Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Identification of some bioactive compounds from Trignonella foenumgraecum as possible inhibitors of PPARϒ for diabetes treatment through molecular docking studies, pharmacophore modelling and ADMET profiling: An in-silico study

Abstract

Oral antidiabetic agents including the peroxisome proliferator-activated receptor gamma (PPARγ) agonists are available for the clinical management of diabetes mellitus (DM) but most are characterized by many adverse effects. In this study, we explore the antidiabetic properties of phytoconstituents from Trigonellafeonumgraecum (Fabaceae) as potential agonist of PPARγ; using in silico molecular docking, molecular mechanics generalized surface area (MM/GBSA)free binding energy prediction, Pharmacophore modeling experiment, and Pharmacokinetic/ toxicity analysis. One hundred and forty (140) compounds derived from Trigonellafeonumgraecum were screened by molecular docking against protein target PDB 3VI8. Results obtained from binding affinity (BA) and that of binding free energy (BFE) revealed five 5 compounds; arachidonic acid (CID_10467, BA -10.029, BFE -58.9), isoquercetin (CID_5280804, BA -9.507kcal/mol, BFE -56.33), rutin (CID_5280805, BA -9.463kcal/mol, BFE -56.33), quercetin (CID_10121947, BA -11.945kcal/mol, BFE -45.89) and (2S)-2-[[4-methoxy-3-[(pyrene-1-carbonylamino)methyl]phenyl]methyl]butanoic acid (CID_25112371, BA -10.679kcal/mol, BFE -45.73); and were superior to the standard; Rosiglitazone with a docking score of -7.672. Hydrogen bonding was notable in the protein-ligand complex interaction, with hydrophobic bond, polar bond and pipi stacking also observed. Their Pharmacokinetic/ toxicity profile showed varying druggable characteristics, but; arachidonic acid had the most favorable characteristics. These compounds are potential agonists of PPARγ and are considered as antidiabetic agents after successful experimental validation.

1. Introduction

Diabetes mellitus (DM) is a chronic metabolic disorder characterized by glucose tolerance, hyperglycemia and insulin resistance, with features such as high fasting and post-prandial blood glucose concentration resulting from defective insulin secretion, insulin action, or both [1]. Around 463 million adults are currently living with diabetes, and this may increase to 578 million and 700 million by 2030 and 2045 respectively [2, 3]. It is ranked among the top ten (10) leading causes of death worldwide [4]. There are three types of diabetes: Type I Diabetes (Insulin Dependent Diabetes Mellitus), Type II Diabetes (Non-Insulin Dependent Diabetes Mellitus) and Gestational diabetes. The most common form of diabetes is Type II diabetes, constituting about 90–95% of the diabetic population [5]. It is characterized by insulin resistance, thereby resulting in hyperglycemia. Hyperglycemia, in turn, leads to glucotoxicity and long-term vascular (including peripheral and coronary artery, and cerebrovascular ailments; retinopathy, neuropathy, and nephropathy) and non-vascular complications (including infections, gastroparesis, and skin changes) of diabetes mellitus [1].

The universally accepted strategy in the management of diabetes is the administration of insulin jabs and antidiabetic prescriptions containing oral hypoglycemic drugs such as insulin secretagogues, incretin agonists, insulin sensitizers, dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors to enhance a controlled blood glucose level [6, 7]. However, the accessibility and affordability of these; coupled with their adverse effects, such as hypoglycemia, gastrointestinal upset, and lactic acidosis, have been reported in patients on antidiabetic drug therapy, and thus, pose a risk in the management of diabetes [8]. These contribute to the immense usage of herbs and plants that have distinct array of phytochemicals; paving more interest in the administration of medicinal plants with their varying pharmacological and biological activities to manage and treat diabetes [9]. One of such antidiabetic plants widely sought after is Fenugreek (Trigonellafoenum-graecum Linn).

