You are on page 1of 11

Journal of Receptors and Signal Transduction

ISSN: 1079-9893 (Print) 1532-4281 (Online) Journal homepage: http://www.tandfonline.com/loi/irst20

Zerumbone binding to estrogen receptors: an in-


silico investigation

Eltayeb E. M. Eid, Faizul Azam, Mahmoud Hassan, Ismail M. Taban &


Mohammad A. Halim

To cite this article: Eltayeb E. M. Eid, Faizul Azam, Mahmoud Hassan, Ismail M. Taban &
Mohammad A. Halim (2018): Zerumbone binding to estrogen receptors: an in-silico investigation,
Journal of Receptors and Signal Transduction, DOI: 10.1080/10799893.2018.1531886

To link to this article: https://doi.org/10.1080/10799893.2018.1531886

Published online: 05 Nov 2018.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=irst20
JOURNAL OF RECEPTORS AND SIGNAL TRANSDUCTION
https://doi.org/10.1080/10799893.2018.1531886

RESEARCH ARTICLE

Zerumbone binding to estrogen receptors: an in-silico investigation


Eltayeb E. M. Eida, Faizul Azama , Mahmoud Hassanb, Ismail M. Tabanc and Mohammad A. Halimd,e
a
Unaizah College of Pharmacy, Qassim University, Unaizah, Saudi Arabia; bSwiss Tropical & Public Health Institute, University of Basel,
Switzerland; cSchool of Biosciences, Cardiff University, Cardiff, United Kingdom; dDivision of Computer-aided Drug Design, The Red-Green
Research Center, BICCB, Dhaka, Bangladesh; eInstitut Lumiere Matiere, Universite Lyon 1–CNRS, Universite de Lyon, Villeurbanne
Cedex, France

ABSTRACT ARTICLE HISTORY


Breast cancer is the most frequent malignancy among females worldwide. Estrogen receptor (ER) Received 23 November 2017
mediate important pathophysiological signaling pathways induced by estrogens, and is regarded as a Revised 15 August 2018
promising target for the treatment of breast cancer. Zerumbone (2,6,9,9-tetramethylcycloundeca- Accepted 21 August 2018
2,6,10-trien-1-one; ZER), a chemical constituent present in the Zingiber zerumbet is known to exhibit
KEYWORDS
anti-breast cancer activity by modulating several proteins to induce apoptosis. Medicinal chemists usu- Breast cancer; docking;
ally exploit lead compounds of natural origin to develop molecules with improved pharmacological molecular dynamics;
properties. Current study is intended to utilize molecular modeling techniques to investigate the inter- density functional
action of ZER with estrogen receptors. AutoDock was used to predict the binding modes of ZER and theory; zerumbone
target receptors. Stability of the ZER-ER complex was verified by molecular dynamics simulation using
Desmond software. Docked ZER was further optimized by density functional theory (DFT) using
Gaussian09 program. Analysis of docked conformations in terms of binding energy disclosed estrogen
receptor-b (ERb) as more promising than estrogen receptor-a (ERa). Evaluation of MD trajectories of
ZER bound to both ERa and ERb showed appreciable stability with minimum Ca-atom root mean
square deviation shifts. DFT based global reactivity descriptors such as electron affinity, hardness,
chemical potential, electronegativity and electrophilicity index, calculated from the energies of highest
occupied and lowest unoccupied molecular orbitals underscored the electronic features governing via-
bility of the ZER for interaction with the target receptors. In conclusion, these findings can be
exploited to design and develop novel anticancer agents based on the lead compound, ZER.

Introduction cells, which resulted in suppression of anti-apoptotic and


metastatic gene expression and, induction of apoptotic cell
Natural compounds obtained from plants represent an
death, as well as inhibition of cell invasion [12]; treatment of
immense resource of structurally diverse chemical entities
cancer cells with ZER modulated Bax/Bcl-2 ratio favoring
possessing high affinity and specificity for various pharmaco-
apoptosis, inhibited Sonic hedgehog/GLI-mediated transcrip-
logical targets. They have proven to be by far the magnifi- tion, and downregulated chemokine receptor CXCR4 con-
cent sources for novel leads in drug discovery and comitant with inhibition of CXCL12-induced breast and
development programs. In recent times, bioactive molecules pancreatic cancer invasion [7,13,14]. In addition, ZER abol-
derived from natural sources have served as a privileged ishes RANKL-induced NF-jB activation, inhibits osteoclasto-
starting point for therapeutic intervention of various human genesis, and suppresses human breast cancer-induced bone
ailments [1–3]. loss in athymic nude mice [15].
Zerumbone (2,6,9,9-tetramethylcycloundeca-2,6,10-trien-1- Estrogens have been recognized as potent mitogens in
one; ZER) is a monocyclic sesquiterpene, isolated from the the mammary gland in which they regulate the growth,
rhizome of ginger plant (Zingiber zerumbet), found in tropical development, and functioning of normal as well as cancer-
and subtropical areas (Figure 1) [4]. ZER is known to exhibit ous breast [16,17]. Role of estrogens in the proliferation and
a diverse range of biological activities including anticancer progression of breast cancer has been further established in
activities involving different signaling pathways in various various epidemiological and animal studies where removal of
cell lines [5–7]. It has exhibited promising activity against the ovaries or the treatment with antiestrogens opposes
breast and other cancers in preclinical models [8–11]. their deleterious effects [18,19]. Estrogen receptors (ERa and
Bounty of preclinical studies involving a variety of cancer ERb) belong to a large family of nuclear receptors, which
cell types have conferred a wealth of mechanistic insights mediate the genomic action of estrogens by acting as ligand
into the anticancer effect of ZER. For example: ZER abolished dependent transcription factors [20,21]. Both of these recep-
NF-jB and IjBa kinase activation in a panel of human cancer tors are encoded by two unique genes and therefore, ERb

