Skip to main content

Molecular Modeling of Tau Proline-Directed Protein Kinase (PDPK) Inhibitors

  • Protocol
  • First Online:
Computational Modeling of Drugs Against Alzheimer’s Disease

Part of the book series: Neuromethods ((NM,volume 132))

Abstract

Proline-directed protein kinases (PDPKs) are protein kinases (PKs) that phosphorylate serine or threonine preceding a proline residue (S/TP motif). It is known that these enzymes are responsible for tau hyperphosphorylation, which is related to the formation of amyloid plaques (APs) and neurofibrillary tangles (NFTs) in Alzheimer’s disease (AD) patients. In this sense, the inhibition of certain PDPKs is considered a promising strategy to elaborate therapies against AD. The structures of the most important PDPKs are available as high-resolution X-ray crystals; many of them are co-complexed with inhibitors. This information is very valuable because it constitutes a source for creating additional structural information with the aid of molecular modeling methods. Molecular modeling includes methodologies such as docking, molecular dynamics (MD), free energy calculations, etc. that allow accessing to other properties such as the effects of water media, conformational sampling, and binding energy estimations. More importantly, these methods allow to use the available structural information for generating the structures of novel complexes, which can contribute to the rational design of novel inhibitors. Here, we extensively reviewed how molecular modeling methods have contributed to the study of complexes between PDPKs and their inhibitors. We also analyzed the interaction fingerprints (IFPs) generated from the available complexes for each PDPK. IFPs capture the most important interactions between proteins and their ligands which can be used for guidance in docking experiments and in silico screening of novel candidates.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 89.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Hickman RA, Faustin A, Wisniewski T (2016) Alzheimer disease and its growing epidemic: risk factors, biomarkers, and the urgent need for therapeutics. Neurol Clin 34:941–953. doi:10.1016/j.ncl.2016.06.009

    Article  PubMed  Google Scholar 

  2. Luo J, Wärmländer SKTS, Gräslund A, Abrahams JP (2016) Cross-interactions between the Alzheimer disease amyloid-β peptide and other amyloid proteins: a further aspect of the amyloid cascade hypothesis. J Biol Chem 291:16485–16493. doi:10.1074/jbc.R116.714576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Edwards G, Moreno-Gonzalez I, Soto C (2016) Amyloid-beta and tau pathology following repetitive mild traumatic brain injury. Biochem Biophys Res Commun 483(4):1137–1142. doi:10.1016/j.bbrc.2016.07.123

    Article  PubMed  CAS  Google Scholar 

  4. Bukar Maina M, Al-Hilaly YK, Serpell LC (2016) Nuclear tau and its potential role in Alzheimer’s disease. Biomol Ther 6:9. doi:10.3390/biom6010009

    Google Scholar 

  5. Martin L, Latypova X, Terro F (2011) Post-translational modifications of tau protein: implications for Alzheimer’s disease. Neurochem Int 58:458–471. doi:10.1016/j.neuint.2010.12.023

    Article  CAS  PubMed  Google Scholar 

  6. Fischer D, Mukrasch MD, Biernat J, Bibow S, Blackledge M, Griesinger C, Mandelkow E, Zweckstetter M (2009) Conformational changes specific for pseudophosphorylation at serine 262 selectively impair binding of tau to microtubules. Biochemistry 48:10047–10055. doi:10.1021/bi901090m

    Article  CAS  PubMed  Google Scholar 

  7. Sengupta A, Kabat J, Novak M, Wu Q, Grundke-Iqbal I, Iqbal K (1998) Phosphorylation of tau at both Thr 231 and Ser 262 is required for maximal inhibition of its binding to microtubules. Arch Biochem Biophys 357:299–309. doi:10.1006/abbi.1998.0813

    Article  CAS  PubMed  Google Scholar 

  8. Hanger DP, Anderton BH, Noble W (2009) Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol Med 15:112–119. doi:10.1016/j.molmed.2009.01.003

    Article  CAS  PubMed  Google Scholar 

  9. Chung S-H (2009) Aberrant phosphorylation in the pathogenesis of Alzheimer’s disease. BMB Rep 42:467–474

    Article  CAS  PubMed  Google Scholar 

  10. Wang JZ, Wu Q, Smith A, Grundke-Iqbal I, Iqbal K (1998) Tau is phosphorylated by GSK-3 at several sites found in Alzheimer disease and its biological activity markedly inhibited only after it is prephosphorylated by A-kinase. FEBS Lett 436:28–34

    Article  CAS  PubMed  Google Scholar 

  11. Liu F, Iqbal K, Grundke-Iqbal I, Gong C-X (2002) Involvement of aberrant glycosylation in phosphorylation of tau by cdk5 and GSK-3beta. FEBS Lett 530:209–214

    Article  CAS  PubMed  Google Scholar 

  12. Johnson GV, Hartigan JA (1999) Tau protein in normal and Alzheimer’s disease brain: an update. J Alzheimers Dis 1:329–351

    Article  CAS  PubMed  Google Scholar 

  13. Godemann R, Biernat J, Mandelkow E, Mandelkow EM (1999) Phosphorylation of tau protein by recombinant GSK-3beta: pronounced phosphorylation at select Ser/Thr-Pro motifs but no phosphorylation at Ser262 in the repeat domain. FEBS Lett 454:157–164

    Article  CAS  PubMed  Google Scholar 

  14. Pei JJ, Grundke-Iqbal I, Iqbal K, Bogdanovic N, Winblad B, Cowburn RF (1998) Accumulation of cyclin-dependent kinase 5 (cdk5) in neurons with early stages of Alzheimer’s disease neurofibrillary degeneration. Brain Res 797:267–277

    Article  CAS  PubMed  Google Scholar 

  15. Feijoo C, Campbell DG, Jakes R, Goedert M, Cuenda A (2005) Evidence that phosphorylation of the microtubule-associated protein tau by SAPK4/p38δ at Thr50 promotes microtubule assembly. J Cell Sci 118:397–408. doi:10.1242/jcs.01655

