Skip to main content
Log in

In silico prediction of a new lead compound targeting enolase of trypanosomatids through structure-based virtual screening and molecular dynamic studies

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

Abstract

Enolase is one of the key glycolytic metalloenzyme in many organisms, and it is a potential therapeutic target including trypanosomatids. Sequence and structural analysis of enolase of Trypanosoma bruzi (TbENO), Trypanosoma cruzi (TcENO) and Leishmania donovani (LdENO) revealed conserved sequence pattern and structural features. Hence identification of an inhibitor against enolase of one trypanosomatid organism may have similar effects on enolase of homologous organisms belonging to same family. In the process to identify potent inhibitor compounds against TbENO by in silico methods, compounds containing the substructures of substrate, i.e. phosphoenolpyruvate (PEP) and the well-known inhibitors, fluoro-2-phosphono-acetohydroxamate (FPAH) and phosphono-acetohydroxamate (PAH), were collected. Virtual screening and induced fit docking studies were carried out to explore compounds that have better binding affinity than PEP and FPAH. PPPi was found to be the top hit exhibiting significant binding affinity towards enolase. Glide energy values of two other compounds represented by PubChem ID: 511392 and 101803456 was in good agreement with PEP and PAH. TbENO-PPPi complex was subjected to molecular orbital analysis and molecular dynamic studies by considering its remarkable binding affinity as it could be a potent inhibitor of enolase. Despite being an endogenous compound, based on the results of this study, we highlight PPPi to be a lead compound, and its structure can be treated as a model for further chemical modifications to obtain more potent antagonists.

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

References

  1. Podlipaev S (2001) The more insect trypanosomatids under study-the more diverse Trypanosomatidae appears. Int J Parasitol 31:648–652. https://doi.org/10.1016/S0020-7519(01)00139-4

    Article  CAS  PubMed  Google Scholar 

  2. Nussbaum K, Honek J, Cadmus C, Efferth T (2010) Trypanosomatid parasites causing neglected diseases. Curr Med Chem 17:1594–1617. https://doi.org/10.2174/092986710790979953

    Article  CAS  PubMed  Google Scholar 

  3. Singh N, Kumar M, Singh RK (2012) Leishmaniasis : current status of available drugs and new potential drug targets. Asian Pac J Trop Med 5:485–497. https://doi.org/10.1016/S1995-7645(12)60084-4

    Article  CAS  PubMed  Google Scholar 

  4. Lebioda L, Stec B (1991) Mechanism of Enolase: the crystal structure of Enolase-Mg2+-2-Phosphoglycerate/Phosphoenolpyruvate complex at 2.2-Å resolution. Biochemistry 30:2817–2822. https://doi.org/10.1021/bi00225a012

    Article  CAS  PubMed  Google Scholar 

  5. Schulz EC, Tietzel M, Tovy A et al (2011) Structure analysis of Entamoeba histolytica enolase. Acta Crystallogr Sect D Biol Crystallogr D67:619–627. https://doi.org/10.1107/S0907444911016544

    Article  CAS  Google Scholar 

  6. Wu Y, Wang C, Lin S et al (2015) Octameric structure of Staphylococcus aureus enolase in complex with phosphoenolpyruvate. Acta Crystallogr Sect D Biol Crystallogr 71:2457–2470. https://doi.org/10.1107/S1399004715018830

    Article  CAS  Google Scholar 

  7. Ehinger S, Schubert WD, Bergmann S et al (2004) Plasmin(ogen)-binding α-Enolase from Streptococcus pneumoniae: crystal structure and evaluation of plasmin(ogen)-binding sites. J Mol Biol 343:997–1005. https://doi.org/10.1016/j.jmb.2004.08.088

    Article  CAS  PubMed  Google Scholar 

  8. Kang HJ, Jung SK, Kim SJ, Chung SJ (2008) Structure of human α-enolase (hENO1), a multifunctional glycolytic enzyme. Acta Crystallogr Sect D Biol Crystallogr D64:651–657. https://doi.org/10.1107/S0907444908008561

    Article  CAS  Google Scholar 

  9. Cork AJ, Ericsson DJ, Law RHP et al (2015) Stability of the octameric structure affects plasminogen-binding capacity of streptococcal enolase. PLoS One 10:1–18. https://doi.org/10.1371/journal.pone.0121764