CONTACT Eltayeb E.M. Eid eem.eid@qu.edu.sa; Faizul Azam faizulazam@gmail.com, f.azam@qu.edu.sa Unaizah College of Pharmacy, PO Box 5888,
Qassim University, Unaizah-51911, Saudi Arabia
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
2 E. E. M. EID ET AL.

active sites. For MOE docking, macromolecular crystal struc-


tures were protonated by employing Amber99 force field
after removing water molecules.

Preparation of ligand
Structure of the ZER was retrieved from PubChem database.
The structure of the ligand was drawn in ChemBioDraw Ultra
14.0 and converted to its three-dimensional coordinate in
ChemBio3D Ultra 14.0. Geometry optimization was done
using MM2 method of energy minimization and saved in
pdb format. The prepared ligand was used as input file for
Figure 1. Chemical structure of zerumbone. AutoDock 4.2 in the next step.

shares a high homology to ERa, in particular in the DNA


binding domain (95%) and the ligand binding domain (55%) Docking simulation
[22]. Among these, ERa-positive breast cancer is the most Molecular docking study was performed by using Lamarckian
common type of breast cancer and therefore, depends on genetic algorithm methodology implemented in AutoDock
estrogen for growth and development. Consequently, target- 4.2 program. The standard docking procedure was adopted
ing estrogen and its receptor is one of the crucial armament- for a rigid protein and a flexible ligand (100 independent
arium in the therapeutics of breast cancer [23] even though runs per ligand). A grid of 60, 60, and 60 points in x, y, and z
only 60% of ER-positive tumors respond to adjuvant therapy directions was built with a grid spacing of 0.375 Å and a dis-
with tamoxifen [24]. Tamoxifen is a non-steroidal estrogen tance-dependent function of the dielectric constant were
antagonist, considered as drug of choice for the treatment of used for the calculation of the energetic map. The default
ERa-positive breast cancer with both premenopausal and settings were used for all other parameters. In MOE, docking
postmenopausal women [25]. It is a selective estrogen recep- parameters were set to default values and 100 poses of ZER
tor modulator, competes with estrogens in binding to ERs, were obtained. All docked poses were ranked based on their
blocking the transactivation site of ER leading to the inhib- binding energies.
ition of estrogen action [26]. Regardless of the beneficial
effects of tamoxifen in the treatment of breast cancer, its
Analysis and visualization of docking simulation results
promoter effect on the gynaecological tract may result in the
At the end of docking, the best poses were analyzed for
development of numerous side effects such as hot flashes,
hydrogen bonding or hydrophobic interactions and root
vaginal discharge and bleeding, suppression of menstruation,
mean square deviation (RMSD) calculations using Discovery
and gastrointestinal disturbances [26]. Therefore, novel
Studio Visualizer 2016 (Accelrys Software Inc.) and PyMol
potential drug candidates are always required to overcome
(The PyMOL Molecular Graphics System) programs. The result
the drawbacks of chemotherapeutic agents.
of molecular docking between estrogen receptors and ZER
Considering the remarkable anti-breast cancer activity of
using AutoDock and MOE programs is summarized in
ZER, and lack of mechanistic insight into the interactions
Table 1.
with estrogen receptors, current research is envisioned to
illuminate its binding modes with ERa and ERb by employing
molecular docking coupled with molecular dynamics simula- Molecular dynamics (MD) simulations
tion studies. DFT based calculations of ZER is intended to
underscore electronic features governing the ligand-receptor In order to inspect the stability of the docked ligand-protein
interactions. complexes the 3 D coordinates of the best docking configur-
ation of ZER in complex with ERa and ERb were subjected to
molecular dynamics simulation using Desmond [27,28]. ZER
Materials and methods in complex with ERa and ERb was immersed in orthorhombic
box with 8476 and 8246 water molecules respectively, using
Molecular docking
simple point charge (SPC) solvent model and the optimized
Preparation of the receptors potential for liquid simulations (OPLS3) force field [29] was
The three-dimensional (3D) crystal structures of ERa (PDB applied. An appropriate number of Naþ counter-ions were
Code 3ERD) and ERb (PDB Code 1QKM), in complex with used to neutralize the complexes. Isothermal-isobaric (NPT)
diethylstilbestrol and genistein, respectively, were retrieved ensemble class was used with temperature and pressure
from the Research Collaboratory for Structural Bioinformatics adjusted to 300 K and 1.01325 bar, respectively. Simulation
(RCSB) Protein Data Bank (PDB; http://www.rcsb.org/pdb/ time was set to 5 ns for each system and the coordinates
home/home.do). For each crystal structure, the crystallo- were saved for every 5 ps. A cutoff radius of 9.0 Å was
graphic water molecules were removed, the missing resi- applied for short-range van der Waals and Coulomb interac-
dues/hydrogen atoms were added and the inhibitors from tions. Temperature and pressure of the systems were main-
the crystal structure were used to define the respective tained by applying Nose–Hoover thermostat [30] and
JOURNAL OF RECEPTORS AND SIGNAL TRANSDUCTION 3