Trigonellafoenum-graecum Linn., also known as Methi in various Indian languages and Fenugreek in English, is an annual and dicotyledonous plant belonging to the Fabaceae family [10]. Compounds contained in Fenugreek include but are not limited to: trigonelline, 4-hydroxy isoleucine, galactomannans, quercetin, kaempferol, luteolin, vitexin, protodioscin, diosgenin, sotolone, calycosin, tricin, apigenin, yamogenine, gentianine, scopoletin, coumarin, and tigogenin [1113]. The antioxidant, anti-inflammatory, hepatoprotective, anti-microbial, anticancer, immunomodulatory, gastroprotective, hypocholesterolemic, and neuroprotective effects of Fenugreek have been reported [14, 15]. The antidiabetic effect of Fenugreek has also been demonstrated [5, 1621]. With the aid of in silicovirtual screening methods, the antidiabetic mechanism of compounds isolated from medicinal plants can be ascertained against multiple diabetes targets rapidly and cost-effectively. Examples of such targets include α-amylase, α-glucosidase, glycogen synthase kinase-3β (GSK-3β), dipeptidyl peptidase-4 (DPP-4), protein tyrosine phosphatase 1B (PTP1B), glucokinase, and peroxisome proliferator-activated receptors (PPARs).

PPARs are a group of nuclear receptors which regulate gene transcription by binding to specific DNA response elements together with the retinoid-X receptor (RXR) as heterodimers [19]. Upon ligand activation, they control the expression of genes related to lipid and glucose homeostasis [17, 22, 23], thereby serving as excellent targets for the treatment of diabetes. They exist in three subtypes: α, δ, and γ. Peroxisome proliferator-activated receptor gamma (PPARγ) consists of two isoforms; PPARγ-1 and PPARγ-2. PPARγ-1 is majorly expressed in the gut, while PPARγ-2 is abundant in the adipose tissue where it is involved in adipocyte proliferation and differentiation. PPARγ agonist like the thiazolidinediones (TZD) exert their action by fully stabilizing the AF2 (activation function 2) of the ligand-binding domain (LBD), in its active conformation, thereby activating the PPARγ receptor, which in turn improves insulin sensitivity [22, 24, 25].

The present study screened and evaluated phytoconstituents obtained from Trigonellafoenum-graecum as a potential agonist of PPARγ for the treatment of diabetes mellitus via an insilicocomputational approach.

2. Materials and methods

2.1 Protein preparation

The crystal structure of peroxisome proliferator-activated receptor gamma (PPARγ) (PDB ID: 3VI8) was retrieved from Protein Data Bank (PDB) repository. The protein was prepared using the protein preparation wizard panel of Glide (Schrödinger Suite 2020–3) [2628] where bond orders were assigned, hydrogen added, disulfide bonds created, while missing side chains and loops were filled using prime. Water molecules beyond 3.0 Å of the heteroatoms were removed and the structure was minimized using OPLS2005 and optimized using PROPKA [29, 30]. Subsequently, the receptor grid file was generated to define the binding pocket for the ligands.

2.2 Ligand preparation

One hundred and forty (140) compounds from Trigonellafeonumgraecum, obtained by gas chromatography mass spectrometry (GCMS) analysis as earlier described [31] were prepared for molecular docking using Ligprep module (Schrödinger Suite 2020–3) [32]. Low-energy 3D structures with correct chiralities were generated. The possible ionization states for each ligand structure were generated at a physiological pH of 7.2 ± 0.2. Stereoisomers of each ligand were computed by retaining specified chiralities while others were varied.

2.3 Receptor grid generation

Receptor grid generation allows defining the position and size of the protein’s active site for ligand docking. The scoring grid was defined based on the co-crystalized ligand (2S)-2-(4-methoxy-3-{[(pyren-1-ylcarbonyl)amino]methyl}benzyl)butanoic acid with Unique ID 13M; Chenusing the receptor grid generation tool of Schrödinger Maestro 12.5. the van der Waals (vdW) radius scaling factor of nonpolar receptor atoms were scaled at 1.0, with a partial charge cut off of 0.25.

2.4 Protein-ligand docking

Glide [2628] tool of Schrödinger Maestro 12.5 [33] was used to perform the molecular docking studies using the generated receptor grid file. The prepared ligands were docked using standard precision (SP), setting ligand sampling to flexible, with the ligand sampling set to none (refine only). The vdW radius scaling factor was scaled at 0.80 with a partial charge cut-off of 0.15 for ligand atoms.

