ArticlePDF Available

Molecular Docking Simulation Studies, Toxicity Study, Bioactivity Prediction and Structure Activity Relationship Reveals Rutin as a Potential Inhibitor of SARS-CoV-2 3CL pro

Authors:

Abstract and Figures

Coronavirus disease 2019 (COVID-19) pandemic has infected billions and has killed millions of people. Despite the advancement in drugs and vaccines, the COVID-19 pandemic rages on. Bioactive compounds of medicinal plants with antiviral properties such as flavonoids might be useful as phytotherapy for COVID-19. The present study aims to evaluate 16 flavonoids and identify a potential inhibitor for the main protease of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 3CL pro) which is responsible for replication of the viral genome in a host cell. Among the 16 flavonoids, molecular docking simulation studies reveal that rutin has the highest binding affinity towards chain A, chain B, chain C, and chain D of SARS-CoV-2 3CL pro. Rutin formed conventional hydrogen bonds and non-covalent hydrophobic bonds with important amino acid residues at the active binding site of SARS-CoV-2 3CL pro. Also, rutin was computed to be non-toxic. Rutin had a better drug-likeness score and protease inhibition than the co-crystal inhibitor of SARS-CoV-2 3CL pro. The structure-activity relationship revealed important moieties of rutin that might be essential for the inhibition of SARS-CoV-2 3CL pro. The in-silico evidence suggests rutin may inhibit SARS-CoV-2 3CL pro and advocates its use as a phytotherapy for COVID-19.
Content may be subject to copyright.
Volume 65, Issue 5, 2021
Journal of Scientific Research
of
The Banaras Hindu University
96
DOI: 10.37398/JSR.2021.650511
Abstract: Coronavirus disease 2019 (COVID-19) pandemic has
infected billions and has killed millions of people. Despite the
advancement in drugs and vaccines, the COVID-19 pandemic rages
on. Bioactive compounds of medicinal plants with antiviral
properties such as flavonoids might be useful as phytotherapy for
COVID-19. The present study aims to evaluate 16 flavonoids and
identify a potential inhibitor for the main protease of severe acute
respiratory syndrome coronavirus 2 (SARS-CoV-2 3CL pro) which
is responsible for replication of the viral genome in a host cell.
Among the 16 flavonoids, molecular docking simulation studies
reveal that rutin has the highest binding affinity towards chain A,
chain B, chain C, and chain D of SARS-CoV-2 3CL pro. Rutin
formed conventional hydrogen bonds and non-covalent
hydrophobic bonds with important amino acid residues at the
active binding site of SARS-CoV-2 3CL pro. Also, rutin was
computed to be non-toxic. Rutin had a better drug-likeness score
and protease inhibition than the co-crystal inhibitor of SARS-CoV-
2 3CL pro. The structure-activity relationship revealed important
moieties of rutin that might be essential for the inhibition of SARS-
CoV-2 3CL pro. The in-silico evidence suggests rutin may inhibit
SARS-CoV-2 3CL pro and advocates its use as a phytotherapy for
COVID-19.
Index Terms: COVID-19, Molecular docking, Rutin, SARS-CoV-
2, Structure-activity relationship
I. INTRODUCTION
At present, the world is facing an unprecedented physical,
mental and emotional burden due to the ongoing coronavirus
disease 2019 (COVID-19) pandemic that is caused by severe
acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (Hu et
al., 2020). As of 11th May 2021, the COVID-19 pandemic has
*
Corresponding Author
infected 1,58,651,638 people and has killed more than 3.29
million people (WHO, 2021a). Many drugs are currently being
used to treat COVID-19 (Singh et al., 2021). Also, many
vaccines are currently being used to treat COVID-19 infection
(WHO, 2021b).
Despite the recent advancements in pharmacotherapy and
vaccines for COVID-19, several countries remained severely
affected by COVID-19. Problems such as adverse effects,
toxicity, and drug interactions seem to be associated with
repurposed drugs that are intended for the treatment of COVID-
19 (Shende et al., 2020). Therefore, additional therapies for
COVID-19 that are safe and effective seem to be the need of the
hour (Shende et al., 2020). Interestingly, bioactive compounds
from medicinal plants might be able to offer a safe and effective
therapy for COVID-19 (Islam et al., 2020; Antonio et al., 2020).
This is because bioactive compounds have been successfully
used to treat many viral diseases (Narkhede et al., 2020; Kim et
al., 2014; Tahir Ul Qamar et al., 2019). Thus, medicinal plants
and their bioactive compounds offer promising phytotherapy for
COVID-19 (Narkhede et al., 2020).
The main protease of SARS-CoV-2 is one of the most
favorable drug targets to treat COVID-19 (Jin et al., 2020;
Prajapat et al., 2020; Robson, 2020). This is because the main
protease of SARS-CoV-2 is essential for the replication of viral
RNA within the host cell (Jin et al., 2020). Once the viral RNA
of SARS-CoV-2 enters a host cell, the polyproteins that are
translated from the viral genome are cleaved by the main
protease of SARS-CoV-2 which is followed by transcription and
replication of the viral genome (Huang et al., 2020; Jin et al.,
2020; Gildenhuys, 2020).
Flavonoids have been extensively studied for their antiviral
properties (Zakaryan et al., 2017; Ninfali et al., 2020). In the
current study, the inhibitory potential of 16 flavonoids against
the main protease of SARS-CoV-2 was evaluated with in-silico
Molecular Docking Simulation Studies,
Toxicity Study, Bioactivity Prediction, and
Structure-Activity Relationship Reveals Rutin
as a Potential Inhibitor of SARS-CoV-2 3CL pro
James H. Zothantluanga*1
Journal of Scientific Research, Volume 65, Issue 5, 2021
97
Institute of Science, BHU Varanasi, India
techniques.
II. MATERIALS AND METHODS
A. Retrieval of a target protein
The main protease of SARS-CoV-2 was used as the target
protein in the study. The crystal structure of SARS-CoV-2 3CL
pro with a protein data bank (PDB) ID of 6M2N was retrieved
from the Research Collaboratory for Structural Bioinformatics
(RCSB)-PDB website (RCSB-PDB, 2021). The main protease of
SARS-CoV-2 was found to have four chains (chain A, chain B,
chain C, and chain D). The co-crystal inhibitor (CCI) was also
identified and retrieved from the PubChem database (PubChem,
2021).
B. Pre-processing of protein
The target protein was pre-processed with BIOVIA Discovery
Studio Visualizer v20.1.0.19295 software. All the chains (A, B,
C, and D) of the target protein were individually processed.
Initially, water was removed from the target protein. Then, the
active binding site was defined with the ‘Define and edit binding
site’ feature of the Discovery Studio visualizer software. The 3-
dimensional attributes (XYZ coordinates) of the active binding
site were identified and saved for further use. Then, the CCI of
the target protein was removed. Finally, polar hydrogen was
added to the target protein with Discovery Studio visualizer
software. The processed target protein was saved in PDB format
for future use.
C. Preparation of compound library
The 3-dimensional (3D) structures of 16 flavonoids were
retrieved from the PubChem database (PubChem, 2021). The
structure of each flavonoid was saved in structure-data file
format and their PubChem compound ID (CID) was also saved.
D. Energy minimization of ligands
Energy minimization of ligands was carried out on the PyRx
0.8 tool. To minimize the energy of the ligands used in the
present study, the default parameters of the PyRx 0.8 tool was
used (Force field = Universal force field; Optimization algorithm
= Conjugate gradients; Total no. of steps = 200; No. of steps for
update = 1; Stop if energy difference is less than = 0.1).
E. Detection and coordinates of the active binding site
In all the four chains (A, B, C, and D) of SARS-CoV-2 3CL
pro, a CCI was found to be present at the active binding site. At
first, chain A of the pre-processed target protein that was
initially prepared with the Discovery Studio visualizer software
was loaded onto the PyRx virtual screening platform. This
particular protein was then converted into ‘pdbqt.’ Format with
the PyRx software.
Then, the original target protein complexed with the CCI
which did not undergo any processing was also loaded onto the
PyRx virtual screening platform. The chains that made up
protein complexed with the CCI were revealed by expanding the
protein. Initially, chain B, chain C, and, chain D were removed
from the scene. Then, chain A of the pre-processed target protein
and chain A of the target protein complexed with the CCI was
automatically made to superimpose by the PyRx tool.
Following this, the chain A of the target protein complexed
with the CCI that was not pre-processed was expanded and the
sequence of all the amino acids residues that made up the chain
including the CCI (3WL) was revealed. The location of the CCI
present at the active binding site of the target protein complexed
with the CCI was detected by labelling the atoms of the CCI.
Then, the 3D affinity gird box was carefully made to align
with the central part of the CCI to cover the entire active site
residues by considering (i) the active binding site coordinates of
the pre-processed target protein previously identified with the
Discovery Studio visualizer software and (ii) the location of the
CCI that was made visible on the protein. In this way, the entire
amino acid residues present at the active binding site of the
protein were covered by the 3D affinity grid box.
