Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

General Research Article

Integrated System Pharmacology and In Silico Analysis Elucidating Neuropharmacological Actions of Withania somnifera in the Treatment of Alzheimer’s Disease

Author(s): Md. Abdul Hannan, Raju Dash, Md. Nazmul Haque, Sung Min Choi and Il Soo Moon*

Volume 19, Issue 7, 2020

Page: [541 - 556] Pages: 16

DOI: 10.2174/1871527319999200730214807

Price: $65

Abstract

Background: Withania somnifera (WS), also referred to as Medhya Rasayana (nootropic or rejuvenating), has traditionally been prescribed for various neurological ailments, including dementia. Despite substantial evidence, pharmacological roles of WS, neither as nootropic nor as an antidementia agent, are well-understood at the cellular and molecular levels.

Objectives: We aimed at elucidating the pharmacological action mechanisms of WS root constituents against Alzheimer’s Disease (AD) pathology.

Methods: Various bioinformatics tools and resources, including DAVID, Cytoscape, NetworkAnalyst and KEGG pathway database were employed to analyze the interaction of WS root bioactive molecules with the protein targets of AD-associated cellular processes. We also used a molecular simulation approach to validate the interaction of compounds with selected protein targets.

Results: Network analysis revealed that β-sitosterol, withaferin A, stigmasterol, withanolide A, and withanolide D are the major constituents of WS root that primarily target the cellular pathways such as PI3K/Akt signaling, neurotrophin signaling and toll-like receptor signaling and proteins such as Tropomyosin receptor Kinase B (TrkB), Glycogen Synthase Kinase-3β (GSK-3β), Toll-Like Receptor 2/4 (TLR2/4), and β-secretase (BACE-1). Also, the in silico analysis further validated the interaction patterns and binding affinity of the major WS compounds, particularly stigmasterol, withanolide A, withanolide D and β-sitosterol with TrkB, GSK-3β, TLR2/4, and BACE-1.

Conclusion: The present findings demonstrate that stigmasterol, withanolide A, withanolide D and β-sitosterol are the major metabolites that are responsible for the neuropharmacological action of WS root against AD-associated pathobiology, and TrkB, GSK-3β, TLR2/4, and BACE-1 could be the potential druggable targets.

Keywords: Network pharmacology, molecular simulation, Alzheimer’s disease, Withania somnifera, withanolides, stigmasterol.

