You are on page 1of 11

Journal of Saudi Chemical Society (2018) 22, 1003–1013

King Saud University

Journal of Saudi Chemical Society


www.ksu.edu.sa
www.sciencedirect.com

ORIGINAL ARTICLE

Structure elucidation, DNA binding, DFT,


molecular docking and cytotoxic activity studies on
novel single crystal (E)-1-(2-fluorobenzylidene)
thiosemicarbazide
Nasima Arshad a,*, Pervaiz Ali Channar b, Aamer Saeed b,**, Shahid Iqbal Farooqi a,
Aneela Javeed c, Fayaz Ali Larik b, Waqar Ahmad Abbasi b, Ulrich Flörke d

a
Department of Chemistry, Allama Iqbal Open University, 44000 Islamabad, Pakistan
b
Department of Chemistry, Quaid-i-Azam University, 45320 Islamabad, Pakistan
c
Healthcare Biotechnology, Atta-ur-Rehman School of Applied Biosciences, National University of Science and
Technology (NUST), Islamabad, Pakistan
d
Department Chemie, Fakultätfür Naturwissenschaften, Universität Paderborn, Warburgerstrasse 100, 33098 Paderborn, Germany

Received 6 February 2018; revised 23 April 2018; accepted 2 May 2018


Available online 21 May 2018

KEYWORDS Abstract Compound 3 {(E)-1-(2-fluorobenzylidene)thiosemicarbazide} – a new Schiff base of


Thiosemicarbazide Schiff thiosemicarbazide has been synthesized, characterized and reported for crystal structure. Planer side
base; chain in the crystal structure was observed co-planer with aromatic ring plane and molecules were
Crystal structure; connected into centrosymmetric dimmers via intermolecular hydrogen bonding. DFT geometry
DNA binding; optimization and the relevant quantum parameters indicated unstable and reactive nature of com-
DFT calculations; pound 3. Experimental and theoretical findings for DNA binding by UV–visible, cyclic voltamme-
Molecular docking; try and molecular docking studies showed consistency in kinetic (Kb) and thermodynamic (DG)
Huh-7 cell line activity parameters and that compound 3 significantly interacted with DNA via intercalation. Viscometric
analysis further comprehended intercalation as possible binding mode of the compound with DNA
and non-denaturing of DNA in the presence of 10% aqueous DMSO. Docked parameters further

* Corresponding author at: Department of Chemistry, Faculty of Sciences, Allama Iqbal Open University, Islamabad 44000, Pakistan.
** Co-corresponding author at: Department of Chemistry, Quaid-i-Azam University, 45320 Islamabad, Pakistan.
E-mail addresses: nasimaa2006@yahoo.com (N. Arshad), aamersaeed@yahoo.com (A. Saeed).
Peer review under responsibility of King Saud University.

Production and hosting by Elsevier

https://doi.org/10.1016/j.jscs.2018.05.002
1319-6103 Ó 2018 King Saud University. Production and hosting by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
1004 N. Arshad et al.

assured the drug like characteristics of the investigated compound as fit in Lipinski’s criteria. Dose
dependant cytotoxic activity of compound 3 against human Huh-7 cell line indicated its anti-cancer
potential at 100 mg/ml concentration.
Ó 2018 King Saud University. Production and hosting by Elsevier B.V. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction 2. Experimental