Table 1. Results obtained after docking of ZER with estrogen receptors.


H-bond interactions Hydrophobic interactions
Program Target DGb (kcal/mol)
a b
RMSD (Å) No. Amino acids No. Amino acids
AutoDock ERa (3ERD) –8.47 1.19 – – 14 Leu-346, Ala-350, Leu-384, Met-388, Leu-391, Phe-
404, Met-421, Ile-424, Phe-425, Leu-428, His-524.
ERb (1QKM) –9.40 0.17 – – 14 Met-295, Leu-298, Ala-302, Met-336, Met-340, Leu-
343, Phe-356, Leu-380, Leu-476
MOE c
ERa (3ERD) –8.34 1.12 – – 15 Leu-346, Thr-347, Ala-350, Trp-383, Leu-384, Met-
388, Leu-391, Phe-404, Met-421, Ile-424, Phe-425,
Leu-428, Gly-521, His-524, Leu-525
ERb (1QKM) –9.25 0.6 – – 13 Met-295, Leu-298, Thr-299, Ala-302, Met-336, Met-
340, Leu-343, Phe-356, Ile-373, Ile-376, Phe-377, Gly-
472, Leu-476
a
Binding free energy.
b
Root mean square deviation.
c
Molecular operating environment.

Figure 2. The validation of accuracy and performance of docking protocol. AutoDock 4.2: The native and docked ligands of ERa, lime green and brown (a); ERb,
cyan and blue (b) respectively. MOE: The native and docked ligands of ERa, pink and yellow (c); ERb, sky blue and red (d) respectively (see colour version of this fig-
ure at www.tandfonline.com/irst).

Martyna–Tobias–Klein [31] methods respectively. The RESPA study in the form of pdb format, served as input structure
integrator was used with a time step of 2.0 fs [32] for the for the DFT calculations.
overall simulations. The systems were minimized and equili-
brated with the default protocols of the Desmond.
Results and discussion
Validation of docking protocol
Density functional theory (DFT) calculations
Accuracy of the docking protocol employed in both
Density functional theory (DFT) employing Becke’s three-par- AutoDock 4.2 and MOE programs was assessed by redocking
ameter hybrid model, Lee–Yang–Parr (B3LYP) correlation of the native co-crystallized ligands (diethylstilbestrol and
functional method at 6–311 G (d,p) level was used to opti- genistein for 3ERD.pdb and 1QKM.pdb respectively) within
mize the minimum energy conformation of the docked ZER. the inhibitor binding cavity (IBC) of human ERa and ERb, and
Gaussian09 software program suite [33] was used for these the docked positions were compared to the crystal structure
calculations. DFT based global reactivity descriptors, such as positions by calculating RMSD values. According to the crite-
electron affinity, hardness, softness, chemical potential, elec- ria of validation cited in literature, RMSD values smaller than
tronegativity, and electrophilicity index were calculated to 2.0 Å indicate that the docking protocol is capable of accur-
inspect the suitability of these descriptors for understanding ately predicting the binding orientation of the co-crystallized
the reactive nature and sites of the ZER molecule. The min- ligand [34]. In the present study, RMSD values of both ERa
imum energy conformer obtained from molecular docking and ERb were within 2.0 Å (Figure 2), which signifies that our
4 E. E. M. EID ET AL.

Figure 3. Molecular docking results employing AutoDock 4.2. Three dimensional structures of proteins showing the binding sites (left), and main residues involved
in the ligand-protein interactions (right) of ZER with ERa (a) and ERb (b). Both proteins are shown as ribbon style carrying docked ZER as mesh in dark pink color.
Residues of binding pocket of ERa are shown as green sticks and docked ZER as purple ball and stick style (a). ERb binding pocket is presented as stick in cyan
color and docked ZER as ball and stick in red color (b). Ligand-protein interactions are marked by broken lines in purple color (see colour version of this figure at
www.tandfonline.com/irst).

