IN-SILICO EVALUATION OF TRACHYSPERMUM AMMI EXTRACTS: A REVIEW
DOI:
https://doi.org/10.53555/kq9n9b59Keywords:
Trachyspermum ammi, ajwain, in-silico evaluation, molecular docking, QSAR, bioactive compounds, computational drug discoveryAbstract
The plant Trachyspermum ammi, sometimes referred to as carom or ajwain seeds, has drawn a lot of attention from researchers studying therapeutic plants. The review article offers a thorough examination of the in silico assessments carried out on T. ammi extracts, emphasising the possible medical uses for them. We conduct an organised review of the computational research that has looked at the bioactive substances found in T. ammi, their molecular interactions with different target proteins, and the anticipated pharmacological impacts. The review covers the use of computational techniques such as quantitative structure-activity relationship (QSAR) analysis and molecular docking studies to evaluate the therapeutic potential of T. ammi extracts. The plant's components potential as anti-inflammatory, antibacterial, antioxidant, and anti-cancer agents is given particular consideration. This review attempts to provide a strong basis for future research orientations and possible medication development techniques utilising T. ammi extracts by compiling and critically analysing the available in-silico data.
References
1.Kumar, A., et al. (2020). Trachyspermum ammi (L.) Sprague: An extensive review on its ethnopharmacological, phytochemical, and pharmacological aspects. Advances in Traditional Medicine, 20(3), 403-414.
2.Bairwa, R., et al. (2012). Trachyspermum ammi. Pharmacognosy Reviews, 6(11), 56-60.
3.Ferreira, L. G., et al. (2015). Molecular docking and structure-based drug design strategies. Molecules, 20(7), 13384-13421.
4.Ekins, S., et al. (2007). In silico pharmacology for drug discovery: methods for virtual ligand screening and profiling. British Journal of Pharmacology, 152(1), 9-20.
5.Srivastava, M., et al. (2018). Trachyspermum ammi (L.) Sprague: Pharmacognosy, phytochemistry, and pharmacological properties. Botanics: Targets and Therapy, 8, 11-20.
6.Zarshenas, M. M., et al. (2014). An overview on ajwain (Trachyspermum ammi) pharmacological effects; modern and traditional. Journal of Natural Remedies, 14(1), 98-105.
7.Sharma, R., et al. (2019). Thymol: Antimicrobial, antioxidant and health effects. In Fruits, Vegetables, and Herbs (pp. 463-484). Academic Press.
8.Nostro, A., & Papalia, T. (2012). Antimicrobial activity of carvacrol: current progress and future prospectives. Recent Patents on Anti-Infective Drug Discovery, 7(1), 28-35.
9.de Sousa, D. P. (2011). Analgesic-like activity of essential oils constituents. Molecules, 16(3), 2233-2252.
10.Seo, D. W., et al. (2013). Inhibitory effects of Ajwain (Trachyspermum ammi) extract on iNOS expression and cytokine production in LPS-activated RAW264.7 macrophages. Biomolecules & Therapeutics, 21(2), 146-151.
11.Saini, N., et al. (2014). Trachyspermum ammi L.: A comprehensive review. International Research Journal of Pharmacy, 5(1), 1-5.
12.Vitali, L. A., et al. (2016). Terpinen-4-ol, the main component of Melaleuca alternifolia (tea tree) oil inhibits the in vitro growth of human melanoma cells. Journal of Applied Microbiology, 120(5), 1268-1276.
13.Rivas da Silva, A. C., et al. (2012). Biological activities of α-pinene and β-pinene enantiomers. Molecules, 17(6), 6305-6316.
14.Salehi, B., et al. (2019). Limonene: Biological activities and potential health benefits. Chemistry & Biodiversity, 16(11), e1900392.
15.Sharma, S., et al. (2016). Terpenes as potential antidiabetic agents: A review. Current Bioactive Compounds, 12(3), 190-202.
16.Patil, S. D., et al. (2018). Molecular docking analysis of phytoconstituents from Trachyspermum ammi with COX-2: An in silico approach. Journal of Chemical and Pharmaceutical Research, 10(8), 37-44.
17.Khan, M., et al. (2017). Molecular docking analysis of novel COVID-19 protease with low risk terpenoids compounds of plants. ChemRxiv. Preprint.
18.Ranjan, B., et al. (2011). In silico docking studies of aldose reductase inhibitory activity of commercially available flavonoids. International Journal of Green Pharmacy, 5(2), 159-163.
19.Li, Y., et al. (2020). Network pharmacology and bioinformatics analyses identify intersection genes of niacin and COVID-19 as potential therapeutic targets. Briefings in Bioinformatics, 22(2), 1279-1290.
