scispace - formally typeset
Open AccessJournal ArticleDOI

6-Methoxyquinoline complexes as lung carcinoma agents: induction of oxidative damage on A549 monolayer and multicellular spheroid model

Reads0
Chats0
TLDR
The results reveal a synergistic activity with a 1:1 ratio of both complexes in the monolayer and multicellular spheroids.
Abstract
The aim of this work was to study the antitumor effects and the mechanisms of toxic action of a series of 6-methoxyquinoline (6MQ) complexes in vitro. The Cu(II) and Zn(II) complexes (Cu6MQ and Zn6MQ) are formulated as M(6MQ)2Cl2; the Co(II) and Ag(I) compounds (Co6MQ and Ag6MQ) are ionic with formulae [Ag(6MQ)2]+NO3− and H(6MQ)+[Co(6MQ)Cl3]− (where H(6MQ)+ is the protonated ligand). We found that the copper complex, outperformed the Co(II), Zn(II) and Ag(I) complexes with a lower IC50 (57.9 µM) in A549 cells exposed for 24 h. Cu6MQ decreased cell proliferation and induced oxidative stress detected with H2DCFDA at 40 µM, which reduces GSH/GSSG ratio. This redox imbalance induced oxidative DNA damage revealed by the Micronucleus test and the Comet assay, which turned into a cell cycle arrest at G2/M phase and induced apoptosis. In multicellular spheroids, the IC50 values tripled the monolayer model (187.3 µM for 24 h). At this concentration, the proportion of live/dead cells diminished, and the spheroids could not proliferate or invade. Although Zn6MQ also decreased GSH/GSSG ratio from 200 µM and the cytotoxicity is related to oxidative stress, the induction of the hydrogen peroxide levels only doubled the control value. Zn6MQ induced S phase arrest, which relates with the increased micronucleus frequency and with the induction of necrosis. Finally, our results reveal a synergistic activity with a 1:1 ratio of both complexes in the monolayer and multicellular spheroids.

read more

Content maybe subject to copyright    Report

Vol.:(0123456789)
1 3
JBIC Journal of Biological Inorganic Chemistry
https://doi.org/10.1007/s00775-019-01644-7
ORIGINAL PAPER
6‑Methoxyquinoline complexes aslung carcinoma agents: induction
ofoxidative damage onA549 monolayer andmulticellular spheroid
model
J.F.Cadavid‑Vargas
1,2
· C.Villa‑Pérez
1
· M.C.Ruiz
1,2
· I.E.León
1
· G.C.Valencia‑Uribe
3
· D.B.Soria
1
·
S.B.Etcheverry
1,2
· A.L.DiVirgilio
1,2
Received: 9 November 2018 / Accepted: 17 January 2019
© Society for Biological Inorganic Chemistry (SBIC) 2019
Abstract
The aim of this work was to study the antitumor effects and the mechanisms of toxic action of a series of 6-methoxyquinoline
(6MQ) complexes invitro. The Cu(II) and Zn(II) complexes (Cu6MQ and Zn6MQ) are formulated as M(6MQ)
2
Cl
2
; the
Co(II) and Ag(I) compounds (Co6MQ and Ag6MQ) are ionic with formulae [Ag(6MQ)
2
