Skip to main content
Log in

Pharmacophore modeling, molecular docking, and molecular dynamics simulation approaches for identifying new lead compounds for inhibiting aldose reductase 2

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

Abstract

Aldose reductase 2 (ALR2), which catalyzes the reduction of glucose to sorbitol using NADP as a cofactor, has been implicated in the etiology of secondary complications of diabetes. A pharmacophore model, Hypo1, was built based on 26 compounds with known ALR2-inhibiting activity values. Hypo1 contains important chemical features required for an ALR2 inhibitor, and demonstrates good predictive ability by having a high correlation coefficient (0.95) as well as the highest cost difference (128.44) and the lowest RMS deviation (1.02) among the ten pharmacophore models examined. Hypo1 was further validated by Fisher’s randomization method (95%), test set (r = 0.91), and the decoy set shows the goodness of fit (0.70). Furthermore, during virtual screening, Hypo1 was used as a 3D query to screen the NCI database, and the hit leads were sorted by applying Lipinski’s rule of five and ADME properties. The best-fitting leads were subjected to docking to identify a suitable orientation at the ALR2 active site. The molecule that showed the strongest interactions with the critical amino acids was used in molecular dynamics simulations to calculate its binding affinity to the candidate molecules. Thus, Hypo1 describes the key structure–activity relationship along with the estimated activities of ALR2 inhibitors. The hit molecules were searched against PubChem to find similar molecules with new scaffolds. Finally, four molecules were found to satisfy all of the chemical features and the geometric constraints of Hypo1, as well as to show good dock scores, PLPs and PMFs. Thus, we believe that Hypo1 facilitates the selection of novel scaffolds for ALR2, allowing new classes of ALR2 inhibitors to be designed.

Pharmacophore model was generated and validated using various potent techniques. The validated Hypo1 was used in virtual screening to find a novel compounds and subjected to molecular docking and dynamics simulations

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
Fig. 10
Fig. 11

Similar content being viewed by others

Abbreviations

ADME:

Absorption, distribution, metabolism, and excretion

ALR2:

Aldose reductase 2

BBB:

Blood–brain barrier

DS:

Discovery Studio v.2.5

EF:

Enrichment factor

GF:

Goodness of fit

HBA:

Hydrogen bond acceptor

HAli:

Hydrophobic aliphatic

HAro:

Hydrophobic aromatic

MD:

Molecular dynamics

NI:

Negative ionization

NADPH:

Nicotinamide dinucleotide

53N:

3-[5-(3-Nitrophenyl) thiophen-2-yl] propanoic acid

PME:

Particle mesh Ewald

PLP:

Piecewise linear potential

PMF:

Potential of mean force

RA:

Ring aromatic

RMS:

Root mean square

RMSD:

Root mean square deviation

RMSF:

Root mean square fluctuation

TIM:

Triose phosphate isomerase

VMD:

Visual molecular dynamics

References

  1. National Institute of Diabetes and Digestive and Kidney Diseases (2011) National Diabetes Information Clearinghouse (NDIC). http://diabetes.niddk.nih.gov

  2. Sato S, Kador PF (1990) Inhibition of aldehyde reductase by aldose reductase inhibitors. Biochem Pharmacol 40:1033–1042

    Article  CAS  Google Scholar 

  3. Carper DA, Wistow G, Nishimura C, Graham C, Watanabe K, Fujii Y, Hayashi H, Hayaishi O (1989) A superfamily of NADPH-dependent reductases in eukaryotes and prokaryotes. Exp Eye Res 49:377–388

    Article  CAS  Google Scholar 

  4. Costantino L, Rastelli G, Cignarella G, Vianello P, Barlocco D (1997) New aldose reductase inhibitors as potential agents for the prevention of long-term diabetic complications. Expert Opin Ther Patents 7:843–858

    Article  CAS  Google Scholar 

  5. Singh SB, Malamas MS, Hohman TC, Nilakantan R, Carper DA, Kitchen D (2000) Molecular modeling of the aldose reductase–inhibitor complex based on the X-ray crystal structure and studies with single-site-directed mutants. J Med Chem 43:1062–1070

    Google Scholar 

  6. Nishimura C, Matsuura Y, Kokai Y, Akera T, Carper D, Morjana N, Lyons C, Flynn TG (1990) Cloning and expression of human aldose reductase. J Biol Chem 17:9788–9792

    Google Scholar 

  7. Sotriffer CA, Krämer O, Klebe G (2004) Probing flexibility and “induced-fit” phenomena in aldose reductase by comparative crystal structure analysis and molecular dynamics simulations. Proteins Struct Funct Bioinf 56:52–66

    Article  CAS  Google Scholar 

  8. Fournier B, Bendeif EE, Bt G, Podjarny A, Lecomte C, Jelsch C (2009) Charge density and electrostatic interactions of fidarestat, an inhibitor of human aldose reductase. J Am Chem Soc 131:10929–10941. doi:10.1021/ja8095015