    Article  CAS  PubMed  Google Scholar 

  16. Wang J-Z, Liu F (2008) Microtubule-associated protein tau in development, degeneration and protection of neurons. Prog Neurobiol 85:148–175. doi:10.1016/j.pneurobio.2008.03.002

    Article  CAS  PubMed  Google Scholar 

  17. Pei J-J, Braak E, Braak H, Grundke-Iqbal I, Iqbal K, Winblad B, Cowburn RF (2001) Localization of active forms of C-jun kinase (JNK) and p38 kinase in Alzheimer’s disease brains at different stages of neurofibrillary degeneration. J Alzheimers Dis 3:41–48

    Article  CAS  PubMed  Google Scholar 

  18. Yoshida H, Hastie CJ, McLauchlan H, Cohen P, Goedert M (2004) Phosphorylation of microtubule-associated protein tau by isoforms of c-Jun N-terminal kinase (JNK). J Neurochem 90:352–358. doi:10.1111/j.1471-4159.2004.02479.x

    Article  CAS  PubMed  Google Scholar 

  19. Cho J-H, Johnson GVW (2004) Primed phosphorylation of tau at Thr231 by glycogen synthase kinase 3beta (GSK3beta) plays a critical role in regulating tau’s ability to bind and stabilize microtubules. J Neurochem 88:349–358

    Article  CAS  PubMed  Google Scholar 

  20. Sabatini S, Manfroni G, Barreca ML, Bauer SM, Gargaro M, Cannalire R, Astolfi A, Brea J, Vacca C, Pirro M, Massari S, Tabarrini O, Loza MI, Fallarino F, Laufer SA, Cecchetti V (2015) The pyrazolobenzothiazine core as a new chemotype of p38 alpha mitogen-activated protein kinase inhibitors. Chem Biol Drug Des 86:531–545. doi:10.1111/cbdd.12516

    Article  CAS  PubMed  Google Scholar 

  21. Boulahjar R, Ouach A, Bourg S, Bonnet P, Lozach O, Meijer L, Guguen-Guillouzo C, Le Guevel R, Lazar S, Akssira M, Troin Y, Guillaumet G, Routier S (2015) Advances in tetrahydropyrido[1,2-a]isoindolone (valmerins) series: potent glycogen synthase kinase 3 and cyclin dependent kinase 5 inhibitors. Eur J Med Chem 101:274–287. doi:10.1016/j.ejmech.2015.06.046

    Article  CAS  PubMed  Google Scholar 

  22. De SK, Stebbins JL, Chen L-H, Riel-Mehan M, Machleidt T, Dahl R, Yuan H, Emdadi A, Barile E, Chen V, Murphy R, Pellecchia M (2009) Design, synthesis, and structure-activity relationship of substrate competitive, selective, and in vivo active triazole and thiadiazole inhibitors of the c-Jun N-terminal kinase. J Med Chem 52:1943–1952. doi:10.1021/jm801503n

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fischer M, Coleman RG, Fraser JS, Shoichet BK (2014) Incorporation of protein flexibility and conformational energy penalties in docking screens to improve ligand discovery. Nat Chem 6:575–583. doi:10.1038/nchem.1954

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Halperin I, Ma B, Wolfson H, Nussinov R (2002) Principles of docking: an overview of search algorithms and a guide to scoring functions. Proteins 47:409–443. doi:10.1002/prot.10115

    Article  CAS  PubMed  Google Scholar 

  25. Ramírez D, Caballero J (2016) Is it reliable to use common molecular docking methods for comparing the binding affinities of enantiomer pairs for their protein target? Int J Mol Sci 17:525. doi:10.3390/ijms17040525

    Article  PubMed Central  CAS  Google Scholar 

  26. Lyne PD, Lamb ML, Saeh JC (2006) Accurate prediction of the relative potencies of members of a series of kinase inhibitors using molecular docking and MM-GBSA scoring. J Med Chem 49:4805–4808. doi:10.1021/jm060522a

    Article  CAS  PubMed  Google Scholar 

  27. Adasme-Carreño F, Muñoz-Gutierrez C, Caballero J, Alzate-Morales J (2014) Performance of the MM/GBSA scoring using a binding site hydrogen bond network-based frame selection: the protein kinase case. Phys Chem Chem Phys 16:14047–14058. doi:10.1039/C4CP01378F

    Article  PubMed  Google Scholar 

  28. Verdonk ML, Berdini V, Hartshorn MJ, Mooij WTM, Murray CW, Taylor RD, Watson P (2004) Virtual screening using protein-ligand docking: avoiding artificial enrichment. J Chem Inf Comput Sci 44:793–806. doi:10.1021/ci034289q

    Article  CAS  PubMed  Google Scholar 

  29. Caballero J, Alzate-Morales JH (2012) Molecular dynamics of protein kinase-inhibitor complexes: a valid structural information. Curr Pharm Des 18:2946–2963

    Article  CAS  PubMed  Google Scholar 

  30. 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. doi:10.1002/jcc.540040211

    Article  CAS  Google Scholar 

  31. Jorgensen WL, Maxwell DS, Tirado-Rives J (1996) Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids. J Am Chem Soc 118:11225–11236. doi:10.1021/ja9621760

    Article  CAS  Google Scholar 

  32. Cornell WD, Cieplak P, Bayly CI, Gould IR, Merz KM, Ferguson DM, Spellmeyer DC, Fox T, Caldwell JW, Kollman PA (1995) A second generation force field for the simulation of proteins, nucleic acids, and organic molecules. J Am Chem Soc 117:5179–5197. doi:10.1021/ja00124a002

    Article  CAS  Google Scholar 

  33. Schuler LD, Daura X, van Gunsteren WF (2001) An improved GROMOS96 force field for aliphatic hydrocarbons in the condensed phase. J Comput Chem 22:1205–1218. doi:10.1002/jcc.1078

