Skip to main content
In Silico Pharmacology logoLink to In Silico Pharmacology
. 2021 Apr 3;9(1):25. doi: 10.1007/s40203-021-00084-z

Exploring the antidiabetic potential of compounds isolated from Anacardium occidentale using computational aproach: ligand-based virtual screening

Victor Okoliko Ukwenya 1,, Sunday Aderemi Adelakun 1, Olusola Olalekan Elekofehinti 2
PMCID: PMC8019409  PMID: 33868895

Abstract

Diabetes mellitus is becoming an important public health challenge worldwide and especially in developing nations. About 8.8 percent of the world adult population has been reported to have diabetes. Glutamine-fructose-6-phosphate amidotransferase 1 (GFAT1) catalyses the first committed step in the pathway for biosynthesis of hexosamines in mammals, and its inhibition has been thought to prevent hyperglycaemia. Dipeptidyl peptidase-4 (DPP-4), on the other hand, degrades hormone glucagon-like peptide-1 (GLP-1), an enzyme that plays a major role in the enhancement of glucose-dependent insulin secretion, making these two proteins candidate targets for diabetes. To find potential inhibitors of DPP-4 and GFAT1 from Anacardium occidentale using a computational approach, glide XP (extra precision) docking, Induced Fit Docking (IFD), Binding free energy of the compounds were determined against prepared crystal structure of DPP-4 and GFAT1 using the Maestro molecular interface of Schrödinger suites. The Lipinski's rule of five (RO5) and ADME properties of the compounds were assessed. Predictive models for both protein targets were built using AutoQSAR. This study identified 8 hit compounds. Most of these compounds passed the RO5 and were within the recommended range for defined ADME parameters. In addition, the predicted pIC50 for the hit compounds were promising. The results obtained from the present study can be used to design an antidiabetic drug.

Supplementary Information

The online version contains supplementary material available at 10.1007/s40203-021-00084-z.

Keywords: Diabetes mellitus, Insulin, Molecular docking studies, DPP-4, GFAT1

Introduction

Diabetes mellitus, also referred to as diabetes, is a group of metabolic disorders characterized by persistent hyperglycaemia. It is associated with long term complications such as retinopathy, neuropathy and angiopathy (Franks et al. 2010). Diabetes is due to either lack of or resistance to insulin action resulting in type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) respectively (Shoback and Gardner 2011).

Diabetes is globally the 7th leading cause of death. An estimated 463 million people worldwide had diabetes as of 2019 (8.8 percent of the adult population), with type 2 diabetes responsible for around 90 percent of the cases (Williams et al. 2019). In the same year, about 4.2 million deaths were caused by diabetes (Hu 2011). Diabetes mellitus is becoming an important public healthchallenge (Fakhri et al. 2016; Hassanzadeh et al. 2016; Hu 2011), and is a significant contributor to the large disparities in life expectancy in Nigeria and the world at large (Nguyen et al. 2016).

Presently, pharmacological therapies are focused on increasing the level of insulin in type I diabetes patients, improving insulin sensitivity, slowing carbohydrates delivery and absorption or increasing urinary glucose excretion (Jha and Bhadoriya 2018).

The gut hormone glucagon-like peptide-1 (GLP-1) has an antidiabetic role. It regulates glucose metabolism through several mechanisms, including enhancement of glucose-dependent insulin secretion, slowed gastric emptying, and reduction of postprandial glucagon and of dietary intake or calorie consumption (Ahrén 2005; Deacon et al. 2004; Koliaki and Doupis 2011). However, this important hormone is degraded by the enzyme dipeptidyl peptidase-4 (DPP-4) very fast. This makes DPP-4 an efficient approach for the management of diabetes.

DPP-4 is a type II integral membrane protein also referred to as adenosine deaminase complexing protein 2 or CD26 and consists of 766 amino acids in humans, positioned on the long arm of chromosome 2 (2q24.3) (Abbott et al. 1994; Misumi et al. 1992). While the intracellular N-terminal hydrophilic segment of the type II integral membrane protein consists of 6 amino acids, domain 22 amino acid has a hydrophobic membrane (Hong and Doyle 1990). Another part of the enzyme is the extracellular part containing 738 amino acid sequence. This protein is a protease and it has been observed that its catalytic sites found at the end of C-terminal form a small pocket for catalytic activity (Ludwig et al. 2003; Ogata et al. 1992).

The hexosamine pathway has been identified as one of the biochemical pathways that play an important role in hyperglycaemia and fat-induced insulin resistance (Hawkins et al. 1997; Marshall et al. 1991). In patients with diabetes, an elevated level of blood sugar is mainly metabolized to glucose-6 phosphate, fructose-6 phosphate, and subsequently to the rest of the glycolytic pathway end products. In addition to this, fructose-6-phosphate is also catalysed by the rate-limiting enzyme glutamine:fructose-6 phosphate amidotransferase (GFAT) into glucosamine-6-phosphate through the hexosamine pathway, which produces UDP-N-acetylglucosamine (UDP-GlcNAc), the end product of hexosamine biosynthesis pathway, a substrate for O-linked glycosylation of serine and threonine residues on a variety of cellular proteins, catalysed by O-GlcNActransferase. These series of events lead to induced transcription of Transforming growth factor alpha, Transforming growth factor beta 1 (Kolm-Litty et al. 1998), and Plasminogen activator inhibitor-1 (Du et al. 2000). Since Glutamine-fructose-6-phosphate amidotransferase (GFAT) catalyses the first committed step in the pathway for biosynthesis of hexosamines in mammals, its inhibition has been thought to prevent hyperglycaemia induced elevation in the transcription of TGF-α, TGF-β1 (Kolm-Litty et al. 1998), and PAI-1 (Du et al. 2000), leading to survival from several diabetic complications. Presently, three human isoforms of GFAT have been identified namely GFAT1, GFAT2, and GFAT1L (McKnight et al. 1992; Niimi et al. 2001; Oki et al. 1999). GFAT1 isoform, however, is the ideal target for regulation of obesity and diabetic conditions since it’s predominantly expressed in liver and fat.