    Google Scholar 

  9. Gual P, Le Marchand-Brustel Y, Tanti JF (2003) Positive and negative regulation of glucose uptake by hyperosmotic stress. Diabetes Metab 29:566–575

    Article  CAS  Google Scholar 

  10. Oates PJ (2002) Polyol pathway and diabetic peripheral neuropathy. Int Rev Neurobiol 50:325–328

    Google Scholar 

  11. Eisenmann M, Steuber H, Zentgraf M, Altenkamper M, Ortmann R, Perruchon J, Klebe G, Schlitzer M (2009) Structure-based optimization of aldose reductase inhibitors originating from virtual screening. Chem Med Chem 4:809–819

    Article  CAS  Google Scholar 

  12. Mc VZ, Doan B, Dr S, Sredy J, Podjarny AD (2009) Discovery of [3-(4,5,7-trifluoro-benzothiazol-2-ylmethyl)-pyrrolo[2,3-b]pyridin-1-yl]acetic acids as highly potent and selective inhibitors of aldose reductase for treatment of chronic diabetic complications. Bioorg Med Chem Lett 7:2006–2008

    Google Scholar 

  13. Petrova T, Lunin VY, Ginell S, Hazemann I, Lazarski K, Mitschler A, Podjarny A, Joachimiak A (2009) X-ray-radiation-induced cooperative atomic movements in protein. J Mol Biol 387:1092–1105

    Google Scholar 

  14. El-Kabbani O, Darmanin C, Schneider TR, Hazemann I, Ruiz F, Oka M, Joachimiak A, Schulze-Briese C, Tomizaki T, Mitschler A, Podjarny A (2004) Ultrahigh resolution drug design II. Atomic resolution structures of human aldose reductase holoenzyme complexed with fidarestat and minalrestat: implications for the binding of cyclic imide inhibitors. Proteins Struct Funct Bioinf 55:805–813

    Google Scholar 

  15. El-Kabbani O, Dk W, Petrash M, Quiocho FA (1998) Structural features of the aldose reductase and aldehyde reductase inhibitor-binding sites. Mol Vision 4:19–25

    CAS  Google Scholar 

  16. Oka M, Matsumoto Y, Sugiyama S, Tsuruta N, Matsushima M (2000) A potent aldose reductase inhibitor, (2S,4S)-6-fluoro-2′,5′-dioxospiro[chroman-4,4′-imidazolidine]-2-carboxamide (fidarestat): its absolute configuration and interactions with the aldose reductase by X-ray crystallography. J Med Chem 43:2479–2483

    Google Scholar 

  17. Steuber H, Zentgraf M, Podjarny A, Heine A, Klebe G (2006) High-resolution crystal structure of aldose reductase complexed with the novel sulfonyl-pyridazinone inhibitor exhibiting an alternative active site anchoring group. J Mol Biol 356:45–56

    Google Scholar 

  18. Ramana KV, Srivastava SK (2010) Aldose reductase: a novel therapeutic target for inflammatory pathologies. Int J Biochem Cell Biol 42:17–20

    Google Scholar 

  19. Miyamoto S (2002) Molecular modeling and structure-based drug discovery studies of aldose reductase inhibitors. Chem-Bio Inf J 2:74–85

    Google Scholar 

  20. Accelrys Software, Inc. (2011) Corporate homepage. http://www.accelrys.com

  21. Mylari BL, Larson ER, Beyer TA, Zembrowski WJ, Aldinger CE, Dee MF, Siegel TW, Singleton DH (1991) Novel, potent aldose reductase inhibitors: 3,4-dihydro-4-oxo-3-[[5-(trifluoromethyl)-2-benzothiazolyl]methyl]-1-phthalazineacetic acid (zopolrestat) and congeners. J Med Chem 34:108–122

    Article  CAS  Google Scholar 

  22. Mylari BL, Zembrowski WJ, Beyer TA, Aldinger CE, Siegel TW (1992) Orally active aldose reductase inhibitors: indazoleacetic, oxopyridazineacetic, and oxopyridopyridazineacetic acid derivatives. J Med Chem 35:2155–2162

    Article  CAS  Google Scholar 

  23. Lipinski CA, Aldinger CE, Beyer TA, Bordner J, Burdi DF, Bussolotti DL, Inskeep PB, Siegel TW (1992) Hydantoin bioisosteres. In vivo active spiro hydroxy acetic acid aldose reductase inhibitors. J Med Chem 35:2169–2177