2.5 Receptor-ligand complex pharmacophore modelling

The first three compounds ranked with highest binding affinity against the target protein was used to develop a receptor-ligand complex pharmacophore model using PHASE [34, 35]. Auto (E-pharmacophore) method was used, hypothesis was set with maximum number of features to be generated at 7, minimum feature-feature distance at 2.00, minimum feature-feature distance for feature of the same type at 4.00 and donors as vectors.

2.6 Pharmacology parameters

The absorption, distribution, metabolism, excretion and toxicity (ADMET) properties of the test compounds were determined using in silico integrative model predictions at the SwissADME and PROTOX-II server respectively.

3. Results

3.1 Virtual screening of compounds

About one hundred and forty (140) compounds derived from Trignonella foenum graecum (Table 1) were screened against 3VI8, in which five (5) top posed compounds exhibited the highest binding affinities against the protein target and were selected for post docking analysis. From the multiple screening analysis, the following compounds arachidonic acid (CID_10467), isoquercetin (CID_5280804), rutin (CID_5280805), quercetin (CID_10121947) and (2S)-2-[[4-methoxy-3-[(pyrene-1-carbonylamino)methyl]phenyl]methyl]butanoic acid (CID_25112371) showed the lowest docking scores (highest binding affinity scores) of -10.029, -9.507, -9.463, -11.945 and -10.679 respectively against PDB 3VI8. This virtual screening based on binding energy gave us a vivid idea of the best ligands having the highest affinity for PDB 3VI8.

thumbnail
Table 1. Dockingscores of screened compounds derived from Trignonella foenum graecum against Peroxisome proliferator-activated receptor-γ.

https://doi.org/10.1371/journal.pone.0284210.t001

3.2 Molecular mechanics generalized surface area (MM/GBSA)

The Molecular mechanics generalized surface area (MM/GBSA) has been more accurate in binding free energy calculation studies and was used in determining the binding free energies of these compounds after virtual screening (multi-ligands docking). These best five (5) posed compounds exhibited lowest binding free energy of; -58.9 (CID_10467), -56.33 (CID_5280804), -56.33 (CID_5280805), -45.89 (CID_10121947) and-45.73 (CID_25112371) respectively with PDB 3VI8 (Table 2).

thumbnail
Table 2. Molecular mechanics generalized surface area (MM/GBSA) binding free energies of screened compounds derived from Trignonella foenum graecum against Peroxisome proliferator-activatedreceptor-γ.

https://doi.org/10.1371/journal.pone.0284210.t002

3.3 MM/GBSA dG binding energy versus docking scores

A plot of MM/GBSA dG binding energy versus docking scores is shown in Fig 1. The graph exhibited a strong correlation between MM/GBSA dG binding energy and docking scores of the binding affinities earlier observed in the multiple docking/virtual screening analysis. Lower binding free energies of top posed compounds.

3.4 Molecular interactions (2D and 3D) analysis

Molecular interaction of the post docking studies of the ligands CID_10467, CID_5280804, CID_5280805, CID_10121947, and CID_25112371 with 3VI8 displayed well defined binding pockets constituted of residues as indicated by their 2D and 3D configuration. Threonine-314, Serine-280, Glutamine-277, Histidine-440, and Tyrosine-464 in CID_10467 ligands exhibiting hydrogen bonding to PDB 3VI8. There were also other bonds such as hydrophobic, polar, and π- π stacking present in these interactions, emphasizing the cause of the observed low binding energy (high binding affinity) between CID_10467 and PDB 3VI8 (Fig 2A and 2B). More so, the ligand CID_25112371 showed hydrogen bonding linked to the residues Histidine-440, tyrosine-464, Serine-280, and Tyrosine-314 alongside other bonds interactions with PDB 3VI8 (Fig 3A and 3B). In the ligand CID_10121947, a hydrogen bond was observed in the ligands Asparagine-219, Methionine-220, Threonine-283, and Glutamate-286 also with other bonds interactions (Fig 4A and 4B). Similarly, the ligands CID_5280804 and CID_5280805 also showed hydrogen bonds between residues Threonine-283, Alanine-333, and Tyrosine-334, Threonine-279, Asparagine-219, and Methionine-355 respectively alongside other bonds interactions with the protein target PDB 3VI8 (Figs 5A and 5B and 6A and 6B). Overall, these bonding interactions corroborated with the observed binding affinities and binding free energies between the ligands and the protein target.

thumbnail
Fig 2.

