ArticlePDF Available

A Co(III) Complex of 1-Amino-4-hydroxy-9,10-anthraquinone Exhibits Apoptotic Action against MCF-7 Human Breast Cancer Cells

Authors:

Abstract and Figures

A Co(III) complex of 1-amino-4-hydroxy-9,10-anthraquinone (QH) (Scheme-1) having the molecular formula CoQ3 (Scheme-2) was prepared and characterized by elemental analysis, FTIR spectroscopy, UV–vis spectroscopy, fluorescence spectroscopy, and mass spectrometry. In the absence of a single crystal, the energy-optimized molecular structure of CoQ3 was determined by employing computational methods that was validated using spectroscopic evidences, elemental analysis, and mass spectrometry data. The electrochemical properties of the complex were analyzed using cyclic voltammetry and indicate a substantial modification of the electrochemical properties of the parent amino-hydroxy-9,10-anthraquinone. CoQ3 was thereafter tested on MCF-7 human breast cancer cells. The IC50 value for a 24 h incubation was found to be (95 ± 0.05) μg/mL. The study showed that such cancer cells underwent both early and late apoptosis following the interaction with CoQ3.
Content may be subject to copyright.
A Co(III) Complex of 1Amino-4-hydroxy-9,10-anthraquinone
Exhibits Apoptotic Action against MCF7 Human Breast Cancer Cells
Somenath Banerjee, Sanjay Roy, Dhanasekaran Dharumadurai, Balaji Perumalsamy,
Ramasamy Thirumurugan, Saurabh Das,*Asoke Prasun Chattopadhyay,*and Partha Sarathi Guin*
Cite This: https://doi.org/10.1021/acsomega.1c06125
Read Online
ACCESS Metrics & More Article Recommendations *
sıSupporting Information
ABSTRACT: A Co(III) complex of 1-amino-4-hydroxy-9,10-anthraquinone (QH) (Scheme-1) having the molecular formula CoQ3
(Scheme-2) was prepared and characterized by elemental analysis, FTIR spectroscopy, UVvis spectroscopy, uorescence
spectroscopy, and mass spectrometry. In the absence of a single crystal, the energy-optimized molecular structure of CoQ3was
determined by employing computational methods that was validated using spectroscopic evidences, elemental analysis, and mass
spectrometry data. The electrochemical properties of the complex were analyzed using cyclic voltammetry and indicate a substantial
modication of the electrochemical properties of the parent amino-hydroxy-9,10-anthraquinone. CoQ3was thereafter tested on
MCF-7 human breast cancer cells. The IC50 value for a 24 h incubation was found to be (95 ±0.05) μg/mL. The study showed that
such cancer cells underwent both early and late apoptosis following the interaction with CoQ3.
1. INTRODUCTION
Anthracycline drugs are anticancer agents used in treating
dierent forms of human carcinoma.
14
Although they enjoy
wide acceptance in chemotherapy, their use is often questioned
for the associated cardiotoxicity and high cost involved,
particularly for people from economically weaker sections of
the society. Hence, there is an eort worldwide
519
to nd
alternative cheaper analogues that are less cardiotoxic.
510
These are either derivatives of anthracyclines that are less
costly or their simpler analogues.
1120
The limitation due to acute and chronic toxicity,
2125
of
which cardiotoxicity receives the maximum attention, is the
most disturbing regarding the use of anthracyclines or their
derivatives and analogues as anticancer agents.
2631
Participat-
ing in reactions of the respiratory chain, they produce
semiquinone radical anions and related intermediates by one-
electron reduction of the quinone. Although a pre-requisite for
chemotherapeutic ecacy, such generation is also responsible
for cardiotoxicity.
2630
Semiquinone upon reaction with O2
generates superoxide radical anion (O2
) that in turn produce
H2O2/OH.
20,3033
These species participate in a wide range
of redox reactions as in oxidative phosphorylation, complex
formation with phospholipid, and in lipid peroxidation.
3032
Previous research on the subject suggests that complex
formation of these drugs with dierent metal ions leads to
decreased toxicity, the magnitude of which depends on the
metal ion. Those metal ions having a stable lower oxidation
state were found to cause maximum decrease in O2
formation
in an assay where NADH was the electron donor and
cytochrome c was the electron acceptor. Hence, studies related
to metal complexes gained a lot of importance regarding this
matter.
7,12,14,31,32
Metal complexes stabilize the semiquinone
radical anion formed. Hence, superoxide formation due to a
reaction between a semiquinone radical anion and molecular
oxygen is either inhibited or decreased drastically. It is
Received: November 1, 2021
Accepted: December 22, 2021
Article
http://pubs.acs.org/journal/acsodf
© XXXX The Authors. Published by
American Chemical Society A
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
Downloaded via 64.137.13.64 on December 29, 2021 at 12:22:17 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
therefore imperative to study such metal complexes,
particularly with regard to their electrochemical behavior
under dierent experimental conditions.
It is worth mentioning that although several metal
complexes of adriamycin, daunorubicin, mitoxantrone, and
their analogues with Fe(III), Al(III), Cu(II), Ni(II), Pd(II),
and Tb(III) were prepared and characterized,
7,1014,3346
comprehensive knowledge on structures of these metal
complexes is lacking due to inherent diculties in obtaining
single crystals for X-ray diraction studies. Single-crystal X-ray
diraction structures of only a few hydroxy-9,10-anthraquinone
complexes are reported.
44,47,48
In this study also, dierent
methods were employed to obtain single crystals of CoQ3
taking dierent compositions of solvents. However, all eorts
in getting an appropriate single crystal for CoQ3failed. The
planarity of the anthraquinone unit in these complexes could
possibly be a hindrance in getting single crystals.
7
For this
reason, we made an eort to characterize CoQ3theoretically
using density functional theory (DFT) based on experimental
data we obtained such as elemental analysis, IR spectroscopy,
mass spectrometry, powder X-ray diraction, molecular
spectroscopy, and electrochemistry. DFT is helpful in
generating the energy optimized structure, and various
essential parameters of the complex may also be obtained
from this study. The thus prepared complex was tested on
MCF-7 human breast cancer cells to see whether it initiates
apoptosis and thus could be considered as a less costly
alternative to anthracyclines already in use.
2. RESULTS AND DISCUSSION
2.1. Analysis of the Mass Spectra of CoQ3.Assuming
that the formula of the complex is CoQ3(Scheme 2), an
analysis of its mass spectrum (Figure S1, SI) was attempted.
The molecular ion peak or that of the protonated molecular
ion expected at m/z = 773.62 was not found. However, a clear
signal at m/z = 689.46 corresponds to a fragment remaining of
the complex following loss of a carbon-bound NH2group
from each ligand (a loss of 28 mass units from each ligand, i.e.,
78 mass units from the complex) to result in a peak
theoretically expected at m/z = 689.62. From this peak, loss
of two quinone oxygens would result in a peak theoretically
expected at m/z = 661.62. The experimental value is 661.45,
which tallies with the expected value. Loss of four quinone
oxygens from the rst fragment results in a peak theoretically
expected at m/z = 633.62 and experimentally found at 633.42.