docking methods are valid for the given structures and both conformation having minimum binding energy and best lig-
AutoDock 4.2 and MOE, therefore can be relied for docking and-receptor interactions. The results in terms of binding
ZER into the IBC of ERa and ERb [35–38]. free energy, RMSD value and number of interactions are pre-
sented in Table 1.
There are twelve a-helices (H1–H12) and a b-hairpin in the
Binding interactions of ZER with estrogen receptors
ligand binding domain of estrogen receptors. However, H12
Estrogen hormones, in addition to regulating multiple is regarded as an indispensable helix for receptor activation
physiological processes, also play a crucial role in promoting and acts as switch of ligand-binding domain, by adopting a
the proliferation of the neoplastic breast epithelium by act- characteristic conformation upon ligand binding [43,44]. The
ing on soluble nuclear estrogen receptors, ERa and ERb [39]. ligand-binding domain of ERb differs at only two amino acid
Molecular docking has been routinely employed for several positions to that of ERa. However, overall smaller size of the
purposes including ligand binding affinity prediction, ligand ERb pocket as well as amino acid differences of the ligand-
pose prediction and lead identification [3,35,40,41]. binding pocket may contribute to the identification of com-
Therefore, this methodology can be exploited for the identifi- pounds displaying receptor selectivity. Unfortunately, ligands
cation of novel anti-breast cancer lead by targeting estrogen having appreciable selectivity are yet to be identified and cur-
receptors, owing to its critical role in gene expression and rently studied ligands only display modest selectivity for ERb
transcription [42]. The X-ray coordinates of ERa (pdb id: versus ERa and certainly there is a necessity to develop novel
3ERD) and ERb (pdb id: 1QKM) co-crystallized with diethylstil- compounds with improved selectivity [45].
bestrol and genistein respectively, were downloaded from The molecular docking results by AutoDock showed that
the protein data bank website (www.rcsb.org), and processed ZER possesses a very similar binding mode as native co-crystal-
further for docking simulations employing AutoDock and lized ligand, diethylstilbesterol by occupying the entire hydro-
MOE programs. After successful completion of molecular phobic pocket of ligand binding domain in ERa (Figure 3).
docking of ZER with each target protein, a total of 100 poses Common residues involving hydrophobic interactions include
belonging to docked ZER were visualized to identify the Leu346, Ala350, Leu384, Phe404, Met421, Gly521, Leu525.
JOURNAL OF RECEPTORS AND SIGNAL TRANSDUCTION 5

Figure 4. Molecular docking results employing MOE. Three-dimensional structures of proteins showing the binding sites (left), and two-dimensional view of main
residues involved in the ligand-protein interactions (right) of ZER with ERa (a) and ERb (b). Docked ZER in both receptors is shown as stick style in green color (see
colour version of this figure at www.tandfonline.com/irst).

However, Thr347, Met388, Ile424, Phe425, and Leu428 were exhibiting binding energies of 9.25 and 8.34 Kcal mol1
observed to anchor additional hydrophobic contact in docked respectively, as shown in Table 1. All of the intermolecular
ZER. Likewise, docked ZER also occupies the binding pocket of interactions were of hydrophobic type while none of the
ERb in similar fashion to the native co-crystallized ligand, gen- amino acid residues were observed to interact with the only
istein and shares hydrophobic contacts through Met295, hydrophilic keto moiety of the ZER. Most of the interacting
Leu298, Leu339, Met340, Phe356, Ile376, and Leu476 (Figure
residues of ERa and ERb were common in both AutoDock and
3). Other residues encircling the docked ZER by hydrophobic
MOE results. However, Thr-347, Trp-383, Gly-521, and Leu-525
links in ERb include Thr299, Ala302, Met336, Leu343, and
residues were found to contribute in the hydrophobic inter-
Leu380. However, keto group, as one and only polar moiety of
ZER could not avail the opportunity to participate in any kind action within ERa binding site but not observed in case of
of hydrophilic interaction within the active sites of both ERa AutoDock (Figure 4). Furthermore, Thr-299, Ile-373, Ile-376,
and ERb. Similarly, results of molecular docking studies by Phe-377, and Gly-472 were also part of the hydrophobic
MOE correspond with AutoDock results, revealing better inter- pocket of ERb receptor but not in case of AutoDock.
action profile of ZER with the ERb in comparison with ERa, Interestingly, Leu-380 residue encircled the binding cavity of
6 E. E. M. EID ET AL.

ERb in AutoDock but MOE result was devoid of this


amino acid.

