Anda di halaman 1dari 16

J Mol Model (2017) 23:239

DOI 10.1007/s00894-017-3396-7

ORIGINAL PAPER

Investigation of naphthofuran moiety as potential dual inhibitor


against BACE-1 and GSK-3: molecular dynamics simulations,
binding energy, and network analysis to identify first-in-class dual
inhibitors against Alzheimers disease
Akhil Kumar 1 & Gaurava Srivastava 1 & Swati Srivastava 1 & Seema Verma 2 &
Arvind S. Negi 2 & Ashok Sharma 1

Received: 13 December 2016 / Accepted: 25 June 2017


# Springer-Verlag GmbH Germany 2017

Abstract BACE-1 and GSK-3 are potential therapeutic good binding affinity towards both the targets. Molecular dy-
drug targets for Alzheimer's disease. Recently, both the targets namics, binding energy, and network analysis were performed
received attention for designing dual inhibitors for to compare their binding with the targets and amino acids
Alzheimers disease. Until now, only two-scaffold triazinone responsible for binding. Naphthofuran series derivatives
and curcumin have been reported as BACE-1 and GSK-3 showed good interaction within the active site residues of both
dual inhibitors. Docking, molecular dynamics, clustering, of the targets. Hydrogen bond occupancy and binding energy
binding energy, and network analysis of triazinone derivatives suggested strong binding with the targets. Dual-inhibitor bind-
with BACE-1 and GSK-3 was performed to get molecular ing was mostly governed by the hydrophobic interactions for
insight into the first reported dual inhibitor. Further, we de- both of the targets. Per residue energy decomposition and
signed and evaluated a naphthofuran series for its ability to network analysis identified the key residues involved in the
inhibit BACE-1 and GSK-3 with the computational ap- binding and inhibiting BACE-1 and GSK-3. The results in-
proaches. Docking study of naphthofuran series showed a dicated that naphthofuran series derivative 11 may be a prom-
ising first-in-class dual inhibitor against BACE-1 and GSK-
3. This naphthofuran series may be further explored to de-
Electronic supplementary material The online version of this article
(doi:10.1007/s00894-017-3396-7) contains supplementary material, sign better dual inhibitors.
which is available to authorized users.
Keywords Alzheimers disease . BACE-1 and GSK-3 .
* Ashok Sharma Dual inhibitor . Molecular dynamics simulation . Binding
ashoksharma@cimap.res.in energy analysis . Network analysis
Akhil Kumar
akhil2687@gmail.com
Gaurava Srivastava Introduction
gaurava1708@gmail.com
Swati Srivastava Alzheimer disease (AD) is the sixth leading cause of death,
swati23.srivastava@gmail.com currently affecting more than 44 million people worldwide
Seema Verma [13]. AD gradually develops and shows symptoms like neu-
seemaverma12@gmail.com ral loss, neurotransmitter depletion, and synaptic dysfunction
Arvind S. Negi
in patients [2]. The most common findings in an AD brain are
arvindcimap@rediffmail.com deposition of senile neuritic plaques (SNPs) and neurofibril-
lary tangles (NFTs) [3]. SNPs are the result of an extracellular
1
Biotechnology Division, CSIR-Central Institute of Medicinal and accumulation of amyloid protein (A), which is generated by
Aromatic Plants, P.O. CIMAP, Lucknow, UP 226015, India the cleavage of amyloid precursor protein (APP) by the -
2
Chemical Sciences Division, CSIR-Central Institute of Medicinal secretase (BACE-1) [4]. On the other hand, NFTs are gener-
and Aromatic Plants, P.O. CIMAP, Lucknow, UP 226015, India ated by the hyperphosphorylation of tau protein by the kinase
239 Page 2 of 16 J Mol Model (2017) 23:239

enzyme, glycogen synthase kinase-3 (GSK-3) [57]. moieties have shown complete inhibition of A oligomerization
These abnormalities lead to the neurotoxic cascades and cyto- and fibrillization [37]. Similarly, arylquinone is also known to
skeletal changes that cause synaptic dysfunction and neural inhibit BACE-1, aggregation of -amyloid peptide, and a
cell death [3]. The synaptic dysfunction and AD pathology destabilizer of preformed -amyloid fibrils [38]. In one of the
have been described by the various hypothesis in which am- studies, furan moiety was reported to inhibit GSK-3 in human
yloid, cholinergic, glutamatergic, oxidative stress, and metal leukemia cancer cells [39]. Another study reported furo-
hypothesis are the most accepted ones [8, 9]. pyrimidines as the potent GSK-3 inhibitor [40]. Furan-based
The currently used drugs based on the cholinergic hypoth- derivatives are also reported to penetrate the bloodbrain barrier
esis are hardly able to modify or halt the disease progression [41]. We therefore, combined the two series naphtho and furan
[10] and most of the current treatments are purely symptom- and selected it for further exploration. We rationally designed
atic based. There is no FDA-approved drug available that naphthofuran series derivatives and identified first-in-class dual
completely halts the diseases progression. Several reports BACE-1 and GSK-3 inhibitor NS11 (Fig. 1b). NS11 showed
are available on targeting single AD targets based on various good binding scores as compared to the first reported dual inhib-
disease causing hypothesis to block the disease progression. itor. It was thus taken as a lead molecule for further study.
[8, 9, 1114]. However, one target-one inhibitor strategies Interaction and molecular dynamics studies for BACE-1 and
seem to be less effective in fighting against multifactorial GSK-3 were performed both in the absence of ligand and in
complex diseases like AD [1517]. A new concept multi- the presence of first reported dual inhibitor DI1 and NS11.
targeted directed ligand (MTDL) has recently emerged for Hydrogen occupancy and binding energy of ligands were calcu-
neurodegenerative diseases like AD [15]. Computational lated to estimate the affinity towards both of the targets. Free-
screening, docking, and molecular dynamics simulations have energy landscape was derived to check the stability of the sys-
been used extensively for the identification of new leads for tems. Networking and clustering methods were also used to
drug discovery [1821]. For multi- or dual-inhibitor design, study the induced conformational changes and interaction be-
various AD target combinations have been explored [2227]. tween protein and dual inhibitors. Our study integrates the con-
Recently, 3,4-dihydro-1,3,5-triazin-2(1H)-ones chemical moi- cept of dual targeting with the help of established computational
ety was reported as a first dual inhibitor (DI1) for BACE-1 and methods to identify the new chemical moiety that might act as a
GSK-3 [28]. These dual inhibitors simultaneously inhibit dual inhibitor for BACE-1 and GSK-3. This model may help in
two of the most validated drug targets in AD drug discovery. developing a potent dual inhibitor for BACE-1 and GSK-3.
GSK-3 has been proposed as the possible link between A
and tau. Further inhibition of GSK-3 downregulates the
amyloidogenic cleavage of APP which suggests the cross talk Materials and methods
between these two disease pathways. [1, 6, 17]. Inhibition of
GSK-3 kinase decreases A production and A-induced neu- Protein 3D structure preparation
rotoxicity by reducing BACE-1 cleavage of APP [29]. Thus,
these two targets are now considered as potential and the most X-ray crystal structures of BACE-1 and GSK-3 complexes
promising dual targets for AD [1, 28]. It was observed that multi- with inhibitor were obtained from the Protein Data Bank, i.e.,
target agents provide a better efficacy profile and effective dis- 1FKN for BACE-1 and 1Q5K for GSK-3. Prior to docking,
ease treatment than the single-target inhibitors [30, 31]. The hy- bond orders were assigned and hydrogen atoms were added to
pothesis of dual inhibitor for BACE-1 and GSK-3 is challeng- the crystal structures and protein structures were prepared by
ing and underexplored until now. Only two scaffolds viz., AutoDock 4.2 software [42]. The solved structure of BACE-1
triazinone [28] (Fig. 1a) and curcumin [1], have been identified. has been (PDB ID 1FKN) previously used in molecular dy-
Thus, there is a need to discover new scaffolds that inhibit both namics and binding energy analysis [43]. 1Q5K is also report-
therapeutic targets. Small molecules and various databases have ed to be successfully used for pose prediction accuracy in
been successfully screened with the help of an in silico approach docking study for GSK-3 [44]. We set the protonation states
[32, 33]. In silico methods have been applied successfully to find of Asp32 and Asp228 dyad of BACE-1 according to one of
dual inhibitors against different targets [21, 34] as well as against the state of art study. Which recommended that protonation
AD [35, 36]. Computational methods have earlier not been used state of these residues are important for effective virtual
to identify dual inhibitors for BACE-1 and GSK-3. In this screening [45].
study, we applied computational methods to obtain molecular
insight into first reported dual inhibitor DI for BACE-1 and Ligand preparation and grid generation
GSK-3.
We screened various chemical moieties in which Reference compounds, i.e., first reported dual inhibitor, for
naphthofuran showed better binding scores towards both the BACE-1 and GSK-3 and naphthofuran molecules referred
targets. 1,4-naphthoquinon combined with the tryptophan to as an (NS) were drawn in 2D with ChemSketch software
J Mol Model (2017) 23:239 Page 3 of 16 239