Anacardium occidentale is a tropical plant found in different parts of the world principally in Northeastern Brazil (Bitu et al. 2015). The fruit of this plant, typically known as cashew, is used industrially to produce juice. Phytochemical constituents such as flavonoids and carotenoids represent an important content which can be related to significant biological activities which have been used as alternative medicines for primary health care (Kubo et al. 2011; Kulis et al. 2012).While anaesthetic, bactericidal, and insecticidal properties of phytochemicals derived from Anacardium occidentale have been reported by different authors (Castillo-Juárez et al. 2007; Olajide et al. 2004, 2013), its antidiabetic property has also been well established (Jaiswal et al. 2017; Olatunji et al. 2005; Palheta and Ferrira 2018; Ukwenya et al. 2012). Drugs are essential for the prevention and treatment of disease. Human life is constantly threatened by many diseases such as cancer and diabetes. Therefore, ideal drugs are always in great demand (Fuller et al. 2009). The processes involved in drug discovery are strenuous and challenging due to time consumption and the running cost (Mandal et al. 2009). In silico study in medicine is thought to increase the speed of drug discovery while reducing the need for expensive laboratory work and clinical trials. The success rate of potential inhibitors of drug target in in silico experiment has been proven to be promising in in vitro studies (Allegra et al. 2015; Guedes et al. 2014; Röhrig et al. 2010). Presently, Computer aided drug design (CADD) has been explored to develop small molecules as inhibitors/activators of different proteins implicated in the pathogenesis of diabetes mellitus (Elekofehinti et al. 2018; Kikiowo et al. 2020), and so far, close to none has been subjected to clinical trials due to a number of issues, among them is unacceptable efficacy of the molecules against protein target. In this study, phytoconstituents isolated from Anacardium occidentale as identified in different literatures were investigated for their antidiabetic properties against two of the prime targets implicated in diabetes. Some studies have shown that compounds from Anacardium occidentale such as kaempferol and quercetin posses hypoglycaemic properties in vivo and in vitro (Zhu et al. 2007). Hence the main motive for investigating Anacardium occidentale phytoconstituents therapeutic potential against GFAT and DPP-4 using an integrative computational approach.

Methodology

Preparation of ligand and protein target structure

The three-dimensional crystal structure of the GFAT1 (PDB ID: 2V4M) and DPP-4 (PDB ID: 3D4L) were downloaded from Protein Data Bank (Berman et al. 2000), and further prepared by protein preparation wizards which is available in Glide(2018c) as previously described by Iwaloye et al. (2020a). The protein preparation wizards panel have two components namely, pre-process and refinement tab. In the pre-process tab, create zero bonds to metals and create disulfide bonds options were selected, bond orders were assigned, waters were deleted from 5.0 Å of het groups, het states was set at pH 7.0 ± 2.0, and hydrogen was added. In the next step, water molecules were removed and hydrogen atoms were added to the structure. Minimization was performed until the average root mean square deviation of the non-hydrogen atoms reached 0.3 A˚. Consequently, a total of 71 phytoconstituents of Anacardium occidentale identified in literatures (Salehi et al. 2019; Bhakyaraj Singaravadive, 2012) were uploaded on the workspace of Maestro (2018a) and prepared for docking using Ligprep panel (2017). 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. While the ligands were minimized using an OPLS3 force field (Harder et al. 2016), other options were set to their default.

Molecular docking

Glide tool in maestro v11.8 (2018a) was employed to perform the molecular docking of the prepared ligands into the active site of the crystal protein structures. Glide utilizes a hierarchical series of filters to search for possible locations of the ligand in the active-site region of the receptor, while the shape and properties of the receptor are represented on a grid by several sets of fields that progressively provide more accurate scoring of the ligand poses. An extensive conformational search is performed using a heuristic screen that rapidly eliminates unsuitable conformations (e.g. long-range internal H-bonds or sterical clashes). The entire amount of poses generated is then hierarchically classified, refined and further minimized into the active site grid before being finally scored using the proprietary GlideScore function, defined as in Eq. (1).

GScore=0.065×vdW+0.130×Coul+Lipo+Hbond+Metal+BurryP+RotB+Site 1

The ligand docking panel used the grid file generated by picking the co-crystallized ligand at the active site of the protein which defines the area around the active site in terms of coordinates x, y and z. Extra precision (XP) was assigned as the scoring function. The XP scoring functions integrate the basic GlideScore functions with additional terms: to model solvation, an explicit water term was used, parameterized on several known protein–ligand complexes; similarly, additional parameterized rewards were calculated for buried hydrophobic pockets occupied by hydrophobic ligand groups.

Validation of molecular docking results

The docking protocol was further validated by docking native ligand (co-crystal ligand) with the prepared crystal structure of both GFAT1 and DPP-4 to validate the docking efficiency by determining the root mean square deviation (RMSD) were determined. An RMSD value of 1.62 Å and 0.935 Å for GFAT1 and DPP-4 (Fig. 1a, b), respectively showed the docking procedure is reproducible (Iwaloye et al. 2020b).

Fig. 1.

Fig. 1

Docking pose of co-crystallized ligands with its native form showed a RSMD value of 0.935 Å for DPP-4, and b RMSD value of 1.62 Å for GFAT1

Induced fit docking (flexible simulation)

Induced Fit Docking (IFD) is an in silico based method that uses Glide and the Refinement module in Prime to accurately predict ligand binding modes and concomitant structural changes in the receptor (Sherman et al. 2006). The conventional molecular docking procedure allows the binding of the flexible ligands into the catalytic site of a rigid receptor while in reality, a receptor freely rotates on binding to a ligand. IFD protocol has made the docking process more realistic than the conventional docking.

Free energy calculation using prime

Prime Molecular Mechanics/Generalized Born Surface Area (MM-GBSA), also known as a post-scoring approach, is a key and appealing computational method used to analyse the accuracy of docking processes. The docked poses with the lowest Glide score (best docking score) were minimized using MM-GBSA approach as implemented in the Prime module of the Schrödinger suite with default settings (Jacobson et al. 2002, 2004). The final output obtained from the above process is ΔGbind, a mixture of different kinds of energies in which polar and nonpolar energies contribute separately. ΔGbind was computed as:

ΔGbind=ΔEMM+ΔGSolv+ΔGSA

where ΔEMM is the difference in energy between the complex structure and the sum of the energies of the ligand and unliganded protein using the OPLS3 force field. ΔGsolv isthe difference in the GBSA solvation energy of the complex and the sum of the solvation energies for the ligand and unliganded protein. Lastly, ΔGSA is the difference in the surfacearea energy for the complex and the sum of the surface area energies for the ligand and uncomplexed protein.

ADME predictions

Absorption, Distribution, Metabolism, and Excretion analysis constitutes the pharmacokinetics and physicochemical properties of a drug molecule (Nisha et al. 2016). This study used Qikprop(2018b) to predict significant descriptors of drug-likeness such as Lipinski rule of 5 (RO5), binding to human serum albumin (QPlogKhsa), IC50 value for the blockage of HERG K + channels (QPlogHERG) and Human Oral Absorption.

Automated QSAR

AutoQSAR is a machine-learning algorithm provided by Schrödinger suite that builds and applies QSAR models through automation (Dixon et al. 2016). In order to build a predictive model, AutoQSAR takes the 1D, 2D and 3D structural data of a molecule along with a property (e.g. IC50) to be modeled, as an input. It will then compute the fingerprints and descriptors using machine-learning statistical methods for creating a predictive QSAR model. The experimental dataset containing 39 DPP-4 inhibitors with their respective IC50 was retrieved from CHEMBL database online server (www.ebi.ac.uk/chembl/), by blasting the FASTA sequence of the DPP-4. In the same vein, 19 known inhibitors of GFAT1 with their respective IC50 were retrieved from different literature reviews (Bobzin et al. 2000; Bolin et al. 2006; Floquet et al. 2007; Le Camus et al. 1998a, b; Qian et al. 2011). The compounds were uploaded into the workspace of Maestro and prepared by Ligprep. QSAR models were built using the structure of inhibitors with their respective IC50.

