Generic placeholder image

Current Drug Discovery Technologies

Editor-in-Chief

ISSN (Print): 1570-1638
ISSN (Online): 1875-6220

Research Article

Systems Pharmacology and Pharmacokinetics Strategy to Decode Bioactive Ingredients and Molecular Mechanisms from Zingiber officinale as Phyto-therapeutics against Neurological Diseases

Author(s): Pavan Gollapalli*, Aditya S.J. Rao, Hanumanthappa Manjunatha, Gnanasekaran Tamizh Selvan*, Praveenkumar Shetty and Nalilu Suchetha Kumari

Volume 20, Issue 1, 2023

Published on: 20 October, 2022

Article ID: e250822207996 Pages: 12

DOI: 10.2174/1570163819666220825141356

Price: $65

Abstract

Background: The bioactive constituents from Zingiber officinale (Z. officinale) have shown a positive effect on neurodegenerative diseases like Alzheimer's disease (AD), which manifests as progressive memory loss and cognitive impairment.

Objective: This study investigates the binding ability and the pharmaco-therapeutic potential of Z. officinale with AD disease targets by molecular docking and molecular dynamic (MD) simulation approaches.

Methods: By coupling enormous available phytochemical data and advanced computational technologies, the possible molecular mechanism of action of these bioactive compounds was deciphered by evaluating phytochemicals, target fishing, and network biological analysis.

Results: As a result, 175 bioactive compounds and 264 human target proteins were identified. The gene ontology and Kyoto Encyclopaedia of Genes and Genomes pathway enrichment analysis and molecular docking were used to predict the basis of vital bioactive compounds and biomolecular mechanisms involved in the treatment of AD. Amongst selected bioactive compounds, 10- Gingerdione and 1-dehydro-[8]-gingerdione exhibited significant anti-neurological properties against AD targeting amyloid precursor protein with docking energy of -6.0 and -5.6, respectively.

Conclusion: This study suggests that 10-Gingerdione and 1-dehydro-[8]-gingerdione strongly modulates the anti-neurological activity and are associated with pathological features like amyloid-β plaques and hyperphosphorylated tau protein are found to be critically regulated by these two target proteins. This comprehensive analysis provides a clue for further investigation of these natural compounds' inhibitory activity in drug discovery for AD treatment.

Keywords: Zingiber officinale, neurological diseases, Alzheimer’s disease, network pharmacology, amyloid precursor protein, natural.