The thiosemicarbazide moiety serves as structural template for 2.1. Materials and methods
the synthesis of a wide variety of N and S containing hetero-
cyclic compounds. Moreover, the presence of NH and C‚S Chemicals and reagents used in synthesis, analytical and bio-
moieties plays essential role for the synthesis of various metal logical studies were of analytical grade. All the necessary
complexes. Sulfur and nitrogen atoms of hydrazine in purifications and drying of solvents were carried out according
thiosemicarbazides provide a platform by acting as chelating to standard methods. The dried solvents were stored over
ligands to form complexes of transition metal ions. Thiosemi- molecular sieves. For monitoring of the synthetic reactions;
carbazides have been evaluated over the last 50 years as antivi- thin layer chromatography (TLC) was conducted on 0.25
ral, antibacterial, and anticancer therapeutics and their mm silica gel plates (60 F254, Merck). Visualization of chro-
biological activities are a function of parent aldehyde or matogram was made under UV-lamp at 365 and 254 nm. Rf
ketone moiety [1–7]. value was calculated using a solvent system of petroleum ether:
Schiff bases are nitrogen analogs of the aldehydes or ethyl acetate in 4:1 ratio. The yield (%) given was on the basis
ketones and their metal complexes are reported to have more of 1.0 mM of each precursor used. ds.DNA extraction from
biological potentials than simple organic compounds [8]. Schiff calf thymus gland was done by using Falcon protocol [28].
bases can be easily synthesized by reacting primary amines Autoclaved apparatus and water was used throughout the
with aldehyde or ketones in acidic or medium at neutral pH. extraction procedure. DNA threads were obtained and dis-
Azomethine group containing lone pair on its nitrogen atom solved in water. Stock solution of DNA was further diluted
plays main role in biological processes via hydrogen bonding and monitored on UV–visible spectrophotometer for absor-
interaction with active cell constituents [9–11]. Many biologi- bance at kmax of 260 nm. DNA concentration was determined
cally important Schiff bases have been reported for possessing from Lambert-Beer’s equation (A = eCL; where e260 (DNA)
antibacterial [1–7], antifungal [12–14], antimicrobial [15–17] = 6600 cm1 M1, C = DNA concentration, L = cuvette
and anti-HIV [18,19] activities. Schiff base have also been path length) [28,29]. Further, absorbance ratio A260/A280 was
investigated for antitumor and anti inflammatory activities evaluated greater than 1.8 which assured the DNA purity
using animal tumors as well as by in-vitro biological and ana- [30]. The stock solution of synthesized compound 3 was pre-
lytical methods [20–22]. Other applications of Schiff bases are pared by dissolving it in 10% aqueous DMSO. For DNA
reported for their chemotherapeutic, catalytic, polymer stabi- binding studies, compound’s concentration was optimized to
lizer and corrosion inhibition activities [10,11,23,24]. 1 mM and kept constant while adding various diluted concen-
DNA damaging – the major cause of pathological changes trations of DNA ranging from 40 mM to 320 mM under body
in living organisms like development of several types of can- temperature of 37 °C. In viscometric experiments 10 lM fixed
cers, may arise due to abnormal changes in the DNA structure concentration of DNA was used with varying concentrations
via metabolic disturbance under natural and environmental of compound 3. For cytotoxicity assay, Human Hepatocellular
influence. Chemotherapeutic drugs available in the market Carcinoma cell line (Huh-7) was cultured in Dulbecco’s Mod-
though prohibit the process of cell division but also have sev- ified Eagle Media DMEM – high glucose with 10% FBS
ere side effects. This drawback is still a major challenge for (Thermo Fischer Scientific, USA) and 1% pen-strep MTT
researcher to replace them with those having no or fewer side (3-[4,5-dimethylthiazol-2-yl]-2,5- diphenyltetrazolium bromide
effects. (Sigma Aldrich, USA).
Covalent binding of a compound with DNA is the main
cause to harm to the healthy cell; therefore research on the 2.2. Instrumentations
compounds that interact with DNA non- covalently and rever-
sibly via electrostatic, groove binding and intercalation has Melting point was recorded using a digital Gallenkamp
gained much attention. In this respect, several new compounds (SANYO) model MPD.BM 3.5 apparatus and is uncorrected.
have been explored to probe their potency as therapeutic 1
H NMR and 13C NMR spectra were determined at 300 MHz
agents by reversible DNA binding interactions. Theoretical using a Bruker AM-300 spectrophotometer. FTIR and mass
and analytical studies are considered more appropriate for in spectra (EI, 70 eV) were recorded, respectively, on a Bio-
vitro investigations of anticancer/or antitumor activities of a Rad-Excalibur Series Mode FTS 3000 MX spectrophotometer
drug/ or drug like compound via interaction with DNA [25– (USA) in range of 4000–400 cm1 and on a GC–MS Agilent
27]. In this paper, we are reporting newly synthesized Schiff Technologies 6890N and an inert mass selective detector
bases of thiosemicarbazide for crystal structure and DFT, its 5973 mass spectrometer technologies. Elemental analysis was
DNA binding by UV–visible spectroscopy, cyclic voltamme- conducted using a LECO-183 CHNS analyzer. Shimadzu1800
try, viscometry and molecular docking and compound’s activ- spectrophotometer (TCC-240A, Japan) armed with tempera-
ity for cancer cell line. ture control device was used to record the electronic
Studies on single crystal (E)-1-(2-fluorobenzylidene)thiosemicarbazide 1005

absorption spectra using 1.0 cm matched quartz cells. Hettich have been deposited with the Cambridge Crystallographic
EBA20 Portable Centrifuge C 2002 (Max. speed: 6000 min1) Data Centre as supplementary publication no. CCDC-
and a vortex machine were used during the extraction of DNA 1567724. Copies of available material can be obtained free of
from chicken blood. ‘‘AUTOLAB” PGSTAT-302 poten- charge via www.ccdc.cam.ac.uk.
tiostat/galvanostat with the electrochemical software package
GPES version 4.9 (Eco Chemie, Utrecht, Netherlands) was 2.6. Experimental procedures for DNA binding investigations
used for cyclic voltammetric experiments. Viscosity experi-
ments were carried out on an automated Schott Gerate vis- 2.6.1. Spectroscopic titrations
cometer (Model; AVS 310). Theoretical quantum chemical
calculations were done by using Gaussian 09 software, while In order to investigate binding of the synthesized compound 3
for molecular docking simulation MOE-dock by Chemical with DNA; spectroscopic titrations were carried out on UV–
Computing Group Inc was used. Molecular modeling studies visible spectrophotometer by maintaining the temperature of
were performed on Pentium 1.6 GHz workstation, 512 MB cell cuvette at 37 °C. Initially, spectra were run separately for
memory with the Windows Operating System that applies a the compound and DNA. Then 40–320 mM DNA was titrated
two stage scoring process to sort out the best conformations in aliquot form with 1 mM fixed concentration of the com-
and orientations of the ligand based on its interaction pattern pound. Variations in the spectral responses were recorded
with the DNA. separately at 37 °C and absorbance data were obtained for
the compound before and after the addition of various concen-
trations of DNA.
2.3. General procedure for synthesis

