You are on page 1of 14

Polyhedron 168 (2019) 113–126

Contents lists available at ScienceDirect

Polyhedron
journal homepage: www.elsevier.com/locate/poly

Energetically favorable anti-electrostatic hydrogen bonded cationic


clusters in Ni(II) 3,5-dimethylpyrazole complexes: Anticancer evaluation
and theoretical studies
Anshuman Gogoi a, Debajit Dutta a, Akalesh K. Verma b, Hiren Nath a, Antonio Frontera c,
Ankur K. Guha a, Manjit K. Bhattacharyya a,⇑
a
Department of Chemistry, Cotton University, Guwahati 781001, Assam, India
b
Department of Zoology, Cell & Biochemical Technology Laboratory, Cotton University, Guwahati 781001, Assam, India
c
Departament de Química, Universitat de les Illes Balears, Crta de valldemossa km 7.7, 07122 Palma de Mallorca (Baleares), Spain

a r t i c l e i n f o a b s t r a c t

Article history: Two new Ni(II) coordination complexes of pyrazole and acetato ligands, involving nitrate and chloride
Received 20 March 2019 counter anions, viz. [Ni(Hdmpz)2(CH3COO)(H2O)3]NO32H2O (1) and [Ni(Hdmpz)2(CH3COO)(H2O)3]Cl
Accepted 14 April 2019 (2) (where Hdmpz = 3,5-dimethylpyrazole), have been synthesized in purely aqueous medium at room
Available online 7 May 2019
temperature. The complexes have been characterized by elemental analysis, FT-IR, electronic
spectroscopy, PXRD, TGA and single crystal X-ray diffraction. The H-bonds established between the
Keywords: isostructural cationic [Ni(Hdmpz)2(CH3COO)(H2O)3]+ moieties of both complexes can be classified as
Synthesis
anti-electrostatic H-bonds (AEHBs) and these have been studied using DFT calculations. Remarkably, in
Anti-electrostatic hydrogen bond
Anticancer
one case these AEHBs exhibit favorable interaction energies, thus indicating that the attractive nature
Apoptosis of the H-bonds is able to compensate the pure electrostatic repulsion. The analyses of the cell viability
Docking results for the DL cell line show a significant concentration dependent decrease by compounds 1 and 2
with a negligible cytotoxic effect observed against normal cells (PBMC) in comparison to the cancer cells
(DL). The cytotoxic activity of these compounds in DL cells occurs via apoptosis. Molecular docking and
SAR analysis reveal that the compounds possess high binding affinities against most common cancer tar-
get proteins.
Ó 2019 Elsevier Ltd. All rights reserved.

1. Introduction chemistry, optics, magnetism and luminescence etc. [13,14]. Nickel


pyrazole complexes with varied pharmacological actions, including
Utilization of supramolecular architectures via non-covalent antifungal [15,16], antibacterial [17] and antiproliferative/anti-
interactions is a vigorous field in crystal engineering, involving cancer properties, have also been reported due to their role in
the synthesis of new functional materials, and it is a powerful tool the development of newer drugs [18,19]. Nickel(II) thiosemicar-
for interesting structure formations [1–4]. The construction of bazone complexes have been reported to have cytotoxicity against
desired architectures with potential applications depends on vari- the K562 leukemic cell line [20]. A wide range of cytotoxic drugs
ous synthetic factors, such as the nature of the metal centers [5,6], with different mechanisms of action have been used as anticancer
counter ions [7,8] and the reaction conditions [9,10]. In this agents, singly or in combination with other drugs [21]. The main
respect, the suitable selection of organic moieties as ligands plays drawback of these drugs is their life threatening side effects that
a decisive role for the construction of architectures of desired are caused mainly by their indiscriminate action against both
dimensionalities [11,12]. cancerous and normal cells in the host body. Therefore, there is
Complexes involving pyrazole ligands, due to their diverse an urgent need to find new alternative drugs with enhanced anti-
structural topologies, are efficient and versatile in various fields cancer activity to fight against this disease [22].
that includes medicine, catalysis, separations, bio-mimetic Among the non-covalent interactions, hydrogen bonds are the
most common type of supramolecular interaction used by organic
ligands to control the secondary coordination sphere of metal ions
⇑ Corresponding author. [23–25]. Hydrogen bonding between closed-shell ions of like
E-mail address: manjit.bhattacharyya@cottonuniversity.ac.in (M.K.
charge where the dominant electrostatic force is strongly opposed
Bhattacharyya).

https://doi.org/10.1016/j.poly.2019.04.043
0277-5387/Ó 2019 Elsevier Ltd. All rights reserved.
114 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

is referred to as anti-electrostatic hydrogen bonding (AEHB) [26]. NO32H2O (1) and [Ni(Hdmpz)2(CH3COO)(H2O)3]Cl (2), involving
The term AEHB was introduced by Weinhold and Klein in 2014, pyrazole and acetate ligands having different counter anions
when they provided new quantum chemical evidence of hydrogen (nitrate and chloride, respectively). The complexes were character-
bonding overcoming electrostatic repulsion [27]. The possibility of ized by single crystal X-ray diffraction, electronic, vibrational spec-
‘‘anti-electrostatic” hydrogen bond (AEHB) formation between troscopy, PXRD and TGA. The counter anions and Hdmpz ligands
like-charged ions has recently been theoretically predicted and are involved in variety of non-covalent interactions resulting in
experimentally confirmed [28,29]. In addition, cation-cation and the self-assembly of the cationic units of 1 and 2 into supramolec-
anion-anion hydrogen bonded clusters can also overcome the elec- ular double chain structures. The crystal structures show
trostatic repulsion which can be influenced by counter ions or sol- supramolecular association involving anti-electrostatic hydrogen
vents [30,31]. bonds that have been analyzed using DFT and NCI plot calculations.
It has also been established that when cationic complexes are It is interesting to observe that the behaviour of the anti-electro-
used as building blocks, the counter anions play a crucial role in static hydrogen bonds in the complexes is influenced by the coun-
the determination of the supramolecular architectures [32,33]. ter anions. The analyses of cell viability results for the DL cell line
For such metal complexes with functionalized hydrogen bonded show a significant concentration dependent decrease for com-
ligands, the contribution of the selected counter ions should be pound 2 compared to compound 1 and interestingly, a negligible
expected to support the self-assembly processes. From studies on cytotoxic effect was observed against normal cells (PBMC) com-
the molecular assembly of several ionic metal complexes based pared to the cancer cells (DL).
on substituted pyrazole ligands, the hydrogen-bonding interaction
between the counter anion and the NAH-pyrazole groups are the
2. Experimental
main tool for the molecular self-assembly, although some other
factors, such as variation of the counter anion, can also drive the
2.1. Materials and physical measurements
supramolecular arrangements [34–36].
In the present work, we have synthesised (Scheme 1) two new
All reagents, viz. nickel(II) nitrate hexahydrate, nickel(II) chlo-
coordination complexes, viz. [Ni(Hdmpz)2(CH3COO)(H2O)3]
ride hexahydrate, 3,5-dimethylpyrazole and sodium acetate trihy-
drate, used in this work were obtained from Sigma Aldrich and
Merk (India) Ltd. and were used without further purification.
Deionized water was used as the reaction medium. Elemental anal-
yses (C, H and N) were carried out on a Perkin Elmer 2400 Series II
CHNS/O Analyzer. KBr phase FT-IR spectra were recorded on a Bru-
ker Alpha FT-IR spectrophotometer in the mid-IR region (4000–
400 cm1). The electronic spectra were recorded using a Shimadzu
UV-2600 spectrophotometer. For UV–Vis-NIR spectra, BaSO4 pow-
der was used as a reference (100% reflectance). Thermogravimetric
studies were carried out under a flow of N2 gas using a Mettler
Toledo TGA/DSC1 STARe system at a heating rate of 10 °C min1.
Scheme 1. Synthesis of 1 and 2. The powder X-ray diffraction (PXRD) data were recorded using

Table 1
Crystal data and structure refinement details for 1 and 2.