Graphical Abstract
[1]
Brahmachari G. Chemistry and pharmacology of naturally occurring bioactive compounds. CRC Press Boca Raton 2013.
[http://dx.doi.org/10.1201/b13867]
[2]
Mao QQ, Xu XY, Cao SY, et al. Bioactive compounds and bioactivities of Ginger (Zingiber officinale Roscoe). Foods 2019; 8(6): 185.
[http://dx.doi.org/10.3390/foods8060185] [PMID: 31151279]
[3]
Prasad S, Tyagi AK. Ginger and its constituents: Role in prevention and treatment of gastrointestinal cancer. Gastroenterol Res Pract 2015; 2015: 1-11.
[http://dx.doi.org/10.1155/2015/142979] [PMID: 25838819]
[4]
Jafarzadeh A, Jafarzadeh S, Nemati M. Therapeutic potential of ginger against COVID-19: Is there enough evidence? J Tradit Chin Med Sci 2021; 8(4): 267-79.
[http://dx.doi.org/10.1016/j.jtcms.2021.10.001]
[5]
Bode AM, Dong Z. The amazing and mighty ginger. In: Benzie IFF, Wachtel GS, Eds. Herbal medicine: Biomolecular and clinical aspects. 2nd ed. Boca Raton, FL: CRC Press 2011.
[http://dx.doi.org/10.1201/b10787-8]
[6]
Liju VB, Jeena K, Kuttan R. Gastroprotective activity of essential oils from turmeric and ginger. J Basic Clin Physiol Pharmacol 2015; 26(1): 95-103.
[http://dx.doi.org/10.1515/jbcpp-2013-0165] [PMID: 24756059]
[7]
Ho SC, Chang KS, Lin CC. Anti-neuroinflammatory capacity of fresh ginger is attributed mainly to 10-gingerol. Food Chem 2013; 141(3): 3183-91.
[http://dx.doi.org/10.1016/j.foodchem.2013.06.010] [PMID: 23871076]
[8]
Akinyemi AJ, Thome GR, Morsch VM, et al. Effect of dietary supplementation of ginger and turmeric rhizomes on angiotensin-1 converting enzyme (ACE) and arginase activities in L-NAME induced hypertensive rats. J Funct Foods 2015; 17: 792-801.
[http://dx.doi.org/10.1016/j.jff.2015.06.011]
[9]
Suk S, Kwon GT, Lee E, et al. Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high fat diet fed mice. Mol Nutr Food Res 2017; 61(10): 1700139.
[http://dx.doi.org/10.1002/mnfr.201700139] [PMID: 28556482]
[10]
Zhang XX, Tian Y, Wang ZT, Ma YH, Tan L, Yu JT. The epidemiology of Alzheimer’s disease modifiable risk factors and prevention. J Prev Alzheimers Dis 2021; 8(3): 1-9.
[http://dx.doi.org/10.14283/jpad.2021.15] [PMID: 34101789]
[11]
Kandlur A, Satyamoorthy K, Gangadharan G. Oxidative stress in cognitive and epigenetic aging: A retrospective glance. Front Mol Neurosci 2020; 13: 41.
[http://dx.doi.org/10.3389/fnmol.2020.00041] [PMID: 32256315]
[12]
Ricciarelli R, Fedele E. The amyloid cascade hypothesis in Alzheimer’s disease: It’s time to change our mind. Curr Neuropharmacol 2017; 15(6): 926-35.
[http://dx.doi.org/10.2174/1570159X15666170116143743] [PMID: 28093977]
[13]
Ando K, Houben S, Homa M, et al. Alzheimer’s disease: Tau pathology and dysfunction of endocytosis. Front Mol Neurosci 2021; 13: 583755.
[http://dx.doi.org/10.3389/fnmol.2020.583755] [PMID: 33551742]
[14]
Chen XQ, Mobley WC. Exploring the pathogenesis of Alzheimer disease in basal forebrain cholinergic neurons: Converging insights from alternative hypotheses. Front Neurosci 2019; 13: 446.
[http://dx.doi.org/10.3389/fnins.2019.00446] [PMID: 31133787]
[15]
Unzeta M, Esteban G, Bolea I, et al. Multi target directed donepezillike ligands for Alzheimer’s disease. Front Neurosci 2016; 10: 205.
[http://dx.doi.org/10.3389/fnins.2016.00205] [PMID: 27252617]
[16]
Girek M. Szymański P. Tacrine hybrids as multi target directed ligands in Alzheimer’s disease: Influence of chemical structures on biological activities. Chem Pap 2019; 73(2): 269-89.
[http://dx.doi.org/10.1007/s11696-018-0590-8]
[17]
Cummings JL, Morstorf T, Zhong K. Alzheimer’s disease drug development pipeline: Few candidates, frequent failures. Alzheimers Res Ther 2014; 6(4): 37.
[http://dx.doi.org/10.1186/alzrt269] [PMID: 25024750]
[18]
Peng X, Xing P, Li X, et al. Towards personalized intervention for Alzheimer’s disease. Genomics Proteomics Bioinformatics 2016; 14(5): 289-97.