2.6.2. Voltammetric titrations


The thiosemicarbazide (0.138 g, 1.0 mmol) was introduced to a
stirred solution of suitably substituted benzaldehyde (1.0 Glassy carbon (d = 3 mm), saturated calomel (SCE; 3.5 M
mmol) in absolute ethanol (10 ml) along with 1–2 drops of KCl) placed in luggin capillary and a Pt wire as working,
concentrated sulfuric acid. The reaction mixture was refluxed reference and counter electrode, respectively, were used in
for 3–6 h and completion of the reaction was checked by double wall Dr. Bob cell for electrochemical measure-
TLC. The mixture was concentrated and resulted solid product ments. Surface of glassy carbon electrode was polished
was separated and recrystallized from ethanol. with powdered a-alumina, washed and dried before run-
ning each voltammetric experiment. Temperature of the
2.4. Characterization data sample solution was maintained at 37 °C by circulating
water in double wall cell. In order to maintain oxygen free
2.4.1. (E)-1-(2-Fluorobenzylidene)thiosemicarbazide – environment in the cell; purging of argon gas (99.999%)
compound 3 for 10–15 min and its blanketing was made before every
electrochemical assay.
Yield: 75%: m.p 235 °C Rf: 0.52; Petroleum ether:ethyl acetate
Cyclic voltammetry of compound 3 alone (1 mM) and in
(6:4) FTIR; (KBr, cm1): 3375 (NH2), 3261 (NAH), 3117 (sp2
the presence of varying DNA concentrations (40–320 mM)
CH), 1624 (C‚N), 1558 (ArAC‚C), 1071 (C‚S), 1H NMR
was carried out within potential range of 0.2 V to 1.4 V
(300 MHz, DMSO d6): d (ppm): 11.56 (s, 1H, NH), 8.27 (s, 1H,
at scan rate of 100 mV/s. Solvent purity was assured by run-
CH‚N), 8.25–7.19 (m, 4H ArAH), 8.08 (s, 2H, ANH2); 13C
ning blank CV within same potential range.
NMR (75 MHz, DMSO d6): d (ppm): 178.5, 162.9, 159.6,
135.2, 127.2, 125.1, 122.2, 116.0. GC–MS (EI, 70 eV): m/z
(%):197 (M+ 100). 2.6.3. Viscosity measurements
Before starting viscosity experiments; various samples with
2.5. Crystal structure determination of compound 3 fixed DNA concentration (10 lM) and varying compound
3 concentrations (10–80 mM; with the difference of 10
C8H8FN3S, Mr = 197.2, colorless crystal, size 0.25  0.18  mM) were prepared separately. Viscosity was measured ini-
0.08 mm3, orthorhombic space group Pbca with Z = 8, a = tially for DNA solution at its fixed concentration as go.
11.845(2), b = 6.6894(14), c = 22.759(5) Å, V = 1803.3(6) Then each sample was run separately and viscosity was
Å3; Dc = 1.453 Mg/m3, l = 0.328 mm1, F(0 0 0) = 816. measured as g. During each measurement, temperature
The intensity data were recorded using a Bruker SMART was kept at 37 °C. The values of cube root of relative
CCD area-detector diffractometer with graphite monochro- specific viscosity i.e., (g/go)1/3 was determined and then
mated MoKa radiation (k = 0.71073 Å) at T = 130(2) K. plotted against compound-DNA concentration ratio i.e.,
15,834 reflections collected 1.8 > > 27.9°; 2151 independent [compound]/[DNA].
reflections I > 2r(I), Rint = 0.052. Structure solution by
direct methods, full-matrix least squares refinement based on 2.7. Theoretical procedures
F2 and 118 parameters [31,32]. All but H-atoms were refined
anisotropically, hydrogen atoms were clearly located from dif- 2.7.1. DFT method
ference Fourier maps and then refined at idealized positions Quantum semi imperial computational studies for com-
riding on the carbon atoms with isotropic displacement param- pound structure were carried out by density functional the-
eters Uiso(H) = 1.2Ueq(C/N) and CAH 0.95 Å, NAH 0.88 Å. ory (DFT) method. Molecular geometry of compound 3
Refinement converged at R1 = 0.039[I > 2r(I)], wR2 = was optimized by using Gaussian 09 at DFT/B3LYP/basis
0.098 [all data] and S = 1.04; min./max. DF 0.32/0.37 e/Å3. set (6-31G) level and quantum computational parameters
Crystallographic data for the structure reported in this paper were obtained.
1006 N. Arshad et al.

Scheme 1 Synthesis of compound 3; (E)-1-(2-fluorobenzylidene)thiosemicarbazide.

2.7.2. Molecular docking method 3. Results and discussion


Structure of compound 3 was drawn and minimized on MOE
window using MOE builder and entered into MOE database. 3.1. Chemistry of synthesized compound 3
The starting point of the docking simulation was the X-ray
structure of the DNA with PDB ID 1BNA obtained through Synthesis of target compound was carried out according to
the protein data bank (PDB) and imported to MOE window. synthetic route outlined in Scheme 1. The thiosemicarbazide
All water molecules were removed from the complex with 12 in dry ethanol was allowed to react with suitably substituted
base pairs running in 50 -30 direction. The base pair sequence hetero aromatic aldehyde in the presence of sulfuric acid as
was (50 -D(CGCGAATTCGCG)-30 ): (50 -D(CGCGAATTCGC catalyst to obtain the desired compound 3 in good to high
G)-30 ) with molecular weight of 7326.84. All hydrogen atoms yields. The compound obtained was purified by recrystalliza-
were added to the structure with their standard geometry fol- tion from aqueous ethanol.
lowed by their energy optimization tool using MOPAC 7.0. In the typical case of compound 3, presence of the C‚S
The resulting DNA model was subjected to systematic confor- functional group was marked by the appearance of stretching
mational search at default parameters with RMS gradient of bands at 1071 cm1, for C‚N at 1624 cm1, for ArAC‚C at
0.01 kcal mol1 using Site Finder. A number of runs were car- 1558 cm1 and a stretching around 3375 and 3261 cm1 indi-
ried out to get a final binding docking pose as accurate as pos- cating the presence of NH2 and NH linkage of the hydrazide
sible. The best conformation was selected based on energetic moiety. In 1H NMR spectrum the appearance of a singlet at
ground and the minimum final docking energy (DG) [25,26]. d 8.27 for azomethine proton (HC‚N) and singlets at d 11.5
and 8.08 were noticed for NH and NH2, respectively (See Sup-
2.8. In vitro cytotoxic assay plementary data). The characteristic azomethine protons were
observed in the range of 8.27 ppm in 1H NMR spectra. In 13C
NMR spectra, the signals at d 178.0 (C‚S) and 162 (C‚N)
Anticancer potential of the synthesized compound 3 was tested
confirmed the formation of the thiosemicarbazones.
by a colorimetric MTT assay as reported earlier [33]. Human
Hepatocellular Carcinoma cell line (Huh-7) was used to evalu-
3.2. Description of crystal structure
ate the cytotoxic effect of compound 3. The cell lines were
grown and maintained in DMEM – high glucose supplemented
with 10% FBS (phosphate buffer saline) and 1% pen-strep The molecular structure of synthesized compound 3 is repre-
antibiotic. For MTT assay; 1  104 cells were plated into each sented in Fig. 1. The molecular structure was found closely
of 96 well plates and incubated for attachment overnight related to that of bis-fluoro substituted analogs [34]. It showed
before incubating them with varying concentration (25– that the side chain is almost co-planar with the aromatic ring
100ug/ml) of compound 3 in triplicates. PBS was added in con- plane; the relevant torsion angles measured as
trol wells. Cells were incubated again for 24 h at 37 °C incuba- C3AC2AC1AN1 9.56(16)° and C2AC1AN1AN2 0.68(14)°.
tor with 5% CO2. MTT (3-[4,5-dimethylthiazol-2-yl]-2,5- The side chain itself was planar with largest deviation of 0.68
diphenyltetrazolium bromide) reagent was added to each well, (1) Å for N2 from the best plane through C1 to S1. Pertinent
including control. The plates were then incubated for 3 h until
purple precipitates of formazan were visible. After aspirating
all solutions from the wells, Formazan were dissolved in 100
ml of DMSO, by keeping the plates covered in the dark for
1 h at room temperature. The absorbance was recorded at
570 nm in a microplate reader. Percentage cytotoxicity was cal-
culated as;