a: 2D configuration of arachidic acid complex with peroxisome proliferator-activated receptor gamma, b: 3D configuration of arachidic acid complex with peroxisome proliferator-activated receptor gamma, c: Parmacophore model of arachidic acid complex with peroxisome proliferator-activated receptor gamma.

https://doi.org/10.1371/journal.pone.0284210.g002

thumbnail
Fig 3.

a: 2D configuration of isoquercitrin complex with peroxisome proliferator-activated receptor gamma, b: 3D configuration of isoquercitrin complex with peroxisome proliferator-activated receptor gamma, c: Parmacophore model of isoquercitrin indicating pharmacophoric sites in the ligand.

https://doi.org/10.1371/journal.pone.0284210.g003

thumbnail
Fig 4.

a: 2D configuration Rutin complex with peroxisome proliferator-activated receptor gamma, b: 3D configuration Rutin complex with peroxisome proliferator-activated receptor gamma, c: Parmacophore model of rutin complexes with peroxisome proliferator-activated receptor gamma.

https://doi.org/10.1371/journal.pone.0284210.g004

thumbnail
Fig 5.

a: 2D configuration of Quercetin complex with peroxisome proliferator-activated receptor gamma, b: 3D configuration of Quercetin complex with peroxisome proliferator-activated receptor gamma, c: Parmacophore model of quercetin complex with peroxisome proliferator-activated receptor gamma.

https://doi.org/10.1371/journal.pone.0284210.g005

thumbnail
Fig 6.

a: 2D configuration of (2s)-2-(4-Methoxy-3-{[(Pyren-1-Ylcarbonyl)amino]methyl}benzyl)butanoic Acid complex with peroxisome proliferator-activated receptor gamma, b: 3D configuration of (2s)-2-(4-Methoxy-3-{[(Pyren-1-Ylcarbonyl)amino]methyl}benzyl)butanoic Acid complex with peroxisome proliferator-activated receptor gamma, c: Parmacophore model of (2s)-2-(4-Methoxy-3-{[(Pyren-1-Ylcarbonyl)amino]methyl}benzyl)butanoic Acid complexes with peroxisome proliferator-activated receptor gamma.

https://doi.org/10.1371/journal.pone.0284210.g006

3.5 Development of pharmacophore hypothesis for the top posed ligands

The top-five pharmacophore hypotheses were generated using these set ligands (Table 3). These hypotheses could be classified into three (3) groups according to the features in the pharmacophore model ADDRR-10121947, ADDRR-5280804, AADDDRR-5280805 (01 and 04 and 05), DHNRRR-25112371 (02), and HHHHN-10467 (03) (Figs 2C, 3C, 4C, 5C and 6C). The set hypotheses were classified by their locations and the direction of their features.

thumbnail
Table 3. Summary of the pharmacophore models for PPAR-γ agonists.

https://doi.org/10.1371/journal.pone.0284210.t003

3.6 Absorption Distribution Metabolism Excretion & Toxicity (ADMET)/Pharmacokinetics predictions analysis of top posed compounds

The analysis of three lead compounds using the Lipinski rule of five, Ghose, and Verber rule is presented in Table 4 for its drug-likeness, while the absorption and metabolism profiles of these compounds are presented in Tables 5 and 6 with the toxicity profile given in Table 7. There were different degrees of violation of the three (3) rules for drug-like properties as indicated especially on the H-bond donors, H-bond Acceptors, and molecular weight. It was only CID_25112371 that showed high human gastrointestinal tract absorption and bioavailability with CID_10121947 and CID_10467 exhibiting low GIT absorption profiles. These ligands also exhibited varying degrees of cytochrome–P variants inhibition profiles with also different toxicological properties towards certain target organs and features.

thumbnail
Table 4. Drug-likeness profile of leads compounds from Trignonella foenum graecum.

https://doi.org/10.1371/journal.pone.0284210.t004

thumbnail
Table 5. Absorption profile of leads compounds from Trignonella foenum graecum.

https://doi.org/10.1371/journal.pone.0284210.t005

thumbnail
Table 6. Metabolism profile of leads compounds from Trignonella foenum graecum.

https://doi.org/10.1371/journal.pone.0284210.t006

thumbnail
Table 7. Toxicity profile of leads compounds from Trignonella foenum graecum.

https://doi.org/10.1371/journal.pone.0284210.t007

4. Discussion

The search for more effective and safer therapeutic alternatives is a noble task. This is necessary to reduce DM-related complications, morbidity, and mortality resulting from multisystem defects. Natural products provide an inexhaustible pool of pharmacologically active compounds with therapeutic potentials in DM treatment. Scientific screening of the vast number of natural compounds against important molecular targets for DM is an enormous task going by the conventional methods of drug development. Recent advances such as the in-silico molecular docking and ADME(T) techniques provide an invaluable tool for time- and resource-efficient screening of potential therapeutic compounds.

