Generic placeholder image

Current Drug Discovery Technologies

Editor-in-Chief

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

Research Article

Nutritional Profile, GC-MS Analysis and In-silico Anti-diabetic Phytocompounds Candidature of Jatropha gossypifolia Leaf Extracts

Author(s): Olasunkanmi Kayode Awote*, Rahmon Ilesanmi Kanmodi, Success Chidera Ebube and Zainab Folashade Abdulganniyyu

Volume 21, Issue 3, 2024

Published on: 10 October, 2023

Article ID: e221223224764 Pages: 14

DOI: 10.2174/0115701638267143230925172207

Price: $65

conference banner
Abstract

Background: Diabetes mellitus (DM) is a metabolic disorder known to impair many physiological functions via reactive oxygen species (ROS). Aldose reductase, sorbitol dehydrogenase, dipeptidyl peptidase IV, α-amylase and α-glucosidase are pharmacotherapeutic protein targets in type-2 diabetes mellitus (T2DM). Inhibitors of these enzymes constitute a new class of drugs used in the management and treatment of T2DM. Some reports have claimed that medicinal plant extracts that serves as food (and as an antioxidant source) can reduce these alterations by eliminating ROS caused by DM. Ethnobotanical survey claims Jatropha gossypifolia commonly called “fignut” and “Lapa-lapa” in the Yoruba land of South-western Nigeria, to be used for the treatment and management of diabetes, in addition to its nutritive value.

Objective: The nutritional composition and in-silico antidiabetic potential of the bioactive constituents of J. gossypifolia leaf extracts were investigated.

Methods: Proximate, minerals and gas chromatography–mass spectroscopy (GC-MS) analysis were carried out using standard procedures. Phytocompounds present in J. gossypifolia methanol (JGM) and ethyl acetate (JGE) leaf extracts were tested as potential antagonists of selected protein targets via in-silico techniques. Drug-likeness, pharmacokinetic properties and toxicity of the promising docked ligands were also predicted.

Results: The proximate and mineral analysis revealed good nutritional composition and mineral content. Additionally, cyclo-pentadecane and dibutyl phthalate from methanol extract, and benzene- 1,2,4,5-tetramethyl, benzene-1,2,3,5-tetramethyl, and benzene-1,3-dimethyl-5-(1-methylethyl) from ethyl acetate extract were present in J. gossypifolia leaf which exhibited a better binding affinity than the clinically prescribed standard, metformin.

Conclusion: Benzene-1,2,4,5-tetramethyl from JGE extracts exhibited the most promising antidiabetic potential in-silico, suggesting its candidature as diabetes-target-protein inhibitor which may be developed for the treatment of type-2 diabetes mellitus.