    Article  CAS  Google Scholar 

  34. Kollman PA, Massova I, Reyes C, Kuhn B, Huo S, Chong L, Lee M, Lee T, Duan Y, Wang W, Donini O, Cieplak P, Srinivasan J, Case DA, Cheatham TE (2000) Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models. Acc Chem Res 33:889–897. doi:10.1021/ar000033j

    Article  CAS  PubMed  Google Scholar 

  35. Zuccotto F, Ardini E, Casale E, Angiolini M (2010) Through the “gatekeeper door”: exploiting the active kinase conformation. J Med Chem 53:2681–2694. doi:10.1021/jm901443h

    Article  CAS  PubMed  Google Scholar 

  36. Deng Z, Chuaqui C, Singh J (2004) Structural interaction fingerprint (SIFt): a novel method for analyzing three-dimensional protein-ligand binding interactions. J Med Chem 47:337–344. doi:10.1021/jm030331x

    Article  CAS  PubMed  Google Scholar 

  37. Shaw PC, Davies AF, Lau KF, Garcia-Barcelo M, Waye MM, Lovestone S, Miller CC, Anderton BH (1998) Isolation and chromosomal mapping of human glycogen synthase kinase-3 alpha and -3 beta encoding genes. Genome 41:720–727

    CAS  PubMed  Google Scholar 

  38. Beurel E, Michalek SM, Jope RS (2010) Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3). Trends Immunol 31:24–31. doi:10.1016/j.it.2009.09.007

    Article  CAS  PubMed  Google Scholar 

  39. Hernández F, Gómez de Barreda E, Fuster-Matanzo A, Lucas JJ, Avila J (2010) GSK3: a possible link between beta amyloid peptide and tau protein. Exp Neurol 223:322–325. doi:10.1016/j.expneurol.2009.09.011

    Article  PubMed  CAS  Google Scholar 

  40. Lucas JJ, Hernández F, Gómez-Ramos P, Morán MA, Hen R, Avila J (2001) Decreased nuclear beta-catenin, tau hyperphosphorylation and neurodegeneration in GSK-3beta conditional transgenic mice. EMBO J 20:27–39. doi:10.1093/emboj/20.1.27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hetman M, Cavanaugh JE, Kimelman D, Xia Z (2000) Role of glycogen synthase kinase-3beta in neuronal apoptosis induced by trophic withdrawal. J Neurosci 20:2567–2574

    CAS  PubMed  Google Scholar 

  42. Hoshi M, Takashima A, Noguchi K, Murayama M, Sato M, Kondo S, Saitoh Y, Ishiguro K, Hoshino T, Imahori K (1996) Regulation of mitochondrial pyruvate dehydrogenase activity by tau protein kinase I/glycogen synthase kinase 3beta in brain. Proc Natl Acad Sci U S A 93:2719–2723

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hooper C, Killick R, Lovestone S (2008) The GSK3 hypothesis of Alzheimer’s disease. J Neurochem 104:1433–1439. doi:10.1111/j.1471-4159.2007.05194.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mou L, Li M, Lu S-Y, Li S, Shen Q, Zhang J, Li C, Lu X (2014) Unraveling the role of Arg4 and Arg6 in the auto-inhibition mechanism of GSK3β from molecular dynamics simulation. Chem Biol Drug Des 83:721–730. doi:10.1111/cbdd.12286

    Article  CAS  PubMed  Google Scholar 

  45. Ilouz R, Kowalsman N, Eisenstein M, Eldar-Finkelman H (2006) Identification of novel glycogen synthase kinase-3beta substrate-interacting residues suggests a common mechanism for substrate recognition. J Biol Chem 281:30621–30630. doi:10.1074/jbc.M604633200

    Article  CAS  PubMed  Google Scholar 

  46. Lu S-Y, Jiang Y-J, Zou J-W, Wu T-X (2011) Molecular modeling and molecular dynamics simulation studies of the GSK3β/ATP/substrate complex: understanding the unique P+4 primed phosphorylation specificity for GSK3β substrates. J Chem Inf Model 51:1025–1036. doi:10.1021/ci100493j

    Article  CAS  PubMed  Google Scholar 

  47. Lu S-Y, Huang Z-M, Huang W-K, Liu X-Y, Chen Y-Y, Shi T, Zhang J (2013) How calcium inhibits the magnesium-dependent kinase gsk3β: a molecular simulation study. Proteins 81:740–753. doi:10.1002/prot.24221

    Article  CAS  PubMed  Google Scholar 

  48. Lu S-Y, Jiang Y-J, Zou J-W, Wu T-X (2011) Dissection of the difference between the group I metal ions in inhibiting GSK3β: a computational study. Phys Chem Chem Phys 13:7014–7023. doi:10.1039/c0cp02498h

    Article  CAS  PubMed  Google Scholar 

  49. Tang X-N, Lo C-W, Chuang Y-C, Chen C-T, Sun Y-C, Hong Y-R, Yang C-N (2011) Prediction of the binding mode between GSK3β and a peptide derived from GSKIP using molecular dynamics simulation. Biopolymers 95:461–471. doi:10.1002/bip.21603

    Article  CAS  PubMed  Google Scholar 

  50. Zhang N, Jiang Y, Zou J, Zhuang S, Jin H, Yu Q (2007) Insights into unbinding mechanisms upon two mutations investigated by molecular dynamics study of GSK3beta-axin complex: role of packing hydrophobic residues. Proteins 67:941–949. doi:10.1002/prot.21359

    Article  CAS  PubMed  Google Scholar 

  51. Lu S-Y, Jiang Y-J, Zou J-W, Wu T-X (2012) Effect of double mutations K214/A-E215/Q of FRATide on GSK3β: insights from molecular dynamics simulation and normal mode analysis. Amino Acids 43:267–277. doi:10.1007/s00726-011-1070-4

    Article  CAS  PubMed  Google Scholar 

  52. Lu S, Jiang Y, Lv J, Zou J, Wu T (2011) Mechanism of kinase inactivation and nonbinding of FRATide to GSK3β due to K85M mutation: molecular dynamics simulation and normal mode analysis. Biopolymers 95:669–681. doi:10.1002/bip.21629