In the present study, a total of one hundred and forty (140) compounds derived from Trigonellafeonumgraecum were screened by molecular docking against peroxisome proliferator-activated receptor gamma (PPARϒ) for potential PPARϒagonistic action. Out of the 140 screened compounds, 5 (namely; arachidonic acid, quercetin, isoquercetin, rutin, and (2S)-2-[[4-methoxy-3-[(pyrene-1-carbonylamino) methyl]phenyl]methyl]butanoic acid) emerged as top pose compounds, whilst 3 compounds (arachidonic acid, quercetin and (2S)-2-[[4-methoxy-3-[(pyrene-1-carbonylamino) methyl]phenyl]methyl]butanoic acid) showed the most drug-like characteristics and overall; arachidonic acid had the most favorable characteristics. Some molecular docking studies reported Trigonelline and Diosgenin as top pose compounds of Trigonellafoecumgraecum with the strongest interactions against PPARϒ [5, 17, 19]. The reports showed that Trigonelline had a moderate affinity for the target while Diosgenin was better [5, 17, 19, 21]. The present study however revealed that arachidonic acid, 2S]… butanoic acid and quercetin are the ligands with superior drug-like characteristics. Trigonelline only showed a moderate binding affinity (docking score of -6.135) whereas Diosgenin had no docking score in the present study. Arachidonic acid had a docking score of -10.029 which indicates a strong affinity for the target protein (PPARϒ). This binding affinity is superior to that of Rosiglitazone (a standard PPARϒ agonist) which showed a docking score of -7.672 in the present study. It’s also superior to Rosiglitazone’s docking score of -7.176 reported by Tharaheswari et al. [17]. In addition, arachidonic acid formed a strong bond with the target with binding free energy (BFE) of -58.9. The docking score and BFE value denote superior interaction and binding efficiency which suggest that arachidonic acid is potentially a superior PPARϒ agonist to Rosiglitazone. Earlier studies suggested that arachidonic acid is an endogenous agonist of the α-subunit of PPARs whereas polyunsaturated fatty acids (PUFA) and PGI2 are endogenous ligands of the ϒ-subunit [36, 37]. This study revealed that arachidonic acid is also a strong agonist of the ϒ-subunit. Agents that activate PPARϒ or enhance the expression of the PPARϒ genes have been associated with enhancement of small insulin-sensitive adipocytes, uptake and storage of fats by adipose tissues, reduction of lipotoxicity on pancreatic β cells and other insulin-sensitive cells, and increased insulin sensitivity of liver and muscle cells [3841]. The post docking analysis revealed hydrogen bond was (mainly) involved in ligands interaction with PPARϒ. Arachidonic acid interacted with the target amino acid residues of Thr 314, Ser 240, Glu 277, His 440, and Tyr 464. Threonine, Serine, and Tyrosine residues confer favorable flexibility to the ligand-protein complex due to their rotatable OH groups [42]. Additional bonds in the interaction included hydrophobic bond, polar bond, and pipi stacking. The detailed druggable characteristics of arachidonic acid ligand revealed in the ADMET analysis are shown in Table 4. Other notable characteristics include strong compliance to Lipinski, Ghose, and Verber’s rules, non-inhibition of important drug-metabolizing enzymes (CYP 2C19, CYP 2D6, CYP 3A4, and CYP 2C9), and low potential for organ toxicity (toxic class 4, Table 7). The post docking analyses also revealed that the other four (4) top pose compounds had characteristics similar to arachidonic acid (Tables 17). Their interaction with the target (PPARϒ) was also due to the hydrogen bond (mainly) with little contributions of other bonds. They showed a less efficient bond with the target compared to arachidonic acid, except for quercetin which is better only with a binding affinity (docking score -11.945). However, their docking scores were all superior to that of Rosiglitazone (-7.672). The docking scores were -11.945, -10.679, -9.507 and -9.467, and BFEs were -45.89, -45.73, -56.33 and -56.33 for quercetin, 2S]… butanoic acid, isoquercetin and rutin respectively (Tables 2 and 3). The ADMET properties of these compounds showed that they have variable oral absorptivity, could inhibit some CYP enzymes and interfere with other drug’s metabolism, and are potentially immunotoxic. Their other druggable features and ADMET properties are shown in the Tables 36.

