Skip to main content

Advertisement

Log in

Identification of the Seaweed Metabolites as Potential Anti-tubercular Agents Against Human Pantothenate synthetase: An In Silico Approach

  • Published:
Current Microbiology Aims and scope Submit manuscript

Abstract

Tuberculosis is the disease which is caused due to the contagion of Mycobacterium tuberculosis. The multidrug resistance Mycobacterium tuberculosis is the main hassle in the treatment of this worldwide health threats. Pantothenate synthase is a legitimate goal for rational drug designing against Mycobacterium tuberculosis. The enzyme is most active in the presence of magnesium or manganese. Marine algal cell wall is rich in sulfated polysaccharides such as fucoidans (brown algae), κ-carrageenans (red algae), and ulvan (green algae) with various favorable biological activities such as anticoagulant, antiviral, antioxidative, anticancer, and immunomodulating activities. In this study, we have modeled binding modes of selected known anti-tubercular compounds and different solvent extract against pantothenate synthase using advanced docking program AutoDock 4.2 tool. In our current study, in silico experiments were carried out to determine if fucoidan, κ-carrageenan, and ulvan sulfated polysaccharides could be a potential target against PANc (pantothenate synthetase), with the goal of identifying potential inhibitors as anti-TB leads targeting PANc for further wet lab validation. Two bioactive compounds were docked to the Mtb pantothenate synthetase protein binding site, with docking scores ranging from − 5.57 to − 2.73. κ-carrageenan had the best pose and docking score, with a Ligand fit score of − 5.815. Ulvan did not dock with the protein. The molecular dynamics simulations were conducted with substrate and ligand bounded fucoidan and κ-carrageenan for 150 ns and the protein Mtb pantothenate synthetase showed a stable conformation in the simulation, with tight amino acid contributions binding to the ligand molecule. RMSD characterizes the conformation and stability of protein ligand complexes, with higher fluctuations indicating low stability and minimal low-level fluctuations indicating equilibration and stability. The graph for RMSF shows significant peaks due to fluctuations in active site regions and other peaks indicating the adaptation of the ligand molecule to the protein binding pocket. From the molecular dynamics study, it is clear that the compounds are having good binding affinity in the active site. The root mean square deviation, root mean square fluctuations, and radius of gyration are supportive evidences which helped us to conclude that the compounds κ-carrageenan and fucoidan are suitable lead molecules for inhibiting pantothenate synthetase. Based on these evidences, the natural compounds from seaweeds can be tested clinically either alone or in combinations against the protein, which could facilitate the designing or the synthesis of new lead molecules as drugs against the tuberculosis.

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

Similar content being viewed by others

References

  1. Chandra P, Grigsby SJ, Philips JA (2022) Immune evasion and provocation by Mycobacterium tuberculosis. Nat Rev Microbiol 20(12):750–766

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. World Health Organization (2023) Tuberculosis. https://www.who.int/news-room/fact-sheets/detail/tuberculosis. Accessed 24 March 2023

  3. Alemu A, Bitew ZW, Seid G, Diriba G, Gashu E, Berhe N, Mariam SH, Gumi B (2022) Tuberculosis in individuals who recovered from COVID-19: a systematic review of case reports. PLoS ONE 17(11):e0277807

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Global Tuberculosis Report 2019 (World Health Organization, Geneva (2019) Licence: CC BY-NC-SA

  5. Shah YB, Mistry PS, Dhameliya TM, Ranch KM, Boddu SH, Jacob S, Mahalakshmi B, Renukuntla J (2023) Tuberculosis: current treatment options and future scope. In: Shegokar R, Pathak Y (eds) Tubercular drug delivery systems: advances in treatment of infectious diseases. Springer, Cham, pp 59–77

    Chapter  Google Scholar 

  6. Lange C, Aaby P, Behr MA, Donald PR, Kaufmann SH, Netea MG, Mandalakas AM (2022) 100 years of Mycobacterium bovis bacille Calmette-Guérin. Lancet Infect Dis 22(1):2–12. https://doi.org/10.1016/S1473-3099(21)00403-5

    Article  Google Scholar 

  7. Hawn TR, Day TA, Scriba TJ, Hatherill M, Hanekom WA, Evans TG, Churchyard GJ, Kublin JG, Bekker LG, Self SG (2014) Tuberculosis vaccines and prevention of infection. Microbiol Mol Biol Rev 78(4):650–671

    Article  PubMed Central  Google Scholar 

  8. Johnston JC, Cooper R, Menzies D (2022) Chapter 5: Treatment of tuberculosis disease. Can J Respir Crit Care Sleep Med 6(1):66–76

    Google Scholar 

  9. Syed RR, Catanzaro DG, Colman RE, Cooney CG, Linger Y, Kukhtin AV, Holmberg RC, Norville R, Crudu V, Ciobanu N, Codreanu A (2023) Clinical evaluation of the XDR-LFC assay for the molecular detection of isoniazid, rifampin, fluoroquinolone, kanamycin, capreomycin, and amikacin drug resistance in a prospective cohort. J Clin Microbiol 61(3):e01478-e1522

    Article  PubMed  PubMed Central  Google Scholar 

  10. Deshkar AT, Shirure PA, Deshkar A, Shirure P (2022) Bedaquiline: A novel diarylquinoline for multidrug-resistant pulmonary tuberculosis. Cureus. https://doi.org/10.7759/cureus.28519

