Department of Pharmacy, Kohat University of Science and Technology, Kohat. Pakistan Author
  • Fawad Ali Department of Pharmacy, Kohat University of Science and Technology, Kohat. Pakistan Author
  • Sajid Hussain Department of Pharmacy, Kohat University of Science and Technology, Kohat. Pakistan Author
  • Neelum Gul Qazi Department of Pharmacy, Iqra University Islamabad, Pakistan Author
  • Abdul Wahab Department of Pharmacy, Kohat University of Science and Technology, Kohat. Pakistan Author
  • Naseebullah Department of Genetics, Hazara University Mansehra, Pakistan Author
  • Abdul Saboor Pirzada Department of Pharmacy, Abdul Wali Khan University Mardan, 23200. Pakistan Author
  • Muhammad Ikram Department of Pharmacy, Abdul Wali Khan University Mardan, 23200. Pakistan Author

DOI:

https://doi.org/10.61919/0tgkzm06

Keywords:

Anchusa arvensis; p-methoxy catechol; uvaol; leishmaniasis; molecular docking; network pharmacology; ADMET; anti-leishmanial agents; drug discovery; natural products

Abstract

Background: Leishmaniasis is a neglected tropical disease with limited treatment options, characterized by high toxicity, rising resistance, and poor affordability of currently available drugs. Plant-derived bioactive compounds represent a promising alternative, offering potential for safer, more cost-effective therapeutics. Anchusa arvensis (Boraginaceae) has long been used in traditional medicine and is reported to possess diverse pharmacological properties, yet its anti-leishmanial potential remains underexplored. Objective: This study aimed to evaluate the anti-leishmanial efficacy, drug-likeness, and safety profiles of two key phytoconstituents from A. arvensis—p-methoxy catechol and uvaol—using an integrated approach combining in silico network pharmacology, molecular docking, ADMET prediction, and in vitro assays. Methods: Computational tools including PASS Online, SwissADME, pkCSM, FAME3, GLORY, Pred-hERG, and Endocrine Disruptome were used to predict pharmacokinetics, toxicity, and drug-likeness. Molecular docking was performed against the Leishmania tropica leishmanolysin (gp63; PDB ID: 1LML), followed by molecular dynamics simulation to assess complex stability. In vitro anti-leishmanial activity was tested against L. tropica promastigotes using MTT assays, with glucantime as a reference drug. Results: p-Methoxy catechol exhibited favorable drug-likeness (Lipinski compliant), low predicted toxicity, and strong activity as an inhibitor of aspulvinone dimethylallyltransferase and chlordecone reductase. Uvaol demonstrated additional hepatoprotective and anti-inflammatory potentials but showed moderate risks of hepatotoxicity and skin sensitization. Molecular docking revealed binding energies of −5.74 kcal/mol (p-methoxy catechol) and −9.0 kcal/mol (uvaol), with stable hydrogen bonding at key gp63 residues. In vitro assays confirmed significant anti-leishmanial activity with IC₅₀ values of 5.60 µg/mL (p-methoxy catechol) and 16.17 µg/mL (uvaol), compared to 0.78 µg/mL for glucantime. Conclusion: The findings establish p-methoxy catechol as a potent, safe, and druggable candidate, while uvaol offers complementary therapeutic value through multi-target pharmacology. Both compounds represent promising leads for the development of affordable plant-based anti-leishmanial therapies, aligning with global health goals to combat neglected tropical diseases.

 

References

1. Stojanoski, N., Development of health culture in Veles and its region from the past to the end of the 20th century. Veles: Society of science and art, 1999. 13: p. 34.

2. Petrovska, B.B., Historical review of medicinal plants’ usage. Pharmacognosy reviews, 2012. 6(11): p. 1.

3. Abu-Rabia, A., Herbs as a food and medicine source in Palestine. Asian Pacific Journal of Cancer Prevention, 2005. 6(3): p. 404.

4. Dafni, A., Z. Yaniv, and D.J.J.o.E. Palevitch, Ethnobotanical survey of medicinal plants in northern Israel. 1984. 10(3): p. 295-310.

5. Ali-Shtayeh, M.S., Z. Yaniv, and J.J.J.o.e. Mahajna, Ethnobotanical survey in the Palestinian area: a classification of the healing potential of medicinal plants. 2000. 73(1-2): p. 221-232.

6. Al-Snafi, A.E.J.I.j.o.p. and p. sciences, The pharmacology of Anchusa italica and Anchusa strigosa–A review. 2014. 6(4): p. 7-10.

7. Perveen, A. and M. Qaiser, Pollen flora of Pakistan-IV. Boraginaceae. Pakistan Journal of Botany, 1995. 27: p. 327-360.