5. Conclusion

Overall, the present study and post docking analyses revealed potential antidiabetic compounds not commonly reported in the screening of Trigonellafoenumgraecum. The compounds showed superior binding affinity and bond strength compared to the standard PPARϒ agonist (rosiglitazone) and are therefore potentially more efficacious. Their pure forms need to be synthesized and tested in invivo studies to validate their efficacy and safety in diabetics. These could emerge as novel therapeutic agents of choice in DM treatment.

Acknowledgments

JARIS Computation Biology Centre, Jos, Nigeria is highly appreciated for running the in silico analysis and for capacity building in in silico analysis.

References

  1. 1. Gwatidzo S. D. and Williams S. J., “Diabetes mellitus medication use and catastrophic healthcare expenditure among adults aged 50+ years in China and India: results from the WHO study on global AGEing and adult health (SAGE),” BMC Geriatr, vol. 17, no. 1, p. 14, 2017. pmid:28077072
  2. 2. International Diabetes Federation, Diabetes Atlas, 9th ed. Brussels, Belgium, 2019.
  3. 3. Gaonkar V. P. and Hullatti K., “Indian Traditional medicinal plants as a source of potent Anti-diabetic agents: A Review,” J. Diabetes Metab. Disord., 2020. pmid:33553046
  4. 4. World Health Organization, “The top 10 causes of deaths:,” Fact sheet 2020, 2020. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death.
  5. 5. Jeyam M., Priyadharsini K., Ravikumar P., and Shalini G., “Evaluating multi-target efficiency of phytocompounds against diabetes mellitus—an in silico approach,” BIOINFO Drug Targets, vol. 2, no. 1, pp. 24–28, 2014.
  6. 6. Lorenzati B., Zucco C., Miglietta S., Lamberti F., and Bruno G., “Oral hypoglycemic drugs: pathophysiological basis of their mechanism of action,” Pharm., vol. 3, no. 9, pp. 3005–3020, 2010, https://doi.org/10.3390/ph3093005.
  7. 7. American Diabetes Association, “Strategies for Improving Care,” Diabetes Care, vol. 39, no. Sppulement 1, p. S6, 2016, doi: 10.2337/dc16-S004.
  8. 8. Mooventhan A. and Nivethitha L., “A Narrative Review on Evidence-based Antidiabetic Effect of Fenugreek (Trigonella Foenum-Graecum),” Int J Nutr Pharmacol Neurol Dis., vol. 7, pp. 84–87, 2017, https://doi.org/10.4103/ijnpnd.ijnpnd.
  9. 9. Campbell-Tofte J. I., Mϕlgaard P., and Winther K., “Harnessing the potential clinical use of medicinal plants as antidiabetic agents,” Bot. Targets Ther., vol. 2, pp. 7–19, 2012.
  10. 10. Chaudhary S., Chaudhary P. S., Chikara S. K., Sharma M. C., and Iriti M., “Review on Fenugreek (Trigonella foenum-graecum L.) and its Important Secondary Metabolite Diosgenin,” Not Bot Horti Agrobo, vol. 46, no. 1, pp. 22–31, 2018, https://doi.org/10.15835/nbha46110996.
  11. 11. Nagulapalli Venkata K. C., Swaroop A., Bagchi D., and Bishayee A., “A small plant with big benefits: Fenugreek (Trigonella foenum-graecum Linn.) for disease prevention and health promotion,” Mol. Nutr. Food Res., vol. 61, no. 6, pp. 1–26, 2017. pmid:28266134
  12. 12. Mahbub J., Mou R. A., Sikta S. A., Rahman A., and Dash P. R., “Biological and medicinal significance of Trigonella foenum-graecum: A review,” Int. J. Life Sci. Rev., vol. 4, no. 2, pp. 15–26, 2018.