    Article  CAS  Google Scholar 

  10. Vanegas G, Quiñones W, Carrasco-López C et al (2007) Enolase as a plasminogen binding protein in Leishmania mexicana. Parasitol Res 101:1511–1516. https://doi.org/10.1007/s00436-007-0668-7

    Article  PubMed  Google Scholar 

  11. Almeida L, Vanegas G, Calcagno M et al (2004) Plasminogen interaction with Trypanosoma cruzi. Mem Inst Oswaldo Cruz 99:63–67. https://doi.org/10.1590/S0074-02762004000100011

    Article  CAS  PubMed  Google Scholar 

  12. Antúnez K, Anido M, Arredondo D et al (2011) Paenibacillus larvae enolase as a virulence factor in honeybee larvae infection. Vet Microbiol 147:83–89. https://doi.org/10.1016/j.vetmic.2010.06.004

    Article  CAS  PubMed  Google Scholar 

  13. Capello M, Ferri-Borgogno S, Cappello P, Novelli F (2011) α-Enolase: a promising therapeutic and diagnostic tumor target. FEBS J 278:1064–1074. https://doi.org/10.1111/j.1742-4658.2011.08025.x

    Article  CAS  PubMed  Google Scholar 

  14. Weng Y, Chen F, Liu Y et al (2016) Pseudomonas aeruginosa enolase influences bacterial tolerance to oxidative stresses and virulence. Front Microbiol 7:1–12. https://doi.org/10.3389/fmicb.2016.01999

    Article  Google Scholar 

  15. Kühnel K, Luisi BF (2001) Crystal structure of the Escherichia coli RNA degradosome component enolase. J Mol Biol 313:583–592. https://doi.org/10.1006/jmbi.2001.5065

    Article  CAS  PubMed  Google Scholar 

  16. Verlinde CLMJ, Hannaert V, Blonski C et al (2001) Glycolysis as a target for the design of new anti-trypanosome drugs. Drug Resist Updat 4:50–65. https://doi.org/10.1054/drup.2000.0177

    Article  CAS  PubMed  Google Scholar 

  17. Hannaert V, Albert MA, Rigden DJ et al (2003) Kinetic characterization, structure modelling studies and crystallization of Trypanosoma brucei enolase. Eur J Biochem 270:3205–3213. https://doi.org/10.1046/j.1432-1033.2003.03692.x

    Article  CAS  PubMed  Google Scholar 

  18. Engel JC, Franke de Cazzulo BM, Stoppani AOM et al (1987) Aerobic glucose fermentation by Trypanosoma cruzi axenic culture amastigote-like forms during growth and differentiation to epimastigotes. Mol Biochem Parasitol 26:1–10. https://doi.org/10.1016/0166-6851(87)90123-X

    Article  CAS  PubMed  Google Scholar 

  19. Albert MA, Haanstra JR, Hannaert V et al (2005) Experimental and in silico analyses of glycolytic flux control in bloodstream form Trypanosoma brucei. J Biol Chem 280:28306–28315. https://doi.org/10.1074/jbc.M502403200

    Article  CAS  PubMed  Google Scholar 

  20. Naderer T, Ellis MA, Sernee MF et al (2006) Virulence of Leishmania major in macrophages and mice requires the gluconeogenic enzyme fructose-1,6-bisphosphatase. Proc Natl Acad Sci 103:5502–5507. https://doi.org/10.1073/pnas.0509196103

    Article  CAS  PubMed  Google Scholar 

  21. Maldonado J, Marina C, Puig J et al (2006) A study of cutaneous lesions caused by Leishmania mexicana in plasminogen-deficient mice. Exp Mol Pathol 80:289–294. https://doi.org/10.1016/j.yexmp.2005.06.005

    Article  CAS  PubMed  Google Scholar 

  22. Cimasoni G (1972) The inhibition of Enolase by fluoride in vitro. Caries Res 6:93–102. https://doi.org/10.1159/000259782

    Article  CAS  PubMed  Google Scholar 

  23. Qin J, Chai G, Brewer JM et al (2006) Fluoride inhibition of Enolase: crystal structure and thermodynamics. Biochemistry 45:793–800. https://doi.org/10.1021/bi051558s