    Article  CAS  PubMed  Google Scholar 

  53. Lee S-C, Shin D, Cho JM, Ro S, Suh Y-G (2012) Structure-activity relationship of the 7-hydroxy benzimidazole analogs as glycogen synthase kinase 3β inhibitor. Bioorg Med Chem Lett 22:1891–1894. doi:10.1016/j.bmcl.2012.01.065

    Article  CAS  PubMed  Google Scholar 

  54. Wang F, Liu M, Liu J (2012) In silico prediction of inhibitory effects of pyrazol-5-one and indazole derivatives on GSK3β kinase enzyme. J Mol Struct 1024:94–103. doi:10.1016/j.molstruc.2012.05.018

    Article  CAS  Google Scholar 

  55. Caballero J, Zilocchi S, Tiznado W, Collina S, Rossi D (2011) Binding studies and quantitative structure-activity relationship of 3-amino-1H-indazoles as inhibitors of GSK3β. Chem Biol Drug Des 78:631–641. doi:10.1111/j.1747-0285.2011.01186.x

    Article  CAS  PubMed  Google Scholar 

  56. Quesada-Romero L, Caballero J (2014) Docking and quantitative structure–activity relationship of oxadiazole derivates as inhibitors of GSK3beta. Mol Divers 18:149–159. doi:10.1007/s11030-013-9483-5

    Article  CAS  PubMed  Google Scholar 

  57. Quesada-Romero L, Mena-Ulecia K, Tiznado W, Caballero J (2014) Insights into the interactions between maleimide derivates and GSK3β combining molecular docking and QSAR. PLoS One 9:e102212. doi:10.1371/journal.pone.0102212

    Article  PubMed  PubMed Central  Google Scholar 

  58. Sangu S, Vema A, Bigala R (2014) 3D-QSAR and molecular docking studies of quinazoline derivatives as glycogen synthase kinase-3β (Gsk-3β) inhibitors. Pharm Lett 6:289–296

    CAS  Google Scholar 

  59. Withers IM, Mazanetz MP, Wang H, Fischer PM, Laughton CA (2008) Active site pressurization: a new tool for structure-guided drug design and other studies of protein flexibility. J Chem Inf Model 48:1448–1454. doi:10.1021/ci7004725

    Article  CAS  PubMed  Google Scholar 

  60. Mazanetz MP, Withers IM, Laughton CA, Fischer PM (2008) Exploiting glycogen synthase kinase 3beta flexibility in molecular recognition. Biochem Soc Trans 36:55–58. doi:10.1042/BST0360055

    Article  CAS  PubMed  Google Scholar 

  61. Lee H-C, Hsu W-C, Liu A-L, Hsu C-J, Sun Y-C (2014) Using thermodynamic integration MD simulation to compute relative protein-ligand binding free energy of a GSK3β kinase inhibitor and its analogs. J Mol Graph Model 51:37–49. doi:10.1016/j.jmgm.2014.04.010

    Article  PubMed  CAS  Google Scholar 

  62. Pradeep H, Rajanikant GK (2012) A rational approach to selective pharmacophore designing: an innovative strategy for specific recognition of Gsk3β. Mol Divers 16:553–562. doi:10.1007/s11030-012-9387-9

    Article  CAS  PubMed  Google Scholar 

  63. Hanumanthappa P, Krishnamurthy RG (2014) A comparative molecular dynamics simulation study to assess the exclusion ability of novel GSK3β inhibitors. Med Chem Res 23:3092–3095. doi:10.1007/s00044-013-0889-5

    Article  CAS  Google Scholar 

  64. Lu S-Y, Jiang Y-J, Lv J, Zou J-W, Wu T-X (2011) Role of bridging water molecules in GSK3β-inhibitor complexes: insights from QM/MM, MD, and molecular docking studies. J Comput Chem 32:1907–1918. doi:10.1002/jcc.21775

    Article  CAS  PubMed  Google Scholar 

  65. Zhang N, Zhong R, Yan H, Jiang Y (2011) Structural features underlying selective inhibition of GSK3β by dibromocantharelline: implications for rational drug design. Chem Biol Drug Des 77:199–205. doi:10.1111/j.1747-0285.2010.01069.x

    Article  CAS  PubMed  Google Scholar 

  66. Li X, Wang X, Tian Z, Zhao H, Liang D, Li W, Qiu Y, Lu S (2014) Structural basis of valmerins as dual inhibitors of GSK3β/CDK5. J Mol Model 20:2407. doi:10.1007/s00894-014-2407-1

    Article  PubMed  CAS  Google Scholar 

  67. Mazanetz MP, Laughton CA, Fischer PM (2014) Investigation of the flexibility of protein kinases implicated in the pathology of Alzheimer’s disease. Molecules 19:9134–9159. doi:10.3390/molecules19079134

    Article  PubMed  CAS  Google Scholar 

  68. Paglini G, Cáceres A (2001) The role of the Cdk5–p35 kinase in neuronal development. Eur J Biochem 268:1528–1533. doi:10.1046/j.1432-1327.2001.02023.x

    Article  CAS  PubMed  Google Scholar 

  69. Quan H, Wu X, Tian Y, Wang Y, Li C, Li H (2014) Overexpression of CDK5 in neural stem cells facilitates maturation of embryonic neurocytes derived from rats in vitro. Cell Biochem Biophys 69:445–453. doi:10.1007/s12013-014-9816-8

    Article  CAS  PubMed  Google Scholar 

  70. Liu S-L, Wang C, Jiang T, Tan L, Xing A, Yu J-T (2016) The role of Cdk5 in Alzheimer’s disease. Mol Neurobiol 53:4328–4342. doi:10.1007/s12035-015-9369-x