Parameters 1 2
Formula C12H29N5NiO10 C12H25ClN4NiO5
Formula weight 462.11 399.52
T (K) 293(2) 293(2)
k (Å) 0.71073 0.71073
Crystal system monoclinic triclinic

Space group P21/c P1
Unit cell dimensions
a (Å) 17.9772 (4) 7.3800 (6)
b (Å) 7.2977 (2) 8.6686 (7)
c (Å) 18.6249 (4) 14.5212 (13)
a (°) 90 101.359 (5)
b (°) 116.4380 (10) 92.449 (5)
c (°) 90 98.857 (5)
V (Å3) 2187.90(9) 897.36 (13)
Z 4 2
Calculated density (Mg m3) 1.403 1.479
F (0 0 0) 976 420
Crystal size (mm) 0.40  0.28  0.16 0.43  0.12  0.11
h range for data collection (°) 2.19 to 25.00 1.43 to 25.00
Limiting indices, h, k, l 21  h  21, 8  k  8, 22  l  22 8  h  8, 10  k  10, 17  l  17
Reflections collected 30 226 8748
Unique data (Rint) 3835 [Rint = 0.0315] 3057 [Rint = 0.0385]
Refinement method full-matrix least-squares on F2 Full-matrix least-squares on F2
Data/restraints/parameters 3853/0/292 3057/11/232
Goodness-of-fit on F2 1.036 1.193
Final R indices [I > 2r(I)] R1 = 0.0287, wR2 = 0.0770 R1 = 0.1146, wR2 = 0.3318
R indices (all data) R1 = 0.0354, wR2 = 0.0813 R1 = 0.1174, wR2 = 0.3327
Largest difference peak and hole (e Å3) 0.377 and 0.306 2.522 and 1.237
A. Gogoi et al. / Polyhedron 168 (2019) 113–126 115

an XPERT-PRO X-ray powder diffractometer with Cu Ka radiation. of 550 nm. The percentage cell viability was calculated from the
Room temperature magnetic susceptibility was measured at 300 K formula given below:
on a Sherwood Mark 1 magnetic susceptibility balance by the
Evans Method. OD of Sample  OD of Blank
Cell viability ð%Þ ¼  100
OD of Control
2.2. Crystallographic data collection and refinement A dose–response curve (% cell viability versus sample concen-
tration) was plotted and the sample concentration that inhibits
Intensity data of 1 and 2 were collected on a Bruker SMART CCD 50% of the cell viability (IC50) was determined.
diffractometer with graphite monochromatised Mo Ka radiation
(k = 0.71073 Å). The crystal structures were solved by the direct 2.5. Apoptosis study using fluorescence microscopy
method and refined by full matrix least squares techniques with
SHELXL-97[37] via the WINGX [38] platform available for personal com- To understand the mechanism of cell death (cytotoxicity), com-
puters. The hydrogen atoms in both the complexes were located in pounds 1 and 2 mediated apoptotic cell death was assessed using
difference Fourier maps and refined with isotropic atomic displace- the acridine orange/ethidium bromide (AO/EB) double staining
ment parameters, except for the lattice water molecules of 1. The method [47]. Acridine orange is taken up by both viable and apop-
structural diagrams were drawn with DIAMOND 3.0 [39]. Data collec- totic cells and emits green fluorescence when exposed to UV light.
tion refinement parameters for complexes 1 and 2 are summarized Ethidium bromide is taken up only by apoptotic cells and emits red
in Table 1. fluorescence. In the present study, control and treated DL cells
were collected after 24 h treatment, washed with PBS and to the
2.3. Computational methods cell suspension, 20 mL of AO/EB dye mixture (100 lg/mL of each
dye in distilled water) was added, mixed gently and incubated
The calculations of the non-covalent interactions were carried for 5 min in the dark. The cells were thoroughly examined under
out using GAUSSIAN-09 [40] and the M06-2X/def2-TZVP level of a fluorescence microscope and photographed. About 1000 cells
theory. The Grimme’s dispersion correction has been used in the were analyzed, and the percentage of apoptotic nuclei was deter-
calculations [41]. To evaluate the interactions in the solid state, mined for three independent determinations. Viable cells were
the crystallographic coordinates were used and only the position identified by bright uniform green nuclei with organized struc-
of the H-bonds has been optimized. This procedure and level of tures; apoptotic cells contain condensed or fragmented chromatin
theory has been successfully used to evaluate similar interactions with red or orange nuclei [48].
[42]. The interaction energies were computed by calculating the
difference between the energies of the isolated monomers and 2.6. Molecular docking
their assembled structures. The NCI plot is a visualization index
that efficiently allows the identification and visualization of In the present study, the molecular interactions between the
non-covalent interactions [43]. It is based on the electron density compounds and cancer target proteins were studied using Molegro
and its derivatives and the isosurfaces correspond to both Virtual Docker (MVD) software (www.molegro.com) along with
favorable and unfavorable interactions. They are easily differenti- the Graphical User Interface (GUI). The 3-dimensional (3D) coordi-
ated by the sign of the second density Hessian eigenvalue and nates of multiple cancer target proteins were downloaded from the
are defined by the isosurface color. NCI analysis is a very web for the Research Collaborator for Structural Bioinformatics
convenient tool to rationalize host–guest complementarity and to (RCSB) protein data bank (PDB). The PDB ids 4XV2 (melanoma, col-
know which interactions stabilize a complex. The color scheme orectal, thyroid and non-small cell lung cancer), 5LWE (ovarian
is a red-yellow-green-blue scale with red for q+cut (repulsive) cancer, prostate cancer, pancreatic cancer, large B cell lymphoma,
and blue for q cut (attractive). Yellow and green surfaces corre- melanoma), 4FLH (colon, brain, gastric, breast and lung cancer),
spond to weak repulsive and weak attractive interactions, 1XKK (non-small cell lung cancer, bladder cancer, breast cancer
respectively [44]. and glioblastoma) were selected based on the potential roles in
multiple cancer types. The molecular arrangement and geometry
2.4. MTT based cell viability assay and IC50 measurement of the complexes were fully optimized using a semi-empirical
quantum chemistry method (PM3). All of the water and cofactor
MTT is a largely used colorimetric assay for measuring cell pro- compounds were deleted from the protein structures and the pro-
liferations, cellular metabolic activity and cell viability [45,46]. tein and complex structures were further prepared using the
The MTT assay is based on the ability of cellular oxidoreductase parameter settings in the same software package [49]. The active
enzymes (MDH and SDH) to reduce the tetrazolium salt into insol- binding site region was defined as a spherical region which encom-
uble formazan crystals in the presence of nicotinamide adenine passes all protein within 15.0 Å of the bound crystallographic
dinucleotide phosphate (NADPH), which later produces a purple ligand atom with selected coordinates of the X, Y and Z axes,
color with DMSO. In the present study, compound 1 and 2 medi- respectively. Default settings were used for all the calculations.
ated cell viability was measured by the MTT assay in DL (cancer Docking was performed using a grid resolution of 0.30 Å and for
cells) and peripheral blood mononuclear cells (PBMC) (normal each of the 10 independent runs, a maximum number of 1500 iter-
cells) as per standard methods [46]. After incubation with differ- ations were executed on a single population of 50 individuals. The
ent doses (0, 0.01, 0.1, 0.5, 1, 5 and 10 mM) of the compounds active binding site was considered as a rigid molecule, whereas the
for 24 h, 10 mL of the MTT labelling reagent (5 mg/mL in phos- complexes were treated as being flexible, i.e. all non-ring torsions
phate-buffered saline) was added into each well, except the were allowed [50]. The protein-complex interaction was further
empty wells. The microtiter plate was then incubated for four analyzed and visualized by Chimera software (https://www.cgl.
hours in a CO2 incubator (5% CO2 and 95% air) at 37 °C. Following ucsf.edu/chimera/). Furthermore, Structure Activity Relationships
that, 100 mL of DMSO was added into each well, with gentle shak- (SARs) of compounds 1 and 2 were examined based on the docking
ing. The plate was checked for complete solubilization of the pur- results as well as bioassay results available in the NCBI database
ple formazan crystals, followed by measurement of the (https://www.ncbi.nlm.nih.gov/) for the parental template (pyra-
absorbance (optical density, OD) of the samples at a wavelength zole complex) of both compounds.
116 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