[http://dx.doi.org/10.1016/j.gpb.2016.01.006] [PMID: 27693548]
[19]
Hampel H. Current insights into the pathophysiology of Alzheimer’s disease: Selecting targets for early therapeutic intervention. Int Psychogeriatr 2012; 24(S1): S10-7.
[http://dx.doi.org/10.1017/S1041610212000579] [PMID: 22784422]
[20]
Schneider LS. Alzheimer disease pharmacologic treatment and treatment research. Continuum 2013; 19(2): 339-57.
[http://dx.doi.org/10.1212/01.CON.0000429180.60095.d0]
[21]
Azam F, Amer A, Abulifa A, Elzwawi M. Ginger components as new leads for the design and development of novel multi targeted anti alzheimer’s drugs: A computational investigation. Drug Des Devel Ther 2014; 8: 2045-59.
[http://dx.doi.org/10.2147/DDDT.S67778] [PMID: 25364231]
[22]
Ru J, Li P, Wang J, et al. TCMSP: A database of systems pharmacology for drug discovery from herbal medicines. J Cheminform 2014; 6(1): 13.
[http://dx.doi.org/10.1186/1758-2946-6-13] [PMID: 24735618]
[23]
Bhowmik A, Khan R, Ghosh MK. Blood brain barrier: A challenge for effectual therapy of brain tumors. BioMed Res Int 2015; 2015: 1-20.
[http://dx.doi.org/10.1155/2015/320941] [PMID: 25866775]
[24]
Daina A, Michielin O, Zoete V. SwissADME: A free web tool to evaluate pharmacokinetics, drug likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017; 7(1): 42717.
[http://dx.doi.org/10.1038/srep42717] [PMID: 28256516]
[25]
Djoumbou Feunang Y, Eisner R, Knox C, et al. ClassyFire: Automated chemical classification with a comprehensive, computable taxonomy. J Cheminform 2016; 8(1): 61.
[http://dx.doi.org/10.1186/s13321-016-0174-y] [PMID: 27867422]
[26]
Backman TW, Cao Y, Girke T. ChemMine tools: An online service for analyzing and clustering small molecules. Nucleic Acids Res 2011; 39: W486-91.
[http://dx.doi.org/10.1093/nar/gkr320]
[27]
Gfeller D, Grosdidier A, Wirth M, Daina A, Michielin O, Zoete V. SwissTargetPrediction: A web server for target prediction of bioactive small molecules. Nucleic Acids Res 2014; 42: W32-38.
[http://dx.doi.org/10.1093/nar/gku293]
[28]
Mlecnik B, Galon J, Bindea G. Automated exploration of gene ontology term and pathway networks with ClueGO-REST. Bioinformatics 2019; 35(19): 3864-6.
[http://dx.doi.org/10.1093/bioinformatics/btz163] [PMID: 30847467]
[29]
Ge SX, Jung D, Yao R, Shiny GO. A graphical gene set enrichment tool for animals and plants. Bioinformatics 2020; 36(8): 2628-9.
[http://dx.doi.org/10.1093/bioinformatics/btz931] [PMID: 31882993]
[30]
Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 2017; 45(D1): D353-61.
[http://dx.doi.org/10.1093/nar/gkw1092] [PMID: 27899662]
[31]
Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: Protein protein association networks with increased coverage, supporting functional discovery in genome wide experimental datasets. Nucleic Acids Res 2019; 47(D1): D607-13.
[http://dx.doi.org/10.1093/nar/gky1131] [PMID: 30476243]
[32]
Otasek D, Morris JH, Bouças J, Pico AR, Demchak B. Cytoscape Automation: Empowering workflow based network analysis. Genome Biol 2019; 20(1): 185.
[http://dx.doi.org/10.1186/s13059-019-1758-4] [PMID: 31477170]
[33]
Wu Z, Li W, Liu G, Tang Y. Network based methods for prediction of drug-target interactions. Front Pharmacol 2018; 9: 1134.
[http://dx.doi.org/10.3389/fphar.2018.01134] [PMID: 30356768]
[34]
Anitha P, Anbarasu A, Ramaiah S. Gene network analysis reveals the association of important functional partners involved in antibiotic resistance: A report on an important pathogenic bacterium Staphylococcus aureus. Gene 2016; 575(2): 253-63.
[http://dx.doi.org/10.1016/j.gene.2015.08.068] [PMID: 26342962]
[35]
Mitra K, Carvunis AR, Ramesh SK, Ideker T. Integrative approaches for finding modular structure in biological networks. Nat Rev Genet 2013; 14(10): 719-32.
[http://dx.doi.org/10.1038/nrg3552] [PMID: 24045689]
[36]