]
+
NO
3
and H(6MQ)
+
[Co(6MQ)
Cl
3
]
(where H(6MQ)
+
is the protonated ligand). We found that the copper complex, outperformed the Co(II), Zn(II) and
Ag(I) complexes with a lower IC
50
(57.9µM) in A549 cells exposed for 24h. Cu6MQ decreased cell proliferation and
induced oxidative stress detected with H
2
DCFDA at 40µM, which reduces GSH/GSSG ratio. This redox imbalance induced
oxidative DNA damage revealed by the Micronucleus test and the Comet assay, which turned into a cell cycle arrest at G2/M
phase and induced apoptosis. In multicellular spheroids, the IC
50
values tripled the monolayer model (187.3µM for 24h). At
this concentration, the proportion of live/dead cells diminished, and the spheroids could not proliferate or invade. Although
Zn6MQ also decreased GSH/GSSG ratio from 200µM and the cytotoxicity is related to oxidative stress, the induction of the
hydrogen peroxide levels only doubled the control value. Zn6MQ induced S phase arrest, which relates with the increased
micronucleus frequency and with the induction of necrosis. Finally, our results reveal a synergistic activity with a 1:1 ratio
of both complexes in the monolayer and multicellular spheroids.
Keywords 6-Methoxyquinoline complexes· Lung carcinoma· A549 cells· Multicellular spheroid model· Oxidative
damage
Introduction
The constant worldwide expansion of a disease such as
cancer is challenging the research and development of new
drugs and is pushing scientists to find new and creative ways
to fight against tumor development.
Oxidative damage is considered a potential therapeutic
approach for the development of novel ROS-based anti-
cancer agents. It is very well established that cancer cells
display an altered metabolism with hallmarks such as an
increase in the glucose uptake, increase in lactate synthe-
sis, and an altered redox homeostasis level [1, 2]. In fact,
tumor cells have higher levels of endogenous reactive oxy-
gen species (ROS) than normal cells, and this difference
makes them more vulnerable to ROS-induced injury [3].
Therefore, further oxidative stress induced by exogenous
agents is a strategy to selectively inhibit tumor prolifera-
tion without producing significant toxicity to normal cells
Electronic supplementary material The online version of this
article (https ://doi.org/10.1007/s0077 5-019-01644 -7) contains
supplementary material, which is available to authorized users.
* A. L. Di Virgilio
aldivirgilio@biol.unlp.edu.ar
1
CEQUINOR (CONICET-UNLP), Bv. 120N 1465, LaPlata,
Argentina
2
Facultad de Ciencias Exactas, Universidad Nacional de La
Plata, 47 y 115, 1900LaPlata, Argentina
3
GIAFOT, Departamento de Química, Facultad de Ciencias,
Universidad Nacional de Colombia-Sede Medellín, Medellín,
Colombia