2.7. Statistical analysis 3. Results and discussion

The experimental results are expressed as mean values ± S.D. 3.1. Synthesis and general aspects
All measurements were replicated three times. The data were ana-
lyzed by an analysis of variance (ANOVA) (P  0.05). The IC50 val- Complexes 1 and 2 have been isolated in high yield by reacting
ues were calculated from dose response curve analysis. one equivalent of NiX26H2O (X = NO 
3 and Cl ) with two equiva-
lents of Hdmpz and one equivalent of the acetato ligand in water.
2.8. Syntheses Complexes 1 and 2 are soluble in water and also in common
organic solvents, viz. methanol, DMSO etc. Complexes 1 and 2
2.8.1. Synthesis of [Ni(Hdmpz)2(CH3COO)(H2O)3]NO32H2O (1) show room temperature meff values of 3.03 and 3.08 BM,
Complex 1 was prepared by adding an aqueous solution of Ni respectively.
(NO3)26H2O (0.498 g, 2 mmol) dropwise to a mixed aqueous solu-
tion of Hdmpz (0.384 g, 4 mmol) and sodium acetate trihydrate
(0.272 g, 2 mmol) in a molar ratio of 1:2:1. The resulting green 3.2. Crystal structures of complexes 1 and 2
solution was mechanically stirred for 2 hours at room temperature.
The clear solution was kept at 2–4 °C, from which block-shaped The structures of the mononuclear cations for 1 and 2 are sim-
green crystals suitable for single crystal X-ray diffraction were ilar, as shown in Fig. 1(a) and (b). In both cases the Ni(II) ions are
obtained after several days. Yield: 89% (0.82 g). Anal. Calc. for C12- coordinated to two pyrazolyl nitrogen atoms of two Hdmpz ligands
H29NiN5O10: C, 31.16; H, 6.27; N, 15.14; Found: C, 31.28; H, 6.31; N, in a cis-fashion, three water molecules and an acetate ligand. Com-
15.17%. FT-IR spectral data (KBr disc, cm1): 3278(s), 3146(w,sh), plex 1 crystallizes in the triclinic centrosymmetric space group
3042(w,sh), 2931(m,sh), 2874(sh), 1624(w,sh) 1580(s), 1470(w, P21/c, with Z = 4, while complex 2 crystallizes in the monoclinic

sh), 1381(m,sh), 1330(m,sh), 1278(m,sh), 1146(s), 1050(s), 1020 crystal system in space group P1, with Z = 2. There are two water
(s), 815(s), 697(m), 653(m,sh), 602(m) [s, strong; m, medium; w, molecules and a nitrate anion in the crystal lattice of 1, while the
weak; br, broad; sh, shoulder]. crystal lattice of 2 contains a chloride anion. The axial positions
of 1 and 2 are occupied by two coordinated water molecules and
2.8.2. Synthesis of [Ni(Hdmpz)2(CH3COO)(H2O)3]Cl (2) the equatorial plane is formed by two Hdmpz ligands, one water
Complex 2 was synthesized by a method similar to that of com- molecule and an acetate anion. The acetate anion is coordinated
plex 1, by adding an aqueous solution of NiCl26H2O (0.474 g, in a monodentate fashion in both complexes with a NiAOacetate dis-
2 mmol) dropwise to an aqueous solution of Hdmpz (0.384 g, tance of 2.065(2) Å in 1 and 2.093(9) Å in 2. The NiAOH2O bond dis-
4 mmol) and sodium acetate trihydrate (0.272 g, 1 mmol) in a tances in 1 range from 2.069(1) to 2.090(2) Å, while the NiAOH2O
molar ratio of 1:2:1. The resulting green solution was stirred distances are found to be in the range 2.057(11)–2.116(8) Å for 2.
mechanically for 2 h at room temperature. Irregular shaped crys- However, only small differences between the molecular cations
tals suitable for single crystal X-ray analysis were obtained after are observed, i.e. the dihedral angle between the pyrazole planes,
a few weeks. Yield: 87.2% (0.65 g). Anal. Calc. for C12H25NiN4O5Cl: which is 66.04(2)° for 1 and 59.95(4)° for 2. This feature could be
C, 36.04; H, 6.25; N, 14.03. Found: C, 36.08; H, 6.28; N, 14.10%. related to the different interactions of the pyrazole NAH groups
FT-IR spectral data (KBr disc, cm1): 3241(s, br), 3146(w,sh), with the corresponding to counter anions, viz. nitrate in 1 and chlo-
3109(w,sh), 3042(w,sh), 2984(w,sh), 2925(m,sh), 2874(sh), 2777 ride in 2 [51]. Table 2 lists the selected bond lengths and bond
(w,sh), 1624(w,sh), 1580(s), 1551(w,sh), 1418(s), 1338(m,sh), angles for 1 and 2. As can be seen from Table 2, the coordination
1285(s), 1258(m,sh), 1153(s), 1042(s), 1014(s), 874(m), 778(s), geometry of the Ni(II) ions in the complexes can be best described
705(m), 661(m,sh), 616(m), 529(m). as slightly distorted octahedral.

Fig. 1. Molecular structure of (a) 1 and (b) 2.


A. Gogoi et al. / Polyhedron 168 (2019) 113–126 117

Table 2 [Ni(Hdmpz)2(H2O)3(CH3COO)]+ units via a variety of hydrogen


Selected bond lengths (Å) and angles (°) of the coordination metal core in 1 and 2. bonding interactions (Table 3), thereby forming a 1D supramolec-
Bond d (Å) Angle Degree (°) ular double chain. A detailed crystal packing analysis of 1 further
1 reveals two symmetrically equivalent CAH  C contacts [52], [C
Ni1AO1 2.065(2) O1ANi1AN3 172.6 (sp3)AH  C(sp2), [CAC = 3.999(4) Å] between carbon atom C(9)
Ni1AO3 2.090(2) O5ANi1AN1 175.8 of a Hdmpz ligand of one complex cation and carbon atom C(11)
Ni1AO4 2.087(2) O3ANi1AO4 172.6 of a Hdmpz ligand of another complex cation. The CAH  C con-
Ni1AO5 2.069(1) N3ANi1AN1 93.5
Ni1AN1 2.077(1) O1ANi1AO4 81.5
tacts extend the double chain into an infinite 2D network structure
Ni1AN3 2.078(2) O3ANi1AN3 94.5 along the ac plane [Fig. 3(c)]. Remarkably, the acetato ligands in the
O3ANi1AN1 88.7 cationic [Ni(Hdmpz)2(CH3COO)(H2O)3]+ units of 1 are involved in
O3ANi1AO5 87.1 strong anti-electrostatic OAH  O hydrogen bonding interactions
O1ANi1AO5 89.5
[Fig. 3(d)]. The hydrogen bonds are formed between the uncoordi-
O1ANi1AO4 81.5
O1ANi1AO3 91.1 nated oxygen atom (O2) of the acetate ligand and the coordinated
water molecules (O4), with a donor acceptor distance of 2.794
2
Ni1AO1 2.093(9) O3ANi1AN1 176.4 (2) Å, which is further analyzed below in the theoretical study.
Ni1AO3 2.116(8) O1ANi1AN3 172.3 As shown in Fig. 3(e) this anti-electrostatic hydrogen bonding
Ni1AO4 2.057(11) O4ANi1AO5 173.1 interaction forms a 1D chain along the crystallographic b-axis,
Ni1AO5 2.058(11) O4ANi1AO3 89.7 assisted by nitrate anions. The nitrate anions in the crystal lattice
Ni1AN1 2.072(10) N3ANi1AO4 90.4
with anti-electrostatic OAH  O hydrogen bonding adds a new
Ni1AN3 2.070(12) N3ANi1AO3 89.7
N3ANi1AN1 92.7 dimension to the crystal structure of 1, which can be described
N1ANi1AO1 90.2 as a 3D supramolecular architecture, thus highlighting the crucial
N1ANi1AO5 89.2 role of the nitrate anion in the crystal packing of 1.
N1ANi1AO4 92.7
As shown in Fig. 4(a), the cationic [Ni(Hdmpz)2(CH3COO)
N3ANi1AO5 96.0
(H2O)3]+ dimeric units in 2 are stablized by the formation of two
symmetrically equivalent OAH  O anti-electrostatic hydrogen
3.3. Crystal packing bonding interactions involving the oxygen atom (O1) of the acetate
ligand and coordinated water molecule (O3). The OAH  O bond
The crystal structure analysis reveals that the nitrate anion in 1 distance was found to be 1.772 Å, which clearly indicates strong
and chloride anion in 2 interact with three proximate complex OAH  O hydrogen bonding interactions between the cationic
units via intermolecular hydrogen bonding interactions (see Fig. 2). units. The presence of the chloride anion in the crystal lattice of
On examining the role of the lattice water molecules in the 2 strengthens the formation of anti-electrostatic OAH  O hydro-
crystal packing of 1, we observed that they interacts with the gen bonding interactions, which are further analyzed below in
nitrate anion to form nitrate water clusters with a discrete [(H2- the theoretical study. The above dimeric units extend into a 1D
O)4(NO3)2]2 core which has a chair-like conformation [Fig. 3(a)]. supramolecular double chain via OAH  Cl and NAH  Cl hydrogen
As shown in Fig. 3(b), these nitrate water clusters interact with bonding interactions (Table 3) involving the chloride anion in the

Fig. 2. Supramolecular synthons formed by [Ni(Hdmpz)2(H2O)3(CH3COO)]+ with (a) nitrate and (b) chloride anions.
118 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

Fig. 3. (a) Perspective representation of a chair-like anion-water cluster with a discrete [(H2O)4(NO3)2]2 core; (b) supramolecular 1D double chain sustained by
nitratewater clusters; (c) 2D layered structure generated by CAH  C contacts in 1; (d) anti-electrostatic OAH  O hydrogen bonding interactions between [Ni(Hdmpz)2(-
CH3COO)(H2O)3]+ dimeric units in 1; (e) role of the nitrate anion in the 3D packing of 1, involving anti-electrostatic OAH  O hydrogen bonding interactions between the [Ni
(Hdmpz)2(CH3COO)(H2O)3]+ cationic moieties.

