In Silico Characterization of a Novel Bioactive Compound derived from Psidium guajava 4-[5-(Pyridin-4-yl)-1,2,4-oxadiazol-3-yl]-1,2,5-oxadiazol-3-amine: A Potential Inhibitor for Targeting Signaling Proteins involved in Diabetes Development

Authors

  • Maya Yadav Department of Botany, Dr. Shyama Prasad Mukherjee Govt. Degree College, Bhadohi-221401, Uttar Pradesh, India
  • RAJ KUMAR SINGH YADAV Department of Physics, Dr. Shyama Prasad Mukherjee Govt. Degree College, Bhadohi-221401, Uttar Pradesh, India

DOI:

https://doi.org/10.25004/IJPSDR.2024.160207

Keywords:

Type 2 diabetes mellitus (T2DM), Molecular docking, ADMET, bioactive compounds

Abstract

Pharmacological interventions for diabetes predominantly involve chemically synthesized compounds, often causing undesirable side effects. This has led to a growing interest in plant-based therapeutic alternatives. Technological advancements have facilitated the discovery of bioactive phytochemicals with medicinal properties. This study employs molecular docking analysis to assess the anti-diabetic potential of a naturally derived compound, 4-[5-(Pyridin-4-yl)-1,2,4-oxadiazol-3-yl]-1,2,5-oxadiazol-3-amine (POA), obtained from Psidium guajava leaf extract. The evaluation focuses on its inhibitory action against four human proteins 11 β -HSD1 (PDB: 4K1L), GFAT (PDB ID: 2ZJ4), SIRT6 (PDB ID: 3K35) and Aldose reductase (PDB ID: 3G5E) associated with diabetes. Physicochemical, pharmacokinetic, and ADMET profiles were computed using online web servers Molinspiration, ADMETLAB 2.0, and SWISSADME. POA demonstrated superior binding affinity (in Kcal/mol) -8.0, -7.5, -8.9  and -9.5, respectively) compared to the widely used diabetic drug Metformin -5.4, -6.0, -5.4 and -7.2 with these receptor proteins. Based on molecular docking studies and pharmacokinetics/ADMET profiles, POA may act as a multi-targeted, less harmful, and more efficacious medication for Type 2 Diabetes Mellitus (T2DM) compared to Metformin.

Downloads

Download data is not yet available.

References

Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract [Internet]. 2018 Apr 1 [cited 2023 Nov 21];138:271–81. Available from: https://pubmed.ncbi.nlm.nih.gov/29496507/

Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022 Jan 1;183:109119.

Holman N, Young B, Gadsby R. Current prevalence of Type 1 and Type 2 diabetes in adults and children in the UK. Diabet Med. 2015;32(9):1119–20.

Unnikrishnan R, Misra A. Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID- 19 . The COVID-19 resource centre is hosted on Elsevier Connect , the company ’ s public news and information . 2020;(January).

Damián MK, Salinas MY, Milenkovic D, Figueroa YL, Marino ME, Higuera CI, et al. In silico analysis of antidiabetic potential of phenolic compounds from blue corn (Zea mays L.) and black bean (Phaseolus vulgaris L.). Heliyon. 2020;6(3):3632.

Hameed I, Masoodi SR, Mir SA, Nabi M, Ghazanfar K, Ganai BA. Type 2 diabetes mellitus: From a metabolic disorder to an inflammatory condition. World J Diabetes [Internet]. 2015 May 5 [cited 2023 Nov 23];6(4):598. Available from: /pmc/articles/PMC4434080/

Elkhalifa AEO, Al-Shammari E, Adnan M, Alcantara JC, Mehmood K, Eltoum NE, et al. Development and characterization of novel biopolymer derived from abelmoschus esculentus l. Extract and its antidiabetic potential. Molecules. 2021;26(12):3609-20.

Ashraf SA, Elkhalifa AEO, Mehmood K, Adnan M, Khan MA, Eltoum NE, et al. Multi-targeted molecular docking, pharmacokinetics, and drug-likeness evaluation of okra-derived ligand abscisic acid targeting signaling proteins involved in the development of diabetes. Molecules. 2021 Oct 1;26(19):5957-79.

Luo W, Deng J, He J, Yin L, You R, Zhang L, et al. Integration of molecular docking, molecular dynamics and network pharmacology to explore the multi-target pharmacology of fenugreek against diabetes. J Cell Mol Med. 2023 Jul 1;27(14):1959–74.

