DISCOVERY OF NOVEL AND SELECTIVE C-JUN NH2-TERMINAL KINASES 2 INHIBITORS BY TWO-DIMENSIONAL QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIP MODEL DEVELOPMENT, MOLECULAR DOCKING AND ABSORPTION, DISTRIBUTION, METABOLISM, ELIMINATION PREDICTION STUDIES: AN IN SILICO APPROACH

Authors

  • Ashima Nagpal Department of Pharmacy, Banasthali Vidyapith, Banasthali – 304 022, Rajasthan, India.
  • Sarvesh Paliwal Department of Pharmacy, Banasthali Vidyapith, Banasthali – 304 022, Rajasthan, India.

DOI:

https://doi.org/10.22159/ajpcr.2018.v11i5.24157

Keywords:

QSAR, Multiple linear regression, Partial least square, Tanimoto index, Absorption, distribution, metabolism, and elimination

Abstract

Objective: Incited by the dearth of selective c-Jun NH2-terminal kinases (JNK) inhibitors, efforts have been made to design novel JNK2 inhibitors with good selectivity profile by utilizing a set of in silico tools.

Methods: The present study involved 2D QSAR model development through multiple linear regression and partial least square methods. Further, the information unveiled through the above model was meticulously utilized to design novel JNK2 inhibitors (compound a and compound b). The selectivity of the novel molecules was ascertained through the molecular docking experiments. Determination of Tanimoto similarity index and absorption, distribution, metabolism, and elimination (ADME) properties was performed to ascertain the novelty and drug-like properties of the designed molecules, respectively.

Results: Four explanatory variables or descriptors, moment of inertia 2 length (subst.1), moment of inertia 3 length (subst.3), Kier Chi4 (path/ cluster) index (whole molecule), and vamp highest occupied molecular orbital (whole molecule), were found to possess profound influence on the biological activity with the values of standard parameters, s: 0.28, r: 0.96, r2: 0.92, r2CV: 0.82, and f=59.69. Tanimoto index values were found to be 0.095 and 0.075 for compound a and compound b, respectively.

Conclusion: The values of the statistical parameters proved the developed model to be of excellent quality. The results obtained for ADME prediction studies proved both the compounds to be better than the most potent compound (compound 45) of the set of JNK2 inhibitors selected for model development. In addition, extremely low Tanimoto similarity index values for both the compounds provided sufficient evidence for the novelty of the designed molecules.

Objective: Incited by the dearth of selective c-Jun NH2-terminal kinases (JNK) inhibitors, efforts have been made to design novel JNK2 inhibitors with good selectivity profile by utilizing a set of in silico tools.

Methods: The present study involved 2D QSAR model development through multiple linear regression and partial least square methods. Further, the information unveiled through the above model was meticulously utilized to design novel JNK2 inhibitors (compound a and compound b). The selectivity of the novel molecules was ascertained through the molecular docking experiments. Determination of Tanimoto similarity index and absorption, distribution, metabolism, and elimination (ADME) properties was performed to ascertain the novelty and drug-like properties of the designed molecules, respectively.

Results: Four explanatory variables or descriptors, moment of inertia 2 length (subst.1), moment of inertia 3 length (subst.3), Kier Chi4 (path/ cluster) index (whole molecule), and vamp highest occupied molecular orbital (whole molecule), were found to possess profound influence on the biological activity with the values of standard parameters, s: 0.28, r: 0.96, r2: 0.92, r2CV: 0.82, and f=59.69. Tanimoto index values were found to be 0.095 and 0.075 for compound a and compound b, respectively.

Conclusion: The values of the statistical parameters proved the developed model to be of excellent quality. The results obtained for ADME prediction studies proved both the compounds to be better than the most potent compound (compound 45) of the set of JNK2 inhibitors selected for model development. In addition, extremely low Tanimoto similarity index values for both the compounds provided sufficient evidence for the novelty of the designed molecules.

Downloads

Download data is not yet available.

Author Biography

Ashima Nagpal, Department of Pharmacy, Banasthali Vidyapith, Banasthali – 304 022, Rajasthan, India.

Department of Pharmacy, Lecturer

References

Yang DD, Kuan CY, Whitmarsh AJ, Rincón M, Zheng TS, Davis RJ, et al. Absence of excitotoxicity-induced apoptosis in the hippocampus of mice lacking the jnk3 gene. Nature 1997;389:865-70.

Hunot S, Vila M, Teismann P, Davis RJ, Hirsch EC, Przedborski S, et al. JNK-mediated induction of cyclooxygenase 2 is required for neurodegeneration in a mouse model of Parkinson’s disease. Proc Natl Acad Sci U S A 2004;101:665-70.

Chang L, Jones Y, Ellisman MH, Goldstein LS, Karin M. JNK1 is required for maintenance of neuronal microtubules and controls phosphorylation of microtubule-associated proteins. Dev Cell 2003;4:521-33.

Bjorkblom B, Ostman N, Hongisto V, Komarovski V, Filen JJ, Nyman TA, et al. Constitutively active cytoplasmic c-Jun N-terminal kinase 1 is a dominant regulator of dendritic architecture: Role of microtubuleassociated protein 2 as an effector. J Neurosci 2005;25:6350-61.

Tararuk T, Ostman N, Li W, Björkblom B, Padzik A, Zdrojewska J, et al. JNK1 phosphorylation of SCG10 determines microtubule dynamics and axodendritic length. J Cell Biol 2006;173:265-77.

Morishima Y, Gotoh Y, Zieg J, Barrett T, Takano H, Flavell R, et al. Beta-amyloid induces neuronal apoptosis via a mechanism that involves the c-Jun N-terminal kinase pathway and the induction of Fas ligand. J Neurosci 2001;21:7551-60.