    Article  CAS  PubMed  Google Scholar 

  71. Camins A, Verdaguer E, Folch J, Canudas AM, Pallàs M (2006) The role of CDK5/P25 formation/inhibition in neurodegeneration. Drug News Perspect 19:453–460. doi:10.1358/dnp.2006.19.8.1043961

    Article  CAS  PubMed  Google Scholar 

  72. Tan VBC, Zhang B, Lim KM, Tay TE (2010) Explaining the inhibition of cyclin-dependent kinase 5 by peptides derived from p25 with molecular dynamics simulations and MM-PBSA. J Mol Model 16:1–8. doi:10.1007/s00894-009-0514-1

    Article  CAS  PubMed  Google Scholar 

  73. Cardone A, Brady M, Sriram R, Pant HC, Hassan SA (2016) Computational study of the inhibitory mechanism of the kinase CDK5 hyperactivity by peptide p5 and derivation of a pharmacophore. J Comput Aided Mol Des 30:513–521. doi:10.1007/s10822-016-9922-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Demange L, Abdellah FN, Lozach O, Ferandin Y, Gresh N, Meijer L, Galons H (2013) Potent inhibitors of CDK5 derived from roscovitine: synthesis, biological evaluation and molecular modelling. Bioorg Med Chem Lett 23:125–131. doi:10.1016/j.bmcl.2012.10.141

    Article  CAS  PubMed  Google Scholar 

  75. Dehbi O, Tikad A, Bourg S, Bonnet P, Lozach O, Meijer L, Aadil M, Akssira M, Guillaumet G, Routier S (2014) Synthesis and optimization of an original V-shaped collection of 4-7-disubstituted pyrido[3,2-d]pyrimidines as CDK5 and DYRK1A inhibitors. Eur J Med Chem 80:352–363. doi:10.1016/j.ejmech.2014.04.055

    Article  CAS  PubMed  Google Scholar 

  76. Shrestha S, Natarajan S, Park J-H, Lee D-Y, Cho J-G, Kim G-S, Jeon Y-J, Yeon S-W, Yang D-C, Baek N-I (2013) Potential neuroprotective flavonoid-based inhibitors of CDK5/p25 from Rhus parviflora. Bioorg Med Chem Lett 23:5150–5154. doi:10.1016/j.bmcl.2013.07.020

    Article  CAS  PubMed  Google Scholar 

  77. Pitchuanchom S, Boonyarat C, Forli S, Olson AJ, Yenjai C (2012) Cyclin-dependent kinases 5 template: useful for virtual screening. Comput Biol Med 42:106–111. doi:10.1016/j.compbiomed.2011.10.014

    Article  CAS  PubMed  Google Scholar 

  78. Chatterjee A, Cutler SJ, Doerksen RJ, Khan IA, Williamson JS (2014) Discovery of thienoquinolone derivatives as selective and ATP non-competitive CDK5/p25 inhibitors by structure-based virtual screening. Bioorg Med Chem 22:6409–6421. doi:10.1016/j.bmc.2014.09.043

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang W, Cao X, Zhu X, Gu Y (2013) Molecular dynamic simulations give insight into the mechanism of binding between 2-aminothiazole inhibitors and CDK5. J Mol Model 19:2635–2645. doi:10.1007/s00894-013-1815-y

    Article  CAS  PubMed  Google Scholar 

  80. Dong K, Wang X, Yang X, Zhu X (2016) Binding mechanism of CDK5 with roscovitine derivatives based on molecular dynamics simulations and MM/PBSA methods. J Mol Graph Model 68:57–67. doi:10.1016/j.jmgm.2016.06.007

    Article  CAS  PubMed  Google Scholar 

  81. Wu Q, Kang H, Tian C, Huang Q, Zhu R (2013) Binding mechanism of inhibitors to CDK5/p25 complex: free energy calculation and ranking aggregation analysis. Mol Inform 32:251–260. doi:10.1002/minf.201200139

    Article  CAS  PubMed  Google Scholar 

  82. Patel JS, Berteotti A, Ronsisvalle S, Rocchia W, Cavalli A (2014) Steered molecular dynamics simulations for studying protein-ligand interaction in cyclin-dependent kinase 5. J Chem Inf Model 54:470–480. doi:10.1021/ci4003574

    Article  CAS  PubMed  Google Scholar 

  83. Ul Haq Z, Uddin R, Wai LK, Wadood A, Lajis NH (2011) Docking and 3D-QSAR modeling of cyclin-dependent kinase 5/p25 inhibitors. J Mol Model 17:1149–1161. doi:10.1007/s00894-010-0817-2

    Article  CAS  PubMed  Google Scholar 

  84. Zhang B, Corbel C, Guéritte F, Couturier C, Bach S, Tan VBC (2011) An in silico approach for the discovery of CDK5/p25 interaction inhibitors. Biotechnol J 6:871–881. doi:10.1002/biot.201100139

    Article  CAS  PubMed  Google Scholar 

  85. Atzori C, Ghetti B, Piva R, Srinivasan AN, Zolo P, Delisle MB, Mirra SS, Migheli A (2001) Activation of the JNK/p38 pathway occurs in diseases characterized by tau protein pathology and is related to tau phosphorylation but not to apoptosis. J Neuropathol Exp Neurol 60:1190–1197

    Article  CAS  PubMed  Google Scholar 

  86. Pei J-J, Braak H, An W-L, Winblad B, Cowburn RF, Iqbal K, Grundke-Iqbal I (2002) Up-regulation of mitogen-activated protein kinases ERK1/2 and MEK1/2 is associated with the progression of neurofibrillary degeneration in Alzheimer’s disease. Brain Res Mol Brain Res 109:45–55

    Article  CAS  PubMed  Google Scholar 

  87. Zhu X, Castellani RJ, Takeda A, Nunomura A, Atwood CS, Perry G, Smith MA (2001) Differential activation of neuronal ERK, JNK/SAPK and p38 in Alzheimer disease: the “two hit” hypothesis. Mech Ageing Dev 123:39–46