Aliyu SM, Salisu N, Muhammad IA, Umar S, Fatima Zahra A, Kurya AU. Ethnobotanical survey of medicinal plants used for the treatment of typhoid fever in Kaduna Metropolis, Nigeria. Int J Curr Res Biosci Plant Biol. 2020;7(7):47–53.

Halimat A, Suleiman Yusuf M, Oladipupo Abdulazeez Yusuf D, Bala Sidi A, Nasiru Usman A, Rabiat Unekwu H, et al. Ethnobotanical survey of medicinal plants used in the treatment of gastrointestinal tract infections in Ebiraland Kogi state, Nigeria. ~ 38 ~ J Med Plants Stud [Internet]. 2020;8(1):38–44. Available from: http://www.plantsjournal.com

Aguoru, C. U & Ogaba JO. Ethnobotanical survey of anti-typhoid plants amongst the Idoma people of Nigeria. Int Sci Res J. 2010;2:34–40.

Ighere DA, Edagbo DE, Borokini TI, Alowonle AA, Michael C. Herbs used by the urhobo people in delta state Nigeria for the treatment of typhoid fever. Int J Recent Sci Res. 2012; 3(6):478–481.

Daskum AM, Chessed G, Dazigau HA. Ethnobotanical Survey of Plants in Folklore Medicine of Selected Communities of Yobe State, North-East Nigeria. 2020;3:6–22. Available from: http://www.ajtcam.ir

Ajibesin KK, Bala DN, Umoh UF. Ethno medicinal survey of plants used by the indigenes of Rivers State of Nigeria. Pharm Biol. 2012;50(9):1123–43.

Tharrun Daniel Paul L, Roshan Kumar Sah, Thirunavukkarasu Palaniyandi. Molecular docking studies of pancreatic cancer expressed proteins with Psidium guajava (guava) derived bioactive compounds. Int J Res Pharm Sci. 2020;11(SPL4):3176–83.

Adetutu A, Olaniyi TD, Owoade OA. GC-MS analysis and in silico assessment of constituents of Psidium guajava leaf extract against DNA gyrase of Salmonella enterica serovar Typhi. Informatics Med Unlocked. 2021 Jan 1;26:1–9.

Shady NH, Abdullah HS, Maher SA, Albohy A, Elrehany MA, Mokhtar FA, et al. Antiulcer Potential of Psidium guajava Seed Extract Supported by Metabolic Profiling and Molecular Docking. Antioxidants. 2022;11(7):1–14.

Olaniyi TD, Adetutu A. In silico anti-quorum sensing activities of phytocompounds of Psidium guajava in Salmonella enterica serovar Typhi. J Umm Al-Qura Univ Appl Sci [Internet]. 2023;9(2):142–56. Available from: https://doi.org/10.1007/s43994-023-00029-6

Habtemariam S. The chemical and pharmacological basis of guava (Psidium guajava L.) as potential therapy for type 2 diabetes and associated diseases. Med Foods as Potential Ther Type-2 Diabetes Assoc Dis. 2019 Jan 1;251–305.

Tella T, Masola B, Mukaratirwa S. Anti-diabetic potential of Psidium guajava leaf in streptozotocin induced diabetic rats. Phytomedicine Plus [Internet]. 2022;2(2):100254. Available from: https://doi.org/10.1016/j.phyplu.2022.100254

Yatoo MI, Saxena A, Gopalakrishnan A, Alagawany M, Dhama K. Promising antidiabetic drugs, medicinal plants and herbs: An update. Int J Pharmacol. 2017;13(7):732–45.

Baliga MS, Palatty PL, Adnan M, Naik TS, Kamble PS, George T, et al. Anti-Diabetic Effects of Leaves of Trigonella foenum-graecum L. (Fenugreek): Leads from Preclinical Studies. J Food Chem Nanotechnol. 2017;03(02):67–71.

Sadasivan Nair AN, Raveendran Nair RV, Rajendran Nair AP, Nair AS, Thyagarajan S, Johnson AJ, et al. Antidiabetes constituents, cycloartenol and 24-methylenecycloartanol, from ficus krishnae. PLoS One [Internet]. 2020;15(6):1–21. Available from: http://dx.doi.org/10.1371/journal.pone.0235221

Alam S, Sarker MMR, Sultana TN, Chowdhury MNR, Rashid MA, Chaity NI, et al. Antidiabetic Phytochemicals From Medicinal Plants: Prospective Candidates for New Drug Discovery and Development. Front Endocrinol (Lausanne). 2022;13(February).