Molecular dynamics (MD) simulation study of ZER and


estrogen receptors
The molecular dynamics simulations studies were performed
for 5 ns to analyze the steadiness and conformational stabil-
ity of ZER–ERa and ZER–ERb complexes, which were
embedded with water molecules in the physiological envir-
onmental conditions, such as temperature and pressure. The
RMSDs for the Ca of the receptor (in blue) and for the ligand
(in red) are presented in Figure 5. The 5 ns simulation studies
indicate that ZER forms a stable complex with both ERa and
ERb receptors. RMSD values with respect to the progression
of time are often used to inspect whether or not a simulated
system attains stability [46]. RMSD values of protein, ranging
from 0.6–1.8 Å and 0.75–2.25 Å for ERa and ERb, respectively,
is based on all backbone Ca atoms relative to the corre-
sponding starting structures of all trajectories for the simu-
lated ZER-ER complex. RMSD values of ZER shown on right
Figure 5. The Root Mean Square Deviations (RMSD) of backbone atoms relative
Y-axis of Figure 5 specifies how stable the ZER is with
to the starting complexes during 5 ns MD simulation of ZER in complex with
respect to the estrogen receptors and their binding pocket. ERa (a) ERb (b). Each plot shows the RMSD of protein on left Y-axis whereas lig-
The plots of both Ca and ‘Lig fit Prot’ (ligand fitting on pro- and RMSD is presented on right Y-axis (see colour version of this figure at
www.tandfonline.com/irst).
tein) shows the RMSD of ZER when the protein-ligand com-
plex is first aligned on the protein backbone of the reference
and then the RMSD of the ligand heavy atoms is measured.
In both ERa and ERb simulated systems, the RMSD values
increased quickly during the initial time of simulation and
then reached plateaus at nearly 0.5 nsec (ERa) and 1.0 nsec
(ERb), which simply implicate that the simulated systems
achieved stability.
Figure 6 portrays the plot of root-mean-square fluctua-
tions (RMSF) in the orientation of side chain of each residue
and backbone of the protein in the MD trajectory. RMSF for
ERa was observed to be within 2.8 Å whereas in case of
ERb, the fluctuations were recorded up to 4.0 Å. Therefore,
it can be implied that the lower atomic fluctuation for resi-
dues and its backbone atoms is due to small conform-
ational changes during the simulation of both ERa and ERb
receptors [47].
At the end of the simulation, each system was independ-
ently scrutinized for per-residue interaction analysis, which is
based on the average value of the interaction occupancies
of the binding site residues and the results were described
in the form of stacked bar plots, normalized over the course
of the trajectory (Figure 7).
In the case of ZER-ERa system, maximum interaction occu-
pancy was noted for Ala350 where hydrophobic interaction Figure 6. The Root Mean Square Fluctuation (RMSF) of complexes during 5 ns
was maintained during approx. 30% of the simulation time. MD simulation of ERa (a) and ERb (b) (see colour version of this figure at www.
tandfonline.com/irst).
However, ZER-ERb system was demonstrated to preserve
similar type of interaction by Ile373, during approx. 20% of
the simulation time. Amino acids such as Leu346, Ala350,
Leu384, Met388, and Leu525 showing hydrophobic interac- crystallized ligand, diethylstilbestrol in ERa. However, add-
tions in docked ZER were also observed to sustain their par- itional residues including Met343, Leu349, Leu387, Leu391,
ticipation in such interactions throughout the simulated Phe404, Met421, Ile424, Phe425, Leu428, Val534, and Leu540
trajectory of ZER–ERa complex. Similar residues are also were also observed to play significant role in hydrophobic
known to constitute the binding pocket of native co- interactions (Figure 8). Similarly, residues Met295, Leu298,
JOURNAL OF RECEPTORS AND SIGNAL TRANSDUCTION 7

Figure 7. Per-residues analysis of the ZER in complex with ERa (a) and ERb (b). The analysis was based throughout the 5-ns MD simulations. Hydrogen bond,
hydrophobic and water bridge interactions are illustrated by green, purple and blue lines, respectively (see colour version of this figure at www.tandfonline.
com/irst).

Leu339, Met340, Phe356, Ile376, and Leu476 showing their The energy separation between frontier molecular orbitals
importance in hydrophobic interactions of docked ZER also constituting the gap between the energies of highest occu-
conserved their contribution in simulated ZER-ERb system. pied molecular orbital (HOMO) and lowest unoccupied
However, it was interesting to note that these residues also molecular orbital (LUMO) reflects the kinetic stability and
outline the binding pocket of native co-crystallized ligand, chemical reactivity of the molecule [47,52,53]. The HOMO
genistein, in ERb. and LUMO energy gap for ZER was calculated at the B3LYP/
6-311G(d,p) level and the graphical representation of these
orbitals is shown in Figure 9. The positive and negative
DFT computations phases of molecular orbitals can be recognized in red and
green color respectively. Detailed inspection of HOMO and
Molecular interactions of ligands with target proteins are
LUMO orbitals indicates that the path of electron density
absolutely crucial to various pharmacological activities, which
transfer from ground to first excited state. Both of these are
in turn depend on the structural features of the molecules.
considered as key orbitals contributing in chemical reactivity
Density functional theory (DFT) calculations furnish informa- as well as bioactivity. In HOMO, the electron distribution is
tion about electronic effects and intermolecular charge trans- scattered over the cross conjugated ketone moiety covering
fer responsible for ligand-protein connections [48–50]. whole a,b-unsaturated carbonyl group, and partially extend-
Therefore, computation of the HOMO (highest occupied ing towards C8 as well as C15 in the 11-membered ring
molecular orbital) and LUMO (lowest unoccupied molecular structure of ZER. However, LUMO has been detected to par-
orbital) energies describes the reactivity, shape and binding tially spread on C13 and C9 in addition to the carbonyl moi-
properties of a complete molecule as well as of molecular ety and both conjugated double bonds. Interestingly,
fragments and substituents. In recent times, the concept of isolated double bond between C10 and C12 conferred not
HOMO and LUMO has been successfully executed in explicat- any participation in HOMO/LUMO transition. According to
ing the biological activity and molecular properties of the Koopmans’ theorem [54] the ionization potential (I) and elec-
drug candidates [47,51,52]. tron affinity (A) of ZER can be expressed through HOMO and
8 E. E. M. EID ET AL.