Fig. 1 2D structures of the


studied inhibitors against BACE-
1 and GSK-3 a 3,4-dihydro-
1,3,5-triazin-2(1H)-ones (DI1)
first reported dual inhibitor of
BACE-1 and GSK-3 and b 1-
(3,4,5 trimethoxyphenyl)-
naphtho [2, 1-b]-furan-2-
aldoxime acetate(NS11)

and saved in a .mol file. These molecules were then converted important for designing new inhibitors. PharmaGist [50] is a
into 3D .pdb file format with the help of Open Babel software freely available web server for pharmacophore detection. It
[46]. Avogadro software was used for structure minimization computes candidate pharmacophore by multiple flexible
using MM3 force field as per the earlier studies [47, 48]. The alignments of the input ligands. The main innovation of this
size of the grid box was 80 80 80 3 with spacing a of approach is that the flexibility of the input ligands is handled
0.375 , so that the active site of both receptors may fit well explicitly and in a deterministic manner within the alignment
into the grid. process. Another important characteristic of the method is the
capability of detecting pharmacophore shared by different
Docking parameters subsets of input molecules. This capability is a key advantage
when the ligands belong to different binding modes or when
AutoDock had been successfully used earlier for both of the the input contains outliers. For multiple alignments, a
targets BACE-1 [24] and GSK-3 [49]. BACE-1 is a protease curcumin molecule was also incorporated into the input file
enzyme with Asp dyad and GSK-3 is a Ser/Thr kinase en- for PharmaGist.
zyme. Their active sites for both the targets are well charac-
terized. The grid box was placed around flap and active site Molecular dynamics simulations for protein ligand
residues of BACE-1, and the grid box dimensions were complex
X = 80, Y = 80, Z = 80 and coordinates were X = +10.069,
Y = 2.417, Z = 0.366 to cover active site residues. For Molecular dynamics simulations were performed using
GSK-3 grid was placed around the active site as well as Gromacs 4.5.5 [51], which was earlier successfully applied
ATP binding site residues. For GSK-3, the grid box dimen- in various studies including BACE-1 [52] and GSK-3 [53]
sions were X = 80, Y = 80, Z = 80 and coordinates were using force field GROMOS 53a6 [54]. The simple point
X = 22.868, Y = 27.175, Z = 8.107 to cover active site resi- charge (SPC) model was used for water molecules and 11
dues. Parameters used during docking experiments with Na+ ions were added in the BACE-1 system and 8 Cl ions
AutoDock 4.2 were - number of GA runs 10, population size were added in GSK-3). For BACE-1, 20,707 water mole-
150, mutation rate 0.02, and crossover rate 0.8. Grid spacing cules whereas were added, for GSK-3 24,517 water mole-
and other parameters were used as default. Docking simula- cule were added in the system. Each system was subjected to
tions were carried out up to 2.5 million energy evaluations energy minimization using the steepest descent integrator
steps with a maximum of 270,000 generations. In without constraints for 1000 steps. Periodic boundary condi-
AutoDock, docking simulation produces ten docked confor- tions were imposed on the system and canonical ensemble
mations. They were clustered on the basis of RMSD. The (NVT) and isothermal-isobaric ensemble (NPT) were used
lowest energy conformations generated by AutoDock were with normal temperature (300 K) and 1 bar of pressure for
regarded as the best binding conformations between ligands 1 ns. Berendsen temperature control [55] (a coupling constant
and the protein. The 3D coordinates of the best conformation of 0.1 ps) and ParrinelloRahman pressure coupling [56] (a
from the docking experiment were taken for the proteinli- coupling constant of 2.0 ps) were used for keeping the system
gand MD simulations. in a stable environment (300 K, 1 bar). The particle mesh
Ewald algorithm [57] was employed to deal with the long-
Pharmacophore search with PharmaGist range Coulomb interactions with Fourier grid spacing of
0.12 nm. Other parameters cutoff such as short range
Pharmacophore, which is the spatial arrangement of features neighborlist cutoff (rlist = 1.0 nm), short-range electrostatic
that is essential for a molecule to interact with the receptor, is cutoff (rcoulomb = 1.0) nm and short-range van der Waals
239 Page 4 of 16 J Mol Model (2017) 23:239

cutoff (rvdw = 1.0 nm) were set. The LINCS [58] algorithm Network analysis
was used to constrain all the bonds. For, each system MD
simulation has been performed for 25 nanosecond (ns) and The study of individual amino acid residues and their molec-
trajectories were collected. An identical simulation strategy ular interactions in protein structures were studied with anoth-
was used in all the cases. In the first set, we performed molec- er computational approach, viz., residue networks analysis
ular dynamics simulations of unbound BACE-1, BACE-1 with the help of RINalyzer [61] and was further visualized
with dual inhibitor (DI 1), and BACE-1 with NS11 and in with the Cytoscape platform [62]. The geometry-based resi-
the second set unbound GSK-3, GSK-3 with dual inhibitor due network was derived from 3D protein structures, which
(DI 1) and GSK-3 with NS11. In total, we had six systems were taken after post molecular dynamics simulation cluster-
and a total of 150 ns molecular dynamics simulations were ing. RINalyzer was used to generate a 2D visualization of
performed to evaluate the binding of NS11 towards both of the network and residue interaction. In this visualization, network
targets. A 2-fs time step was applied and 2-ps final coordinates nodes represent amino acid residues and edges depict non-
were saved. Most of the analyses were performed using covalent residue interactions. To visualize this non-covalent
Gromacs inbuilt tools. All visualizations were performed interaction, UCSF Chimera [63] was used. Communication
using Chimera (http://www.rbvi.ucsf.edu/chimera), and between Cytoscape and UCSF Chimera was handled by struc-
PyMOL (The PyMOL Molecular Graphics System, Version ture, viz., the structure-based layout algorithm, which is im-
0.99 Schrdinger, LLC). Further, hydrogen bonds were plemented in RINalyzer, and uses structureViz to retrieve the
calculated by proton donor and acceptor distance 3.5 and current spatial coordinates.
the angle between acceptordonor hydrogen 30. Hydrogen
bond occupancy between protein and ligands molecule were
calculated via a Python script. Principal component analysis (PCA) and the Gibbs
free-energy landscape
Binding energy calculation MM_PBSA
PCA was performed on each of the BACE-1 and GSK-3
system trajectories obtained from the MD simulation. PCA
Gibbs free energy was calculated using Gromacs and the
is based on the construction of a mass-weighted covariance
method developed by Rashmi kumari (http://rashmikumari.
matrix of the atom displacement, and this covariance matrix is
github.io/g_mmpbsa/) [59]. Interaction energy for BACE-1
diagonalized to extract a set of eigenvectors and eigenvalues
with DI 1, BACE-1 with NS11, GSK-3 with DI 1 and
that reflect the concerted motion of the molecule. First, the
GSK-3 with NS11 were calculated via MM_PBSA methods
g_covar tool was used to extract eigenvectors and its corre-
for 20 to 25 ns. For each simulated system, total snapshots
sponding eigenvalues of Calpha atoms of each unbound pro-
were taken from the last 5 ns of the trajectory at the interval of
tein and protein ligand complex trajectories obtained from
20 ps. This methodology includes electrostatic interactions,
MD. To view the most dominant mode and 2D projection of
van der Waals interactions, polar solvation energy, and non-
eigenvector1 with eigenvector 2, we used the g_anaeig tool.
polar solvation energy. The MM_PBSA analysis also offers
First, two dominant modes were used to plot Gibbs free ener-
an individual contribution to the binding energy and per-
gy by applying Boltzmann relation using g_anaeig and
residue contribution that provides the important amino acid,
g_sham module of Gromacs.
which can be helpful in designing dual inhibitors.

Clustering analysis Blood-brain barrier (BBB) prediction and drug-likeness


prediction
In order to select a reduced set of representative models for all
six systems, root-mean-square deviation (RMSD) conforma- Bloodbrain barrier prediction of NS11 was performed by
tional clustering was performed using the gromos option [60] applying two algorithms, viz., AdaBoost and SVM, combined
implemented in GROMACS (g_cluster) with a 0.1-nm cutoff with four different fingerprints (MACCS, Open Babel (FP2),
value for RMSD. The RMSD clusters analysis uses the pro- Molprint 2D, PubChem). Both AdaBoost and SVM are ad-
cess of structural-fitting, which includes superimposition (to vanced machine learning algorithms used earlier to detect
remove overall translation) and a least-squares-fitting proce- Alzheimers disease [64]. The online BBB predictor [65] pre-
dure (to remove overall rotations). The RMSD matrices were dicts if a compound can pass the BBB(BBB+) or cannot pass
computed on each of the trajectories by the least square fitting the BBB (BBB-). Drug-likeness prediction was performed
of backbone atoms and the matrices were generated. Then, with the help of the PreADMET server [66]. PreADMET is
based on RMSD, similar conformations were clustered into a web-based application for predicting ADME data and build-
groups. ing the drug-like library using in silico method [67].
J Mol Model (2017) 23:239 Page 5 of 16 239