  13. 13. Banerjee S., Bhattacharjee P., Kar A., and Mukherjee P. K., “LC–MS / MS analysis and network pharmacology of Trigonella foenum- graecum–A plant from Ayurveda against hyperlipidemia and hyperglycemia with combination synergy,” Phytomedicine, vol. 60, no. 152944, 2019, [Online]. Available: https://doi.org/10.1016/j.phymed.2019.152944. pmid:31178235
  14. 14. Arunabha M. and Bhattacharjee C., “Trigonella foenum -graecum: A review on its traditional uses, phytochemistry and pharmacology,” Int. J. Adv. Sci. Res., vol. 5, no. 5, pp. 1–12, 2019, [Online]. Available: https://doi.org/10.7439/ijasr.v5i5.5217.
  15. 15. Almatroodi S. A., Almatroudi A., Alsahli M. A., and Rahmani A. H., “Fenugreek (Trigonella Foenum-Graecum) and its Active Compounds: A Review of its Effects on Human Health through Modulating Biological Activities,” Pharmacogn J., vol. 13, no. 3, pp. 813–821, 2021.
  16. 16. Kumar P., Kale R. K., and Aquer N. Z., “Antihyperglycemic and protective effects of Trigonella foenum graecum seed powder on biochemical alterations in alloxan diabetic rats,” Eur. Rev. Med. Pharmacol. Sci., vol. 16, no. 3, pp. 18–27, 2012. pmid:22957414
  17. 17. Tharaheswari M., Kumar J. S. P., Reddy N. J., Subhashree S., and Rani S. S., “Fenugreek Seed Extract Stabilize Plasma Lipid Levels in Type 2 Diabetes by Modulating PPARs and GLUT4 in Insulin Target Tissues,” Am. J. Phytomedicine Clin. Ther., vol. 2, no. 5, pp. 587–602, 2014.
  18. 18. Baliga M. S. et al., “Anti-Diabetic Effects of Leaves of Trigonella foenum- graecum L. (Fenugreek): Leads from Preclinical Studies,” J. Food Chem. Nanotechnol., vol. 3, no. 2, pp. 67–71, 2017.
  19. 19. Begum S., Fathima S., Priya S., Sundararajan R., and Hemalatha S., “Screening Indian Medicinal Plants to Control Diabetes–An In silico and In vitro Approach,” Gen. Med. (Los Angeles), vol. 5, p. 289, 2017, https://doi.org/10.4172/2327-5146.1000289.
  20. 20. Geberemeskel G. A., Debebe Y. G., and Nguse N. A., “Antidiabetic Effect of Fenugreek Seed Powder Solution (Trigonella foenum-graecum L.) on Hyperlipidemia in Diabetic Patients,” J Diabetes Res., vol. 8507453, 2019.
  21. 21. Mohammadi A., Gholamhosseinian A., and Fallah H., “Trigonella foenum-graecum water extract improves insulin sensitivity and stimulates PPAR and γ gene expression in high fructose-fed insulin-resistant rats,” Adv Biomed Res, vol. 5, p. 54, 2016.
  22. 22. Frkic R. L. et al., “PPARγ in Complex with an Antagonist and Inverse Agonist: a Tumble and Trap Mechanism of the Activation Helix,” iScience, vol. 5, pp. 69–79, 2018, https://doi.org/10.1016/j.isci.2018.06.012.
  23. 23. Chen T. H., Tsai M. J., Fu Y. S., and Weng C. F., “The Exploration of Natural Compounds for Anti-Diabetes from Distinctive Species Garcinia linii with Comprehensive Review of the Garcinia Family,” Biomolecules, vol. 9, no. 11, p. 641, 2019, https://doi.org/10.3390/biom9110641. pmid:31652794
  24. 24. Brusotti G. et al., “Betulinic acid is a PPARγ antagonist that improves glucose uptake, promotes osteogenesis and inhibits adipogenesis,” Sci. Rep., vol. 7, no. 1, p. 5777, 2017, https://doi.org/10.1038/s41598-017-05666-6.
  25. 25. Kume S., Uzu T., Isshiki K., and Koya D., “Peroxisome proliferator-activated receptors in diabetic nephropathy,” PPAR Res., vol. 879523, 2008, https://doi.org/10.1155/2008/879523. pmid:19277201