Okoh OS, Yakubu AH, Adegboyega AE, Uti DE, Obeten UN, Agada SA, et al. Identification of some bioactive compounds from Trignonella foenumgraecumas possible inhibitors of PPARγ for diabetes treatment through molecular docking studies, pharmacophore modelling and ADMET profiling: An in-silico study. PLoS One [Internet]. 2023;18(5 May):1–15. Available from: http://dx.doi.org/10.1371/journal.pone.0284210

Joshi DD, Deb L, Somkuwar BG, Rana VS. Relevance of Indian traditional tisanes in the management of type 2 diabetes mellitus: A review. Saudi Pharm J [Internet]. 2023;31(5):626–38. Available from: https://doi.org/10.1016/j.jsps.2023.03.003

Kumar V, Sharma N, Sourirajan A, Khosla PK, Dev K. Docking studies in targeting proteins involved in cardiovascular disorders using phytocompounds from Terminalia arjuna. bioRxiv [Internet]. 2020;173229:2020.06.22.164129. Available from: https://www.biorxiv.org/content/10.1101/2020.06.22.164129v1%0Ahttps://www.biorxiv.org/content/10.1101/2020.06.22.164129v1.abstract

Abdellatiif MH, Ali A, Ali A, Hussien MA. Computational studies by molecular docking of some antiviral drugs with COVID-19 receptors are an approach to medication for COVID-19. Open Chem. 2021;19(1):245–64.

Souza HCA, Souza MDA, Sousa CS, Viana EKA, Alves SKS, Marques AO, et al. Molecular Docking and ADME-TOX Profiling of Moringa oleifera Constituents against SARS-CoV-2. Adv Respir Med. 2023;91(6):464–85.

Mendie LE, Hemalatha S. Molecular Docking of Phytochemicals Targeting GFRs as Therapeutic Sites for Cancer: an In Silico Study. Appl Biochem Biotechnol. 2022 Jan 1;194(1):215–31.

Protein Data Bank [Internet]. Available from: https://www.rcsb.org/

Morris GM, Ruth H, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, et al. Software news and updates AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J Comput Chem. 2009;30(16):2785–91.

PubChem [Internet]. [cited 2023 Nov 24]. Available from: https://pubchem.ncbi.nlm.nih.gov/

BIOVIA, Dassault Systèmes, Discovery Studio , 2021, San Diego: Dassault Systèmes, 2021. - Google Search [Internet]. Available from: https://discover.3ds.com/discovery-studio-visualizer

Trott O, Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem [Internet]. 2009;31(2). Available from: https://vina.scripps.edu/downloads/

Morris GM, Goodsell DS, Halliday RS, Huey R, Hart WE, Belew RK, et al. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J Comput Chem. 1998;19(14):1639–62.

Molinspiration Cheminformatics Publications [Internet]. [cited 2023 Nov 23]. Available from: https://www.molinspiration.com/cgi-bin/properties

SwissADME [Internet]. [cited 2023 Nov 24]. Available from: http://www.swissadme.ch/

ADMETlab 2.0 [Internet]. [cited 2023 Nov 24]. Available from: https://admetmesh.scbdd.com/

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 [Internet]. 2012;64(SUPPL.):4–17. Available from: http://dx.doi.org/10.1016/j.addr.2012.09.019

Tomlinson JW, Walker EA, Bujalska IJ, Draper N, Lavery GG, Cooper MS, et al. 11β-Hydroxysteroid dehydrogenase type 1: A tissue-specific regulator of glucocorticoid response. Endocr Rev. 2004;25(5):831–66.

Morton NM, Paterson JM, Masuzaki H, Holmes MC, Staels B, Fievet C, et al. Novel Adipose Tissue – Mediated Resistance to Dehydrogenase Type 1 – Deficient Mice. Mol Med [Internet]. 2004;53:931–8. Available from: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=15047607

Morgan SA, Sherlock M, Gathercole LL, Lavery GG, Lenaghan C, Bujalska IJ, et al. 11Β-Hydroxysteroid Dehydrogenase Type 1 Regulates Glucocorticoid- Induced Insulin Resistance in Skeletal Muscle. Diabetes. 2009;58(11):2506–15.

Hollis G, Huber R. 11β-Hydroxysteroid dehydrogenase type 1 inhibition in type 2 diabetes mellitus. Diabetes, Obes Metab. 2011;13(1):1–6.

Alberts P, Nilsson C, Selén G, Engblom LOM, Edling NHM, Norling S, et al. Selective Inhibition of 11β-Hydroxysteroid Dehydrogenase Type 1 Improves Hepatic Insulin Sensitivity in Hyperglycemic Mice Strains. Endocrinology. 2003;144(11):4755–62.