Here also, the agreement is close. Similarly, loss of six quinone
oxygens from all the three ligands of the rst fragment should
result in a theoretical peak at m/z = 605.62. This was
experimentally observed at 605.39, again pointing to a close
agreement. At this stage of fragment formation in mass
analysis, the metal center is bound to three ligands via the
three phenolic OH groups on each of them. The peaks
identied above therefore categorically indicate the formation
of a 1:3 complex. Subsequent to the fragmentations mentioned
above, further loss of two carbon atoms and a few hydrogens at
a time explains peaks at an m/zvalue of 577.35 and also the
cluster of peaks at m/z values of 533.99, 532.99, and 531.98,
respectively. Peaks at lower m/z values correspond to smaller
fragments. Therefore, from an analysis of the mass spectrum of
the cobalt complex, it may be concluded that the complex has
the formula CoQ3as shown in Scheme 2.
2.2. Analysis of the IR Spectra of CoQ3.The FTIR
spectrum for QH (Figure S2, SI) shows a peak at 3431 cm1,
which is due to NH bond stretching, while that at 3300 cm1
is due to stretching of OH bonds.
6
The OH stretching is
modied signicantly in the complex (Figure S3,SI),
indicating an involvement of the OH group during complex
formation. Since there is deprotonation of OH during
complex formation, the molecule ceases to show intra-
molecular hydrogen bonding identied in QH. Peaks in this
region do not disappear completely in the complex when
compared with QH, indicating the presence of free NH2on
each ligand (just as that observed or the IR spectrum of QH).
In the IR spectrum of CoQ3(Figure S3, SI), peaks at 1625,
1586, and 1537 cm1are attributed to stretching due to free
carbonyl and CC or a combination of both, respectively. In
an earlier study,
6
we showed that peaks obtained in the region
14641031 cm1in the IR spectrum of the ligand (QH) may
be attributed to combinations of OH, NH, and CH
bending modes. Natures of the peaks in this region are
somewhat dierent in the complex. More specically, the peak
at 1121 cm1is reduced signicantly, probably due to binding
of oxygen of the OH group to Co(III) following its
deprotonation.
2.3. Powder X-ray Diraction of CoQ3.The powder X-
ray diraction (PXRD) pattern of CoQ3is shown in Figure 1.
All peaks can be indexed with the space group R32(155), and
Cu Kα= 1.5406 Å using the WINPLOTR program. Rened
cell parameters were found to be a= 7.45 Å, b= 6.52 Å, and c
= 27.8 Å. The unit cell volume was 1352 Å3;α= 33.43°,β=
90°, and γ=90°. Thus, PXRD analysis provides information
about the dimension of the unit cell of a crystalline CoQ3.
2.4. Structure of CoQ3from Density Functional
Theory Methods. The energy-optimized molecular structure
of CoQ3is shown in Figure 2, and structural parameters are
summarized in Table S1 (SI). Figure 2 shows three QH
molecules coordinated to Co(III) through phenolic Oand
quinone oxygen, forming CoQ3.
Scheme 1. 1-Amino-4-hydroxy-9,10-anthraquinone (QH)
Scheme 2. Structure of CoQ3
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
B
The energy level diagrams of QH and CoQ3are shown in
Figure S3 (SI). The HOMO (H) and LUMO (L) are
indicated in each case (Figure 3). Red lines indicate the π
orbitals, black lines indicate σ, and blue lines represent mixed
metalligand (M-L) orbitals. Some M-L type MOs may have
mixed σand πcharacters as the three ligands are arranged in
such a manner that σof one may mix with πof another. Metal
orbitals are mainly dπ, with some pπmixed. Co(III) orbitals
are much lower in energy to be shown in the above diagram. It
should also be noted that the HOMO and LUMO are M-L
type orbitals.
2.5. UVvis Spectroscopy of CoQ3.The absorption
spectrum of QH (Figure 4a) in 30% ethanol
6,7
shows four
absorption bands (at 250, 290, 530, and 565 nm) due to ππ*
and nπ*transitions of its various tautomeric forms in rapid
equilibrium in aqueous solution.
6,7,49
From the UVvis
spectrum of CoQ3(Figure 4b), it is clear that the absorption
peaks at 250, 290, 530, and 565 nm remain almost unaltered,
which indicate that the electronic absorption spectrum of
CoQ3depends weakly on the nature of the metal and is
primarily dened by the ligand (QH).
49
However, the
appearance of a new peak at 600 nm is characteristic of the
complex (CoQ3). It is important to mention here that
tautomeric structures found for free QH
49
in aqueous media
are not possible for CoQ3since phenolic OH groups in the
QH molecule are deprotonated owing to coordination of
Co(III) by phenolic oxygens.
2.6. Fluorescence Spectroscopy of CoQ3.Fluorescence
spectra of QH and CoQ3are shown in Figure S4 (SI) recorded
following an excitation at 530 nm. The emission spectrum
exhibits a maximum at 590 nm for QH and 594 nm for CoQ3.
The dierence in the emission peak of CoQ3compared to QH
is due to the metal ligand bond.
2.7. Electrochemical Reduction of CoQ3in Organic
Polar Solvents. Electrochemical behavior of CoQ3was
studied in anhydrous DMSO and DMF in the presence of
TBAB as the supporting electrolyte using cyclic voltammetry.
In anhydrous DMSO, CoQ3undergoes successive three one-
electron reductions having peak potentials (Epc)at0.795,
1.010, and 1.295 V, respectively, vs Ag/AgCl/saturated
KCl (Figure 5 and Table 1). In this case, the rst reduction is
reversible, while the other two are quasi-reversible at dierent
scan rates. These three one-electron reduction steps are due to
the reduction of the three free quinone centers of the three Q
bound to Co(III) inCoQ3(Scheme 3). For these reductions,
the formal potentials (E) of the respective reduction steps were
found at 0.750, 0.987, and 1.255 V, respectively. It is
noted that although there are three equivalent free quinone
sites in CoQ3(Scheme 3), there exists a dierence in their
formal potential values, which is quite appreciable. Thus, after
reduction at the rst free quinone in CoQ3, reduction of the
second and third quinone sites is signicantly delayed. In other
words, the reduced species (semiquinone radical anion) that
formed due to the rst or second reduction is stabilized in the
metal ion environment due to delocalization of the negative
charge. This is important with regard to the compounds
biochemical action since a stabilized semiquinone would delay
the reaction between semiquinone and molecular oxygen
3034
within cells where it would be employed.
In anhydrous DMF, under similar experimental conditions,
CoQ3undergoes three-one electron reductions having peak
potentials (Epc)at1.025, 1.225, and 1.475 V, respectively,
with the corresponding formal potentials (E) being 0.950,
1.195, and 1.405 V, respectively (Figure 6 and Table 1).
Considering the fact that the polarity of DMF is less than that
of DMSO
51
and comparing the three reduction potentials of
CoQ3in the two solvents, it can be said that with the
increasing polarity of the medium, reduction potentials move
in a positive direction and that reductions become more
feasible as the polarity of solvent increases. This means stability
of the formed semiquinone species is increased with an
increase in the polarity of the medium. Stabilization of the
Figure 1. Powder X-ray diraction patterns of CoQ3.
Figure 2. Energy optimized structure of CoQ3.
Figure 3. Dierent HOMOs (H) and LUMOs (L) of CoQ3.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
C
semiquinone is also reected in the formal reduction potential
data. This aspect is important with respect to its chemo-
therapeutic eciency.
3034
Owing to stabilization of the
semiquinone radical anion, the probability for reaction of a
semiquinone radical anion with molecular oxygen would be
delayed and that may reduce cardiotoxicity if the molecule
were to be employed as an anticancer agent.
3034
Under similar experimental conditions, a cyclic voltammo-
gram of QH shows two reversible waves at 0.816 and 1.355
V in anhydrous DMSO and at 0.832 and 1.309 V in
anhydrous DMF vs Ag/AgCl, with saturated KCl forming a
semiquinone radical anion and quinone dianion, respec-
tively.