Results the strong binding and support of our docking result as one of
the earlier reports on the study of inhibitors of BACE-1 had
Interaction study through molecular docking of DI1 revealed that residues Tyr71, Thr72, Thr231, Thr232, and
and NS11 with BACE-1 and GSK-3 Gln73 provide van der Waals and Arg235, Arg128, and
Lys224, and Lys321 provides electrostatic contributions for the
The AutoDock docking results analysis showed that the estimat- binding of the ligand [69, 70].
ed binding energy for BACE-1 with DI1 and NS11 was 6.70 For another target (GSK-3), estimated binding energy
kcal/mol and 8.98 kcal/mol, respectively. It was found that DI1 from docking of DI 1 and NS11 was found to be 5.84 kcal/
interacts with BACE-1 through a hydrogen bond with one of the mol and 9.73 kcal/mol, respectively. Docking of DI 1
catalytic dyad (Asp228) and other amino acids Tyr71, Thr72, showed a total of three hydrogen bonds (two with Asp200
Gly230, Thr231, and Ile118 through hydrophobic interaction and one with Ser66). Amino acid residues Gly68, Gly65,
(Fig. 2a). NS11 formed three hydrogen bonds with Arg235 (with and Phe67 were involved in hydrophobic interaction with
different atoms of Arg235) and one hydrogen bond with Gln 73. the DI 1 inhibitor. In the case of NS11, four hydrogen bonds
BACE-1 residues (Phe108, Ile226, and Tyr198) showed hydro- were predicted in the docking studies with Lys85, Asp200,
phobic interaction with NS11. Superimposition of protein-ligand Cys199, and Phe 67. Gly65, Gly68, Ser66, Lys85, Asp200,
docked complex generated with the help of ligplot+ suggested and Phe67 were found to be common amino acid residues for
that Tyr71, Thr 72, Asp228, Gly230, Thr231 were the common binding of DI 1 and NS11 (Fig. 2b). Strong binding of GSK-
amino acid residues for binding of both the ligand DI1 and NS11 3 with pyrimidylpyrrole, oxadiazole, and halomethyl ke-
with BACE-1(Fig. 2a). These residues might be the core residues tones was reported and they formed a hydrogen bond with
for the binding of BACE-1 and can be further considered for the Asp200 and showed interaction with Cys199 located in ATP
design of dual inhibitors. Binding of peptide inhibitor OM992 binding pocket [13, 71]. In GSK-3, Ile62, Gly63, Phe67, and
with BACE-1 has already revealed various subsites for binding Val70 formed a hydrophobic pocket and docking study re-
like S1, S2, S3, and S4 [68]. Our docking results also showed vealed that NS 11 was located in this pocket. In the docking
NS11 binding at S1 (Asp32, Tyr71, Gln73, Phe108, Asp228, results, it was found that ligand NS11 also interacted with the
Gly230), S2 (Tyr71, Thr72, Gln73, Gly230, Thr231, Arg235) polar region formed by the Lys85, Cys199, and Asp200.
subsites and interaction with few residues of the S3 subsite. Previous studies had revealed that the ATP-competitive inhib-
These BACE-1 amino acid interactions with NS11 suggested itors establish hydrogen bonds with the backbone atoms of

Fig. 2 2D protein (BACE-1 and GSK-3) ligand (DI 1 and NS11) Superimposed plot of GSK-3 with DI 1 and NS11. Red circle shows
interaction plot were generated by LigPlot+ and superimposed with shared/common amino acid residues present in the binding of ligand DI1
each other. a Superimposed plot of BACE-1 with DI 1 and NS11. b and NS11 with BACE-1 and GSK-3
239 Page 6 of 16 J Mol Model (2017) 23:239

Asp133 and Val135 [72] and interactions with these ami- Molecular dynamics simulations
no acids are key for enhancing affinity to GSK-3, but it
does not provide the selectivity over other kinases [71, Structure and stability analysis of the MD-simulated BACE-1
72]. Ligand NS11 did not show any interaction with these and GSK-3 systems
residues. Another region of GSK-3 was also reported to
enhance activity and selectivity [71, 72] and this region is Despite the initial structural rearrangements, the trajectories
characterized by the amino acids Lys85, Glu97, and eventually stabilized as indicated by the RMSD plot of back-
Asp200 where ligand NS11 binds. Interaction of an inhib- bone atoms for all the simulated systems. Average RMSD for
itor with this region increases activity and selectivity for unbound BACE-1 backbone atoms was 0.21 nm. In an earlier
GSK-3. Thus, the ligand might behave as a selective and report of BACE-1 (1FKN) 20-ns simulation, it was observed
potent kinase inhibitor. that the system equilibrated well after 5-ns simulation and the
RMSD was reported to be 0.2 nm [76]. Molecular dynamics
of DI1 and NS11 with BACE-1 showed the average value of
Alignment of DI1 and NS11 to study the common spatial RMSD to be 0.20 and 0.23 nm, respectively. These results
arrangement of pharmacophore in both DI1 and NS11 were in agreement for structural stability of the system. In
GSK-3 systems, average RMSD for unbound GSK-3
Alignment of two reported scaffold triazinones and backbone atoms was 0.23 nm. Molecular dynamics of GSK-
curcumin with NS11 was performed to know the similar 3 with DI1 and NS11 showed that the average values of
pharmacophores among these compounds. The results RMSD were 0.260 and 0.267 nm, respectively, which was
revealed common pharmacophoric features, i.e., one slightly higher than unbound protein. The RMSD for all
aromatic, one hydrogen bond acceptor, and one hydro- BACE-1 and GSK-3 systems was nearly similar, thus indi-
gen bond donor in both of the ligands. Distances of cating the stable trajectory of both of the complexes. The
hydrogen bond acceptor and donor from the aromatic radius of gyration (Rg) provides insight into the overall di-
center were 5.91 and 3.66 , respectively, and mension and the shape of the protein. The average value of Rg
distance between donor and acceptor was 3.97 (Fig. was higher for BACE-1 with DI12.14 nm than unbound
3b). Alignment of DI1, NS11, and curcumin (Fig. 3a) BACE-1 protein - 2.13 nm, and BACE-1 with NS11
suggested the same features as reported earlier where 2.11 nm. The average value of Rg for unbound GSK-3 pro-
one of the pharmacophore analyses of B ACE-1 tein was 2.14 nm, with DI12.14 nm and with NS11 was
suggested similar distance required to inhibit BACE-1 2.18 nm. Average RMSD and Rg values for the BACE-1
[73, 74]. In the marine natural-derived inhibitors of system were nearly comparable and RMSD and Rg plot ver-
glycogen synthase kinase-3 phenylmethylene sus time indicated no structural instability or uncoiling for the
hydantoins, pharmacophoric study also showed nearly backbone of any of the systems. Thus, both RMSD (Fig. 4a)
similar distance. [75]. and Rg (Fig. 4b) indicated structural stability.

Fig. 3 Common feature after alignment from PharmaGist. a Spatial arrangement of three features. b Alignment of DI1 and NS 11 with common
pharmacophore features aromatic (light blue) acceptors (green) and donor (yellow)
J Mol Model (2017) 23:239 Page 7 of 16 239

Fig. 4 RMSD profile of protein backbone and Rg profile of whole (green). a RMSD of backbone of unbound BACE-1 and with DI and
protein calculated over the course of 25-ns molecular dynamics simula- NS11. b Rg of backbone of unbound BACE-1 and with DI and NS11.
tion of unbound BACE-1 and GSK-3 (black)/ BACE-1 and GSK-3 c RMSD of backbone of unbound GSK-3 and with DI and NS11. d Rg
bound with DI 1(red)/ and NS11 bound with BACE-1 and GSK-3 of backbone of unbound GSK-3 and with DI and NS11

Comparison of mobility of various flexible regions of unbound various conformational changes (Fig. 5). Binding of NS11
BACE-1/ BACE-1 with DI1 and NS11 and unbound GSK-3 / showed higher fluctuation in inserts B, F, E, D, C, whereas
GSK-3 with DI1 and NS11 the third strand and insert A showed less fluctuation as com-
pared to BACE-1 and bound with DI1 (SF1). In GSK-3 loop
BACE-1 is a flexible protein and its loop region and various C (8995), C helix region (96104), activation loop (200
inserts (i.e., A, B, C, D, E, and F) show considerable mobility 226), and hinge region (133138) were considered as impor-
in molecular dynamics simulations and substrate binding [76]. tant regions for substrate binding. RMSF plot for GSK-3
Careful observation of the flap region and inserts showed indicated that hinge domain and C helix showed a similar