    Article  CAS  Google Scholar 

  24. Advanced Chemistry Development, Inc. (2010) Homepage. http://www.acdlabs.com

  25. Brooks BR, Brooks CL 3rd, Mackerell AD Jr, Nilsson L, Petrella RJ, Roux B, Won Y, Archontis G, Bartels C, Boresch S, Caflisch A, Caves L, Cui Q, Dinner AR, Feig M, Fischer S, Gao J, Hodoscek M, Im W, Kuczera K, Lazaridis T, Ma J, Ovchinnikov V, Paci E, Pastor RW, Post CB, Pu JZ, Schaefer M, Tidor B, Venable RM, Woodcock HL, Wu X, Yang W, York DM, Karplus M (2009) CHARMM: the biomolecular simulation program. J Comput Chem 30:1545–1614

    Google Scholar 

  26. Brooks BR, Bruccoleri RE, Olafson BD, States DJ, Swaminathan S, Karplus M (1983) CHARMM: a program for macromolecular energy, minimization, and dynamics calculations. J Comput Chem 4:187–217

    Google Scholar 

  27. MacKerell AD, Brooks CL, Nilsson L, Roux B, Won Y, Karplus M (1998) CHARMM: the energy function and its paramerterization with an overview of the program. In: Schleyer (ed) The encyclopedia of computational chemistry. Wiley, Chichester, pp 271–277

  28. Smellie A, Teig SL, Towbin P (1995) Poling: promoting conformational variation. J Comput Chem 16:171–187

    Google Scholar 

  29. Sakkiah S, Thangapandian S, John S, Lee KW (2011) Pharmacophore based virtual screening, molecular docking studies to design potent heat shock protein 90 inhibitors. Eur J Med Chem 46:2937–2947

    Article  CAS  Google Scholar 

  30. 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

    Google Scholar 

  31. Sakkiah S, Thangapandian S, John S, Kwon YJ, Lee KW (2010) 3D QSAR pharmacophore based virtual screening and molecular docking for identification of potential HSP90 inhibitors. Eur J Med Chem 45:2132–2140

    Google Scholar 

  32. Berman HM (2008) The Protein Data Bank: a historical perspective. Acta Crystallogr A 64:88–95

    Article  Google Scholar 

  33. The PDB Team (2003) The Protein Data Bank. Methods Biochem Anal 44:181–198

  34. Berendsen HJC, van der Spoel D, van Drunen R (1995) GROMACS: a message-passing parallel molecular dynamics implementation. Comput Phys Commun 91:43–56

    Google Scholar 

  35. Spoel DVD, Patriksson A, Seibert MM (2007) Protein folding properties from molecular dynamics simulations. In: Proc 8th Int Conf on Applied Parallel Computing: State of the Art in Scientific Computing (PARA 2006), Umeå, Sweden, 18–21 June 2006

  36. Schuttelkopf AW, van Aalten DMF (2004) PRODRG: a tool for high-throughput crystallography of protein–ligand complexes. Acta Crystallogr D 60:1355–1363

    Google Scholar 

  37. Aalten DMF, Bywater R, Findlay JBC, Hendlich M, Hooft RWW, Vriend G (1996) PRODRG, a program for generating molecular topologies and unique molecular descriptors from coordinates of small molecules. J Comput Aided Mol Des 10:255–262

    Article  Google Scholar 

  38. Ryckaert JP, Ciccotti G, Berendsen HJC (1977) Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes. J Comput Phys 23:327–341

    Google Scholar 

  39. Darden T, York D, Pedersen L (1993) Particle mesh Ewald: an N.log(N) method for Ewald sums in large systems. J Chem Phys 98:10089–10092

    Google Scholar 

  40. Hess B, Bekker H, Berendsen H, Fraaije J (1997) LINCS: A linear constraint solver for molecular simulations. J Comput Chem 18:1463–1472

    Article  CAS  Google Scholar 

  41. Venkatachalam CM, Jiang X, Oldfield T, Waldman M (2003) LigandFit: a novel method for the shape-directed rapid docking of ligands to protein active sites. J Mol Graph Model 21:289–307

    Article  CAS  Google Scholar 

Download references

Acknowledgments

This research was supported by the Basic Science Research Program (2009–0073267), the Pioneer Research Center Program (2009–0081539), and the Management of Climate Change Program (2010–0029084) through the National Research Foundation of Korea (NRF), as funded by the Ministry of Education, Science and Technology (MEST) of the Republic of Korea. This work was also supported by the Next-Generation BioGreen21 Program (PJ008038) from the Rural Development Administration (RDA) of the Republic of Korea.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Keun Woo Lee.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sakkiah, S., Thangapandian, S. & Lee, K.W. Pharmacophore modeling, molecular docking, and molecular dynamics simulation approaches for identifying new lead compounds for inhibiting aldose reductase 2. J Mol Model 18, 3267–3282 (2012). https://doi.org/10.1007/s00894-011-1247-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00894-011-1247-5

Keywords

Navigation