7,8
Formal potentials for such reductions were evaluated
as 0.770 and 1.308 V in anhydrous DMSO and 0.785 and
1.258 V in anhydrous DMF.
8
Comparing electrochemical
parameters and cyclic voltammograms (Figures 5 and 6)of
CoQ3with those of QH in anhydrous DMSO and anhydrous
DMF,
8
one can say that the electrochemical behavior of QH
bound to a metal ion as Q(as in CoQ3) is signicantly altered.
It is seen that the reduction peak currents (Ipc) for three
successive reductions of CoQ3in both DMSO and DMF have
a linear relationship with the square root of the scan rate and
that it passes through the origin (Figures 5 and 6). This
suggests that such reductions are fully diusion controlled and
that there is no adsorption on the electrode surface. The
diusion coecient (DO) of CoQ3was determined by the
relation shown in eq 1
50
and found as 3.04 ×105and 6.31 ×
105cm2s1in DMSO and DMF, respectively (summarized in
Table 1).
I
nD ACv(2.69 10 )
pc 53/2
O1/2 1/2
(1)
where Ipc = cathodic peak current (A), n= number of electron
involved in the reduction, A= area of the electrode (cm2), C=
concentration (mol·cm3), and v= scan rate (V·s1).
From values of diusion coecients of CoQ3in two
dierent solvents (Table 1), it is evident that DOincreases as
the polarity of the solvent decreases, clearly indicating greater
solvation of CoQ3in a more polar solvent that causes lower
diusion onto the surface of the electrode. Thus, CoQ3is more
solvated in DMSO due to hydrogen bonding and other
electrostatic interactions.
8
Intermolecular hydrogen bonding
between one of the two hydrogen of aromatic amino group
(NH2) of QH and negatively charged oxygen of the solvent
(DMSO) is very strong.
8
This type of hydrogen bonding
would be weak in the case of DMF since for this solvent,
oxygen has a less partial negative charge than that on oxygen in
DMSO.
8
Figure 4. UVvis spectrum of (a) QH and (b) CoQ3in aqueous ethanol.
Figure 5. (a) Cyclic voltammogram of CoQ3in anhydrous DMSO media. Scan rate: 0.10 Vs1. [CoQ3]=1×103M, [TBAB] = 0.1 M, T=
298.15 K. (b) Plot of cathodic peak current vs square root of scan rate for rst (solid circles), second (open squares), and third reduction (solid
squares) of CoQ3in anhydrous DMSO.
Table 1. Electrochemical Parameters of CoQ3
a
media Epc-1 (V) Epc-2 (V) Epc-3 (V) E-1 (V) E-2 (V) E-3 (V) D0(cm2s1)
DMSO 0.795 1.010 1.295 0.750 0.987 1.255 3.04 ×105
DMF 1.025 1.225 1.475 0.950 1.195 1.405 6.31 ×105
a
Potentials were measured with respect to vs Ag/AgCl/saturated KCl.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
D
2.8. Eect of CoQ3on Viability of MCF-7 Human
Breast Cancer Cells by the MTT Assay. Using the MTT
assay, the cytotoxic activity of CoQ3was analyzed against
MCF-7 human breast cancer cells (Figure 7). It was estimated
according to dose values of exposure of CoQ3required to
reduce the survival to 50% (IC50) in comparison to that of
untreated cells. The IC50 value for 24 h was found to be (95 ±
0.05) μg/mL. This indicates that CoQ3is cytotoxic against
MCF-7 breast cancer cells.
2.9. AO/EB Staining. Apoptosis is the hallmark of cell
death and can be characterized by cellular morphological
Scheme 3. Three Step One-Electron Reductions of CoQ3in Organic Polar Solvents Like DMSO and DMF
Figure 6. (a) Cyclic voltammogram of CoQ3in anhydrous DMF
media. Scan rate: 0.10 Vs1. [CoQ3]=1×103M, [TBAB] = 0.1 M,
T= 298.15 K. (b) Plot of cathodic peak current vs square root of scan
rate for rst (open circles), second (open squares), and third
reduction (solid squares) of CoQ3in anhydrous DMF. Figure 7. Cytotoxic eect of CoQ3on MCF-7 human breast cancer
cells after exposure for 24 h.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
E
changes observed during the process of cell death. The dual
staining method of AO/EB detects such morphological
changes. Figure 8 corresponds to AO/EB staining of
control/non-treated and CoQ3-treated MCF7 breast cancer
cells. Based on uorescence emission and nucleus morphology,
cells were distinguished to have viable, apoptotic, or necrotic
characteristics. The viable cells were observed to have uniform
green-colored nuclei with a typical cell morphology and intact
membrane. On the other hand, apoptotic cells showed
irregular cell morphologies with orange to red condensed
chromatin and/or fragmented nuclei. Furthermore, the large
orange to red uorescent swollen cells with no fragmented
nuclei were dierentiated as necrotic cells. The results from
AO/EB staining reveal that the control group contains more
viable cells and a few apoptotic and necrotic cells. In contrast,
CoQ3-treated MCF7 breast cancer cells induced majority of
cell death through the apoptosis mode and actually very few by
necrosis. Furthermore, condensed and fragmented morpholo-
gies were mostly observed in the CoQ3treatment group. The
results of calculating the percentage of apoptotic cell death
induced by CoQ3and analyzed by uorescent images of AO/
EB staining revealed that AQS-treated cells induced a higher
percentage of apoptotic cells and a lower percentage of
necrotic cells than untreated cells (Figure 9). The graph
depicts a percentage count of apoptotic normal and abnormal
cells. The error bar represents the standard deviation across
three replicates.
3. CONCLUSIONS
A Co(III) complex of 1-amino-4-hydroxy-9,10-anthraquinone
(QH) with the molecular formula CoQ3was synthesized and
characterized by dierent methods. The optimized molecular
structure of CoQ3was estimated using computational
methods. The HOMO and LUMO of CoQ3were also
characterized by this method. Electrochemical properties of
CoQ3were studied in anhydrous DMSO and anhydrous DMF
media using cyclic voltammetry, and the mechanism of
reduction was established. It showed that dierent reduced
anions of CoQ3are stabilized in a metal surrounding
environment and that reductions would therefore be delayed.
Polarity of the solvents also aects stability of the reduced
anion. A signicant modication of electrochemical properties
of QH was also seen when it was bound to Co(III) in CoQ3.
The IC50 value of CoQ3for 24 h of incubation corresponding
to cytotoxicity of CoQ3on human breast cancer cells MCF-7
was evaluated as 95 ±0.05 μg/mL. The study revealed that
such cancer cells underwent both early and late apoptosis due
to CoQ3.
4. EXPERIMENTAL SECTION
4.1. Materials. 1-Amino-4-hydroxy-9, 10-anthraquinone
(QH) (Scheme 1) (96%) purchased from Alfa Aesar, Germany
was recrystallized from an ethanolmethanol mixture and
characterized as mentioned earlier.
69
The quinone moiety
being sensitive to light, solutions were prepared either just
before an experiment or very carefully stored in the dark.
CoCl2·6H2O purchased from Merck, India was used to prepare
the Co(III) complex. KCl and tetrabutyl ammonium bromide
[TBAB] (both are AR grade, Spectrochem, India) were used as
supporting electrolytes in aqueous and non-aqueous media,
respectively.