    Article  CAS  PubMed  Google Scholar 

  88. Munoz L, Ammit AJ (2010) Targeting p38 MAPK pathway for the treatment of Alzheimer’s disease. Neuropharmacology 58:561–568. doi:10.1016/j.neuropharm.2009.11.010

    Article  CAS  PubMed  Google Scholar 

  89. Zhu X, Rottkamp CA, Boux H, Takeda A, Perry G, Smith MA (2000) Activation of p38 kinase links tau phosphorylation, oxidative stress, and cell cycle-related events in Alzheimer disease. J Neuropathol Exp Neurol 59:880–888

    Article  CAS  PubMed  Google Scholar 

  90. Thornton TM, Pedraza-Alva G, Deng B, Wood CD, Aronshtam A, Clements JL, Sabio G, Davis RJ, Matthews DE, Doble B, Rincon M (2008) Phosphorylation by p38 MAPK as an alternative pathway for GSK3beta inactivation. Science 320:667–670. doi:10.1126/science.1156037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Yang Y, Liu H, Yao X (2012) Understanding the molecular basis of MK2-p38α signaling complex assembly: insights into protein-protein interaction by molecular dynamics and free energy studies. Mol BioSyst 8:2106–2118. doi:10.1039/c2mb25042j

    Article  CAS  PubMed  Google Scholar 

  92. Ma Z, Pan Y, Huang W, Yang Y, Wang Z, Li Q, Zhao Y, Zhang X, Shen Z (2014) Synthesis and biological evaluation of the pirfenidone derivatives as antifibrotic agents. Bioorg Med Chem Lett 24:220–223. doi:10.1016/j.bmcl.2013.11.038

    Article  CAS  PubMed  Google Scholar 

  93. Heo J, Shin H, Lee J, Kim T, Inn K-S, Kim N-J (2015) Synthesis and biological evaluation of N-cyclopropylbenzamide-benzophenone hybrids as novel and selective p38 mitogen activated protein kinase (MAPK) inhibitors. Bioorg Med Chem Lett 25:3694–3698. doi:10.1016/j.bmcl.2015.06.036

    Article  CAS  PubMed  Google Scholar 

  94. de Oliveira LR, Romeiro NC, de Lima CKF, Louback da Silva L, de Miranda ALP, Nascimento PGBD, Cunha FQ, Barreiro EJ, Lima LM (2012) Docking, synthesis and pharmacological activity of novel urea-derivatives designed as p38 MAPK inhibitors. Eur J Med Chem 54:264–271. doi:10.1016/j.ejmech.2012.05.006

    Article  CAS  Google Scholar 

  95. Khoshneviszadeh M, Ghahremani MH, Foroumadi A, Miri R, Firuzi O, Madadkar-Sobhani A, Edraki N, Parsa M, Shafiee A (2013) Design, synthesis and biological evaluation of novel anti-cytokine 1,2,4-triazine derivatives. Bioorg Med Chem 21:6708–6717. doi:10.1016/j.bmc.2013.08.009

    Article  CAS  PubMed  Google Scholar 

  96. Vinh NB, Devine SM, Munoz L, Ryan RM, Wang BH, Krum H, Chalmers DK, Simpson JS, Scammells PJ (2015) Design, synthesis, and biological evaluation of tetra-substituted thiophenes as inhibitors of p38α MAPK. ChemistryOpen 4:56–64. doi:10.1002/open.201402076

    Article  CAS  PubMed  Google Scholar 

  97. Choi H, Park HJ, Shin JC, Ko HS, Lee JK, Lee S, Park H, Hong S (2012) Structure-based virtual screening approach to the discovery of p38 MAP kinase inhibitors. Bioorg Med Chem Lett 22:2195–2199. doi:10.1016/j.bmcl.2012.01.104

    Article  CAS  PubMed  Google Scholar 

  98. Irwin JJ, Shoichet BK (2005) ZINC–a free database of commercially available compounds for virtual screening. J Chem Inf Model 45:177–182. doi:10.1021/ci049714+

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Cabrera ÁC, Gil-Redondo R, Perona A, Gago F, Morreale A (2011) VSDMIP 1.5: an automated structure- and ligand-based virtual screening platform with a PyMOL graphical user interface. J Comput Aided Mol Des 25:813–824. doi:10.1007/s10822-011-9465-6

    Article  CAS  PubMed  Google Scholar 

  100. Willemen HLDM, Campos PM, Lucas E, Morreale A, Gil-Redondo R, Agut J, González FV, Ramos P, Heijnen C, Mayor F, Kavelaars A, Murga C (2014) A novel p38 MAPK docking-groove-targeted compound is a potent inhibitor of inflammatory hyperalgesia. Biochem J 459:427–439. doi:10.1042/BJ20130172

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Poon J-F, Alao JP, Sunnerhagen P, Dinér P (2013) Azastilbenes: a cut-off to p38 MAPK inhibitors. Org Biomol Chem 11:4526–4536. doi:10.1039/c3ob27449g

    Article  CAS  PubMed  Google Scholar 

  102. Pinsetta FR, Taft CA, de Paula da Silva CHT (2014) Structure- and ligand-based drug design of novel p38-alpha MAPK inhibitors in the fight against the Alzheimer’s disease. J Biomol Struct Dyn 32:1047–1063. doi:10.1080/07391102.2013.803441

    Article  CAS  PubMed  Google Scholar 

  103. He L, Dai R, Zhang XR, Gao SY, He YY, Wang LB, Gao X, Yang LQ (2015) Ligand-based 3D pharmacophore design, virtual screening and molecular docking for novel p38 MAPK inhibitors. Med Chem Res 24:797–809. doi:10.1007/s00044-014-1158-y

    Article  CAS  Google Scholar 

  104. Chang H-W, Chung F-S, Yang C-N (2013) Molecular modeling of p38α mitogen-activated protein kinase inhibitors through 3D-QSAR and molecular dynamics simulations. J Chem Inf Model 53:1775–1786. doi:10.1021/ci4000085