« Previous
Graphical Abstract
[1]
Anastasio TJ. Editorial: computational and experimental approaches in multi-target pharmacology. Front Pharmacol 2017; 8(443): 443.
[http://dx.doi.org/10.3389/fphar.2017.00443] [PMID: 28713280]
[2]
Archana R, Namasivayam A. Antistressor effect of Withania somnifera. J Ethnopharmacol 1999; 64(1): 91-3.
[http://dx.doi.org/10.1016/S0378-8741(98)00107-X] [PMID: 10075127]
[3]
Bhattacharya A, Ghosal S, Bhattacharya SK. Anti-oxidant effect of Withania somnifera glycowithanolides in chronic footshock stress-induced perturbations of oxidative free radical scavenging enzymes and lipid peroxidation in rat frontal cortex and striatum. J Ethnopharmacol 2001; 74(1): 1-6.
[http://dx.doi.org/10.1016/S0378-8741(00)00309-3] [PMID: 11137343]
[4]
Ziauddin M, Phansalkar N, Patki P, Diwanay S, Patwardhan B. Studies on the immunomodulatory effects of Ashwagandha. J Ethnopharmacol 1996; 50(2): 69-76.
[http://dx.doi.org/10.1016/0378-8741(95)01318-0] [PMID: 8866726]
[5]
Sehgal N, Gupta A, Valli RK, et al. Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci USA 2012; 109(9): 3510-5.
[http://dx.doi.org/10.1073/pnas.1112209109] [PMID: 22308347]
[6]
Gupta M, Kaur G. Withania somnifera (L.) Dunal ameliorates neurodegeneration and cognitive impairments associated with systemic inflammation. BMC Complement Altern Med 2019; 19(1): 217.
[http://dx.doi.org/10.1186/s12906-019-2635-0] [PMID: 31416451]
[7]
Alzoubi KH, Al Hilo AS, Al-Balas QA, El-Salem K, El-Elimat T, Alali FQ. Withania somnifera root powder protects against post-traumatic stress disorder-induced memory impairment. Mol Biol Rep 2019; 46(5): 4709-15.
[http://dx.doi.org/10.1007/s11033-019-04915-3] [PMID: 31218539]
[8]
Kuboyama T, Tohda C, Komatsu K. Neuritic regeneration and synaptic reconstruction induced by withanolide A. Br J Pharmacol 2005; 144(7): 961-71.
[http://dx.doi.org/10.1038/sj.bjp.0706122] [PMID: 15711595]
[9]
Kuboyama T, Tohda C, Zhao J, Nakamura N, Hattori M, Komatsu K. Axon- or dendrite-predominant outgrowth induced by constituents from Ashwagandha. Neuroreport 2002; 13(14): 1715-20.
[http://dx.doi.org/10.1097/00001756-200210070-00005] [PMID: 12395110]
[10]
Tohda C, Kuboyama T, Komatsu K. Dendrite extension by methanol extract of Ashwagandha (roots of Withania somnifera) in SK-N-SH cells. Neuroreport 2000; 11(9): 1981-5.
[http://dx.doi.org/10.1097/00001756-200006260-00035] [PMID: 10884056]
[11]
Zhao J, Nakamura N, Hattori M, Kuboyama T, Tohda C, Komatsu K. Withanolide derivatives from the roots of Withania somnifera and their neurite outgrowth activities. Chem Pharm Bull (Tokyo) 2002; 50(6): 760-5.
[http://dx.doi.org/10.1248/cpb.50.760] [PMID: 12045329]
[12]
Kuboyama T, Tohda C, Komatsu K. Withanoside IV and its active metabolite, sominone, attenuate Abeta(25-35)-induced neurodegeneration. Eur J Neurosci 2006; 23(6): 1417-26.
[http://dx.doi.org/10.1111/j.1460-9568.2006.04664.x] [PMID: 16553605]
[13]
Joyashiki E, Matsuya Y, Tohda C. Sominone improves memory impairments and increases axonal density in Alzheimer’s disease model mice, 5XFAD. Int J Neurosci 2011; 121(4): 181-90.
[http://dx.doi.org/10.3109/00207454.2010.541571] [PMID: 21329473]
[14]
Pandey A, Bani S, Dutt P, Kumar Satti N, Avtar Suri K, Nabi Qazi G. Multifunctional neuroprotective effect of Withanone, a compound from Withania somnifera roots in alleviating cognitive dysfunction. Cytokine 2018; 102: 211-21.