  26. 26. Halgren T. A. et al., “Glide: A New Approach for Rapid, Accurate Docking and Scoring. 2. Enrichment Factors in Database Screening,” J. Med. Chem., vol. 47, pp. 1750–1759, 2004. pmid:15027866
  27. 27. Friesner R. A. et al., “Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand Complexes,” J. Med. Chem., vol. 49, pp. 6177–6196, 2006. pmid:17034125
  28. 28. Friesner R. A. et al., “Glide: A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy,” J. Med. Chem., vol. 47, pp. 1739–1749, 2004. pmid:15027865
  29. 29. Chresten R. S., Ollson M. H. M., Rostkowski M., and Jensen J. H., “Improved Treatment of Ligands and Coupling Effects in Empirical Calculation and Rationalization of pKa Values,” J. Chem. Theory Comput., vol. 7, no. 7, pp. 2284–2295, 2011, pmid:26606496
  30. 30. Ollson M. H. M., Chresten R. S., Rostkowski M., and Jensen J. H., “PROPKA3: consistent treatment of internal and surface residues in empirical pKa predictions,” J. Chem. Theory Comput., vol. 7, no. 2, pp. 525–537, 2011, pmid:26596171
  31. 31. D.E. Uti, I.J. Atangwho, U.E. Eyong, G.U Umoru, Egbung G.E., Rotimi S.O., et al, (2020). African Walnuts (Tetracarpidium conophorum) Modulate Hepatic Lipid Accumulation in Obesity via Reciprocal Actions on HMG-CoA Reductase and Paraoxonase. Endocrine, Metabolic & Immune Disorders—Drug Targets EMIDDT: 20 (3) 365–379. pmid:31339080
  32. 32. Schrödinger Release, “LigPrep.” New York, 2021.
  33. 33. Schrödinger, “Schrödinger Release 2021–3: Maestro.” New York, 2021.
  34. 34. Dixon S. L., Smondyrev A. M., Knoll E. H., Rao S. N., Shaw D. E., and Friesner R. A., “PHASE: A New Engine for Pharmacophore Perception, 3D QSAR Model Development, and 3D Database Screening. 1. Methodology and Preliminary Results,” J. Comput. Aided Mol. Des., vol. 20, pp. 647–671, 2006.
  35. 35. Dixon S. L., Smondyrev A. M., and Rao S. N., “PHASE: A Novel Approach to Pharmacophore Modeling and 3D Database Searching,” Chem. Biol. Drug Des., vol. 67, pp. 370–272, 2006. pmid:16784462
  36. 36. Desvergne B. and Wahli W., “Peroxisome proliferator-activated receptors: nuclear control of metabolism,” Endocr Rev, vol. 20, pp. 649–688, 1999. pmid:10529898
  37. 37. Schoonians K., Staels B., and Auwerx J., “The peroxisome prolif- erator activated receptors (PPARs) and their effects on lipid metabolism and adipocyte differentiation,” Biochim. Biophys. Acta., vol. 1302, pp. 93–109, 1996. pmid:8695669
  38. 38. Kintscher U. and Law R. E., “PPAR gamma- mediated insulin sensitization: The importance of fat versus muscle,” Am J Physiol Endocrinol Metab., vol. 288, pp. 287–291, 2005. pmid:15637349
  39. 39. Yki-Jarvinen H., “Thiazolidinediones,” N Engl J Med, vol. 351, pp. 1106–1018, 2004. pmid:15356308
  40. 40. Rosen E. D. et al., “PPARc is required for the differentiation of adipose tissue in vivo and in vitro,” Mol Cell, vol. 4, pp. 611–617, 1999.
  41. 41. Berthiaume M. et al., “Actions of PPARgamma agonism on adipose tissue remodeling, insulin sensitivity and lipemia in absence of glucocorticoids,” Am J Physiol Regul Integr Comp Physiol, vol. 287, no. 5, pp. R1116 –R1123, 2004. pmid:15256367
  42. 42. Tatu P. and Antti P., “Binding Affinity via Docking: Fact andFiction,” Molecules, vol. 23, no. 8, p. 1899, 2018, pmid:30061498