Fogarty MP, Downer EJ, Campbell V. A role for c-Jun N-terminal kinase 1 (JNK1), but not JNK2, in the beta-amyloid-mediated stabilization of protein p53 and induction of the apoptotic cascade in cultured cortical neurons. Biochem J 2003;371:789-98.

Kimberly WT, Zheng JB, Town T, Flavell RA, Selkoe DJ. Physiological regulation of the beta-amyloid precursor protein signalling domain by c-Jun N-terminal kinase JNK3 during neuronal differentiation. J Neurosci 2005;25:5533-43.

Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action and Type 2 diabetes. Science 2004;306:457-61.

Jaeschke A, Rincón M, Doran B, Reilly J, Neuberg D, Greiner DL, et al. Disruption of the jnk2 (Mapk9) gene reduces destructive insulitis and diabetes in a mouse model of type I diabetes. Proc Natl Acad Sci U S A 2005;102:6931-5.

Li Y, Schwabe RF, DeVries-Seimon T, Yao PM, Gerbod-Giannone MC, Tall AR, et al. Free cholesterol-loaded macrophages are an abundant source of tumor necrosis factor-alpha and interleukin-6: Model of NF-kappaB- and map kinase-dependent inflammation in advanced atherosclerosis. J Biol Chem 2005;280:21763-72.

Mohit AA, Martin JH, Miller CA. P493F12 kinase: A novel MAP kinase expressed in a subset of neurons in the human nervous system. Neuron 1995;14:67-78.

Dérijard B, Hibi M, Wu IH, Barrett T, Su B, Deng T, et al. JNK1: A protein kinase stimulated by UV light and ha-ras that binds and phosphorylates the c-jun activation domain. Cell 1994;76:1025-37.

Kallunki T, Su B, Tsigelny I, Sluss HK, Dérijard B, Moore G, et al. JNK2 contains a specificity-determining region responsible for efficient c-Jun binding and phosphorylation. Genes Dev 1994;8:2996-3007.

Gupta S, Barrett T, Whitmarsh AJ, Cavanagh J, Sluss HK, Dérijard B, et al. Selective interaction of JNK protein kinase isoforms with transcription factors. EMBO J 1996;15:2760-70.

Karin M, Gallagher E. From JNK to pay dirt: Jun kinases, their biochemistry, physiology and clinical importance. IUBMB Life 2005;57:283-95.

Karin M, Lawrence T, Nizet V. Innate immunity gone awry: Linking microbial infections to chronic inflammation and cancer. Cell 2006;124:823-35.

Han Z, Boyle DL, Chang L, Bennett B, Karin M, Yang L, et al. C-jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis. J Clin Invest 2001;108:73-81.

Sumara G, Belwal M, Ricci R. Jnking†atherosclerosis. Cell Mol Life Sci 2005;62:2487-94.

Ricci R, Sumara G, Sumara I, Rozenberg I, Kurrer M, Akhmedov A, et al. Requirement of JNK2 for scavenger receptor A-mediated foam cell formation in atherogenesis. Science 2004;306:1558-61.

Bowers S, Truong AP, Neitz RJ, Neitzel M, Probst GD, Hom RK, et al. Design and synthesis of a novel. Orally active, brain penetrant, tri-substituted thiophene based JNK inhibitor. Bioorg Med Chem Lett 2011;21:1838-43.

Zhivkova Z, Doytchinova I. In silico quantitative structure-pharmacokinetic relationship modeling on acidic drugs: Half Life. Int J Pharm Pharm Sci 2014;6:283-9.

Cronin MT, Schultz TW. Development of quantitative structure-activity relationships for the toxicity of aromatic compounds of Tetrahymena pyriformis: Comparative assessment of the methodologies. Chem Res Toxicol 2001;14:1284-95.

Dessalew N. Investigation of the structural requirement for inhibiting HIV integrase: QSAR study. Acta Pharm 2009;59:31-43.

Kovatcheva A, Buchbauer G, Golbraikh A, Wolschann P. QSAR modelling of r-campholenic derivatives with sandalwood odor. J Chem Inf Comput Sci 2003;43:259-66.

Kubinyi H. QSAR and 3D-QSAR in Drug Design Part 1: Methodology. Drug Discov Today 1997;2:457-67.

Jagdale D, Ramaa CS. Quantitative structure-activity relationship analysis of novel pyrazoline derivatives using k nearest neighbour molecular field analysis method. Int J Pharm Pharm Sci 2017;9:87-91.

Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001;46:3-26.

Daina A, Michielin O, Zoete V. Swiss ADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017;7:42717.

Egan WJ, Kenneth MM, Baldwin JJ. Prediction of drug absorption using multivariate statistics. J Med Chem 2000;43:3867-77.

Veber DF, Stephen R, Cheng JH, Smith BR, Ward KW, Kopple KW. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem 2002;45:2615-623.

Published

01-05-2018

How to Cite

Nagpal, A., and S. Paliwal. “DISCOVERY OF NOVEL AND SELECTIVE C-JUN NH2-TERMINAL KINASES 2 INHIBITORS BY TWO-DIMENSIONAL QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIP MODEL DEVELOPMENT, MOLECULAR DOCKING AND ABSORPTION, DISTRIBUTION, METABOLISM, ELIMINATION PREDICTION STUDIES: AN IN SILICO APPROACH”. Asian Journal of Pharmaceutical and Clinical Research, vol. 11, no. 5, May 2018, pp. 100-8, doi:10.22159/ajpcr.2018.v11i5.24157.

Issue

Section

Original Article(s)