Finally, the chain A of the target protein complexed with the
CCI that was not pre-processed was removed from the scene.
However, the chain A of the pre-processed target protein was
kept for MDSS. The active binding site of chain B, chain C, and,
chain D of SARS-CoV-2 3CL pro was detected similarly. The
size of the 3D affinity grid box was kept default at 25 Å for the
entire simulation process. The coordinates of the active binding
site of each chain are described below.
1) Chain A of SARS-CoV-2 3CL pro
In the Vina search space, the coordinates at the active binding
site of the protein were x = -33.9203241113, y = -
64.4918230137, z = 40.5838306859.
2) Chain B of SARS-CoV-2 3CL pro
In the Vina search space, the coordinates at the active binding
site of the protein was x = -48.4728236411, y = -
1.05170930781, z = -6.07895092506.
3) Chain C of SARS-CoV-2 3CL pro
In the Vina search space, the coordinates at the active binding
site of the protein were x = -40.7113525071, y = -
21.7108783003, z = 55.108989351.
4) Chain D of SARS-CoV-2 3CL pro
In the Vina search space, the coordinates at the active binding
site of the protein were x = -61.8523694685, y = -
35.3575321951, z = 22.9711975727.
F. Molecular docking simulation studies
In the present study, molecular docking simulation studies
(MDSS) were carried out for 16 flavonoids and the CCI using
AutoDock Vina (Trott & Olson, 2010) on a virtual screening
tool known as PyRx 0.8 tool. Other than those specified, the
standard operating procedure of PyRx tool was followed for the
MDSS (Dallakyan & Olson, 2015).
The flavonoid which showed the highest binding affinity
towards chain A, chain B, chain C, and chain D of SARS-CoV-2
3CL pro was selected for further analysis.
G. Visualization and analysis of ligand interactions
The ligand interactions formed by the flavonoid with the
amino acid residues at the active binding site of SARS-CoV-2
3CL pro were analyzed with BIOVIA Discovery Studio
Visualizer v20.1.0.19295 software. Visualization of the ligand
interactions was done in a 2-dimensional (2D) and 3D view. The
3D binding pose of the flavonoid and the CCI at the active
binding site of the target protein was generated using PyMOL
Journal of Scientific Research, Volume 65, Issue 5, 2021
98
Institute of Science, BHU Varanasi, India
molecular graphics system, Version 2.4.1 Schrodinger, LLC
software. The ligand interactions between the CCI and SARS-
CoV-2 3CL pro were also visualized and analyzed using the
same technique as described above.
H. Validation of molecular docking via re-docking,
superimposing, and RMSD calculation
The molecular docking protocol used in the present study was
validated by a re-docking method (Shivanika et al., 2020). To
validate the docking protocol, the CCI was re-docked into the
active binding site of each chain of the target protein using the
same protocols that were previously used for docking the
flavonoids to the target protein. After re-docking was completed,
the ‘pdbqt.’ output of the re-docked ligand was opened with a
text document and all the information on the ligand was copied.
The ‘pdbqt.’ output of the target protein was opened with a text
document and the information on the re-docked ligand was
pasted at the end. This text document was saved for further use.
Finally, the re-docked protein-CCI complex was converted to
PDB file format from the pdbqt.’ file format. This process was
carried out for all the chains of the target protein.
At the same time, the original protein-CCI complex was
processed with the Discovery Studio Visualizer software to
obtain chain A complexed with the CCI, chain B complexed
with the CCI, chain C complexed with the CCI and chain D
complexed with the CCI. It may be noted that all the chains of
the target protein are originally complexed with CCI. The
original protein-CCI complex (chain A) and the re-docked
protein-ligand complex (chain A) were opened simultaneously
on the Discovery Studio visualizer software. The sequence
alignment of the target proteins was created following which
they were made to superimpose on each other. The root mean
square deviation (RMSD) between the original protein-CCI
complex and the re-docked protein-CCI complex was calculated.
Also, the superimposed protein-CCI image was generated with
the Discovery Studio Visualizer software. This process was
repeated for all the chains of SARS-CoV-2 3CL pro.
I. Toxicity analysis and drug-likeness
The toxicity and drug-likeness of the flavonoid were
evaluated with Data Warrior v.5.2.1 software. Toxicities such as
mutagenicity, tumorigenic, reproductive effective and irritant of
the flavonoid were analyzed. The toxicity and drug-likeness of
the CCI were also analyzed.
J. Protease inhibition
The protease inhibition of the flavonoid with the highest
binding affinity towards SARS-CoV-2 3CL pro was predicted
with the Molinspiration Chemoinformatics web tool
(Molinspiration, 2021). The protease inhibition of the CCI was
also calculated.
K. Structure-activity relationship
The contribution of chemical groups, atoms, and sugar moiety
of the flavonoid towards protease inhibition was studied. The
structure of the flavonoid was modified at certain points using
Marvin Sketch 20.10 software. The SMILES ID of each
modified flavonoid structure was generated and its protease
inhibition was evaluated with the Molinspiration
Chemoinformatics web tool (Molinspiration, 2021). In this way,
a preliminary structure-activity relationship (SAR) study was
carried out for rutin.
III. RESULTS AND DISCUSSION
A. Features of the target protein
The crystal structure of the SARS-CoV-2 3CL pro is available
from the RCSB-PDB website. It is made up of four chains i.e.
chain A, chain B, chain C and chain D. The sequence length of
SARS-CoV-2 3CL pro is 306. All the chains are complexed with
a co-crystal inhibitor viz. 3WL. 3WL is a flavonoid known as
baicalein and bears a PubChem CID of 5281605. In the present
study, 3WL was used as a standard drug whenever and wherever
necessary.
B. Details of the flavonoid library
A total of 16 flavonoids were selected to be screened against
SARS-CoV-2 3CL pro. The library of flavonoids to be used in
the study included apigenin (5280443), aromadendrin (122850),
eriodictyol (440735), fisetin (5281614), hesperetin (72281),
isorhamnetin (5281654), kaempferol (5280863), luteolin
(5280445), myricetin (5281672), naringenin (932), pachypodol
(5281677), quercetin (5280343), rhamnazin (5320945), rutin
(5280805), tangeretin (68077) and taxifolin (439533). The co-
crystal inhibitor that is complexed with the target protein is also
a flavonoid.
C. Molecular docking and binding affinities of flavonoids
Molecular docking is a computational technique that predicts
possible interactions between a drug and a protein. It gives an
idea of the inhibitory potential of a drug against a protein
involved in a disease network (Meng et al., 2012). When MDSS
is carried out on a PyRx tool, it provides a binding affinity value
(kcal/mol) for each ligand so that the binding potential of ligands
toward a protein can be ranked (Dallakyan & Olson, 2015).
The binding affinities of all the flavonoids towards chain A,
chain B, chain C, and chain D of SARS-CoV-2 3CL pro are
given in Table I. The binding affinity of the 3WL is also
included in Table I. For each ligand, PyRx tool generates a total
of 9 poses at the active binding site of the target protein. A more
negative binding affinity value suggests a better binding between
a compound and a protein (Dallakyan & Olson, 2015). A low
binding affinity value also indicates the low energy requirement
for protein-ligand binding (Azam & Abbasi, 2013). In all cases,
the first pose is considered the best pose since it has the highest
binding affinity towards the target protein. The ninth pose has
the lowest binding affinity towards the target protein. Based on
this, the first pose and its binding affinity value were considered
for the study.
Table I. Binding affinities of the flavonoids and the CCI towards the
active binding site of different chains of SARS-CoV-2
Compounds
Binding affinity (kcal/mol)
Chain A
Chain B
Chain C
Chain D
3WL
-7.3
-7.5
-7.4
-7.3
Apigenin
-7.3
-7.6
-7.1
-7.2
Journal of Scientific Research, Volume 65, Issue 5, 2021
99
Institute of Science, BHU Varanasi, India
Aromadendrin
-7.5
-7.5
-7.0
-6.9
Eriodictyol
-7.6
-8.2
-7.4
-7.4
Fisetin
-7.7
-8.6
-7.1
-7.3
Hesperetin
-7.6
-8.0
-7.0
-7.1
Isorhamnetin
-7.5
-7.5
-7.2
-7.2
Kaempferol
-7.3
-7.4
-7.1
-6.8
Luteolin
-7.8
-8.6
-7.3
-7.4
Myricetin
-7.6
-8.4
-7.4
-7.6
Naringenin
-7.2
-7.6
-6.6
-7.1
Pachypodol
-7.0
-7.0
-7.3
-7.5
Quercetin
-7.9
-8.7
-7.4
-7.4
Rhamnazin
-7.4
-7.2
-7.1
-7.5
Rutin
-8.5
-9.3
-8.7
-9.7
Tangeretin
-7.1
-6.4
-6.4
-6.9
Taxifolin
-7.6
-8.1
-7.3
-7.1
Among all the flavonoids, rutin has the highest binding
affinity towards chain A (-8.5 kcal/mol), chain B (-9.3 kcal/mol),
chain C (-8.7 kcal/mol), and chain D (-9.3 kcal/mol) of SARS-
CoV-2 3CL pro. This suggests that in comparison to other
flavonoids, rutin will bind easily to the active binding site of
different chains of the target protein with minimum energy
involvement.