Fig. 5 RMSF of protein amino


acid residue calculated over the
course of 25-ns molecular
dynamics simulation of unbound
BACE-1 and GSK-3 (black), /
DI1 with BACE-1 and GSK-3
(red)/, and NS11 with BACE-1
and GSK-3 (green). a BACE-1
RSMF plot with time. b GSK-3
RSMF plot with time
239 Page 8 of 16 J Mol Model (2017) 23:239

pattern of fluctuation for DI1 and NS11 binding (SF. 2). flap. Van der Waals and hydrogen bond interaction between the
Activation and C loop were more fluctuating in unbound inhibitors and Tyr71 plays a key role in flap movement [79]. The
GSK-3 as compared to bound with DI1 and NS11, suggest- C (Thr72)- C(Thr329) and (Thr72)- NH1(Arg235) distance is
ing that both of the ligands interact with hinge domain and important, as it has been observed earlier that these two residues
provide stability to the DI1 and NS11. It is therefore suggested are involved in ligand BACE-1 interaction [76]. The distance
that hinge domain may provide a platform for inhibitors to between OG1(Thr72)- NH1(Arg235) did not show any signifi-
bind. Studies of RMSF fluctuations suggested that binding cant change. The distance of C (Thr72-C(Asp32), C
of both the ligands stabilize the movement of loop A, C, and (Thr72)- C(Thr329) was comparable to earlier reports [76],
hinge domain as compared to unbound GSK-3. whereasthedistanceofOG1(Thr72)-NH1(Arg235))waslonger,
suggesting thatthe active site was more open uponNS11 binding.
Possible mechanism of BACE-1 inactivation upon ligand
binding: Dynamics of flap region for unbound BACE-1/ bond Possible mechanisms of GSK-3 inactivation upon ligand
with DI1 and NS11 binding: Dynamics of spine residue and distance
between Lys85-Glu97 and Asp200-Gly 202 of GSK-3
The flap is an important aspect for protease inhibitor design [77].
The movement of the flap can be monitored by the changes in the We analyzed the RMSD of hydrophobic spine residues, the
three interatomic distances C(Thr72)- C(Asp32), distance between hydrogen bond (Asp200-Gly202) and salt
C(Thr72)- C(Thr329) and OG1(Thr72)- NH1(Arg235)) bridge (Lys85-Glu97) within the active site of unbound GSK-
[76]. The distance between C(Thr72)-C(Asp32) defines the 3 and bound with DI1 and NS11. In earlier reports, it has
motion of the flap and substrate recognition [76]. Unbound been suggested that if spine residue RMSD is very low, hy-
BACE-1 C(Thr72)-C (Asp32) distance indicate that the flap drogen bond and salt bridge exist between residues Asp200-
was switching between open and closed conformations through- Gly202 and Lys85-Glu97, respectively. If these three condi-
out the simulation run. It was earlier reported that the flap of HIV tions exist then GSK-3 may be considered in an active state
protease and BACE 1 switch between open and closed confor- or active conformation [82]. Spine residue showed a higher
mations [77, 78]. The average C(Thr72)-C(Asp32) distance RMSD value when bound with both of the inhibitors. A large
was 1.24 nm (Table 1), representing the close conformation of distance gap between (Asp200-Gly202) and (Lys85-Glu97)
BACE-1 [29] as previously reported in dynamic studies of flaps was observed, indicating that the there was no possibility to
[52]. In the case of the BACE-1 binding with DI1, the mean value form a hydrogen bond as well as the salt bridge between the
of distance was 1.29 nm, indicating that the flap also adopts stable above-mentioned residues (Fig. 7). The average value of
close conformation. Binding of NS11 with BACE-1 increased RMSD and distance between (Asp200-Gly202) and salt
the distance further and the mean value for C (Thr72)- bridge (Lys85-Glu97) in the active site along 25 ns of simu-
C(Asp32) was 1.64 nm. This indicates that the flap adopts an lations suggested the inactivation of GSK-3 after binding of
open conformation when binding to NS11 and a closed confor- NS11 and DI1 [83].
mation with DI1 (Fig. 6). Adaptation of open conformation of
flap upon inhibitor binding was earlier reported in BACE- SC6, Hydrogen bond network and occupancy during MD
aminopyridine and 2-aminoquinazole inhibitor binding [79, 80]. simulation for ligand DI1 and NS11 with the target BACE-1
Docking results showed that flap residue Tyr71 interacted with and GSK-3
van der Waals interaction, resulting in an open flap after NS11
binding, whereas DI1 interacted with hydrogen bound with Hydrogen bond numbers were plotted against the time for DI1
Tyr71, resulting in a closed conformation of the flap. Hydrogen and NS11 for BACE-1 and GSK-3 complex (SF3). To de-
bond was earlier observed to stabilize the closed conformation of termine the overall residual contribution in the formation of
flap [81]. The variation in the extent of the flap closing and open- hydrogen bond between protein and ligand, we calculated the
ing was caused by the differences in interaction pattern with the hydrogen bond occupancy. The percentage existence of

Table 1 Computed interatomic


distance (nm) between amino acid C (Thr72-C(Asp32) C(Thr72)- (Thr329) OG1(Thr72)-NH1(Arg235)
(C (Thr72-C(Asp32), C
(Thr72)- C(Thr329) and Distance (nm) St. Dev. Distance (nm) St. Dev. Distance (nm) St. Dev.
OG1(Thr72)- NH1(Arg235))
from molecular simulation data to BACE-1 1.24 0.18 1.29 0.17 0.90 0.17
monitor the movement of the flap BACE-1 with DI 1 1.29 0.12 1.38 0.13 0.95 0.16
along 25 ns of simulations BACE-1 with NS11 1.64 0.12 1.45 0.17 0.91 0.18
J Mol Model (2017) 23:239 Page 9 of 16 239

Fig. 6 Dynamics of flap region


for unbound BACE-1 (black)/
bond with DI 1 (red) and NS11
(green) over the course of 25-ns
molecular dynamics simulation.
Interatomic distance between
(C(Thr72-C(Asp32)) atom was
plotted against time to monitor the
flap movement

hydrogen bond in the case of DI1 for Gln12, Val31, Asp32, between GSK-3 and DI1 was 11% Arg141, 5% Thr138,
Thr72, and Gly230 was 17%, 11%, 16%, 4%, and 2%, 4% Phe67,4% Tyr 134, and 4% Lys 85, whereas for NS11
respectively, whereas for NS11 hydrogen bond with Thr232 it was 76% Phe201, 48% Lys85, 11% Asp200, and 5% Arg96.
showed existence of 30%, Asn233 16%, Arg235 23%, These interaction results with GSK-3 were in accordance of
Thr72 10%, Trp115 6%. NS11 showed better binding in earlier findings [84].
terms of a hydrogen bond with BACE-1. Earlier comparison
of the binding modes of the BACE-1 inhibitors suggested that MM_PBSA analysis for BACE-1 and GSK-3 complex
hydrogen bonds with the backbones of Gly230, Arg 235, with DI 1 and NS11
Thr72, or Gln73 were frequently present and this interaction
appears to be vital for high BACE-1 inhibitory activity [79]. To estimate the strength of interaction between target proteins
These results suggest the strong inhibitory activity of NS11 and inhibitors, energy contribution of binding for each amino
against BACE-1. Similarly, number of hydrogen bond and acid was calculated. MM_PBSA method was applied to DI1
occupancy calculated for GSK-3. NS11 showed better hy- and NS11 complexes. Binding energy calculations were per-
drogen bond profile with GSK-3. The hydrogen bond occu- formed from 20 to 25 ns. Binding energies for DI1 and NS11
pancy for both of the ligands revealed that DI1 showed Lys85, ligands were 72.15 kJ/mol and 99.34 kJ/mol, respec-
Phe67, Arg141, and Thr138 as the major residues involved in tively. Energy decomposition to individual component
a hydrogen bond. In the case of NS11, Arg96, Lys85, Phe67, showed that van der Waals and electrostatic forces were the
and Phe201 and Asp200 made a good contribution to hydro- most dominant contributors towards energy contribution
gen bond interactions. Percentage existence of hydrogen bond (Table 2). The van der Waals force was more prominent in

Fig. 7 Structural analyses of functional residues in the active site of GSK-3 along 25 ns of simulations. a RMSD of hydrophobic spine residues. b
Distance between hydrogen bond (Asp200-Gly202). c Salt bridge (Lys85-Glu97) forming residues
239 Page 10 of 16 J Mol Model (2017) 23:239

Table 2 Binding energy values and individual component energy calculated with MM-PBSA method for BACE-1 with DI 1 and NS11

Ligands van der Waals energy Electrostatic energy Polar solvation energy SASA energy Binding energy
(kJ/mol) (kJ/mol) (kJ/mol) (kJ/mol) (kJ/mol)

BACE-1 with DI1 -106.107 9.843 -48.2510.33 94.3918.84 -12.190.67 -72.159.40


BACE-1 with NS11 -164.8916.72 -34.8512.86 118.1319.25 -17.721.07 -99.3414.96
GSK3 with DI1 -95.799.35 -28.4916.92 78.8516.02 -11.370.65 -56.809.06
GSK3 with NS11 -218.4013.26 -33.638.57 134.6919.92 -19.871.01 -137.2019.97

NS11 binding, showing the hydrophobic interaction being a Finally, energy partition for individual residues of the
key player in effective binding. Predicted binding energy BACE-1 and GSK-3 was calculated to identify the contribu-
through MM-PBSA suggested the strong binding of DI 1 tions to the total energy (Fig. 8). Mostly residues belonged to
and NS11 with both the targets. In MM-PBSA analysis the active site and flap region of BACE-1. Asp228 (4.44kJ/
NS11 showed better binding than DI1 towards both the mol), Trp115 (4.22kJ/mol), Ile110 (3.68kJ/mol), Leu30 (2.67
targets. kJ/mol), Thr231 (2.44kJ/mol) and Phe108 (1.24kJ/mol), Tyr71

Fig. 8 Binding energy graph for individual amino acid contributing to binding energy y-axis show the energy contribution per residue in kJ/mol and x-
axis residue number: a for BACE-1 with DI 1(blue) and NS11 (red) and b GSK-3 with DI 1(blue) and NS11 (red)
J Mol Model (2017) 23:239 Page 11 of 16 239