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Anderson VE, Weiss PM, Cleland WW (1984) Reaction intermediate analogues for Enolase. Biochemistry 23:2779–2786. https://doi.org/10.1021/bi00307a038

    Article  CAS  PubMed  Google Scholar 

  25. de AS Navarro MV, Gomes Dias SM, Mello LV et al (2007) Structural flexibility in Trypanosoma brucei enolase revealed by X-ray crystallography and molecular dynamics. FEBS J 274:5077–5089. https://doi.org/10.1111/j.1742-4658.2007.06027.x

    Article  CAS  Google Scholar 

  26. Leonard PG, Satani N, Maxwell D et al (2016) SF2312 is a natural phosphonate inhibitor of Enolase Paul. Nat Chem Biol 12:1053–1058. https://doi.org/10.1038/nchembio.2195

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Muller et al (2018) Enolase inhibitors and methods of treatment therewith. Global Patent Index - EP 3268376 A1

  28. Avilán L, Gualdrón-López M, Quiñones W et al (2011) Enolase: a key player in the metabolism and a probable virulence factor of trypanosomatid parasites - perspectives for its use as a therapeutic target. Enzyme Res 2011. https://doi.org/10.4061/2011/932549

    Article  Google Scholar 

  29. Cáceres AJ, Portillo R, Acosta H et al (2003) Molecular and biochemical characterization of hexokinase from Trypanosoma cruzi. Mol Biochem Parasitol 126:251–262. https://doi.org/10.1016/S0166-6851(02)00294-3

    Article  CAS  PubMed  Google Scholar 

  30. Sievers F, Wilm A, Dineen D et al (2011) Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal omega. Mol Syst Biol 7. https://doi.org/10.1038/msb.2011.75

    Article  Google Scholar 

  31. Robert X, Gouet P (2014) Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res 42:320–324. https://doi.org/10.1093/nar/gku316

    Article  CAS  Google Scholar 

  32. Protein Preparation Wizard [Schrödinger Suite 2017–2] Schrödinger, LLC, NewYork, NY, 2017

  33. Jorgensen WL, Tirado-rives J (1988) The OPLS potential functions for proteins. Energy minimizations for crystals of cyclic peptides and Crambin. J Am Chem Soc 110:1657–1666. https://doi.org/10.1021/ja00214a001

    Article  CAS  Google Scholar 

  34. Da Silva Giotto MT, Hannaert V, Vertommen D et al (2003) The crystal structure of Trypanosoma brucei enolase: visualisation of the inhibitory metal binding site III and potential as target for selective, irreversible inhibition. J Mol Biol 331:653–665. https://doi.org/10.1016/S0022-2836(03)00752-6

    Article  CAS  PubMed  Google Scholar 

  35. SiteMap [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  36. LigPrep [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  37. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46:3–26. https://doi.org/10.1016/S0169-409X(96)00423-1

    Article  CAS  PubMed  Google Scholar 

  38. Epik [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  39. Glide [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  40. Induced Fit Docking [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  41. Yang Z, Lasker K, Schneidman-duhovny D et al (2012) UCSF chimera, MODELLER, and IMP : an integrated modeling system. J Struct Biol J 179:269–278. https://doi.org/10.1016/j.jsb.2011.09.006

    Article  CAS  Google Scholar 

  42. Melo F, Sánchez R, Sali A (2002) Statistical potentials for fold assessment. Protein Sci 11:430–448. https://doi.org/10.1002/pro.110430

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pieper U, Webb BM, Barkan DT et al (2011) ModBase, a database of annotated comparative protein structure models, and associated resources. Nucleic Acids Res 39:465–474. https://doi.org/10.1093/nar/gkq1091

    Article  CAS  Google Scholar 

  44. Laskowski AR (1993) PROCHECK: a program to check the stereochemical quality of protein structures 26:283–291. https://doi.org/10.1107/S0021889892009944

    Article  CAS  Google Scholar 

  45. Arnold K, Bordoli L, Kopp J, Schwede T (2006) The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling. Bioinformatics 22:195–201. https://doi.org/10.1093/bioinformatics/bti770

