Sie sind auf Seite 1von 12

Polyhedron 156 (2018) 320–331

Contents lists available at ScienceDirect

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

The induction of apoptosis in BEL-7402 cells by an iridium(III) complex


through lysosome–mitochondria pathway
Yang-Jie Wang a, Qiao-Yan Yi a, Wen-Yao Zhang a, Fan Du a, Miao He a, Yun-Jun Liu a,b,⇑
a
School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
b
Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou 510006, PR China

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

Article history: A new iridium(III) polypyridyl complex [Ir(ppy)2(DPBD)](PF6) (ppy = 2-phenylpyridine, DPBD = 4-(dipyr-
Received 4 September 2018 ido[3,2-a:20 ,30 -c]phenazin-11-yl)benzene-1,2-diamine, Ir-1) was synthesized and characterized by ele-
Accepted 19 September 2018 ment analysis, ESI-MS, IR, 1H NMR and 13C NMR. The cytotoxicity in vitro of the complex against
Available online 28 September 2018
cancer BEL-7402, A549, Eca-109 and normal LO2 cells was evaluated by 3-(4,5-dimethylthiazole)-2,5-
diphenyltetraazolium bromide (MTT) method. The IC50 values of the complex toward BEL-7402, A549
Keywords: and Eca-109 are 13.8 ± 0.7, 14.5 ± 0.6 and 15.2 ± 2.4 mM, respectively. The apoptosis was studied with
Iridium(III) complex
acridine orange (AO) and ethidium bromide (EB) staining method and comet assay. The reactive oxygen
Apoptosis
Intracellular Ca2+ levels
species including superoxide anion and NO, the location of the complex at lysosomes, lysosomal mem-
Lysosomes brane permeabilization, location of the complex at mitochondria and the change of mitochondrial mem-
Bcl-2 family proteins brane potential were assayed under a fluorescent microscope. The complex can enhance the intracellular
Ca2+ levels and induce a release of cytochrome c. In addition, the complex can cause autophagy and
upregulate the expression of LC-3 and Beclin-1 proteins. The complex inhibits the cell growth at S phase.
The effects of the complex on the expression of caspase 3 and Bcl-2 family proteins were explored. The
results show that the complex induces apoptosis in BEL-7402 cells through lysosome–mitochondrial dys-
function pathway, which was accompanied by the regulation of Bcl-2 family proteins.
Ó 2018 Elsevier Ltd. All rights reserved.

1. Introduction attention. In recent years, organometallic iridium complexes have


emerged as potential candidates for new metallo-anticancer
One of the features of cancer is the rapid generation of abnor- drugs, a number of iridium(III) complexes have shown interesting
mal cells that grow beyond their usual boundaries, which can then biological effect [17–23]. Complex [Ir(ppy)2(FBPIP)]+ (ppy = 2-phe-
invade adjoining parts of the body and spread to other organs. Plat- nylpyridine, FBPIP = 2-(4-formyl)benzeno[4,5-f][1,9]phenanthro-
inum based drugs are effectively used in the treatment of various line) showed no cytotoxic activity against SGC-7901 cells.
types of cancer [1–4]. Unfortunately, cisplatin exhibits some draw- However, upon irradiation, the complex displayed potent anti-pro-
backs including severe side effects [5], drug resistance [2] and low liferative activity toward SGC-7901 cells, with a low IC50 value of
water-solubility [6], which seriously limit its clinical applications. 6.1 ± 0.6 mM, and showed selectivity between cancer and normal
These limitations have motivated to search for other transition cells [24]. Complex [Ir(ppy)2(BTCP)]+ (BTCP = 2-bicyclo[2.2.1]
metal complexes with beneficial biological properties, wide reac- hept-5-en-yl-1H-1,3,7,8-tetraazacyclopenta[l]phenanthrene) inhi-
tivity ranges, and lower systemic toxicities [7–9]. Among these bits the cell growth at G0/G1 phase in SGC-7901 cells. The complex
metal complexes, anticancer activity of ruthenium complexes has induces apoptosis through a ROS-mediated mitochondrial dysfunc-
made great achievement [10–15]. On the other hand, cyclometa- tion pathway [25]. Mao et al. reported that iridium(III) complex [Ir
lated iridium(III) complexes have rich photophysical properties, (dfppy)2(4,40 -bis(chloromethyl)-2,20 -bipyridine)]+ (dfppy = 2-(2,4-
e.g., high quantum yields, large Stokes shifts, long lived lumines- difluorophenyl)pyridine) induced caspase-dependent apoptosis
cence, good photostability and cell permeability [16], the anti- through mitochondrial damage, cellular ATP depletion, mitochon-
cancer activity of iridium(III) complexes have also attracted great drial respiration inhibition and reactive oxygen species (ROS)
elevation [16]. To obtain more information on anticancer activity
of iridium(III) complex, in this paper, a new iridium(III) complex
⇑ Corresponding author at: School of Pharmacy, Guangdong Pharmaceutical
[Ir(ppy)2(DPBD)](PF6) (ppy = 2-phenylpyridine, DPBD = 4-(dipyr-
University, Guangzhou 510006, PR China.
E-mail address: lyjche@gdpu.edu.cn (Y.-J. Liu). ido[3,2-a:20 ,30 -c]phenazin-11-yl)benzene-1,2-diamine, Scheme 1)

https://doi.org/10.1016/j.poly.2018.09.057
0277-5387/Ó 2018 Elsevier Ltd. All rights reserved.
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 321