JBIC Journal of Biological Inorganic Chemistry
1 3
[4]. Growing evidence suggests that increased amounts of
ROS can trigger oxidative damage to lipids, proteins, and
DNA. Severe permanent DNA injury leads to a mitotic
catastrophe, which may then be followed by apoptosis or
necrosis [5].
On the other hand, epigenetic control reversibly influ-
ences on the onset and progression of cancer [6]. This
reason led to the development of new drugs that target
histone deacetylases [6]. In fact, it has been reported that
these enzymes may act as oncogenes since they have been
found overexpressed in solid tumors and it is a point of
vulnerability for cancer cells [6]. Indeed, histone deacety-
lases inhibition significantly alters tumor cells, inducing
cell cycle arrest, differentiation, cell death, reduction of
angiogenesis and also can induce an increase in the level
of intracellular oxygen reactive species [7, 8]. Moreover,
it has been highlighted that histone deacetylase inhibitors
provoke genomic instability contributing to the cytotoxic
effects of these drugs [9].
Many quinoline-based drugs that have been used in the
treatment of malaria, arthritis, and lupus, showed to inhibit
histone deacetylase activity [10, 11]. In addition, it has been
demonstrated that quinolines induce DNA damage and apop-
tosis [10] and display antiproliferative activity in invitro and
invivo systems [11, 12] Significant oxidative stress induced
in cells by quinolone derivatives might contribute to the anti-
tumor effect [13, 14].
Previously, it has been reported the synthesis, thermal,
spectral and magnetic studies of metal coordination com-
pounds with 6-methoxyquinoline (6MQ) as ligand [15, 16].
Moreover, the crystal structure of many complexes with
6-methoxyquinoline as ligand and transition metals have
been recently reported by some of us [17]. The synthesis of
these complexes has been undertaken since it is known that
coordination with metals may reinforce therapeutic activity
of the compounds or may allow the acquisition of beneficial
actions. These complexes have shown to improve the anti-
bacterial effect on Gram-positive and Gram-negative bac-
teria after complexation, although nothing is known about
their activity as anticancer drugs [17].
On these bases, we are interested to evaluate if the com-
plexation process of 6-methoxyquinoline with Ag(I), Co(II),
Cu(II) and Zn(II) generates compounds with antitumor activ-
ity for lung carcinoma. Our study was carried out on mon-
olayer and in a multicellular spheroid model of human lung
carcinoma A549 cells, considering cell viability as a starting
point to study, and the mechanisms of action involved in
their antiproliferative effects. We focused our attention on
the role of oxidative stress, and the cytotoxicity and geno-
toxicity actions of Cu(II) and Zn(II) complexes (Cu6MQ and
Zn6MQ) whose formula is M(6MQ)
2
Cl
2
(Fig.1 shows the
crystallographic structure) since these two resulted to be the
most active and to differ from the cation effect.
Materials andmethods
Materials
Tissue culture materials were purchased from Corning
(Princeton, NJ, USA) and APBiotech (Buenos Aires,
Argentina), Dulbeccos modified Eagle medium (DMEM),
TrypLE™ from Gibco (Gaithersburg, MD, USA), and
fetal bovine serum (FBS) from Internegocios SA (Bue-
nos Aires, Argentina). 2,7-Dichlorodihydrofluorescein
diacetate (H
2
DCFDA) and dihydroethidium (DHE) were
obtained from Molecular Probes
®
(Eugene, OR, USA).
Annexin V, Fluorescein isothiocyanate (FITC), propid-
ium iodide (PI) were bought from Invitrogen Corporation
(Buenos Aires, Argentina). Reduced glutathione (GSH),
o-phthalaldehyde (OPT), n-ethylmaleimide (NEM), vita-
min E (α-tocopherol), cytochalasin and the agaroses were
acquired from Sigma Aldrich (St. Louis, MO, USA). Vita-
min C (ascorbic acid) from Merck (Buenos Aires, Argen-
tina). Fluorescein diacetate and Resazurin sodium salt
were purchased from Santa Cruz Biotechnology (Santa
Cruz, CA, USA). A549 (CCL-185) and MRC-5 (CCL-175)
cell lines were purchased from ATCC
®
.
Synthesis oftransition metal complexes
of6‑methoxyquinoline andaqueous stability
Four monomeric complexes of Cu(II), Zn(II), Co(II)
and Ag(I) with 6-methoxyquinoline (6MQ) as ligand
have been prepared and identified according to Villa-
Pérez etal. [17]. The Cu(II) and Zn(II) complexes are
formulated as Cu(6MQ)
2
Cl
2
and Zn(6MQ)
2
Cl
2
; the
Co(II) and Ag(I) compounds are ionic with formulae
[Ag(6MQ)
2
]
+
NO
3
and H(6MQ)
+
[Co(6MQ)Cl
3
]
(where
H(6MQ)
+
is the protonated ligand). Hereafter, the com-
pounds will be referred as Cu6MQ, Zn6MQ, Co6MQ, and
Ag6MQ, respectively.
Aqueous stability in 1000µM solutions of the com-
plexes was measured in phosphate-buffered saline (PBS)
using a Shimadzu UV–Vis spectrophotometer UV-2600
Fig. 1 ORTEP plots of Cu6MQ and Zn6MQ