Graphical Abstract

[1]
American Diabetes Association. Standards of medical care in diabetes—2022 abridged for primary care providers. Clin Diabetes 2022; 40(1): 10-38.
[http://dx.doi.org/10.2337/cd22-as01] [PMID: 35221470]
[2]
Rana A, Samtiya M, Dhewa T, Mishra V, Aluko RE. Health benefits of polyphenols: A concise review. J Food Biochem 2022; 46(10): e14264.
[http://dx.doi.org/10.1111/jfbc.14264] [PMID: 35694805]
[3]
Wu Q, Patocka J, Nepovimova E, Kuca K. Jatropha gossypiifolia L. and its biologically active metabolites: A mini review. J Ethnopharmacol 2019; 234: 197-203.
[http://dx.doi.org/10.1016/j.jep.2019.01.022] [PMID: 30695706]
[4]
Bharathy V, Sumathy BM, Uthayakumari F. Determination of phytocomponents by GC-MS in leaves of Jatropha gossypifolia L. Science Research Reporter 2012; 2(3): 286-90.
[5]
Singh P, Dabas A, Srivastava R, Nagpure NS, Singh A. Evaluation of Genotoxicity Induced by Medicinal Plant Jatropha gossypifolia in Freshwater Fish Channa punctatus (Bloch). Turk J Fish Aquat Sci 2014; 14(2): 421-8.
[http://dx.doi.org/10.4194/1303-2712-v14_2_12]
[6]
Ilavarasan R, Vijayakumar AR, Daniel EP, Venkataraman S, Vijayakumar S. Ulcer protective activity of Jatropha gossypiifolia Linn. in wistar rats. Pharmacognosy Res 2016; 8(5) (Suppl. 1): 61.
[http://dx.doi.org/10.4103/0974-8490.178640] [PMID: 27114695]
[7]
Saleem H, Ahmad I, Gill MSA. Evaluation of lipoxygenase inhibition of Jatropha gossypifolia, a medicinal plant from Pakistan. Bangladesh J Pharmacol 2016; 11(2): 319-20.
[http://dx.doi.org/10.3329/bjp.v11i2.26516]
[8]
Karalliedde J, Gnudi L. Diabetes mellitus, a complex and heterogeneous disease, and the role of insulin resistance as a determinant of diabetic kidney disease. Nephrol Dial Transplant 2016; 31(2): 206-13.
[PMID: 25550448]
[9]
Petrash JM. All in the family: aldose reductase and closely related aldo-keto reductases. Cell Mol Life Sci 2004; 61(7-8): 737-49.
[http://dx.doi.org/10.1007/s00018-003-3402-3] [PMID: 15094999]
[10]
Barski OA, Tipparaju SM, Bhatnagar A. The aldo-keto reductase superfamily and its role in drug metabolism and detoxification. Drug Metab Rev 2008; 40(4): 553-624.
[http://dx.doi.org/10.1080/03602530802431439] [PMID: 18949601]
[11]
Tang WH, Martin KA, Hwa J. Aldose reductase, oxidative stress, and diabetic mellitus. Front Pharmacol 2012; 3: 87.
[http://dx.doi.org/10.3389/fphar.2012.00087] [PMID: 22582044]
[12]
Alexiou P, Pegklidou K, Chatzopoulou M, Nicolaou I, Demopoulos V. Aldose reductase enzyme and its implication to major health problems of the 21(st) century. Curr Med Chem 2009; 16(6): 734-52.
[http://dx.doi.org/10.2174/092986709787458362] [PMID: 19199934]
[13]
Srikanth KK, Orrick JA. Biochemistry, polyol or sorbitol pathways. StatPearls. StatPearls Publishing 2022. Internet
[14]
Sofowora A. Research on medicinal plants and traditional medicine in Africa. J Altern Complement Med 1996; 2(3): 365-72.
[http://dx.doi.org/10.1089/acm.1996.2.365] [PMID: 9395671]
[15]
Ojewunmi O, Oshodi T, Ogundele O, Chijioke M, Adenekan S. Toxicity screening and in vitro antioxidant activities of aqueous extracts of Morinda lucida and Saccharum officinarum leaves. Biokemistri 2013; 25(2): 72-8.
[16]
Behlil F, Ullah S, Khan N, et al. Phytochemical screening and antioxidant activity determination of some medicinally important plants of Balochistan. Pak J Bot 2019; 51(2): 1-8.
[http://dx.doi.org/10.30848/PJB2019-2(14)]
[17]
AOCS. Official Methods and Recommended Practices of the American Oil Chemists Society. Champaing 1993.
[18]
AOAC. Official and Tentative Methods of the AOAC International Maryland 1995.
[19]
AOAC. Official method of Analysis. 18th ed. Washington, DC: Association of Officiating Analytical Chemists 2005.