    Article  CAS  PubMed  Google Scholar 

  105. Vinh NB, Simpson JS, Scammells PJ, Chalmers DK (2012) Virtual screening using a conformationally flexible target protein: models for ligand binding to p38α MAPK. J Comput Aided Mol Des 26:409–423. doi:10.1007/s10822-012-9569-7

    Article  CAS  PubMed  Google Scholar 

  106. Ebadi SA, Razzaghi-Asl N, Khoshneviszadeh M, Miri R (2015) Detailed atomistic molecular modeling of a potent type ΙΙ p38α inhibitor. Struct Chem 26:1125–1137. doi:10.1007/s11224-015-0568-x

    Article  CAS  Google Scholar 

  107. Lan P, Huang Z-J, Sun J-R, Chen W-M (2010) 3D-QSAR and molecular docking studies on fused pyrazoles as p38α mitogen-activated protein kinase inhibitors. Int J Mol Sci 11:3357–3374. doi:10.3390/ijms11093357

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Astolfi A, Iraci N, Sabatini S, Barreca ML, Cecchetti V (2015) p38α MAPK and type I inhibitors: binding site analysis and use of target ensembles in virtual screening. Molecules 20:15842–15861. doi:10.3390/molecules200915842

    Article  CAS  PubMed  Google Scholar 

  109. Medina MG, Ledesma MD, Domínguez JE, Medina M, Zafra D, Alameda F, Dotti CG, Navarro P (2005) Tissue plasminogen activator mediates amyloid-induced neurotoxicity via Erk1/2 activation. EMBO J 24:1706–1716. doi:10.1038/sj.emboj.7600650

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Sergeant N, Bretteville A, Hamdane M, Caillet-Boudin M-L, Grognet P, Bombois S, Blum D, Delacourte A, Pasquier F, Vanmechelen E, Schraen-Maschke S, Buée L (2008) Biochemistry of tau in Alzheimer’s disease and related neurological disorders. Expert Rev Proteomics 5:207–224. doi:10.1586/14789450.5.2.207

    Article  CAS  PubMed  Google Scholar 

  111. Misiura MM, Kolomeisky AB (2016) Theoretical investigation of the mechanisms of ERK2 enzymatic catalysis. J Phys Chem B 120:10508–10514. doi:10.1021/acs.jpcb.6b08435

    Article  CAS  PubMed  Google Scholar 

  112. Barr D, Oashi T, Burkhard K, Lucius S, Samadani R, Zhang J, Shapiro P, MacKerell AD, van der Vaart A (2011) Importance of domain closure for the autoactivation of ERK2. Biochemistry 50:8038–8048. doi:10.1021/bi200503a

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Blake JF, Gaudino JJ, De Meese J, Mohr P, Chicarelli M, Tian H, Garrey R, Thomas A, Siedem CS, Welch MB, Kolakowski G, Kaus R, Burkard M, Martinson M, Chen H, Dean B, Dudley DA, Gould SE, Pacheco P, Shahidi-Latham S, Wang W, West K, Yin J, Moffat J, Schwarz JB (2014) Discovery of 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidine inhibitors of Erk2. Bioorg Med Chem Lett 24:2635–2639. doi:10.1016/j.bmcl.2014.04.068

    Article  CAS  PubMed  Google Scholar 

  114. Choi W-K, El-Gamal MI, Choi HS, Baek D, Oh C-H (2011) New diarylureas and diarylamides containing 1,3,4-triarylpyrazole scaffold: synthesis, antiproliferative evaluation against melanoma cell lines, ERK kinase inhibition, and molecular docking studies. Eur J Med Chem 46:5754–5762. doi:10.1016/j.ejmech.2011.08.013

    Article  CAS  PubMed  Google Scholar 

  115. Jin F, Gao D, Wu Q, Liu F, Chen Y, Tan C, Jiang Y (2013) Exploration of N-(2-aminoethyl)piperidine-4-carboxamide as a potential scaffold for development of VEGFR-2, ERK-2 and Abl-1 multikinase inhibitor. Bioorg Med Chem 21:5694–5706. doi:10.1016/j.bmc.2013.07.026

    Article  CAS  PubMed  Google Scholar 

  116. Kinoshita T, Sugiyama H, Mori Y, Takahashi N, Tomonaga A (2016) Identification of allosteric ERK2 inhibitors through in silico biased screening and competitive binding assay. Bioorg Med Chem Lett 26:955–958. doi:10.1016/j.bmcl.2015.12.056

    Article  CAS  PubMed  Google Scholar 

  117. Larif S, Ben Salem C, Hmouda H, Bouraoui K (2014) In silico screening and study of novel ERK2 inhibitors using 3D QSAR, docking and molecular dynamics. J Mol Graph Model 53:1–12. doi:10.1016/j.jmgm.2014.07.001

    Article  CAS  PubMed  Google Scholar 

  118. Niu Y, Pan D, Yang Y, Liu H, Yao X (2016) Revealing the molecular mechanism of different residence times of ERK2 inhibitors via binding free energy calculation and unbinding pathway analysis. Chemom Intell Lab Syst 158:91–101. doi:10.1016/j.chemolab.2016.08.002

    Article  CAS  Google Scholar 

  119. Wu K-W, Chen P-C, Wang J, Sun Y-C (2012) Computation of relative binding free energy for an inhibitor and its analogs binding with Erk kinase using thermodynamic integration MD simulation. J Comput Aided Mol Des 26:1159–1169. doi:10.1007/s10822-012-9606-6

    Article  CAS  PubMed  Google Scholar 

  120. Rastelli G, Rosenfeld R, Reid R, Santi DV (2008) Molecular modeling and crystal structure of ERK2-hypothemycin complexes. J Struct Biol 164:18–23. doi:10.1016/j.jsb.2008.05.002