[http://dx.doi.org/10.1016/j.cyto.2017.10.019] [PMID: 29108796]
[15]
Choudhary D, Bhattacharyya S, Bose S. Efficacy and safety of ashwagandha (Withania somnifera (l.) dunal) root extract in improving memory and cognitive functions. J Diet Suppl 2017; 14(6): 599-612.
[http://dx.doi.org/10.1080/19390211.2017.1284970] [PMID: 28471731]
[16]
Mandloi S, Chakrabarti S. PALM-IST: Pathway assembly from literature mining--an information search tool. Sci Rep 2015; 5: 10021.
[http://dx.doi.org/10.1038/srep10021] [PMID: 25989388]
[17]
Safran M, Dalah I, Alexander J, et al. GeneCards Version 3: the human gene integrator. Database 2010; 2010baq020
[18]
Wu CH, Apweiler R, Bairoch A, et al. The Universal Protein Resource (UniProt): an expanding universe of protein information. Nucleic Acids Res 2006; 34(Database issue): D187-91.
[http://dx.doi.org/10.1093/nar/gkj161] [PMID: 16381842]
[19]
Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003; 13(11): 2498-504.
[http://dx.doi.org/10.1101/gr.1239303] [PMID: 14597658]
[20]
Piñero J, Bravo À, Queralt-Rosinach N, et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic Acids Res 2017; 45(D1): D833-9.
[http://dx.doi.org/10.1093/nar/gkw943] [PMID: 27924018]
[21]
Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009; 4(1): 44-57.
[http://dx.doi.org/10.1038/nprot.2008.211] [PMID: 19131956]
[22]
Xia J, Gill EE, Hancock REW. NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat Protoc 2015; 10(6): 823-44.
[http://dx.doi.org/10.1038/nprot.2015.052] [PMID: 25950236]
[23]
Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000; 28(1): 27-30.
[http://dx.doi.org/10.1093/nar/28.1.27] [PMID: 10592173]
[24]
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(1-3): 3-26.
[http://dx.doi.org/10.1016/S0169-409X(00)00129-0] [PMID: 11259830]
[25]
Jorgensen WL, Duffy EM. Prediction of drug solubility from structure. Adv Drug Deliv Rev 2002; 54(3): 355-66.
[http://dx.doi.org/10.1016/S0169-409X(02)00008-X] [PMID: 11922952]
[26]
Liu J-F, Hu A-N, Zan J-F, Wang P, You Q-Y, Tan A-H. Network pharmacology deciphering mechanisms of volatiles of wendan granule for the treatment of Alzheimer's disease. eCAM 2019; 20197826769
[http://dx.doi.org/10.1155/2019/7826769]
[27]
Wiesmann C, Ultsch MH, Bass SH, de Vos AM. Crystal structure of nerve growth factor in complex with the ligand-binding domain of the TrkA receptor. Nature 1999; 401(6749): 184-8.
[http://dx.doi.org/10.1038/43705] [PMID: 10490030]
[28]
Chao MV, Rajagopal R, Lee FS. Neurotrophin signalling in health and disease. Clin Sci (Lond) 2006; 110(2): 167-73.
[http://dx.doi.org/10.1042/CS20050163] [PMID: 16411893]
[29]
Pattarawarapan M, Burgess K. Molecular basis of neurotrophin-receptor interactions. J Med Chem 2003; 46(25): 5277-91.
[http://dx.doi.org/10.1021/jm030221q] [PMID: 14640536]
[30]
Ibáñez CF. Emerging themes in structural biology of neurotrophic factors. Trends Neurosci 1998; 21(10): 438-44.
[http://dx.doi.org/10.1016/S0166-2236(98)01266-1] [PMID: 9786342]
[31]
Shoemark DK, Williams C, Fahey MS, et al. Design and Nuclear Magnetic Resonance (NMR) structure determination of the second extracellular immunoglobulin tyrosine kinase A (TrkAIg2) domain construct for binding site elucidation in drug discovery. J Med Chem 2015; 58(2): 767-77.