(1.88kJ/mol), Gly230 (1.07kJ/mol), Gln12 (1.75kJ/mol) Network analysis of BACE-1, GSK-3, with DI1 and NS11
were observed to be involved in effective energy con- to determine the important residues essential for dual
tribution in BACE-1 with DI1 complex. Similarly effec- inhibition activity
tive energy contribution, per residue analysis of NS11
with BACE-1 revealed, Asp228 (1.12kJ/mol), Trp115 The protein ligand network was generated from the structure
(1.01kJ/mol), Ile110 (3.62kJ/mol), Leu30 (2.30kJ/mol) obtained from post molecular dynamics run after the clustering.
Thr231 (2.52 kJ/mol ), Phe108 (1.45 kJ/mol ), Tyr71 The most representative structure of the cluster was taken and the
(4.88kJ/mol), Gly230 (0.18kJ/mol), Gln12 (0.91kJ/mol). ligand was retrieved from the trajectory for BACE-1 and GSK-
Binding energy for interacting BACE-1 amino acids 3 (Fig. 9). BACE-1 with DI1 complex network showed that
with 5HA (isophthalamide) were in accordance with residues Gly11, Gln12, Gly13, Leu 30, Gln73, Gly74, Trp115,
earlier reports [71, 82]. Residue analysis indicated that and Thr231 were non-covalently interacting with the target pro-
binding was majorly governed by the amino acid with tein. BACE-1 with NS11 complex network showed more inter-
hydrophobic and polar uncharged side chain. It was ear- action as compared to DI1. In this network, Val69, Tyr71, Gln73,
lier suggested that Asn233, Arg235, and Ser325, which Trp76, Lys107, Phe108, Ile110, Trp115, Gly230, Thr231,
was located in the S2 open region, a hydrophobic phe- Thr232, Asn233, and Arg235 were non-covalently interacting
nyl ring of the inhibitors can interact with these sur- with BACE-1. Network analysis also supported that docking
rounding residues to improve the inhibitory activity interaction at subsite S1, S2, and S3 residues [85, 86] were pres-
[79]. In binding of DI1 with GSK-3, Leu188 ent in the network of BACE-1 with NS11. One of the state-of-the-
(6.12 kJ/mol ), Val70 (6.01 kJ/mol ), Ile62 (2.59kJ/mol ), art studies, encompassing BACE-1 flexibility and ensemble
Thr138 (2.40 kJ/mol ), Tyr134 (2.35 kJ/mol ), Asp200 docking-based hybrid QSAR approach, suggested the essential
(2.09kJ/mol), Leu132 (2.09kJ/mol) were major contribu- interacting residues [87]. Our results were also in agreement with
tors to the binding energy. Val70 (10.47kJ/mol), Leu188 the earlier study. These results were also supported by the binding
(8.45kJ/mol), Cys199 (6.46kJ/mol), Leu132 (5.79kJ/mol), energy contribution to binding for each residue from molecular
Val110 (4.18kJ/mol), Ile62 (3.05kJ/mol) were major con- dynamics and docking results. The DI1 network showed direct
tributors in NS 11 binding with GSK-3. The energy interaction with the flap residues Gln73 and Gly74 and this may
contributing per residues was similar to previous molec- be the reason for the closed flap conformation while binding to
ular dynamics studies of various inhibitors against GSK- DI1. Third strand residue Trp115 also showed interaction with
3 [84]. These interactions with amino acid and indi- DI1 as shown in an RMSF plot and clustering.
vidual amino acid energy contribution were in accor- BACE-1 is a protease enzyme having a large pocket with
dance with our results, further suggesting the good hydrophobicsurfaces.Thus, hydrophobic interactionsare impor-
binding against GSK-3. tant for proteinligand interactions. Dispersion forces, pipi

Fig. 9 2D visualization and interactive analysis of residue interaction, amino acid residues and edges depict non-covalent residue interactions.
BACE-1 with DI1 (a) and BACE-1 with NS11 (b), and GSK-3 binding To visualize this non-covalent interaction, UCSF Chimera was used (yel-
with DI1 (c), and NS11 (d). In this visualization, network nodes represent low line showed non-covalent residue interactions)
239 Page 12 of 16 J Mol Model (2017) 23:239

stacking, and Hpi stacking are an important force in showed stable hinge backbone atoms after ligand
the development of BACE-1 inhibitors [88]. These in- binding.
teractions are non-covalent interactions and may occur
between aromatic rings since they contain pi bonds. Ligand-induced conformation changes during MD
The interaction depended upon the arrangement of simulation: clustering of MD trajectory of mobile region
rings such as sandwich, parallel displaced, T-shaped, of BACE-1 and GSK-3 before and after binding of DI1
face to face, or edge to face. In this case, the aromatic and NS11
ring of Tyr71 and aromatic ring of NS11 were arranged
in parallel displaced orientation that stabilizes the We studied the top clusters for all the systems to observe the
interaction. As per earlier reports, these interactions structural changes in backbone atoms of BACE-1 and GSK-3
are most abundant in arginine and tyrosine among in unbound and bound with DI and NS11. The most representa-
BACE-1 inhibitors. In acylguanidine-based inhibitors tive structure of cluster 1 for all the protein and protein-ligand
pyrrole ring formed a Pi stacking interaction with the complex ligand system was retrieved and aligned (Fig. 10). The
flap Tyr 71 residue and stabilize the enzyme in an open alignment showed that the flap region adopted a relatively closed
conformation [12, 88]. Interaction of Tyr71 with NS11 conformation in unbound BACE-1 and bound with DI1, whereas
was also supported by the free energy per residue con- it was open in NS11. Binding NS11 shifted the third strand
tribution graph (Fig. 9). Residues Lys107, Phe108, (Lys107- Gly117) into the active site and that shifting was oppo-
Ile110, and Trp115 showed interaction with the NS11 site with respect to what was observed in unbound BACE-1 and
and this interaction may explain the shifting of the third bound with DI1. Insert A (Gly158-Leu167) showed extended
strand towards active site. Thus it can be said that bind- conformation with both the DI 1 and NS11. Insert B (Lys218-
ing of DI1 and NS11 involved third-strand shifting Asn221) E (Glu290-Ser295), and C (Ala251-Pro258) followed a
towards the active site. Substrate binding in BACE-1 similar pattern as observed with unbound BACE-1. Insert D
was guided by the third strand and downwards move- (Trp270- Thr274) showed slight shifting in the case of NS11.
ment of A and F strand [76]. GSK-3 DI 1 complex Insert F (Asp311- Asp 317) showed considerable shifting in the
n e t w o r k s h o w e d , I l e 6 2 , G l y 6 3 , Va l 7 0 , A l a 8 3 , DI 1 binding. NS11 binding affected the insert A, D, and the third
Asp133, Tyr134, Val135, Pro136, Thr138, Leu 188, strand whereas in DI 1 major changes occurred in insert F. In
whereas the GSK-3 NS11 complex showed interaction DI1, slight shifting of insert A and third strand was also observed
with Ile62, Gly63, Gly68, Val70, Ala83, Lys85, Glu97, when compared to free BACE-1.GSK-3 was made up of N-
Val110, Leu132, Asp133, Val135, Gln185, Asn186, lobe (35138) and large C-lobe (139386). For an enzymatic
Leu188, Cys 199, and Asp200. These results were in activity majorly three regions i.e., P loop (6469), activation loop
accordance with the docking and free-energy contribu- (200226) and C-helix (96104) were responsible which also
tion of each residue in binding energy. The network contain basic grove (Arg 96, Arg180, and Lys205), Glu97,
showed the hinge domain residues interacting through Lys85, Tyr216 present at the catalytic site [83]. The activation
both of the ligands. These network results were also loop was highly dislocated when it was bound to DI-1 and NS11
supported by the RMSF and clustering results as as compared to free GSK-3. C- helix adopted a similar pat-
RMSF of hinge domain decrease and clustering tern, but Arg 96 seems to shift away in the NS11 in the

Fig. 10 Superimposed backbone structure of the various mobile region of free (a) BACE-1 (black), BACE-1 with DI1 (red) BACE-1 with NS11 (green)
and (b) GSK-3 (black) and bound with DI 1 (red) and NS11 (green) obtained after clustering of 25-ns simulation trajectory
J Mol Model (2017) 23:239 Page 13 of 16 239