3WL is the CCI that is present at the active binding site of all
the chains of the target protein. In comparison to 3WL, it was
also observed that rutin had a better binding affinity towards all
the chains of SARS-CoV-2 3CL pro. Thus, the MDSS study
revealed that rutin has the best potential to bind and interact with
the amino acid residues at the active binding site of the target
protein. Based on this evidence, rutin was selected for further
analysis.
D. Molecular interactions between different chains of SARS-
CoV-2 3CL pro and rutin
A good understanding of the interaction between a protein and
a ligand is important in the field of drug development (Du et al.,
2016). The 2D and 3D ligand interactions along with the 3D
binding pose of rutin and 3WL at the active binding site of chain
A, chain B, chain C, and chain D of SARS-CoV-2 3CL pro are
given in Fig. 1, 2, 3, and 4 respectively. The ligand interactions
of rutin and 3WL with the amino acid residues at the active
binding site of different chains of SARS-CoV-2 3CL pro are
given in Table II. Generally, rutin was found to interact with
more amino acid residues at the active binding site than 3WL.
Table II. Interacting active site residues of SARS-CoV-2 3CL pro with
3WL and rutin
SARS-CoV-2
3CL pro
Ligand interactions
3WL
Rutin
Chain A
GLY143, GLU166,
MET165, CYS145,
ASN142, HIS163
MET49, GLU166,
ASP187, ARG188,
GLN189, THR190,
GLN192, HIS41, CYS44
Chain B
LEU141, SER144,
CYS145, GLU166,
HIS41, CYS44, MET49
HIS41, THR24, CYS44,
MET49, CYS145,
ASN142
Chain C
ASN142, GLY143,
SER144, GLU166,
CYS145, MET165
LEU141, SER144,
CYS145, MET165,
GLU166
Chain D
GLY143, SER144,
HIS163, GLU166,
CYS145, MET165
THR26, HIS41, TYR54,
LEU141, ASN142,
GLU166, ASP187,
MET49, CYS44,
HIS164, MET165,
ARG188
In chain A, rutin interacted with 9 amino acid residues
(MET49, GLU166, ASP187, ARG188, GLN189, THR190,
GLN192, HIS41, and CYS44) while 3WL interacted with 6
amino acid residues (GLY143, GLU166, MET165, CYS145,
ASN142, and HIS163) (Fig. 1, Table II). Both rutin and 3WL
interacted with GLU166 (Table II). The hydroxyl (-OH) group at
position 13 of rutin interacted with MET49 and ASP187 through
a conventional hydrogen bond. The -OH at position 19 of rutin
interacted with GLU166 through a conventional hydrogen bond.
The -OH group at positions 28 and 29 of rutin interacted with
ARG188 and GLN192 through conventional hydrogen bonds
respectively. The -OH group at position 30 of rutin interacted
with GLN189 and THR190 through a conventional hydrogen
bond. The A-ring of the C6-C3-C6 scaffold of rutin formed pi-
sigma interaction with MET49, pi-pi interaction with HIS41, and
pi-sulfur interaction with CYS44. The C-ring of the C6-C3-C6
scaffold of rutin also formed pi-alkyl interaction with MET49
and pi-pi interaction with HIS41.
In chain B, rutin (HIS41, THR24, CYS44, MET49, CYS145,
ASN142) and 3WL (LEU141, SER144, CYS145, GLU166,
HIS41, CYS44, MET49) interacted with 6 and 7 amino acid
residues respectively (Fig. 2, Table II). Both rutin and 3WL
interacted with CYS44, HIS41, MET49, and CYS145 (Table II).
The -OH group at position 13 and the oxygen atom at position
43 of rutin interacted with HIS41 through a conventional
hydrogen bond. The -OH group at position 40 of rutin interacted
with THR24 through a conventional hydrogen bond. The A-ring
and C-ring of the C6-C3-C6 scaffold of rutin formed pi-pi
interaction with HIS41. The A-ring of the C6-C3-C6 scaffold of
rutin formed pi-alkyl interaction with MET49 while the B-ring
and C-ring of rutin formed pi-alkyl interaction with CYS145.
The A-ring of rutin formed pi-sulfur interaction with CYS44.
In chain C, rutin interacted with 5 amino acid residues
(LEU141, SER144, CYS145, MET165, GLU166) while 3WL
interacted with 6 amino acid residues (ASN142, GLY143,
SER144, GLU166, CYS145, MET165) (Fig. 3, Table II). Both
rutin and 3WL interacted with SER144, CYS145, MET165, and
GLU166 (Table II). The -OH group at position 19 of rutin
interacted with SER144 through a conventional hydrogen bond.
The -OH group at position 20 of rutin interacted with LEU141
through a conventional hydrogen bond. The B-ring and C-ring of
the C6-C3-C6 scaffold of rutin formed pi-alkyl interaction with
CYS145 and MET165 respectively.
In addition to hydrogen bonds, non-covalent bonds such as
hydrophobic interactions and electrostatic interactions are also
considered important for the stabilization of protein-ligand
complexes (de Freitas & Schapira, 2017; Kumar & Nussinov,
2002). Rutin showed non-covalent interactions (pi-pi, pi-alkyl,
and pi-sulfur) with different amino acid residues at the active
binding site of different chains of SARS-CoV-2 3CL pro (Fig. 1,
Journal of Scientific Research, Volume 65, Issue 5, 2021
100
Institute of Science, BHU Varanasi, India
Fig. 2, Fig. 3 and Fig. 4). Thus, rutin might be able to inhibit
SARS-CoV-2 3CL pro as it was found to have a good binding
affinity towards all the different chains. In comparison to 3WL,
rutin showed better molecular interactions with different amino
acid residues at the active binding site of SARS-CoV-2 3CL pro.
Based on the above findings, rutin was subjected to further
analysis.
Fig. 1. Chain A: 2D ligand interactions (a, c), 3D ligand interactions (b,
d), and 3D binding pose (e) (blue=3WL, red=Rutin) of 3WL (a, b, e)
and rutin (c, d, e) at the active binding site of SARS-CoV-2 3CL pro.
Fig. 2. Chain B: 2D ligand interactions (a, c), 3D ligand interactions (b,
d), and 3D binding pose (e) (blue=3WL, red=Rutin) of 3WL (a, b, e)
and rutin (c, d, e) at the active binding site of SARS-CoV-2 3CL pro.
Fig. 3. Chain C: 2D ligand interactions (a, c), 3D ligand interactions (b,
d), and 3D binding pose (e) (blue=3WL, red=Rutin) of 3WL (a, b, e)
and rutin (c, d, e) at the active binding site of SARS-CoV-2 3CL pro.
Fig. 4. Chain D: 2D ligand interactions (a, c), 3D ligand interactions (b,
d), and 3D binding pose (e) (blue=3WL, red=Rutin) of 3WL (a, b, e)
and rutin (c, d, e) at the active binding site of SARS-CoV-2 3CL pro.
E. Validation of docking
The purpose of this study was to evaluate the efficiency of the
docking protocol used in the present study. The docking protocol
was validated through re-docking of the CCI to the active
binding site of the target protein using the same parameters that
were used to dock the flavonoids. The re-docked protein-ligand
complex was then superimposed to the original protein-ligand
complex. The superimposed image of the protein-ligand
complex is given in Fig. 5.
Journal of Scientific Research, Volume 65, Issue 5, 2021
101
Institute of Science, BHU Varanasi, India
The RMSD of the re-docked protein-ligand complex was
calculated by using the original protein-ligand complex as the
reference protein. Several researchers had also validated their
docking protocol with this method (Shivanika et al. 2020). A
low RMSD value is preferred over a higher value to provide
validation to the docking protocol.
In the present study, an RMSD value of 0.00 was obtained for
all the docking procedures used for docking the CCI towards the
active binding site of chain A, chain B, chain C, and chain D.
Re-docking showed that the CCI had a binding affinity of -7.3
kcal/mol, -7.5 kcal/mol, -7.4 kcal/mol, and -7.3 kcal/mol
towards chain A, chain B, chain C, and chain D respectively.
The binding affinity values obtained from re-docking are similar
to the value previously obtained in the first docking (Table I).
Also, the docking protocol used for the present study allowed the
binding of CCI into the active binding site of the target protein
as evidence by re-docking (Fig. 5). This proves the efficiency
and validity of the docking protocol followed in the present
study.
Fig. 5. Superimposition of re-docked protein-3WL complex (3WL=red
color) with the original co-crystallized complex (3WL=blue color) for
chain A (a), chain B (b), chain C (c) and chain D (d) of SARS-CoV-2
3CL pro.
F. Toxicity analysis and drug-likeness
Toxicity is one of the main reasons behind the withdrawal of
many drugs from the market (Guengerich, 2011). Drug-likeness
is a parameter that indicates the possibility of a molecule to
become an orally bioavailable drug (Daina et al., 2017).