% cell viability ¼ ðAs  Ab Þ=ðAc  Ab Þ  100

where As, Ab and Ac corresponds to absorbance value of sam-


ple, blank and control respectively. The IC50 value was extrap-
olated from the dose-response graph. The concentrations of
compound 3 that reduced the viability of the cells by 50% were
determined by plotting the triplicate data sets over various
concentration ranges. The values were calculated using regres- Fig. 1 Molecular structure of compound 3. Anisotropic dis-
sion analysis of PRISM program. placement ellipsoids are drawn at the 50% probability level.
Studies on single crystal (E)-1-(2-fluorobenzylidene)thiosemicarbazide 1007

3.3. DNA binding studies by UV–visible spectroscopy

UV–visible spectroscopy is the most significant technique to


learn the binding mode of small molecules and has been
reported several times for DNA binding interactions, kinetics
and mechanism [20,25–27]. Compound–DNA interaction can
easily be assessed by monitoring spectral changes in the com-
pound’s spectra after DNA addition; hence is considered an
effective tool for recognition of their interplay, understanding
of binding mechanism and complex formation. The changes
observed in the UV-spectra of a compound upon DNA titra-
tions have provided evidences of the interactional modes. In
case of intercalative binding mode, p-p* stacking interactions
may appear and resultant spectral changes are observed as
decrease in the absorbance peak (hypochromism) [35]. Fur-
ther, bathochromic (red) shift in the spectral response after
Fig. 2 Unit cell with intermolecular H-bonding pattern as dotted DNA addition confirm stabilization of the DNA duplex [35].
lines, viewed along b-axis. H atoms not involved are omitted. The UV-spectra of investigated compound 3 were recorded
in 10% aqueous DMSO at physiological temperature 37 °C
before and after the addition of various concentrations of
bond geometries were N1AN2 1.3784(19), N1AC1 1.279(2), CT-DNA, Fig. 3. Compound’s absorbance peak appeared at
N2-C8 1.347(2), N3AC8 1.325(2), C8AS1 1.6953(17) and kmax = 270.5 nm. Upon addition of various aliquots of
C3AF1 1.358(2) Å. The crystal packing is given in Fig. 2 which DNA, peak intensity drop up to 12.7% as calculated by
showed intermolecular N3-H32A. . .S(-x + 1, y, z + 1) the following formula.
hydrogen bonds with NAH 0.88 Å, H. . .S 2.53 Å and
Afree  Abound
NAH. . .S 170.4° that connected the molecules into centrosym- H% ¼  100:
metric dimers which are stacked along the b-axis. The crystal Afree
data and structure refinement for compound 3 are provided A slight red shift of 1.5 nm was also observed which along
in Table 1 and selected bond lengths and angles are listed in with hypochromic effect validated the formation of complex
Table 2. via p-p* stacking interaction between the chromophoric part

Table 1 Crystal data and structure refinement for compound 3.


Empirical formula C8 H8 F N3 S
Formula weight 197.23
Temperature 130(2) K
Wavelength 0.71073 Å
Crystal system Orthorhombic
Space group Pbca
Unit cell dimensions a = 11.845(2) Å a = 90°
b = 6.6894(14) Å b = 90°
c = 22.759(5) Å c = 90°
Volume 1803.3(6) Å3
Z 8
Density (calculated) 1.453 Mg/m3
Absorption coefficient 0.328 mm1
F(0 0 0) 816
Crystal size 0.25  0.18  0.08 mm3
Theta range for data collection 1.79 to 27.87°.
Index ranges 15h11, 8k8, 29l29
Reflections collected 15,834
Independent reflections 2151 [R(int) = 0.0523]
Completeness to theta = 27.87° 100.0%
Absorption correction Semi-empirical from equivalents
Max. and min. transmission 0.9742 and 0.9225
Refinement method Full-matrix least-squares on F2
Data/restraints/parameters 2151/0/118
Goodness-of-fit on F2 1.038
Final R indices [I > 2sigma(I)] R1 = 0.0393, wR2 = 0.0909
R indices (all data) R1 = 0.0550, wR2 = 0.0987
Largest diff. peak and hole 0.368 and 0.323 e.Å3
1008 N. Arshad et al.