Table 3
Selected hydrogen bond distances (Å) and angles (°) for 1 and 2.

DAH  A d(DAH) d(D  A) d(H  A) —(DHA) Symmetry code

1
O3AH3OA  O8 0.954(1) 2.857(2) 1.914(2) 168.7 x, y + 1/2, +z + 1/2
O5AH5OB  O9 0.820(2) 2.665(3) 1.923(3) 149.9 x + 1, +y  1/2, z + 1/2
O4AH4OA  O2 0.789(3) 2.794(2) 2.007(3) 175.5 x, +y + 1, +z
O9AH9C  O10 0.890(4) 2.776(4) 1.902(4) 166.8 x + 1, y + 1, z
O9AH9D  O7 0.803(5) 2.803(4) 2.013(6) 167.2 x, y, z
O10AH10C  O8 0.986(5) 2.852(2) 1.882(5) 166.4 x, y, z
O10AH10D  O2 0.883(4) 2.778(2) 1.937(4) 158.6 x + 1, +y + 1/2, z + 1/2
N2AH2N  O6 0.796(3) 2.834(3) 2.090(3) 155.5 x, y, z
N4AH4N  O8 0.796(2) 3.137(3) 2.352(2) 168.3 x, y + 1/2, +z + 1/2
2
O3AH3OA  O1 0.932(16) 2.701(14) 1.771(16) 174.4 x, y, z
O4AH4D  O2 0.962(10) 2.681(13) 1.982(10) 127.7 x  1, +y, +z
O4AH4C  Cl 0.965(38) 3.634(10) 2.899(64) 133.5 x, y, z
O5AH5OB  Cl 0.931(8) 3.090(10) 2.294(4) 97.8 x, +y  1, +z
O3AH3OB  Cl 0.956(10) 3.275(12) 2.355(11) 161.2 x, +y  1, +z
N2AH2N  Cl 0.971(13) 3.205(14) 2.250(12) 167.3 x, y, z
N4AH4N  Cl 0.972(13) 3.183(13) 2.255(14) 159.2 x, +y  1, +z

crystal lattice of 2, thus again highlighting the crucial role of the held together by weak CAH  C contacts [52], resulting in an infi-
counter anion in the self-assembly process [Fig. 4(b)]. A detailed nite layered structure parallel to the crystallographic bc plane
structural analysis further reveals that the 1D double chains are [Fig. 4(c)]. CAH  C contacts [C(sp3)AH  C(sp2), [CAC = 3.867(2)
A. Gogoi et al. / Polyhedron 168 (2019) 113–126 119

Fig. 4. (a) Anti-electrostatic OAH  O hydrogen bonding interactions between cationic [Ni(Hdmpz)2(CH3COO)(H2O)3]+ units; (b) supramolecular double chain generated by
anti-electrostatic OAH  O hydrogen bonding and chloride induced OAH  Cl and NAH  Cl hydrogen bonding interactions (blue dotted lines represent AEHB and green
dotted lines represent OAH  Cl and NAH  Cl hydrogen bonding interactions); (c) 2D layered structure generated by CAH  C contacts in 2; (d) 3D supramolecular
architecture generated by OAH  O hydrogen bonding interactions in 2. (Color online.)

Table 4
Comparison of the CAH  C contacts (Å, °) observed in 1 and 2.

DAH  A d(D–H) d(D  A) d(H  A) <(DHA) Symmetry code


1
C11AH11A  C9 0.959(3) 3.999(4) 3.045(3) 172.9 x, y + 1, z
2
C1AH1A  C8 0.959(15) 3.867(20) 2.942(14) 162.3 x, y, z + 1

Å] were observed between carbon atom C(1) of the Hdmpz ligand the simulated (Mercury Software) and as-synthesized PXRD for 2
of one complex cation and carbon atom C(8) of the Hdmpz ligand (see Fig. 5) are in agreement with each other, demonstrating the
of another complex cation from an adjacent double chain. As good phase purity of the complex. Small differences in the reflec-
clearly shown in Fig. 4(c), the CAH  C bridges are symmetrically tion intensities and peak positions are observed between the sim-
equivalent, similar to that in 1. A comparison of the CAH  C con- ulated and experimental patterns, which can be attributed to the
tacts in 1 and 2 is given in Table 4. The uncoordinated oxygen atom variation in crystal orientation or particle size of the powder sam-
O(2) of the acetate ligand of one complex unit is involved in a ple [53,54]. However, in the PXRD of 1, an appreciable difference in
strong intermolecular hydrogen bonding interaction with hydro- 2h has been observed (see supplementary Fig. S1). The evacuation
gen atom H4A of the coordinated water molecule (O4) of a nearby of the weakly bounded outer sphere water molecules in the crystal
complex unit [1.982 Å and 127.7°]. This OAH  O hydrogen bond- lattice of complex 1 may contribute to the difference in the 2h val-
ing interaction involving the oxygen atom O(2) of the acetate ues of the PXRD lines [55].
ligand propagates along the crystallographic a-axis, which further
extends the 2D layer into a 3D supramolecular architecture, similar 3.5. Theoretical studies
to that of 1 [Fig. 4(d)].
The theoretical study is devoted to the analysis of the inter-
3.4. Powder X-ray diffraction (PXRD) molecular H-bonding interactions that are formed in compounds
1 and 2 between the cationic [Ni(Hdmpz)2(CH3COO)(H2O)3]+ units,
The powder X-ray diffraction (PXRD) patterns of complexes 1 together with the influence of the counter ions. The H-bonds are
and 2 were measured at room temperature. The peak positions of formed between the acetate ligands and the coordinated water
120 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

tive (DE1 = +13.4 kcal/mol) due to the Columbic repulsion between


the two moieties of the same charge. In the case where the counter
ions are included in the theoretical model [see Fig. 6(c)], the inter-
action becomes favorable (DE2 = 14.7 kcal/mol), thus highlight-
ing the crucial role of the anion. In order to further characterize
the AEHB interactions, we have used the NCI plot index computa-
tional tools. The NCI plot enables the visualization and identifica-
tion of non-covalent interactions efficiently, because it allows an
assessment of host–guest complementarity and the extent to
which weak interactions stabilize a complex. A representation of
the NCI plot is shown in Fig. 6(d). It can be observed a small and
bluish isosurface is located between the H atom of the coordinated
water molecule and the O atom of the acetate ligand, thus charac-
terizing the short H-bond, which indicates that it is energetically
favorable in spite of the overall positive interaction energy. In addi-
tion, a more extended green isosurface is located between the H
atoms of the methyl groups, thus revealing the existence of weak
Fig. 5. Powder X-ray diffraction patterns of 2: as-synthesized (red) and simulated
van der Waals interactions that further stabilize the assembly.
from MERCURY software (black). (Color online.) Fig. 7(a) shows a partial view of the X-ray of compound 2
(anions omitted) where an infinite 1D chain is represented. It is
formed by the infinite propagation of self-assembled dimers that
molecules, as detailed below. Previous theoretical studies have are stabilized by the formation of two symmetrically equivalent
suggested the possibility of finding cation–cation hydrogen OAH  O AEHBs [see blue dashed lines in Fig. 7(b)]. The interaction
bonded cluster minima in the absence of solvent [56]. The presence energy of the self-assembled dimer is attractive, even though the
of a dissociation barrier makes these minima observable, even assembly is dicationic (DE3 = 5.9 kcal/mol). This clearly indicates
though their overall binding energy is usually repulsive. Evidently, that the H-bonds are very strong since their stabilization energy is
the Coulombic energy governs an interaction that is repulsive in able to compensate the electrostatic repulsion. This agrees with the
the case of systems with the same charge. The analysis of previ- short H-bonding distance (1.76 Å). In the case where the counter
ously reported minima structures in anti-electrostatic hydrogen ions are included in the calculations [see Fig. 7(c)], the interaction
bonded systems indicates that attractive forces between the strengthens significantly (DE4 = 88.1 kcal/mol), thus highlighting
groups involved in the HB interaction are responsible for the pres- again the crucial role of the anion. A representation of the NCI plot
ence of such minima [42,57]. is shown in Fig. 7(d). Two dark blue isosurfaces between the H
Fig. 6(a) shows a partial view of the X-ray structure of atom of the coordinated water molecule and the O atom of the
compound 1 (anions omitted), where an infinite 1D chain of is acetate ligand can be observed, thus characterizing the strong H-
represented that is governed by the formation of OAH  O anti- bonds. The NCI plot agrees well with the energetic and geometric
electrostatic H-bonding interactions. The interaction energy of a analysis and confirms the strong nature of the AEHBs in this partic-
dimer extracted from this infinite 1D chain [see Fig. 6(b)] is posi- ular assembly. In addition, two green isosurfaces characterize two

Fig. 6. (a) Partial view of the X-ray structure of compound 1. H atoms and anion omitted for clarity; (b and c) theoretical models used to evaluate the AEHB interactions
(distances in Å); (d) NCI plot of the fragment of the complex. The gradient cut-off is s = 0.35 au, and the color scale is 0.04 < q < 0.04 au. (Color online.)
A. Gogoi et al. / Polyhedron 168 (2019) 113–126 121

Fig. 7. (a) Partial view of the X-ray structure of compound 2. H-atoms and anion omitted for clarity; (b and c) theoretical models used to evaluate the AEHB interactions (blue
dashed lines). Distances in Å; (d) NCI plot of the fragment of the complex. The gradient cut-off is s = 0.35 au, and the color scale is 0.04 < q < 0.04 au. (Color online.)

additional and weaker H-bonds involving the other coordinated defined band at 3278 cm1 for complex 1 and 3241 cm1 for com-
water molecule that further stabilize the assembly. plex 2 was also observed, which corresponds to m(OAH) vibrations
and is generally associated with coordinated water molecules.
Weak absorptions observed at 2900–2770 cm1 can be attributed
3.6. Spectral studies
to the m(CAH) vibration of the Hdmpz groups. The peaks at 1476,
1292 and 1146 cm1 in the complexes are attributable to pyrazole
3.6.1. FT-IR spectroscopy
ring stretching vibrations (CAN, NAN and C@N respectively) [59].
The FT-IR spectra of the prepared Ni(II) distorted octahedral
On the other hand, the peaks observed in the range 1020–1014 and
complexes are given in Fig. 8. The spectra of 1 and 2 are fully con-
778–697 cm1 for the complexes can be attributed to in plane
sistent with their formulations. The IR spectra of the complexes
deformation vibrations of the pyrazole ring [59]. Sharp peaks at
show peaks around 3146 cm1 for both complexes which are
1580 cm1 for 1 and 1572 cm1 for 2 are attributed to in plane
assigned to m(NAH) absorptions coming from the monodentate
stretching vibrations of the pyrazole ring that has been shifted to
coordination mode of the Hdmpz ligands [58]. A strong and well-
lower frequencies on coordination of the Hdmpz ligands to the
metal centres [59(c)]. The asymmetric mas(COO) stretching for
the carboxyl vibrations of the acetato ligand appear at 1624 cm1
as weak shoulder for both the complexes, while the symmetric ms(-
COO) stretching appears at 1330 and 1338 cm1 for 1 and 2
respectively. The Dm values in the complexes are larger than
200 cm1, indicating monodentate coordination of the acetato
ligand [60]. Furthermore, the presence of the nitrate ion in com-
plex 1 was confirmed by a peak at 1385 cm1, attributed to the
asymmetric mas(NAO) stretching vibrations [61].

3.6.2. Electronic spectroscopy


The UV–Vis–NIR spectra of powdered samples obtained from
crystals of 1 and 2 are presented in Fig. 9. Complex 1 show three
ligand field bands at 1112, 654 and 390 nm, while these bands
for 2 are observed at 1068, 636 and 381 nm, respectively. These
are associated with the spin allowed transitions from the 3A2g
ground term to the three excited triplet terms, viz. 3T2g(F), 3T1g(F)
Fig. 8. FT-IR spectra of 1 and 2. and 3T1g(P) [62]. The first spin-allowed transition, to the 3T2g(F)
122 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

Fig. 9. UV–Vis–NIR spectra of 1 and 2.

Fig. 11. TGA curves of 1 and 2.

state, occurs at 1112 and 1068 nm for 1 and 2 respectively. The


electronic transition, 3A2g ? 3T1g(F) gives rise to the 654 nm N2 atmosphere at a heating rate of 10 °C min1 (Fig. 11). The TG
absorption for 1 and 636 nm for 2. It is observed that former analysis of complex 1 showed a weight loss in the temperature
absorption band, corresponding to the 3A2g(F) ? 3T1g(F) transition, range 70–138 °C corresponding to the release of two lattice water
shows a shoulder which may be a consequence of the transition molecules (observed weight loss 10.7%, calculated 11.2%) [65]. A
from the 3A2g(F) to 1Eg level, gaining intensity through a configura- second weight loss in the temperature range 150–250 °C corre-
tion interaction with the 3T1g(F) level, although some investigators sponds to the loss of three coordinated water molecules, two
prefer to interpret the doublet as arising from spin-orbit coupling Hdmpz ligands [66] and CO2 from the acetate ligand [67] (observed
[63]. The peaks observed at 390 and 381 nm for 1 and 2 respec- weight loss 65%, calculated 65.7%). A similar continuous weight
tively are due to the 3A2g ? 3T1g(P) transition. The UV band origi- loss is also observed in the temperature range 80–400 °C for com-
nating from the p ? p* transition of the Hdmpz ligand was found plex 2. The loss of three coordinated water molecules takes place
at the expected position (285 nm) in both complexes [64]. between 75 and 125 °C. The experimental mass loss of 13.5% fits
The UV–Vis spectrum of 1 in water exhibits absorption bands at well with the calculated mass loss of 12.9%. The remaining mass
231 and 269 nm, while for 2 a peak at 231 nm was observed. These undergoes decomposition in the temperature range 136–390 °C,
absorption peaks are assigned as the p–p* transition [64] of the which corresponds to the loss of two Hdmpz ligands and a CO2
pyrazole group (Fig. 10). The absorption bands in the range 396– molecule from the acetate ligand (observed weight loss 55.2%, cal-
400 nm for both complexes can be assigned as 3A2g(F) ? 3T2g(F) culated 56.1%) [68].
transitions for octahedral Ni(II) ions and the bands at around
700 nm for 1 and 693 nm for 2 can be assigned to 3A2g ? 3T1g(P)
transitions. 3.8. MTT based cytotoxicity and IC50 measurements

3.7. Thermogravimetric analyses of the complexes Transition metal complexes have been largely studied by
researchers for developing new chemotherapeutic agents because
In order to examine the thermal stability of the two complexes, of their multiple biological activities, such as antioxidant, antibac-
TGA experiments were performed between 25 and 700 °C under an terial, antifungal and anticancer [69–72]. Fig. 12 shows the cell via-
bility results of both complexes; the IC50 values were calculated
from the percentage of cytotoxicity by dose–response curve fitting

Fig. 12. Cell viability results after compound 1, 2 and cisplatin (reference drug)
treatment for 24 h. A peripheral blood mononuclear cell (PBMC) was used as a
normal blood cell. Both compounds significantly decrease the cell viability in DL
Fig. 10. UV–Vis spectra of 1 and 2 in water (103 M). cells as compared to the control group.
A. Gogoi et al. / Polyhedron 168 (2019) 113–126 123

Table 5 and are presented in Table 5. The analyses of cell viability results
IC50 values of cisplatin, compounds 1 and 2 on DL and PBMC cells. obtained for both compounds in the DL cell line showed a signifi-
Sl. No. Reference drug/Compounds IC50 (lM) cant concentration dependent decrease (P  0.05) in cell viability.
DL cells PBMC Interestingly, a negligible cytotoxic effect (10–15%) was observed
(Cancer cells) (Normal cells) against normal cells (PBMC) as compared to the cancer cell (DL) for
1 Cisplatin 0.45 06.31
both complexes. Pyrazole rings, which exhibited the highest ability
2 Compound 1 8.31 41.11 to form bifunctional adducts with DNA in vitro, may be a possible
3 Compound 2 5.22 43.21 reason for inducing cytotoxicity [73]. Furthermore, the DNA repair

Fig. 13. The upper panel shows the morphological features of apoptotic and viable cells with acridine orange and ethidium bromide (AO/EB) staining. Control DL cells showed
mostly viable cells, represented by green nuclei. Cisplatin treatment (24 h) showing apoptotic features with membrane damage and nuclear fragmentation. Treatment with
compounds 1 and 2 showing appearance of cells blebbing, chromatin condensation and fragmented nuclei (under 400) within the cytoplasm of the DL cells. The lower panel
presents the percentage of apoptotic cells after treatment with the reference drug (cisplatin), 1 and 2. Data are mean ± S.D., n = 3, ANOVA, P  0.05.

Fig. 14. Docking structure of compound 1 with EGFR kinase domain (a) PI3K- gamma receptors (b) B-RAF kinase (c) and CC chemokine receptor (d) hydrogen bonds between
the ligand and the receptors are shown in blue lines.
124 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

mechanism is very poor in cancer cells [74] as compared to normal domain and PI3K- gamma receptors) that are involved in initiation,
cells, which may additionally contribute to compounds 1 and 2 progression and metastasis of several cancer types (melanoma, col-
mediating selective cytotoxicity in DL cells. Moreover, cancer cells orectal, thyroid, non-small cell lung cancer, ovarian cancer, pros-
have a higher oxidative status than normal cells and therefore suf- tate cancer, pancreatic cancer, large B cell lymphoma, brain,
fer more oxidative DNA damage [75]. gastric and glioblastoma) [81–84]. The docking scores of 1 and 2
with all the receptors under study showed comparable results with
3.9. Apoptosis study using fluorescence microscope their respective reference inhibitors (Figs. 14 and 15). Compound 1
interacts with B-RAF kinase, CC chemokine receptor, EGFR kinase
Treatment of DL cells with both complexes led to cell apoptosis domain and PI3K- gamma receptors by 4, 7, 6 and 2 hydrogen
bonds with various amino acid residues in the active site as, B-
as the mechanism of cell death, which was investigated morpho-
logically by the acridine orange/ethidium bromide dual staining RAF kinase: Phe595(1), Asp594(1), Lys483(2), CC chemokine:
Arg323(2), Asp327(1), Asp84(3), Glu322(1); EGFR kinase: Asn842
method (Fig. 13). Viable cells exhibited a uniform green fluores-
cence (acridine orange staining) whereas apoptotic cells show an (1), Lys745(1), Arg841(1), Asp855(3) and PI3K- gamma: Tyr867
(1), ASP964(1), whereas, compound 2 interacts with B-RAF kinase,
orange-red nuclear fluorescence (ethidium bromide staining) by
intercalation of ethidium bromide into damaged DNA in the apop- CC chemokine receptor, EGFR kinase domain and PI3K- gamma
totic cells. Cells treated with the compounds for 24 h demonstrated receptors by 6, 8, 5 and 4 hydrogen bonds with various amino acid
the morphological characteristics of apoptosis, such as cell bleb- residues, such as, B-RAF kinase: Asp594(1), Lys483(2), Phe595(1),
bing, irregular chromatin structure, followed by condensation
and the appearance of several apoptotic bodies near the cell mem-
brane. Moreover, results from several studies suggest that the
majority of metal-pyrazole complexes possess anticancer activities
due to formation of stable DNA adducts [76–78]. Pyrazoloacridine
was identified as a DNA topoisomerase inhibitor, induced apopto-
sis in myeloma and leukemia cells, and displayed pre-clinical activ-
ity in myeloma and leukemia cells [79]. We hypothesized that the
Ni(II) complexes of the present study may exert apoptosis in DL
cells through a similar pathway.

3.10. Molecular docking study

To gain more insight into the biological mode of action of 1 and


2, molecular docking studies were carried out with the common
isostructural cationic complex moiety of the compounds. Compu-
tational tools, such as molecular docking simulation and mod-
elling, have been widely used for drug development and the Fig. 16. Docking scores of compounds 1 and 2 with B-RAF kinase, CC chemokine
discovery of multi-targeted inhibitors against many overexpressed receptor, EGFR kinase domain and PI3K- gamma receptors. The reference inhibitors
for B-RAF kinase, CC chemokine receptor, EGFR kinase domain and PI3K- gamma
cancer target proteins [80]. The studies reveal that complexes 1
receptors are Dabrafenib, vercirnon, quinazoline and AMG-511 respectively. As per
and 2 possess high binding affinities against most common cancer the MVD docking score algorithm, the lower the score, the better is the interaction.
target proteins (B-RAF kinase, CC chemokine receptor, EGFR kinase Data are mean ± S.D., n = 3, ANOVA, P  0.05.

Fig. 15. Docking structure of compound 2 with EGFR kinase domain (a) PI3K- gamma receptors (b) B-RAF kinase (c) and CC chemokine receptor (d) hydrogen bonds between
the ligand and the receptors are shown in blue lines.
A. Gogoi et al. / Polyhedron 168 (2019) 113–126 125

Table 6
Docking results of 1 and 2 with B-RAF kinase, CC chemokine receptor, EGFR kinase domain and PI3K- gamma receptors. Amino acids in bold show the common amino acids
involved in the molecular interactions between the reference inhibitors and the compounds under study; the number in brackets indicates the number of H-bonds.

Sl Nos. Receptors Reference ligands/Complexes No. of H-bond H-bond scores Interacting receptors amino acids in active site
1 B-RAF kinase Dabrafenib 4 2.55 Cys 532(2), Phe 595(1), Lys 483(1)
(4xv2) (Reference ligand)
Compound 1 4 2.51 Phe595(1), Asp594(1), Lys483(2)
Compound 2 6 2.72 Asp594(1), Lys483(2), Phe595(1), Asp594(2)
2 CC chemokine Vercirnon 6 1.91 Phe 324(1), Arg 323(1), Thr 81(1), Asp 84(1), Arg 78(1), Thr 81(1)
(5lwe) (Reference ligand)
Compound 1 7 1.99 Arg323(2), Asp327(1),
Asp84(3), Glu322(1)
Compound 2 8 2.91 Arg323(2), Arg78(1), Asp84(3), Asp327(1), Glu322(1)
3 PI3K- gamma AMG-511 5 2.50 Lys 833(1), Lys 802(1), Ala 805(1), Val 882(2)
(4flh) (Reference ligand)
Compound 1 2 1.81 Tyr867(1), ASP964(1)
Compound 2 4 2.41 ASP964(1), Lys833(2), Ser806(1)
4 EGFR kinase domain Quinazoline 3 2.50 Met 793(1), Thr 854(1), Asp 855(1)
(1xkk) (Reference ligand)
Compound 1 6 2.80 Asn842(1), Lys745(1), Arg841(1), Asp855(3)
Compound 2 5 2.73 Lys745(1), Asp855(3), Arg841(1)

The complexes were characterized by single crystal X-ray diffrac-


tion, electronic, vibrational spectroscopy, PXRD and TGA. The acet-
ato ligands in the isostructural cationic moiety of both the
complexes are involved in strong anti-electrostatic hydrogen
bonding interactions that have been studied using DFT calculations
and NCI plot index analysis. The interaction energy of the cationic
moieties, extracted from the infinite 1D chain of 1, is positive and,
interestingly, the interaction becomes favorable in the presence of
the nitrate ion, thus highlighting the crucial role of the anion.
Remarkably, in the case of compound 2, these AEHBs are able to
compensate the electrostatic repulsion between the cationic units.
The anticancer activity of both complexes against DL cells showed
significant cytotoxicity, mostly contributed by apoptotic cell death.
Analyses of cell viability results for the DL cell line show a signifi-
cant concentration dependent decrease by the compounds with a
negligible cytotoxic effect against normal cells (PBMC) in compar-
ison to the cancer cells (DL). Docking based SAR analysis reveals
that the oxygen and nitrogen atoms in the isostructural cationic
complex unit of both complexes are involved in the interactions
Fig. 17. SAR analysis of the isostructural cationic complex moiety of 1 and 2 with the cancer target proteins. Cytotoxic and apoptotic effects of
involving key atoms during biological activities.
these compounds in DL malignant cancer cells indicated that both
compounds could be good candidates for further pharmacological
Asp594(2); CC chemokine: Arg323(2), Arg78(1), Asp84(3), Asp327 studies to develop effective anticancer agents in the future.
(1), Glu322(1); EGFR kinase: Lys745(1), Asp855(3), Arg841(1) and
PI3K- gamma: ASP964(1), Lys833(2), Ser806(1). The number in the
Acknowledgements
brackets indicates the number of H-bonds with the respective
amino acid (Fig. 16). The different interaction behaviors of 1 and
The authors acknowledge the financial support from University
2 with B-RAF (Table 6) are due to differences in the structural dis-
Grants Commission (UGC), New Delhi [Grant number: F. No. 42-
tortion and orientation of the cationic moieties of the complexes
377/2013]. Financial support from the MINECO of Spain (project
[85]. The SAR analysis (Fig. 17) for common structural parts of both
CTQ2017-85821-R FEDER funds) is gratefully acknowledged. The
compounds enables understanding about the specific active atomic
authors thank the Department of Chemistry, IIT, Guwahati for pro-
sites in the molecular structures that result in the biological activ-
viding the single crystal X-ray diffraction data. The authors are also
ities [86]. From the docking based SAR analyses of the isostructural
thankful to the CSIR-NEIST, Jorhat for providing the PXRD data and
cationic units of 1 and 2, it has been observed that the oxygen and
the Department of Chemistry, Gauhati University for TGA data.
nitrogen atoms with atom labels O1, O2, O3, O4, O5, N1, N2, N3 and
N4 take part in the interactions with B-RAF kinase, CC chemokine
receptor, EGFR kinase domain and PI3K- gamma receptors. These
Appendix A. Supplementary data
results provide a specific molecular level understanding of the pos-
sible biological activities of the reported complexes.
CCDC 1838104 and 1868598 contains the supplementary crys-
tallographic data for 1 and 2 respectively. These data can be
4. Conclusion obtained free of charge via http://www.ccdc.cam.ac.uk/conts/re-
trieving.html, or from the Cambridge Crystallographic Data Centre,
Two new Ni(II) coordination complexes involving pyrazole and 12 Union Road, Cambridge CB2 1EZ, UK; fax: (+44) 1223-336-033;
acetato ligands with different counter anions have been reported. or e-mail: deposit@ccdc.cam.ac.uk.
126 A. Gogoi et al. / Polyhedron 168 (2019) 113–126

Supplementary data to this article can be found online at [41] S. Grimme, J. Antony, S. Ehrlich, H. Krieg, J. Chem. Phys. 132 (2010) 154104.
[42] I. Alkorta, I. Mata, E. Molins, E. Espinosa, Chem. Eur. J. 22 (2016) 9226.
https://doi.org/10.1016/j.poly.2019.04.043.
[43] J. Contreras-García, E.R. Johnson, S. Keinan, R. Chaudret, J.P. Piquemal, D.N.
Beratan, W. Yang, J. Chem. Theory Comput. 7 (2011) 625.
References [44] E.R. Johnson, S. Keinan, P. Mori-Sanchez, J. Contreras-Garcia, A.J. Cohen, W.
Yang, J. Am. Chem. Soc. 132 (2010) 6498.
[1] G.R. Desiraju, Nature 412 (2001) 397. [45] T. Mosmann, J. Immunol. Methods 65 (1983) 55.
[2] C.B. Aakeroy, N.R. Champness, C. Janiak, CrystEngComm. 12 (2010) 22. [46] (a) A.K. Verma, S.B. Prasad, J. Ethnopharmacol. 148 (2013) 869;
[3] H.J. Schneider, Angew. Chem. Int. Ed. 48 (2009) 3924. (b) T. Mosmann, Immunol. Methods 65 (1983) 55.
[4] H. Eshtiagh-Hosseini, M. Mirzaei, M. Biabani, V. Lippolis, M. Chahkandi, C. [47] M.K. Squier, J.J. Cohen, Mol. Biotechnol. 19 (2001) 305.
Bazzicalupi, CrystEngComm. 15 (2013) 6752. [48] A.K. Verma, S.B. Prasad, Anti-Cancer Agents Med. Chem. 13 (2013) 1096.
[5] J.F. Ma, J.F. Liu, X. Yan, H.Q. Jia, Y.H. Lin, J. Chem. Soc., Dalton Trans. (2000) [49] A. Swargiary, A.K. Verma, K. Sarma, Med. Chem. Res. 22 (2013) 2870.
2403. [50] R. Thomsen, M.H. Christensen, J. Med. Chem. 49 (2006) 3315.
[6] C.B. Aakeröy, N.R. Champness, C. Janiak, CrystEngComm. 12 (2010) 22. [51] M. Cano, J.V. Herasa, M. Luz Gallego, J. Perles, C. Ruiz-Valero, E. Pinillaa, M.
[7] M. Mirzaei, H. Eshtiagh-Hosseini, Z. Bolouri, Z. Rahmati, A. Esmaeilzadeh, A. Rosario Torres, Helv. Chim. Acta 86 (2003) 3194.
Hassanpoor, A. Bauza, P. Ballester, M. Barceló-Oliver, J.T. Mague, B. Notash, A. [52] (a) S. Chakravorty, J.A. Platts, B.K. Das, Dalton Trans. 40 (2011) 11605;
Frontera, Cryst. Growth Des. 15 (2015) 1351. (b) S.M.N. Islam, D. Dutta, A.K. Guha, M.K. Bhattacharyya, J. Mol. Struct. 1175
[8] L. Carlucci, G. Ciani, D.M. Proserpio, S. Rizzato, Chem. Eur. J. 5 (1999) 237. (2019) 130.
[9] H.R. Khavasi, B.M.M. Sadegh, Inorg. Chem. 49 (2010) 5356. [53] P. Ghosh, A. Roychowdhury, M. Corbella, A. Bhaumik, P. Mitra, S.M. Mobin, A.
[10] O.S. Jung, S.H. Park, K.M. Kim, H.G. Jang, Inorg. Chem. 37 (1998) 5781. Mukherjee, S. Basu, P. Banerjee, Dalton Trans. 43 (2014) 13500.
[11] P. Ovejero, M.J. Mayoral, M. Cano, J.A. Campo, J.V. Heras, E. Pinilla, M.R. Torres, [54] (a) L. Croitor, D. Chisca, E.B. Coropceanu, G.F. Volodina, O. Petuhov, M.S. Fonari,
J. Organomet. Chem. 692 (2007) 4093. J. Mol. Struct. 1137 (2017) 136;
[12] (a) B.H. Ye, M.L. Tong, X.M. Chen, Coord. Chem. Rev. 249 (2005) 545; (b) D. Dutta, S.M.N. Islam, U. Saha, S. Chetry, A.K. Guha, M.K. Bhattacharyy, J.
(b) D. Dutta, H. Nath, A. Frontera, M.K. Bhattacharyya, Inorg. Chim. Acta 487 Chem. Crystallogr. 48 (2019) 156.
(2019) 354. [55] (a) D. Dutta, S. Chetry, A. Gogoi, B. Choudhury, A.K. Guha, M.K. Bhattacharyya,
[13] M.V. Lunagariya, K.P. Thakor, R.R. Varma, B.N. Waghela, C. Pathak, M.N. Patel, Polyhedron 151 (2018) 381;
MedChemComm 9 (2018) 282. (b) R. Bala, A. Kaur, M. Kashyap, D. Janzen, J. Mol. Struct. 1063 (2014) 203.
[14] K. Karrouchi, E.B. Yousfi, N.K. Sebbar, Y. Ramli, J. Taoufik, Y. Ouzidan, M. Ansar, [56] F. Weinhold, Angew. Chem. Int. Ed. 56 (2017) 14577.
Y.N. Mabkhot, H.A. Ghabbour, S. Radi, Int. J. Mol. Sci. 18 (2017) 2215. [57] G. Frenking, G.F. Caramori, Angew. Chem. Int. Ed. 54 (2015) 2596.
[15] J.R. Eedijk, Heterocyclic nitrogen-donor ligands, Pergamon, Oxford, 1987, p. 73. [58] J. Li, Y.H. Xing, H.Y. Zhao, Z.P. Li, C.G. Wang, X.Q. Zeng, M.F. Ge, S.Y. Niu, Inorg.
[16] S. Abdolmaleki, M. Ghadermazi, A. Fattahi, S. Shokraii, M. Alimoradi, B. Chim. Acta 362 (2009) 2788.
Shahbazi, A.R.J. Azar, J. Coord. Chem. 70 (2017) 1406. [59] (a) A. Direm, M. Tursun, C. Parlak, N. Benali-Cherif, J. Mol. Struct. 1093 (2015)
[17] D. Ajo, A. Bencini, F. Mani, Inorg. Chem. 27 (1988) 2437. 208;
[18] T. Harit, F. Malek, B. El Bali, A. Khan, K. Dalvandi, B.P. Marasini, S. Noreen, R. (b) N. Sundaraganesan, E. Kavitha, S. Sebastian, J.P. Cornard, M. Martel,
Malik, S. Khan, M.I. Choudhary, Med. Chem. Res. 21 (2012) 2772. Spectrochim. Acta, Part A: Mol. Biomol. Spectrosc. 74 (2009) 788;
[19] A. Chauhan, P.K. Sharma, N. Kaushik, Int. J. Chem. Technol. Res. 3 (2011) 11. (c) M.K. Ehlert, S.J. Rettig, A. Storr, R.C. Thompson, J. Trotter, Can. J. Chem. 68
[20] H. Yu, W. Zhang, Q. Yu, F.P. Huang, H.D. Bian, H. Liang, Molecules 22 (2017) (1990) 1494.
1772. [60] G.B. Deacon, R.J. Phillips, Coord. Chem. Rev. 33 (1980) 227.
[21] P. Kumar, B. Narasimhan, K. Ramasamy, V. Mani, R.K. Mishra, A.B.A. Majeed, [61] P.F. Barron, J.C. Dyason, P.C. Healy, J. Chem. Soc., Dalton Trans. (1986) 1965.
Curr. Top. Med. Chem. 15 (2015) 1050. [62] A.B.P. Lever, Inorganic Electronic Spectroscopy, 557, Elsevier, New York, 1984.
[22] H. Bahron, S.S. Khaidir, A.M. Tajuddin, K. Ramasamy, B.M. Yamin, Polyhedron [63] B.N. Figgis, M.A. Hitchman, Ligand Field Theory and Its Applications, 209,
161 (2019) 84. Wiley-VCH, New York, 2000.
[23] S.A. Cook, A.S. Borovik, Acc. Chem. Res. 48 (2015) 2407. [64] P. Ovejero, M.J. Mayoral, M. Cano, M.C. Lagunas, J. Organomet. Chem. 692
[24] H.W. Roesky, M. Andruh, Coord. Chem. Rev. 236 (2003) 91. (2007) 1690.
[25] J. Reedijk, Chem. Soc. Rev. 42 (2013) 1776. [65] Y. Ma, B. Mu, R.D. Huang, Transit. Met. Chem. 43 (2018) 103.
[26] F. Weinhold, R.A. Klein, Angew. Chem. Int. Ed. 53 (2014) 1. [66] A. Gogoi, S.M.N. Islam, A. Frontera, M.K. Bhattacharyya, Inorg. Chim. Acta 484
[27] F. Weinhold, R.A. Klein, Angew. Chem. 126 (2014) 11396. (2019) 133.
[28] (a) E.M. Fatila, E.B. Twum, A. Sengupta, M. Pink, J.A. Karty, K. Raghavachari, A. [67] S.J. Bora, B.K. Das, J. Solid State Chem. 192 (2012) 93.
H. Flood, Angew. Chem., Int. Ed. 55 (2016) 14057; [68] S. Demir, G.K. Kantar, Y. Topcu, Q. Li, Transit. Met. Chem. 37 (2012) 257.
(b) W. Zhao, B. Qiao, C.H. Chen, A.H. Flood, Angew. Chem., Int. Ed. 56 (2017) [69] A.K. Yudin, Chem. Sci. 6 (2015) 30.
13083; [70] P. Rathi, D.P. Singh, Spectrochim. Acta, Part A: Mol. Biomol. Spectrosc. 136
(c) E.M. Fatila, E.B. Twum, J.A. Karty, A.H. Flood, Chem. Eur. J. 23 (2017) 10652. (2015) 381.
[29] D. Mungalpara, A. Valkonen, K. Rissanen, S. Kubik, Chem. Sci. 8 (2017) 6005. [71] R.A. Shiekh, I.A. Rahman, M.A. Malik, N. Luddin, S.A.M. Masudi, S.A. Al-Thabaiti,
[30] (a) I. Mata, I. Alkorta, E. Molins, E. Espinosa, Chem. Phys. Lett. 555 (2013) 106; Int. J. Electrochem. Sci. 8 (2013) 6972.
(b) I. Mata, E. Molins, I. Alkorta, E. Espinosa, J. Phys. Chem. A 119 (2015) 183. [72] P. Arthi, A. Haleel, P. Srinivasan, D. Prabhu, C. Arulvasu, A.K. Rahiman,
[31] A. Shokri, M. Ramezani, A. Fattahi, S.R. Kass, J. Phys. Chem. A 117 (2013) 9252. Spectrochim. Acta, Part A: Mol. Biomol. Spectrosc. 129 (2014) 400.
[32] M. Munakata, M. Wen, Y. Suenaga, T. Kuroda-Sowa, M. Maekawa, M. Anahata, [73] E. Ciesielska, A. Szulawska, K. Studzian, J. Ochocki, K. Malinowska, K. Kik, L.
Polyhedron 20 (2001) 2037. Szmigiero, J. Inorg. Biochem. 100 (2006) 1579.
[33] M. El-Massaoudi, S. Radi, Y.N. Mabkhot, S.S. Al-Showiman, H.A. Ghabbour, M. [74] R.S. Day, C.H. Ziolkowski, D.A. Scudiero, S.A. Meyer, A.S. Lubiniecki, A.J. Girardi,
Ferbinteanu, N.N. Adarsh, Y. Garcia, Inorg. Chim. Acta 482 (2018) 411. S.M. Galloway, G.D. Bynum, Nature 288 (1980) 724.
[34] S. Bhattacharya, A. Goswami, B. Gole, S. Ganguly, S. Bala, S. Sengupta, S. Khanra, [75] T. Helleday, N. Engl, J. Med. 373 (2015) 1570.
R. Mondal, Cryst. Growth Des. 14 (2014) 2853. [76] N. Ribeiro, S. Roy, N. Butenko, I. Cavaco, T. Pinheiro, I. Alho, F. Marques, F.
[35] S.W. Jin, X.H. Ye, L. Jin, L. Zheng, J.W. Li, B.P. Jin, D.Q. Wang, Polyhedron 81 Avecilla, J.C. Pessoa, I. Correia, J. Inorg. Biochem. 174 (2017) 63.
(2014) 382. [77] S. Thota, S. Vallala, R. Yerra, D.A. Rodrigues, N.M. Raghavendra, E.J. Barreiro, Int.
[36] (a) M. Guerrero, J. Pons, M. Font-Bardia, T. Calvet, J. Ros, Polyhedron 29 (2010) J. Biol. Macromol. 82 (2016) 663.
1083; [78] J. Quirante, D. Ruiz, A. Gonzalez, C. López, M. Cascante, R. Cortés, R. Messeguer,
(b) R. Mondal, T. Basu, D. Sadhukhan, T. Chattopadhyay, M.K. Bhunia, Cryst. C. Calvis, L. Baldomà, A. Pascual, J. Inorg. Biochem. 105 (2011) 1720.
Growth Des. 9 (2009) 1095. [79] D. Atamanyuk, B. Zimenkovsky, R. Lesyk, J. Sulfur Chem. 29 (2008) 151.
[37] G.M. Sheldrick, Acta Crystallogr., Sect: A 64 (2008) 11. [80] H. El Hamdani, M. Amane, B.B. Mohammed, K. Yamni, J. Mol. Struct. 1181
[38] L.J. Farrugia, J. Appl. Crystallogr. 32 (1999) 837. (2019) 627.
[39] K. Brandenburg, Diamond 3.1f, Crystal Impact GbR, Bonn, Germany, 2008. [81] N. Li, D. Batt, M. Warmuth, Curr. Opin. Invest. Drugs (London, England) 8
[40] M.J. Frisch, G.W. Trucks, H.B. Schlegel, G.E. Scuseria, M.A. Robb, J.R. Cheeseman, (2007) (2000) 452.
G. Scalmani, V. Barone, G.A. Petersson, H. Nakatsuji, X. Li, M. Caricato, A. [82] Z. Tu, R. Xiao, J. Xiong, K.M. Tembo, X. Deng, M. Xiong, P. Liu, M. Wang, Q.
Marenich, J. Bloino, B.G. Janesko, R. Gomperts, B. Mennucci, H.P. Hratchian, J.V. Zhang, J. Hematol. Oncol. 9 (2016) 10.
Ortiz, A.F. Izmaylov, J.L. Sonnenberg, D. Williams-Young, F. Ding, F. Lipparini, F. [83] J.N. Gallant, J.H. Sheehan, T.M. Shaver, M. Bailey, D. Lipson, R. Chandramohan,
Egidi, J. Goings, B. Peng, A. Petrone, T. Henderson, D. Ranasinghe, V.G. M.R. Brewer, S.J. York, M.G. Kris, J.A. Pietenpol, M. Ladanyi, V.A. Miller, S.M. Ali,
Zakrzewski, J. Gao, N. Rega, G. Zheng, W. Liang, M. Hada, M. Ehara, K. J. Meiler, C.M. Lovly, Cancer Discov. (2015) CD-15-0654.
Toyota, R. Fukuda, J. Hasegawa, M. Ishida, T. Nakajima, Y. Honda, O. Kitao, H. [84] J.V. Fresno, E. Casado, P. Cejas, C. Belda-Iniesta, M. González-Barón, Cancer
Nakai, T. Vreven, K. Throssell, J.A. Montgomery Jr., J.E. Peralta, F. Ogliaro, M. Treat. Rev. 30 (2004) 193.
Bearpark, J.J. Heyd, E. Brothers, K.N. Kudin, V.N. Staroverov, T. Keith, R. [85] P. Krishnamoorthy, P. Sathyadevi, R.R. Butorac, A.H. Cowley, N.S.P. Bhuvanesh,
Kobayashi, J. Normand, K. Raghavachari, A. Rendell, J.C. Burant, S.S. Iyengar, J. N. Dharmaraj, Dalton Trans. 41 (2012) 4423.
Tomasi, M. Cossi, J.M. Millam, M. Klene, C. Adamo, R. Cammi, J.W. Ochterski, R. [86] M.A. Peterson, M. Oliveira, M.A. Christiansen, C.E. Cutler, Bioorg. Med. Chem.
L. Martin, K. Morokuma, O. Farkas, J.B. Foresman, D.J. Fox, Gaussian 09, Lett. 19 (2009) 6775.
Revision C.02, Gaussian, Inc., Wallingford CT, 2016.

You might also like