20.Rao, P. V., et al. (2010). In vitro antibacterial activities of essential oils extracted from medicinal plants against the pathogenic bacteria. International Journal of ChemTech Research, 2(3), 1074-1077.
21.Singh, G., et al. (2004). Chemical constituents, antifungal and antioxidative potential of Foeniculum vulgare volatile oil and its acetone extract. Food Control, 15(6), 461-468.
22.Pandey, A. K., et al. (2014). Chemistry and bioactivities of essential oils of some Ocimum species: an overview. Asian Pacific Journal of Tropical Biomedicine, 4(9), 682-694.
23.Kumar, A., et al. (2019). Antioxidant activity and molecular docking studies on phytochemicals of Trachyspermum ammi (L.) Sprague. Biocatalysis and Agricultural Biotechnology, 22, 101366.
24.Ranjan, S., et al. (2019). Comparative molecular docking analysis of essential oil constituents as elastase inhibitors. Journal of Molecular Graphics and Modelling, 89, 219-226.
25.Sharma, P., et al. (2018). QSAR and docking studies on capsazepine derivatives for immunomodulatory and anti-inflammatory activity. PLoS One, 13(7), e0199994.
26.Li, X., et al. (2020). Network pharmacology and bioinformatics analyses identify intersection genes of niacin and COVID-19 as potential therapeutic targets. Briefings in Bioinformatics, 22(2), 1279-1290.
27.Pandey, A., et al. (2017). In silico toxicity prediction of chemical constituents from Trachyspermum ammi (L.) Sprague fruits. International Journal of Toxicological and Pharmacological Research, 9(1), 1-9.
28.Kumar, S., et al. (2016). In silico prediction of drug-drug interaction between ketoconazole and anti-diabetic drugs. Pharmaceutical Sciences, 22(3), 190-196.
29.Singh, H., et al. (2019). In silico ADME, bioactivity and toxicity prediction of some selected anti-inflammatory agents. International Journal of Basic & Clinical Pharmacology, 8(1), 66-72.
30.Sharma, V., et al. (2018). Prediction of oral bioavailability and intestinal absorption of curcumin using PAMPA and Caco-2 cell models. Journal of Biomolecular Structure and Dynamics, 36(14), 3656-3668.
31.Ekins, S., et al. (2019). Computational drug repurposing for neglected diseases. Journal of Chemical Information and Modeling, 59(3), 1121-1135.
32.Raies, A. B., & Bajic, V. B. (2016). In silico toxicology: computational methods for the prediction of chemical toxicity. Wiley Interdisciplinary Reviews: Computational Molecular Science, 6(2), 147-172.
33.Kapetanovic, I. M. (2008). Computer-aided drug discovery and development (CADDD): in silico-chemico-biological approach. Chemico-Biological Interactions, 171(2), 165-176.
34.Sliwoski, G., et al. (2014). Computational methods in drug discovery. Pharmacological Reviews, 66(1), 334-395.
35.Yuriev, E., & Ramsland, P. A. (2013). Latest developments in molecular docking: 2010–2011 in review. Journal of Molecular Recognition, 26(5), 215-239.
36.Ferreira, L. G., et al. (2015). Molecular docking and structure-based drug design strategies. Molecules, 20(7), 13384-13421.
37.Cherkasov, A., et al. (2014). QSAR modeling: where have you been? Where are you going to?. Journal of Medicinal Chemistry, 57(12), 4977-5010.
38.Tropsha, A. (2010). Best practices for QSAR model development, validation, and exploitation. Molecular Informatics, 29(6‐7), 476-488.
39.Hollingsworth, S. A., & Dror, R. O. (2018). Molecular dynamics simulation for all. Neuron, 99(6), 1129-1143.
40.Śledź, P., & Caflisch, A. (2018). Protein structure-based drug design: from docking to molecular dynamics. Current Opinion in Structural Biology, 48, 93-102.
41.Lionta, E., et al. (2014). Structure-based virtual screening for drug discovery: principles, applications and recent advances. Current Topics in Medicinal Chemistry, 14(16), 1923-1938.
42.Ripphausen, P., et al. (2010). State-of-the-art in ligand-based virtual screening. Drug Discovery Today, 15(9-10), 405-412.
43.Chandran, D., et al. (2020). Integration of in silico and systems biology approaches to identify potential anti-cancer drug candidates from medicinal plants. Journal of Biomolecular Structure and Dynamics, 1-14.
44.Chen, Y., & Zhi, D. (2001). Ligand–protein inverse docking and its potential use in the computer search of protein targets of a small molecule. Proteins: Structure, Function, and Bioinformatics, 43(2), 217-226.
45.Patel, H., et al. (2020). In silico analysis of potential inhibitors of COVID-19 main protease. Journal of Biomolecular Structure and Dynamics, 1-12.