    Article  CAS  PubMed  Google Scholar 

  46. Wiederstein M, Sippl MJ (2007) ProSA-web : interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Res 35:407–410. https://doi.org/10.1093/nar/gkm290

    Article  Google Scholar 

  47. Jaguar [Schrödinger Suite 2017–2] Schrödinger, LLC, New York, NY, 2017

  48. Becke AD (2014) A new mixing of Hartree–Fock and local density-functional theories. J Chem Phys 98:1372–1377. https://doi.org/10.1063/1.464304

    Article  Google Scholar 

  49. Lindahl E, Hess B (2001) GROMACS 3.0 : a package for molecular simulation and trajectory analysis. J Mol Model 7:306–317. https://doi.org/10.1007/s008940100045

    Article  CAS  Google Scholar 

  50. Van Der Spoel D, Lindahl E, Hess B, Groenhof G (2005) GROMACS: fast, flexible, and free. J Comput Chem 26:1701–1718. https://doi.org/10.1002/jcc.20291

    Article  CAS  Google Scholar 

  51. Jo S, Kim T, Iyer VG, Im W (2008) Software news and updates CHARMM-GUI : a web-based graphical user Interface for CHARMM. J Comput Chem 29:1859–1865. https://doi.org/10.1002/jcc.20945

    Article  CAS  PubMed  Google Scholar 

  52. Lee J, Cheng X, Swails JM et al (2016) CHARMM-GUI input generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM simulations using the CHARMM36 additive force field. J Chem Theory Comput 12:405–413. https://doi.org/10.1021/acs.jctc.5b00935

    Article  CAS  PubMed  Google Scholar 

  53. Vanommeslaeghe K, Hatcher E, Acharya C et al (2010) CHARMM general force field (CGenFF): a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J Comput Chem 31:671–690. https://doi.org/10.1002/jcc.21367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Floden AM, Watt JA, Brissette CA (2011) Borrelia burgdorferi Enolase is a surface-exposed plasminogen binding protein. PLoS One 6:1–9. https://doi.org/10.1371/journal.pone.0027502

    Article  CAS  Google Scholar 

  55. Hsiao K, Shih N, Fang H et al (2013) Surface α-Enolase promotes extracellular matrix degradation and tumor metastasis and represents a new therapeutic target. PLoS One 8:1–15. https://doi.org/10.1371/journal.pone.0069354

    Article  CAS  Google Scholar 

  56. Fujii A, Yoneda M, Ito T et al (2005) Autoantibodies against the amino terminal of α-enolase are a useful diagnostic marker of Hashimoto’s encephalopathy. J Neuroimmunol 162:130–136. https://doi.org/10.1016/j.jneuroim.2005.02.004

    Article  CAS  PubMed  Google Scholar 

  57. Saranya M, Ayyappan S, Nithya R et al (2018) Molecular structure, NBO and HOMO-LUMO analysis of quercetin on single layer graphene by density functional theory. Dig J Nanomater Biostruct 13:97–105

    Google Scholar 

  58. Pearson RG (1986) Absolute electronegativity and hardness correlated with molecular orbital theory. Proc Natl Acad Sci U S A 83:8440–8441

    Article  CAS  Google Scholar 

Download references

Funding

KP gratefully acknowledges the Department of Biotechnology (DBT), Government of India for the financial support in the form of grants (No.BT/273/NE/TBP/2011) under North Eastern Region Twinning Programme. KP thanks DST-FIST, Government of India for computing facility sanctioned to the department (No: SR/FST/LSII-037/2014 (C) dt.29.03.2016). VMV thanks DBT for the fellowship. Dr. BSL acknowledges the financial support of the DBT – BUILDER (BT/PR12153/INF/22/200/2014) programme for the computational resources and the HPC computing facility at Anna University.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Karthe Ponnuraj.

Additional information

Publisher’s note

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

A.Electronic supplementary material

ESM 1

(DOCX 4161 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vidhya, V.M., Lakshmi, B.S. & Ponnuraj, K. In silico prediction of a new lead compound targeting enolase of trypanosomatids through structure-based virtual screening and molecular dynamic studies. J Mol Model 26, 23 (2020). https://doi.org/10.1007/s00894-019-4284-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00894-019-4284-0

Keywords

Navigation