Figure 8. Ligplot diagrams of docked ZER into the binding cavity of ERa (a) and ERb (c). A representation of detailed ligand atom interactions during MD simula-
tion of ZER with the protein residues of ERa (b) and ERb (d) (see colour version of this figure at www.tandfonline.com/irst).

LUMO orbital energies as: I ¼ EHOMO ¼ 6.46 eV and Conclusion


A ¼ ELUMO ¼ 1.33 and the HOMO-LUMO gap ¼ 5.13 eV. Based
on this, chemical potential, l ¼ ðELUMO þ2 EHOMOÞ ¼ 3.90 eV Molecular interaction of ZER and estrogen receptors was
and hardness, g ¼ ELUMO2 EHOMO ¼ 2.56 eV were computed. A studied by employing docking, molecular dynamics simula-
large HOMO/LUMO energy gap is associated with a hard tion and DFT based optimization. Docking results showed
molecule and small HOMO/LUMO gap with a soft molecule. appreciable affinities with both ERa and ERb receptors and
So, a molecule with smaller HOMO/LUMO gap is expected to demonstrated similar binding interactions with native co-
be more reactive. Electrophilicity index (x) of ZER was calcu- crystallized ligands of the respective receptors exhibiting
lated by implementing the formula of [55] as exclusively hydrophobic interactions. However, keto group,
l2
x ¼ 2g ¼ 2.96 eV, which defines a quantitative classification of as one and only polar moiety of ZER could not afford any
global electrophilic nature of a compound. They anticipated kind of hydrophilic interaction within the active sites of
x as a measure of energy lowering due to maximal electron both estrogen receptor types. Results of MD simulation of
flow between donor and acceptor. docked-ERa/ERb systems in terms of RMSD, RMSF and per-
JOURNAL OF RECEPTORS AND SIGNAL TRANSDUCTION 9

Figure 9. DFT optimized structure of ZER (a) generated at B3LYP/6-311G (d,p). Visualization of LUMO (lowest unoccupied molecular orbital; (b) and HOMO (highly
occupied molecular orbital; (c) (see colour version of this figure at www.tandfonline.com/irst).

residue interaction analyses authenticate the stability of the [5] Prasannan R, Kalesh KA, Shanmugam MK, et al. Key cell signaling
studied complexes throughout the simulated trajectories. pathways modulated by zerumbone: role in the prevention and
treatment of cancer. Biochem Pharmacol. 2012;84:1268–1276.
DFT-based global reactivity descriptors, such as electron
[6] Yodkeeree S, Sung B, Limtrakul P, et al. Zerumbone enhances
affinity, hardness, chemical potential, electronegativity, and TRAIL-induced apoptosis through the induction of death recep-
electrophilicity index can be implemented to better under- tors in human colon cancer cells: evidence for an essential role of
stand the ligand-receptor interactions of ZER. These results reactive oxygen species. Cancer Res. 2009;69:6581–6589.
provide an insight into the design and development of novel [7] Sung B, Jhurani S, Ahn KS, et al. Zerumbone down-regulates che-
mokine receptor CXCR4 expression leading to inhibition of
anticancer agents based on the lead compound, ZER.
CXCL12-induced invasion of breast and pancreatic tumor cells.
Cancer Res. 2008;68:8938–8944.
[8] Sehrawat A, Arlotti JA, Murakami A, et al. Zerumbone causes Bax-
Disclosure statement and Bak-mediated apoptosis in human breast cancer cells and
No potential conflict of interest was reported by the authors inhibits orthotopic xenograft growth in vivo. Breast Cancer Res
Treat. 2012;136:429–441.
[9] Tanaka T, Shimizu M, Kohno H, et al. Chemoprevention of azoxy-
ORCID methane-induced rat aberrant crypt foci by dietary zerumbone
isolated from Zingiber zerumbet. Life Sci. 2001;69:1935–1945.
Faizul Azam http://orcid.org/0000-0002-2927-8167 [10] Kim M, Miyamoto S, Yasui Y, et al. Zerumbone, a tropical ginger
sesquiterpene, inhibits colon and lung carcinogenesis in mice. Int
J Cancer. 2009;124:264–271.
[11] Abdelwahab SI, Abdul AB, Devi N, et al. Regression of cervical
References intraepithelial neoplasia by zerumbone in female Balb/c mice pre-
natally exposed to diethylstilboestrol: involvement of mitochon-
[1] Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, et al. dria-regulated apoptosis. Exp Toxicol Pathol. 2010;62:461–469.
Discovery and resupply of pharmacologically active plant-derived [12] Takada Y, Murakami A, Aggarwal BB. Zerumbone abolishes NF-
natural products: A review. Biotechnol Adv. 2015;33:1582–1614. kappaB and IkappaBalpha kinase activation leading to suppres-
[2] Azam F. Therapeutic potential of free radical scavengers in neuro- sion of antiapoptotic and metastatic gene expression, upregula-
logical disorders. In: Kozyrev D, Slutsky V, editors. Handbook of tion of apoptosis, and downregulation of invasion. Oncogene.
free radicals: formation, types and effects. New York: Nova 2005;24:6957–6969.
Publishers; 2010. p. 57–97. [13] Sakinah SA, Handayani ST, Hawariah LP. Zerumbone induced
[3] Azam F, Mohamed N, Alhussen F. Molecular interaction studies of apoptosis in liver cancer cells via modulation of Bax/Bcl-2 ratio.
green tea catechins as multitarget drug candidates for the treat- Cancer Cell Int. 2007;7:4.
ment of Parkinson’s disease: computational and structural [14] Hosoya T, Arai MA, Koyano T, et al. Naturally occurring small-mol-
insights. Network. 2015;26:97–115. ecule inhibitors of hedgehog/GLI-mediated transcription.
[4] Yob NJ, Jofrry SM, Affandi MM, et al. Zingiber zerumbet (L.) Smith: Chembiochem. 2008;9:1082–1092.
A review of its ethnomedicinal, chemical, and pharmacological [15] Sung B, Murakami A, Oyajobi BO, et al. Zerumbone Abolishes
uses. Evid Based Complement Alternat Med. 2011;2011:1. RANKL-Induced NF-kappaB activation, inhibits osteoclastogenesis,
10 E. E. M. EID ET AL.