8. Hussain, S., et al., In silico, cytotoxic and antioxidant potential of novel ester, 3-hydroxyoctyl-5-trans-docosenoate isolated from anchusa arvensis (L.) m. bieb. against hepg-2 cancer cells. Drug Design, Development and Therapy, 2019: p. 4195-4205.

9. Tsermentseli, S., et al., Phytochemical analysis of Anchusa arvensis roots. 2008. 74(09): p. PC53.

10. Talib, W.H., et al., Natural Products and Altered Metabolism in Cancer: Therapeutic Targets and Mechanisms of Action. International Journal of Molecular Sciences, 2024. 25(17): p. 9593.

11. Hussain, S., et al., Cytotoxicity of Anchusa arvensis against HepG-2 cell lines: Mechanistic and computational approaches. 2019. 19(30): p. 2805-2813.

12. Domingo-Fernández, D., et al., Natural Products Have Increased Rates of Clinical Trial Success throughout the Drug Development Process. 2024. 87(7): p. 1844-1851.

13. Domingo-Fernández, D., et al., Modern drug discovery using ethnobotany: a large-scale cross-cultural analysis of traditional medicine reveals common therapeutic uses. 2023. 26(9).

14. Chen, L., et al., From laptop to benchtop to bedside: structure-based drug design on protein targets. Current pharmaceutical design, 2012. 18(9): p. 1217-1239.

15. Billur Engin, H., et al., Network-based strategies can help mono-and poly-pharmacology drug discovery: a systems biology view. Current pharmaceutical design, 2014. 20(8): p. 1201-1207.

16. Bennett, J.E., R. Dolin, and M.J. Blaser, Mandell, Douglas, and Bennett's principles and practice of infectious diseases. 2014: Elsevier Inc.

17. Gupta, A.K., et al., The pathogenicity and virulence of Leishmania-interplay of virulence factors with host defenses. Virulence, 2022. 13(1): p. 903-935.

18. Chang, C.Y., et al., Gabapentin in acute postoperative pain management. 2014. 2014(1): p. 631756.

19. Jamal, Q., et al., Prevalence and comparative analysis of cutaneous leishmaniasis in Dargai Region in Pakistan. 2013. 45(2).

20. Ullah, N., et al., Plants as antileishmanial agents: current scenario. 2016. 30(12): p. 1905-1925.

21. Elawad, M.A., et al., Natural products derived steroids as potential anti-leishmanial agents; disease prevalence, underlying mechanisms and future perspectives. 2023. 193: p. 109196.

22. Kim, S., Exploring chemical information in PubChem. Current protocols, 2021. 1(8): p. e217.

23. Ru, J., et al., TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. Journal of cheminformatics, 2014. 6: p. 1-6.

24. Filimonov, D., et al., Prediction of the biological activity spectra of organic compounds using the PASS online web resource. Chemistry of Heterocyclic Compounds, 2014. 50: p. 444-457.

25. Lipinski, C.A., et al., Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced drug delivery reviews, 1997. 23(1-3): p. 3-25.

26. Husain, A., et al., Synthesis, molecular properties, toxicity and biological evaluation of some new substituted imidazolidine derivatives in search of potent anti-inflammatory agents. Saudi Pharmaceutical Journal, 2016. 24(1): p. 104-114.

27. Pires, D.E., T.L. Blundell, and D.B. Ascher, pkCSM: predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. Journal of medicinal chemistry, 2015. 58(9): p. 4066-4072.

28. Lohohola, P.O., et al., In silico ADME/T properties of quinine derivatives using SwissADME and pkCSM webservers. 2021. 42(11): p. 1-12.

29. Banerjee, P., et al., ProTox 3.0: a webserver for the prediction of toxicity of chemicals. 2024: p. gkae303.

30. Rocha, E.C.M.d., et al., High-Throughput Molecular Modeling and Evaluation of the Anti-Inflammatory Potential of Açaí Constituents against NLRP3 Inflammasome. 2024. 25(15): p. 8112.

31. Lagunin, A.A., et al., CLC-Pred: A freely available web-service for in silico prediction of human cell line cytotoxicity for drug-like compounds. 2018. 13(1): p. e0191838.

32. Šícho, M., et al., FAME 3: predicting the sites of metabolism in synthetic compounds and natural products for phase 1 and phase 2 metabolic enzymes. Journal of chemical information and modeling, 2019. 59(8): p. 3400-3412.

33. Stork, C., et al., NERDD: a web portal providing access to in silico tools for drug discovery. Bioinformatics, 2020. 36(4): p. 1291-1292.

34. Braga, R.C., et al., Pred‐hERG: A novel web‐accessible computational tool for predicting cardiac toxicity. Molecular informatics, 2015. 34(10): p. 698-701.

35. Borba, J.V., et al., Pred-skin: a web portal for accurate prediction of human skin sensitizers. 2020. 34(2): p. 258-267.