[20]
Oduntan AO, Fasoyiro SB, Akinfasoye JA, Adeboyejo FO, Akintoye HA. Antioxidant and proximate properties of underutilized vegetables in western Nigeria. III All Africa Horticultural Congress 1225. 255-60.
[21]
Martínezherrera J, Siddhuraju P, Francis G, Davilaortiz G, Becker K. Chemical composition, toxic/antimetabolic constituents, and effects of different treatments on their levels, in four provenances of L. from Mexico. Food Chem 2006; 96(1): 80-9.
[http://dx.doi.org/10.1016/j.foodchem.2005.01.059]
[22]
Kongkiatpaiboon S, Gritsanapan W. Optimized extraction for high yield of insecticidal didehydrostemofoline alkaloid in Stemona collinsiae root extracts. Ind Crops Prod 2013; 41: 371-4.
[http://dx.doi.org/10.1016/j.indcrop.2012.04.047]
[23]
Mehmood A, Javid S, Khan MF, Ahmad KS, Mustafa A. In vitro total phenolics, total flavonoids, antioxidant and antibacterial activities of selected medicinal plants using different solvent systems. BMC Chem 2022; 16(1): 64.
[http://dx.doi.org/10.1186/s13065-022-00858-2] [PMID: 36030245]
[24]
Dias MC, Pinto DCGA, Silva AMS. Plant flavonoids: Chemical characteristics and biological activity. Molecules 2021; 26(17): 5377.
[http://dx.doi.org/10.3390/molecules26175377] [PMID: 34500810]
[25]
Sulaimon LA, Anise EO, Obuotor EM, et al. In vitro antidiabetic potentials, antioxidant activities and phytochemical profile of african black pepper (Piper guineense). Clinical Phytoscience 2020; 6(1): 90.
[http://dx.doi.org/10.1186/s40816-020-00236-2]
[26]
Hbika A, Daoudi NE, Bouyanzer A, et al. Artemisia absinthium L. aqueous and ethyl acetate extracts: Antioxidant effect and potential activity in vitro and in vivo against pancreatic α-amylase and intestinal α-glucosidase. Pharmaceutics 2022; 14(3): 481.
[http://dx.doi.org/10.3390/pharmaceutics14030481] [PMID: 35335858]
[27]
Riyaphan J, Pham DC, Leong MK, Weng CF. In silico approaches to identify polyphenol compounds as α-glucosidase and α-amylase inhibitors against type-II diabetes. Biomolecules 2021; 1(12): 1877.
[28]
Fagbohun OF, Oriyomi OV, Adekola MB, Msagati TAM. Biochemical applications of Kigelia africana (Lam.) Benth. fruit extracts in diabetes mellitus. Comp Clin Pathol 2020; 29(6): 1251-64.
[http://dx.doi.org/10.1007/s00580-020-03179-9]
[29]
Perumal PC, Sowmya S, Pratibha P, et al. Identification of novel PPARγ agonist from GC-MS analysis of ethanolic extract of Cayratia trifolia (L.): a computational molecular simulation studies. J Appl Pharm Sci 2014; 4(9): 006-11.
[30]
Deng T, Zhang Y, Wu Y, et al. Dibutyl phthalate exposure aggravates type 2 diabetes by disrupting the insulin-mediated PI3K/AKT signaling pathway. Toxicol Lett 2018; 290: 1-9.
[http://dx.doi.org/10.1016/j.toxlet.2018.03.004] [PMID: 29526571]
[31]
Yang R, Zheng J, Qin J, et al. Dibutyl phthalate affects insulin synthesis and secretion by regulating the mitochondrial apoptotic pathway and oxidative stress in rat insulinoma cells. Ecotoxicol Environ Saf 2023; 249: 114396.
[http://dx.doi.org/10.1016/j.ecoenv.2022.114396] [PMID: 36508788]
[32]
Williams MJ, Wiemerslage L, Gohel P, Kheder S, Kothegala LV, Schiöth HB. Dibutyl phthalate exposure disrupts evolutionarily conserved insulin and glucagon-like signaling in drosophila males. Endocrinology 2016; 157(6): 2309-21.
[http://dx.doi.org/10.1210/en.2015-2006] [PMID: 27100621]
[33]
Bohara M, Ghaju S, Sharma K, Kalauni SK, Khadayat K. In vitro and In silico analysis of Bergenia ciliata and Mimosa pudica for inhibition of α-Amylase. J Chem 2022; 2022: 1-10.
[http://dx.doi.org/10.1155/2022/6997173]

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