and suppresses human breast cancer-induced bone loss in athy- [37] Hussain MS, Azam F, Ahamed KF, et al. Anti-endotoxin effects of
mic nude mice. Cancer Res. 2009;69:1477–1484. terpenoids fraction from Hygrophila auriculata in lipopolysacchar-
[16] Russo J, Russo IH. Differentiation and breast cancer. Medicina (B ide-induced septic shock in rats. Pharm Biol. 2016;54:628–636.
Aires) 1997;57:81–91. [38] Shushni MA, Azam F, Lindequist U. Oxasetin from Lophiostoma
[17] Haslam SZ, Counterman LJ, Nummy KA. Effects of epidermal sp. of the Baltic Sea: identification, in silico binding mode predic-
growth factor, estrogen, and progestin on DNA synthesis in tion and antibacterial evaluation against fish pathogenic bacteria.
mammary cells in vivo are determined by the developmental Nat Prod Commun. 2013;8:1223–1226.
state of the gland. J Cell Physiol. 1993;155:72–78. [39] Russo J, Russo IH. The role of estrogen in the initiation of breast
[18] Hilakivi-Clarke L, Clarke R, Lippman M. The influence of maternal cancer. J Steroid Biochem Mol Biol. 2006;102:89–96.
diet on breast cancer risk among female offspring. Nutrition. [40] Azam F, Amer AM, Abulifa AR, et al. Ginger components as new
1999;15:392–401. leads for the design and development of novel multi-targeted
[19] Liao DZ, Pantazis CG, Hou X, et al. Promotion of estrogen- anti-Alzheimer’s drugs: a computational investigation. Drug Des
induced mammary gland carcinogenesis by androgen in the
Devel Ther. 2014;8:2045–2059.
male Noble rat: probable mediation by steroid receptors.
[41] Azam F. Ginger components as anti-Alzheimer drugs: focus on
Carcinogenesis. 1998;19:2173–2180.
drug design. In: Farooqui T, Farooqui AA, editor. Neuroprotective
[20] Nuclear Receptors Nomenclature Commitee. A unified nomencla-
effects of phytochemicals in neurological disorders. New Jersey:
ture system for the nuclear receptor superfamily [letter]. Cell.
Wiley & Sons; 2017. p. 149–161.
1999;97:161–163.
[42] Kaur G, Mahajan MP, Pandey MK, et al. Design, synthesis, and
[21] Beato M, Herrlich P, Schutz G. Steroid hormone receptors: many
actors in search of a plot. Cell. 1995;83:851–857. anti-breast cancer evaluation of new triarylethylene analogs bear-
[22] Hoffmann J, Sommer A. Anti-hormone therapy: principles of ing short alkyl- and polar amino-/amido-ethyl chains. Bioorg Med
endocrine therapy of cancer. In: Bradbury RH, editor. Cancer. Chem Lett. 2016;26:1963–1969.
topics in medicinal chemistry. Vol. 1. Berlin, Heidelberg: Springer; [43] Nettles KW, Bruning JB, Gil G, et al. Structural plasticity in the
2006. p. 19–82. oestrogen receptor ligand-binding domain. EMBO Rep. 2007;8:
[23] Macgregor J, Jordan VC. Basic guide to the mechanisms of anties- 563–568.
trogen action. Pharmacol Rev. 1998;50:151–196. [44] Pike AC. Lessons learnt from structural studies of the oestrogen
[24] McGuire WL. Endocrine therapy of breast cancer. Annu Rev Med. receptor. Best Pract Res Clin Endocrinol Metab. 2006;20:1–14.
1975;26:353–363. [45] Haldos en LA, Zhao C, Dahlman-Wright K. Estrogen receptor beta
[25] Peng J, Sengupta S, Jordan VC. Potential of selective estrogen in breast cancer. Mol Cell Endocrinol. 2014;382:665–672.
receptor modulators as treatments and preventives of breast can- [46] Maiorov VN, Crippen GM. Significance of Root-Mean-Square devi-
cer. Anticancer Agents Med Chem. 2009;9:481–499. ation in comparing three-dimensional structures of globular pro-