    Article  PubMed  PubMed Central  Google Scholar 

  11. Dover LG, Coxon GD (2011) Current status and research strategies in tuberculosis drug development: miniperspective. J Med Chem 54(18):6157–6165

    Article  CAS  PubMed  Google Scholar 

  12. Perveen S, Sharma R (2022) Screening approaches and therapeutic targets: the two driving wheels of tuberculosis drug discovery. Biochem Pharmacol 197:114906

    Article  CAS  PubMed  Google Scholar 

  13. Pal A, Kamthania MC, Kumar A (2014) Bioactive compounds and properties of seaweeds—a review. Open Access Library J 1(4):1–7

    Google Scholar 

  14. Pereira L (2018) Therapeutic and nutritional uses of algae. CRC Press, Boca Raton

    Book  Google Scholar 

  15. Knutsen SH, Myslabodski DE, Larsen B, Usov AI (1994) A modified system of nomenclature for red algal galactans. Bot Mar 37:163–169

  16. Conchie J, Percival EG (1950) 167. Fucoidin. Part II. The hydrolysis of a methylated fucoidin prepared from Fucus vesiculosus. J Chem Soc (Resumed) 827–832

  17. Zheng R, Blanchard JS (2001) Steady-state and pre-steady-state kinetic analysis of Mycobacterium tuberculosis pantothenate synthetase. Biochemistry 40(43):12904–12912

    Article  CAS  PubMed  Google Scholar 

  18. Norgan AP, Coffman PK, Kocher JP, Katzmann DJ, Sosa CP (2011) Multilevel parallelization of AutoDock 4.2. J Cheminformatics 3:1–9

    Article  Google Scholar 

  19. Lemkul JA (2018) From proteins to perturbed hamiltonians: a suite of tutorials for the GROMACS-2018 molecular simulation package [article v1. 0]. Living J Comput Mol Sci 1(5068.10):33011

  20. Suresh A, Srinivasarao S, Khetmalis YM, Nizalapur S, Sankaranarayanan M, Sekhar KV (2020) Inhibitors of pantothenate synthetase of Mycobacterium tuberculosis—a medicinal chemist perspective. RSC Adv 10(61):37098–37115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR (2012) Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J Cheminformatics 4(1):1–7

    Article  Google Scholar 

  22. https://autodock.scripps.edu/download-autodock4/

  23. Cheeseright T, Mackey M, Rose S, Vinter A (2006) Molecular field extrema as descriptors of biological activity: definition and validation. J Chem Inf Model 46(2):665–676

    Article  CAS  PubMed  Google Scholar 

  24. Muhammed MT, Aki-Yalcin E (2022) Molecular docking: principles, advances, and its applications in drug discovery. Lett Drug Des Discov 19

  25. Stroganov OV, Novikov FN, Stroylov VS, Kulkov V, Chilov GG (2008) Lead finder: an approach to improve accuracy of protein−ligand docking, binding energy estimation, and virtual screening. J Chem Inf Model 48(12):2371–2385

    Article  CAS  PubMed  Google Scholar 

  26. Naz S, Farooq U, Khan S, Sarwar R, Mabkhot YN, Saeed M, Alsayari A, Muhsinah AB, Ul-Haq Z (2021) Pharmacophore model-based virtual screening, docking, biological evaluation and molecular dynamics simulations for inhibitors discovery against α-tryptophan synthase from Mycobacterium tuberculosis. J Biomol Struct Dyn 39(2):610–620

    Article  CAS  Google Scholar 

  27. Berendsen HJ, Hess B, Lindahl E, Van Der Spoel D, Mark AE, Groenhof G (2005) GROMACS: fast, flexible, and free. J Comput Chem 26(16):1701–1718

    Article  PubMed  Google Scholar 

  28. Lindahl E, Hess B, Van Der Spoel D (2001) GROMACS 3.0: a package for molecular simulation and trajectory analysis. Mol Model Annu 7:306–317

    Article  CAS  Google Scholar 

  29. Elofsson A, Hess B, Lindahl E, Onufriev A, Van der Spoel D, Wallqvist A (2019) Ten simple rules on how to create open access and reproducible molecular simulations of biological systems. PLoS Comput Biol 15(1):e1006649

    Article  PubMed  PubMed Central  Google Scholar 

  30. Website. PDB available online: http://www.rcsb.org/pdb/

Download references

Acknowledgements

The authors wish to thank the researchers supporting Project number RSP-2023R110 at King Saud University Riyadh Saudi Arabia. The authors are grateful to the Science and Engineering Research Board (SERB-Early Career Research Award-ECR/2015/000460), Government of India, New Delhi, for the financial support, and also to the Management of Sathyabama Institute of Science and Technology Chennai, Tamilnadu, for providing the necessary infrastructure facilities to carry out the research work.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rajasekar Thirunavukkarasu.

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 556 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

Arokia Rajan, M., Thirunavukkarasu, R., Joseph, J. et al. Identification of the Seaweed Metabolites as Potential Anti-tubercular Agents Against Human Pantothenate synthetase: An In Silico Approach. Curr Microbiol 80, 318 (2023). https://doi.org/10.1007/s00284-023-03422-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00284-023-03422-w

Navigation