respectively. From the plot of Ao/(A  Ao) to 1/[DNA],


Table 2 Bond lengths (Å) and angles (°) for Compound 3.
the ratio of the intercept to the slope gave the value of
Bond distance (Å) binding constant, Kb.
S(1)AC(8) 1.6953(17) C(2)AC(3) 1.389(2) The evaluated binding constant (Kb = 6.02  103 M1)
F(1)AC(3) 1.358(2) C(2)AC(7) 1.398(3) further depicted intercalative binding of compound 3 with
N(1)AC(1) 1.279(2) C(3)AC(4) 1.378(3) DNA as this value was found comparable with reported inter-
N(1)AN(2) 1.3784(19) C(4)AC(5) 1.377(3) calator, isoxazocucumine (6.3  103 M1) [37]. Using the value
N(2)AC(8) 1.347(2) C(5)AC(6) 1.391(3) of binding constant in equation (DG = RT lnKb); free
N(3)AC(8) 1.325(2) C(6)AC(7) 1.384(3) energy change, DG, was evaluated as negative value which
C(1)AC(2) 1.460(2)
indicated spontaneous binding of the compound with DNA.
Bond angles (°)
C(1)AN(1)AN(2) 114.75(14) C(4)AC(3)AC(2) 123.40(18) 3.4. DNA binding studies by cyclic voltammetry
C(8)AN(2)AN(1) 119.91(14) C(5)AC(4)AC(3) 118.25(18)
N(1)AC(1)AC(2) 120.14(16) C(4)AC(5)AC(6) 120.62(17)
C(3)AC(2)AC(7) 116.93(16) C(7)AC(6)AC(5) 119.92(19) Cyclic voltammetry is the most versatile, flexible and conve-
C(3)AC(2)AC(1) 120.26(16) C(6)AC(7)AC(2) 120.85(17) nient way for kinetic, analytical, thermodynamic and mech-
C(7)AC(2)AC(1) 122.80(15) N(3)AC(8)AN(2) 117.80(15) anistic studies. Interactional studies in particular to
F(1)AC(3)AC(4) 118.15(16) N(3)AC(8)AS(1) 123.30(13) compound binding with DNA could easily be assessed by
F(1)AC(3)AC(2) 118.43(15) N(2)AC(8)AS(1) 118.90(13) cyclic voltammetry in term of current - potential variations
in the voltammetric response of the redox active compound
[20,25,27].
DNA binding of compound 3 was studied in 10% aqueous
DMSO at 37 °C. Cyclic voltammogram of compound showed
a single irreversible reduction peak at 0.727 V, while scan-
ning the voltammograms in the presence of DNA concentra-
tions showed a significant decrease in the reduction peak
current, Fig. 4. The current drop was evaluated 10.3% using
following equation;
Io  I
%Current ¼  100
Io

Decrease in the peak current showed less availability of


free compound in the bulk solution due to formation of
slowly diffusing compound–DNA complex. A small shift
toward more negative potential (0.20 V) has indicated the
difficulty in reduction process at the surface due to com-
pound–DNA interaction [38]. Binding constant (Kb) for
the compound–DNA complex by cyclic voltammetry was
evaluated through peak current variations using following
equation [39];
1
Fig. 3 Absorption spectrum of compound 3 in the absence (a) I2p ¼ ðI2  I2p Þ þ I2po  ½DNA
Kb ½DNA po
and presence of 40(b), 80(c), 120(d), 160(e), 200(f), 240(g), 280(h),
320(i) mM DNA. The arrow direction indicates the increasing where Kb is the binding constant, Ipo and Ip are peak currents
conc. of DNA. The inset graph represents the plot of A0/A  A0 before and after the addition of DNA. A plot of I2p vs. I2po  I2p/
vs. 1/[DNA](mM)1 for the calculation of binding constant (Kb) [DNA] gave a straight line with a slope equal to the reciprocal
and Gibb’s free energy(DG). of binding constant, Kb. The binding constant and free energy
change were evaluated 6.86  103 M1 and 18.74 kJ mol1,
respectively. These values were found comparable with that
obtained from spectroscopic studies.
of compound 3 and DNA base pairs [35]. Spectral findings
obtained from UV-experiments advised that the compound 3.5. Viscometric analysis for DNA binding confirmation
may bind to CT-DNA via intercalation [20,25,27]. Further-
more, binding constant (Kb) for the compound–DNA complex
Viscosity determination of DNA in the presence of com-
was obtained by plotting Ao/(A  Ao) vs. 1/[DNA] and by fit-
pound’s concentration is the most appropriate and simple
ting the data in Benesi-Hildebrand equation [36].
way to verify the possible mode of interaction of a compound
Ao eG eG 1 through which it could bind with DNA. The results obtained
¼ þ from viscometric analysis provide best compliments to other
A  Ao eHG  eG eHG  eG Kb ½DNA
experimental techniques used in DNA binding studies. Fur-
where, Ao and A are the absorbance of free and DNA ther, this technique could justify the state of DNA either dena-
bound complex respectively, eG and eHG the molar tured or not if the medium mostly consists of non aqueous
extinction coefficients of free and DNA bound complex solvent like DMSO [40].
Studies on single crystal (E)-1-(2-fluorobenzylidene)thiosemicarbazide 1009

Fig. 4 Voltammetric responses of compound 3 in the absence (a) and presence of 40(b), 80(c), 120(d), 160(e), 200(f), 240(g), 280(h), 320
(i) mM DNA. Arrow direction indicates the increasing conc. of DNA. The inset graph represents the plot of I2p vs. I2po- I2p/[DNA] for the
calculation of binding constant (Kb) and Gibb’s free energy (DG).

3.6. Computational structure analysis by DFT studies

Density functional theory (DFT) is a quantum mechanical


semi – imperial approach for theoretical investigations on elec-
tronic structure of a molecule. Maximum charge distribution
and various computational parameters including HOMO-
LUMO energies, HOMO-LUMO energy gap, binding energy,
dipole moment, ionization potential, electron affinity, elec-
tronegativity, global hardness etc can be calculated from the
optimized geometries. Interpretation of these theoretical
results could help to understand the molecular structure and
possibilities of interactions with biological macromolecules
like DNA.
The molecular geometry of compound 3 was optimized by
DFT/ B3LYP/6-31G level using Gaussian 09 software. The
charge distributions on the optimized geometry and HOMO-
Fig. 5 Plot of relative specific viscosity vs. [Compound]/[DNA] LUMO orbital’s are provided in Fig. 6, while computed
by adding increasing concentrations of compound 3 i.e., 10–80 mM parameters are given in Table 3. Optimized structure indicated
upon fixed 10 mM DNA concentration at 37 °C. that compound 3 has planar geometry which is property of an
intercalator, Fig. 5. The greater negative charge (0.317) on
Due to non-dissolving nature of investigated compound fluorine atom substituted on ring indicated the possibility of
3 in aqueous medium; present studies were carried out in its interaction with hydrogen of DNA base pairs while planar
mixed solvent of 10% aqueous DMSO (DMSO-H2O in part of the molecule intercalated between the DNA base pairs.
9:1v/v ratio). As denaturation of DNA (i.e., ds.DNA con- HOMO and LUMO energies are directly related to ioniza-
verted into ss.DNA) could arise in the presence of DMSO; tion potential and electron affinity, respectively, as electron is
viscosity measurement was considered mandatory in present donated by HOMO to the LUMO orbital. Stability,
work. polarizability and reactivity of a molecule could be decided
Upon increasing compound 3 concentration, the rela- on the basis of HOMO-LUMO energy gap and binding
tive viscosity of DNA was increased gradually, Fig. 5. energy. If energy gap is small and binding energy is negative
This increase in viscosity represented widening of DNA in value, the molecule is considered unstable/or less stable,
duplex as compound 3 molecules accommodated into more polarizable and more reactive [42]. The greater value of
the base pairs pockets [41]. Further, linear rise in the rel- HOMO (2.07 eV) than LUMO energy and very small value
ative viscosity was suggestive of intercalative binding of for HOMO-LUMO energy gap showed electron donation abil-
compound 3 with DNA [40]. Since DNA base pairs are ity from HOMO to LUMO and that the investigated molecule
separated during its denaturing; there should be lowering is less stable and more reactive [42]. The negative binding
in the viscosity values. In present work enhancement in energy further assured compound’s instability. More reactive
the viscosity was observed with the investigated com- nature of the tested compound 3 could further be rationalized
pound 3; hence the possibility of DNA denaturing could on the basis of small value of hardness and greater value of
be excluded. dipole moment [43].
1010 N. Arshad et al.

Fig. 6 The frontier molecule orbital density distribution of compound 3 after optimization at DFT/B3LYP/6-31G level. Top; optimized
structure, Bottom left; Bonding molecular orbital (HOMO), Bottom right; Anti-bonding molecular orbital (LUMO).

evaluated as 2.89 Å and 2.99 Å respectively, which may render


Table 3 Quantum-chemical parameters of compound 3 com-
its greater binding strength and complex stability [26].
puted with DFT at B3LYP/6-31G level.
Theoretical values of binding constant (Kb) and free energy
Parameters Computed values change (DG) were evaluated 2.51  103 M1 and 20.18 kJ
E Homo(ev) 0.207 mol1, respectively and showed consistency with that calcu-
E Lumo(ev) 0.072 lated experimentally form UV–visible spectroscopy and cyclic
DE (eV) 0.135 voltammetry. The other electronic and steric descriptors were
Ionization potential (eV) 0.207 calculated from molecular docking data and given in Table 4.
Electron affinity (eV) 0.072 The values evaluated for these descriptors revealed significant
Binding energy (a.u) 971.76401827 potential of compound 3 for binding with one or the more
Dipole moment (D) 9.969
DNA base pairs.
Electronegativity (X) 0.1395
Global hardness (H) 0.0675
Point group C1 3.8. Drug like characters of compound 3

Theoretical descriptors evaluated from docking data, Table 4,


could further help to investigate the pharmacological behavior
3.7. Molecular docking studies of a compound if obeying the criteria of Lipinski’s rule of five
[44]. Compound 3 was found to have hydrogen bond (HB)
Molecular docking is the most suitable way for theoretical donor atoms (the total number of nitrogen–hydrogen and oxy-
understanding of molecular mechanism and for the elucidation gen–hydrogen bonds) < 5, HB acceptor atoms (all nitrogen or
of binding mode/modes of a compound with DNA through oxygen atoms) < 10, molecular mass < 500 g/mol and octa-
non-covalent interactions. This theoretical approach is consid- nol–water partition coefficient S logP < 5. These values were
ered most appropriate for structure based- drug design. Molec- in accordance with Lipinski’ criteria for a compound to behave
ular docking studies along with experimental studies could like a drug. However, molar refractivity (MR) value was eval-
help to explore a compound as a potential drug candidate. uated 5.3916 which were lesser than Lipinski’s limits i.e., 40–
Molecular docking of compound 3 was performed to pre- 130. Since Lipinski’s criteria allow only one violation of the
dict its binding with DNA. The docked conformation of the five rules for an orally active drug; compound 3 may exhibit
compound with lowest free energy and pose view image are drug like characteristics. However, further studies on its phar-
shown in Fig. 7. In Fig. 7 (right) dotted lines in curve shapes macological and pharmaceutical activities may lead it to be
showed solvent contact and blue blurred regions explained used as drug.
direct exposure of the ligand to hydrophobic core of 1BNA.
Docked complex indicated intercalation of the compound with 3.9. Cytotoxic activity
1BNA as shown in Fig. 7 (left). However, in 2D lig plot (Fig. 7,
right) two additional hydrogen bonds were observed between The synthesized compound 3 was tested for in vitro anti-cancer
two hydrogen atoms at NH2 of thiosemicarbazide moiety of activity against Human Hepatocellular Carcinoma (Huh-7)
compound 3 and cytosine (DC 13) and adenine (DA 14) bases cell line. Incorporation of cell culture studies offer a gold stan-
of DNA. The bond lengths H  DC13 and H  DA14 were dard to evaluate the anticancer potential of the compounds
Studies on single crystal (E)-1-(2-fluorobenzylidene)thiosemicarbazide 1011

Fig. 7 Molecular docked complex of compound 3 with IBNA 3D complex (left); 2D lig plot showing interactions of compounds with
base pairs of 1BNA (right).

Table 4 Electronic and steric descriptors calculated from molecular docking data.
Electronic descriptors
Complex EHOMO ELUMO Eele Evander EIP ETotal
kcal/mol kcal/mol kcal/mol kcal/mol kcal/mol kcal/mol
Comp. 3 – 1BNA 8.8178 1.1053 254066.29 19.5233 8.8178 50316.14
Steric descriptors
Complex Hf Mr S logP Vsurf Dipole HB donor atoms HB acceptor atoms
kcal/mol
Comp. 3 – 1BNA 48.0713 5.3916 0.9928 210.8906 6.3717 2 2

Fig. 8 In vitro cell viability (left) and cytotoxicity (right) of compound 3 on Huh-7 Cell line at various concentrations.

due to controlled conditions and automated procedures. The all other concentrations showed greater percentage viability
% cytotoxicity of test compound was evaluated by using (above 50%). The results clearly indicated that the compound
MTT assays using various concentrations of compound 3, has shown anticancer (cytotoxic) potential in dose dependent
Fig. 8 and Table 5. The % viability of the cells decreased with manner. The maximum anticancer potential was observed with
increasing concentrations of the compound and at 100 mg/ml 100 mg/ml concentration and the extent of cytotoxicity was
concentration cell viability reduced to less than 50%, while 58.556%. The dose dependent inhibition in cell growth was
1012 N. Arshad et al.