Results and discussion

Molecular docking analysis

The primary aim of docking study is to obtain accurate predictions of the ligand conformation and orientation within a targeted binding site (Guedes et al. 2014). Prepared compounds were refined using a two-step hierarchical process, which glide SP and XP are docking. The use of these hierarchical filters and associated parameters was earlier discussed in different studies (James and Ramanathan 2018; Rohini and Shanthi 2018; iwaloye et al. 2020c). The co-crystallized ligand of each protein target was used for the basis of comparison. Figure 2 showed the chemical structure of hit compounds against GFAT1 and DPP-4. These compounds were proved to be more active than other phyto-compounds derived from Anacardium occidentale. Docking score and Induced fit score of the hits compounds against GFAT1and DPP-4 are shown in Table S2 and S3. Kaemferol 3-O-Beta-d-Xyloside and myricetin recorded the highest binding affinity with GFAT1 with docking score of − 7.521 kcal/mol and − 7.440 kcal/mol, respectively. The results of glide XP docking score of hit compound with DPP-4, however, showed that delphinidin and myricetin recorded the highest affinity (− 9.332 kcal/mol and − 8.906 kcal/mol). Consequently, we further made use of Schrodinger’s innovative method known as induced fit docking for modeling conformational changes induced by ligand binding (Tables 1, 2, 3).

Fig. 2.

Fig. 2

Chemical structure of hit compounds

Table 1.

Induced fit docking and interacting residues of hit molecules and co-crystallized ligand GFAT

S/no. Pubchem ID Docking score Induced fit docking No of H− bond Interacting residues Predicted pIC50 ΔGBind
1 kaemferol 3-O-Beta-d-Xyloside − 7.521 − 748.09 4 ASP446, GLN440, THR394, GLU579 6.107 − 40.58
2 Myricetin − 7.440 − 748.41 3 ASP446, THR394, SER395 6.209 − 36.99
3 Quercetin-3-O-beta-d-arabinofuranoside − 7.140 − 752.41 7 THR444, ASP446, SER441, CYS392, VAL490, SER492 6.044 − 36.56
4 Delphinidin − 6.907 − 747.80 4 THR394, GLU579, GLU487 6.203 − 26.06
5 Gallic acid − 6.880 − 746.13 5 GLN440, SER395, SER441, VAL490 6.236 − 31.51
6 Quercetin-3-O-d-galactopyranoside − 6.628 − 749.46 7 ASP446, SER441, GLN440, SER395, THR394, GLU579 6.044 − 34.67
7 ( +)-Catechin − 6.117 − 747.80 3 SER395, GLU487, VAL490 6.025 − 42.60
8 Protocatechuic acid − 6.020 − 743.55 5 THR444, SER441, GLN440, SER439, THR394 6.371 − 24.69
9 Epigallocatechin − 5.982 − 749.75 8 THR394, SER395, GLN440, SER441, GLU579, SER492 5.997 − 45.73
10 Naringenin − 4.841 − 750.82 4 THR394, CYS392, SER492, GLU487 6.269 − 33.29
11 (−) epicatechin − 4.788 − 748.11 6 CYS392, SER395, GLN440, GLU487, GLY489 6.025 − 45.72
12 Co-crystallized ligand − 8.698 − 749.73 8 GLN440, SER395, VAL490, SER492, GLU579 − 5.031 − 41.80

The boldened amino acid residues represent pi-pi iteraction

Table 2.

Induced fit docking and interacting residues of hit molecules and co- crystallized ligand with DPP-4

S/no. Pubchem ID Docking score (3D4L) Induced fit docking No of H-bond Interacting residues Predicted pIC50 ΔGBind
1 kaemferol 3-O-Beta-d-Xyloside − 8.081 − 1335.01 4 PRO550, GLY549, TYR666, SER209 5.076 − 40.16
2 Myricetin − 8.906 − 1336.63 6 GLU206, VAL207, TYR547, TYR666, PRO550, PHE357 4.628 − 38.58
3 Quercetin-3-O-beta-d-arabinofuranoside − 8.857 − 1336.66 6 GLU206, GLU205, PHE357, TYR585, TYR547, TYR662 5.028 − 31.75
4 Delphinidin − 9.332 − 1335.39 6 GLU205, GLU206, ASN710, ARG125, TYR666, TYR547, TYR666 4.546 − 57.95
5 Gallic acid − 4.891 − 1322.10 5 TYR670, TYR666, TYR547 4.830 19.05
6 Quercetin-3-O-d-galactopyranoside − 8.416 − 1335.78 6 GLU206, ARG125, TYR547, PRO550, TYR662, PHE357 5.156 − 42.64
7 ( +)-Catechin − 6.448 − 1333.57 4 GLU205, GLU206, ARG122, TYR666 4.883 − 39.42
8 Protocatechuic acid − 4.386 − 1321.16 3 PRO550, TYR666, TYR547, TYR547 4.877 19.44
9 Epigallocatechin − 7.681 − 1335.56 6 GLU206, TYR670, PRO550, SER630, TYR662 4.779 − 16.01
10 Naringenin − 5.723 − 1334.81 4 GLU206, TYR670, GLN553, TYR585 4.989 − 30.88
11 (−) epicatechin − 7.204 − 1336.04 3 GLU205, GLU206, HIS740, TYR666 4.883 − 21.22
12 Co-crystallized ligand − 7.670 − 1335.63 6 GLU206, VAL207, ARG125, TYR662, TYR662, TYR666 − 4.102 − 40.46

Table 3.

Prediction of ADME properties by QIkprop

S/no. Pubchem ID RuleOfFive HOA QPlogKhsa QPlogHERG QPPMDCK PSA
1 kaemferol 3-O-Beta-d-Xyloside 1 2 − 0.606 − 5.427 4.151 177.65
2 Myricetin 1 2 − 0.488 − 5.008 2.149 164.858
3 Quercetin-3-O-beta-d-arabinofuranoside 2 1 − 0.746 − 5.265 1.171 199.057
4 Delphinidin
5 Gallic acid 0 2 − 0.985 − 1.413 4.294 115.079
6 Quercetin-3-O-d-galactopyranoside 2 1 − 0.895 − 5.375 0.953 217.198
7 ( +)-Catechin 0 2 − 0.415 − 4.774 19.439 116.961
8 Protocatechuic acid 0 2 − 0.903 − 1.493 12.821 93.683
9 Epigallocatechin 1 2 − 0.552 − 4.543 7.055 137.168
10 Naringenin 0 3 − 0.031 − 4.975 54.4 100.802
11 (−) epicatechin 0 3 − 0.035 − 4.963 55.288 100.26

aNumber of violations of Lipinski’s rule of five (Range: maximum is 4)

bHumanOralAbsorption: Predicted qualitative human oral absorption: 1, 2, or 3 for low, medium, or high

cQPlogKhsa: Prediction of binding to human serum albumin. Range from –1.5 to – 1.5

dQPlogHERG: Predicted IC50 value for blockage of HERG K + channels. concern below –5

eQPPMDCK: Predicted apparent MDCK cell permeability in nm/sec. MDCK cells are considered to be a

good mimic for the blood–brain barrier. QikProp predictions are for non-active transport. < 25 poor, > 500 great

fPSA Van der Waals surface area of polar nitrogen and oxygen atoms. Range from 7.0 – 200.0