Therefore, it may be considered logical to assess the potential
toxicity and drug-likeness of a compound at the initial stage of a
study. The potential mutagenicity, tumorigenic, reproductive
effective and irritant of rutin and 3WL are given in Table III.
The drug-likeness of rutin and 3WL are also included in Table
III. It was observed that rutin was free from all possible
toxicities. Also, the drug-likeness of rutin was higher than 3WL.
Based on these findings, rutin was subjected to further analysis.
Table III. Toxicity and drug-likeness of 3WL and rutin
Property
3WL
Rutin
Mutagenicity
None
None
Tumorigenic
None
None
Reproductive effective
None
None
Irritant
None
None
Drug-likeness
0.28194
1.9337
G. Protease inhibition
The protease inhibition of rutin was predicted and it was
compared to that of 3WL (Table IV). SARS-CoV-2 3CL pro is a
viral protease (Prajapat et al., 2020). Although the predicted
protease inhibition might not necessarily correlate to the
inhibition of the main protease of SARS-CoV-2, it provides a
preliminary idea of the potential bioactivity of rutin. With a
bioactivity score of -0.07, rutin has a better protease inhibition
than 3WL (-0.35). Based on these findings, the structure-activity
relationship of rutin concerning its protease inhibition was
studied.
Table IV. Protease inhibitory potential of 3WL and rutin
Compound
Protease inhibition
3WL
-0.35
Rutin
-0.07
H. Structure-activity relationship
Preliminary SAR studies can be used to identify potential
chemical groups that might be responsible for eliciting the
bioactivity of a compound (McKinney et al., 2000). The SAR of
rutin concerning its protease inhibition was studied with the
Molinspiration Chemoinformatics web tool. The contribution of
different atoms, groups, and the sugar moiety towards protease
inhibition of rutin are given as numerical value in Table V.
Table V. SAR of rutin
Structure No.
Structure
Protease inhibition
1 (Rutin)
-0.07
Journal of Scientific Research, Volume 65, Issue 5, 2021
102
Institute of Science, BHU Varanasi, India
2
-0.06
3
-0.25
4
0.05
5
-0.08
6
-0.03
7
-0.04
8
-0.05
From structures no. 2 and 3 (Table V), the sugar moiety of
rutin might be necessary for its protease inhibition activity. Also,
the -OH groups of the sugar moiety of rutin were significantly
involved in the formation of conventional hydrogen bonds with
the amino acid residues at the active binding site of different
chains of SARS-CoV-2 3CL pro (Fig. 1, 2, and 4). The removal
of an oxygen atom at position 43 attached to the C-ring of rutin
significantly increases the protease inhibition activity (Table V,
Structure no. 4). Also, the oxygen atom at position 43 attached
to the C-ring of rutin was not involved in any molecular
interactions (Fig. 1, 3, and 4). In structures no. 5 to 8 (Table V),
the removal of -OH groups from different positions does not
seem to have a positive effect on the protease inhibition activity
of rutin. The aglycone moiety (C6-C3-C6 scaffold) was actively
involved in the formation of different types of protein-ligand
interactions (Fig. 1, 2, 3, and 4).
Based on these findings, it may be assumed that the sugar
moiety and the aglycone moiety of rutin are essential for binding
to the active binding site of SARS-CoV-2 3CL pro. The sugar
moiety of rutin may also be essential for the protease inhibition
activity of rutin (Table V). On the other hand, the oxygen atom
at position 43 attached to the C-ring of rutin does not seem to
contribute to the formation and stabilization of the protein-ligand
complex (Fig. 1, 2, 3, and 4).
Interestingly, molecular docking and molecular dynamics
simulation studies also showed rutin as a potential inhibitor of
SARS-CoV-2 3CL pro (Cherrak et al., 2020). In the study, chain
A of SARS-CoV-2 3CL pro was used. Cherrak et al. (2020)
showed that the sugar moiety of rutin formed two hydrogen
bonds with GLU166. Cherrak et al. (2020) also showed that the
A-ring of rutin formed hydrophobic interactions with MET49.
Similarly, in our study, rutin formed a conventional hydrogen
bond with GLU166. Rutin also formed conventional hydrogen
Journal of Scientific Research, Volume 65, Issue 5, 2021
103
Institute of Science, BHU Varanasi, India
bonds and non-covalent interactions with MET49 of chain A of
SARS-CoV-2 3CL pro. In addition, our study also provides data
for chain B, chain C, and chain D.
Rutin is a citrus flavonoid (Kreft et al., 1997) that is
abundantly found in apples, tea, buckwheat, and passionflowers
(Harborne, 1986). Rutin exerts several pharmacological
activities such as anticarcinogenic, antioxidant, vasoprotective,
cytoprotective, cardioprotective, and neuroprotective activities
(Ganeshpurkar & Saluja, 2017). An analog of rutin known as
sodium rutin sulfate showed anti-retroviral activity against
various strains of human immunodeficiency virus 1 (Tao et al.,
2007). Rutin also showed antiviral activity against vesicular
stomatitis virus (Wacke & Eilmes, 1978), canine distemper virus
(Carvalho et al., 2013), and avian influenza (H5N1) virus
(Ibrahim et al., 2013). The literature review revealed the potent
anti-viral activity of rutin against different pathogens of viral
origin. Considering the anti-viral activity of rutin against other
RNA viruses, there is a possibility that rutin might elicit anti-
viral activity against SARS-CoV-2 by inhibiting its main
protease (3CL pro) which is a key component for viral
replication.
CONCLUSION
The present in-silico study revealed rutin as a potential
inhibitor of SARS-CoV-2 3CL pro. Rutin showed a good
binding affinity at the active binding pocket of all the four chains
of SARS-CoV-2 3CL pro. Rutin also showed better ligand
interactions with SARS-CoV-2 3CL pro than 3WL. In many
cases, rutin and 3WL showed similar molecular interactions with
the same amino acid residues at the active binding site of SARS-
CoV-2 3CL pro. Rutin formed more conventional hydrogen
bonding than 3WL. Rutin is non-toxic and had a better drug-
likeness score than 3WL. Also, rutin was predicted to be a better
protease inhibitor than 3WL. SAR revealed that the aglycone
moiety and the sugar moiety of rutin might be essential for its
protease inhibition activity. SAR also revealed that the oxygen
atom at position 43 of rutin does not contribute to protein-ligand
binding and protease inhibition activity. Therefore, the present
in-silico study advocates rutin as a potential inhibitor of SARS-
CoV-2 3CL pro and as promising phytotherapy for COVID-19.
The C6-C3-C6 scaffold of rutin may also be used to design and
develop potent anti-viral agents against SARS-CoV-2.
ACKNOWLEDGMENTS
The author acknowledges the Department of Pharmaceutical
Sciences, Dibrugarh University for providing the necessary
facilities to carry out the in-silico work.
REFERENCES
Antonio, A.D.S., Wiedemann, L.S.M., & Veiga-Junior, V.F.
(2020). Natural products’ role against COVID-19. RSC
Advances, 10, 23379-23393.
Azam, S.S., & Abbasi, S.W. (2013). Molecular docking studies for
the identification of melatoninergic inhibitors for
acetylserotonin-O-transferase using different docking routines.
Theoretical biology and medical modeling, 10, 63.
Carvalho, O.V., Botelho, C.V., Ferreira, C.G., Ferreira, H.C.,
Santos, M.R., Diaz, M.A., Oliveira, T.T., Soares-Martins, J.A.,
Almeida, M.R., & Silva, A. (2013). In vitro inhibition of
canine distemper virus by flavonoids and phenolic acids:
implications of structural differences for antiviral
design. Research in Veterinary Science, 95(2), 717-724.
Cherrak, S.A., Merzouk, H., Mokhtari-Soulimane, N. (2020).
Potential bioactive glycosylated flavonoids as SARS-CoV-2
main protease inhibitors: A molecular docking and simulation
studies. PLoS ONE, 15(10), e0240653.
Chen, D., Oezguen, N., Urvil, P., Ferguson, C., Dann, S.M., &
Savidge, T.C. (2016). Regulation of protein-ligand binding
affinity by hydrogen bond pairing. Science Advances, 2,
e1501240.
Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: a free
web tool to evaluate pharmacokinetics, drug-likeness and
medicinal chemistry friendliness of small molecules. Scientific
Reports, 7, 42717.
Dallakyan, S., & Olson, A.J. (2015). Small-molecule library
screening by docking with PyRx. Methods in Molecular
Biology, 1263, 243-250.
de Freitas, R.F., & Schapira, M. (2017). A systematic analysis of
atomic protein-ligand interactions in the PDB. Medicinal
Chemistry Communications, 8, 1970-1981.
Du, X., Li, Y., Xia, Y.L., Ai, S.M., Liang, J., Sang, P., Ji, X.L., &
Liu, S.Q. (2016). Insights into Protein-Ligand Interactions:
Mechanisms, Models, and Methods. International Journal of
Molecular Sciences, 17(2), 144.