[3] H.A. Tang, L.F. Wang, R.D. Yang, Transit. Met. Chem. 28
Table 5 % cell viability and % cytotoxicity data. (2003) 395–398.
Conc. (mg/ml) % Cell Viability % Cytotoxicity [4] M. Aggarwal, B. Kondeti, R. McKenna, Expert Opin Ther Pa.
23 (2013) 717–724.
0 100 0
[5] V. Alterio, A. Di Fiore, K. D’Ambrosio, C.T. Supuran, G. De
25 98.17949 1.82051
Simone, Chem. Rev. 112 (2012) 4421–4468.
50 71.78632 28.21368
[6] A.R. Brash, J. Biol Chem. 274 (1999) 23679–23682;
75 62.23077 37.76923
A.B. Camargo, E. Marchevsky, J.M. Luco, J. Agric. Food
100 41.444 58.556
Chem. 55 (2007) 3096–3103.
[7] L. Dailey, P. Imming, Curr. Med. Chem. 6 (1999) 389–398.
[8] Z.H. Chohan, M. Praveen, A. Ghaffar, Met. Based Drugs 4
(1997) 267–272.
found more significant at 100 mg/ml of test compound and [9] S. Patai, The Chemistry of Carbon–Nitrogen Double Bond,
revealed its potency against liver cancer cell line at this Interscience, New York, 1970, pp. 149–180.
[10] K.N. Venugopala, B.S. Jayashree, Indian J. Heterocycl. Chem.
concentration.
12 (2003) 307–310.
IC50 value of test compound was determined to be 72.94 [11] K. Vashi, H.B. Naik, Eur. J. Chem. 1 (2004) 272–276.
mg/ml (14.37 mM) which is greater than that reported for refer- [12] A. Nunez-Montenegro, R. Carballo, E.M. Vazquez-Lopez,
ence standard anticancer drug Tamoxifen (IC50 mM; 5.8 and Polyhedron 27 (2008) 2867.
10.5 for the cell growth inhibition of MCF-7, SaOS-2 cell lines, [13] A. Nunez-Montenegro, R. Carballo, E.M. Vazquez-Lopez,
respectively) [45]. The result thus clearly indicated that Polyhedron 27 (2008) 3915.
compound 3 has a potential anticancer activity against the [14] K. Alomar, M.A. Khan, M. Allain, G. Bouet, Polyhedron 28
hepatocellular carcinoma and could be further considered for (2009) 1273.
its anticancer potential evaluation for other type of cancers [15] P.M. Krishna, K.H. Reddy, Inorg. Chim. Acta 362 (2009) 4185.
using other relevant cell lines. Furthermore; compound 3 can [16] M. Belicchi-Ferrari, F. Bisceglie, E. Buluggiu, G. Pelosi, P.
Tarasconi, Polyhedron 28 (2009) 1160.
also be tested in vivo for its anticancer potential using animal
[17] T.S. Lobana, P. Kumari, R.J. Butcher, Inorg. Chem. Commun.
models. 11 (2008) 11.
[18] T.S. Lobana, R. Sharma, R.J. Butcher, Polyhedron 28 (2009)
4. Conclusions 1103.
[19] T.S. Lobana, S. Khanna, G. Hundal, R.J. Butcher, A.
Castineiras, Polyhedron 28 (2009) 3899.
Compound 3 {(E)-1-(2-fluorobenzylidene)thiosemicarbazide}
[20] N. Arshad, M. Ahmad, M.Z. Ashraf, H. Nadeem, J.
was synthesized, characterized and its structure was deter-
Photochem. Photobiol B: Biol. 138 (2014) 331–346.
mined by X-ray single crystal analysis. DFT charge distribu- [21] E.M. Eudnett, W.J. Dunn, J. Med. Chem. 15 (1972) 339.
tion and chemical reactivity descriptors indicated that the [22] E.M. Eudnett, P.D. Mooney, J. Med. Chem. 13 (1970) 786.
synthesized compound is reactive. Spectroscopic, voltammet- [23] D.N. Dhar, C.L. Taploo, J. Sci.i Ind. Res. 41 (1982) 501–506.
ric, viscometric and docking studies revealed intercalation as [24] S. Li, S. Chen, S. Lei, H. Ma, R. Yu, Corrosion Sci. 41 (1999)
a prominent mode for compound 3-DNA interaction. Experi- 1273–1287.
mental and theoretical findings were in agreement. Computed [25] N. Arshad, M. Zafran, Z. Ashraf, F. Perveen, J. Photochem.
parameters showed drug likeness of compound 3. IC50 value Photobiol. B: Biol. 169 (2017) 134–147.
evaluated for 100 mg/ml of compound 3 against Human Hepa- [26] N. Arshad, F. Perveen, P.A. Channar, S.I. Farooqi, A. Saeed, F.
A. Larik, H. Ismail, B. Mirza, J. Mol. Struct. 1139 (2017) 371–
tocellular Carcinoma cell line (Huh-7) showed its potency
380.
against liver cancer.
[27] N. Arshad, M.H. Bhatti, S.I. Farooqi, S. Saleem, B. Mirza,
Arab. J. Chem. 9 (2016) 451–462.
Acknowledgments [28] T. Maniatis, E.F. Fritsch, J. Sambrook, Molecular Cloning: A
Laboratory Manual, Cold Spring Harbor, New York, 1989.
The authors would like to thanks Chemistry Departments of [29] M.E. Reichmann, S.A. Rice, A. Thomas, P. Doty, J. Am. Chem.
Allama Iqbal Open University and Quaid-i-Azam University Soc. 76 (1954) 3047–3053.
and Healthcare Biotechnology Department of NUST for [30] S.S. Babkina, N.A. Ulakhovich, Anal. Chem. 77 (2005) 5678–
experimental and theoretical facilities in Pakistan and Depart- 5685.
[31] Bruker, SMART (Version 5.63), SAINT (Version 6.02), Bruker
ment Chemie Paderborn Germany for crystal structure.
AXS Inc., Madison, Wisconsin, USA, 2002.
[32] G.M. Sheldrick, Acta Cryst. 112 (2008) A64.
Appendix A. Supplementary data [33] J. Carmichael, W.G. DeGraff, A.F. Gazdar, J.D. Minna, J.B.
Mitchell, Cancer Res. 47 (1987) 936–942.
Supplementary data associated with this article can be found, [34] J.L. Bautista, M. Flores-Alamo, J. Tiburcio, R. Vieto, H.
in the online version, at https://doi.org/10.1016/j.jscs.2018.05. Torrens, Molecules 18 (2013) 13111.
[35] M. Sirajuddin, S. Ali, N.A. Shah, M.R. Khan, N.M. Tahir,
002.
Spectrochim. Acta Part A: Mol. Biomol. Spectros. 94 (2012)
134–142.
References [36] H.A. Benesi, J.H. Hildebrand, J. Am. Chem. Soc. 71 (1949)
2703–2707.
[1] H. Beraldo, D. Gambino, Mini-Rev. Med. Chem. 4 (2004) 31– [37] N. Shahabadi, A. Fatahi, J. Mol. Struct. 970 (2010) 90–95.
39. [38] I. Kocak, U. Yildiz, B. Coban, A. Sengul, J. Solid State
[2] J.S. Casas, M.S. Garcı́a-Tasende, J. Sordo. Coord. Chem. Rev. Electrochem. 19 (2015) 2189–2197.
209 (2000) 197–261. [39] J. Niu, G. Cheng, S. Dong, Electrochim. Acta 39 (1994) 2455.
Studies on single crystal (E)-1-(2-fluorobenzylidene)thiosemicarbazide 1013

[40] N.K. Janjuaa, A. Shaheen, A. Yaqub, F. Perveen, et al, [43] S. Špirtović-Halilović, M. Salihović, E. Veljović, A. Osmanović,
Spectrochim. Acta Part A 79 (2011) 1600–1604. S. Trifunović, D. Završnik, Bull. Chem. Techool. Bosnia
[41] G. Zhang, J. Guo, J. Pan, X. Chen, J. Wang, J. Mol. Struct. 923 Herzegovina 43 (2014) 57–60.
(2009) 114–119. [44] A. Leo, C. Hansch, D. Elkins, Chem. Rev. 71 (1971) 525–616.
[42] C.S. Abraham, J.C. Prasana, S. Muthu, Spectrochim. Acta Part [45] D. Chowrasia, C. Karthikeyan, L. Choure, M.G. Sahabjada, M.
A: Mol. Biomol. Spectros. 181 (2017) 153–163. d. Arshad, P. Trivedi, Arab. J. Chem. 10 (2017) S2424–S2428.

You might also like