alignment. Shifting of P-loop was observed for DI 1 and NS11. inhibitors for BACE-1 and GSK-3. We combined two
The most prominent sign for activation of GSK-3 was where fragments- first naptho - which are already reported to inhibit
C-helix came closer to both the P-loop and activation loop [83]. BACE-1 and second- furan reported to inhibit GSK-3.
P-loop and activation loop shifted away from the active site in a Computational studies of dual inhibitor DI1 and NS11 bearing
ligand-bound state in comparison to unbound state of GSK-3, naphthofuran moiety against BACE-1 and GSK-3 showed po-
which indicate the inactivation of protein. Hinge domain (133 tential to inhibit both the targets. Various computational methods
138) was reported to provide stability to binding ligand following were applied to compare the results between unbound target pro-
a similar pattern of backbone. teins and bound structure of BACE-1 and GSK-3 with DI1 and
NS11. The use of docking with molecular dynamics, free-energy
Principal component analysis (PCA) and the Gibbs calculation lead to identification of a new series of first-in-class
free-energy landscape dual inhibitor (1-(3,4,5 trimethoxyphenyl)-naphtho [2,1-b]-
furan-2 aldoxime acetate) of BACE-1 and GSK-3. The docking
PCA of apo and docked systems of both BACE-1 and GSK-3 of both the DI and NS11 with BACE-1 was found to share com-
beta have shown that more than 60% of total fluctuations in the mon binding residues Asp32, Tyr71, Thr72, Asp228, Gly230,
systems were governed by initial ten eigenvectors (SF4). and Thr231, whereas in binding with GSK-3, Gly65, Gly68,
Calculated PC1 and PC2 (SF5) used to derive free-energy land- Lys85, Asp200, Ser 66, Phe67 were found as common amino
scape (SF6) indicated that all protein ligand systems were able to acid residues. These common residues were previously reported
fold smoothly and achieved stable conformation, and indicated in binding of BACE-1 and GSK-3 with potent inhibitors [71,
an arrangement of amino acid on the surface, which narrows the 78, 79]. Thus these residues can be target for dual inhibitor design
free-energy landscape, leading to convergence of landscape at for BACE-1 and GSK-3.
the bottom. All the systems were in their stable form. Initial conformation obtained by docking experiments was
subjected to molecular dynamics simulation. NS11 bound
The bloodbrain barrier (BBB) prediction with BACE-1 showed hydrogen bond percentage existence
with Thr232 > Arg 235 > Asn233 > Thr72 > Trp115 and
Any molecule working in central nervous system (CNS) drug hydrophobic interaction with Gly74, Lys107, Phe108,
discovery project has to be an optimal candidate that can cross Ile226, Asp32, Asp228, Gly34, Tyr198, Thr72, Val332,
the BBB. The bloodbrain barrier (BBB) is the separation of Thr231, and Tyr71. Similarly, NS11 interacted with GSK-3
circulating blood and cerebrospinal fluid (CSF) in the CNS. It through Phe201 > Lys85 > Asp 200 > Arg96 residues with
consists of tight junctions around the capillaries formed by endo- hydrogen bond. In GSK-3 binding with NS11 residues
thelial cells that specifically restrict the diffusion of microscopic Gly65, Ser 66 Gly68, Val70, Gly97, Met 101, Val110,
objects and large or hydrophilic molecules into the CSF, while Leu130, Thr138, lys183, Gln185, Leu188 showed hydropho-
allowing the diffusion of small hydrophobic molecules. Cells of bic interaction. These interactions suggested that Gly, Lys,
the barrier actively transport metabolic products such as glucose Thr, Val, and Arg were common amino acids for both the
across the barrier with specific proteins.NS11,the leadcandidate, targets and may be further used to design dual inhibitors.
was checked for its ability to cross the bloodbrain barrier. The binding energies MM-PBSA suggested that binding of
AdaBoost and SVM algorithms were used, combined with NS11 was good with BACE-1 and GSK-3. Analysis of the
MACCS, Open Babel (FP2), Molprint 2D, PubChem finger- individual components of energy contribution to binding en-
prints to predict the NS11 bloodbrain permeability. All methods ergy suggested that binding of DI 1 and NS11 with BACE-1
showed +BBB predictions of NS11 except the SVM combine and GSK-3 was mostly governed by the van der Waals en-
with the MACCS fingerprint (SF7, SF8). PreADMET prediction ergy. Van der Waals interactions were governed by the inter-
suggested that NS11 had good drug-like properties, as it followed action of hydrophobic active site residues with both the inhib-
the CMC, MDDR, and WDI like rule (SF9). itors DI1 and NS11. Van der Waals interaction is prominent in
BACE-1 and GSK-3 as reported in earlier docking and dy-
namics studies [89, 90]. Further, per-residue energy contribu-
Discussion tion for NS11 and DI1 in binding with BACE-1 showed sim-
ilar residues, except Tyr71, Asp32, and Asn233, as these were
AD is a multifactorial neurodegenerative disease and various found more prominent in energy contribution with NS11 bind-
reports indicate that amyloid and tau hypothesis are not mutually ing. These residues may provide the high binding affinity of
exclusive [1, 15, 28]. Thus, the development of dual inhibitors for NS11 with BACE-1. Similarly, per-residue energy contribu-
these targets might be an important therapeutic strategy. tion for NS11 and DI1 binding in GSK-3 showed that Val70,
Recently, the first dual inhibitors for BACE-1 and GSK-3 have Leu132, Leu188, Cys199 were more prominent in NS11. The
been reported. The present study was performed to screen and network between inhibitors and both the targets mostly
identify the potential of various chemical moieties as dual showed hydrophobic and polar amino acid residues. These
239 Page 14 of 16 J Mol Model (2017) 23:239

interacting amino acid residues as well as shared common Diagn Ther Rehabil Organ Ges Osterreichischer Nervenarzte
Psychiater 22:153171
amino acid residues involved in binding of both BACE-1
10. Nguyen T-P, Priami C, Caberlotto L (2015) Novel drug target iden-
and GSK-3, together with the spatial arrangement of tification for the treatment of dementia using multi-relational asso-
pharmacophore, may be useful in designing new potent inhib- ciation mining Sci Rep 5:11104. doi:10.1038/srep11104
itors against BACE-1 and GSK-3. The observed results in- 11. Martinez A, Castro A, Dorronsoro I, Alonso M (2002) Glycogen
synthase kinase 3 (GSK-3) inhibitors as new promising drugs for
dicate the potential of NS11 as a lead molecule against AD.
diabetes, neurodegeneration, cancer, and inflammation Med Res
These shared residues between both the targets and their phys- Rev 22:373384. doi:10.1002/med.10011
iochemical properties may be used to design potent inhibitors 12. Ghosh AK, Brindisi M, Tang J (2012) Developing -secretase in-
for both the targets belonging to different families, i.e., prote- hibitors for treatment of Alzheimers disease J Neurochem
120(Suppl 1):7183. doi:10.1111/j.1471-4159.2011.07476.x
ase and kinase. These computational studies provide a plat-
13. Arfeen M, Bharatam PV (2013) Design of glycogen synthase
form to design potent and selective inhibitors against these kinase-3 inhibitors: an overview on recent advancements Curr
two targets. Pharm Des 19:47554775
14. Eldar-Finkelman H, Martinez A (2011) GSK-3 inhibitors: preclin-
Acknowledgements The research work was carried out at the ical and clinical focus on CNS Front Mol Neurosci. doi:10.3389/
Bioinformatics laboratory, Biotechnology Division of the CSIR- fnmol.2011.00032
CIMAP. Akhil Kumar is grateful to the Council of Scientific & 15. Cavalli A, Bolognesi ML, Minarini A, et al. (2008) Multi-target-
Industrial Research (CSIR), India, for the Senior Research Fellowship, directed ligands to combat neurodegenerative diseases J Med Chem
Gaurava Srivastava and Swati Srivastava to BTISnet, DBT, New Delhi. 51:347372. doi:10.1021/jm7009364
16. Prati F, Uliassi E, Bolognesi ML (2014) Two diseases, one ap-
proach: multitarget drug discovery in Alzheimers and neglected
Author contributions A.K., A.S. and A.S.N. conceived the study, de-
tropical diseases Med Chem Commun 5:853861. doi:10.1039/
signed the methodology and wrote the initial draft of the paper, G.S.
C4MD00069B
performed PCA and FEL analysis, S.S. performed network analysis.
17. Iturria-Medina Y, Sotero RC, Toussaint PJ, et al. (2014) Epidemic
A.K., S.V., A.S.N., A.S. contributed to interpreting results, discussion
spreading model to characterize misfolded proteins propagation in
of the associated dynamics, and improvement of this paper.
aging and associated neurodegenerative disorders PLoS Comput
Biol 10:e1003956. doi:10.1371/journal.pcbi.1003956
Compliance with ethical standards 18. Okimoto N, Futatsugi N, Fuji H, et al. (2009) High-performance
drug discovery: computational screening by combining docking
Competing financial interests The authors declare no competing fi- and molecular dynamics simulations PLoS Comput Biol 5:
nancial interests. e1000528. doi:10.1371/journal.pcbi.1000528
19. Shehu A, Nussinov R (2015) Computational methods for explora-
tion and analysis of macromolecular structure and dynamics PLoS
Comput Biol 11:e1004585. doi:10.1371/journal.pcbi.1004585
References 20. Yang S-C, Chang S-S, Chen H-Y, Chen CY-C (2011) Identification
of potent EGFR inhibitors from TCM database@Taiwan PLoS
1. Di Martino RMC, De Simone A, Andrisano V, et al. (2016) Comput Biol 7:e1002189. doi:10.1371/journal.pcbi.1002189
Versatility of the curcumin scaffold: discovery of potent and bal- 21. Chang S-S, Huang H-J, Chen CY-C (2011) Two birds with one
anced dual BACE-1 and GSK-3 inhibitors J Med Chem 59:531 stone? Possible dual-targeting H1N1 inhibitors from traditional
544. doi:10.1021/acs.jmedchem.5b00894 Chinese medicine PLoS Comput Biol 7:e1002315. doi:10.1371/
2. Monczor M (2005) Diagnosis and treatment of Alzheimers disease journal.pcbi.1002315
Curr Med Chem - Cent Nerv Syst Agents 5:513. doi:10.2174/ 22. Bajda M, Guzior N, Ignasik M, Malawska B (2011) Multi-target-
1568015053202723 directed ligands in Alzheimers disease treatment Curr Med Chem
3. Goedert M, Spillantini MG (2006) A century of Alzheimers dis- 18:49494975
ease Science 314:777781. doi:10.1126/science.1132814 23. Tin G, Mohamed T, Gondora N, et al. (2015) Tricyclic phenothia-
zine and phenoselenazine derivatives as potential multi-targeting
4. Cole SL, Vassar R (2008) BACE1 structure and function in health
agents to treat Alzheimers disease Med Chem Commun 6:1930
and Alzheimers disease Curr Alzheimer Res 5:100120
1941. doi:10.1039/C5MD00274E
5. Avila J (2006) Tau phosphorylation and aggregation in Alzheimers 24. Bansode SB, Jana AK, Batkulwar KB, et al. (2014) Molecular
disease pathology FEBS Lett 580:29222927. doi:10.1016/j. investigations of protriptyline as a multi-target directed ligand in
febslet.2006.02.067 Alzheimers disease PLoS One 9:e105196. doi:10.1371/journal.
6. Ghosal K, Fan Q, Dawson HN, Pimplikar SW (2016) Tau protein pone.0105196
mediates APP intracellular domain (AICD)-induced Alzheimers- 25. Su T, Xie S, Wei H, et al. (2013) Synthesis and biological evaluation
like pathological features in mice PLoS One 11:e0159435. doi:10. of berberine-thiophenyl hybrids as multi-functional agents: inhibi-
1371/journal.pone.0159435 tion of acetylcholinesterase, butyrylcholinesterase, and A aggre-
7. Yuraszeck TM, Neveu P, Rodriguez-Fernandez M, et al. (2010) gation and antioxidant activity Bioorg Med Chem 21:58305840.
Vulnerabilities in the tau network and the role of ultrasensitive doi:10.1016/j.bmc.2013.07.011
points in tau pathophysiology PLoS Comput Biol 6:e1000997. 26. Len R, Garcia AG, Marco-Contelles J (2013) Recent advances in the
doi:10.1371/journal.pcbi.1000997 multitarget-directed ligands approach for the treatment of Alzheimers
8. Silva T, Reis J, Teixeira J, Borges F (2014) Alzheimers disease, enzyme disease Med Res Rev 33:139189. doi:10.1002/med.20248
targets and drug discovery struggles: from natural products to drug pro- 27. Capurro V, Busquet P, Lopes JP, et al. (2013) Pharmacological
totypes Ageing Res Rev 15:116145. doi:10.1016/j.arr.2014.03.008 characterization of memoquin, a multi-target compound for the
9. Schmidt R, Neff F, Lampl C, et al. (2008) Therapy of Alzheimers treatment of Alzheimers disease PLoS One 8:e56870. doi:10.
disease: current status and future development Neuropsychiatr Klin 1371/journal.pone.0056870
J Mol Model (2017) 23:239 Page 15 of 16 239