Dimethyl sulfoxide (DMSO) (99.0%, Spectrochem, India)
was rst dried over fused CaCl2for 34 days, decanted, and
then distilled under reduced pressure.
51
The distilled sample
was preserved in a well-stoppered Jena bottle in desiccators
and redistilled again before use. N,N-Dimethyl formamide
(DMF) (99.5%, Spectrochem, India) (LR, BDH) was
puried
52
rst by distilling under reduced pressure in a N2
atmosphere and then preserving the distillate over dry K2CO3
(Merck) for a week. Then, the DMF was decanted and allowed
to mix with dry P2O5(Riedel) and distilled again to be able to
use it under anhydrous conditions. Anhydrous DMF and
DMSO were used as solvents in electrochemistry experiments.
All aqueous solutions were prepared in triple-distilled water.
4.2. Synthesis of CoQ3.An aqueous solution of 0.5 mmol
CoCl2·6H2O and a solution of 1.5 mmol QH in acetonitrile
were mixed and stirred for about 6 h using a magnetic stirrer.
Co(II) was oxidized to Co(III) by purging air into the reaction
media. The solution was kept for 7 days in air to allow it to
evaporate till it was almost 5 mL. A violet-colored complex was
separated by ltration followed by washing with acetonitrile.
The complex was recrystallized from a methanolacetonitrile
mixture and dried in air. Results of elemental analysis showed
that it has the formula CoQ3. Found: C, 65.09%; H, 3.08%; N,
5.51%. Calculated: C, 65.13%; H, 3.10%; N, 5.43%. In 25%
aqueous ethanol solution, 0.1 mM CoQ3showed a
conductance less than 5 μS/cm at 298.15 K, indicating that
it is neutral.
4.3. Computational Studies. The structure of CoQ3was
optimized using DFT with Ahlrich SV basis
53,54
and B3LYP
functional
5557
using the Orca program suite.
58
Electronic
transitions were calculated by the time-dependent DFT
method with the same basis set and functional using Orca.
Pictures of molecular orbitals (MOs) were generated with the
same basis set and functional using Gaussian 09W
59
and MaSK
software.
60
4.4. Analytical Methods. With the help of a Perkin-Elmer
2400 II elemental analyzer, the carbon, hydrogen, and nitrogen
Figure 8. AO/EB staining of the control and CoQ3-treated MCF-7
human breast cancer cells.
Figure 9. Comparison of percentage of cells in apoptotic death
compared to healthy cells and necrotic death.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
F
analyses were done. FTIR analysis was performed on a Perkin
Elmer RX-I spectrophotometer. Spectra were obtained using
KBr pellets in the range 4000400 cm1. The mass spectrum
was recorded on Micromass Q-Tofmicro, Waters Corporation.
CoQ3was dissolved in an anhydrous acetonitrile solvent, and
the MS data were recorded by using ESI positive mode. The
instrument applies a focusing voltage to the electrospray probe
to promote mobile phase evaporation as part of the ionization
process. PXRD data was collected on a Bruker AXS D8 powder
diractometer using Cu Kαradiation (λ= 1.548 Å) generated
at 40 kV and 40 mA. UVvisible spectroscopy was done on a
spectrophotometer (model: MECASYS OPTIZEN POP).
Experiments related to cyclic voltammetry were performed
using the conventional three-electrode system. The temper-
ature was maintained at 25 °C with the help of a circulating
water bath. The working electrode was glassy carbon, the
surface area of which was 0.07065 cm2, the counter electrode
was a platinum wire, and the reference electrode was Ag/AgCl
in satd. KCl. Using a potentiostat (model DY2312, Digi-Ivy),
all electrochemical studies were performed. The range of
concentrations of dierent solutions was 5 ×105moldm3to
1.5 ×103moldm3. Before the solutions were subjected to
cyclic voltammetry, they were degassed for nearly30 min using
highly pure Ar.
4.5. Cell Culture. MCF7 human breast cancer cells were
procured from National Center for Cell Science, Pune, India.
Cells were cultured and maintained in DMEM high-glucose
medium (Sigma-Aldrich, USA) supplemented by 10% fetal
bovine serum (Gibco, Thermo Fisher, USA) and 20 mL of
penicillin/streptomycin as antibiotics (Gibco, Thermo Fisher,
USA), and incubated at 37C with 5% CO2in a CO2
incubator (Thermo scientic, USA). All experiments were
carried out using cells from the passage of 15 or less.
4.6. Cell Viability Assay. CoQ3was dissolved in DMSO
and a stock solution was prepared. It was then diluted to obtain
dierent concentrations of the compound in the range 0200
μg/mL. Two hundred microliters of such solutions was added
to wells containing 5 ×103MCF-7 cells per well of a 96-well
culture plate. DMSO was used as the control solvent. Twenty
microliters of MTT solution (5 mg/mL in PBS) was
transferred to each well following 24 h of incubation, and
the plate was incubated at 37 °C for 4 h in the dark. To
dissolve formazan crystals, 100 μL of DMSO was added to
each well and the absorbance of the nal solution was
measured at 570 nm using a microplate reader (Bio-Rad,
iMark, USA). Data was collected for three replicates each, and
the respective mean was used in the following formula to
calculate percentage inhibition:
percentage inhibition = ([mean OD of untreated cells
(control) mean OD of treated cells (treated)] ×100)/
(mean OD of untreated cells (control))
4.7. Acridine Orange (AO) and Ethidium Bromide (EB)
Staining. CoQ3-induced apoptosis was examined using the
uorescent-based dual staining method AO/EB as dened by
Spector et al.
61
with some modications. In brief, cells treated
for 24 h with the IC50 concentration of CoQ3were harvested
and washed with cold PBS. Cell pellets were resuspended and
diluted with PBS. The cell suspension (5000 in number) was
mixed with AO/EB solution (3.8 μM AO and 2.5 μMEBin
PBS) and transferred to a clean microscope slide. Morpho-
logical features of the cells were examined under a uorescent
microscope (Carl Zeiss, Axioscope2plus) with a UV lter
(450490 nm).
ASSOCIATED CONTENT
*
sıSupporting Information
The Supporting Information is available free of charge at
https://pubs.acs.org/doi/10.1021/acsomega.1c06125.
(Figure S1) Mass Spectrum of CoQ3, (Figure S2) IR
spectrum of QH and CoQ3, (Figure S3) energy level
diagram of QH and CoQ3, (Figure S4) uorescence
spectra of QH and CoQ3in aqueous ethanol, and (Table
S1) structural parameters of CoQ3(PDF)
AUTHOR INFORMATION
Corresponding Authors
Saurabh Das Department of Chemistry, Jadavpur University,
Kolkata 700032, India; orcid.org/0000-0002-0455-
8760; Phone: +91 9123865911; Email: dasrsv@yahoo.in,
saurabh.das@jadavpuruniversity.in; Fax: +91 33
24146223
Asoke Prasun Chattopadhyay Department of Chemistry,
University of Kalyani, Nadia 741235 West Bengal, India;
orcid.org/0000-0002-2411-1384;
Phone: +919836156800; Email: asoke@klyuniv.ac.in;
Fax: : +91 33 2582 8282
Partha Sarathi Guin Department of Chemistry, Shibpur
Dinobundhoo Institution (College), Howrah 711102 West
Bengal, India; orcid.org/0000-0001-9258-9227;
Phone: +91 9330083036; Email: parthasg@gmail.com;
Fax: +91 33 2688 0344
Authors
Somenath Banerjee Department of Chemistry, Shibpur
Dinobundhoo Institution (College), Howrah 711102 West
Bengal, India; Department of Chemistry, Jadavpur
University, Kolkata 700032, India
Sanjay Roy Department of Chemistry, Netaji Subhas Open
University, Nadia 741235, India; orcid.org/0000-0001-
6841-4961
Dhanasekaran Dharumadurai Department of Microbiology,
School of Life Sciences, Bharathidasan University,
Tiruchirappalli 620 024, India
Balaji Perumalsamy National Centre for Alternatives to
Animal Experiments, Bharathidasan University,
Tiruchirappalli 620 024, India
Ramasamy Thirumurugan National Centre for Alternatives
to Animal Experiments, Bharathidasan University,
Tiruchirappalli 620 024, India
Complete contact information is available at:
https://pubs.acs.org/10.1021/acsomega.1c06125
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
P.S.G. is grateful to UGC, New Delhi, India, for the nancial
support through the Major Research Project (le no. 41-225/
2012(SR) dated 18 July 2012).
ABBREVIATIONS
QH, 1-amino-4-hydroxy-9,10-anthraquinone; CoQ3, Co(III)
complex of 1-amino-4-hydroxy-9,10-anthraquinone; TBAB,
tetrabutyl ammonium bromide; DMSO, dimethyl sulfoxide:;
DMF, N,N-dimethyl formamide
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
G
REFERENCES
(1) Hardman, J. -G.; Gilman, A.-G.; Limbird, L.-E. Goodman and
Gilmans The Pharmacological Basis of Therapeutics, 9th ed.; McGraw-
Hill Companies: New York, 1996.
(2) Lim, K. H.; Kim, H. S.; Yang, Y. M.; Lee, S. D.; Kim, W. B.;
Yang, J.; Park, J. G. Cellular uptake and antitumor activity of the new
anthracycline analog DA-125 in human cancer cell lines. Cancer
Chemother. Pharmacol. 1997,40,2330.
(3) Preobrazhenskaya, M. N.; Tevyashova, A. N.; Olsufyeva, E. N.;
Huang, K.-F.; Huang, H.-S. Second generation drugs-derivatives of
natural antitumor anthracycline antibiotics daunorubicin, doxorubicin
and carminomycin. J. Med. Sci. 2006,26, 119128.
(4) Hu, F. Q.; Liu, L. N.; Du, Y. Z.; Yuan, H. Synthesis and
antitumor activity of doxorubicin conjugated stearic acid-g-chitosan
oligosaccharide polymeric micelles. Biomaterials 2009,30, 6955
6963.
(5) Das, A.; Roy, S.; Mondal, P.; Datta, A.; Mahali, K.; Loganathan,
G.; Dharumadurai, D.; Sengupta, P. S.; Akbarsha, M. A.; Guin, P. S.
Studies on the interaction of 2-amino-3-hydroxy-anthraquinone with
surfactant micelles reveal its nucleation in human MDA-MB-231
breast adinocarcinoma cell. RSC Adv. 2016,6, 2820028212.
(6) Mondal, P.; Roy, S.; Loganathan, G.; Mandal, B.; Dharumadurai,
D.; Akbarsha, M. A.; Sengupta, P. S.; Chattopadhyay, S.; Guin, P. S. 1-
amino-4-hydroxy-9,10-anthraquinone - An analogue of anthracycline
anticancer drugs, interacts with DNA and induces apoptosis in human
MDA-MB-231 breast adinocarcinoma cells: Evaluation of structure-
activity relationship using computational, spectroscopic and bio-
chemical studies. Biochem. Biophys. Rep. 2015,4, 312323.
(7) Roy, S.; Mondal, P.; Sengupta, P. S.; Dhak, D.; Santra, R. C.;
Das, S.; Guin, P. S. Spectroscopic, computational and electrochemical
studies on the formation of the copper complex of 1-amino-4-
hydroxy-9,10-anthraquinone and effect of it on superoxide formation
by NADH dehydrogenase. Dalton Trans. 2015,44, 54285440.
(8) Roy, S.; Guin, P. S. Solvation of 1-amino-4-Hydroxy-9,10-
anthraquinone governs its electrochemical behavior in non-aqueous
and aqueous media: A cyclic voltammetry study. J. Electrochem. Soc.
2015,162, H124H131.
(9) Roy, S.; Guin, P. S. Investigation on the interaction of 1-amino-
4-hydroxy-9,10 anthraquinone with calf thymus DNA and CTAB
micelles. J. Mol. Liq. 2015,211, 846853.
(10) Guin, P. S.; Das, S.; Mandal, P. C. Studies on the formation of a
complex of Cu(II) with sodium 1,4-dihydroxy-9,10-anthraquinone-2-
sulphonate- An analogue of the core unit of anthracycline anticancer
drugs and its interaction with calf thymus DNA. J. Inorg. Biochem.
2009,103, 17021710.
(11) Guin, P. S.; Mandal, P. C.; Das, S. The binding of a hydroxy-
9,10-anthraquinone CuII complex to calf thymus DNA : Electro-
chemistry and UV/Vis Spectroscopy. ChemPlusChem. 2012,77, 361
369.
(12) Rossi, S.; Tabolacci, C.; Lentini, A.; Provenzano, B.;
Carlomosti, F.; Frezzotti, S.; Beninati, S. Anthraquinones danthron
and quinizarin exert antiproliferative and antimetastatic activity on
murine B16-F10 melanoma cells. Anticancer Res. 2010,30, 445449.
(13) Das, P.; Jain, C. K.; Dey, S. K.; Saha, R.; Chowdhury, A. D.;
Roychoudhury, S.; Kumar, S.; Majumder, H. K.; Das, S. Synthesis,
crystal structure, DNA interaction and in vitro anticancer activity of a
Cu(II) complex of purpurin: Dual poison for human DNA
topoisomerase I and II. RSC Adv. 2014,4, 5934459357.
(14) Das, P.; Bhattacharya, D.; Karmakar, P.; Das, S. Influence of
ionic strength on the interaction of THA and its Cu (II) complex with
DNA helps to explain studies on various breast cancer cells. RSC Adv.
2015,5, 7309973111.
(15) Das, P.; Guin, P. S.; Mandal, P. C.; Paul, M.; Paul, S.; Das, S.
Cyclic voltammetric studies of 1,2,4-trihydroxy-9,10-anthraquinone,
its interaction with calf thymus DNA and anti-leukemic activity on
MOLT-4 cell lines: A comparison with anthracycline anticancer
drugs. J. Phys. Org. Chem. 2011,24, 774785.
(16) Mandal, B.; Singha, S.; Dey, S. K.; Mazumdar, S.; Mondal, T.
K.; Karmakar, P.; Kumar, S.; Das, S. Synthesis, crystal structure from
PXRD of a MnII(purp)2complex, interaction with DNA at different
temperatures and pH and lack of stimulated ROS formation by the
complex. RSC Adv. 2016,6, 5152051532.
(17) Nakayama, T.; Okumura, N.; Uno, B. Complementary Effect of
Intra- and Intermolecular Hydrogen Bonds on Electron Transfer in β-
Hydroxy-Anthraquinone Derivatives. J. Phys. Chem. B. 2020,124,
848860.
(18) Tian, W.; Wang, C.; Li, D.; Hou, H. Novel anthraquinone
compounds as anticancer agents and their potential mechanism.
Future Med. Chem. 2020, 627.
(19) Tikhomirov, A. S.; Shtil, A. A.; Shchekotikhin, A. E. Advances
in the Discovery of Anthraquinone-Based Anticancer Agents. Recent
Pat. Anti-Cancer Drug Discovery 2018,13, 159183.
(20) Mukherjee Chatterjee, S.; Jain, C. K.; Singha, S.; Das, P.;
Roychoudhury, S.; Majumder, H. K.; Das, S. Activity of CoII
quinalizarin: A novel analogue of anthracyclinebased anticancer
agents targets human DNA topoisomerase, whereas quinalizarin itself
acts via formation of semiquinone on acute lymphoblastic leukemia
MOLT-4 and HCT 116 cells. ACS Omega 2018,3, 1025510266.
(21) Błasiak, J.; Gloc, E.; Warszawski, M. A comparison of the in
vitrogenotoxicity of anticancer drugs idarubicin and mitoxantrone.
Acta Biochim. Pol. 2002,49, 145155.
(22) Kapuscinski, J.; Daizynkiewicz, Z. Relationship between the
pharmacological activity of antitumor drugs ametantrone and
mitoxantrone (novatrone) and their ability to condense nucleic
acids. Proc. Natl. Acad. Sci. U. S. A. 1986,83, 63026306.
(23) Ellis, A. L.; Randolph, J. K.; Conway, B. R.; Gewirtz, D. A.
Biochemical lesions in DNA associated with the antiproliferative
effects of mitoxantrone in the hepatoma cell. Biochem. Pharmacol.
2009,39, 15491556.
(24) Li, N.; Ma, Y.; Yang, C.; Guo, L.; Yang, X. Interaction of
anticancer drug mitoxantrone with DNA analyzed by electrochemical
and spectroscopic methods. Biophys. Chem. 2005,116, 199205.
(25) Riahi, S.; Reza Ganjali, M.; Dinarvand, R.; Karamdoust, S.;
Bagherzadeh, K.; Norouzi, P. A theoretical study on interactions
between mitoxantrone as an anticancer drug and DNA: application in
drug design. Chem. Biol. Drug. Des. 2008,71, 474482.
(26) Trachtenberg, B. H.; Landy, D. C.; Franco, V. I.; Henkel, J. M.;
Pearson, E. J.; Miller, T. L.; Lipshultz, S. E. Anthracycline-associated
cardiotoxicity in survivors of childhood cancer. Pediatr. Cardiol. 2011,
32, 342353.
(27) Shi, Y.; Moon, M.; Dawood, S.; McManus, B.; Liu, P. P.
Mechanisms and management of doxorubicin cardiotoxicity. Herz.
2011,36, 296305.
(28) Outomuro, D.; Grana, D. R.; Azzato, F.; Milei, J. Adriamycin-
induced myocardial toxicity: New solutions for an old problem? Int. J.
Cardiol. 2007,117,615.
(29) Ferrans, V. J. Overview of cardiac pathology in relation to
anthracycline cardiotoxicity. Cancer Treat. Rep. 1978,62, 955961.
(30) Barasch, D.; Zipori, O.; Ringel, I.; Ginsburg, I.; Samuni, A.;
Katzhendler, J. Novel anthraquinone derivatives with redox-active
functional groups capable of producing free radicals by metabolism:
are free radicals essential for cytotoxicity? Eur. J. Med. Chem. 1999,34,
597.
(31) Kumbhar, A.; Padhye, S.; Ross, D. Cytotoxic properties of iron-
hydroxynaphthoquinone complex in rat hepatocytes. BioMetals 1996,
9, 235240.
(32) Bartoszek-Pa̧czkowska, A. Metabolic activation of adriamycin
by NADPH cytochrome P450 reductase; overview of its biological
and biochemical effects. Acta Biochim. Pol. 2002,49, 323331.
(33) Mandal, B.; Mondal, H. K.; Das, S. In situ reactivity of
electrochemically generated semiquinone on emodin and its CuII/
MnII complexes with pyrimidine based nucleic acid bases and calf
thymus DNA: Insight into free radical induced cytotoxicity of
anthracyclines. Biochem. Biophys. Res. Commun. 2019,515, 505509.
(34) Das, S.; Saha, A.; Mandal, P. C. Radiation-induced double-
strand modification in calf thymus DNA in the presence of 1,2-
dihydroxy-9,10-anthraquinone and its Cu(II) complex. J. Radio-
anal.Nucl. Chem. 1996,196,5763.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
H
(35) Feng, M.; Yang, Y.; He, P.; Fang, Y. Spectroscopic studies of
copper(II) and iron(II) complexes of adriamycin. Spectrochim. Acta
2000,56, 581587.
(36) Wang, H.; Hua, E.; Yang, P. The polarographic and
voltammetric behaviour of the copper(II) mitoxantrone complex
and its analytical application. Talanta 1995,42, 15191524.
(37) Yang, P.; Wang, H.; Gao, F.; Yang, B. Antitumor activity of the
Cu(II)-mitoxantrone complex and its interaction with deoxyribonu-
cleic acid. J. Inorg. Biochem. 1996,62, 137145.
(38) Pereira, E.; Fiallo, M. M. L.; Garnier-Suillerot, A.; Kiss, T.;
Kozłowski, H. Impact of aluminium ions on adriamycin-type ligands.
J. Chem. Soc., Dalton Trans. 1993, 455459.
(39) Italia, T.; Liegro, D. I.; Cestelli, A.; Matzanke, B. F.; Bill, E.;
Trautwein, A. X. The interaction of Fe(III), adriamycin and
daunomycin with nucleotides and DNA and their effects on cell
growth of fibroblasts (NIH-3T3). BioMetals 1996,9, 121130.
(40) Massoud, S. S.; Jordan, R. B. Kinetic and equilibrium studies of
the complexation of aqueous iron(III) by daunomycin, quinizarin, and
quinizarin-2-sulfonate. Inorg. Chem. 1991,30, 48514856.
(41) Maroney, M. J.; Day, R. O.; Psyris, T.; Fleury, L. M.;
Whitehead, J. P. Structural model for the binding of iron by
anthracycline drugs. Inorg. Chem. 1989,28, 173175.
(42) Kolodziejczyk, P.; Garnier-Suillerot, A. Circular dichroism
study of the interaction of mitoxantrone, ametantrone and their
Pd(II) complexes with deoxyribonucleic acid. Biochim. Biophys. Acta
1987,926, 249257.
(43) Kadarkaraisamy, M.; Mukjerjee, D.; Soh, C. C.; Sykes, A. G.
Complexation, crystallography, and electrochemistry of new chelating
nicotinyl and thiazolylanthraquinone ligands. Polyhedron 2007,26,
40854092.
(44) Di Vaira, M.; Orioli, P.; Piccioli, F.; Bruni, B.; Messori, L.
Structure of a terbium(III)quinizarine complex: The first crystallo-
graphic model for metalloanthracyclines. Inorg. Chem. 2003,42,
31573159.
(45) Jabłońska-Trypuć,A.;S
́widerski, G.; Krętowski,R.K.;
Lewandowski, W. Newly Synthesized Doxorubicin Complexes with
Selected MetalsSynthesis, Structure and Anti-Breast Cancer
Activity. Molecules 2017,22, 1106.
(46) Rumyantseva, T. A.; Alekseeva, A. A.; Tkachenko, M. A.
Synthesis and Properties of Metal Phthalocyanines Containing
Anthraquinone Chromophores. Rus. J. Gen. Chem. 2020,90, 1660
1663.
(47) Du, S.; Feng, J.; Lu, X.; Wang, G. The synthesis and
characterizations of vanadium complexes with 1,2-dihydroxyanthra-
quinone and the structure-effect relationship in their in vitro
anticancer activities. Dalton Trans. 2013,42, 96999705.
(48) Yuan, H.; Cheng, B.; Lei, J.; Jiang, L.; Han, Z. Promoting
photocatalytic CO2reduction with a molecular copper purpurin
chromophore. Nat. Commun. 2021,12, 1835.
(49) Fain, V. Y.; Zaitsev, B. E.; Ryabov, M. A. Tautomerism of the
metal complexes with 1-amino-4-hydroxyanthaquinone. Russ. J. Coord.
Chem. 2010,36, 396400.
(50)Bard,A.J.;Faulkner,L.R.Electrochemical Methods:
Fundamentals and Applications, 2nd ed.; John Wiley & Sons: New
York , 1980.
(51) Undre, P. B.; Khirade, P. W.; Rajenimbalkar, V. S.; Helambe, S.
N.; Mehrotra, S. C. Dielectric Relaxation in Ethylene Glycol -
Dimethyl Sulfoxide Mixtures as a Function of Composition and
Temperature. J. Korean Chem. Soc. 2012,56, 416423.
(52) Bose, K.; Kundu, K. Free energies of transfer of some single
ions from ethylene glycol to its isodielectric mixtures with acetonitrile
at 25°C. Can. J. Chem. 1978,57, 2476.
(53) Schäfer, A.; Horn, H.; Ahlrichs, R. Fully Optimized Contracted
Gaussian Basis Sets for Atoms Li to Kr. J. Chem. Phys. 1992,97,
25712577.
(54) Schäfer, A.; Huber, C.; Ahlrichs, R. Fully optimized contracted
Gaussian basis sets of triple zeta valence quality for atoms Li to Kr. J.
Chem. Phys. 1994,100, 58295835.
(55) Becke, A. D. Perspective on Density functional thermochem-
istry. III. The role of exact exchange. J. Chem. Phys. 1993,98, 5648
5652.
(56) Becke, A. D. Density-functional exchange-energy approxima-
tion with correct asymptotic behavior. Phys. Rev. A 1988,38, 3098
3100.
(57) Lee, C.; Yang, W.; Parr, R. G. Development of the Colle-
Salvetti correlation-energy formula into a functional of the electron
density. Phys. Rev. B 1988,37, 785789.
(58) Orca an ab initio, DFT and Semi-empirical and SCFMO
package, F. Neese, Max Planck Institute for Chemical Energy
Conversion, Ruhr, Germany, version 3.0.1.
(59) Gaussian 09, Revision A.02, Frisch, M. J. et al., Gaussian, Inc.,
Wallingford, CT, 2009.
(60) Podolyan, Y.; Leszczynski, J. MaSK: A visualization tool for
teaching and research in computational chemistry. Int. J. Quantum
Chem. 2009,109,816.
(61) Spector, D. L.; Goldman, R. D.; Leinwand, L. A., Cell: A
Laboratory Manual, Culture and Biochemical Analysis of Cells, Cold
Spring Harbor Laboratory Press: Cold Spring Harbor, CSHL Press:
New York,1998, pp.341349.
ACS Omega http://pubs.acs.org/journal/acsodf Article
https://doi.org/10.1021/acsomega.1c06125
ACS Omega XXXX, XXX, XXXXXX
I
... The phenolic OH groups in the free QH molecule are deprotonated due to the coordination of Co(III) by the phenolic oxygens, hence it is vital to note that tautomeric structures observed for free QH in aqueous conditions are not conceivable for CoQ3. [69] Content courtesy of Springer Nature, terms of use apply. Rights reserved. ...
... The evaluation of 3.10 104 M for the critical micelle concentration (CMC) of CTAB in the presence of AHAQ was made by observing the change in AHAQ's absorption spectra (the CTAB concentration corresponding to the first point highlighting the increase in absorbance). This number is comparable to the CTAB CMC value obtained from the electrochemical measurements [69] discussed above in the presence of AHAQ. Figure 11b depicts the fluctuation in absorbance at 530 nm as a function of surfactant concentration. It is evident that the absorbance falls as CTAB concentration rises and achieves a saturation level at a certain CTAB concentration. ...
Article
Full-text available
Anthraquinones constitute an important class of compounds with wide applications. The solubility of derivatives at 298.15 K was discussed in ethanol–water solution and at atmospheric pressure, the solubility of 1-amino-4-hydroxy-9,10-anthraquinone (AHAQ) in binary solvents (ethanol–water combinations) was determined. Colour strength and fastening properties depend upon the kind and position of a hydrophobic group connected to the phenoxy ring of Anthraquinone moiety. There is a continuing interest in the creation of novel anthraquinone derivatives with biological activities since they have demonstrated potential for treating multiple sclerosis. For this purpose, by utilizing voltammetric and absorption studies, interactions of various derivatives with calf thymus DNA (ct-DNA) and the cationic surfactant cetyltrimethylammoniumbromide (CTAB) were examined. Here prominent Hydrophobic interaction and electron transfer resulting in binding to CTAB micelles were observed. The polarity index of the media was assessed and associated with the electrochemical parameters. The medicinal behaviour of Anthraquinone derivatives was a result of electron transfer reactions with DNA. UV–Visible and fluorescence properties were due to the transitions between n* and π* orbitals. Large absorption band with low dichroic ratio was characteristic of various derivatives of Anthraquinone. Presence of –NH group proves various derivatives remarkable calorimetric and anionic sensors. Graphical Abstract
Article
Full-text available
CO 2 reduction through artificial photosynthesis represents a prominent strategy toward the conversion of solar energy into fuels or useful chemical feedstocks. In such configuration, designing highly efficient chromophores comprising earth-abundant elements is essential for both light harvesting and electron transfer. Herein, we report that a copper purpurin complex bearing an additional redox-active center in natural organic chromophores is capable to shift the reduction potential 540 mV more negative than its organic dye component. When this copper photosensitizer is employed with an iron porphyrin as the catalyst and 1,3-dimethyl-2-phenyl-2,3-dihydro-1 H -benzo[ d ]imidazole as the sacrificial reductant, the system achieves over 16100 turnover number of CO from CO 2 with a 95% selectivity (CO vs H 2 ) under visible-light irradiation, which is among the highest reported for a homogeneous noble metal-free system. This work may open up an effective approach for the rational design of highly efficient chromophores in artificial photosynthesis.
Article
Full-text available
Quinalizarin (THAQ), a hydroxy-9,10-anthraquinone analogue of the family of anthracycline anticancer drugs and an inhibitor of protein kinase, was observed for its anticancer activity. Because apart from showing anticancer activity, anthracyclines and their analogues also show cardiotoxic side effects, believed to be addressed through metal complex formation; an effort was made to realize this by preparing a CoII complex of THAQ. The aim of this study was to find out if complex formation leads to a decrease in the generation of intermediates that are responsible for toxic side effects. However, because this also meant that efficacy on cancer cells would be compromised, studies were undertaken on two cancer cell lines, namely, acute lymphoblastic leukemia (ALL) MOLT-4 and HCT116 cells. The complex decreases the flow of electrons from NADH to molecular oxygen (O2) in the presence of NADH dehydrogenase forming less semiquinone than THAQ. It showed increased affinity toward DNA with binding constant values remaining constant over the physiological pH range unlike THAQ (for which decrease in binding constant values with increase in pH was observed). The complex is probably a human DNA topoisomerase I and human DNA topoisomerase II poison acting by stabilizing the covalent topoisomerase-cleaved DNA adduct, a phenomenon not observed for THAQ. Activity of the compounds on cancer cells suggests that THAQ was more effective on ALL MOLT-4 cells, whereas the complex performed better on HCT116 cells. Results suggest that the formation of semiquinone probably dominates the action because of THAQ, whereas the performance of the complex is attributed to increased DNA binding, inhibition of topoisomerase, and so forth. Inspite of a decrease in the generation of superoxide by the complex, it did not hamper efficacy on either cell line, probably compensated by improved DNA binding and inhibition of topoisomerase enzymes which are positive attributes of complex formation. A decrease in superoxide formation suggests that the complex could be less cardiotoxic, thus increasing its therapeutic index.
Article
Full-text available
Doxorubicin (DOX) is very effective chemotherapeutic agent, however it has several major drawbacks. Therefore the motivation for developing novel drug complexes as anticancer agents with different mechanism of action has arisen. The aim of the present study was to evaluate the influence of newly synthesized DOX complexes with selected metals (Mg, Mn, Co, Ni, Fe, Cu, Zn) on apoptosis, cell cycle, viability, proliferation and cytotoxicity in the breast cancer cell line MCF-7. Complexation of DOX with metals has likewise been the subject of our research. The current work showed that the tested bivalent metals at a given pH condition formed metal:DOX complexes in a ratio of 2:1, while iron complexes with DOX in a ratio of 3:1. The studies also showed that selected metal-DOX complexes (Mg-DOX, Mn-DOX, Ni-DOX) at 0.5 µM concentration significantly decreased cell viability and proliferation, however they increased caspase 7 activity. Results also indicated that studied metal-DOX complexes showed high cytotoxicity in MCF-7 cells. Therefore they were chosen for cell cycle check-points and apoptosis/necrosis analysis studied by flow cytometry. Obtained results suggest that doxorubicin complexed by specified metals can be considered as a potential anti-breast cancer agent, which is characterized by a higher efficacy than a parent drug.
Article
Anthraquinones exhibit a unique anticancer activity. Since their discovery, medicinal chemists have made several structural modifications, resulting in the design and synthesis of a large number of novel anthraquinone compounds with different biological activities. In general, anthraquinone compounds have been considered to have anticancer activity mainly through DNA damage, cycle arrest and apoptosis. However, recent studies have shown that novel anthraquinone compounds may also inhibit cancer through paraptosis, autophagy, radiosensitising, overcoming chemoresistance and other methods. This Review article provides an overview of novel anthraquinone compounds that have been developed as anticancer agents in recent years and focuses on their anticancer mechanism.
Article
We have carried out an electrochemical and theoretical study on the relationship between electron transfer (ET) and hydrogen bonding in 11 different 9,10-anthraquinone (AQ) derivatives, including β–hydroxy AQs and their methoxylated analogs, in the presence of hydrogen donors in acetonitrile (ACN). The complementary effects of the intra- and intermolecular hydrogen bonds (HBs) on ET were studied by analyzing the complex formation constants derived from the intermolecular HB. Our results revealed that the inductive effect of the β–substituent which indirectly controls the charge distribution on the AQ carboxyl oxygen, and steric hindrance at the β–position, affect the complex formation constants. Furthermore, analysis of ET in different isomeric dihydroxy AQs suggested that the position of the hydroxy groups affects charge distribution and stabilizes structures through conjugation of the quinone moiety including ipso–ring, controlling the intra- and intermolecular HBs complementarily.
Article
There is enough proof to believe that free-radical intermediates of anthracycline based anticancer agents are involved in different stages of drug action. Subtle therapeutic differences observed in the actions of different anthracyclines largely influence the mechanism of action of the drugs that distinguish one member from another. Redox properties control biological responses related either to the one-electron quinone/semiquinone couple or the two-electron quinone/quinone-dianion couple. Comproportionation also leads to generation of semiquinone. Hence, whatever the form of reduction of the quinone moiety, a substantial amount of semiquinone is eventually formed in the system. Immediately after formation, there is competition between natural radical-decay pathways and one-electron transfer reactions that generate the superoxide-radical anion. Prototropic properties control rate of radical decay while redox properties control rate of electron transfer to molecular oxygen. In aerated medium, semiquinone-radical anion and superoxide-radical anion co-exist while in de-aerated medium semiquinone-radical anion predominates. All the radicals are damaging to the biological system. Through this study, attempt was made to detect changes induced by the radicals on pyrimidine based nucleic acid bases and calf thymus DNA in aerated and de-aerated (Ar saturated) medium to know the mechanism by which Emodin, its CuII/MnII complexes might affect DNA. Semiquinone-radical anion was generated electrochemically maintaining a glassy carbon electrode at the first reduction potential of each compound. Since the chosen compound (Emodin), its complexes are analogues of anthracyclines, findings on them can be extrapolated to understand the differences in anticancer activity or of adverse drug reactions reported in an innumerable number of clinical studies related to anthracyclines where the difference in structure of different members is due to differences in the relative positioning of hydroxy groups on the hydroxy-9, 10-anthraquinone moiety of anthracyclines. The study helps to realize action of compounds of this class as anticancer agents.
Article
Background: The anthracene-9,10-dione (anthraquinone) derivatives represent an exceptionally valuable class in anticancer drug development. An outstanding antitumor potency of the anthracycline antibiotics attracted the attention of medicinal chemists since the discovery of these chemotypes. The prominent anthraquinone-based drugs doxorubicin, mitoxantrone as well as more recent epirubicin, idarubicin, and valrubicin are successfully used in chemotherapy of hematological malignancies and solid tumors. The anthraquinone core remains a promising scaffold for the search of new optimized drug candidates. Objective: In this study we analyze the progress in discovery and development of antitumor anthracene-9,10-diones based on patent and journal publications in 2008-2017. The main goal is to dissect novel chemotypes of anthraquinone derivatives; other important issues such as the successes in bioconjugate chemistry of anthraquinone containing agents as well as the patents on new applications of anthracyclines are beyond the scope of this review. Conclusion: A number of newly discovered natural products, the perspective directions for chemical modifications to optimize the anticancer properties, and novel intracellular targets demonstrate that anthracene-9,10-diones deserve further in-depth investigation as an important source of drug candidates.
Article
The formation of reactive oxygen species (ROS) by anthracycline anticancer drugs is essential for their antitumor activity but they also make these drugs cardiotoxic. When complexed with metal ions there is a decrease in ROS formation and therefore in cardiotoxicity. Interestingly, in spite of producing fewer ROS, some of the complexes are effective antitumor agents, often better than the parent anthracycline. Purpurin (LH3), a hydroxy-9,10-anthraquinone, resembles doxorubicin at the core. An MnII complex of LH3 [MnII(LH2)2] was synthesized to see the extent to which the complex resembles metal–anthracyclines with regard to structure and function. The crystal structure was determined by Rietveld refinement of PXRD data using an appropriate structural model developed on the basis of spectroscopic information. This is only the second report on the crystal structure of a hydroxy-9,10-anthraquinone with a 3d-transition metal ion. Bond lengths and bond angles were obtained by structural refinement. The structure is supported by DFT calculations. DNA binding of the complex is slightly better than for purpurin but more importantly unlike purpurin, the binding constant values remained constant even with an increase in the pH of the medium. The NADH dehydrogenase assay and the DCFDA-ROS generation assay showed that generation of superoxide in the former and ROS in general in the latter were significantly less for the complex than for purpurin. Even with decreased ROS formation, the complex is able to maintain the biological activity of purpurin.