This method offered more accuracy in the binding of a ligand with its protein target because it enables the receptor to change its binding pocket so that it conforms more closely to the ligand shape and binding mode (Venkatachalam et al. 2003). The results of the IFD score revealed that quercetin-3-O-beta-d-arabinofuranoside and naringenin bind better with GFAT1 than other hit compounds. Additionally, myricetin and quercetin-3-O-beta-d-arabinofuranoside showed a more favourable binding affinity with DPP-4 than other compounds, recording an IFD score of − 1336.66 kcal/mol and − 1336.63 kcal/mol respectively. The results of the molecular docking analysis clearly showed the inhibitory potential of the hit compounds against GFAT1 and DPP-4.

Pose orientation of hit compounds in the binding pocket of GFAT1 and DPP-4

It is important to investigate the interaction of proposed inhibitors with crucial amino acids residue located at the active site of the proteins, as this will provide evidence of their inhibitory properties. The Ligand Interaction panel of Maestro was used to visualize the 2D interaction of the compounds with the protein target. GFAT1 active sites are known to be conserved from bacteria to humans. The human GFAT1 isomerase domain consists of two sugar isomerase sub-domain: both are composed of five stranded parallel beta-sheets surrounded by two or three alpha-helices (Ruegenberg et al. 2020). An experiment by Ruegenberget al. demonstrated that residues of both isomerase domains contribute to the binding to inhibitors (2020). Here, the hits showed similarity in binding by interacting with hydrophobic, negatively charged and polar amino acid residues and 2D ligand interaction diagram of hit compounds are shown in Table S1 and Figs. 3, 4, 5.

Fig. 3.

Fig. 3

The 2D ligand interaction diagram of kaemferol 3-O-Beta-d-Xyloside with GFAT1

Fig. 4.

Fig. 4

The 2D ligand interaction diagram of Epigallocatechin with GFAT1

Fig. 5.

Fig. 5

The 2D ligand interaction diagram of Naringenin with GFAT1

Two binding sites are found in the human DPP-4 namely; S1 site and S2 site. S1 site is a long hydrophobic region that allows uncharged amino acids to fit in receptor and it has a catalytic triad Ser-630, Asn-710 and His-740. In contrast, the long lipophilic region called S2 site involves key interaction with Glu-205, Glu-206 dyad and Arg-125 (Chakraborty et al. 2014; Kuhn et al. 2007; Watanabe et al. 2015). The substrate-specific S1 site is made up of highly hydrophobic residues such as Tyr-547, Tyr- 631, Val-656, Trp-659, Tyr-662, Tyr-666, and Val-711 (Chakraborty et al. 2014). In our present study, three compounds (Quercetin-3-O-d-galactopyranoside, Quercetin-3-O-beta-d-arabinofuranoside and Quercetin-3-O-d-galactopyranoside) formed pi-pi stacking with the amino acid residue Phe-357, while ( +)-Catechin, kaemferol 3-O-Beta-d-Xyloside and Delphinidin also showed pi–pi interaction with Tyr-666 (Figs. 6, 7, 8).

Fig. 6.

Fig. 6

The 2D ligand interaction diagram of kaemferol 3-O-Beta-d-Xyloside with DPP-4

Fig. 7.

Fig. 7

The 2D ligand interaction diagram of Quercetin-3-O-d-galactopyranoside with DPP-4

Fig. 8.

Fig. 8

The 2D ligand interaction diagram of Quercetin-3-O-beta-d-arabinofuranoside with DPP-4

These interactions were demonstrated by a similar study (Jha and Bhadoriya 2018). We further observed hydrogen bond interaction of two positively charged Glu-205 and Glu-206 with the hit, which has been reported to be involved in active site construction (Aertgeerts et al. 2004). Additional interactions of the proposed inhibitors with Arg-125, Tyr-662 and Tyr-547have been previously established as key residues responsible for DPP-4 inhibition (Pantaleão et al. 2015).

Binding free energy calculation

The post docking program called MMGBSA which calculates ligand binding energies and ligand strain energies for a set of ligands and a single receptor by employing the combination of molecular mechanics calculations and salvation models was used to validate the binding affinity of respective compounds with the target protein. Since Genheden and Ryde argued that output obtain through MMGBSA for binding energies calculations were found to be highly efficient (Genheden and Ryde 2015), several studies have demonstrated MMGBSA post docking program as the most reliable for rating the affinity of a ligand on binding to its protein target (Maffucci et al. 2018; Sun et al. 2014). Table S2 and S3 showed the evaluation of the free energy of binding for each hit-protein complex. All the compounds except protocatechuic acid have a favourable binding with the proteins. Epigallocatechin and (−) epicatechin, however, recorded the most favourable ΔGBind with GFAT1 (− 45.73 kcal/mol and − 45.72 kcal/mol), while quercetin-3-O-d-galactopyranoside in complex with DPP-4 was more favourable than others (− 42.64 kcal/mol). In terms of binding free energy, the major energy contributors were identified as van der Waals (∆Gvdw), Coulomb interaction (∆GColulomb), Hydrogen bond (ΔGHbond) and lipophilic energy (∆GsolLipo) that enhance the binding affinity of the compounds towards the binding pocket of the protein (Table S1 and S2).

Prediction of ADME by Qikprop

Besides the evaluation of binding affinity and binding free energy, other properties including drug-like properties such as the number of hydrogen donor, hydrogen bond acceptor and molecular weight were also determined. These parameters are important molecular properties as formulated by Lipinski et al. (1997). The formulated rule known as Lipinski’s rule of five (RO5) stated that for a compound to be considered as druglike, the drug must not violate more than one of the following attribute: molecular weight < 500 Da, octanol–water partition coefficient < 5, hydrogen bond donor ≤ 5 and hydrogen bond acceptor ≤ 10 (Table S3) (Lipinski et al. 1997). As Gombar et al. reckoned, a prospective drug that obeys the RO5 in a clinical trial has lower attrition rates and has increased chance of passing all phases of the clinical trial. Here it’s interesting to note that all the hit compounds with the exception of quercetin-3-O-beta-d-arabinofuranoside and quercetin-3-O-d-galactopyranoside are considered as druglike. Furthermore, other parameters were further scrutinized and predicted by Qikprop and the results are shown in Fig. 3. Among them are Human Oral Absorption, Prediction of binding to human serum albumin, Predicted apparent MDCK cell permeability in nm/sec and Van der Waals surface area of polar nitrogen and oxygen atoms. Interestingly, all these parameters fell within the recommended range. The compounds’ pharmacokinetics properties were aslso predicted via SWISSADME server (Table 4); (−) epicatechin, Naringenin, Protocatechuic acid, ( +)-Catechin, Gallic acid and Delphinidin had high GI absorption which simplifies that that can be easily absorpbed by gastro intestinal tract. Five most important Iso-enzyme of cytochrom p450 (CYP450) which are responsible for the metabolism of drugs/metabolites were also taken into consideration. They are CYP1A2, CYP2C19, CYP2C9, CYP2D6 and CYP3A4. All the lead compounds with the exception of Myricetin and Naringenin showed moderation against these iso-enzymes (Table 5).