28. Prati F, De Simone A, Armirotti A, et al. (2015) 3,4-Dihydro-1,3,5- 46. OBoyle NM, Banck M, James CA, et al. (2011) Open Babel: an
triazin-2(1H)-ones as the first dual BACE-1/GSK-3 fragment hits open chemical toolbox J Cheminformatics 3:33. doi:10.1186/1758-
against Alzheimers disease ACS Chem Neurosci 6:16651682. 2946-3-33
doi:10.1021/acschemneuro.5b00121 47. Hanwell MD, Curtis DE, Lonie DC, et al. (2012) Avogadro: an
29. Llorens-Martn M, Jurado J, Hernndez F, Avila J (2014) GSK-3, advanced semantic chemical editor, visualization, and analysis plat-
a pivotal kinase in Alzheimer disease Front Mol Neurosci 7:46. doi: form J Cheminformatics 4:17. doi:10.1186/1758-2946-4-17
10.3389/fnmol.2014.00046 48. Cosconati S, Forli S, Perryman AL, et al. (2010) Virtual screening
30. Zheng H, Fridkin M, Youdim M (2014) From single target to with AutoDock: theory and practice Expert Opin Drug Discov 5:
multitarget/network therapeutics in Alzheimers therapy 597607. doi:10.1517/17460441.2010.484460
Pharmaceuticals 7:113135. doi:10.3390/ph7020113 49. Zhong H, Zou H, Semenov MV, et al. (2009) Characterization and
31. Viayna E (2013) Human disease and drug pharmacology, complex development of novel small-molecules inhibiting GSK3 and acti-
as real life Curr Med Chem 20:16231634 vating Wnt signaling Mol BioSyst 5:13561360. doi:10.1039/
32. Wishart DS (2012) Chapter 3: small molecules and disease PLoS b905752h
Comput Biol 8:e1002805. doi:10.1371/journal.pcbi.1002805 50. Schneidman-Duhovny D, Dror O, Inbar Y, et al. (2008)
33. Li YY, An J, Jones SJM (2011) A computational approach to find- PharmaGist: a webserver for ligand-based pharmacophore detec-
ing novel targets for existing drugs PLoS Comput Biol 7:e1002139. tion Nucleic Acids Res 36:W223W228. doi:10.1093/nar/gkn187
doi:10.1371/journal.pcbi.1002139 51. Pronk S, Pll S, Schulz R, et al. (2013) GROMACS 4.5: a high-
34. Arooj M, Sakkiah S, Cao G, Lee KW (2013) An innovative strategy throughput and highly parallel open source molecular simulation toolkit
for dual inhibitor design and its application in dual inhibition of Bioinformatics 29:845854. doi:10.1093/bioinformatics/btt055
human thymidylate synthase and Dihydrofolate reductase enzymes 52. Kumar A, Roy S, Tripathi S, Sharma A (2016) Molecular docking
PLoS One 8:e60470. doi:10.1371/journal.pone.0060470 based virtual screening of natural compounds as potential BACE1
35. Allen BK, Mehta S, Ember SWJ, et al. (2015) Large-scale compu- inhibitors: 3D QSAR pharmacophore mapping and molecular dy-
tational screening identifies first in class multitarget inhibitor of namics analysis J Biomol Struct Dyn 34:239249. doi:10.1080/
EGFR kinase and BRD4 Sci Rep 5:16924. doi:10.1038/srep16924 07391102.2015.1022603
36. Darras FH, Pockes S, Huang G, et al. (2014) Synthesis, biological 53. Kitamura K, Tamura Y, Ueki T, et al. (2014) Binding free-energy
evaluation, and computational studies of tri- and tetracyclic calculation is a powerful tool for drug optimization: calculation and
nitrogen-bridgehead compounds as potent dual-acting AChE inhib- measurement of binding free energy for 7-Azaindole derivatives to
itors and hH3 receptor antagonists ACS Chem Neurosci 5:225 glycogen synthase kinase-3 J Chem Inf Model 54:16531660.
242. doi:10.1021/cn4002126 doi:10.1021/ci400719v
37. Scherzer-Attali R, Pellarin R, Convertino M, et al. (2010) Complete 54. Oostenbrink C, Villa A, Mark AE, van Gunsteren WF (2004) A
phenotypic recovery of an Alzheimers disease model by a biomolecular force field based on the free enthalpy of hydration and
Quinone-tryptophan hybrid aggregation inhibitor PLoS One 5: solvation: the GROMOS force-field parameter sets 53A5 and
e11101. doi:10.1371/journal.pone.0011101 53A6 J Comput Chem 25:16561676. doi:10.1002/jcc.20090
38. Ortega A, Rincn , Jimnez-Aliaga KL, et al. (2011) Synthesis 55. Berendsen HJC, Postma JPM, van Gunsteren WF, DiNola A, Haak
and evaluation of arylquinones as BACE1 inhibitors, -amyloid JR (1984) Molecular dynamics with coupling to an external bath J
peptide aggregation inhibitors, and destabilizers of preformed - Chem Phys 81(8):36843690. doi:10.1063/1.448118
amyloid fibrils Bioorg Med Chem Lett 21:21832187. doi:10. 56. Parrinello M, Rahman A (1981) Polymorphic transitions in single
1016/j.bmcl.2011.03.023 crystals: a new molecular dynamics method J Appl Phys 52(12):
39. Wang F, Lin J, Hou W, et al. (2015) 5-Benzylidene-3,4-dihalo-fu- 71827190. doi:10.1063/1.328693
ran-2-one derivatives inhibit human leukemia cancer cells through 57. Darden T, York D, Pedersen L (1993) Particle mesh Ewald: an
suppression of NF-B and GSK-3 Anti Cancer Agents Med Nlog(N) method for Ewald sums in large systems J Chem Phys
Chem 15:744754 98(12):1008910092. doi:10.1063/1.464397
40. Miyazaki Y, Maeda Y, Sato H, et al. (2008) Rational design of 4- 58. Hess B, Bekker H, Berendsen HJC, Fraaije JGEM (1997) LINCS: a
amino-5,6-diaryl-furo[2,3-d]pyrimidines as potent glycogen syn- linear constraint solver for molecular simulations J Comput Chem
thase kinase-3 inhibitors Bioorg Med Chem Lett 18:19671971. 18:14631472. doi:10.1002/(SICI)1096-987X(199709)18:
doi:10.1016/j.bmcl.2008.01.113 12<1463::AID-JCC4>3.0.CO;2-H
41. Takle AK, Bamford MJ, Davies S, et al. (2008) The identification of 59. Kumari R, Kumar R, Lynn A (2014) g_mmpbsaa GROMACS
potent, selective and CNS penetrant furan-based inhibitors of B-Raf tool for high-throughput MM-PBSA calculations J Chem Inf Model
kinase Bioorg Med Chem Lett 18:43734376. doi:10.1016/j.bmcl. 54:19511962. doi:10.1021/ci500020m
2008.06.070 60. Keller B, Daura X, van Gunsteren WF (2010) Comparing geomet-
42. Morris GM, Huey R, Lindstrom W, et al. (2009) AutoDock4 and ric and kinetic cluster algorithms for molecular simulation data J
AutoDockTools4: automated docking with selective receptor flex- Chem Phys 132:74110. doi:10.1063/1.3301140
ibility J Comput Chem 30:27852791. doi:10.1002/jcc.21256 61. Doncheva NT, Klein K, Domingues FS, Albrecht M (2011)
43. Kim MO, Blachly PG, McCammon JA (2015) Conformational Analyzing and visualizing residue networks of protein structures
dynamics and binding free energies of inhibitors of BACE-1: from Trends Biochem Sci 36:179182. doi:10.1016/j.tibs.2011.01.002
the perspective of protonation equilibria PLoS Comput Biol 11: 62. Shannon P, Markiel A, Ozier O, et al. (2003) Cytoscape: a software
e1004341. doi:10.1371/journal.pcbi.1004341 environment for integrated models of biomolecular interaction net-
44. Gadakar PK, Phukan S, Dattatreya P, Balaji VN (2007) Pose pre- works Genome Res 13:24982504. doi:10.1101/gr.1239303
diction accuracy in docking studies and enrichment of actives in the 63. Pettersen EF, Goddard TD, Huang CC, et al. (2004) UCSF
active site of GSK-3beta J Chem Inf Model 47:14461459. doi:10. chimeraa visualization system for exploratory research and anal-
1021/ci6005036 ysis J Comput Chem 25:16051612. doi:10.1002/jcc.20084
45. Polgr T, Keser GM (2005) Virtual screening for -secretase 64. Morra JH, Tu Z, Apostolova LG, et al. (2010) Comparison of
(BACE1) inhibitors reveals the importance of protonation states AdaBoost and support vector Machines for Detecting Alzheimers
at Asp32 and Asp228 J Med Chem 48:37493755. doi:10.1021/ disease through automated hippocampal segmentation IEEE Trans
jm049133b Med Imaging 29:3043. doi:10.1109/TMI.2009.2021941
239 Page 16 of 16 J Mol Model (2017) 23:239