[http://dx.doi.org/10.1021/jm501307e] [PMID: 25454499]
[32]
Naylor RL, Robertson AGS, Allen SJ, et al. A discrete domain of the human TrkB receptor defines the binding sites for BDNF and NT-4. Biochem Biophys Res Commun 2002; 291(3): 501-7.
[http://dx.doi.org/10.1006/bbrc.2002.6468] [PMID: 11855816]
[33]
Blatt JM, Weisskopf VF. Theoretical nuclear physics. Courier Corporation 1991.
[34]
Arifuzzaman M, Mitra S, Das R, Hamza A, Absar N, Dash R. In silico analysis of nonsynonymous single‐nucleotide polymorphisms (nsSNPs) of the SMPX gene. Ann Hum Genet 2020; 84: 54-71.
[PMID: 31583691]
[35]
Dash R, Arifuzzaman M, Mitra S, Abdul Hannan M, Absar N, Hosen SMZ. Unveiling the structural insights into the selective inhibition of protein kinase D1. Curr Pharm Des 2019; 25(10): 1059-74.
[http://dx.doi.org/10.2174/1381612825666190527095510] [PMID: 31131745]
[36]
Dash R, Junaid M, Mitra S, Arifuzzaman M, Hosen SMZ. Structure-based identification of potent VEGFR-2 inhibitors from in vivo metabolites of a herbal ingredient. J Mol Model 2019; 25(4): 98.
[http://dx.doi.org/10.1007/s00894-019-3979-6] [PMID: 30904971]
[37]
Hosen SMZ, Dash R, Junaid M, Mitra S, Absar N. Identification and structural characterization of deleterious non-synonymous single nucleotide polymorphisms in the human SKP2 gene. Comput Biol Chem 2019; 79: 127-36.
[http://dx.doi.org/10.1016/j.compbiolchem.2019.02.003] [PMID: 30802828]
[38]
Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-Kinase/Akt cell survival pathway. J Biol Chem 1998; 273(32): 19929-32.
[http://dx.doi.org/10.1074/jbc.273.32.19929] [PMID: 9685326]
[39]
Cao ZQ, Wang XX, Lu L, et al. β-Sitosterol and gemcitabine exhibit synergistic anti-pancreatic cancer activity by modulating apoptosis and inhibiting epithelial-mesenchymal transition by deactivating Akt/GSK-3β Signaling. Front Pharmacol 2019; 9(1525): 1525.
[http://dx.doi.org/10.3389/fphar.2018.01525] [PMID: 30670971]
[40]
Haque MN, Moon IS. Stigmasterol upregulates immediate early genes and promotes neuronal cytoarchitecture in primary hippocampal neurons as revealed by transcriptome analysis. Phytomedicine 2018; 46: 164-75.
[http://dx.doi.org/10.1016/j.phymed.2018.04.012]
[41]
Song Z, Yin F, Xiang B, Lan B, Cheng S. Systems pharmacological approach to investigate the mechanism of Acori Tatarinowii Rhizoma for Alzheimer’s Disease. Evid Based Complement Alternat Med 2018; 20185194016
[http://dx.doi.org/10.1155/2018/5194016] [PMID: 30050590]
[42]
Kamkwalala AR, Newhouse PA. Beyond acetylcholinesterase inhibitors: novel cholinergic treatments for Alzheimer’s disease. Curr Alzheimer Res 2017; 14(4): 377-92.
[PMID: 27697062]
[43]
Grover A, Shandilya A, Agrawal V, Bisaria VS, Sundar D. Computational evidence to inhibition of human acetyl cholinesterase by withanolide a for Alzheimer treatment. J Biomol Struct Dyn 2012; 29(4): 651-62.
[http://dx.doi.org/10.1080/07391102.2012.10507408] [PMID: 22208270]
[44]
Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol 2014; 5: 461.
[http://dx.doi.org/10.3389/fimmu.2014.00461] [PMID: 25309543]
[45]
Brüll F, Mensink RP, van den Hurk K, Duijvestijn A, Plat J. TLR2 activation is essential to induce a Th1 shift in human peripheral blood mononuclear cells by plant stanols and plant sterols. J Biol Chem 2010; 285(5): 2951-8.