Ganeshpurkar, A., & Saluja, A.K. (2016). The Pharmacological
Potential of Rutin. Saudi Pharmaceutical Journal, 25(2), 149-
164.
Gildenhuys, S. (2020). Expanding our understanding of the role
polyprotein conformation plays in the coronavirus life cycle.
Biochemical Journal, 477, 1479-1482.
Guengerich, F.P. (2011). Mechanisms of drug toxicity and
relevance to pharmaceutical development. Drug Metabolism
and Pharmacokinetics, 26, 3-14.
Harborne, J.B. (1986). Nature, distribution and function of plant
flavonoids. Progress in Clinical and Biological Research, 213,
15-24.
Hu, B., Guo, H., Zhou, P., & Shi, Z.L. (2021). Characteristics of
SARS-CoV-2 and COVID-19. Nature Reviews Microbiology,
19(3), 141-154.
Huang, Y., Yang, C., Xu, X., Xu, W., & Liu, S. (2020). Structural
and functional properties of SARS-CoV-2 spike protein:
potential antivirus drug development for COVID-19. Acta
Pharmacologica Sinica, 41, 1141-1149.
Ibrahim A.K., Youssef A.I., Arafa A.S., & Ahmed S.A. (2013).
Anti-H5N1 virus flavonoids from Capparis
sinaica Veill. Natural Product Research, 27(22), 2149-2153.
Islam, M.T., Sarkar, C., El-Kersh, D.M., Jamaddar, S., Uddin, S.J.,
Shilpi, J.A., & Mubarak, M.S. (2020). Natural products and
their derivative against coronavirus: A review of the non-
Journal of Scientific Research, Volume 65, Issue 5, 2021
104
Institute of Science, BHU Varanasi, India
clinical and pre-clinical data. Phytotherapy Research, 34,
2471-2492.
Jin, Z., Du, X., Xu, Y., Deng, Y., Liu, M., Zhao, Y., Zhang, B., Li,
X., Zhang, L., Peng, C., Duan, Y., Yu, J., Wang, L., Yang, K.,
Liu, F., Jiang, R., Yang, X., You, T., Liu, X., Yang, X., Bai, F.,
Liu, H., Liu, X., Guddat, L.W., Xu, W., Xiao, G., Qin, C., Shi,
Z., Jiang, H., Rao, Z., & Yang, H. (2020). Structure of Mpro
from SARS-CoV-2 and discovery of its inhibitors. Nature,
582, 289-293.
Kim, D.W., Seo, K.H., Curtis-Long, M.J., Oh, K.Y., Oh, J.W.,
Cho, J.K., Lee, K.H., & Park, K.H. (2014). Phenolic
phytochemical displaying SARS-CoV papain-like protease
inhibition from the seeds of Psoralea corylifolia. Journal of
Enzyme Inhibition and Medicinal Chemistry, 29, 59-63.
Kumar, S., & Nussinov, R. (2002). Close-range electrostatic
interactions in proteins. Chembiochem, 3, 604-617.
Kreft, S., Knapp, M., & Kreft, I. (1997). Extraction of rutin from
buckwheat (Fagopyrum esculentum Moench) seeds and
determination by capillary electrophoresis. Journal of
Agricultural and Food Chemistry, 47(11), 4649-4652.
McKinney, J.D., Richard, A., Waller, C., Newman, M.C., &
Gerberick, F. (2000). The Practice of Structure Activity
Relationships (SAR) in Toxicology. Toxicological Sciences,
56(1), 8-17.
Meng, X.Y., Zhang, H.X., Mezei, M., & Cui, M. (2011).
Molecular docking: a powerful approach for structure-based
drug discovery. Current computer-aided drug design, 7(2),
146-157.
Molinspiration. (2021). Calculation of molecular properties and
bioactivity score. Retrieved May 7, 2021, from
https://www.molinspiration.com/cgi-bin/properties
Narkhede, R.R., Pise, A.V., Cheke, R.S., Shinde, S.D. (2020).
Recognition of natural products as potential inhibitors of
COVID-19 main protease (Mpro): In-Silico evidences. Natural
Products and Bioprospecting, 10, 297-306.
Ninfali, P., Antonelli, A., Magnani, M., & Scarpa, E.S. (2020).
Antiviral Properties of Flavonoids and Delivery Strategies.
Nutrients, 12(9), 2534.
PubChem. (2021). Retrieved May 5, 2021, from
https://pubchem.ncbi.nlm.nih.gov/
Prajapat, M., Sarma, P., Shekhar, N., Avti, P., Sinha, S., Kaur, H.,
Kumar, S., Bhattacharyya, A., Kumar, H., Bansal, S., &
Medhi, B. (2020). Drug targets for corona virus: A systematic
review. Indian Journal of Pharmacology, 52, 56-65.
Robson, B. (2020). Computers and viral diseases. Preliminary
bioinformatics studies on the design of a synthetic vaccine and
a preventative peptidomimetic antagonist against the SARS-
CoV-2 (2019-nCoV, COVID-19) coronavirus. Computers in
Biology and Medicine. 119, 103670.
RCSB-PDB. (2021). SARS-CoV-2 3CL protease (3CL pro) in
complex with a novel inhibitor. Retrieved May 5, 2021, from
https://www.rcsb.org/structure/6M2N
Shivanika, C., Deepak Kumar, S., Ragunathan, V., Tiwari, P.,
Sumitha, A., Brindha Devi, P. (2020). Molecular docking,
validation, dynamics simulations, and pharmacokinetic
prediction of natural compounds against the SARS-CoV-2
main-protease. Journal of Biomolecular Structure and
Dynamics, 8:1-27.
Shende, P., Khanolkar, B., Gaud, R.S. (2020). Drug repurposing:
new strategies for addressing COVID-19 outbreak. Expert
Review of Anti Infective Therapy, 3, 1-18.
Singh, S.P., Pritam, M., Pandey, B., & Yadav, T.P. (2021).
Microstructure, pathophysiology, and potential therapeutics of
COVID-19: A comprehensive review. Journal of Medical
Virology, 93(1), 275-299.
Tahir Ul Qamar, M., Maryam, A., Muneer, I., Xing, F., Ashfaq,
U.A., Khan, F.A., Anwar, F., Geesi, M.H., Khalid, R.R., Rauf,
S.A., & Siddiqi, A.R. (2019). Computational screening of
medicinal plant phytochemicals to discover potent pan-
serotype inhibitors against dengue virus. Scientific Reports, 9,
1433.
Tao J., Hu Q., Yang J., Li R., Li X., Lu C., Chen C., Wang L.,
Shattock R., & Ben K. (2007). In vitro anti-HIV and -HSV
activity and safety of sodium rutin sulfate as a microbicide
candidate. Antiviral Research, 75(3), 227-233.
Trott, O., & Olson, A.J. (2010). AutoDock Vina: improving the
speed and accuracy of docking with a new scoring function,
efficient optimization, and multithreading. Journal of
Computational Chemistry, 31, 455-461.
Wacker A., & Eilmes H.G. (1978). Antiviral activity of plant
components. 1st communication: Flavonoids.
Arzneimittelforschung, 28(3), 347-350.
World Health Organization. (2021a). WHO Coronavirus (COVID-
19) Dashboard. Retrieved May 11, 2021, from
https://covid19.who.int/
World Health Organization. (2021b). Coronavirus disease
(COVID-19) pandemic. Retrieved May 11, 2021, from
https://www.who.int/emergencies/diseases/novel-coronavirus-
2019
Zakaryan, H., Arabyan, E., Oo, A., & Zandi, K. (2017).
Flavonoids: promising natural compounds against viral
infections. Archives of Virology, 162(9), 2539-2551.
***
... Nowadays, owing to its accuracy, precision, and robustness, in-silico techniques are increasingly used to study the potential inhibitory effects of phytocompounds against different drug targets. [10][11][12][13][14] Molecular docking simulation studies (MDSS) are used to study the affinity of a ligand toward a target protein including their potential molecular interactions. 15,16 Molecular dynamics (MD) simulations are used to study the stability of a ligand at the active binding pocket of a protein in a computationally simulated physiological environment. ...
... As described in our previous research works, 11,15 MDSS were carried out with a validated docking protocol with the AutoDock Vina on the PyRx version 0.8 virtual screening tool and the AutoDock Vina neutralizes the charge of the ligands before docking. The molecular docking studies were carried out with two different approaches. ...
... Molecular docking is a reliable computational technique to study the binding pose, binding affinity, and molecular interactions of chemical compounds toward a target protein. 11 Add reference 40 here EA has a high binding affinity for the active site of PfDHFR-TS and was found to interact with ALA16, LEU40, GLY41, GLY44, LEU46, ASP54, MET55, PHE58, THR107, SER108, SER111, ILE112, ILE164, GLY166, SER167, and TYR170 of PfDHFR-TS (refer Tables 1 and 2). From a literature review, we found that synthetic compounds with reported activity against Pf interacted with different amino acid residues at the active binding site of PfDHFR-TS, namely, ALA16, LEU40, GLY41, GLY44, MET55, PHE58, ILE112, LEU119, GLY166, and TYR170. ...