JBIC Journal of Biological Inorganic Chemistry
1 3
in the range 200–400nm every hour for 24h. To ensure
the stability in biological conditions, 250µM solutions
of Cu6MQ and Zn6MQ were prepared in DMEM and the
spectra were recorded in the range 200–400nm every hour
for 24h as well. The UV spectra were analyzed using the
software SpectraGryph (version 1.2.7, Oberstdorf, Ger-
many), determining the area under the curve in the whole
range, followed by the evaluation of the change of area
compared with the area at time 0.
Cell culture (monolayer andmulticellular spheroids)
The A549 human lung carcinoma (passages 15–35) and
MRC-5 normal lung fibroblasts (passages 5–10) cell lines
were cultured in DMEM supplemented with 10% FBS, 100
U/mL penicillin, and 100µg/mL streptomycin at 37°C in a
humidified atmosphere with 5% of CO
2
. Cells were seeded
in a T75 flask, and when 80–90% of confluence was reached,
cells were subcultured using TrypLE™.
Experiments were carried out in multiwell plates, where
cells were allowed to attach and were washed with DMEM
before each treatment.
A549 carcinoma multicellular spheroids (MCS) were
cultured using the liquid overlay method [18]. Briefly, a 96
wells plate was coated with 50µL 1% (w/v) sterile agarose
in PBS; the gel was allowed to solidify for 20min. 10
4
cells/
mL (150µL) were seeded in each well and incubated at
37°C. Half of the culture medium was replaced with com-
plete fresh medium every other day. On the eighth day, MCS
reached an average diameter between 350 and 400µm and
were suitable to be treated with the complexes [19].
Cell viability assay
Monolayer cell viability was determined using
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-
mide (MTT), which is reduced by mitochondria in viable
cells to a purple formazan dye [20]. Briefly, 2.5 × 10
4
cells
were seeded on 96-well plates and incubated at 37°C. After
24h, cells were exposed to different dilutions of each com-
plex, metallic salt, and ligand for 24, 48 or 72h. Afterward,
the monolayers were washed and incubated with 0.5mg/mL
of MTT in DMEM for 3h. The absorbance of the formazan
extracted with DMSO (100 µL/well) was recorded at a wave-
length of 570nm using a multiplate reader Multiskan FC
(Thermo Scientific). The cell viability is shown graphically
as a percent of the control value (cells treated with DMSO
as vehicle).
To evaluate the role of ROS levels on cell viability, a mix-
ture of 50µM ROS scavengers (vitamin C and E) was simul-
taneously added to the culture medium with the complexes.
After the incubation, the cell viability was determined by the
MTT assay as previously described.
With the goal of achieving a complete outlook of the
harmful effect exerted by the complexes, cell morphology
was also studied. A549 cells were cultured in 6-well plates
(2.5 × 10
5
cells/well), and different concentrations of the
complexes were added for 24h. To observe cell morphologi-
cal changes, the monolayer was fixed with absolute ice-cold
methanol for 5min and stained with Giemsa (1:20 in PBS).
The morphological changes were recorded using an inverted
microscope Olympus BX-51 coupled to a digital camera.
Clonogenic assay
To explore if the compounds affect cell proliferation, a
clonogenic assay was conducted according to [21]. 5 × 10
2
exponential growing A549 cells were plated on 6-well plates
and allowed to attach overnight under standard culture
conditions. The cells were washed twice with sterile PBS
and treated with the complexes for 24h. Next, cells were
washed with PBS twice and incubated with complete culture
medium for 10days. Fixation and the staining process were
conducted with glutaraldehyde 6.0% (v/v) and crystal violet
0.25% (w/v). Colonies formed by more than 50 cells were
recorded for the calculations. The surviving fraction of cells
was plotted versus concentration.
Oxidative stress determination
Reduced (GSH) and oxidized (GSSG) glutathione lev-
els were determined as described by Hissin and Hilf [22].
Confluent A549 monolayer cultured in 24-well plates were
treated with different concentrations of Cu6MQ and Zn6MQ
for 6 and 24h. Then, the monolayer was washed with PBS,
and the cells were lysed with 250µL 0.1% Triton X-100
for 30min at 4°C. For GSH determination, 100µL of the
cellular lysate were added to 1.8mL of ice-cold phosphate
buffer (Na
2
HPO
4
0.1M EDTA 0.005 pH 8.0) and 100µL
o-phthaldialdehyde (OPT) (0.1% in methanol). For the deter-
mination of GSSG, 100µL of the cell lysate were mixed
with 20µL 0.04M of N-ethylmaleimide (NEM) for 20min
at 4°C, then 1.8mL of NaOH 0.1M and 100µL OPT 0.1%
were added. Fluorescence was registered using a fluorometer
Shimadzu RF-6000, the samples were excited at 350nm, and
the emission signal was acquired at 420nm. GSH/GSSG
ratio was calculated as % of the basal for all the experimental
conditions.
Transition metal complexes were tested for reactive oxy-
gen species (ROS) induction as a mechanism of death cell
by flow cytometry. 3 × 10
5
A549 cells were seeded in 12
well plates and incubated overnight. The culture medium
was replaced with different concentrations of the complexes
for 24h. H
2
O
2
0.75mM for 20min was employed as a posi-
tive control. Then, the cellular monolayer was washed with
PBS and detached with Tryple. The cells were centrifuged,