Böhme T, Engel CK, Farjot G, Güssregen S, Haack T, Tschank G, et al. 1,1-Dioxo-5,6-dihydro-[4,1,2]oxathiazines, a novel class of 11ß-HSD1 inhibitors for the treatment of diabetes. Bioorganic Med Chem Lett [Internet]. 2013;23(16):4685–91. Available from: http://dx.doi.org/10.1016/j.bmcl.2013.05.102

Ryu JH, Kim S, Lee JA, Han HY, Son HJ, Lee HJ, et al. Synthesis and optimization of picolinamide derivatives as a novel class of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors. Bioorganic Med Chem Lett [Internet]. 2015;25(8):1679–83. Available from: http://dx.doi.org/10.1016/j.bmcl.2015.03.003

Koike T, Shiraki R, Sasuga D, Hosaka M, Kawano T, Fukudome H, et al. Discovery and biological evaluation of potent and orally active human 11β-hydroxysteroid dehydrogenase type 1 inhibitors for the treatment of type 2 diabetes mellitus. Chem Pharm Bull. 2019;67(8):824–38.

Adasme MF, Linnemann KL, Bolz SN, Kaiser F, Salentin S, Haupt VJ, et al. PLIP 2021: Expanding the scope of the protein-ligand interaction profiler to DNA and RNA. Nucleic Acids Res. 2021;49(W1):W530–4.

Nakaishi Y, Bando M, Shimizu H, Watanabe K, Goto F, Tsuge H, et al. Structural analysis of human glutamine:fructose-6-phosphate amidotransferase, a key regulator in type 2 diabetes. FEBS Lett [Internet]. 2009;583(1):163–7. Available from: http://dx.doi.org/10.1016/j.febslet.2008.11.041

Shanak S, Saad B, Zaid H, Carvalho JCT. Metabolic and Epigenetic Action Mechanisms of Antidiabetic Medicinal Plants. Evidence-based Complement Altern Med. 2019;2019:1–19.

Belete TM. A recent achievement in the discovery and development of novel targets for the treatment of type-2 diabetes mellitus. J Exp Pharmacol. 2020;12:1–15.

Lindsley JE, Rutter J. Nutrient sensing and metabolic decisions. Comp Biochem Physiol - B Biochem Mol Biol. 2004;139(4):543–59.

Zhang H, Jia Y, Cooper JJ, Hale T, Zhang Z, Elbein SC. Common Variants in Glutamine:Fructose-6-Phosphate Amidotransferase 2 (GFPT2) Gene Are Associated with Type 2 Diabetes, Diabetic Nephropathy, and Increased GFPT2 mRNA Levels. J Clin Endocrinol Metab. 2004;89(2):748–55.

Buse MG. Hexosamines, insulin resistance, and the complications of diabetes: Current status. Am J Physiol - Endocrinol Metab. 2006;290(1):1-8.

Pan PW, Feldman JL, Devries MK, Dong A, Edwards AM, Denu JM. Structure and biochemical functions of SIRT6. J Biol Chem. 2011;286(16):14575–87.

Kugel S, Mostoslavsky R. Chromatin and beyond: the multitasking roles for SIRT6. Trends Biochem Sci. 2015;39(1):72–81.

Gertman O, Omer D, Hendler A, Stein D, Onn L, Khukhin Y, et al. Directed evolution of SIRT6 for improved deacylation and glucose homeostasis maintenance. Sci Rep [Internet]. 2018;8(1):1–11. Available from: http://dx.doi.org/10.1038/s41598-018-21887-9

Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004;73:417–35.

El-Kabbani O, Ruiz F, Darmanin C, Chung RPT. Aldose reductase structures: Implications for mechanism and inhibition. Cell Mol Life Sci. 2004;61(7–8):750–62.

Kaul CL, Ramarao P. The role of aldose reductase inhibitors in diabetic complications: Recent trends. Methods Find Exp Clin Pharmacol. 2001;23(8):465–75.

Downloads

Published

30-03-2024

Issue

Section

Research Article

How to Cite

“In Silico Characterization of a Novel Bioactive Compound Derived from Psidium Guajava 4-[5-(Pyridin-4-Yl)-1,2,4-Oxadiazol-3-Yl]-1,2,5-Oxadiazol-3-Amine: A Potential Inhibitor for Targeting Signaling Proteins Involved in Diabetes Development”. International Journal of Pharmaceutical Sciences and Drug Research, vol. 16, no. 2, Mar. 2024, pp. 180-9, https://doi.org/10.25004/IJPSDR.2024.160207.