Article
Full-text available
Ellagic acid (EA) is an antiplasmodial polyphenol with reported in-vitro activity against Plasmodium falciparum. Studies have reported that EA acts in the late erythrocytic stages of P. falciparum (Pf) when DNA synthesis is taking place. Pf dihydrofolate reductase-thymidylate synthase (PfDHFR-TS) is an important enzyme for DNA synthesis as its inhibition can kill the parasite. As there is no reported study on the molecular interactions between EA and PfDHFR-TS, we aim to study the molecular interactions between EA and PfDHFR-TS (PDB ID: 3DGA) through molecular docking, molecular dynamics (MD) simulations, binding free energy (MM-GBSA) calculations, and density functional theory (DFT) studies. Site-specific and blind docking revealed that EA has a high binding affinity for the active site of PfDHFR-TS. EA formed hydrogen bonds with multiple active site residues. MD simulations for 100 ns revealed that the PfDHFR-TS-EA complex was stable. The average binding free energy of the PfDHFR-TS-EA complex throughout the 100 ns MD simulations was −39.84 kcal/mol. The energy difference (ΔE = 0.04089 eV) obtained from DFT studies indicates the electrical stability and reactivity of EA at the active site of PfDHFR-TS. We conclude that the antiplasmodial activity of EA might be attributed to its ability to potentially bind with PfDHFR-TS.
... According to the protocols described in our previous studies, the AutoDock Vina embedded in the PyRx tool was used to execute the MDSS. 14 The molecular interactions of the phytocompounds that showed a better binding affinity than the CCI were further analyzed with the DSV software. The 2D ligand interactions and the 3D binding poses were also generated with the DSV software. ...
... The CCI (positive control) has a binding affinity of -5.7 kcal/mol. Among the 29 phytocompounds, a total of 23 phytocompounds (Compound code: 1, 2, 3, 4, 5, 6,8,9,10,14,15,17,18,19,20,21,22, 23, 24, 25, 26, 28 and 29) showed a better binding affinity than the CCI. Among the 23 phytocompounds, the compound (2R)-4',7dihydroxyflavan-(4aà8)-(2R,3S)-3,5,7trihdyroxyflavan-3"-O-α-L-rhamnopyranoside bearing a compound code 25 has the best binding affinity (-9.2 kcal/mol) towards the active binding site of α-glucosidase. ...
Article
Full-text available
The aqueous extract of the shoot tips of Acacia pennata showed a high enzyme inhibitory activity against α-glucosidase. However, the enzyme inhibiting phytocompound is not known. Identification of the antidiabetic phytocompound will be beneficial in designing drugs with higher efficacy to treat diabetes mellitus. We attempted to identify the compound using molecular docking simulation studies (MDSS). Among the 29 phytocompounds present in A. pennata, MDSS revealed that 23 phytocompounds outperformed the co-crystal inhibitor (CCI) of α-glucosidase (PDB ID: 5NN5) in terms of binding affinity. Among the 23 phytocompounds, (2R)-4',7-dihydroxyflavan-(4aà8)-(2R,3S)-3,5,7-trihdyroxyflavan-3"-O-α-L-rhamnopyranoside (compound 25) has the highest binding affinity (-9.2 kcal/mol). Analysis of the protein-ligand interactions revealed that compound 25 formed 5 conventional hydrogen bonds (ASP 282, TRP 481, ARG 600, ASP 616, HIS 674), 3 hydrophobic interactions (TRP 376, TRP 481, LEU 650), 3 electrostatic interactions (MET 519, ASP 616 (n=2)), and 1 carbon-hydrogen bond (ASP 518). The binding pose analysis further revealed that the docking protocol applied in the study was able to re-dock the CCI and dock compound 25 exactly at the active binding site where the CCI was originally positioned. Our in-silico study showed that compound 25 is the phytocompound of A. pennata that is responsible for potentially inhibiting the α-glucosidase enzyme. The structure of compound 25 may be modified to design more potent inhibitors of α-glucosidase.
... The potent anti-viral activity of Ru against different pathogens of viral origin makes it to be used as a phytotherapy for COVID-19. Among various flavonoids that have been evaluated to oppose COVID-19, Ru had the highest binding affinity to the protease of severe acute respiratory syndrome of SARS-CoV-2, proposing it as the most favorable drug target to treat COVID-19 [14]. ...
Article
Full-text available
Rutin (Ru) is a flavonol glycoside, has significant scavenging characteristics against various reactive oxygen species, which exploits for widespread pharmacological activities such as anti-inflammatory and anti-allergic effects. This study aims to improve the bioavailability of iron oxide nanoparticles (IONPs) by coating them with Ru (Ru-IONPs). Considering this, IONPs and Ru-IONPs were prepared and characterized via various physical techniques. In addition, atomic absorption spectroscopy (AAS) has been used to assess the bio-distribution of the prepared formulation in vivo. Results revealed that the prepared IONPs and Ru-IONPs have a spherical shape with an average diameter of about 8 nm and 10 nm, respectively. Their surface potentials were found to be −19 ± 3.9 and −29.5 ± 5.4 mV, respectively. In vivo results revealed that single dose of IV administration of IONPs (16 mg/kg) elevated oxidative stress, indicated by the reduction of GSH in both heart and liver tissues, and increased their MDA contents. Also, IONPs administration resulted in hepatotoxicity characterized by a significant increase in ALT and AST levels in addition to cardiotoxicity characterized by a significant increase in CK-MB and LDH, in addition to the downregulation of heat shock protein 70 (HSP70) expression, while upregulation of TNF-α expression in both heart and liver tissues. Conversely, nearly all of these alterations were significantly ameliorated for animal groups administrated Ru-IONPs and the morphological injury’s counteraction in both heart and liver tissues. Therefore, we suggest that Ru could be used to alleviate the harmful effects and damages associated with IONPs administration due to its antioxidant, anti-inflammatory, and free radical scavenging properties.
... Then, the structure file has been loaded into PyRx AutoDock Tools, followed by usual processes to obtain binding affinities (Dallakyan & Olson, 2015). Finally, BIOVIA Discovery Studio Visualizer was used to investigate the interaction between the ligands and the protein (Zothantluanga, 2021). ...
... Then, the structure file has been loaded into PyRx AutoDock Tools, followed by usual processes to obtain binding affinities (Dallakyan & Olson, 2015). Finally, BIOVIA Discovery Studio Visualizer was used to investigate the interaction between the ligands and the protein (Zothantluanga, 2021). ...
Article
Full-text available
The natural Fisetin and its derivatives have been shown to have effective bioactivity and strong pharmacological profile, which is continuously drawing the interest of therapeutic applications to the development of new biomolecules against Breast cancer and Monkeypox, and Marburg viral infection, while computational approaches and the study of their structure-activity relationship (SAR) are the most eloquent and reliable platform for performing their hypothetical profile renovation. So, the main perspective of this investigation is to evaluate dual function of Fisetin and its derivatives against both virus and cancerous target. First and foremost, the prediction of activity spectra for materials (PASS) valuation has provided preliminary data on the antiviral, antibacterial, antiparasitic, and anti-cancer possibilities of the mentioned compounds. According to the evidence, PASS predicted scores were shown to perform better in antineoplastic and antiviral than antibacterial, and antiparasitic efficiency; as evidenced by their higher PASS scores in antineoplastic and antiviral drug tests. Breast cancer, Monkeypox, and Marburg virus have been selected as targeted pathogens, and different in silico studies were conducted to determine the dual function of mention derivatives. The “Lipinski five rules,” on the other hand, has been subjected to extensive testing for drug-like characteristics. Molecular docking against Breast cancer, Monkeypox, and Marburg virus have been accomplished after confirmation of their bioactivity. The molecular docking evaluation against targeted disease displayed re-markable binding affinity and non-bonding engagement, with most of the results indicating that derivatives are more effective than the FDA approved standard antiviral, and antineoplastic drugs. Finally, the ADMET characteristics have been computed, and they indicate that the substance is suitable to use and did not have any chance to produce adverse effects on aquatic or non-aquatic environment, as well as having a highly soluble capacity in water medium, high G.I absorption rate, with outstanding bioavailability index. Therefore, these mentioned Fisetin derivatives could be suggested as potential medication against Breast cancer and newly reported Monkeypox, and Marburg virus, and may further proceed for laboratory experiment, synthesis, and clinical trials to evaluate their practical value.
... Similarly to that, we planned the design of the current in-silico study to increase the likelihood of getting positive results in bioassays. One of the quickest and most accurate in-silico methods for analyzing the molecular interactions and chemical bonding between a ligand and a protein is molecular docking (41). To observe and analyze the molecular interaction and ligand binding of compounds with studied biomarkers, molecular docking research, and in-vivo studies are often combined (42)(43)(44)(45)(46)(47)(48). ...