[26] Martinkovich S, Shah D, Planey SL, et al. Selective estrogen recep- teins. J Mol Biol. 1994;235:625–634.
tor modulators: tissue specificity and clinical utility. Clin Interv [47] Azam F, Alabdullah NH, Ehmedat HM, et al. NSAIDs as potential
Aging. 2014;9:1437–1452. treatment option for preventing amyloid b toxicity in Alzheimer’s
[27] Bowers KJ, Chow E, Xu H, et al. 2006. Scalable algorithms for disease: an investigation by docking, molecular dynamics, and
molecular dynamics simulations on commodity clusters. DFT studies. J Biomol Struct Dyn. 2018;36:2099–2117.
Proceedings of the ACM/IEEE Conference on Supercomputing [48] Franke R. 1984. Theoretical drug design methods. Amsterdam:
(SC06), Tampa, Florida, November 11–17. Elsevier. p. 115–123.
[28] Desmond Molecular Dynamics System. 2016. D. E. Shaw Research, [49] Osmiałowski K, Halkiewicz J, Radecki A, et al. Quantum chemical
New York (NY), 2016. Maestro-Desmond Interoperability Tools, parameters in correlation analysis of gas-liquid chromatographic
Schro€dinger, New York (NY), 2016. retention indices of amines. J Chromatogr A. 1985;346:53–60.
[29] Harder E, Damm W, Maple J, et al. OPLS3: a force field providing [50] Zhou Z, Parr RG. Activation hardness: new index for describing
broad coverage of drug-like small molecules and proteins. J the orientation of electrophilic aromatic substitution. J Am Chem
Chem Theory Comput. 2016;12:281–296. Soc. 1990;112:5720–5724.
[30] Hoover WG. Canonical dynamics: equilibrium phase-space distri- [51] Latosinska JN, Latosin
ska M, Maurin JK, et al. Quantum-chemical
butions. Phys Rev A Gen Phys. 1985;31:1695–1697.
insight into structure-reactivity relationship in 4,5,6,7-tetrahalo-
[31] Martyna GJ, Tobias DJ, Klein ML. Constant pressure molecular
geno-1H-benzimidazoles: a combined X-ray, DSC, DFT/QTAIM,
dynamics algorithms. J Chem Phys. 1994;101:4177–4189.
Hirshfeld surface-based, and molecular docking approach. J Phys
[32] Tuckerman M, Berne BJ, Martyna GJ. Reversible multiple time
Chem A. 2014;118:2089–2106.
scale molecular dynamics. J Chem Phys. 1992;97:1990–2001.
[52] Azam F, El-Gnidi BA, Alkskas IA. Combating oxidative stress in
[33] Frisch MJ, Trucks GW, Schlegel HB, et al. 2010. Gaussian 09, revi-
sion E. 01. Wallingford (CT): Gaussian, Inc. epilepsy: design, synthesis, quantum chemical studies and anti-
[34] Wang R, Lu Y, Wang S. Comparative evaluation of 11 scoring convulsant evaluationof 1-(substituted benzylidene/ethylidene)-4-
functions for molecular docking. J Med Chem. 2003;46: (naphthalen-1-yl) semicarbazides. Eur J Med Chem. 2010;45:
2287–2303. 2817–2826.
[35] Azam F, Prasad MVV, Thangavel N. Structure-based design, syn- [53] Aihara J. Reduced HOMO-LUMO gap as an index of kinetic stabil-
thesis and molecular modeling studies of 1-(benzo[d]thiazol-2-yl)- ity for polycyclic aromatic hydrocarbons. J Phys Chem A. 1999;
3-(substituted aryl)urea derivatives as novel anti-Parkinsonian 103:7487–7495.
agents. Med Chem Res. 2012;21:2630–2643. [54] Koopmans T. About the allocation of wave functions and eigen-
[36] Ahmed M, Azam F, Gbaj A, et al. Ester prodrugs of ketoprofen: values of the individual electrons one atom. Physica 1934;1:
synthesis, in vitro stability, in vivo biological evaluation and in sil- 104–113.
ico comparative docking studies against COX-1 and COX-2. Curr [55] Parr RG, Szentpaly LV, Liu S. Electrophilicity index. J Am Chem
Drug Discov Technol. 2016;13:41–57. Soc. 1999;121:1922–1924.

You might also like