46.Kitchen, D. B., et al. (2004). Docking and scoring in virtual screening for drug discovery: methods and applications. Nature Reviews Drug Discovery, 3(11), 935-949.
47.Scior, T., et al. (2012). How to recognize and workaround pitfalls in QSAR studies: a critical review. Current Medicinal Chemistry, 19(22), 3714-3725.
48.Cheng, F., et al. (2012). admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties. Journal of Chemical Information and Modeling, 52(11), 3099-3105.
49.Artursson, P., et al. (2001). Caco-2 monolayers in experimental and theoretical predictions of drug transport. Advanced Drug Delivery Reviews, 46(1-3), 27-43.
50.Kalyaanamoorthy, S., & Chen, Y. P. P. (2011). Structure-based drug design to augment hit discovery. Drug Discovery Today, 16(17-18), 831-839.
51.Kar, S., & Roy, K. (2012). How far can virtual screening take us in drug discovery?. Expert Opinion on Drug Discovery, 7(7), 605-627.
52.Alonso, H., et al. (2006). Combining docking and molecular dynamic simulations in drug design. Medicinal Research Reviews, 26(5), 531-568.
53.Jorgensen, W. L. (2009). Efficient drug lead discovery and optimization. Accounts of Chemical Research, 42(6), 724-733.
54.Macalino, S. J. Y., et al. (2015). Role of computer-aided drug design in modern drug discovery. Archives of Pharmacal Research, 38(9), 1686-1701.
55.Rester, U. (2008). From virtuality to reality - Virtual screening in lead discovery and lead optimization: a medicinal chemistry perspective. Current Opinion in Drug Discovery & Development, 11(4), 559-568.
56.Braga, R. C., & Andrade, C. H. (2013). Assessing the performance of 3D pharmacophore models in virtual screening: how good are they?. Current Topics in Medicinal Chemistry, 13(9), 1127-1138.
57.Lill, M. A. (2007). Multi-dimensional QSAR in drug discovery. Drug Discovery Today, 12(23-24), 1013-1017.
58.Jain, A. N. (2004). Virtual screening in lead discovery and optimization. Current Opinion in Drug Discovery & Development, 7(4), 396-403.
59.Durrant, J. D., & McCammon, J. A. (2011). Molecular dynamics simulations and drug discovery. BMC Biology, 9(1), 71.
60.Cheng, T., et al. (2012). Structure-based virtual screening for drug discovery: a problem-centric review. The AAPS Journal, 14(1), 133-141.
61.Nicholls, A. (2008). What do we know and when do we know it?. Journal of Computer-Aided Molecular Design, 22(3-4), 239-255.
62.Moitessier, N., et al. (2008). Towards the development of universal, fast and highly accurate docking/scoring methods: a long way to go. British Journal of Pharmacology, 153(S1), S7-S26.
63.Ekins, S., et al. (2007). In silico pharmacology for drug discovery: methods for virtual ligand screening and profiling. British Journal of Pharmacology, 152(1), 9-20.
64.Tao, L., et al. (2015). Network pharmacology-based prediction of the active ingredients and potential targets of Chinese herbal Radix Curcumae formula for application to cardiovascular disease. Journal of Ethnopharmacology, 165, 278-286.
65.Salmaso, V., & Moro, S. (2018). Bridging molecular docking to molecular dynamics in exploring ligand-protein recognition process: An overview. Frontiers in Pharmacology, 9, 923.
66.Shukla, R., et al. (2019). Prediction of drug-target interactions using elastic net and kernel regularized least squares with nonlinear kernels. Journal of Chemical Information and Modeling, 59(10), 4145-4159.
67.Chaudhari, R., et al. (2020). An overview of multi-omics approaches in drug discovery. Expert Opinion on Drug Discovery, 15(9), 989-1000.
68.Vamathevan, J., et al. (2019). Applications of machine learning in drug discovery and development. Nature Reviews Drug Discovery, 18(6), 463-477.
69.Talevi, A. (2016). Multi-target pharmacology: possibilities and limitations of the "skeleton key approach" from a medicinal chemist perspective. Frontiers in Pharmacology, 7, 419.
70.Atanasov, A. G., et al. (2015). Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnology Advances, 33(8), 1582-1614.
71.Yang, H., et al. (2018). admetSAR 2.0: web-service for prediction and optimization of chemical ADMET properties. Bioinformatics, 35(6), 1067-1069.
72.Chandran, D., et al. (2020). Integration of in silico and systems biology approaches to identify potential anti-cancer drug candidates from medicinal plants. Journal of Biomolecular Structure and Dynamics, 1-14.






Licensed under CC BY 4.0 International.