Article
Full-text available
The prevalence of Diabetes mellitus (DM) is continuously rising worldwide. Among its types, type I is characterized by the destruction of beta cells triggered by various mechanisms, including the activation of Caspase 3. Studies have demonstrated the crucial role of Caspase 3 in initiating the apoptosis of beta cells in DM. Our research aims to identify possible phytocompounds inhibitors of Caspase 3 using computational approach. We obtained 3D structures of Caspase 3 and 6511 phytocompounds from the Protein Data Bank and the African Natural Products Database, respectively. The phytocompounds were assessed for druglikeness properties, topological polar surface area, and preliminary toxicity using DataWarrior. The phytocompounds were subjected to molecular docking simulation (MDS) at Caspase 3 active site using AutoDock-Vina. The frontrunner phytocompounds obtained from the MDS were subjected to protease inhibition prediction on Molinspiration. The pharmacokinetics of the phytocompounds were assessed on SwissADME. The in-depth computational toxicity profile of the phytocompounds was evaluated on the pkCSM web. The binding interactions of the phytocompounds with Caspase 3 were assessed with Discovery Studio Visualizer and Maestro. Seventeen phytocompounds were found to have no violation of Lipinski's rule and had no toxicity based on the preliminary assessment, have better binding affinity and protease inhibitory prediction scores than the references, have optimistic bioactivity radar prediction and similar amino acids interaction, in comparison with the references. Further studies, which include in-vitro and in-vivo studies, will be carried out to validate the results of this study.
... Similarly, we have prepared the study design of the present in-silico study to maximize the chance of obtaining fruitful results in bioassays. Molecular docking is one of the fastest and most reliable in-silico techniques to study the binding affinity, binding pose, and molecular interactions between a ligand and a protein [93]. Molecular docking studies are sometimes combined with in-vivo studies to study the molecular interaction and binding affinity of compounds with studied biomarkers [94,95]. ...
Article
Full-text available
The continuous destruction of normal insulin-producing pancreatic beta-cells is a contributing factor in all common forms of diabetes, due to insufficient production of insulin, especially in type 1 diabetes. There are attempts at beta-cells transplantation, but the cost and availability of donors pose a great challenge to the process. Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase A (DYRK1A) plays a crucial role in beta-cells destruction. Our research targets to identify plants that can be utilized as a possible alternative approach to beta-cell replacement through a pharmacologically induced regeneration of new beta-cells in-silico. The 3D structure DYRK1A and 6511 phytochemicals were obtained from the Protein Data Bank and the African Natural Products Database respectively. They were duly prepared for molecular docking simulations (MDS). MDS was implemented, after validation of docking protocols, in AutoDock-Vina®, with virtual screening scripts. Phytocompounds with good binding affinities for DYRK1A were selected as frontrunners. The compounds were screened for toxicity, Lipinski’s rule confirmation with Data Warrior software followed by kinase inhibitory bioactivity prediction with the Molinspiration Chemoinformatics web tool. Twelve phytocompounds were found to be predictably highly active in-silico against DYRK1A with good drug-like property based on Lipinski’s rule, non-mutagenic, non-tumorigenic, no reproductive effect, and non-irritant, with high predicted bioactivity. In-silico active phytocompounds against DYRK1A with their plant sources and physicochemical parameters were identified. Further studies will be carried out in-vitro and in-vivo to validate the results of this study using plants containing the identified phytocompounds.
Article
In-silico techniques offer a fast, accurate, reliable, and economical approach to studying the molecular interactions between compounds and proteins. In this study, our main aim is to use in-silico techniques as a rational approach for the prediction of the molecular drug targets for luteolin against Plasmodium falciparum. Multi-target molecular docking, 100 nanoseconds (ns) molecular dynamics (MD) simulations, and Molecular Mechanics-Generalized Born Surface Area (MM-GBSA) binding free energy calculations were carried out for luteolin against dihydrofolate reductase thymidylate synthase (PfDHFR-TS), dihydroorotate dehydrogenase (PfDHODH), and falcipain-2. The native ligands of each protein were used as a reference to evaluate the performance of luteolin. Luteolin outperformed the native ligands of all proteins at molecular docking and MD simulations studies. However, in the MM-GBSA calculations, luteolin outperformed the native ligand of only PfDHFR-TS but not PfDHODH and falcipain-2. Among the studied proteins, the in-silico approach predicted PfDHFR-TS as the most favorable drug target for luteolin.
Article
Full-text available
The β-lactamase of Pseudomonas aeruginosa is known to degrade β-lactam antibiotics such as penicillins, cephalosporins, monobactams, and carbapenems. With the discovery of an extended-spectrum β-lactamase in a clinical isolate of P. aeruginosa, the bacterium has become multi-drug resistant. In this study, we aim to identify new β-lactamase inhibitors by virtually screening a total of 43 phytocompounds from two Indian medicinal plants. In the molecular docking studies, pinocembrin-7-O-β-D-glucopyranoside (P7G) (−9.6 kcal/mol) from Acacia pennata and ellagic acid (EA) (−9.2 kcal/mol) from Bridelia retusa had lower binding energy than moxalactam (-8.4 kcal/mol). P7G and EA formed 5 (Ser62, Asn125, Asn163, Thr209, and Ser230) and 4 (Lys65, Ser123, Asn125, and Glu159) conventional hydrogens bonds with the active site residues. 100 ns MD simulations revealed that moxalactam and P7G (but not EA) were able to form a stable complex. The binding free energy calculations further revealed that P7G (−59.6526 kcal/mol) formed the most stable complex with β-lactamase when compared to moxalactam (−46.5669 kcal/mol) and EA (−28.4505 kcal/mol). The HOMO-LUMO and other DFT parameters support the stability and chemical reactivity of P7G at the active site of β-lactamase. P7G passed all the toxicity tests and bioavailability tests indicating that it possesses drug-likeness. Among the studied compounds, we identified P7G of A. pennata as the most promising phytocompound to combat antibiotic resistance by potentially inhibiting the β-lactamase of P. aeruginosa.
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is a global pandemic infecting the respiratory system through a notorious virus known as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Due to viral mutations and the risk of drug resistance, it is crucial to identify new molecules having potential prophylactic or therapeutic effect against SARS-CoV-2 infection. In the present study, we aimed to identify a potential inhibitor of SARS-CoV-2 through virtual screening of a compound library of 470 quercetin derivatives by targeting the main protease-Mpro (PDB ID: 6LU7). The study was carried out with computational techniques such as molecular docking simulation studies (MDSS), molecular dynamics (MD) simulations, and molecular mechanics generalized Born surface area (MMGBSA) techniques. Among the natural derivatives, compound 382 (PubChem CID 65604) showed the best binding affinity to Mpro (−11.1 kcal/mol). Compound 382 interacted with LYS5, TYR126, GLN127, LYS137, ASP289, PHE291, ARG131, SER139, GLU288, and GLU290 of the Mpro protein. The SARS-CoV-2 Mpro-382 complex showed acceptable stability during the 100 ns MD simulations. The SARS-CoV-2 Mpro-382 complex also showed an MM-GBSA binding free energy value of-54.0 kcal/mol. The binding affinity, stability, and free energy results for 382 and Mpro were better than those of the native ligand and the standard inhibitors ledipasvir and cobicistat. The conclusion of our study was that compound 382 has the potential to inhibit SARS-Cov-2 Mpro. However, further investigations such as in-vitro assays are recommended to confirm its in-silico potency.
Article
Full-text available
A novel coronavirus responsible of acute respiratory infection closely related to SARS-CoV has recently emerged. So far there is no consensus for drug treatment to stop the spread of the virus. Discovery of a drug that would limit the virus expansion is one of the biggest challenges faced by the humanity in the last decades. In this perspective, to test existing drugs as inhibitors of SARS-CoV-2 main protease is a good approach. Among natural phenolic compounds found in plants, fruit, and vegetables; flavonoids are the most abundant. Flavonoids, especially in their glycosylated forms, display a number of physiological activities, which makes them interesting to investigate as antiviral molecules. The flavonoids chemical structures were downloaded from PubChem and protease structure 6LU7 was from the Protein Data Bank site. Molecular docking study was performed using AutoDock Vina. Among the tested molecules Quercetin-3-O-rhamnoside showed the highest binding affinity (-9,7 kcal/mol). Docking studies showed that glycosylated flavonoids are good inhibitors for the SARS-CoV-2 protease and could be further investigated by in vitro and in vivo experiments for further validation. MD simulations were further performed to evaluate the dynamic behavior and stability of the protein in complex with the three best hits of docking experiments. Our results indicate that the rutin is a potential drug to inhibit the function of Chymotrypsin-like protease (3CL pro) of Coronavirus.
Article
Full-text available
Coronavirus disease 2019 is a newly emerging infectious disease currently spreading across the world. It is caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The spike (S) protein of SARS-CoV-2, which plays a key role in the receptor recognition and cell membrane fusion process, is composed of two subunits, S1 and S2. The S1 subunit contains a receptor-binding domain that recognizes and binds to the host receptor angiotensin-converting enzyme 2, while the S2 subunit mediates viral cell membrane fusion by forming a six-helical bundle via the two-heptad repeat domain. In this review, we highlight recent research advance in the structure, function and development of antivirus drugs targeting the S protein.