JBIC Journal of Biological Inorganic Chemistry
1 3
and the pellet was incubated with DHE or H
2
DCFDA
(0.8µM) protected from light for 30min. Afterward, cells
were washed twice with PBS, resuspended in 250µl PBS
and transferred to flow cytometry tubes. 2 × 10
4
events were
acquired in FL1 for H
2
DCFDA, or FL2 for DHE using a BD
FACscalibur™ flow cytometer (BD Biosciences, USA) and
further analyses were performed using FlowJo 7.6 software.
Apoptosis
Cells going through different stages of apoptosis were
detected with Annexin V–FITC and propidium iodide (PI)
staining by measuring the externalization of phosphatidyl-
serine (PS) and the cellular membrane integrity, respectively.
Cells exposed to different concentrations of Cu6MQ and
Zn6MQ for 24h were detached using Tryple™ and cen-
trifuged at 2500 RPM for 5min. Afterward, the cellular
pellet was resuspended in 100 µL of binding buffer, and 2
µL of Annexin V-FITC were added, cells were incubated for
20min at room temperature protected from light, and before
de measurement 1µL of PI (50µM) was added. For each
sample, 2 × 10
4
events were analyzed using a BD FACscali-
bur™ flow cytometer (BD Biosciences, USA) and further
analyses were performed using FlowJo 7.6 software.
Cell cycle
DNA content in G1/G0, S, and G2/M phases was analyzed
using flow cytometry. Cells were seeded on 6-well plates and
treated with different concentrations of Cu6MQ and Zn6MQ
for 24h. The harvested cells were washed with PBS, fixed
and permeabilized with 70% ice-cold ethanol for 2h. After-
ward, cells were suspended in 300 µL staining buffer (PBS/
EDTA 2mM, pH 8.0) and 15 µL of RNAse (1mg/mL) and
incubated at 37°C for 15min. Cells were stained with PI (15
µL of a solution 1mg/mL) overnight at 8°C. 10
4
single cells
were analyzed with a BD FACscalibur™ flow cytometer;
histograms depicted the relative DNA distribution within
each sample. The percentage of cells in the G1/G0, S, G2/M
phases and the sub-G1 peak was then calculated using the
cell cycle analysis module in the FlowJo 7.6 software.
Genotoxicity studies
The cytokinesis-block micronucleus (MN) assay was set up
with cultures in the log phase of growth. A549 cells were
seeded onto 6-well plates and incubated at 37°C for 24h.
Then, the cells were treated with different concentrations of
the complexes along with cytochalasin B (4.5μg/mL). After
24h, cells were rinsed and subjected to hypotonic conditions
with 0.075% KCl at 37°C for 5min, fixed with pure metha-
nol at − 20°C for 10min and stained with a 5% Giemsa
solution. For the MN assay, 500 binucleated (BN) cells were
scored at 400× magnification per experimental point from
each experiment. The examination criteria employed were
reported by Fenech [23]. A pulse of 30min of 0.5μg/mL
bleomycin was employed as the positive control.
For detection of DNA damage, the single cell gel elec-
trophoresis assay (Comet assay) was employed based on
the method of Singh etal. [23] with minor modifications.
Briefly, A549 cells were treated with different concentra-
tions of the complexes. After 24h, cells were suspended
in 0.5% low melting point agarose and immediately poured
onto microscope slides precoated with 0.5% normal melting
point agarose. Two slides were prepared for each condition;
one slide was used to observe single-strand DNA breaks and
the other, to obtain information on the presence of oxidized
DNA bases using digestion with the enzyme EndoIII [23].
Slides were immersed in ice-cold lysis solution (2.5M NaCl,
100mM Na2–EDTA, 10mM Trizma–HCl, pH 10 and 1%
Triton X-100, 10% DMSO at 4°C, pH 10) for 1h to lyse the
cells, remove cellular proteins and to allow DNA unfolding.
After that, the slides were washed three times (5min each
time) with enzyme buffer (0.1M KCl, 0.5mM Na
2
–EDTA,
40mM HEPES–KOH, 0.2mg/ml BSA, pH 8.0) and incu-
bated for 45min at 37°C with EndoIII in the enzyme buffer
or with buffer alone. Then, the slides were placed on a hori-
zontal gel electrophoresis tank, and the DNA was allowed
to unwind for 20min in freshly prepared alkaline electro-
phoresis buffer (300mM NaOH and 1mM Na
2
-EDTA, pH
12.7). Electrophoresis was carried out in the same buffer for
30min at 25V (≈ 0.8V/cm across the gels and 300mA)
in an ice bath condition. Afterward, slides were neutralized
and stained with Syber Green. The analysis was performed
in an Olympus BX50 fluorescence microscope. A total of
150 randomly captured cells per experimental point were
used to determine the tail moment using Comet Score ver-
sion 1.5 software. A pulse of 20min of 10μg/mL bleomycin
just before the cells were harvested was employed as the
positive control.
Multicellular spheroids (MCS) viability assay
The spheroid viability was assessed using the resazurin dye,
which is irreversibly reduced by intracellular oxidoreduc-
tases to a pink-red fluorescent dye known as resorufin [24].
The spheroids were cultured as described and incubated with
different concentrations of the complexes for 24 or 48h.
After the exposure, the medium was replaced with 50µM
resazurin solution in DMEM, and the spheroids were incu-
bated overnight at 37°C. Fluorescence was registered using
a fluorometer Shimadzu RF-6000 (excitation at 570nm,
emission at 585nm). Results were corrected by subtraction
of the fluorescence of resazurin and DMEM alone incubated
under the same conditions. Cell viability was plotted as a
percentage of the basal condition (solvent control).