36. Kolšek, K., et al., Endocrine Disruptome An Open Source Prediction Tool for Assessing Endocrine Disruption Potential through Nuclear Receptor Binding. 2014, ACS Publications.

37. Ali, F., et al., Reprofiling analysis of FDA approved drugs with upregulated differential expression genes found in hypertension. Informatics in Medicine Unlocked, 2022. 29: p. 100895.

38. Ullah, I., Z.K. Shinwari, and A.T.J.P.J.B. Khalil, Investigation of the cytotoxic and antileishmanial effects of Fagonia indica L. extract and extract mediated silver nanoparticles (AgNPs). 2017. 49(4): p. 1561-1568.

39. Takahashi, I., et al., Purification and characterization of dimethylallyl pyrophosphate: aspulvinone dimethylallyltransferase from Aspergillus terreus. Biochemistry, 1978. 17(13): p. 2696-2702.

40. Zhang, L.-H., et al., Polyketide butenolide, diphenyl ether, and benzophenone derivatives from the fungus Aspergillus flavipes PJ03-11. Bioorganic & medicinal chemistry letters, 2016. 26(2): p. 346-350.

41. Molowa, D., A. Shayne, and P. Guzelian, Purification and characterization of chlordecone reductase from human liver. Journal of Biological Chemistry, 1986. 261(27): p. 12624-12627.

42. Bonel-Pérez, G.C., et al., Antiproliferative and pro-apoptotic effect of uvaol in human hepatocarcinoma HepG2 cells by affecting G0/G1 cell cycle arrest, ROS production and AKT/PI3K signaling pathway. Molecules, 2020. 25(18): p. 4254.

43. Lynch, T. and A. Price, The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. American family physician, 2007. 76(3): p. 391-396.

44. de Bruyn Kops, C., et al., GLORY: generator of the structures of likely cytochrome P450 metabolites based on predicted sites of metabolism. Frontiers in chemistry, 2019. 7: p. 402.

45. De Coster, S. and N. Van Larebeke, Endocrine-disrupting chemicals: associated disorders and mechanisms of action. Journal of environmental and public health, 2012. 2012.

46. Nielsen, E., G. Ostergaard, and J.C. Larsen, Toxicological risk assessment of chemicals: A practical guide. 2008: CRC Press.

47. Sharma, S.V., D.A. Haber, and J. Settleman, Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nature reviews cancer, 2010. 10(4): p. 241-253.

48. Borba, J.V., et al., Pred-skin: a web portal for accurate prediction of human skin sensitizers. Chemical Research in Toxicology, 2020. 34(2): p. 258-267.

49. Chen, D., et al., Regulation of protein-ligand binding affinity by hydrogen bond pairing. 2016. 2(3): p. e1501240.

50. Dhorajiwala, T.M., et al., Computer-aided docking studies of phytochemicals from plants Salix subserrata and Onion as inhibitors of glycoprotein G of rabies virus. 2019. 3(4): p. 269-276.

51. Siddiqui, S., et al., Kaneric acid, a new triterpene from the leaves of Nerium oleander. Journal of Natural Products, 1986. 49(6): p. 1086-1090.

52. Babalola, I.T. and E.A. Adelakun, Isolation and Characterization of Two Ursane-Skeleton Triterpenoids from Eucalyptus grandis (Myrtaceae). 2017.

53. Du, S.-y., et al., Anti-inflammatory properties of uvaol on DSS-induced colitis and LPS-stimulated macrophages. Chinese Medicine, 2020. 15(1): p. 1-13.

54. Lafi, O., et al., Synergistic antileishmanial activity of erythrodiol, uvaol, and oleanolic acid isolated from olive leaves of cv. Chemlali. 3 Biotech, 2023. 13(12): p. 395.

55. Saudagar, P. and V.K. Dubey, Cloning, expression, characterization and inhibition studies on trypanothione synthetase, a drug target enzyme, from Leishmania donovani. 2011.

56. da Silva Filho, A., et al., In vitro antileishmanial, antiplasmodial and cytotoxic activities of phenolics and triterpenoids from Baccharis dracunculifolia DC (Asteraceae). Fitoterapia, 2009. 80(8): p. 478-482.

Downloads

Published

2025-09-30

Issue

Section

Articles

How to Cite

1.
Maryam, Fawad Ali, Sajid Hussain, Neelum Gul Qazi, Abdul Wahab, Naseebullah, et al. Investigating the Anti-Leishmanial Potential of Phytoconstituents Derived from Anchusa arvensis: In-vitro and Network Pharmacology Study. JHWCR [Internet]. 2025 Sep. 30 [cited 2025 Nov. 29];3(13):e824. Available from: https://jhwcr.com/index.php/jhwcr/article/view/824

Most read articles by the same author(s)