Article
Full-text available
COVID-19 is a viral disease caused by a new severe acute respiratory syndrome (SARS-CoV-2), which has quickly resulted in a pandemic. As a great threat to global public health, the development of a treatment has become vital, and a rush to find a cure has mobilized researchers from all areas across the world. Synthetic drugs, such as hydroxychloroquine, have gained attention. However, the efficacy of repositioned drugs is still under evaluation, and besides, some severe side effects are a cause for concern. This emphasizes the urgency for treatment options, which can be both safe and effective. With this in mind, natural products could be an important resource in the development of COVID-19 treatment, as they have already contributed in the past to treatments against other viruses, such as HIV, MERS-CoV, and influenza. Natural products are described long term as bioactive substances and some phytochemical classes such as flavonoids, alkaloids, and peptides are known antiviral bioproducts, and have been virtually tested with success against COVID-19. However, important issues still need to be addressed as to their bioavailability and true efficacy in vivo. This review intends to systematically evaluate the natural metabolites that could potentially be used against this new disease looking at their natural sources, mechanism of action and previous pharmacological usages. The aim is to provide a starting point for this research area in order to speed up the establishment of anti-SARS-CoV-2 bioproducts.
Article
Full-text available
SARS-CoV-2 (2019-nCoV) emerged in 2019 and proliferated rapidly across the globe. Scientists are attempting to investigate antivirals specific to COVID-19 treatment. The 2019-nCoV and SARS-CoV utilize the same receptor of the host which is COVID-19 of the main protease (Mpro).COVID-19 caused by SARS-CoV-2 is burdensome to overcome by presently acquired antiviral candidates. So the objective and purpose of this work was to investigate the plants with reported potential antiviral activity. With the aid of in silico techniques such as molecular docking and druggability studies, we have proposed several natural active compounds including glycyrrhizin, bicylogermecrene, tryptanthrine, β-sitosterol, indirubin, indican, indigo, hesperetin, crysophanic acid, rhein, berberine and β-caryophyllene which can be encountered as potential herbal candidate exhibiting anti-viral activity against SARS-CoV-2. Promising docking outcomes have been executed which evidenced the worthy of these selected herbal remedies for future drug development to combat coronavirus disease. Graphic Abstract
Article
Full-text available
Coronavirus are the causative agents in many globally concerning respiratory disease outbreaks such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and coronavirus disease-2019 (COVID-19). It is therefore important that we improve our understanding of how the molecular components of the virus facilitate the viral life cycle. These details will allow for the design of effective interventions. Krichel and coauthors in their article in the Biochemical Journal provide molecular details of how the viral polyprotein (nsp7–10) produced from the positive single stranded RNA genome, is cleaved to form proteins that are part of the replication/transcription complex. The authors highlight the impact the polyprotein conformation has on the cleavage efficiency of the main protease (Mpro) and hence the order of release of non-structural proteins 7–10 (nsp7–10) of the SARS-CoV. Cleavage order is important in controlling viral processes and seems to have relevance in terms of the protein–protein complexes formed. The authors made use of mass spectrometry to advance our understanding of the mechanism by which coronaviruses control nsp 7, 8, 9 and 10 production in the virus life cycle.
Article
Full-text available
The 2019‑novel coronavirus (nCoV) is a major source of disaster in the 21th century. However, the lack of specific drugs to prevent/treat an attack is a major need at this current point of time. In this regard, we conducted a systematic review to identify major druggable targets in coronavirus (CoV). We searched PubMed and RCSB database with keywords HCoV, NCoV, corona virus, SERS‑CoV, MERS‑CoV, 2019‑nCoV, crystal structure, X‑ray crystallography structure, NMR structure, target, and drug target till Feb 3, 2020. The search identified seven major targets (spike protein, envelop protein, membrane protein, protease, nucleocapsid protein, hemagglutinin esterase, and helicase) for which drug design can be considered. There are other 16 onstructural proteins (NSPs), which can also be considered from the drug design perspective. The major structural proteins and NSPs may serve an important role from drug design perspectives. However, the occurrence of frequent recombination events is a major deterrent factor toward the development of CoV‑specific vaccines/drugs.
Article
Introduction: COVID-19 outbreak has infected 34.20 million people with 1019 thousand deaths in more than 125 countries till 30 September 2020. Due to the unavailability of vaccine or targeted novel drug therapy against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), screening of existential medical treatments facilitates identification of promising drugs for the treatment and management of COVID-19. Areas covered: The review article highlights repurposing of antiviral, antimalarial, antineoplastic, antidiabetic, analgesic, and immunomodulatory drugs. Furthermore, clinical trials, in-vitro studies, benefits, adverse effects, toxicities, mechanisms of action, and regulatory status of drugs are covered in this article. Expert opinion: Lack of conclusive results from randomized clinical trials indicates absence of specific drugs for treatment of COVID-19. Unavailability of complete data regarding safety, efficacy, and adverse reactions of drugs restricts the recommendation of clinical advice on dose and duration of the drug therapy. Remdesivir and favipiravir show promising outcomes but more clinical evidence is required for use in large populations. Experimental and repurposed drug therapies targeting spike and envelope proteins, Mpro, 3CLpro and PLpro enzymes, and RdRp and TMPRSS2 genes show capability to produce effective anti-SARS-CoV-2 action. Development of vaccine against SARS-CoV-2 will offer long-term solution to terminate spread of this global pandemic.
Article
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible and pathogenic coronavirus that emerged in late 2019 and has caused a pandemic of acute respiratory disease, named ‘coronavirus disease 2019’ (COVID-19), which threatens human health and public safety. In this Review, we describe the basic virology of SARS-CoV-2, including genomic characteristics and receptor use, highlighting its key difference from previously known coronaviruses. We summarize current knowledge of clinical, epidemiological and pathological features of COVID-19, as well as recent progress in animal models and antiviral treatment approaches for SARS-CoV-2 infection. We also discuss the potential wildlife hosts and zoonotic origin of this emerging virus in detail. In this Review, Shi and colleagues summarize the exceptional amount of research that has characterized acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 2019 (COVID-19) since this virus has swept around the globe. They discuss what we know so far about the emergence and virology of SARS-CoV-2 and the pathogenesis and treatment of COVID-19.
Article
The study aims to evaluate the potency of two hundred natural antiviral phytocompounds against the active site of the Severe Acquired Respiratory Syndrome - Coronavirus − 2 (SARS-CoV-2) Main-Protease (Mpro) using AutoDock 4.2.6. The three- dimensional crystal structure of the Mpro (PDB Id: 6LU7) was retrieved from the Protein Data Bank (PDB), the active site was predicted using MetaPocket 2.0. Food and Drug Administration (FDA) approved viral protease inhibitors were used as standards for comparison of results. The compounds theaflavin-3-3’-digallate, rutin, hypericin, robustaflavone, and (-)-solenolide A with respective binding energy of −12.41 (Ki = 794.96 pM); −11.33 (Ki = 4.98 nM); −11.17 (Ki = 6.54 nM); −10.92 (Ki = 9.85 nM); and −10.82 kcal/mol (Ki = 11.88 nM) were ranked top as Coronavirus Disease − 2019 (COVID-19) Mpro inhibitors. The interacting amino acid residues were visualized using Discovery Studio 3.5 to elucidate the 2-dimensional and 3-dimensional interactions. The study was validated by i) re-docking the N3-peptide inhibitor-Mpro and superimposing them onto co-crystallized complex and ii) docking decoy ligands to Mpro. The ligands that showed low binding energy were further predicted for and pharmacokinetic properties and Lipinski’s rule of 5 and the results are tabulated and discussed. Molecular dynamics simulations were performed for 50 ns for those compounds using the Desmond package, Schrödinger to assess the conformational stability and fluctuations of protein-ligand complexes during the simulation. Thus, the natural compounds could act as a lead for the COVID-19 regimen after in-vitro and in- vivo clinical trials. Communicated by Ramaswamy H. Sarma
Article
Several corona viral infections have created serious threats in the last couple of decades claiming the death of thousands of human beings. Recently, corona viral epidemic raised the issue of developing effective antiviral agents at the earliest to prevent further losses. Natural products have always played a crucial role in drug development process against various diseases which resulted in screening of such agents to combat emergent mutants of corona virus. This review focuses on those natural compounds that showed promising results against corona viruses. Although inhibition of viral replication is often considered as a general mechanism for antiviral activity of most of the natural products, studies have shown that some natural products can interact with key viral proteins that are associated with virulence. In this context, some of the natural products have antiviral activity in the nanomolar concentration (e.g., lycorine, homoharringtonine, silvestrol, ouabain, tylophorine and 7‐methoxycryptopleurine), and could be leads for further drug development on their own or as a template for drug design. In addition, a good number of natural products with anti‐corona virus activity are the major constituents of some common dietary supplements which can be exploited to improve the immunity of the general population in certain epidemics. This article is protected by copyright. All rights reserved.