65. The blood-brain barrier (BBB) Prediction Server. http://www. decomposition analysis Med Chem Res 22:32593269. doi:10.
cbligand.org/BBB/. Accessed 6 Feb 2017 1007/s00044-012-0277-6
66. PreADMET |. In: PreADMET Predict. ADMETox. https:// 79. Baxter EW, Conway KA, Kennis L, et al. (2007) 2-amino-3,4-
preadmet.bmdrc.kr/. Accessed 6 Feb 2017 dihydroquinazolines as inhibitors of BACE-1 (beta-site APP cleav-
67. Singh SK, Gaur R, Kumar A, et al. (2014) The flavonoid derivative ing enzyme): use of structure based design to convert a micromolar
2-(4 Benzyloxyphenyl)-3-hydroxy-chromen-4-one protects against hit into a nanomolar lead J Med Chem 50:42614264. doi:10.1021/
A42-induced neurodegeneration in transgenic drosophila: insights jm0705408
from in silico and in vivo studies Neurotox Res 26:331350. doi:10. 80. Hong L, Tang J (2004) Flap position of free Memapsin 2 (-
1007/s12640-014-9466-z secretase), a model for flap opening in aspartic protease catalysis
68. Hong L, Turner RT, Koelsch G, et al. (2002) Crystal structure of Biochemistry (Mosc) 43:46894695. doi:10.1021/bi0498252
memapsin 2 (beta-secretase) in complex with an inhibitor OM00-3 81. Kumalo HM, Soliman ME (2016) Per-residue energy footprints-
Biochemistry (Mosc) 41:1096310967 based pharmacophore modeling as an enhanced in silico approach
69. Liu S, Fu R, Cheng X, et al. (2012) Exploring the binding of BACE-1 in drug discovery: a case study on the identification of novel -
inhibitors using comparative binding energy analysis (COMBINE) Secretase1 (BACE1) inhibitors as anti-Alzheimer agents Cell Mol
BMC Struct Biol 12:21. doi:10.1186/1472-6807-12-21 Bioeng 9:175189. doi:10.1007/s12195-015-0421-8
70. Wu Q, Li X, Gao Q, et al. (2016) Interaction mechanism explora- 82. Buch I, Fishelovitch D, London N, et al. (2010) Allosteric regula-
tion of HEA derivatives as BACE1 inhibitors by in silico analysis tion of glycogen synthase kinase 3: a theoretical study
Mol BioSyst 12:11511165. doi:10.1039/C5MB00859J Biochemistry (Mosc) 49:1089010901
71. Perez DI, Palomo V, Prez C, et al. (2011) Switching reversibility to 83. Khanfar MA, Hill RA, Kaddoumi A, El Sayed KA (2010)
irreversibility in glycogen synthase kinase 3 inhibitors: clues for Discovery of novel GSK-3 inhibitors with potent in vitro and
specific design of new compounds J Med Chem 54:40424056. in vivo activities and excellent brain permeability using combined
doi:10.1021/jm1016279 ligand- and structure-based virtual screening J Med Chem 53:
72. Kramer T, Schmidt B, Lo Monte F (2012) Small-molecule inhibi- 85348545. doi:10.1021/jm100941j
tors of GSK-3: structural insights and their application to 84. Arfeen M, Patel R, Khan T, Bharatam PV (2015) Molecular dy-
Alzheimers disease models Int J Alzheimers Dis 2012:e381029. namics simulation studies of GSK-3 ATP competitive inhibitors:
doi:10.1155/2012/381029 understanding the factors contributing to selectivity J Biomol Struct
73. Palakurti R, Sriram D, Yogeeswari P, Vadrevu R (2013) Multiple e- Dyn 33:25782593. doi:10.1080/07391102.2015.1063457
pharmacophore modeling combined with high-throughput virtual 85. Kacker P, Bottegoni G, Cavalli A (2012) Computational methods in
screening and docking to identify potential inhibitors of - the discovery and design of BACE-1 inhibitors Curr Med Chem 19:
secretase(BACE1) Mol Inf 32:385398. doi:10.1002/minf. 60956111. doi:10.2174/092986712804485746
201200169 86. Chakraborty S, Ramachandran B, Basu S (2014) Encompassing
74. Hossain T, Mukherjee A, Saha A (2015) Chemometric design to receptor flexibility in virtual screening using ensemble docking-
explore pharmacophore features of BACE inhibitors for controlling based hybrid QSAR: discovery of novel phytochemicals for
Alzheimers disease Mol BioSyst 11:549557. doi:10.1039/ BACE1 inhibition Mol BioSyst 10:26842692. doi:10.1039/
C4MB00540F c4mb00307a
75. Khanfar MA, Asal BA, Mudit M, et al. (2009) The marine natural- 87. Cole DC, Stock JR, Chopra R, et al. (2008) Acylguanidine inhibi-
derived inhibitors of glycogen synthase kinase-3 tors of beta-secretase: optimization of the pyrrole ring substituents
phenylmethylene hydantoins: in vitro and in vivo activities and extending into the S1 and S3 substrate binding pockets Bioorg Med
pharmacophore modeling Bioorg Med Chem 17:60326039. doi: Chem Lett 18:10631066. doi:10.1016/j.bmcl.2007.12.010
10.1016/j.bmc.2009.06.054 88. Xiong B, Huang X-Q, Shen L-L, et al. (2004) Conformational
76. Barman A, Schrer S, Prabhakar R (2011) Computational modeling flexibility of beta-secretase: molecular dynamics simulation and
of substrate specificity and catalysis of the -secretase (BACE1) essential dynamics analysis Acta Pharmacol Sin 25:705713
enzyme Biochemistry (Mosc) 50:43374349. doi:10.1021/ 89. Tounge BA, Rajamani R, Baxter EW, et al. (2006) Linear interac-
bi200081h tion energy models for -secretase (BACE) inhibitors: role of van
77. Hornak V, Okur A, Rizzo RC, Simmerling C (2006) HIV-1 protease der Waals, electrostatic, and continuum-solvation terms J Mol
flaps spontaneously open and reclose in molecular dynamics simu- Graph Model 24:475484. doi:10.1016/j.jmgm.2005.10.002
lations Proc Natl Acad Sci U S A 103:915920. doi:10.1073/pnas. 90. Licht-Murava A, Plotkin B, Eisenstein M, Eldar-Finkelman H
0508452103 (2011) Elucidating substrate and inhibitor binding sites on the sur-
78. Razzaghi-Asl N, Ebadi A, Edraki N, et al. (2013) Ab initio model- face of GSK-3 and the refinement of a competitive inhibitor J Mol
ing of a potent isophthalamide-based BACE-1 inhibitor: amino acid Biol 408:366378. doi:10.1016/j.jmb.2011.02.036

Anda mungkin juga menyukai