Table 4.

Prediction of compounds’ pharmacokinetics by SWISSADME server

S/no. Entry name MW GI absorption Rotatable bonds CYP1A2 inhibitor CYP2C19 inhibitor CYP2C9 inhibitor CYP2D6 inhibitor CYP3A4 inhibitor
1 Kaemferol 3-O-Beta-d-Xyloside 418.35 Low 3 No No No No No
2 Myricetin 418.24 Low 1 Yes No No No Yes
3 Quercetin-3-O-beta-d-arabinofuranoside 366.36 low 4 No No No No No
4 Delphinidin 303.24 High 1 Yes No No No No
5 Gallic acid 170.2 High 1 No No No No Yes
6 Quercetin-3-O-d-galactopyranoside 463.38 Low 4 No No No No No
7 ( +)-Catechin 290.27 High 1 No No No No No
8 Protocatechuic acid 306.27 High 1 No No No No No
9 Epigallocatechin 458.37 low No No No No No
10 Naringenin 272.25 High 1 Yes No No No Yes
11 (−) epicatechin 290.27 High 1 No No No No No

Table 5.

Best model generated for GFAT1 and DPP-4

Best model generated for GFAT1
 S/n Model code R2 RMSE Q2
 1 Kpls_desc_47 0.8208 0.1887 0.8658
Best model generated for DPP-4
 S/n Model code R2 RMSE Q2
 1 Kpls_linear_15 0.7771 0.5799 0.7303

QSAR analysis

The purpose of the AutoQSAR is to automatically produce robust QSAR models with minimal user input or understanding. This program splits the curated dataset randomly into the desired percentage of the training set and test set. The models are built on each training set from all possible combination of machine learning method, and sets of independent variables that are supported by each machine learning methods, and it could produce up to 400 models where each mode assigns a quality model score to each model based on its performance on the training and test set (An et al. 2013).Models are scored based on the equation below.

Score M = accuracy test [1.0 – (accuracy train – accuracy test)] (Li et al. 2015).

Where: Accuracy is defined between intervals 0–1.

A perfect prediction is represented by 1 and incorrect prediction as 0.00. The equation accepts models that exhibit high accuracy with respect to the test and train and penalize inaccurate models (Sastry et al. 2010).

As previously described, the autoQSAR program was used to build a different model for GFAT1 and DPP-4. The observed activities and predicted activities of the training set and test set in the negative logarithm of inhibitor concentration(pIC50) for both GFAT1 and DPP-4 was represented in Tables S4 and S5. The top model represented in Table S4 was selected, and itwas employed in this study. For GFAT1, the top model Kpls_desc_47 recorded R2 of 0.8208, RMSE of 0.1887 and Q2 of 0.8658. Consequently, the top model for DPP-4 (Kpls_linear_15) recorded R2 of 0.7771, RMSE of 0.5799 and Q2 of 0.7303. The scatter plot depicting predicted pIC50 versus experimental pIC50 for the best-generated model is shown in Figs. 9 and 10. It is worth noting that the hit compounds are observed to have promising predicted pIC50. kaemferol 3-O-Beta-d-Xyloside, Quercetin-3-O-beta-d-arabinofuranoside and Quercetin-3-O-d-galactopyranoside had predicted pIC50 of 5.076, 5.028 and 5.156 for DPP-4, and predicted pIC50 of 6.107, 6.044 and 6.044 for GFAT1.

Fig. 9.

Fig. 9

Scatter plot analysis of best model predicted from AutoQSAR for known inhibitors of DPP-4

Fig. 10.

Fig. 10

Scatter plot analysis of best model predicted from AutoQSAR for known inhibitors of GFAT1

Conclusion

Here we identified hit compounds from Anacardium occidentale as potential inhibitors of two prime therapeutic targets (GFAT1 and DPP-4) in the management of diabetes. Although molecular docking, binding free energy, QSAR and ADME properties revealed gallic acid, naringenin and kaemferol 3-O-Beta-d-Xyloside as promising compounds, these results are not sufficient to affirm their therapeutic potential against GFAT1 and DPP-4, further experiments are needed to attest to the inhibitory potentials of these compounds.

Supplementary Information

Below is the link to the electronic supplementary material.

Acknowledgement

We would like to express our sincere gratitude to Mr. Oluwole Israel Ogunsola for his efforts in proof reading and type-setting the manuscript.

Author contributions

VOU: conceptualization, experiments design, data analysis, data presentation, writing and reviewing. SAA: data analysis, data presentation, writing and reviewing. OOE: Experiments design, data analysis, data presentation, writing and reviewing.

Declarations

Conflict of interest

Authors declare no conflict of interest.

Ethics approval

Not Applicable

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  1. Abbott CA, McCaughan GW, Baker E, Sutherland GR. Genomic organization, exact localization, and tissue expression of the human CD26 (dipeptidyl peptidase IV) gene. Immunogenetics. 1994;40:331–338. doi: 10.1007/BF01246674. [DOI] [PubMed] [Google Scholar]
  2. Aertgeerts K, et al. Crystal structure of human dipeptidyl peptidase IV in complex with a decapeptide reveals details on substrate specificity and tetrahedral intermediate formation. Protein Sci. 2004;13:412–421. doi: 10.1110/ps.03460604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ahrén B. Inhibition of dipeptidyl peptidase-4 (DPP-4)-a novel approach to treat type 2 diabetes. Curr Enzym Inhib. 2005;1:65–73. doi: 10.2174/1573408052952667. [DOI] [Google Scholar]
  4. Allegra M, et al. Indicaxanthin from Opuntia ficus-indica crosses the blood–brain barrier and modulates neuronal bioelectric activity in rat hippocampus at dietary-consistent amounts. J Agric Food Chem. 2015;63:7353–7360. doi: 10.1021/acs.jafc.5b02612. [DOI] [PubMed] [Google Scholar]
  5. An Y, Sherman W, Dixon SL. Kernel-based partial least squares: application to fingerprint-based QSAR with model visualization. J Chem Inf Model. 2013;53:2312–2321. doi: 10.1021/ci400250c. [DOI] [PubMed] [Google Scholar]
  6. Berman HM, et al. The protein data bank. Nucl Acids Res. 2000;28:235–242. doi: 10.1093/nar/28.1.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bhakyaraj R, Singaravadive K. Minerals and bioactive compounds in cashew apple (Anacardium occidentale L.) J Food Resour Sci. 2012;1:32–36. doi: 10.3923/jfrs.2012.32.36. [DOI] [Google Scholar]
  8. Bitu VdCN, Bitu VdCN, Matias EFF, de Lima WP, da Costa PA, Coutinho HDM, de Menezes IRA. Ethnopharmacological study of plants sold for therapeutic purposes in public markets in Northeast Brazil. J Ethnopharmacol. 2015;172:265–272. doi: 10.1016/j.jep.2015.06.022. [DOI] [PubMed] [Google Scholar]
  9. Bobzin SC, Yang S, Kasten TP. Application of liquid chromatography–nuclear magnetic resonance spectroscopy to the identification of natural products. J Chromatogr B Biomed Sci Appl. 2000;748:259–267. doi: 10.1016/S0378-4347(00)00289-9. [DOI] [PubMed] [Google Scholar]
  10. Bolin DR, Chen S, Mischke SG, Qian Y (2006) Glutamine fructose-y-phosphate amidotransferase (GFAT) inhibitors. Google Patents [DOI] [PubMed]
  11. Castillo-Juárez I, Rivero-Cruz F, Celis H, Romero I. Anti-Helicobacter pylori activity of anacardic acids from Amphipterygium adstringens. J Ethnopharmacol. 2007;114:72–77. doi: 10.1016/j.jep.2007.07.022. [DOI] [PubMed] [Google Scholar]
  12. Chakraborty C, Hsu MJ, Agoramoorthy G. Understanding the molecular dynamics of type-2 diabetes drug target DPP-4 and its interaction with Sitagliptin and inhibitor Diprotin-A. Cell Biochem Biophys. 2014;70:907–922. doi: 10.1007/s12013-014-9998-0. [DOI] [PubMed] [Google Scholar]
  13. Deacon CF, Ahrén B, Holst JJ. Inhibitors of dipeptidyl peptidase IV: a novel approach for the prevention and treatment of type 2 diabetes? Expert Opin Investig Drugs. 2004;13:1091–1102. doi: 10.1517/13543784.13.9.1091. [DOI] [PubMed] [Google Scholar]
  14. Dixon SL, Duan J, Smith E, Von Bargen CD, Sherman W, Repasky MP. AutoQSAR: an automated machine learning tool for best-practice quantitative structure–activity relationship modeling. Fut Med Chem. 2016;8:1825–1839. doi: 10.4155/fmc-2016-0093. [DOI] [PubMed] [Google Scholar]
  15. Du X-L, et al. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc Natl Acad Sci. 2000;97:12222–12226. doi: 10.1073/pnas.97.22.12222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Elekofehinti OO, Ejelonu O, Famuti A, et al. Discovery of potential visfatin activators using in silico docking and ADME predictions as therapy for type 2 diabetes. Beni-suef Univ J Basic Appl Sci. 2018;7:241–249. doi: 10.1016/j.bjbas.2018.02.007. [DOI] [Google Scholar]
  17. Fakhri Y, et al. Association between Dichlorodiphenyldichloroethylene in the serum and adipose tissue with Type 2 diabetes: a systematic review and meta-analysis. Global J Health Sci. 2016;9:43–54. doi: 10.5539/gjhs.v9n2p43. [DOI] [Google Scholar]
  18. Fang XK, Gao J, Zhu DN. Kaempferol and quercetin isolated from Euonymus alatus improve glucose uptake of 3T3-L1 cells without adipogenesis activity. Life Sci. 2008;82(11–12):615–622. doi: 10.1016/j.lfs.2007.12.021. [DOI] [PubMed] [Google Scholar]
  19. Floquet N, Richez C, Durand P, Maigret B, Badet B, Badet-Denisot M-A. Discovering new inhibitors of bacterial glucosamine-6P synthase (GlmS) by docking simulations. Bioorg Med Chem Lett. 2007;17:1966–1970. doi: 10.1016/j.bmcl.2007.01.052. [DOI] [PubMed] [Google Scholar]
  20. Franks PW, Hanson RL, Knowler WC, Sievers ML, Bennett PH, Looker HC. Childhood obesity, other cardiovascular risk factors, and premature death. N Engl J Med. 2010;362:485–493. doi: 10.1056/NEJMoa0904130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Fuller JC, Burgoyne NJ, Jackson RM. Predicting druggable binding sites at the protein–protein interface. Drug Discov Today. 2009;14:155–161. doi: 10.1016/j.drudis.2008.10.009. [DOI] [PubMed] [Google Scholar]
  22. Genheden S, Ryde U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opin Drug Discov. 2015;10:449–461. doi: 10.1517/17460441.2015.1032936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Guedes IA, de Magalhães CS, Dardenne LE. Receptor–ligand molecular docking. Biophys Rev. 2014;6:75–87. doi: 10.1007/s12551-013-0130-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Harder E, et al. OPLS3: a force field providing broad coverage of drug-like small molecules and proteins. J Chem Theory Comput. 2016;12:281–296. doi: 10.1021/acs.jctc.5b00864. [DOI] [PubMed] [Google Scholar]
  25. Hassanzadeh V, et al. Association between polychlorinated biphenyls in the serum and adipose tissue with type 2 diabetes mellitus: a systematic review and meta-analysis International. J Med Res Health Sci. 2016;5:13–21. [Google Scholar]