    Article  CAS  PubMed  Google Scholar 

  121. Liu B, Fu L, Zhang C, Zhang L, Zhang Y, Ouyang L, He G, Huang J (2015) Computational design, chemical synthesis, and biological evaluation of a novel ERK inhibitor (BL-EI001) with apoptosis-inducing mechanisms in breast cancer. Oncotarget 6:6762–6775. doi:10.18632/oncotarget.3105

    Article  PubMed  PubMed Central  Google Scholar 

  122. Mehan S, Meena H, Sharma D, Sankhla R (2011) JNK: a stress-activated protein kinase therapeutic strategies and involvement in Alzheimer’s and various neurodegenerative abnormalities. J Mol Neurosci 43:376–390. doi:10.1007/s12031-010-9454-6

    Article  CAS  PubMed  Google Scholar 

  123. Dias-Santagata D, Fulga TA, Duttaroy A, Feany MB (2007) Oxidative stress mediates tau-induced neurodegeneration in Drosophila. J Clin Invest 117:236–245. doi:10.1172/JCI28769

    Article  CAS  PubMed  Google Scholar 

  124. Colombo A, Repici M, Pesaresi M, Santambrogio S, Forloni G, Borsello T (2007) The TAT-JNK inhibitor peptide interferes with beta amyloid protein stability. Cell Death Differ 14:1845–1848. doi:10.1038/sj.cdd.4402202

    Article  CAS  PubMed  Google Scholar 

  125. Ferrer I, Barrachina M, Puig B (2002) Anti-tau phospho-specific Ser262 antibody recognizes a variety of abnormal hyper-phosphorylated tau deposits in tauopathies including Pick bodies and argyrophilic grains. Acta Neuropathol 104:658–664. doi:10.1007/s00401-002-0600-2

    CAS  PubMed  Google Scholar 

  126. Christopher JA, Atkinson FL, Bax BD, Brown MJB, Champigny AC, Chuang TT, Jones EJ, Mosley JE, Musgrave JR (2009) 1-Aryl-3,4-dihydroisoquinoline inhibitors of JNK3. Bioorg Med Chem Lett 19:2230–2234. doi:10.1016/j.bmcl.2009.02.098

    Article  CAS  PubMed  Google Scholar 

  127. Dykstra DW, Dalby KN, Ren P (2013) Elucidating binding modes of zuonin A enantiomers to JNK1 via in silico methods. J Mol Graph Model 45:38–44. doi:10.1016/j.jmgm.2013.08.008

    Article  CAS  PubMed  Google Scholar 

  128. Ngoei KRW, Ng DCH, Gooley PR, Fairlie DP, Stoermer MJ, Bogoyevitch MA (2013) Identification and characterization of bi-thiazole-2,2′-diamines as kinase inhibitory scaffolds. Biochim Biophys Acta 1834:1077–1088. doi:10.1016/j.bbapap.2013.02.001

    Article  CAS  PubMed  Google Scholar 

  129. Zhuo Z-H, Sun Y-Z, Jin P-N, Li F-Y, Zhang Y-L, Wang H-L (2016) Selective targeting of MAPK family kinases JNK over p38 by rationally designed peptides as potential therapeutics for neurological disorders and epilepsy. Mol BioSyst 12:2532–2540. doi:10.1039/C6MB00297H

    Article  CAS  PubMed  Google Scholar 

  130. Katari SK, Natarajan P, Swargam S, Kanipakam H, Pasala C, Umamaheswari A (2016) Inhibitor design against JNK1 through e-pharmacophore modeling docking and molecular dynamics simulations. J Recept Signal Transduct Res 36:558–571. doi:10.3109/10799893.2016.1141955

    Article  CAS  PubMed  Google Scholar 

  131. Shaikh AR, Ismael M, Del Carpio CA, Tsuboi H, Koyama M, Endou A, Kubo M, Broclawik E, Miyamoto A (2006) Three-dimensional quantitative structure-activity relationship (3 D-QSAR) and docking studies on (benzothiazole-2-yl) acetonitrile derivatives as c-Jun N-terminal kinase-3 (JNK3) inhibitors. Bioorg Med Chem Lett 16:5917–5925. doi:10.1016/j.bmcl.2006.06.039

    Article  CAS  PubMed  Google Scholar 

  132. Kim M-H, Ryu J-S, Hah J-M (2013) 3D-QSAR studies of 1,2-diaryl-1H-benzimidazole derivatives as JNK3 inhibitors with protective effects in neuronal cells. Bioorg Med Chem Lett 23:1639–1642. doi:10.1016/j.bmcl.2013.01.082

    Article  CAS  PubMed  Google Scholar 

  133. Wu X-X, Dai D-S, Zhu X, Li X-F, Yuan J, Wu X-F, Miao M-S, Zeng H-H, Zhao C-L (2014) Molecular modeling studies of JNK3 inhibitors using QSAR and docking. Med Chem Res 23:2456–2475. doi:10.1007/s00044-013-0782-2

    Article  CAS  Google Scholar 

  134. Madhavan T, Chung JY, Kothandan G, Gadhe CG, Cho SJ (2012) 3D-QSAR studies of JNK1 inhibitors utilizing various alignment methods. Chem Biol Drug Des 79:53–67. doi:10.1111/j.1747-0285.2011.01168.x

    Article  CAS  PubMed  Google Scholar 

  135. Hierold J, Baek S, Rieger R, Lim T-G, Zakpur S, Arciniega M, Lee KW, Huber R, Tietze LF (2015) Design, synthesis, and biological evaluation of quercetagetin analogues as JNK1 inhibitors. Chemistry 21:16887–16894. doi:10.1002/chem.201502475

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Julio Caballero .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media LLC

About this protocol

Cite this protocol

Navarro-Retamal, C., Caballero, J. (2018). Molecular Modeling of Tau Proline-Directed Protein Kinase (PDPK) Inhibitors. In: Roy, K. (eds) Computational Modeling of Drugs Against Alzheimer’s Disease. Neuromethods, vol 132. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-7404-7_13

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7404-7_13

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-7403-0

  • Online ISBN: 978-1-4939-7404-7

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics