ArticlePDF Available

Molecular dynamics simulation and docking analysis of NF-κB protein binding with sulindac acid

Authors:

Abstract

It is of interest to document the Molecular Dynamics Simulation and docking analysis of NF-κB target with sulindac sodium in combating COVID-19 for further consideration. Sulindac is a nonsteroidal anti-inflammatory drug (NSAID) of the arylalkanoic acid class that is marketed by Merck under the brand name Clinoril. We show the binding features of sulindac sodium with NF-κB that can be useful in drug repurposing in COVID-19 therapy.
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
170
www.bioinformation.net
Research Article
Volume 18(3)
Received February 24, 2022; Revised March 24, 2022; Accepted March 31, 2022, Published March 31, 2022 DOI: 10.6026/97320630018170
Declaration on Publication Ethics:
The author’s state that they adhere with COPE guidelines on publishing ethics as described elsewhere at https://publicationethics.org/.
The authors also undertake that they are not associated with any other third party (governmental or non-governmental agencies) linking
with any form of unethical issues connecting to this publication. The authors also declare that they are not withholding any information
that is misleading to the publisher in regard to this article.
Declaration on official E-mail:
The corresponding author declares that lifetime official e-mail from their institution is not available for all authors
License statement:
This is an Open Access article which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly credited. This is distributed under the terms of the Creative Commons Attribution License
Comments from readers:
Articles published in BIOINFORMATION are open for relevant post publication comments and criticisms, which will be published
immediately linking to the original article without open access charges. Comments should be concise, coherent and critical in less than 1000
words.
Edited by P Kangueane
Citation: Ahmad et al. Bioinformation 18(3): 170-179 (2022)
Molecular dynamics simulation and docking analysis
of NF-κB protein binding with sulindac acid
Shaban Ahmad1, 2,#, Piyush Bhanu3,#, Jitendra Kumar4,#, Ravi Kant Pathak5, Dharmendra Mallick6,
Akshay Uttarkar7, Vidya Niranjan8 & Vachaspati Mishra9,*
1International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; 2Department of
Computer Science, Jamia Milia Islamia, New Delhi 110025, India; 3Xome Life Sciences, Bangalore Bioinnovation Centre, Helix Biotech Park,
Bengaluru 560100, Karnataka, India; 4Bangalore Bioinnovation Centre (BBC), Helix Biotech Park, Electronics City Phase 1, Bengaluru
560100, Karnataka, India; 5School of Bioengineering and Biosciences, Lovely Professional University, JalandharDelhi Grand Trunk Rd,
Phagwara 144001, Punjab, India; 6Department of Botany, Deshbandhu College, University of Delhi, Delhi 110019, India; 7Department of
Biotechnology, RV College of Engineering, RV Vidyanikethan Post, Mysuru Road, Bengaluru 560059, India; 8Department of
Biotechnology, RV College of Engineering, RV Vidyanikethan Post, Mysuru Road, Bengaluru 560059, India; 9Department of Botany,
Hindu College, University of Delhi, Delhi 110007, India;*Correspondence, #equal contribution.
Author contacts:
Shaban Ahmad - E-mail: shaban.12082000@gmail.com
Piyush Bhanu - E-mail: pb.inbusiness@gmail.com
Jitendra Kumar - E-mail: director@bioinnovationcentre.com
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
171
Ravi Kant Pathak - E-mail: ravibt36@gmail.com
AkshayUttarkar - E-mail: akshayuc@rvce.edu.in
Vidya Niranjan - E-mail: vidya.n@rvce.edu.in
Vachaspati Mishra - E-mail: mishravachaspati31@gmail.com
Abstract:
It is of interest to document the Molecular Dynamics Simulation and docking analysis of NF-κB target with sulindac sodium in combating
COVID-19 for further consideration. Sulindac is a nonsteroidal anti-inflammatory drug (NSAID) of the arylalkanoic acid class that is
marketed by Merck under the brand name Clinoril. We show the binding features of sulindac sodium with NF-κB that can be useful in
drug repurposing in COVID-19 therapy.
Keywords: NF-κB; molecular dynamics simulation; COVID-19; SARS-CoV2; sulindac sodium
Background:
Sulindac is a non-steroidal anti-inflammatory agent (NSAIA) that
can block nuclear factor-κB (NF-κB), a transcription factor (TF)
located primarily in the cytoplasm in a complex while in an inactive
state. Activation of this TF makes it transition from a latent
cytoplasmic form to a nuclear DNA binding state [1] and has been
demonstrated to have a role in the prevention of colon cancer [2] by
its inhibition [1, 3].This TF is known to selectively inhibit
interferon-gamma-induced expression of the chemokine CXCL9
gene in mouse macrophages. [4] The growth inhibitory and anti-
inflammatory properties of sulindac are possibly due to its ability
to reduce prostaglandin synthesis by cyclooxygenase inhibition [2].
Furthermore, Yamamoto et al. [2] have demonstrated that aspirin
and sodium salicylate inhibited the activity of an IκB kinase β
(IKKβ) that is required to activate the NF-κB pathway, which leads
to nuclear translocation of NF-κB, where it binds to its cognate
DNA and activates transcription of a wide variety of genes
involved in host immunity, inflammation, cell proliferation and
apoptosis [2, 3]. Various NF-κB inducers are known to be highly
variable and include aggregates of bacterial lipopolysaccharides,
ionizing radiation, reactive oxygen species (ROS), cytokines, such
as tumour necrosis factor alpha (TNF-α) and interleukin 1-beta (IL-
1β) and viral DNA and RNA [4]. Upon activation, NF-κB promotes
the gene expression of a broad range of cytokines (e.g., IL-1, IL-2,
IL-6, IL-12, TNF-α, LT-α, LT-β, and GM-CSF), chemokines (e.g., IL-
8, MIP-1, MCP1, RANTES, and eotaxin), adhesion molecules (e.g.,
ICAM, VCAM, and E-selectin), acute phase proteins (e.g., serum
amyloid A: SAA), and inducible effector enzymes (e.g., inducible
nitric oxide synthase: iNOS and cyclooxygenase-2: COX-2). The TF
NF-κB can be considered as a “quick action” primary transcription
factor that is able to regulate myriad of cellular responses,
including the host’s early innate immune response to infection, and
is also associated with chronic inflammatory states, viral infections,
septic shock syndrome and multiorgan failure [5, 6] viz-a-viz
ascribed as an anti-apoptotic TF [7].The constitutive activation of
NF-κB pathways has been responsible for stimulation of
inflammatory diseases, such as multiple sclerosis and rheumatoid
arthritis [8, 9]. Furthermore, p38 MAPK-based activation of NF-kB
has also been reported [9]. Drug induced suppression of NF-kB has
been found to have a strong potential in proapoptotic signal
modulation therapy [10].
Recently, a study utilizing molecular dynamics simulations (MDS) -
based analysis targeting human angiotensin-converting enzyme 2
(hACE2) as a receptor showed interesting results on development
of a lead candidate to be used later as a therapeutic drug against
COVID-19 [11] Though in the current scenario, the spread of
COVID 19-based pandemic has slowed down to a great extent, but
its resurgence cannot be ruled out. Due to the lack of adequate
specific treatments for COVID-19, there is an urgency to develop or
repurpose drugs to help end the epidemic completely. The drug
discovery process has received a great impetus with the emergence
of computer-aided drug discovery (CADD) and has been
instrumental over the last decade in exploring protein inhibitors in
protein-drug interactions and proteinprotein interactions [12].
Since the process involved in the development of a candidate drug
into an approved drug is lengthy and expensive, a combination of
computational methodologies such as virtual screening, docking,
molecular dynamics simulation, and binding free energy evaluation
contributes to identifying potential drug candidates from
compound libraries [13].To date, MDS and structure-based virtual
screening studies have been carried out to understand the SARS-
CoV-2 spike protein functions [14-16], role of repurposed protease
inhibitors in blocking SARS-CoV-2 [17], effects of some antiviral
drugs on SARS-CoV-2 [18], effect of temperature on the structure of
the spike protein [19], SARS-CoV-2-targetted potent drugs that can
be effective in controlling COVID-19 [20, 21] as well as many other
aspects of biology qualifying to control SARS-CoV-2 progression.
However, the rate of occurrence of different pathological human
phenotypes and their heterogeneous lethality rates arising from
constant mutations of SARS-CoV-2 [22, 23] indicate serious
problems that need to be appropriately dealt with. A paradigm
shift in the strategy on targeting COVID-19 borne biomolecules-for
therapeutic purposes to human-based molecular biomarkers is
much necessitated in the current scenario, since the current
approaches based on former fail to yield appreciable outcomes. A
recent study revealed the importance of the NF-κB pathway in
developing therapy regimens for critical COVID-19 patients [24].
Other reports have also substantiated the above fact by arguing that
therapeutic benefits of NF-κB inhibitors, including dexamethasone,
a synthetic form of glucocorticoid, have increasingly been realized
now [17]. Abnormal activation of NF-κB resulting from SARS-CoV-
2 infection might be associated with the pathogenic profile of
immune cells, cytokine storms and multiorgan defects [23].
Therefore, we currently explored the structural details of NF-κB via
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
172
an in silico approach wherein we pinpointed the protein pockets for
the binding of SARS-CoV-2 spike proteins. A hypothetical
explanation currently generated is that upon entering the human
body, the COVID-19 viral protein might be triggering the action of
the NF-κB directly or through activation of IKK, the kinase
phosphorylating IκBs that otherwise remain bound with NF-κB to
keep it in an inactive state in the cytoplasm. This argument is made
in the light of the fact that another complex member, IκBα is known
to be phosphorylated by specific kinases at two sites near the N-
terminus (Ser-32 and Ser-36), while the phosphorylated protein is
then ubiquitinated at Lys-21 and Lys-22, leading to proteosome-
mediated degradation [24] and release of NF-κB from the complex.
The removal of IκB unmasks the nuclear localization sequence
(NLS) of NF-κB and allows its translocation to the nucleus [24].
Thus, IκBα is a multifunctional inhibitor of NF-κB that blocks
nuclear translocation, DNA binding, and phosphorylation by
protein kinase A (PKA). Inhibition of NF-kB blocks the NF-κB-
mediated deactivation of immune and inflammatory responses to
stimuli, such as cytokines or bacterial/viral infection products [24].
Along with NF-κB, sulindac has been of interest because of its
many roles that includes also as a chemo preventive agent for
adenomatous colorectal polyps and colon cancer [25] We show here
that sulindac is able to bind specifically to a pocket on NF-κB, a
concept that becomes theoretically important for exploring its
therapeutic potential [26- 29].
Figure 1: Root mean square deviation (RMSD) of the protein and
ligand after the initial RMSD values were stabilized. This plot
shows RMSD values for the protein on the left Y-axis, whereas for
the ligand, these values are indicated on the right Y-axis. The
RMSD graph for c-αis shown in blue colour, and for ligand fit on
the protein, it is in red colour.
Materials and Methods:
Protein and Ligand Preparation:
Coordinates of the X-ray diffraction-based crystal structure of the I-
κ-B-α/NF-κB complex with solvents (PDB ID: 1NFI) at a resolution
of 2.70 Å were downloaded from the Protein Data Bank (PDB).
Solvent molecules and chains A, B, and F were removed during
protein preparation using the Protein Preparation wizard from
Schrodinger Maestro (Schrodinger, LLC, and New York, NY, USA).
The remaining structures were processed using the Protein
Preparation wizard using appropriate methodologies. The SARS-
CoV-2 structure possessing a loop (residues 376381) was missing
in the PDB structure and hence was modelled using Schrödinger’s
Prime module. Hydrogen atoms were incorporated, and a standard
protonation state at pH 7 was used. Bond orders were assigned
using the chemical components dictionary (CCD) database. The
heterostate was generated using Epik with a pH of 7 (±2.0), and
Prime was used to fill the missing chains and loops. While refining
the structure, PROPKA with pH 7.0 was used along with the
sample water orientation. While minimizing the structure, a root
mean square deviation (RMSD) of 0.30 Å with the OPLS_2005 force
field was used. The ligand was selected based on published
information regarding its use as a therapeutic molecule for various
human diseases. The ligand was prepared using the Ligprep
wizard of Schrodinger with the OPLS_2005 force field; the Epik job
was submitted with the metal-binding site and included the
original state. Thirty-two stereoisomers were assigned per ligand
with specified chirality’s. Glide was used to filter the search to
locate the ligand in the active-site region of the receptor. The shape
and properties of the receptor were represented on a grid to
provide a more accurate scoring of the ligand poses. The docked
complexes were superimposed on the original crystal structure to
calculate the RMSD (Figure 1).
Virtual Screening and Molecular Docking:
The Glide grid generator was used for generating the grid for blind
docking. A Schrodinger virtual screening workflow (VSW) was
used to score the virtual screening with default parameters
employing the Glide program of Schrödinger. Use of HTVS mode
allowed the elimination of most of the stereoisomers, and only 10%
of the total ligands could be retained that passed the screening.
Ligands following QikProp and Lipinski’s rule were filtered for
docking. Docking in the VSW was performed with Epik state
penalties and further passed through HTVS, SP and XP docking
modes. The interactions of the selected ligand and protein docked
complexes were analyzed by a pose viewer.
Molecular Dynamics Simulation:
To study the dynamic behavior of the protein complex under
simulated physiological conditions, MDSs of the protein-ligand
(PL) complex were performed using Desmond, which is available
with Schrodinger Maestro (v12.5). The PL complex (9873 atoms)
was solvated in a 10 × 10 × 10 Å orthorhombic periodic box built
with TIP3P water molecules [30]. The whole system was
neutralized by adding an appropriate number of 6 Na+ counterions.
This solvated system was energy minimized and position
restrained with OPLS_2005 as the force field. Furthermore, 100 ns of
MDS was carried out at 1 atm pressure and 300 K temperature,
implementing the NPT ensemble with a recording interval of 100
ps, resulting in a total of 1000 read frames. Finally, various
parameters of the MDS, such as ligand-binding site analysis,
RMSD, root mean square fluctuation (RMSF), PL contacts,
secondary structure element (SSE) analysis, etc., were also analyzed
to check the stability, compactness, structural fluctuations and PL
interactions in the solvated system (Figure 2).
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
173
Interaction Analyses
Structural Data:
The PDB was used to extract all 3-D structural information. The X-
ray diffraction-based crystal structure of the I-κ-B-α/NF-κB
complex with solvents (PDB ID: 1NFI) at a resolution of 2.70 Å was
used for this study. Solvents and chains A, B, and F were removed
during protein preparation. The structure of the SARS-CoV-2 spike
glycoprotein (closed state) solved using electron microscopy (PDB
ID: 6VXX) with a resolution of 2.80 Å was obtained. Furthermore,
all water molecules were removed from both structural data sets.
The educational version of PyMOL (The PyMOL Molecular
Graphics System, v1.2r3pre, Schrödinger, LLC) was used to
generate the images derived to understand and analyse the
structure and inter chain interaction information.
Molecular Docking:
The ligand sulindac chosen in the current study for the mentioned
reasons has its docking score and binding affinity shown in Figure
3. The complex of the drug molecule docked in the pocket of NF-κB
was designated S0 (Figure 4A) and was taken as the reference for
further interaction analysis.
Pocket Analysis:
The molecule NF-κB was found to having multiple pockets, as
calculated using the CASTp 3.0 server [30, 31] Three largest pockets
(based on the accessible area) of these were considered for analysis.
The residues present in these pockets interacted with the spike
protein on exposure changing the area of the pockets significantly
due to this interaction and also upon the drug interaction or the
combination of the spike protein and the drug together.
Protein-Protein Docking:
Protein-protein docking studies were carried out using ClusPro 2.0
[32] with minor changes following the protocols used in Jorgensen
et al. [33]. This job was carried out to understand the interaction of
the SARS-CoV-2 spike glycoprotein with NF-κB in the presence and
absence of the drug molecule. The best pose of the spike protein
binding with the NF-κB-IkB complex was selected based on the
cluster size, which is how the models are ranked in ClusPro. S1
(Figure 4B) represents the complex of NF-κB with the SARS-CoV-2
spike glycoprotein, and S2 (Figure 4C) represents the complex of S0
with the SARS-CoV-2 spike glycoprotein.
LigPlot+ v.2.2 [34] was used to find the ligand-protein interaction of
complex S0 (Figure 2). PDBsum was used to calculate the
interaction between NF-κB and IkB (Figure 5A, 5B), the interaction
between the spike protein and NF-κB (Figure 5B) and the
interaction between the spike protein and NF-κB docked with drug
molecules. PyMOL was used to analyse the structural differences
imposed on NF-κB when it interacted with the drug molecule and
the spike protein in the presence of the drug molecule.
Figure 2: RMSF of the protein backbone and ligand complex. Red
colour shows the B factor means of the PDB structure, and green
colour indicates the interaction of the ligand with the protein.
MM-GBSA Studies:
Prime MM-GBSA (molecular mechanics, the generalized born
model and solvent accessibility method) calculations were
performed with the docked complex of receptor and sulindac. The
implicit solvent model used is VSGB [35] which has an efficient
approximation of the solvation free energy with Surface
Generalized Born (SGB) over Vacuum and chloroform solvation
models. The following calculations were made
1 Rec Strain = Receptor (from optimized complex) − Receptor
2 Lig Strain = Ligand (from optimized complex) − Ligand
3 MMGBSA dG Bind = Complex − Receptor − Ligand
4 MMGBSA dG Bind (NS) = Complex Receptor (from optimized complex)
Ligand (from optimized complex) = MMGBSA dG Bind
Rec Strain - Lig Strain:
NS here refers to binding or interaction energy without any
involvement of conformational changes occurring in the receptor or
ligand. The potential energy of the complex will be reported in
kcal/mol.
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
174
Figure 3: (A) Diagram showing detailed ligand atom interactions
with the protein residues. Interactions that occur more than 7.0% of
the simulation time in the selected trajectory from 0.00 through
100.00 ns. The highlighted structure represents the ligand (drug)
molecule. The interacting residues are shown in circular shapes
with the representation of chains as C (representing residues of the
NFkB molecule) and E (representing residues of IkBα). Circles with
H2O represent the water-mediated H-bond interactions. (B) A
timeline representation of the interactions and contacts (H-bond,
hydrophobic, ionic, and Water Bridge) summarized in the above
figure. The top panel shows the total number of specific contacts
that the protein makes with the ligand over the course of the
trajectory. The bottom panel shows specific residues interacting
with the ligand in each trajectory frame. Some residues make more
than one specific contact with the ligand, which is represented by a
darker shade of orange according to the scale shown to the right of
the plot.
Results and Discussion:
MD Simulation Studies:
The complex RMSD in the initial phase at 1.63 Å went to 4.11 Å
while stabilizing the structure for 100 ns of simulation. Initially, at
up to 50 ns, the RMSD did not fluctuate much, but after that, the
stability fluctuated for 10 ns and stabilized subsequently (Figure 1).
The RMSF plot analysis displayed minimal fluctuations in the
protein structures (Figure 2). Furthermore, minimal fluctuations
were also observed in the PL complex, and the RMSF plot showed
fluctuations in some regions of the protein. The results of the
residue interaction analysis after docking are highlighted with
specific colours based on the self-explanatory features (Figure3).
The docking interactions of sulindac (Figure 4A, 4B) showed that it
docked in the largest pocket of the NF-κB-IkB complex that
contains most of the residues interacting with the spike protein. It is
possible that drug binding to the complex interferes with the above
interaction of the spike protein, however, needs to be ascertained
with more robust wet laboratory-based experimental designs. The
sulindac was found to bind to other pocket than the one occupied
by NF-κB-IkB complex (Figure 4C). Moreover, data shows the
importance of NF-κB as a suitable drug receptor for COVID-19 for
its proven roles as molecular targets in drug discovery-based in
silico studies (Table 1).
The interaction made by the drug molecule with the residues of the
active site cavity of NF-kB, IkB and P50 peptide chains is dynamic
in nature and formed, broke and reformed during the simulation
duration (Figure 3). A brief duration of 20 ns was observed when
no interaction occurred between the drug molecule and the NF-kB
active site residues. Although there was an interaction between the
ligand (drug) molecule and residues of NF-kB (chain C) and IkB
(chain E) in the first 50 ns, the interaction between the drug and IkB
(chain E) disappeared after 50 ns. This loss of interaction
strengthened the ligand-NFkB interaction.
Table 1: The binding affinities of various drugs with NF-κB are shown. This shows that NF-κB is a candidate receptor in various in silico drug-based interventions that may have
potential for being a therapeutic target.
Sl.
No.
Bioactive compound/ligand
Binding pocket
Outcomes achieved/remarks
References
1
Piperine and piperlongumine
DNA binding sites
Docking results of the analogues had higher affinity towards NF-
κB and IL-1β than natural molecules
[36]
2
71 anti-cancer compounds along with
dipeptidyl boronic acid, betulinic acid, and
glycyrrhetinic acid inhibitors
Active sites: Gly47, Ala49, Thr21, and Thr1 of the receptor
Docking was at par with the drug, Bortezomib against
proteasome target of NF-κB pathway
[37]
3
Compound series designed based on
quinazoline scaffold pharmacophore model
p50 subunit of NF-κB
Compounds can inhit NF-κB function and reduce the
proliferation of numerous tumor cell lines especially compound
2a
[38]
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
175
4
P100 peptide
Binding sites of IκB Kinase α(IKK α)
Highlightes the different roles of NIK and IKK in regulating p100
processing and understands the mechanisms that mediate
control of p100 processing
[39]
5
Four series of 24 thienopyrimidine/N-
methylpicolinamide derivatives substituted
with pyrimidine
Active sites of TAK1 kinases
Compound 38 inhibited TAK1 phosphorylation, I κ
degradation, TNF-α-induced IκBα phosphorylation, , reduce the
expression of p65 and p65 phosphorylation
[40]
6
Growth arrest specific facgtor 6 (Gas6)
Autophosphorylation docking site- Tyr867
Mertk receptor showed disti nct effects for phagocytosis
[41]
7
1, 8-dihydroxy-4-methylanthracene-9, 10-
dione (DHMA) from Luffa acutangal a
DNA-binding region (DBR) of NF-κB
DHMA plays a role in cancer prevention and treatment by
having an altered binding affinities against NF-κB and DNA
[42]
8
Natural compounds found in Cupressus
pyramidalis and Aegle marmelos extracts
DNA binding sites of proteins
These lead molecules down-regulated the expression of IL8 gene
and inhibited the activity of NF-kappaB-50
[43]
9
Phenolic fraction of Citru s maxima
(hesperidin, naringenine, naringin and
dexamethasone)
DNA binding region of NF-κB
Docking studies of the phenolic fraction compounds had an
inhibitory effect against NF-κB
[44]
10
Diphosphorylated 21-mer p100 peptide
model
Four argenine residues Arg285, Arg410, Arg431, Arg521 and
Arg474 in hydrophobe B-TrCP
Peptide p100 disrupts the IκB-α/NF-κB signalling pathway
[45]
11
Small molecule, NSC-127102
Phosphorylation of p65 at serine 276
NSC-127102 hinders IL-8, VCAM-1 expression and
phosphorylation at serine 276
[46]
12
Scopoletin (Pharmacologically active
coumarin)
Keap1, NF-kB, HO-1 and p38MAPk are the targets against
vancomycin nephrotoxicity
Scopoletin helps in protecting renal inury by interferring with
Nrf2/HO-1 and IkBa/p65 NF-kB signalling pathway as well as
resorting the antioxidant defense system
[47]
13
Ergosta-7, 9 (11), 22-trien--ol (EK100)
Interferes with Lipopolysaccharide (LPS) docking to the LPS-
binding protein (LBP), transferred to the cluster of differenti ation 14
(CD14), and bonded to TLR4/myeloid differentiation-2 (MD-2) co-
receptors
EK100 equally attenuated NF-κB gene expression and TLR4/NF-
κB inflammatory pathway in Drosophila
[48]
Figure 4: X-ray diffraction-based crystal structure of the IκB-α/NF-
κB complex with solvents (PDB ID: 1NFI) at resolution of 2.70 Å,
showing various chains in colour formats representing different
molecules employed in the current study. For example, chain C
(shown as a green cartoon model) represents NF-κB-P65, chain D
(shown in a cyan cartoon model) represents NF-κB-P50, and chain
E (shown in a magenta cartoon model) represents IkBα. (A)
Structure of the complex formed after docking of the drug molecule
with NF-κB. The highlighted region represents the binding pocket
of the drug molecule in the NF-κB molecule. The binding pocket is
represented by the labelled residues (G209, D210, E211, D257, R253,
R259, S269, E285, etc., of the NFkB molecule and I102, N182, Q183,
H184, etc., of the IkBα molecule). (B) Structure of the complex
formed after protein-protein docking of the SARS-CoV-2 spike
glycoprotein (closed state) (PDB ID: 6VXX, shown in orange colour
at a resolution of 2.80 Å) with the NF-κB molecule. The highlighted
region represents the binding pocket of the spike protein in the NF-
κB molecule. The binding pocket is represented by the labelled
residues (G209, D210, E211, D257, R253, R259, S269, E285, etc., of
the NFkB molecule and N182, Q183, H184, etc., of the IkBα
molecule). This information indicates that the drug molecule binds
in the same pocket that the spike protein used to interact with NF-
κB, possibly to activate its translocation to the nucleus. (C)
Structure of the complex formed after protein-protein docking of
the SARS-CoV-2 spike glycoprotein (shown in orange colour) with
the NF-κB molecule (already docked with the drug molecule). The
highlighted region represents the binding pocket of the spike
protein in the NF-κB molecule in the presence of the drug molecule.
The drug molecule occupies the same binding pocket on NF-κB
that is also used by the spike protein during docking interaction,
thus inhibiting the spike protein from interacting with NF-κB at its
designated site. In the interim, the spike protein interacts with NF-
κB at a different site and hence may not be able to activate it.
Currently, these assumptions need validating experimental proof,
which is lacking because of the strict restraint on experimentation
with live samples from active human COVID-19 cases.
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
176
Figure 5: A 3-D structural alignment of the NF-κB-drug complexes
(represented in a red cartoon model) and spike-protein-NF-κB-drug
complexes (represented in a green cartoon model), with the NF-κB
molecule represented in an orange cartoon model. (A) These
alignments show the conformational changes imposed in the
structure of the NF-κB molecule upon binding of the drug and
spike protein. (B) Sequence-based comparison of the interacting
residues of the NF-κB molecule with the drug sulindac sodium
(yellow coloured bar), IkB molecule (green coloured bar) and spike
protein (red coloured bar).
The changes in the 3-D structure of the NF-κB-IkB complex due to
the binding of the drug molecule and spike protein in the presence
of the drug molecule were compared to that of the unbound
complex (Figure 5A). The binding of the drug molecule to the NF-
κB-IkB complex led to an increase in the compactness of the
complex. The area and volume data of the pockets presented in
Figure 5A corroborated this observation. The binding of the spike
protein in the presence of the drug molecule further increased the
compactness of this complex, possibly due to increased interaction
between the chains of the NF-κB-IkB complex. Additionally, we
made a comparison of the amino acid (aa) residues of the spike
protein, MAP kinase and pLC gamma during their interactions
with the residues of the NF-κB molecule to identify if any common
pattern existing in aa selection by sulindac during these
interactions. However, all three sets of interactions, such as NF-κB-
sulindac docked with spike protein, NF-κB sulindac docked with
MAPK and NF-κB sulindac docked with pLC gamma had different
aa residues interacting with the chosen ligand (sulindac)(Figure 6),
revealing all these as independent events. In our studies, MM-
GBSA studies on docked complexes revealed dG bind values that
were reasonably in agreement with ranking based experimental
scores, i.e., binding values We first calculated the MMGBSA dG
Bind = Prime Energy (Optimized Complex) Prime Energy
(Optimized Free Ligand) Prime Energy (Optimized Free
Receptor) and second, MMGBSA dG Bind(NS) = Prime Energy
(Optimized Complex) − Prime Energy (Ligand Geometry From
Optimized Complex) − Prime Energy (Receptor Geometry From
Optimized Complex). The values were found to be −2.15 kcal/mol
for MMGBSA dG bind and 2.71 kcal/mol for MMGBSA dG bind
(NS). The atoms contributing to the binding affinities are shown in
Figure 7.
Figure 6: Comparison of the residues of the spike protein, MAP
kinase and pLC gamma interacting with the residues of the NF-κB
molecule. The underlined residues are in common (black: common
in the s protein and pLC gamma, red: common in the spike protein
and MAP kinase, and blue: common in all three).
Our understanding on the involvement of NF-κB signalling
pathway in COVID-19 is limited, but NF-κB inhibitors have been
increasingly utilized to gain therapeutic benefits in many human
diseases [49, 50]. Abnormal activation of NF-κB in the manifestation
of SARS-CoV-2 infection is reported for the pathogenic profile of
immune cells, cytokine storms and multi organ defects and
cytokine storm alone is capable of triggering excessive
inflammatory response to SARS-CoV-2 and is extremely
responsible for disease severity [51]. Thus, pharmacological
inactivation of the NF-κB, such as proposed currently in the use of
sulindac, NF-κB-kinase interaction either directly or by inhibiting
the dephosphorylation of its inhibitors, such as IKKα, IKKβ and
other associated molecules, strongly represents a potential
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
177
therapeutic target to treat the symptoms of COVID-19 in clinical
trials. A survey of literature since the emergence of COVID-19
clearly reveals that majority of the drugs formulated to tackle
SARS-CoV-2 thus far, have been structured to target virus [52].
However, given the current situation where the virus mutates so
rapidly, our conceptual paradigm on targeting the key viral
proteins through drug intervention need to get changed towards
the host proteins, such as human proteins that hold key portfolio in
SARS-CoV-2 mediated infections. This would possibly take care
of the various mutants of SARS-CoV-2 that have been difficult to
control until now. Furthermore, cytokine storm in COVID-19 [53]
and various drugs that were initially used for therapy [54] need to
be revisited in light of modern researches based on drug
repurposing, which are potentially very effective in treatments of
even the anomalies arising post-COVID-19 infection and can thus
counteract the many aberrant responses thereof.
Figure 7: MM-GBSA energy based visualization of docked complex
highlighting the docked complex of receptor and sulindac sodium
are represented with colour highlights on the ligand atoms
showing contributions towards binding affinity. Blue represents the
receptor and adaptive colors on ligand from Red to Green
showcasing higher negative values to least values, respectively. The
yellow dashed line represents the hydrogen bond with the
receptor.
Author contributions:
S.A.: MDS analysis, Original Draft; P.B.: Docking analysis and
Report; J.K.: Docking analysis supervision, Review and Editing;
R.P.: MDS analysis confirmation and Report; D.M.: Review and
Editing; A.U.: MM-GBSA analysis; V.N.: MM-GBSA report; V.M.:
Conceptualization, Revision and Editing, Conclusion and
Discussion, Formatting the MS, Overseeing the project and the MS
Funding:
The funding acquisition was made from the Bangalore Bio
Innovation Centre, Karanataka Innovation and Technology Society,
Department of Electronics, IT, BT and S&T, Government of
Karnataka, India, towards paying the publication cost.
Acknowledgments:
The authors acknowledge the International Centre for Genetic
Engineering and Biotechnology, New Delhi, for providing facilities
for MDS studies and the Bangalore Bio Innovation Centre,
Department of Electronics, IT, BT and S&T, Government of
Karnataka, India, for funding acquisition towards paying the
publication cost.
Conflicts of interest: The authors declare that there is no conflict of
interest.
References:
[1] Housby JN et al. Cytokine. 1999 11:347. [PMID: 10328874]
[2] Yamamoto Y et al. J Biol Chem. 1999 274:27307. [PMID:
10480951]
[3] Sakaeda Y et al. Biochem Biophys Res Commun. 2006
350:339. [PMID: 17010317]
[4] Tegeder I et al. FASEB J. 2001 15:2057. [PMID: 11641233]
[5] Beg AA et al. Genes Dev. 1992 6:1899. [PMID: 1340770]
[6] Oeckinghaus A & Ghosh S Cold Spring Harb Perspect Biol.
2009 1:a000034. [PMID: 20066092]
[7] Sakaeda Y et al. Biochem Biophys Res Commun. 2006
350:339. [PMID: 17010317]
[8] Li Q & Verma IM Nat. Rev. Immunol. 2002 2:725. [PMID:
12360211]
[9] Liu T et al. Signal Transduct Target Ther. 2017 2:17023.
[PMID: 29158945]
[10] Washo-Stultz D et al. Cancer Lett. 2002 177:129. [PMID:
11825660]
[11] Neufeldt CJ et al. Commun Biol. 2022 5:45. [PMID:
35022513]
[12] Bradbury CM et al. Cancer Res. 2001 61:7689. [PMID:
11606413].
[13] Keretsu S et al. Sci. Rep.2020 10:17716. [PMID: 33077821]
[14] Elmezayen AD et al. J Biomol Struct Dyn. 2020 39:2980.
[PMID: 32306862]
[15] McCarty MF et al. Integr Cancer Ther. 2006 5:252. [PMID:
16880431]
[16] Pablo R Arantes et al. ACS Cent. Sci. 2020 6:1654. [PMID:
33140032]
[17] Al-Karmalawy AA et al. Front Chem. 2021 9:661230. [PMID:
34017819]
[18] Selvaraj C et al. J. Biomol. Struct. Dyn. 2020 39:4582. [PMID:
32567979]
[19] Cardoso WB & Mendanha S J. Mol. Struct. 2021 1225:129.
[PMID: 32863430]
[20] Rath SL & Kumar K Front Mol Biosci. 2020 7:583. [PMID:
33195427]
[21] Lokhande KB et al. J Biomol Struct Dyn.2020 1:12. [PMID:
32815481]
[22] Justo A et al. Sci Rep. 2021 11:17755. [PMID: 34493762]
[23] Hariharan A et al. Inflammopharmacology 2021 29:91.
[PMID: 33159646]
[24] Catanzaro M. Signal Transduct Target Ther. 2020 5:84.
[PMID: 32467561]
[25] Wang X et al. Front. Cell Dev. Biol.2020 8: 574706. [PMID:
33224945]
[26] Wang Q et al. Cell 2020 894:904. [PMID: 32275855]
[27] https://go.drugbank.com/drugs/DB00605
[28] Brunell D et al. Drug Metab. Dispos. 2011 1014:1021. [PMID:
21383205]
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
178
[29] Marchetti M et al. PLoS ONE 2009 4:e5804. [PMID:
19503837]
[30] Binkowski TA et al. Nucleic Acids Res. 2003 31:3352. [PMID:
12824325]
[31] Tian W et al. Nucleic Acids Res. 2018 46:W363. [PMID:
29860391]
[32] Kozakov D et al. Nat Protoc. 2017 12:255. [PMID: 28079879]
[33] Thompson PA et al. Clin Cancer Res. 2021 27:5660. [PMID:
34112707]
[34] Laskowski RA & Swindells MB J. Chem. Inf. Model.2011
51:2778. [PMID: 21919503]
[35] Li J et al. Proteins. 2011 79:2794. [PMID: 21905107]
[36] Zazeri G et al. Molecules. 2020 25:2841 [PMID: 32575582]
[37] Yadav D et al. J Biomol Struct Dyn. 2020 38:3621. [PMID:
31514715]
[38] Xu L & Russu WA Bioorg Med Chem. 2013 21:540. [PMID:
23219854]
[39] Xiao G et al. J Biol Chem. 2004 279:30099. [PMID: 15140882]
[40] Wang L et al. Eur J Med Chem. 2021 223:113576 [PMID:
34153577]
[41] Tibrewal N et al. J Biol Chem. 2008 283:3618. [PMID:
18039660]
[42] Ramar V & Pappu S Comput Biol Chem. 2016 29:35. [PMID:
27061144]
[43] Piccagli L et al. BMC Struct Biol. 2008 8:38. [PMID:
18768082]
[44] Nandeesh R et al. Biomed Pharmacother. 2018 108:1535.
[PMID: 30372855]
[45] Melikian M et al. J Chem Inf Model. 2017 57:223.
[PMID:28004927]
[46] Law M et al. Cancer Lett. 2010 291:217. [PMID: 19910110].
[47] Khalaf MM et al. Int Immunopharmacol. 2022 102:108382.
[PMID: 34848155]
[48] Hsieh WT et al. Int J Mol Sci. 2021 22:6511. [PMID:
34204506]
[49] Li J et al. Proteins 2011 79:2794. [PMID: 21905107]
[50] Park M H & Hong JT Cells 2016 5:15. [PMID: 27043634]
[51] Yu H et al. Signal Transduct. Target Ther. 2020 5:209. [PMID:
32958760]
[52] Castelli V et al. Frontiers in Immunology 2020 11:2132.
[PMID: 32983172]
[53] McKee DL et al. Pharmacological Research 2020 157:104859
[PMID: 32360480]
ISSN 0973-2063 (online) 0973-8894 (print)
©Biomedical Informatics (2022)
Bioinformation 18(3): 170-179 (2022)
179
... The complex nature of this disease demands innovative approaches to treatment and intervention. One promising avenue in cancer therapy is the development of multitargeted inhibitors that simultaneously target various cellular survival and stress response proteins involved in cancer progression [9][10][11][12]. Lung cancer remains a formidable global health challenge, necessitating innovative therapeutic strategies [13]. ...
... The QikProp filter kept filtering the compounds, applied Lipinski's rule, and browsed the receptor grids as a source of protein [50,51]. Furthermore, in the Docking tab, we used the [10,35,36]. We passed the complete filtered compound's library to HTVS, top 10% of HTVS to SP, top 10% of SP to XP, and 100% of XP to MM\GBSA to compute the binding free energy and filter the poses. ...
Article
Full-text available
Lung cancer is a disease in which lung cells grow abnormally and uncontrollably, and the cause of it is direct smoking, secondhand smoke, radon, asbestos, and certain chemicals. The worldwide leading cause of death is lung cancer, which is responsible for more than 1.8 million deaths yearly and is expected to rise to 2.2 million by 2030. The most common type of lung cancer is non-small cell lung cancer (NSCLC), which accounts for about 80% and small cell lung cancer (SCLC), which is more aggressive than NSCLC and is often diagnosed later and accounts for 20% of cases. The global concern for lung cancer demands efficient drugs with the slightest chance of developing resistance, and the idea of multitargeted drug designing came up with the solution. In this study, we have performed multitargeted molecular docking studies of Drug Bank compounds with HTVS, SP and XP algorithms followed by MM\GBSA against the four proteins of lung cancer cellular survival and stress responses, which revealed Mitoglitazone as a multitargeted inhibitor with a docking and MM\GBSA score ranging from − 5.784 to − 7.739 kcal/mol and − 25.81 to − 47.65kcal/mol, respectively. Moreover, we performed pharmacokinetics studies and QM-based DFT analysis, showing suitable candidate and interaction pattern analysis revealed the most count of interacting residues was 4GLY, 5PHE, 6ASP, 6GLU, 6LYS, and 6THR. Further, the results were validated with SPC water model-based MD simulation for 100ns in neutralised condition, showing the cumulative deviation and fluctuation < 2Å with many intermolecular interactions. The whole analysis has suggested that Mitoglitazone can be used as a multitargeted inhibitor against lung cancer—however, experimental studies are needed before human use.
... Ras and related G proteins are relied upon for more broad applications to different compounds that catalyze phosphoryl (-PO(3)2-), including kinases and phosphatases. [13][14][15][16] To comprehend the molecular mechanisms of hidden bortezomib obstruction, we should recognize the potential objective genes differentially expressed in bortezomib-safe leukemia cells versus organ controls. [17][18][19][20] In this study, we extricate the short sequence repeats (SSRs) to comprehend this issue and focus on the medication component with new methodologies. ...
... ese SSRs can serve as biomarkers and be utilized in various drug-related strategies. 13,33,34 e dataset includes various types of trinucleotide repeats, dinucleotide repeats, mononucleotide repeats, 9-nucleotide repeats, and other SSRs, along with their corresponding sequences and positions. ...
Article
Full-text available
Objectives: To compare Ras-related associated with diabetes (RRAD) across different species and to identify specific biomarkers for cancer therapy. Methods: The study involves comparing the coding sequences, genes, messenger ribonucleic acid (RNA), non-coding RNA, open reading frame, short- and long-sequence repeats, and transcription factors of RRAD genes from 82 species. Various tools and software are employed for these comparisons, and evolutionary analysis was carried out to understand the gene's evolutionary history. The data are classified based on forward and reverse sequences. Results: Our analysis indicates that ACTG1 may function as a downstream effector of RRAD, offering potential avenues for diabetes and cancer treatments. By collecting RRAD sequences from 82 species and carrying out comparative genomics, this study provides diverse strategies for developing biomarker-based therapeutics. Furthermore, it suggests using RRAD in other organisms as a model for studying the knockdown effects of specific sequence sets. The study presents RRAD sequences from 82 organisms across different families, contributing to a diverse knowledge base for identifying drug-designing biomarkers. Conclusion: This research offers insights into the potential of RRAD as a therapeutic target in various organisms and highlights the importance of biomarker identification in drug development.
... While they provided some benefits to specific patients, they often fell short of addressing the full spectrum of the disease. The limitations of single-target drugs became apparent as researchers grappled with lung cancer's profound heterogeneity [2][3][4][5]. Lung cancer is not uniform; it encompasses various subtypes with unique genetic and molecular characteristics [6][7][8]. Moreover, lung cancer cells are notorious for their ability to adapt and develop resistance to targeted therapies. ...
Article
Full-text available
Lung cancer, a relentless and challenging disease, demands unwavering attention in drug design research. Single-target drugs have yielded limited success, unable to effectively address this malignancy’s profound heterogeneity and often developed resistance. Consequently, the clarion call for lung cancer drug design echoes louder than ever, and multitargeted drug design emerges as an imperative approach in this landscape, which is done by concurrently targeting multiple proteins and pathways and offering a beacon of hope. This study is focused on the multitargeted drug designing approach by identifying drug candidates against human cyclin-dependent kinase-2, SRC-2 domains of C-ABL, epidermal growth factor and receptor extracellular domains, and insulin-like growth factor-1 receptor kinase. We performed the multitargeted molecular docking studies of Drug Bank compounds using HTVS, SP and XP algorithms and poses filter with MM\GBSA against all proteins and identified DB02504, namely [3-(1-Benzyl-3-Carbamoylmethyl-2-Methyl-1h-Indol-5-Yloxy)-Propyl-]-Phosphonic Acid (3-1-BCMIYPPA) as multitargeted lead with docking and MM\GBSA score range from -8.242 to -6.274 and -28.2 and -44.29 Kcal/mol, respectively. Further, the QikProp-based pharmacokinetic computations and QM-based DFT showed acceptance results against standard values, and interaction fingerprinting reveals that THR, MET, GLY, VAL, LEU, GLU and ASP were among the most interacting residues. The NPT ensemble-based 100ns MD simulation in a neutralised state with an SPC water model has also shown a stable performance and produced deviation and fluctuations <2Å with huge interactions, making it a promising multitargeted drug candidate—however, experimental studies are suggested.
... This comprehensive approach aims to synergistically disrupt multiple cancer-promoting pathways, potentially offering a more effective and holistic treatment strategy. Moreover, by targeting interconnected proteins, a multitargeted drug may reduce the likelihood of cancer cells developing resistance [21][22][23][24]. It is essential to underscore that, while this conceptual approach holds promise, rigorous experimental validation is imperative before any drug can be considered for clinical application. ...
Article
Full-text available
Breast cancer begins in the breast cells, mainly impacting women. It starts in the cells that line the milk ducts or lobules responsible for producing milk and can spread to nearby tissues and other body parts. In 2020, around 2.3 million women across the globe received a diagnosis, with an estimated 685,000 deaths. Additionally, 7.8 million women were living with breast cancer, making it the fifth leading cause of cancer-related deaths among women. The mutational changes, overexpression of drug efflux pumps, activation of alternative signalling pathways, tumour microenvironment, and cancer stem cells are causing higher levels of drug resistance, and one of the major solutions is to identify multitargeted drugs. In our research, we conducted a comprehensive screening using HTVS, SP, and XP, followed by an MM/GBSA computation of human-approved drugs targeting HER2/neu, BRCA1, PIK3CA, and ESR1. Our analysis pinpointed IRESSA (Gefitinib-DB00317) as a multitargeted inhibitor for these proteins, revealing docking scores ranging from −4.527 to −8.809 Kcal/mol and MM/GBSA scores between −49.09 and −61.74 Kcal/mol. We selected interacting residues as fingerprints, pinpointing 8LEU, 6VAL, 6LYS, 6ASN, 5ILE, and 5GLU as the most prevalent in interactions. Subsequently, we analysed the ADMET properties and compared them with the standard values of QikProp. We extended our study for DFT computations with Jaguar and plotted the electrostatic potential, HOMO and LUMO regions, and electron density, followed by a molecular dynamics simulation for 100 ns in water, showing an utterly stable performance, making it a suitable drug candidate. IRESSA is FDA-approved for lung cancer, which shares some pathways with breast cancers, clearing the hurdles of multitargeted drugs against breast and lung cancer. This has the potential to be groundbreaking; however, more studies are needed to concreate IRESSA’s role.
... It acted as NF-κB inhibitor and ABCC1/3 expression enhancer [45]. Molecular docking studies demonstrate that sulindac sodium has NF-κB binding properties that might be valuable for medication repurposing purposes in COVID-19 treatment [46]. Silibinin was launched in 1972 by Madaus. ...
Article
A necessary immune response that promotes survival and preserves tissue homeostasis, inflammation is a frequent characteristic of many disorders. In other circumstances, however, the inflammatory process turns harmful and aids in the progression of a disease. The use of nanomedicines (nanoparticles that are loaded with a therapeutic active principle) to target inflammation through the identification of molecules overexpressed on the surface of activated macrophages or endothelial cells, through increased vasculature permeability, or even through biomimicry offers a promising treatment for inflammatory diseases. The present review study will discuss the main novel nanomedicine approaches, nanoinformatics used in drug discovery and nanomedicine development and also the impact of nanoinformatics on different sectors of healthcare research that have been proposed in the previous decades for the resolution of inflammatory disorders after giving a brief overview of the pathophysiology of inflammation and current therapeutic approaches. It will then concentrate on those currently in clinical trials and ethical considerations. The nanomedicines, along with nanoinformatics, could help to improve delivery efficiency. The nanoinformatics can play a critical role in accelerating research and development in nanomedicine. We suggest that the application of nanoinformatics in this field can lead to novel breakthroughs, better understanding of underlying mechanisms, and ultimately drive innovation. In particular, we observe a potential for the use of nanoinformatics in drug design, delivery systems, and diagnostics. By leveraging the power of data-driven approaches, computational modelling, and artificial intelligence, we can unlock the full potential of nanomedicine and transform the way we approach healthcare.
... To isolate the pure crystal structure of the target protein, heteroatoms and molecules other than amino acids were removed using UCSF Chimera. Protein preparation was conducted with Schr€ odinger's 2022-3 embedded Protein Preparation Wizard (Ahmad et al., 2022). Further proceeds with three necessary steps; import and preprocess, review and modify and refine. ...
... Lung cancer has earned its reputation as one of the most pervasive and challenging diseases worldwide. It is the leading cause of cancer-related mortality, responsible for more deaths than breast, prostate, and colon cancers combined [4][5][6][7]. The disease's pervasiveness knows no boundaries, affecting men and women across diverse populations and age groups. ...
Article
Full-text available
Lung cancer is a pervasive and challenging disease with limited treatment options, with global health challenges often present with complex molecular profiles necessitating the exploration of innovative therapeutic strategies. Single-target drugs have shown limited success due to the heterogeneity of this disease. Multitargeted drug designing is imperative to combat this complexity by simultaneously targeting multiple target proteins and pathways, which can enhance treatment efficacy and overcome resistance by addressing the dynamic nature of the disease and stopping tumour growth and spread. In this study, we performed the molecular docking studies of Drug Bank compounds with a multitargeted approach against crucial proteins of lung cancer such as heat shock protein 5 (BIP/GRP78) ATPase, myosin 9B RhoGAP, EYA2 phosphatase inhibitor, RSK4 N-terminal kinase, and collapsin response mediator protein-1 (CRMP-1) using HTVS, SP with XP algorithms, and poses were filtered using MM\GBSA which identified [3-(1-Benzyl-3-Carbamoylmethyl-2-Methyl-1h-Indol-5-Yloxy)-Propyl-]-Phosphonic Acid (3-1-BenCarMethIn YlPro-Phosphonic Acid) (DB02504) as multitargeted drug candidate with docking and MM\GBSA score ranges from −5.83 to −10.66 and −7.56 to −50.14 Kcal/mol, respectively. Further, the pharmacokinetic and QM-based DFT studies have shown complete acceptance results, and interaction fingerprinting reveals that ILE, GLY, VAL, TYR, LEU, and GLN were among the most interacting residues. The 100 ns MD simulation in the SPC water model with NPT ensemble showed stable performance with deviation and fluctuations <2 Å with huge interactions, making it a promising multitargeted drug candidate; however, experimental studies are needed before use.
Chapter
Computer-aided drug design (CADD) is a rapidly growing field that combines the knowledge of computational chemistry, bioinformatics, and pharmacology to aid in the discovery and development of new drugs. CADD utilizes various computational techniques and tools such as molecular modeling, docking, and machine learning (ML) to predict the properties and interactions of potential drug compounds with their biological targets. One of the significant advantages of CADD is its ability to significantly reduce the time and cost of drug development by allowing for the virtual screening of many compounds in silico instead of relying solely on experimental methods, which can also reduce the number of compounds that must be tested in animal models and clinical trials. As a result, CADD has become an essential tool in modern pharmaceutical research. Molecular dynamics (MD) simulations, which may foretell the motion and interactions of atoms and molecules in a biological system, and docking, which can foretell the binding of a medicinal compound to its target protein, are two examples of CADD techniques. Artificial neural networks (ANNs) and random forests are two ML techniques used in CADD for various tasks, including virtual screening, lead optimization, absorption, distribution, metabolism, and excretion (ADMET) prediction.
Article
Full-text available
Cervical cancer poses a substantial worldwide health challenge, especially in low- and middle-income nations, caused by high-risk types of human papillomavirus. It accounted for a significant percentage of cancer-related deaths among women, particularly in areas with limited healthcare resources, necessitating innovative therapeutic approaches, and single-targeted studies have produced significant results, with a considerable chance of developing resistance. Therefore, the multitargeted studies can work as a beacon of hope. This study is focused on performing the multitargeted molecular docking of FDA-approved drugs with the three crucial proteins TBK1, DNA polymerase epsilon, and integrin α-V β-8 of cervical cancer. The docking studies using multisampling algorithms HTVS, SP, and XP reveal Pixantrone Maleate (DB06193) as a multitargeted inhibitor with docking scores of -8.147, -8.206 and -7.31 Kcal/mol and pose filtration with MM\GBSA computations with scores -40.55, -33.67, and -37.64 Kcal/mol. We also have performed QM-based DFT and pharmacokinetics studies of the compound and compared it with the standard values, which results in the compound being entirely suitable against cervical cancer proteins. The interaction fingerprints have revealed that PHE, VAL, SER and ALA are the residues among most interactions. We also explore the stability of the multitargeted potential of Pixantrone Maleate through 100ns MD simulations and investigate the RMSD, RMSF and intermolecular interactions between all three proteins-ligand complexes. All computational studies favour Pixantrone Maleate as a multitargeted inhibitor of the TBK1, DNA polymerase epsilon, and integrin α-V β-8 and can be validated experimentally before use.
Article
Full-text available
SARS-CoV-2 is a novel virus that has rapidly spread, causing a global pandemic. In the majority of infected patients, SARS-CoV-2 leads to mild disease; however, in a significant proportion of infections, individuals develop severe symptoms that can lead to long-lasting lung damage or death. These severe cases are often associated with high levels of pro-inflammatory cytokines and low antiviral responses, which can cause systemic complications. Here, we have evaluated transcriptional and cytokine secretion profiles and detected a distinct upregulation of inflammatory cytokines in infected cell cultures and samples taken from infected patients. Building on these observations, we found a specific activation of NF-κB and a block of IRF3 nuclear translocation in SARS-CoV-2 infected cells. This NF-κB response was mediated by cGAS-STING activation and could be attenuated through several STING-targeting drugs. Our results show that SARS-CoV-2 directs a cGAS-STING mediated, NF-κB-driven inflammatory immune response in human epithelial cells that likely contributes to inflammatory responses seen in patients and could be therapeutically targeted to suppress severe disease symptoms.
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This disease has spread globally, causing more than 161.5 million cases and 3.3 million deaths to date. Surveillance and monitoring of new mutations in the virus’ genome are crucial to our understanding of the adaptation of SARS-CoV-2. Moreover, how the temporal dynamics of these mutations is influenced by control measures and non-pharmaceutical interventions (NPIs) is poorly understood. Using 1,058,020 SARS-CoV-2 from sequenced COVID-19 cases from 98 countries (totaling 714 country-month combinations), we perform a normalization by COVID-19 cases to calculate the relative frequency of SARS-CoV-2 mutations and explore their dynamics over time. We found 115 mutations estimated to be present in more than 3% of global COVID-19 cases and determined three types of mutation dynamics: high-frequency, medium-frequency, and low-frequency. Classification of mutations based on temporal dynamics enable us to examine viral adaptation and evaluate the effects of implemented control measures in virus evolution during the pandemic. We showed that medium-frequency mutations are characterized by high prevalence in specific regions and/or in constant competition with other mutations in several regions. Finally, taking N501Y mutation as representative of high-frequency mutations, we showed that level of control measure stringency negatively correlates with the effective reproduction number of SARS-CoV-2 with high-frequency or not-high-frequency and both follows similar trends in different levels of stringency.
Article
Full-text available
Ergosta-7, 9 (11), 22-trien-3β-ol (EK100) was isolated from Cordyceps militaris, which has been used as a traditional anti-inflammatory medicine. EK100 has been reported to attenuate inflammatory diseases, but its anti-inflammatory mechanism is still unclear. We were the first to investigate the effect of EK100 on the Toll-like receptor 4 (TLR4)/nuclear factor of the κ light chain enhancer of B cells (NF-κB) signaling in the lipopolysaccharide (LPS)-stimulated RAW264.7 cells and the green fluorescent protein (GFP)-labeled NF-κB reporter gene of Drosophila. EK100 suppressed the release of the cytokine and attenuated the mRNA and protein expression of pro-inflammatory mediators. EK100 inhibited the inhibitor kappa B (IκB)/NF-κB signaling pathway. EK100 also inhibited phosphatidylinositol-3-kinase (PI3K)/Protein kinase B (Akt) signal transduction. Moreover, EK100 interfered with LPS docking to the LPS-binding protein (LBP), transferred to the cluster of differentiation 14 (CD14), and bonded to TLR4/myeloid differentiation-2 (MD-2) co-receptors. Compared with the TLR4 antagonist, resatorvid (CLI-095), and dexamethasone (Dexa), EK100 suppressed the TLR4/AKT signaling pathway. In addition, we also confirmed that EK100 attenuated the GFP-labeled NF-κB reporter gene expression in Drosophila. In summary, EK100 might alter LPS docking to LBP, CD14, and TLR4/MD-2 co-receptors, and then it suppresses the TLR4/NF-κB inflammatory pathway in LPS-stimulated RAW264.7 cells and Drosophila.
Article
Full-text available
The rapid and global spread of a new human coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has produced an immediate urgency to discover promising targets for the treatment of COVID-19. Here, we consider drug repurposing as an attractive approach that can facilitate the drug discovery process by repurposing existing pharmaceuticals to treat illnesses other than their primary indications. We review current information concerning the global health issue of COVID-19 including promising approved drugs, e.g., human angiotensin-converting enzyme inhibitors (hACEIs). Besides, we describe computational approaches to be used in drug repurposing and highlight examples of in-silico studies of drug development efforts against SARS-CoV-2. Alacepril and lisinopril were found to interact with human angiotensin-converting enzyme 2 (hACE2), the host entranceway for SARS-CoV-2 spike protein, through exhibiting the most acceptable rmsd_refine values and the best binding affinity through forming a strong hydrogen bond with Asn90, which is assumed to be essential for the activity, as well as significant extra interactions with other receptor-binding residues. Furthermore, molecular dynamics (MD) simulations followed by calculation of the binding free energy were also carried out for the most promising two ligand-pocket complexes from docking studies (alacepril and lisinopril) to clarify some information on their thermodynamic and dynamic properties and confirm the docking results as well. These results we obtained probably provided an excellent lead candidate for the development of therapeutic drugs against COVID-19. Eventually, animal experiments and accurate clinical trials are needed to confirm the potential preventive and treatment effect of these compounds.
Article
Full-text available
The nuclear factor-kappa B (NF-κB) signaling pathway regulates a variety of biological functions in the body, and its abnormal activation contributes to the pathogenesis of many diseases, such as cardiovascular and respiratory diseases and cancers. Therefore, to ensure physiological homeostasis of body systems, this pathway is strictly regulated by IκBα transcription, IκBα synthesis, and the IκBα-dependent nuclear transport of NF-κB. Particularly, the post-translational modifications of IκBα including phosphorylation, ubiquitination, SUMOylation, glutathionylation and hydroxylation are crucial in the abovementioned regulatory process. Because of the importance of the NF-κB pathway in maintaining body homeostasis, understanding the post-translational modifications of IκBα can not only provide deeper insights into the regulation of NF-κB pathway but also contribute to the development of new drug targets and biomarkers for the diseases.
Article
Full-text available
In the rapidly evolving coronavirus disease (COVID-19) pandemic, repurposing existing drugs and evaluating commercially available inhibitors against druggable targets of the virus could be an effective strategy to accelerate the drug discovery process. The 3C-Like proteinase (3CLpro) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been identified as an important drug target due to its role in viral replication. The lack of a potent 3CLpro inhibitor and the availability of the X-ray crystal structure of 3CLpro (PDB-ID 6LU7) motivated us to perform computational studies to identify commercially available potential inhibitors. A combination of modeling studies was performed to identify potential 3CLpro inhibitors from the protease inhibitor database MEROPS (https://www.ebi.ac.uk/merops/index.shtml). Binding energy evaluation identified key residues for inhibitor design. We found 15 potential 3CLpro inhibitors with higher binding affinity than that of an α-ketoamide inhibitor determined via X-ray structure. Among them, saquinavir and three other investigational drugs aclarubicin, TMC-310911, and faldaprevir could be suggested as potential 3CLpro inhibitors. We recommend further experimental investigation of these compounds.
Article
Full-text available
Molecular dynamics simulations revealed a promising immune target on the SARS-CoV-2 spike protein, proposing novel strategies for vaccine development.
Article
Nephrotoxicity is an indication for the damage of kidney-specific detoxification and excretion mechanisms by exogenous or endogenous toxicants. Exposure to vancomycin predominantly results in renal damage and losing the control of body homeostasis. Vancomycin-treated rats (200 mg/kg/once daily, for seven consecutive days, i.p.) revealed significant increase in serum pivotal kidney function, oxidative stress, and inflammatory biomarkers. Histologically, vancomycin showed diffuse acute tubular necrosis, denudation of epithelium and infiltration of inflammatory cells in the lining tubular epithelium in cortical portion. In the existing study, the conservative consequences of scopoletin against vancomycin nephrotoxicity was investigated centering on its capacity to alleviate oxidative strain and inflammation through streamlining nuclear factor (erythroid-derived-2) like 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling and prohibiting the nuclear factor kappa B (NF-κB)/mitogen-activated protein kinase (p38 MAPK) pathway. With respect to vancomycin group, scopoletin pretreatment (50 mg/kg/once daily, i.p.) efficiently reduced kidney function, oxidative stress biomarkers and inflammatory mediators. Moreover, histological and immunohistochemical examination of scopoletin-treated group showed remarkable improvement in histological structure and reduced vancomycin-induced renal expression of iNOS, NF-κB and p38 MAPK. In addition, scopoletin downregulated (Kelch Like ECH Associated Protein1) Keap1, P38MAPK and NF-κB expression levels while upregulated renal expression levels of regulatory protein (IκBα), Nrf2 and HO-1. Furthermore, molecular docking and network approach were constructed to study the prospect interaction between scopoletin and the targeted proteins that streamline oxidative stress and inflammatory pathways. The present investigations elucidated that scopoletin co-treatment with vancomycin may be a rational curative protocol for mitigation of vancomycin-induced renal intoxication.
Article
Purpose: To evaluate the effect of sulindac, a non-selective anti-inflammatory drug, for activity to reduce breast density (BD), a risk factor for breast cancer. Experimental design: An open-label Phase 2 study was conducted to test the effect of 12 months' daily sulindac at 150 mg bid on change in percent BD in postmenopausal hormone receptor-positive breast cancer patients on aromatase inhibitor (AI) therapy. Change in percent BD in the contralateral, unaffected breast was measured by non-contrast MRI and reported as change in MRI percent BD (MRPD). A non-randomized patient population on AI therapy (observation group) with comparable baseline BD was also followed for 12-months. Changes in tissue collagen after 6 months of sulindac treatment were explored using second-harmonic generated (SHG) microscopy in a subset of women in the sulindac group who agreed to repeat breast biopsy. Results: In 43 women who completed 1 year of sulindac (86% of those accrued), relative MRPD significantly decreased by 9.8% (95% CI, -14.6 to -4.7) at 12 months; an absolute decrease of -1.4% (95% CI, -2.5 to -0.3). A significant decrease in mean breast tissue collagen fiber straightness (p=0.032), an investigational biomarker of tissue inflammation, was also observed. MRPD (relative or absolute) did not change in the AI only observation group (N = 40). Conclusions: This is the first study to indicate that the NSAID sulindac may reduce BD. Additional studies are needed to verify these findings and determine if prostaglandin E2 inhibition by NSAIDs is important for BD or collagen modulation.
Article
Using cheminformatics tools RDKit and literature investigation, four series of 24 thienopyrimidine/N-methylpicolinamide derivatives substituted with pyrimidine were designed, synthesized and evaluated for activities against three cancer cell lines (MDA-MB-231, HCT116 and A549), TAK1 kinase and NF-κB signaling pathway. Almost all compounds showed selectivity toward the A549 cell lines and the most promising compound 38 could inhibit TAK1 kinase and NF-κB signaling pathway with the IC50 values of 0.58 and 0.84 μM. Moreover, 38 can induce cell cycle arrest of A549 cells at the G2/M checkpoint with 30.57% and induce apoptosis (34.94%) in a concentration-dependent manner. And western blot showed that compound 38 could inhibit TNF-α-induced IκBα phosphorylation, IκBα degradation, p65 phosphorylation and TAK1 phosphorylation, and reduce the expression of p65. What’s more, the studies of docking, molecular dynamics, MM/PBSA and frequency analysis theoretically supported the conclusions of the bioevaluation.