JBIC Journal of Biological Inorganic Chemistry
1 3
Moreover, morphological changes were studied with
a live–death cell staining. Multicellular spheroids treated
with different concentrations of the complexes were incu-
bated for 24 or 48h and stained with fluorescein diacetate
(8 × 10
−3
mg/mL) and propidium iodide (2 × 10
−2
mg/mL).
The spheroids were incubated in the dark for 5min at room
temperature. The fluorescence was registered using an epi-
fluorescence inverted microscope Nikon Ti Eclipse with
FITC and Texas Red filters. The raw images were processed
using ImageJ
®
software, and composite RGB images were
obtained.
Multicellular spheroids spreading assay
To evaluate if the cells in the spheroids can migrate and
proliferate after the exposure to Cu6MQ and Zn6MQ for
24h, the spheroids were transferred into a 96-well plate
containing 150µL of DMEM supplemented with 10% FBS
and were incubated at 37°C in a humidified atmosphere with
5% of CO
2
. The development of outward cellular projec-
tions from the spheroids into the well surface was registered
through phase contrast microscopy after 24 and 72h.
Synergistic calculations
To determine the existence of a synergistic effect between
Cu6MQ and Zn6MQ on A549 cells, the concentration fixed
ratios 1:1, 1:3 and 1:4 of the complexes were tested. Fol-
lowing the same procedure applied in the cell viability assay
(see “Cell viability assay”), and the data were analyzed using
the Chou–Talalay method through the CompuSyn software.
The results are expressed as the combination index (CI):
synergistic effect (CI < 1), additive effect (CI = 1) and antag-
onism effect (CI > 1) [5].
Statistical analysis
Results are expressed as the mean of three independent
experiments and plotted as mean ± standard error of the
mean (SEM). The total number of repeats (n) is specified in
the legends of the figures. The Tukey test (two way ANOVA)
was employed to compare means in all the experiments
performed.
Results
Stability ofthecomplexes
The stability of the complexes was evaluated using UV–Vis
spectroscopy (Fig.1 from Supplementary Material shows
the electronic absorption spectra of Cu6MQ and Zn6MQ
in DMEM and Co6MQ and Ag6MQ in PBS). After 24h
in PBS, all the complexes kept their spectroscopic charac-
teristics and showed a degradation rate below the 10%. The
stability follows: Cu6MQ = Ag6MQ > Zn6MQ> Co6MQ
(Fig.2A from Supplementary Material). Moreover, in bio-
logical conditions, Cu6MQ and Zn6MQ remain stable for
24 h (Fig.2B from Supplementary Material).
Eect of6‑methoxyquinoline complexes oncell
viability
Results from the MTT assay (Fig.2a) in A549 cell line
show that Co6MQ caused no harmful effect on the tumor
cells, and Ag6MQ was the most active antiproliferative
compound. However, despite the remarkable effect of
the silver compound, it did not show a differential cyto-
toxic effect when compared to the cation Ag
+
in the same
range of concentrations (data not shown). On the other
Fig. 2 a Effect of Cu6MQ, Zn6MQ, Co6MQ and Ag6MQ on A549
cell viability. Cells were incubated alone (control) or with differ-
ent concentrations of the compounds at 37 °C for 24h. The results
are expressed as the percentage of the basal level and represent the
mean ± the standard error of the mean (SEM) (N = 9). Asterisks
represent a statistically significant difference in comparison with
the basal level *(p < 0.05) **(p < 0.001). b Differential behavior of
Cu6MQ and Zn6MQ on A549 and MRC-5 cell viability. The results
are expressed as the percentage of the basal level and represent the
mean ± SEM (N = 9). Asterisks represent a statistically significant dif-
ference in comparison with the basal level *(p < 0.05) **(p < 0.001).
Number sign (#) represents a statistically significant difference when
the same complex concentration is evaluated on A549 and MRC-5
cell lines (p < 0.05)

Citations
More filters
Journal ArticleDOI

Enhanced antitumor effect of l-buthionine sulfoximine or ionizing radiation by copper complexes with 2,2´-biquinoline and sulfonamides on A549 2D and 3D lung cancer cell models

TL;DR: Results reveal an antitumor effect of both complexes in monolayer and multicellular spheroids and an improvement with the addition of BSO, however, only SDQCu was the best adjuvant of ionizing radiation treatment.
Journal ArticleDOI

Physicochemical and biological studies of Ni(II), Cu(II) and Zn(II) ternary complexes of sulfaquinoxaline and 2,2’-bipyrimidine.

TL;DR: In this article , three monomeric ternary complexes of Ni(II), Cu(II) and Zn(II)-with sulfaquinoxaline (SQO), and 2,2'-bipyrimidine (bpm) as ligands have been prepared and their crystal structures have been determined.
References
More filters
Journal ArticleDOI

Global cancer statistics

TL;DR: A substantial proportion of the worldwide burden of cancer could be prevented through the application of existing cancer control knowledge and by implementing programs for tobacco control, vaccination, and early detection and treatment, as well as public health campaigns promoting physical activity and a healthier dietary intake.
Journal ArticleDOI

Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays

TL;DR: A tetrazolium salt has been used to develop a quantitative colorimetric assay for mammalian cell survival and proliferation and is used to measure proliferative lymphokines, mitogen stimulations and complement-mediated lysis.
Journal ArticleDOI

Global cancer statistics, 2012

TL;DR: A substantial portion of cancer cases and deaths could be prevented by broadly applying effective prevention measures, such as tobacco control, vaccination, and the use of early detection tests.
Journal ArticleDOI

Ultrastructural Characterization of the Lower Motor System in a Mouse Model of Krabbe Disease.

TL;DR: The data further characterize the ultrastructural analysis of the KD mouse model, and support recent theories of a dying-back mechanism for neuronal degeneration, which is independent of demyelination.
Journal ArticleDOI

A fluorometric method for determination of oxidized and reduced glutathione in tissues.

TL;DR: Results for GSH levels agreed well with earlier reports but levels of GSSG estimated here were higher than earlier reported values, and the reasons for the apparently higher levels ofGSSG are discussed.
Related Papers (5)
Frequently Asked Questions (21)
Q1. What is the effect of zinc on the DNA repair enzymes in mammals?

It is known that copper generates oxygen species that ultimately lead to double ruptures in the DNA and that zinc harms DNA-repair enzymes in mammals potentiating the effect of Cu and triggering apoptosis [58]. 

The aim of this work was to study the antitumor effects and the mechanisms of toxic action of a series of 6-methoxyquinoline ( 6MQ ) complexes in vitro. The authors found that the copper complex, outperformed the Co ( II ), Zn ( II ) and Ag ( I ) complexes with a lower IC50 ( 57. 9 μM ) in A549 cells exposed for 24 h. Cu6MQ decreased cell proliferation and induced oxidative stress detected with H2DCFDA at 40 μM, which reduces GSH/GSSG ratio. 

Cu6MQ resulted to be an interesting candidate for further in vivo studies. 

The relationship between dissolved copper and MN frequency is supported by oxidative-stress mechanisms, and more particularly by the production of reactive oxygen species, which attack DNA on the sugar residue and induce base loss and strand breaks [49]. 

histone deacetylases inhibition significantly alters tumor cells, inducing cell cycle arrest, differentiation, cell death, reduction of angiogenesis and also can induce an increase in the level of intracellular oxygen reactive species [7, 8]. 

Surgery is the most recommended treatment for patients in an early-stage followed by thoracic radiotherapy and chemotherapy [28]. 

For each sample, 2 × 104 events were analyzed using a BD FACscalibur™ flow cytometer (BD Biosciences, USA) and further analyses were performed using FlowJo 7.6 software. 

Zn6MQ produced a live cell staining up to 400 µM and very little staining with propidium iodide in accordance with the MCS viability assay. 

Cu6MQ at 50 and 100 µM induced an accumulation of cells in the G2/M phase, 23.2% and 21.5%, respectively, (p < 0.001), the increase of events at this phase was at expense of the G1 population which was reduced 15% in average. 

Cu6MQ impaired MCS viability from 100 µM in a 24 h treatment showing a concentration-dependent manner and from 50 µM when doubling the exposure time (p < 0.001). 

According to their findings, several copper complexes with promising anticancer activity displayed remarkable effects against spheroids and tumor xenografts in vivo in a murine model [38] and anti-metastatic properties by inhibiting the migratory and invasive ability of cancer cells [56]. 

It has been shown that Zn cation induces micronuclei in human leucocytes in the same range of concentrations and not in a dose-dependent manner [51]. 

In accordance with the morphological features, Cu6MQ could be established as the most promising candidate since it reduces tumor cell viability affecting non-tumor cells less severely. 

the fraction of necrotic cells followed a concentration-dependent increase, i.e., 16.7, 53.3 and 76.0% for 150, 250 and 300 µM, respectively. 

As the authors expected, both compounds showed higher IC50 in the MCS model than in the monolayer cell model, with a correlation with the proportion of live–dead cells and with the inhibition of the spreading. 

at 24 h the IC50 for Zn6MQ on spheroids doubled the IC50 in the 2D model, while for Cu6MQ the IC50 on 3D raised three times the IC50 found in the cellular monolayer. 

These complexes have shown to improve the antibacterial effect on Gram-positive and Gram-negative bacteria after complexation, although nothing is known about their activity as anticancer drugs [17]. 

it has been demonstrated that micronuclei can be induced by chemicals that are known to cause DNA replication stress and S phase arrest [54]. 

In fact, tumor cells have higher levels of endogenous reactive oxygen species (ROS) than normal cells, and this difference makes them more vulnerable to ROS-induced injury [3]. 

Their results reveal that a ratio 1:1 of both Cu(II) and Zn(II) complexes in the monolayer and the 3D model offers the best synergistic effect as a novel strategy for anti-cancer therapy. 

Two slides were prepared for each condition; one slide was used to observe single-strand DNA breaks and the other, to obtain information on the presence of oxidized DNA bases using digestion with the enzyme EndoIII [23].