[http://dx.doi.org/10.1074/jbc.M109.036343] [PMID: 19948716]
[46]
Heyninck K, Lahtela-Kakkonen M, Van der Veken P, Haegeman G, Vanden Berghe W. Withaferin A inhibits NF-kappaB activation by targeting cysteine 179 in IKKβ. Biochem Pharmacol 2014; 91(4): 501-9.
[http://dx.doi.org/10.1016/j.bcp.2014.08.004] [PMID: 25159986]
[47]
Kaileh M, Vanden Berghe W, Heyerick A, et al. Withaferin a strongly elicits IkappaB kinase beta hyperphosphorylation concomitant with potent inhibition of its kinase activity. J Biol Chem 2007; 282(7): 4253-64.
[http://dx.doi.org/10.1074/jbc.M606728200] [PMID: 17150968]
[48]
Oh JH, Kwon TK. Withaferin A inhibits tumor necrosis factor-α-induced expression of cell adhesion molecules by inactivation of Akt and NF-kappaB in human pulmonary epithelial cells. Int Immunopharmacol 2009; 9(5): 614-9.
[http://dx.doi.org/10.1016/j.intimp.2009.02.002] [PMID: 19236958]
[49]
Pawar P, Gilda S, Sharma S, et al. Rectal gel application of Withania somnifera root extract expounds anti-inflammatory and muco-restorative activity in TNBS-induced inflammatory bowel disease. BMC Complement Altern Med 2011; 11(1): 34.
[http://dx.doi.org/10.1186/1472-6882-11-34] [PMID: 21527003]
[50]
Cole SL, Vassar R. The Alzheimer’s disease β-secretase enzyme, BACE1. Mol Neurodegener 2007; 2(1): 22.
[http://dx.doi.org/10.1186/1750-1326-2-22] [PMID: 18005427]
[51]
Burg VK, Grimm HS, Rothhaar TL, et al. Plant sterols the better cholesterol in Alzheimer’s disease? A mechanistical study. J Neurosci 2013; 33(41): 16072-87.
[http://dx.doi.org/10.1523/JNEUROSCI.1506-13.2013] [PMID: 24107941]
[52]
Vanmierlo T, Weingärtner O, van der Pol S, et al. Dietary intake of plant sterols stably increases plant sterol levels in the murine brain. J Lipid Res 2012; 53(4): 726-35.
[http://dx.doi.org/10.1194/jlr.M017244] [PMID: 22279184]
[53]
Martins IJ, Berger T, Sharman MJ, Verdile G, Fuller SJ, Martins RN. Cholesterol metabolism and transport in the pathogenesis of Alzheimer’s disease 2009; 1111(6): 1275-308.
[54]
St-Onge M-P, Jones PJH. Phytosterols and human lipid metabolism: efficacy, safety, and novel foods. Lipids 2003; 38(4): 367-75.
[http://dx.doi.org/10.1007/s11745-003-1071-3] [PMID: 12848281]
[55]
Campanella C, Pace A, Caruso Bavisotto C, et al. Heat shock proteins in Alzheimer’s disease: role and targeting. Int J Mol Sci 2018; 19(9): 2603.
[http://dx.doi.org/10.3390/ijms19092603] [PMID: 30200516]
[56]
Abuznait AH, Kaddoumi A. Role of ABC transporters in the pathogenesis of Alzheimer’s disease. ACS Chem Neurosci 2012; 3(11): 820-31.
[http://dx.doi.org/10.1021/cn300077c] [PMID: 23181169]
[57]
Wolf A, Bauer B, Hartz AM. ABC transporters and the Alzheimer’s disease enigma. Front Psychiatry 2012; 3(54): 54.
[http://dx.doi.org/10.3389/fpsyt.2012.00054] [PMID: 22675311]
[58]
Siegel GJ, Chauhan NB. Neurotrophic factors in Alzheimer’s and Parkinson’s disease brain. Brain Res Brain Res Rev 2000; 33(2-3): 199-227.
[http://dx.doi.org/10.1016/S0165-0173(00)00030-8] [PMID: 11011066]
[59]
Zerbinatti CV, Wahrle SE, Kim H, et al. Apolipoprotein E and low density lipoprotein receptor-related protein facilitate intraneuronal Abeta42 accumulation in amyloid model mice. J Biol Chem 2006; 281(47): 36180-6.
[http://dx.doi.org/10.1074/jbc.M604436200] [PMID: 17012232]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy