You are on page 1of 13

Journal of Molecular Graphics and Modelling 126 (2024) 108640

Contents lists available at ScienceDirect

Journal of Molecular Graphics and Modelling


journal homepage: www.elsevier.com/locate/jmgm

Finding structural requirements of structurally diverse α-glucosidase and


α-amylase inhibitors through validated and predictive 2D-QSAR and
3D-QSAR analyses
Soumya Mitra a, Subhadas Chatterjee a, Shobhan Bose a, Parthasarathi Panda a, Souvik Basak a,
Nilanjan Ghosh b, **, Subhash C. Mandal b, Saroj Singhmura a, Amit Kumar Halder a, *
a
Dr. B. C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, 713206, India
b
Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India

A R T I C L E I N F O A B S T R A C T

Keywords: Diabetes mellitus (DM) is a chronic metabolic disorder characterized by hyperglycemic state. The α-glucosidase
T2DM and α-amylase are considered two major targets for the management of Type 2 DM due to their ability of
α-glucosidase metabolizing carbohydrates into simpler sugars. In the current study, cheminformatics analyses were performed
α-amylase
to develop validated and predictive models with a dataset of 187 α-glucosidase and α-amylase dual inhibitors.
Dual inhibitors
Separate linear, interpretable and statistically robust 2D-QSAR models were constructed with datasets containing
QSAR
the activities of α-glucosidase and α-amylase inhibitors with an aim to explain the crucial structural and phys­
icochemical attributes responsible for higher activity towards these targets. Consequently, some descriptors of
the models pointed out the importance of specific structural moieties responsible for the higher activities for
these targets and on the other hand, properties such as ionization potential and mass of the compounds as well as
number of hydrogen bond donors in molecules were found to be crucial in determining the binding potentials of
the dataset compounds. Statistically significant 3D-QSAR models were developed with both α-glucosidase and
α-amylase inhibition datapoints to estimate the importance of 3D electrostatic and steric fields for improved
potentials towards these two targets. Molecular docking performed with selected compounds with homology
model of α-glucosidase and X-ray crystal structure of α-amylase largely supported the interpretations obtained
from the cheminformatic analyses. The current investigation should serve as important guidelines for the design
of future α-glucosidase and α-amylase inhibitors. Besides, the current investigation is entirely performed by using
non-commercial open-access tools to ensure easy accessibility and reproducibility of the investigation which may
help researchers throughout the world to work more on drug design and discovery.

1. Introduction polyphagia, loss of weight and blurred vision. According to a report by


the International Diabetes Federation, IDF Diabetes Atlas, 9th Edition
Diabetes mellitus (DM) is a chronic metabolic disorder characterized 2019; approximately 436 million people are suffering from DM and the
by hyperglycemic state. Hyperglycemia results from insufficient insulin number would reach up to 700 million by the end of 2045 [3]. Inter­
secretion or loss of insulin activity or as a result of both. The disease estingly enough, the prevalence of Type II Diabetes mellitus (T2DM) is
burden of diabetes mellitus is complicated by the generation of different much higher in human beings which results from insulin resistance as
microvascular complications i.e., neuropathy, nephropathy, retinopathy well as defects in insulin secretion. The role of oxidative stress in
or macrovascular complications like peripheral and autonomic neu­ aggravating pathological complications in T2DM is crucial, considering
ropathy, increased incidence of atherosclerosis, peripheral and cere­ the generation of reactive oxygen species (ROS) which inadvertently
brovascular diseases [1,2]. Chronic hyperglycemia is considered to be causes damage to different organs and interrupts enzymatic pathways.
the primary characteristic trait of DM which includes polyuria, The highly volatile nature of the free radical causes oxidative stress to

* Corresponding author.
** Corresponding author.
E-mail addresses: nilanjanghosh.phamacy@jadavpuruniversity.in (N. Ghosh), amitcsir2011@gmail.com (A.K. Halder).

https://doi.org/10.1016/j.jmgm.2023.108640
Received 7 April 2023; Received in revised form 9 September 2023; Accepted 26 September 2023
Available online 27 September 2023
1093-3263/© 2023 Elsevier Inc. All rights reserved.
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

the cellular machinery by the formation of advanced glycation products triazinoindole-thiazolidinone classes were collected from the recent
(AGEs) which play a key role in causing cellular damage. The excess studies conducted by Khan et al. [12–19]. The compounds were
production of ROS through the polyol pathway leads to retrained anti­ renamed with numerical codes and suffixes as AT [12], BT [13], IZ [14],
oxidant enzymatic activity of the system resulting in enhanced oxidative TZ [15], DC [16], ID [17], TT [18] and TH [19]. The inhibitory activities
stress in hyperglycemic conditions [4,5]. The uphill management of of these compounds were tested against α-glucosidase enzyme obtained
T2DM has brought a plethora of probable approaches ranging from from brewer’s yeast Saccharomyces cerevisiae and while α-amylase was
conventional lifestyle modifications to single-regime drug therapies. obtained from human pancreas. The half maximum inhibitory concen­
However, recent developments have suggested the involvement of tration (IC50 in μM) values of these compounds were calculated into
multiple therapies to be more beneficial. pIC50 (=-log10(IC50/106)) values and were subsequently used as the
One of the most coveted approaches in combatting the relentless response variables for 2D-QSAR and 3D-QSAR analyses. The structures,
damages caused by T2DM is through dual inhibition of α-glucosidase in the form of SMILES, and biological responses of these ligands are
and α-amylase enzymes. The α-glucosidase is one of the major given in the supplementary material of this work. The 2D structures of
carbohydrate-digesting enzymes that is present in the brush borders of the compounds were drawn by using Chemsketch academic version and
the intestine and contributes in breaking down of complex oligosac­ they were changed to.sdf files by utilization of Discovery Studio Visu­
charides into simple monosaccharides. The α-glucosidase inhibitors alization tools and were numbered subsequently. The structural opti­
delay the absorption of carbohydrates into the small intestines which mization of the compounds was carried out with Chemaxon standardizer
effectively allows pancreas to secrete enough insulin. These inhibitors tool by using the following options: (a) explicit hydrogen atoms were
exert moderate antihyperglycemic action without interfering with the added, (b) compounds were aromatized, (c) cleaned 2D, (d) cleaned 3D,
insulin secretion directly, thus monotherapy is not recommended. (e) neutralized and (f) stripped of salts. The standardized structures were
However, there are several adverse effects associated with α-glucosidase further processed for 2D-QSAR and 3D-QSAR analyses [20].
inhibitors like acarbose; such as nausea, bloating and flatulence [6,7].
The α-amylase, a digestive endo-amylase secreted from the saliva and 2.2. 2D-QSAR analyses
pancreas, is crucial in metabolism of long chain polysaccharides into
simple carbohydrate like glucose. The enzyme catalyzes hydrolysis of 2.2.1. Descriptor calculation
the α1→4 linkage of the polysaccharides into monosaccharides while Descriptor calculation of the 187 compounds was carried out by
retaining the α-monomeric form of the same. The prompt hydrolysis of using alvaDesc v.2.0.4 (https://www.alvascience.com/alvadesc/) [21]
the carbohydrates in simple glucose by α-amylase may also be consid­ under OCHEM webserver [22]. Corina tool was utilized during
ered to be one of the leading causes for T2DM which ultimately brings geometrical optimization of the structures for 3D descriptor calculation
forth the strategy of inhibiting the enzyme as a probable approach. In [23]. The calculated descriptors of these ligands were then integrated
this quest, triazinoindole, thiazolidin-4-one derivatives have been with respective pIC50 values of the compounds to develop the complete
extensively investigated for their possible α-amylase inhibitory potential dataset for 2D-QSAR model generation.
[8–10]. The findings suggested key structural scaffolds playing signifi­
cant role in offering inhibitory potential of the enzymes. This, coupled 2.2.2. Dataset division and model development
with extensive literature survey from our group of researchers, have led Dataset division plays a key role towards generation of 2D QSAR
the notion for exploring scaffolds with dual inhibitory potential towards modelling. The dataset was split into a training set and a test set by using
the carbohydrate metabolizing enzymes; i.e. α-glucosidase and k-means cluster analysis (kMCA) technique of Python based open-access
α-amylase through ligand and structure based in silico studies and sub­ SFS-QSAR tool (https://github.com/ncordeirfcup/SFS-QSAR-tool) [24].
sequent design and development of the dual inhibitors [11]. Dataset division was carried out by keeping 20% of the structures in test
Research investigations conducted by Khan et al. on design and set while k-MCA supplied a rational data-distribution strategy in which
development of dual inhibitors of these enzymes reported a series of the response variables as well as the independent variables are initially
compounds with potent inhibitory action [12–19]. These investigators clustered into eight groups and then the dataset was randomly divided
focused on diverse scaffolds such as triazole, triazine, indazole, indole, (random seed 42 in SFS-QSAR) into training set (80%) and test set
dicyanoaniline, quinolinyl bearing thiadiazole-4-one, and (20%). The 2D-QSAR models were developed by deploying two sets of
triazinoindole-thiazolidinone moieties for determining inhibitory action alvaDesc descriptors. First, only some selected interpretable descriptors
against the enzymes. In their investigation, the synthesized compounds were used and these descriptors belong to the categories of constitu­
were tested for their activity with maximum half inhibitory concentra­ tional descriptors, functional group counts, 2D-atom pairs, drug-like
tion (IC50) against both the enzymes and values ranged between 1.1 μM indices, ring descriptors, atom-centred fragments, pharmacophore de­
and 66.77 μM for α-glucosidase while for α-amylase the values ranged scriptors and molecular properties. Next, all alvaDesc descriptors were
between 0.9 μM and 66.50 μM. Such wide range of biological activity for applied to check whether inclusion of additional descriptors (relatively
focused library of diverse ligands require detailed in silico study to less interpretable) is genuinely required for characterization of the
identify structural features contributing to higher inhibitory action structural requirements of the dataset ligands. Two distinct feature se­
against these enzymes. lection tactics were used for 2D-QSAR model development - (a)
In this current study, cheminformatic modelling approaches like 2D sequential forward selection (SFS) and (b) genetic algorithm (GA). The
and 3D quantitative structure activity relationship (2D-QSAR and 3D- first SFS mediated model was developed by using python based open
QSAR) have been performed to analyse the structural features in order access tool, SFS-QSAR (https://github.com/ncordeirfcup/SFS-QSAR-
to enhance inhibitory potency against α-glucosidase and α-amylase. tool) which administers ‘Feature Selector’ module of Mlxtend library
From the enzyme kinetic studies by Khan et al., it was found that the (http://rasbt.github.io/mlxtend/) [24]. Constant and near-constant
ligands can bind to the enzymes in competitive, non-competitive and descriptors were eliminated by keeping the variance cut-off at 0.0001
uncompetitive manner [12–19]. while highly inter-correlated descriptors were eliminated by setting the
correlation cut-off at 0.99. Models were developed by using four scoring
2. Materials and methods functions from ‘Sequential Feature Selector’ module which included R2:
determination coefficient, NMAE: negative mean absolute error, NMPD:
2.1. Dataset collection and structure preparation negative mean Poisson deviance and NMGD: negative mean gamma
deviance. The models were generated with keeping 0-fold cross vali­
A total of 187 compounds of triazole, triazine, indazole, indole, dation initially and 5-fold cross validation subsequently. GeneticAlgor­
dicyanoaniline, quinolinyl bearing thiadiazole-4-one, and ithm v.4.1_2 (accessed from https://dtclab.webs.com/software-tools)

2
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

[25], an open access tool was employed to establish GA-based models. standardized errors of prediction (SDEP) values. To confirm whether the
SFS uses a non-stochastic feature selection method which is distinctive generated model was distinct and not generated by serendipity, pro­
from GA which follows stochastic algorithms to develop randomized gressive scrambling operations were run for selected models with crit­
models while implementing techniques like cross-over and mutations to ical value: 0.80, type: LMO groups = 5, runs = 20 and scrambling = 20
improve fitting of the independent variable with the response variables. as set criteria. ‘Fitted Q2 values’ in Open3DQSAR resulted as the
Descriptor pre-treatment was carried out for GA which is similar to outcome of the scrambling test, which is denoted as Q2s in this study. Low
SFS-QSAR. Five descriptors were allowed for development of 2D-QSAR value of Q2s as compared to Q2LMO in this study justifies the robust nature
model. of the developed 3D-QSAR model. The contour maps were visualized
with isocontour values at PLS coefficients of +0.005 (green) and − 0.005
2.2.3. Statistical analyses of the models (yellow) for the steric field and the +0.003 (blue) and − 0.003 (red) for
The predictability of the eventual 2D-QSAR model was justified with electrostatic field. Matplotlib software has been used to generate all the
a number of well-established statistical parameters like determination of 2D-QSAR and 3D-QSAR plots.
R2, adjusted R2 (R2A), F-statistics, mean absolute error (MAE), and in­
ternal cross-validation parameter Q2LOO [26,27]. R2Pred, an external vali­ 2.4. Structure based modelling
dation parameter was utilized to validate predictivity of the generated
models [28]. Parameters related to rm2 matrices like rm2LOO and Δrm2LOO 2.4.1. Homology modelling
were also used for internal validation while external validation was The homology model of Saccharomyces cerevisiae α-glucosidase was
accomplished by rm2test and Δrm2test [29]. Inter-collinearity among the developed using SWISS-MODEL webserver [35]. The FASTA sequence
descriptors of the best models were estimated from cross-correlation was obtained from Uniprot (access code P53341) [36]. The template
matrices from the descriptors. Furthermore, variation inflation factor which was used for developing the homology model is Saccharomyces
(VIF) was calculated to estimate multi-collinearity of the generated cerevisiae isomaltase (PDB ID: 3AJ7) that possesses satisfactory sequence
models (VIF = 1/1-R2i , where R2i is the determination coefficient (R2) similarity (72.4%) with α-glucosidase. Notably, in one of the previously
determined by regressing the ith descriptor on the other descriptors) published report, Wang et al. also used the same FASTA sequence and
[30]. In addition to this, Y-randomization test was carried out 1000 template for developing homology model of Saccharomyces cerevisiae
times to generate 1000 randomized response variables. cR2p value was α-glucosidase [37]. After developing the model, its detailed structural
calculated to confirm the fact that the final model was not created by characterization was performed with the help of Molprobity webserver
chance, which could be predicted by its higher value [31]. (http://molprobity.biochem.duke.edu/index.php) [38].

2.2.4. Applicability domain of the models 2.4.2. Molecular docking


Applicability domain of the generated model was estimated by Molecular docking was performed with homology model of Saccha­
Williams plot which is a plot generated between leverage values and romyces cerevisiae α-glucosidase as well as human α-amylase, which was
standardized coefficients. If leverage value of any data point exceeds the retrieved from Protein Data Bank (PDB:3BAJ). For detection of cavity for
hat value h* (h* = 3*p/n, where p is the number of model’s descriptors molecular docking, we explored CB-Dock2 that implements protein-
+1 and n is the number of data-points in the training set), it is considered surface-curvature-based cavity detection technique (10.1093/nar/
to be structural outlier whereas, when standardized residual of the data- gkac394). The molecular docking was performed with Autodock 4.2
point is > ±2.5, it is deemed to be a response outlier [32]. [39]. For α-glucosidase, a grid was generated at X = 16.0, Y = − 14.0 and
Z = 15.0 with size of 40Å X 40Å X 40Å. On the other hand, the grid was
located at X = 8.3, Y = 15.1 and Z = 41.1 with the same extensions. The
2.3. 3D-QSAR analyses docking steps involved preparation of protein that includes addition of
hydrogen atoms, Gasteiger charges and assigning the atoms with AD4
2.3.1. Alignment method types. After preparing the PDBQT files (of proteins and ligands), Auto­
In this study, 3D-QSAR model development was carried out by atom- grid4 was run to generate the affinity maps for each atom type of the
based alignment or unsupervised rigid-body molecular alignment [33]. ligands and subsequently, Autodock4 was run by setting genetic algo­
For atom-based alignment, ‘obminimize’ function from OpenBabel soft­ rithm as ‘Search parameter’ and Lamarckian GA (4.2) as output
ware was employed which used steepest descent method for carrying out parameter file. Default settings were used for the remaining docking
the alignment. 100 conformations of the 3D structures of the ligands parameters. The docked poses were analysed using Discovery Studio
were then generated and the alignment was accomplished by using Visualizer tool.
rdMolAlign.GetCrippenO3A program of RDKit. The python scripts for the
rigid-body based molecular alignment have been provided in https://gi 3. Results and discussions
thub.com/ncordeirfcup/InsilicoModeling_RdRp.git.
3.1. 2D-QSAR models: α-glucosidase inhibitors
2.3.2. Model development
An open-source software, Open3DQSAR, was used to perform 3D- As described earlier in the study, the 2D-QSAR model was developed
QSAR analyses [34]. This tool utilizes a carbon and volume-less posi­ by using all alvaDesc descriptors by deploying two feature selection
tively (+1) charged probe to calculate steric and electrostatic field of the techniques i.e., sequential forward selection (SFS) and genetic algorithm
provided ligands. Pre-treatment of the fields were carried out by keeping (GA). The generated models were compared for best statistical quality it
smart region definition (SRD) cut-off level at 2.0 with removal of N-level was found that 2D-QSAR model generated with SFS by R2 scoring and 5-
variables. The Open3DQSAR employs SRD for grouping variables based fold cross-validation had slightly better statistical quality than those
on closeness of the variables in 3D space as well as two distinct algo­ generated by GA-MLR. The findings have been presented in Table 1.
rithms for variable selection are deployed, which are Fractional Facto­ From Table 1, it can be deduced that as compared to the models
rial Design-based variable SELection (FFD-SEL) and Uninformative developed with selected interpretable descriptors, significant improve­
Variable Elimination-based Partial Least Square (UVE-PLS). Predictive ment is noticed in the statistical quality of the model when all de­
quality of the PLS models developed from the ligands was assessed by R2 scriptors are used. When all descriptors are employed, SFS based feature
with standardized errors of calibration (SDEC), F-test results, selection strategy coupled with R2 scoring function and 5-fold cross-
leave-one-out Q2 (Q2LOO), leave-two-out Q2 (Q2LTO), leave-many-out Q2 validation produced the model with maximum overall predictivity.
(Q2LMO, with 5 groups and 20 runs) and R2Pred with associated Moreover, we observed that this model was indeed generated with five

3
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Table 1

= 0.841,
2D-QSAR analyses of α-glucosidase inhibitors with some interpretable and all
descriptors (the most significant model is marked in bold).
Methoda Scoreb Foldc Interpretable descriptorsd All descriptorse

Ntraining = 147, R2 = 0.899, R2Adj = 0.896, F(F(5,141)) = 251.47, Q2LOO = 0.886, MAE = 0.133, r2LOO
m
Q2LOO R2Pred Q2LOO R2Pred

SFS R2 0 0.812 0.765 0.879 0.81


NMAE 0 0.83 0.765 0.885 0.837
NMPD 0 0.812 0.765 0.87 0.845
NMGD 0 0.812 0.765 0.87 0.845
R2 5 0.808 0.821 0.886 0.859
NMAE 5 0.79 0.764 0.876 0.786
NMPD 5 0.805 0.777 0.886 0.859
NMGD 5 0.805 0.777 0.886 0.859

m = 0.075
GA NA NA 0.835 0.806 0.872 0.866

a: Feature selection methods, SFS: sequential forward selection, GA: Genetic

m = 0.823, Δrtest
algorithm, b: Scoring functions used in SFS, check the main text for full names, c:

2
cross-validation methods used for SFS based feature selection, 0: No cross-
validation, 5: 5-fold cross validation, d: Only selected categories of descriptors
with high mechanistic interpretability are used (see the main text), e: all alva­

= 0.076, Ntest = 40, R2Pred = 0.859, r2test


Desc descriptors were employed for model development.

descriptors with significantly high interpretability. Therefore, this


model is therefore discussed in the current work. The equation and the
statistical results of the SFS-MLR model is presented in Table 2.
The 2D-QSAR model explains 89.6% and predicts 88.6% variances of
the response variable with five descriptors. The models depicted satis­
factory external predictivity with R2Pred of 0.859, r2test 2
m of 0.823 and Δrtest

Statistical resultsa
m
of 0.075. The maximum intercollinearity of the model was found as
0.495 indicating that the descriptors of the models are independent from
each other. We performed Y-randomization test with the model with

Δr2LOO
1000 run to determine the cR2p value as 0.881 suggesting the model is

m
itself unique and was not developed by chance. After confirming sta­
tistical robustness, we determined the applicability domain of the model

Ntraining: Number of data-points present in the training set; Ntest: Number of data-points present in the test set.
pIC50_glc = 1.055( ± 0.155)*TDB03i + 0.46( ± 0.051)*C-043 + 0.581( ± 0.02)*SaasN + 0.685( ± 0.127)
with the help of Williams plot, which is a plot of leverage vs standardized
residual. Three training set compounds and one test set compound were
found as structural outliers. Removal of the outlier from the test set,
improves the external predictivity of the model with R2Pred of 0.867. The
observed vs predicted activity plot and Williams plot of the model are
presented in Fig. S1 of supplementary materials. The relative signifi­
Summary of the SFS-MLR model developed with all descriptors for α-glucosidase inhibitors.

cance of each descriptor of 2D-QSAR model presented in Table 2 is


shown in Fig. 1.
As deduced from Table 2 and Fig. 1, the most significant descriptor of
the model was TDB03i, which belongs to 3D type of descriptors which
indicates 3D topological distance-based descriptors - lag 3 weighted by
*B09[S–Cl] + 0.529( ± 0.064)*CATS3D_07_DD + 0.44( ± 0.594)

ionization potential. This descriptor indicates that specific 3D geometry


and topology of the compounds play crucial role in determining the
biological activity of the compounds against α-glucosidase enzyme. At
the same time, the ionizability of these compounds significantly
contribute to the variations in the biological activity. The next most
significant descriptor is found to be B09[S–Cl], a 2-D atom pair
descriptor, which simply signifies the presence or absence of sulphur
and chlorine at topological distance of 9 [40]. Positive correlation of the
descriptor in the equation indicates that compounds with higher value of
this descriptor is found in some higher active compounds, as depicted in
Fig. 2. For instance, higher active compounds 6TT and 3TH possess atom
pairs S and Cl at topological distance of 9 whereas lower active com­
pounds 19DC and 12TZ do not possess the said atom pair, owing to
which, they may have lower inhibitory potential.
Equation (1)

The next most significant descriptor of the model is SaasN, an atom-


type E-state indices descriptor, which depicts the sum of total number of
nitrogen atom connected with one single bond along with two aromatic
bonds [41]. Being positively correlated with the response variable, the
greater value of this descriptor clearly favours the higher activity against
descriptors

α-glucosidase. The phenomenon is represented in Fig. 3 which shows


Descriptors

higher active compounds 9IZ and 15IZ with nitrogen containing aro­
Table 2

matic scaffolds (i.e., pyrazole moiety). On the other hand, the pair of
All

4
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 1. Relative significance of individual descriptor in the 2D-QSAR model for α-glucosidase inhibitors.

Fig. 2. Importance of the B09[S–Cl] descriptor with respect to selected dataset compounds.

compounds 5ID and 14TZ do not contain the said scaffolds, resulting in compounds lacked such structural features (i.e., CATS3D_07_DD = 0).
potentially lower inhibitory activity. The last descriptor, C-043, is an atom centred fragment descriptor
Chemically advanced template search (CATS) descriptors have been which is easily interpreted. The positive correlation of the descriptor
particularly useful in determining structural requirements in compounds depicts the presence of the fragments with higher biological activity as
with greater biological activity. The fourth most significant descriptor seen in Fig. 5. The C-043 atom centred fragment descriptor, represented
from the dataset was found to be CATS3D_07_DD, a 3D-pharmacophore by X–CR..X which depicts X as any electronegative atom (halogens or S,
descriptor that signifies topological distance of 7 between two hydrogen O, N etc.) while R can be any group linked through C [43]. For example,
bond donor features [42]. Having positive correlation with the pIC50, compounds 19TH and 10TT have atom centre C attached with electro­
higher values of these descriptors were found in compounds with greater negative atom while lower active compounds like 17ID and 4DC do not
inhibitory potentials. Indeed, some highly active compounds of the possess the aforementioned atom centre.
datasets were found to possess higher CATS3D_07_DD values (i.e., 1), as
exemplified in Fig. 4, whereas less and even moderately active

5
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 3. Significance of the SaasN descriptor with respect to selected dataset compounds.

Fig. 4. Significance of the CATS3D_07_DD descriptor with respect to selected dataset compounds.

3.2. 2D-QSAR models: α-amylase inhibitors independent of each other. The Y-randomization test of the model with
1000 runs was performed to establish whether the model was developed
A similar approach was taken to develop the 2-D QSAR model of the by luck, but the cR2p value of 0.862 suggests its unique nature. It is to be
α-amylase inhibitors. All AlvaDesc descriptors were used for generation observed that for both α-glucosidase and α-amylase 2D-QSAR model
of the 2D-QSAR model by deploying SFS-QSAR tool and GeneticAlgor­ development, 147 data points were considered for training set (Ntraining),
ithm v.4.1_2. Findings from these two different types of 2D-QSAR model while 40 data points were considered for test set (Ntest). Afterwards,
generation are given in Table 3, which suggests that GA based 2D-QSAR Williams plot was deployed to determine the applicability domain of the
model was better perceived in terms of statistical significance. developed model and only one training set compound was found as
From the developed models, it could be seen that the statistical structural outlier in the model.
quality of model produced by GA with all alvaDesc descriptors possesses As shown in Fig. 6, the most significant descriptor of Equation 2was
the most satisfactory statistical predictivity. This 2D-QSAR model with found to be RDF010m, which is a radial distribution function type
five descriptors is presented below in Table 4 and the plot between descriptor used for defining 3D molecular structure with atomic
observed and predicted values along with Williams plot have been numbers weighted by mass [40]. Negative correlation of this descriptor
provided in Fig. S2 of Supplementary material. with the dataset compounds imply that lower values can be associated
Equation in Table 4 demonstrates the 2D-QSAR models of α-amylase with compounds having greater α-amylase inhibitory potency. The
inhibitors provided satisfactory statistical quality that is similar to the presence of this descriptor clearly indicates that 3D-geometry weighed
model developed with α-glucosidase inhibitors. For example, the model by atomic mass should play significant roles in determining the bio­
afforded high internal predictivity as suggested by Q2LOO of 0.866. At the logical activity. The second most significant descriptor from Equation 2
same time, high value of r2LOO
m (=0.814) and low values of MAE (=0.14) was found to be CATS3D_07_DD that has been explained previously. This
as well as Δr2LOO
m (0.091) further indicate the same. As far as the external 3D-pharmacophore descriptor, being positively correlated to the dataset
predictivity is concerned, high R2Pred of 0.875 indicates that the model compounds imply that the 3D geometry of the compounds plays pivotal
achieved satisfactory predictivity, whereas the parameters determined role in determining inhibitory action of the compounds. Interestingly,
from r2m metrics also approved that. Maximum inter-collinearity of the the next significant descriptor of the model is SaasN, which was also
model was found to be 0.565 which suggested that the descriptors are found in the most predictive 2D-QSAR model of α-glucosidase [41]. This

6
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 5. Significance of the C-043 descriptor with respect to selected dataset compounds.

distance of 10 while compounds with relatively lower activity i.e. 16DC


Table 3
and 14TZ do not possess the atom pair as represented in Fig. 8. This
2D-QSAR analyses of α-amylase inhibitors with some interpretable and all de­
implies that having only S in the structure may not be enough to impart
scriptors (the most significant model is marked in bold).
greater biological activity. The last descriptor with the lowest signifi­
Methoda Scoreb Foldc Interpretable descriptorsd All descriptorse
cance from Equation 2is HVcpx, graph vertex complexity vertex, which
Q2LOO R2Pred Q2LOO R2Pred is an information indices descriptor calculated on the basis of contents of
SFS R2 0 0.756 0.704 0.881 0.775 molecules, based on the calculation of equivalence classes from the
NMAE 0 0.823 0.783 0.877 0.838 molecular graph [44].
NMPD 0 0.807 0.766 0.872 0.831
NMGD 0 0.800 0.780 0.872 0.831
R2 5 0.801 0.803 0.880 0.854 3.3. 3D-QSAR analyses
NMAE 5 0.777 0.768 0.875 0.838
NMPD 5 0.801 0.780 0.878 0.795
With rigid body alignment technique, the structures of both
NMGD 5 0.795 0.786 0.878 0.795
GA NA NA 0.815 0.808 0.866 0.875
α-glucosidase inhibitors and α-amylase inhibitors of the current dataset
were aligned. The aligned structures were thereafter used in developing
a: Feature selection methods, SFS: sequential forward selection, GA: Genetic 3D-QSAR models with Open3DQSAR tool. Two different feature selec­
algorithm, b: Scoring functions used in SFS, check the main text for full names, c:
tion techniques, such as FFD-SEL and UVE-PLS were utilized to develop
cross-validation methods used for SFS based feature selection, 0: No cross-
3D-QSAR models, the results of which are presented in Table 5.
validation, 5: 5-fold cross validation, d: Only selected categories of descriptors
with high mechanistic interpretability are used (see the main text), e: all alva­ The UVE-PLS technique provided the models with higher statistical
Desc descriptors were employed for model development. qualities for both α-glucosidase and α-amylase datasets. Satisfactory
internal predictivity of α-glucosidase 3D-QSAR model is perceived from
electrotopological state indices type descriptor shows positive correla­ Q2LOO of 0.773, Q2LTO of 0.773 and Q2LMO of 0.763. With 37 test set mol­
tion with the dataset compounds, revealing the presence of N atoms ecules, this model yielded R2Pred of 0.732. The model was subjected to
connected with one single along with two aromatic bonds being more randomization test that provided Q2s of 0.656 indicating that the model
significant towards α-amylase inhibitory action. The dataset compounds was not developed by luck. The contour maps of this model are pre­
like 8IZ and 12IZ have greater value corresponding to SaasN descriptor sented below in Fig. 9.
while less active compounds like 8ID and 7TZ show low value of the said From the 3D-QSAR model of α-glucosidase, it was found that the
descriptor as can be depicted from Fig. 7. Noticeably, these two de­ contribution of steric and electrostatic fields was 0.497 and 0.503,
scriptors are also seen in 2D-QSAR analysis for α-glucosidase inhibitors respectively. It is evident from the model that both fields have
implying that there is significant correlation existing between the contributed almost equally towards the model development. When the
α-amylase and α-glucosidase inhibition of the dataset compounds. most potent α-glucosidase inhibitor 19TH is compared with the least
The fourth most significant descriptor from Equation 2 has been potent compound 17DC, it was observed that 19TH could escape the
identified as positively correlated descriptor B10[C–S]. This 2D-atom steric unfavourable contour due to conformational flexibility that is less
pair descriptor depicts the presence or absence of C and S at topologi­ for 17DC. The methyl group on the thiazole ring in 19TH lies in very
cal distance of 09 [40]. Compounds with higher α-amylase inhibitory close proximity to the steric favourable contour maps, while the absence
potential i.e., 17TH and 11IZ contain S and C atoms at topological of bulky group/s in the steric unfavourable region also contributes to the
higher inhibitory potential of the ligand. A stark contrast may be seen in

7
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

case of the least active compound 17DC which is deprived of bulky


moieties in the steric favourable region and cyano groups can be found

= 0.814,
lying closely to steric unfavourable contour maps. Additionally, furan
moiety in 17DC can been seen inserted in the steric unfavourable con­

Ntraining = 147, R2 = 0.879, R2Adj = 0.875, F(5,147) = 205.71, Q2LOO = 0.866, MAE = 0.14, r2LOO
tour region which may result in low inhibitory potential of the ligand.

m
Upon studying the electrostatic contour maps, it has been found that
electron deficient thiazole moiety and the hydrogen present in hydroxyl
(OH) groups of the best active compound 19TH are nearer to electro­
positive contour maps while the electron-rich acetohydrazide skeleton
lies in very close proximity of the electronegative favourable contour
map contributing to the higher activity of the dataset ligand. Conse­

m = 0.095.
quently, the lowest active compound 17DC contains electron deficient
aromatic ring at electropositive contour map, resulting in low inhibitory
potential of the compound.
m = 0.832, Δrtest
2

In case of α-amylase inhibitors, satisfactory internal predictivity of


the 3D-QSAR model has been achieved from Q2LOO of 0.779, Q2LTO of
0.779 and Q2LMO of 0.769. For 37 test set molecules, R2Pred of the model
was calculated to be 0.738. The model was further subjected to
= 0.091, Ntest = 40, R2Pred = 0.875, r2test

randomization test that provided Q2s of 0.651 implying that the model
development was not a fluke. The contour maps of this model are pre­
sented below in Fig. 10.
For 3D-QSAR model of α-amylase, 0.53 and 0.47 were found to be
the contributions of the steric and electrostatic fields, respectively. From
this model, it can be comprehended that contribution of both the fields
are equally important for the model generation though steric contribu­
Statistical resultsa

tion slightly higher. Upon observing the contour maps of the most potent
α-amylase inhibitor 17TH, it can be observed that the bulky 4-methyl
thiazole moiety is inserted into the steric favourable region of the con­
Δr2LOO

tour map while the rest of the remaining bulky structure escapes the
m

steric unfavourable region. This might be considered as one of the major


reasons behind the higher activity of the compound. On the contrary, the
pIC50_aml=0.442( ± 0.045)*B10[C–S] + 0.551( ± 0.068)*CATS3D_07_DD + 0.444( ± 0.023)*SaasN + (− 0.93)(

lowest active ligand 26DC with α-amylase inhibitory potential contains


Ntraining: Number of data-points present in the training set; Ntest: Number of data-points present in the test set.

no bulky moieties in the steric favourable contour region. Additionally,


bulky aromatic moieties can be seen inserted into the steric unfav­
ourable region of the contour maps which contributes to its low activity.
The electrostatic contour maps also reveal significant contributions
regarding the α-amylase inhibitory potential of the dataset ligands. As
seen in Fig. 10, electron-deficient thiazole moiety and electron-deficient
groups of the highest active ligand 17TH are close to electropositive
Summary of the GA-MLR models developed with all descriptors for α-amylase inhibitors.

contour map while the electron rich hydrazide skeleton and hydroxyl
(OH) group are almost embedded in the electronegative contour region.
A contrasting structural attribute can be observed in the lowest active
compound 26DC which lacks the favourable electrostatic characteristics
± 0.091)*RDF010 m + 0.217( ± 0.079)*HVcpx + 4.559( ± 0.238)

to induce higher inhibitory potential.

3.4. Molecular docking analyses

No crystallographic structure of Saccharomyces cerevisiae of


α-glucosidase enzyme is available yet. The homology model of Saccha­
romyces cerevisiae α-glucosidase was developed with GMQE (Global
Model Quality Estimate) of 0.95 and QMEANDisCo of 0.91 indicating
that the model was produced with satisfactory quality. Furthermore,
when assessed with Molprobity, a score of 1.11 was obtained that clearly
indicates that model fulfils most of the criteria for being a homology
model with good structural qualities. In spite of having a slightly higher
but acceptable clash score of 1.39, favoured region of Ramachandran
plot was found as 96.36%. The Ramachandran plot of the homology
model developed for Saccharomyces cerevisiae α-glucosidase is presented
Equation (2)

in Fig. S3 of supplementary materials.


Two dataset compounds (17TH and 7DC) with significant variations
in their biological activity against these enzymes were used for docking
analyses with these two enzymes and interactions of their best docked
descriptors

poses are shown in Fig. 11.


Descriptors

The higher active compound 17TH depicted binding energies of


Table 4

− 9.62 kcal/mol and − 9.78 kcal/mol with α-glucosidase and α-amylase


All

enzymes, respectively. On the other hand, for the lower active


a

8
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 6. Relative importance of each descriptor in the 2D-QSAR model for α-amylase inhibitors.

Fig. 7. Significance of the SaasN descriptor with respect to selected dataset compounds.

compound 7DC, these values are − 6.91 kcal/mol and − 7.42 kcal/mol, the hydroxyl groups of phenyl ring were clearly depicted in 3D-QSAR
respectively. Therefore, the docking methodology at least discriminates models developed with α-glucosidase and α-amylase enzymes. The
the binding behaviours of these two dataset compounds. However, the involvement of the acetohydrazide moiety of 17TH for polar in­
main objective of performing the docking is to compare the ligand- teractions were also insinuated by the 3D-QSAR models because of the
receptor interactions with the mechanistic interpretations obtained closeness of electrostatic contours near this moiety. As per the 3D-QSAR
from 2D- and 3D-QSAR methodologies. The significance of the 4-meth­ models, unfavourable steric interactions played a significant role in
ylthiazole scaffold and hydroxyl groups of phenyl moiety was obtained lowering the activity of 7DC whereas molecular docking suggested that
from 2D-QSAR analyses and from the docking interactions these in­ a comparatively lower number of interactions at the binding site may be
teractions are clearly found to be important. The hydroxyl groups of responsible for relatively low binding energy of such compounds against
17TH formed hydrogen bond interactions with the binding site amino these enzymes. The central aromatic moiety of 7DC failed to show sig­
acids while the pi-alkyl interactions (that depends both on the hydro­ nificant interactions with the receptors whereas the interactions of
phobic and electronic environment of the aromatic ring) were observed amino as well as cyano groups varied considerably. Among three aro­
between 4-methylthiazole scaffold and surrounding amino acids. matic moieties, only pyridine moiety interacted strongly with sur­
Noticeably, 3D-QSAR modelling hinted that both steric and electrostatic rounding amino acids.
interactions of 4-methylthiazole scaffold (of 19TH, which is close to
17TH both structurally and biologically) are important for higher ac­
tivity against these enzymes. Similarly, the electronic contributions of

9
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 8. Significance of the B10[C–S] descriptor with respect to selected dataset compounds.

with small number of data-points. Investigations carried out with rela­


Table 5 tively large number of data-points were based on the classification an­
Statistical results of 3D-QSAR analyses of α-glucosidase and α-amylase
alyses. The current study describes the regression-based validated and
inhibitors.
predictive 2D- and 3D-QSAR models in order to understand the struc­
Parameter* α-glucosidase α-amylase tural requirements of 187 compounds reported as α-glucosidase and
FFD-SEL UVE-PLS FFD-SEL UVE-PLS α-amylase inhibitors by the same group of research investigators. A few
PC 2 2 2 2 interesting facts encouraged us to perform receptor-independent and
NTraining 150 150 150 150 ligand-based cheminformatic analyses. First, no X-ray crystal structure
F-test 318.36 396.50 285.51 411.67 of α-glucosidase is available that leaves better scopes for ligand-based
R2/SDEC 0.814/0.219 0.844/0.201 0.795/0.231 0.848/0.198 methodologies. Second, the inhibitory potencies against these two en­
Q2LOO/SDEP 0.730/0.264 0.773/0.242 0.709/0.275 0.779/0.239
Q2LTO/SDEP 0.730/0.264 0.773/0.242 0.709/0.275 0.779/0.239
zymes are highly correlated and each dataset compound may thus in fact
Q2LMO/SDEP 0.718/0.269 0.763/0.247 0.699/0.280 0.769/0.245 be considered as dual inhibitors of α-glucosidase and α-amylase.
NTest 37 37 37 37 Nevertheless, separate cheminformatic models were developed for these
R2Pred/SDEP 0.705/0.265 0.732/0.252 0.693/0.280 0.738/0.259 activities and this is why, the models developed here may be considered
Q2s ND 0.545 ND 0.651
significant for both these activities. The 2D-QSAR models were devel­
oped with significantly reliable statistical qualities with a smaller
3.5. Design of new compounds number of descriptors and at the same time, these specifically pointed
out the structural attributes responsible for higher inhibitory potentials
The current work mainly focuses on the development of linear pre­ against these enzymes. It was found that apart from specific 2D- and
dictive models to understand structural requirements of diverse sets of 3D-topologies, the physicochemical properties such as mass and ioni­
ligands for their activity against α-amylase and α-glucosidase activity. zation potential play crucial role in determining the activity of the
This is why we performed feature selection techniques in 2D-QSAR in compounds. Considerably high statistical predictions were obtained
order to collect the most significant descriptors to describe structural from the 3D-QSAR models as well and the contour maps generated in the
features responsible for higher inhibition. Nevertheless, these models models justified the variations observed in the activities among the
may be utilized to design new molecules. In this work, we demonstrate α-glucosidase and α-amylase inhibitors. Moreover, we found consis­
one such example NC1 (Fig. 12). It was observed that the most potent tencies among the interpretations of 2D- and 3D-QSAR models. For
α-glucosidase inhibitor of the dataset had low value of SaasN descriptor example, the importance of hydroxyl groups and acetohydrazide moi­
whereas it had CATS_07_DD value of 1. We designed a new compound eties as well as topological distance between two aromatic moieties in
which increased the value of SaasN at the expense of CATS_07_DD since higher active compounds (19TH, 17TH and 7TH) are shown by both the
the former had slightly higher significance than the latter. As expected, 2D-QSAR and 3D-QSAR models. In the α-glucosidase 2D-QSAR model,
the predicted pIC50 value against α-glucosidase improved and at the the descriptor weighed by ionization potential (i.e., TDB03i) and in the
same time, the designed molecule NC1 remained inside the applicability α-amylase 2D-QSAR model, the descriptor weighted by atomic mass (i.
domain of the model. When this compound was docked with α-gluco­ e., RDF010m) remained the most crucial descriptors. Such information
sidase, a binding energy of − 8.74 kcal/mol was obtained. is reflected by the fact that in their 3D-QSAR models, steric and elec­
trostatic interactions were found to be equally important in determining
the biological activity. More interestingly, 3D-QSAR models highlighted
4. Conclusion
importance of interactions with unfavourable steric contours with bulky
atoms for lowering the activity and this information is consistent with
Previously, a number of investigations described cheminformatics
the fact that RDF010m appeared to have a negative influence on
analyses of α -glucosidase and α-amylase inhibitors [45–50]. These in­
inhibitory potency against α-amylase. Additionally, this work reports
vestigations were either reported with only one of these enzymes and/or

10
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 9. Contour maps obtained from the best 3D-QSAR model for α-glucosidase data (Green: Steric favourable, Yellow: Steric unfavourable, Blue: Electropositive
favourable; Red: Electronegative favourable). (A) Steric maps of the best active compound 19TH, (B) Steric maps of the least active compound 17DC, (C) Electrostatic
maps of the best active compound 19TH and (D) Electrostatic maps of the least active compound 17DC. (For interpretation of the references to colour in this figure
legend, the reader is referred to the Web version of this article.)

Fig. 10. Contour maps obtained from the best 3D-QSAR model for α-amylase data (Green: Steric favourable, Yellow: Steric unfavourable, Blue: Electropositive
favourable; Red: Electronegative favourable). (A) Steric maps of the best active compound 17TH, (B) Steric maps of the least active compound 26DC, (C) Electrostatic
maps of the best active compound 17TH and (D) Electrostatic maps of the least active compound 26DC. (For interpretation of the references to colour in this figure
legend, the reader is referred to the Web version of this article.)

molecular docking with selected number of ligands and ligand receptor α-amylase inhibitors. More importantly, the current work is entirely
interactions complied well with QSAR interpretations. Even though this performed by using non-commercial open-access tools to ensure easy
work emphasized mainly on the structural characterization, as it is accessibility and reproducibility of the investigation which may help
shown in the design of a new compound NC1, this investigation should researchers throughout the world to work more on drug design and
serve as important guidelines for the design of future α-glucosidase and discovery.

11
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

Fig. 11. Binding interactions between α-glucosidase enzyme and dataset compounds 17TH (A) and 7DC (B) and between α-amylase and dataset compounds 17TH
(C) and 7DC (D).

Declaration of competing interest

The authors declare that they have no known competing financial


interests or personal relationships that could have appeared to influence
the work reported in this paper.

Data availability

I have shared the data at the Attach File step of submission

Acknowledgement

The authors are thankful to Dr. B. C. Roy College of Pharmacy and


Allied Health Sciences for providing infrastructures and support for the
current work.

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.


org/10.1016/j.jmgm.2023.108640.

References

[1] J. de F. Maraschin, Classification of diabetes, Adv. Exp. Med. Biol. 771 (2012)
12–19.
[2] B. Fletcher, M. Gulanick, C. Lamendola, Risk factors for type 2 diabetes mellitus,
J. Cardiovasc. Nurs. 16 (2002) 17–23.
Fig. 12. Structure of the newly designed compound NC1. [3] American Diabetes Association, 2. Classification and diagnosis of diabetes:
standards of medical care in diabetes-2020, Diabetes Care 43 (Suppl 1) (2020)
S14–S31, https://doi.org/10.2337/dc20-S002.

12
S. Mitra et al. Journal of Molecular Graphics and Modelling 126 (2024) 108640

[4] F. Giacco, M. Brownlee, Oxidative stress and diabetic complications, Circ. Res. 107 [25] P. Ambure, R.B. Aher, A. Gajewicz, T. Puzyn, K. Roy, “NanoBRIDGES” software:
(2010) 1058–1070. open access tools to perform QSAR and nano-QSAR modelling, Chemometr. Intell.
[5] M.A. Darenskaya, L.I. Kolesnikova, S.I. Kolesnikov, Oxidative stress: pathogenetic Lab. Syst. 147 (2015) 1–13.
role in diabetes mellitus and its complications and therapeutic approaches to [26] I.V. Tetko, V.Y. Tanchuk, A.E. Villa, Prediction of n-octanol/water partition
correction, Bull. Exp. Biol. Med. 171 (2021) 179–189. coefficients from PHYSPROP database using artificial neural networks and E-state
[6] U. Ghani, Re-exploring promising α-glucosidase inhibitors for potential indices, J. Chem. Inf. Comput. 41 (2011) 1407–1421.
development into oral anti-diabetic drugs: finding needle in the haystack, Eur. J. [27] A.K. Halder, Finding the structural requirements of diverse HIV-1 protease
Med. Chem. 103 (2015) 133–162. inhibitors using multiple QSAR modelling for lead identification, SAR QSAR
[7] J.J. DiNicolantonio, J. Bhutani, J.H. O’Keefe, Acarbose: safe and effective for Environ. Res. 29 (2018) 911–933.
lowering postprandial hyperglycaemia and improving cardiovascular outcomes, [28] A. Golbraikh, A. Tropsha, Beware of q2, J. Mol. Graph. 20 (2002) 269–276.
Open heart 2 (2015), e000327. [29] P.P. Roy, S. Paul, I. Mitra, K. Roy, On two novel parameters for validation of
[8] N. Kaur, V. Kumar, S.K. Nayak, P. Wadhwa, P. Kaur, S.K. Sahu, Alpha-amylase as predictive QSAR models, Molecules 14 (2009) 1660–1701.
molecular target for treatment of diabetes mellitus: a comprehensive review, [30] W. Yoo, R. Mayberry, S. Bae, K. Singh, Q.P. He, J.W. Lillard, A study of effects of
Chem. Biol. Drug Des. 98 (2021) 539–560, https://doi.org/10.1111/cbdd.13909. MultiCollinearity in the multivariable analysis, Int. J. Appl. Sci. Technol. 4 (2014)
[9] S. Chahal, J. Punia, P. Rani, R. Singh, P.K. Mayank, R. Kataria, G. Joshi, J. Sindhu, 9–19.
Development of thiazole-appended novel hydrazones as a new class of α-amylase [31] P.K. Ojha, K. Roy, Comparative QSARs for antimalarial endochins: importance of
inhibitors with anticancer assets: an in silico and in vitro approach, RSC med. 14 descriptor-thinning and noise reduction prior to feature selection, Chemometr.
(2023) 757–781. Intell. Lab. Syst. 109 (2011) 146–161.
[10] K. Date, A. Satoh, K. Iida, H. Ogawa, Pancreatic α-amylase controls glucose [32] K. Roy, S. Kar, P. Ambure, On a simple approach for determining applicability
assimilation by duodenal retrieval through N-Glycan-specific binding, endocytosis, domain of QSAR models, Chemometr. Intell. Lab. Syst. 145 (2015) 22–29.
and degradation, J. Biol. Chem. 290 (2015) 17439–17450. [33] P. Tosco, T. Balle, F. Shiri, Open3DALIGN: an open-source software aimed at
[11] R. Singh, P. Kumar, J. Sindhu, M. Devi, A. Kumar, S. Lal, D. Singh, Parsing unsupervised ligand alignment, J. Comput. Aided Mol. Des. 25 (2011) 777–783.
structural fragments of thiazolidin-4-one based α-amylase inhibitors: a combined [34] P. Tosco, T. Balle, Open3DQSAR: a new open-source software aimed at high-
approach employing in vitro colorimetric screening and GA-MLR based QSAR throughput chemometric analysis of molecular interaction fields, J. Mol. Model. 17
modelling supported by molecular docking, molecular dynamics simulation and (2011) 201–208, https://doi.org/10.1007/s00894-010-0684-x.
ADMET studies, Comput. Biol. Med. 157 (2023), 106776. [35] A. Waterhouse, M. Bertoni, S. Bienert, G. Studer, G. Tauriello, R. Gumienny, F.
[12] E. Oyeye, Kanwal, K.M. Khan, S. Chigurupati, A. Wadood, A.U. Rehman, T. Heer, T.A.P. de Beer, C. Rempfer, L. Bordoli, R. Lepore, T. Schwede, SWISS-
S. Perveen, M.K. Maharajan, S. Shamim, S. Hameed, S.A. Aboaba, M. Taha, MODEL: homology modelling of protein structures and complexes, Nucleic Acids
Syntheses, in vitro α-amylase and α-glucosidase dual inhibitory activities of 4- Res. 46 (2018) W296–W303.
amino-1,2,4-triazole derivatives their molecular docking and kinetic studies, [36] UniProt Consortium, UniProt: the universal protein knowledgebase in 2023,
Bioorg. Med. Chem. 28 (2020), 115467. Nucleic Acids Res. 51 (2023) D523–D531.
[13] S. Hameed, Kanwal, F. Seraj, R. Rafique, S. Chigurupati, A. Wadood, A.U. Rehman, [37] G. Wang, J. Z., Peng, X. Li Wang, J. Li, Synthesis, in vitro evaluation and molecular
V. Venugopal, U. Salar, M. Taha, K.M. Khan, Synthesis of benzotriazoles docking studies of novel triazine-triazole derivatives as potential α-glucosidase
derivatives and their dual potential as α-amylase and α-glucosidase inhibitors in inhibitors, Eur. J. Med. Chem. 125 (2017) 423–429.
vitro: structure-activity relationship, molecular docking, and kinetic studies, Eur. J. [38] J. Christopher, et al., MolProbity: more and better reference data for improved all-
Med. Chem. 183 (2019), 111677, 2019. atom structure validation, Protein Sci. 27 (2018) 293–315.
[14] R. Rafique, K.M. Khan, Kanwal Arshia, S. Chigurupati, A. Wadood, A.U. Rehman, [39] G.M. Morris, R. Huey, W. Lindstrom, M.F. Sanner, R.K. Belew, D.S. Goodsell, A.
A. Karunanidhi, S. Hameed, M. Taha, M. Al-Rashida, Synthesis of new indazole J. Olson, AutoDock4 and AutoDockTools4: automated docking with selective
based dual inhibitors of α-glucosidase and α-amylase enzymes, their in vitro, in receptor flexibility, J. Comput. Chem. 30 (2009) 2785–2791.
silico and kinetics studies, Bioorg. Chem. 94 (2020), 103195. [40] R. Todeschini, V. Consonni, Handbook of Molecular Descriptors, John Wiley &
[15] S. Shamim, K.M. Khan, N. Ullah, S. Chigurupati, A. Wadood, A.U. Rehman, M. Ali, Sons, Ltd, WILEY-VCH, Germany, 2000, 2000.
U. Salar, A. Alhowail, M. Taha, S. Perveen, Synthesis and screening of (E)-3-(2- [41] K. Roy, I. Mitra, Electrotopological state atom (E-state) index in drug design, QSAR,
benzylidenehydrazinyl)-5,6-diphenyl-1,2,4-triazine analogs as novel dual property prediction and toxicity assessment, Curr. Comput. Aided Drug Des. 8
inhibitors of α-amylase and α-glucosidase, Bioorg. Chem. 101 (2020), 103979. (2012) 135–158, https://doi.org/10.2174/157340912800492366.
[16] S. Faiza, A. Kanwal, K.M. Khan, S. Chigurupati, Y. Andriani, M. Solangi, S. Hameed, [42] M. Reutlinger, C.P. Koch, D. Reker, N. Todoroff, P. Schneider, T. Rodrigues,
A.A. Monem, A. Hafez, F. Begum, M.A. Lodhi, M. Taha, F. Rahim, T.S. G. Schneider, Chemically advanced template search (CATS) for scaffold-hopping
T. Muhammad, S. Perveen, Dicyanoanilines as potential and dual inhibitors of and prospective target prediction for ‘orphan’ molecules, Mol. Inform. 32 (2013)
α-amylase and α-glucosidase enzymes: synthesis, characterization, in vitro, in 133–138, https://doi.org/10.1002/minf.201200141.
silico, and kinetics studies, Arab, J. Chem. 15 (2022), 103651. [43] R. Kühne, R.U. Ebert, G. Schüürmann, Chemical domain of QSAR models from
[17] A.N. Kawde, M. Taha, R.S. Alansari, N.B. Almandil, E.H. Anouar, N. Uddin, atom-centered fragments, J. Chem. Inf. Model. 49 (2009) 2660–2669.
F. Rahim, S. Chigurupati, M. Nawaz, S. Hayat, M. Ibrahim, P.K. Elakurthy, [44] M.E. Di Ianni, M.E. Del Valle, A.V. Enrique, M.A. Rosella, F. Bruno, L.E. Bruno-
V. Vijayan, M. Morsy, H. Ibrahim, N. Baig, K.M. Khan, Exploring efficacy of indole- Blanch, A. Talevi, Computer-aided identification of anticonvulsant effect of natural
based dual inhibitors for α-glucosidase and α-amylase enzymes: in silico, nonnutritive sweeteners stevioside and rebaudioside A, Assay Drug Dev. Technol.
biochemical and kinetic studies, Int. J. Biol. Macromol. 154 (2020) 217–232. 13 (2015) 313–318.
[18] A.A. Khan, H. Ullah, F. Rahim, M. Taha, F. Khan, W. Rehman, A. Wadood, K. [45] N.S.H. Moorthy, M.J. Ramos, P.A. Fernandes, Comparative structural analysis of
M. Khan, Synthesis, in vitro α-glucosidase and α-amylase activities, and an in silico α-glucosidase inhibitors on difference species: a computational study, Arch.
molecular docking study of triazinoindole-thiazolidinone hybrid derivatives, Pharmazie 345 (2012) 265–274.
Chem. Data Collect 45 (2023), 101035, https://doi.org/10.1016/j. [46] M. Duhan, J. Sindhu, P. Kumar, M. Devi, R. Singh, R. Kumar, S. Lal, A. Kumar,
cdc.2023.101035. S. Kumar, K. Hussain, Quantitative structure activity relationship studies of novel
[19] M. Taha, S. Hayat, F. Rahim, N. Uddin, A. Wadood, M. Nawaz, M. Gollapalli, A. hydrazone derivatives as α-amylase inhibitors with index of ideality of correlation,
U. Rehman, K.M. Khan, R.K. Farooq, Exploring thiazole-based Schiff base analogs J. Biomol. Struct. Dyn. 40 (2020) 4933–4953.
as potent α-glucosidase and α-amylase inhibitor: their synthesis and in-silico study, [47] K. Diéguez-Santana, A. Puris, O.M. Rivera-Borroto, G.M. Casanola-Martin,
J. Mol. Struct. 1287 (2023), 135672. B. Rasulev, H. González-Díaz, A fuzzy system classification approach for QSAR
[20] ChemAxon Standardizer, Version 15.9.14.0 Software, ChemAxon, Budapest, modeling of α-amylase and α-glucosidase inhibitors, Curr. Comput. Aided Drug
Hungary, 2010. Des. 18 (2022) 469–479.
[21] A. Mauri, AlvaDesc: A Tool to Calculate and Analyze Molecular Descriptors and [48] K. Dieguez-Santana, H. Pham-The, O.M. Rivera-Borroto, A. Puris, H. Le-Thi-Thu, G.
Fingerprints in Ecotoxicological QSARs, 2020, pp. 801–820. M. Casanola-Martin, A two QSAR way for antidiabetic agents targeting using
[22] I. Sushko, et al., Online chemical modeling environment (OCHEM): web platform α-amylase and α-glucosidase inhibitors: model parameters settings in artificial
for data storage, model development and publishing of chemical information, intelligence techniques, Lett. Drug Des. Discov. 14 (2017) 8.
J. Comput. Aided Mol. Des. 25 (2011) 533–554. [49] S. Ahmadi, Z. Moradi, A. Kumar, A. Almasirad, SMILES-based QSAR and molecular
[23] J. Sadowski, J. Gasteiger, G. Klebe, Comparison of automatic three-dimensional docking study of xanthone derivatives as α-glucosidase inhibitors, J. Recept. Signal
model builders using 639 X-ray structures, J. Chem. Inf. Model. 34 (2002) Transduct. Res. 42 (2021) 361–372.
1000–1008. [50] O. Abchir, O. Daoui, S. Belaidi, M. Ouassaf, F.A. Qais, S. ElKhattabi, S. Belaaouad,
[24] A.K. Halder, A.H.S. Delgado, M.N.D.S. Cordeiro, First multi-target QSAR model for S. Chtita, Design of novel benzimidazole derivatives as potential α-amylase
predicting the cytotoxicity of acrylic acid-based dental monomers, Dent. Mater. J. inhibitors using QSAR, pharmacokinetics, molecular docking, and molecular
38 (2022) 333–346. dynamics simulation studies, J. Mol. Model. 28 (2022) 4.

13

You might also like