  26. Hawkins M, Barzilai N, Liu R, Hu M, Chen W, Rossetti L. Role of the glucosamine pathway in fat-induced insulin resistance. J Clin Investig. 1997;99:2173–2182. doi: 10.1172/JCI119390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Hong W, Doyle D. Molecular dissection of the NH2-terminal signal/anchor sequence of rat dipeptidyl peptidase IV. J Cell Biol. 1990;111:323–328. doi: 10.1083/jcb.111.2.323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Hu FB. Globalization of diabetes: the role of diet, lifestyle, and genes. Diabet Care. 2011;34:1249–1257. doi: 10.2337/dc11-0442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Iwaloye O, Elekofehinti OO, Oluwarotimi EA, Babatomiwa K, Momoh IA. In silico molecular studies of natural compounds as possible anti-Alzheimer’s agents: ligand-based design. Netw Model Anal Health Inform Bioinform. 2020;9:54. doi: 10.1007/s13721-020-00262-7. [DOI] [Google Scholar]
  30. Iwaloye O, Elekofehinti OO, Oluwarotimi EA, et al. Insight into glycogen synthase kinase-3β inhibitory activity of phyto-constituents from Melissa officinalis: in silico studies. Silico Pharmacol. 2020;8:2. doi: 10.1007/s40203-020-00054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Iwaloye O, Elekofehinti, OO, Babatomiwa K, Fadipe TM, Akinjiyan MO, Ariyo EO, Aiyeku OO, Adewumi NA (2020b) Discovery of TCM derived compounds as wild type and mutant Plasmodium falciparum dihydrofolate reductase inhibitors: induced fit docking and ADME studies. Curr Drug Discov Technol. 10.2174/1570163817999200729122753 [DOI] [PubMed]
  32. Jacobson MP, Friesner RA, Xiang Z, Honig B. On the role of the crystal environment in determining protein side-chain conformations. J Mol Biol. 2002;320:597–608. doi: 10.1016/S0022-2836(02)00470-9. [DOI] [PubMed] [Google Scholar]
  33. Jacobson MP, Pincus DL, Rapp CS, Day TJ, Honig B, Shaw DE, Friesner RA. A hierarchical approach to all-atom protein loop prediction. Protein Struct Funct Bioinform. 2004;55:351–367. doi: 10.1002/prot.10613. [DOI] [PubMed] [Google Scholar]
  34. Jaiswal Y, Tatke P, Gabhe S, Vaidya A. Antidiabetic activity of extracts of Anacardium occidentale Linn. leaves on n-streptozotocin diabetic rats. J Tradit Comp Med. 2017;7:421–427. doi: 10.1016/j.jtcme.2016.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. James N, Ramanathan K. Discovery of potent ALK inhibitors using pharmacophore-informatics strategy. Cell Biochem Biophys. 2018;76:111–124. doi: 10.1007/s12013-017-0800-y. [DOI] [PubMed] [Google Scholar]
  36. Jha V, Bhadoriya KS. Synthesis, pharmacological evaluation and molecular docking studies of pyrimidinedione based DPP-4 inhibitors as antidiabetic agents. J Mol Struct. 2018;1158:96–105. doi: 10.1016/j.molstruc.2018.01.014. [DOI] [Google Scholar]
  37. Kikiowo B, Ogunleye J, Iwaloye O, Ijatuyi T. Therapeutic potential of Chromolaena odorata phyto-constituents against human pancreatic α-amylase Therapeutic potential of Chromolaena odorata phyto-constituents against human pancreatic a-amylase. J Biomol Struct Dyn. 2020 doi: 10.1080/07391102.2020.1833758. [DOI] [PubMed] [Google Scholar]
  38. Koliaki C, Doupis J. Incretin-based therapy: a powerful and promising weapon in the treatment of type 2 diabetes mellitus. Diabet Therapy. 2011;2:101–121. doi: 10.1007/s13300-011-0002-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Kolm-Litty V, Sauer U, Nerlich A, Lehmann R, Schleicher E. High glucose-induced transforming growth factor beta1 production is mediated by the hexosamine pathway in porcine glomerular mesangial cells. J Clin Investig. 1998;101:160–169. doi: 10.1172/JCI119875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kubo I, Nitoda T, Tocoli FE, Green IR. Multifunctional cytotoxic agents from Anacardium occidentale. Phytother Res. 2011;25:38–45. doi: 10.1002/ptr.3109. [DOI] [PubMed] [Google Scholar]
  41. Kuhn B, Hennig M, Mattei P. Molecular recognition of ligands in dipeptidyl peptidase IV. Curr Top Med Chem. 2007;7:609–620. doi: 10.2174/156802607780091064. [DOI] [PubMed] [Google Scholar]
  42. Kulis M, MacQueen I, Li Y, Guo R, Zhong X-P, Burks AW. Pepsinized cashew proteins are hypoallergenic and immunogenic and provide effective immunotherapy in mice with cashew allergy. J Allergy Clin Immunol. 2012;130:716–723. doi: 10.1016/j.jaci.2012.05.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Le Camus C, Badet-Denisot M-A, Badet B. Arabinose-5-phosphate oxime vs its methylenephosphonate mimetic as high energy intermediate of the glucosamine-6P synthase catalyzed reaction. Tetrahedron Lett. 1998;39:2571–2572. doi: 10.1016/S0040-4039(98)00263-9. [DOI] [Google Scholar]
  44. Le Camus C, Chassagne A, Badet-Denisot M-A, Badet B. Stereoselective synthesis of 5-methylphosphono-d-arabino hydroximolactone, inhibitor of glucosamine-6-phosphate synthase and phosphoglucose isomerase. Tetrahedron Lett. 1998;39:287–288. doi: 10.1016/S0040-4039(97)10514-7. [DOI] [Google Scholar]
  45. Li J, et al. Synthesis of many different types of organic small molecules using one automated process. Science. 2015;347:1221–1226. doi: 10.1126/science.aaa5414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. 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. 1997;23:3–25. doi: 10.1016/S0169-409X(96)00423-1. [DOI] [PubMed] [Google Scholar]
  47. Ludwig K, Yan S, Fan H, Reutter W, Böttcher C. The 3D structure of rat DPPIV/CD26 as obtained by cryo-TEM and single particle analysis. Biochem Biophys Res Commun. 2003;304:73–77. doi: 10.1016/S0006-291X(03)00539-4. [DOI] [PubMed] [Google Scholar]
  48. Maffucci I, Hu X, Fumagalli V, Contini A. An efficient implementation of the Nwat-MMGBSA method to rescore docking results in medium-throughput virtual screenings. Front Chem. 2018;6:43. doi: 10.3389/fchem.2018.00043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Mandal S, Mn M, Mandal SK. Rational drug design. Eur J Pharmacol. 2009;625:90–100. doi: 10.1016/j.ejphar.2009.06.065. [DOI] [PubMed] [Google Scholar]
  50. Marshall S, Bacote V, Traxinger R. Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem. 1991;266:4706–4712. doi: 10.1016/S0021-9258(19)67706-9. [DOI] [PubMed] [Google Scholar]
  51. McKnight G, Mudri S, Mathewes S, Traxinger R, Marshall S, Sheppard P, O'hara P (1992) Molecular cloning, cDNA sequence, and bacterial expression of human glutamine: fructose-6-phosphate amidotransferase. J Biol Chem 267:25208–25212 [PubMed]