Bader GD, Hogue CWV. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics 2003; 4(1): 2.
[http://dx.doi.org/10.1186/1471-2105-4-2] [PMID: 12525261]
[37]
Abraham MJ, Murtola T, Schulz R, et al. GROMACS: High performance molecular simulations through multi level parallelism from laptops to supercomputers. SoftwareX 2015; 1-2: 19-25.
[http://dx.doi.org/10.1016/j.softx.2015.06.001]
[38]
Schmid N, Eichenberger AP, Choutko A, et al. Definition and testing of the GROMOS force field versions 54A7 and 54B7. Eur Biophys J 2011; 40(7): 843-56.
[http://dx.doi.org/10.1007/s00249-011-0700-9] [PMID: 21533652]
[39]
Fang ZZ, Zhang YY, Wang XL, Cao YF, Huo H, Yang L. Bioactivation of herbal constituents: Simple alerts in the complex system. Expert Opin Drug Metab Toxicol 2011; 7(8): 989-1007.
[http://dx.doi.org/10.1517/17425255.2011.586335] [PMID: 21609190]
[40]
Grobelny BT, London D, Hill TC, North E, Dugan P, Doyle WK. Betweenness centrality of intracranial electroencephalography networks and surgical epilepsy outcome. Clin Neurophysiol 2018; 129(9): 1804-12.
[http://dx.doi.org/10.1016/j.clinph.2018.02.135] [PMID: 29981955]
[41]
Wang J, Li Y, Yang Y, et al. A new strategy for deleting animal drugs from traditional chinese medicines based on modified Yimusake Formula. Sci Rep 2017; 7(1): 1504.
[http://dx.doi.org/10.1038/s41598-017-01613-7] [PMID: 28473709]
[42]
Kumar N, Srivastava R, Prakash A, Lynn AM. Structure based virtual screening, molecular dynamics simulation and MM-PBSA toward identifying the inhibitors for two component regulatory system protein NarL of Mycobacterium tuberculosis. J Biomol Struct Dyn 2020; 38(11): 3396-410.
[http://dx.doi.org/10.1080/07391102.2019.1657499] [PMID: 31422761]
[43]
Hampel H, Mesulam MM, Cuello AC, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018; 141(7): 1917-33.
[http://dx.doi.org/10.1093/brain/awy132] [PMID: 29850777]
[44]
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides 2015; 52: 1-18.
[http://dx.doi.org/10.1016/j.npep.2015.06.008] [PMID: 26149638]
[45]
Gianvito G, Andrea D. Molecular simulations of amyloid beta assemblies. Adv Phys X 2020; 5: 1.
[46]
Choi JG, Kim SY, Jeong M, Oh MS. Pharmacotherapeutic potential of ginger and its compounds in age related neurological disorders. Pharmacol Ther 2018; 182: 56-69.
[http://dx.doi.org/10.1016/j.pharmthera.2017.08.010] [PMID: 28842272]
[47]
Rahmani AH, Shabrmi FM, Aly SM. Active ingredients of ginger as potential candidates in the prevention and treatment of diseases via modulation of biological activities. Int J Physiol Pathophysiol Pharmacol 2014; 6(2): 125-36.
[PMID: 25057339]
[48]
Mohd Yusof YA. Gingerol and its role in chronic diseases. Adv Exp Med Biol 2016; 929: 177-207.
[http://dx.doi.org/10.1007/978-3-319-41342-6_8] [PMID: 27771925]
[49]
Lee C, Park GH, Kim CY, Jang JH. [6]-Gingerol attenuates β-amyloid-induced oxidative cell death via fortifying cellular antioxidant defense system. Food Chem Toxicol 2011; 49(6): 1261-9.
[http://dx.doi.org/10.1016/j.fct.2011.03.005] [PMID: 21396424]
[50]
Zeng G, Zong S, Zhang Z, et al. The role of 6-Gingerol on inhibiting amyloid beta protein-induced apoptosis in PC12 cells. Rejuvenation Res 2015; 18(5): 413-21.
[http://dx.doi.org/10.1089/rej.2014.1657] [PMID: 25811848]
[51]
Chen CY, Kao CL, Liu CM. The cancer prevention, anti inflammatory and anti-oxidation of bioactive phytochemicals targeting the TLR4 signaling pathway. Int J Mol Sci 2018; 19(9): 2729.
[http://dx.doi.org/10.3390/ijms19092729] [PMID: 30213077]
[52]
Lee HY, Park SH, Lee M, et al. 1-Dehydro-[10]-gingerdione from ginger inhibits IKKβ activity for NF-κB activation and suppresses NF-κB-regulated expression of inflammatory genes. Br J Pharmacol 2012; 167(1): 128-40.
[http://dx.doi.org/10.1111/j.1476-5381.2012.01980.x] [PMID: 22489648]

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