  52. Misumi Y, Hayashi Y, Arakawa F, Ikehara Y (1992) Molecular cloning and sequence analysis of human dipeptidyl peptidase IV, a serine proteinase on the cell surface. Biochimica et Biophysica Acta (BBA)-Gene Struct Express 1131:333–336 [DOI] [PubMed]
  53. Nguyen H, Chitturi S, Maple-Brown L. Management of diabetes in Indigenous communities: lessons from the Australian Aboriginal population. Intern Med J. 2016;46:1252–1259. doi: 10.1111/imj.13123. [DOI] [PubMed] [Google Scholar]
  54. Niimi M, et al. Identification of GFAT1-L, a novel splice variant of human glutamine: fructose-6-phosphate amidotransferase (GFAT1) that is expressed abundantly in skeletal muscle. J Hum Genet. 2001;46:566–571. doi: 10.1007/s100380170022. [DOI] [PubMed] [Google Scholar]
  55. Nisha CM, Kumar A, Vimal A, Bai BM, Pal D, Kumar A. Docking and ADMET prediction of few GSK-3 inhibitors divulges 6-bromoindirubin-3-oxime as a potential inhibitor. J Mol Graph Model. 2016;65:100–107. doi: 10.1016/j.jmgm.2016.03.001. [DOI] [PubMed] [Google Scholar]
  56. Ogata S, Misumi Y, Tsuji E, Takami N, Oda K, Ikehara Y. Identification of the active site residues in dipeptidyl peptidase IV by affinity labeling and site-directed mutagenesis. Biochemistry. 1992;31:2582–2587. doi: 10.1021/bi00124a019. [DOI] [PubMed] [Google Scholar]
  57. Oki T, Yamazaki K, Kuromitsu J, Okada M, Tanaka I. cDNA cloning and mapping of a novel subtype of glutamine: fructose-6-phosphate amidotransferase (GFAT2) in human and mouse. Genomics. 1999;57:227–234. doi: 10.1006/geno.1999.5785. [DOI] [PubMed] [Google Scholar]
  58. Olajide OA, Aderogba MA, Adedapo AD, Makinde JM. Effects of Anacardium occidentale stem bark extract on in vivo inflammatory models. J Ethnopharmacol. 2004;95:139–142. doi: 10.1016/j.jep.2004.06.033. [DOI] [PubMed] [Google Scholar]
  59. Olajide OA, Aderogba MA, Fiebich BL. Mechanisms of anti-inflammatory property of Anacardium occidentale stem bark: Inhibition of NF-κB and MAPK signalling in the microglia. J Ethnopharmacol. 2013;145:42–49. doi: 10.1016/j.jep.2012.10.031. [DOI] [PubMed] [Google Scholar]
  60. Olatunji LA, Okwusidi JI, Soladoye AO. Antidiabetic effect of Anacardium occidentale. Stem-bark in fructose-diabetic rats. Pharmac Biol. 2005;43:589–593. doi: 10.1080/13880200500301712. [DOI] [Google Scholar]
  61. Palheta I, Ferreira L. Hypoglycemic potential of Anacardium occidentale L. J Anal Pharm Res. 2018;7:152–153. [Google Scholar]
  62. Pantaleão SQ, Maltarollo VG, Araujo SC, Gertrudes JC, Honorio KM. Molecular docking studies and 2D analyses of DPP-4 inhibitors as candidates in the treatment of diabetes. Mol Bio Syst. 2015;11:3188–3193. doi: 10.1039/c5mb00493d. [DOI] [PubMed] [Google Scholar]
  63. Qian Y, et al. Discovery of 1-arylcarbonyl-6, 7-dimethoxyisoquinoline derivatives as glutamine fructose-6-phosphate amidotransferase (GFAT) inhibitors. Bioorg Med Chem Lett. 2011;21:6264–6269. doi: 10.1016/j.bmcl.2011.09.009. [DOI] [PubMed] [Google Scholar]
  64. Rohini K, Shanthi V. Hyphenated 3D-QSAR statistical model-drug repurposing analysis for the identification of potent neuraminidase inhibitor. Cell Biochem Biophys. 2018;76:357–376. doi: 10.1007/s12013-018-0844-7. [DOI] [PubMed] [Google Scholar]
  65. Röhrig UF, et al. Rational design of indoleamine 2, 3-dioxygenase inhibitors. J Med Chem. 2010;53:1172–1189. doi: 10.1021/jm9014718. [DOI] [PubMed] [Google Scholar]
  66. Ruegenberg S, Horn M, Pichlo C, Allmeroth K, Baumann U, Denzel MS. Loss of GFAT-1 feedback regulation activates the hexosamine pathway that modulates protein homeostasis. Nat Commun. 2020;11:1–16. doi: 10.1038/s41467-020-14524-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Salehi B, Gültekin-Özgüven M, Kırkın C, Özçelik B, Morais-Braga MFB, et al. Anacardium plants: chemical, nutritional composition and biotechnological applications. Biomolecules. 2019;9:465. doi: 10.3390/biom9090465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Sastry M, Lowrie JF, Dixon SL, Sherman W. Large-scale systematic analysis of 2D fingerprint methods and parameters to improve virtual screening enrichments. J Chem Inf Model. 2010;50:771–784. doi: 10.1021/ci100062n. [DOI] [PubMed] [Google Scholar]
  69. Schrödinger (2017) Schrödinger release 2017-2: LigPrep, Schrödinger, LLC, New York, NY, USA
  70. Schrödinger (2018a) Schrödinger release 2018–2: Maestro, version 11.8. Schrödinger, LLC, New York, NY, USA.
  71. Schrödinger (2018b) Schrödinger Release 2018-2: QikProp, Schrödinger LLC New York, NY, USA
  72. Schrödinger (2018c) Schrödinger Release 2018-4: Glide, Schrödinger, LLC, New York, NY, USA
  73. Sherman W, Day T, Jacobson MP, Friesner RA, Farid R. Novel procedure for modeling ligand/receptor induced fit effects. J Med Chem. 2006;49:534–553. doi: 10.1021/jm050540c. [DOI] [PubMed] [Google Scholar]
  74. Shoback D, Gardner D (2011) Chapter 17 Greenspan's basic and clinical endocrinology (9th ed). McGraw-Hill Medical, New York, 217–236
  75. Sun H, Li Y, Tian S, Xu L, Hou T. Assessing the performance of MM/PBSA and MM/GBSA methods. 4. Accuracies of MM/PBSA and MM/GBSA methodologies evaluated by various simulation protocols using PDBbind data set. Phys Chem Chem Phys. 2014;16:16719–16729. doi: 10.1039/C4CP01388C. [DOI] [PubMed] [Google Scholar]
  76. Ukwenya VO, Ashaolu JO, Adeyemi AO, Akinola OA, Caxton-Martins EA (2012) Antihyperglycemic activities of methanolic leaf extract of Anacardium occidentale (Linn.) on the pancreas of streptozotocin-induced diabetic rats. J Cell Anim Biol 6(11):169–174
  77. Venkatachalam CM, Jiang X, Oldfield T, Waldman M. LigandFit: a novel method for the shape-directed rapid docking of ligands to protein active sites. J Mol Graph Model. 2003;21:289–307. doi: 10.1016/S1093-3263(02)00164-X. [DOI] [PubMed] [Google Scholar]
  78. Watanabe YS, Yasuda Y, Kojima Y, Okada S, Motoyama T, Takahashi R, Oka M. Anagliptin, a potent dipeptidyl peptidase IV inhibitor: its single-crystal structure and enzyme interactions. J Enzyme Inhib Med Chem. 2015;30:981–988. doi: 10.3109/14756366.2014.1002402. [DOI] [PubMed] [Google Scholar]
  79. Williams R, Colagiuri S, Chan J, Gregg E, Ke C, Lim L-L, Yang X (2019) IDF Atlas 9th Edition 2019.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from In Silico Pharmacology are provided here courtesy of Springer

RESOURCES