Scheme 1. Synthetic route of complex [Ir(ppy)2(DPBD)]PF6.

was synthesized and characterized by elemental analysis, ESI-MS, 1583m, 1532m, 1492s, 1479s, 1438m, 1416s, 1358s, 1316s,
IR, 1H NMR and 13C NMR. The cytotoxicity in vitro of the complex 1268m, 1164m, 1079m, 843s, 758s, 731m, 557s. 1H NMR (DMSO-
against BEL-7402, A549, Eca-109 and normal LO2 cells was inves- d6): 9.54 (d, 1Ha, J = 8.0 Hz), 9.49 (d, 1Hj, J = 8.0 Hz), 8.33–8.27
tigated by MTT method. The apoptosis, comet assay, reactive oxy- (m, 4H3,14,b,i), 8.24 (d, 2Hc,h, J = 5.0 Hz), 8.12–8.05 (m, 2H8,19),
gen species (ROS), mitochondrial membrane potential, 7.97 (d, 2H1,12, J = 8.0 Hz), 7.90 (d, 3Hn,7,18, J = 7.0 Hz), 7.70 (d,
intracellular Ca2+ levels, release of cytochrome c, cell cycle arrest, 2H4,15, J = 5.5 Hz), 7.68 (d, 1Hl, J = 6.0 Hz), 7.26 (d, 1Hk, J = 5.0 Hz),
cell invasion and Bcl-2 family proteins expression were studied 7.06 (d, 4H9,10,20,21, J = 8.0 Hz), 6.97 (t, 2H2,13, J = 6.0 Hz), 6.70 (d,
in detail. 1Hp, J = 8.0 Hz), 6.30 (dd, 2Hs,t, J = 7.0, J = 7.5 Hz), 3.47 (s, 4HNH2).
13
C NMR (DMSO-d6, 125 MHz): 166.84 (Ca,j), 151.67 (C1), 151.34
2. Experimental (C12), 149.73 (C7), 149.61 (C18), 149.29 (Cw), 148.89 (Cx), 144.59
(C5), 144.06 (C6), 142.82 (Cq), 141.08 (Cr), 139.97 (Cm), 138.87
2.1. Materials and methods (Cb), 138.40 (Ci), 137.29 (Co), 134.95 (C3), 134.65 (C14), 131.52
(Cc), 131.22 (Ch), 130.69 (Cd), 130.58 (Cg), 130.36 (Ce,f), 129.37
All reagents and solvents were purchased commercially and (C4,15), 128.37 (Ck), 126.14 (Cl), 125.16 (C9,20), 123.90 (C10,21),
used without further purification unless otherwise noted. Ultra- 122.56 (C2), 121.98 (C13), 120.06 (Cn), 117.87 (Ct), 115.02 (Cs),
pure MilliQ water was used in all experiments. DMSO and RPMI 113.34 (Cp). ESI-MS (CH3CN): m/z 889.3 ([MPF6]+).
1640 were purchased from Sigma. 1,10-Phenanthroline was
obtained from the Guangzhou Chemical Reagent Factory. Cancer 2.3. Cytotoxic activity assay in vitro
cell lines of BEL-7402 (human hepatocellular carcinoma), A549
(human lung carcinoma), Eca-109 (human esophagus cancer cell 3-(4,5-Dimethylthiazole)-2,5-diphenyltetraazolium bromide
line) and normal cell LO2 (human liver cell) were purchased from (MTT) method [28] was used to evaluate the cytotoxicity in vitro
the American Type Culture Company. IrCl33H2O was purchased of the complex against cancer cells. Cells were placed in 96-well
from the Kunming Institution of Precious Metals. microassay culture plates (8  103 cells per well) and grown over-
Microanalysis (C, H, and N) was carried out with a Perkin-Elmer night at 37 °C in a 5% CO2 incubator. The tested complexes were
240Q elemental analyzer. Electrospray ionization mass spectra dissolved in DMSO and then added to the wells to achieve final
(ESI-MS) were recorded on a LCQ system (Finnigan MAT, USA) concentrations ranging from 106 to 104 M (final concentration
using acetonitrile as mobile phase. The spray voltage, tube lens off- of DMSO of 0.05%, v/v). Negative control wells were prepared by
set, capillary voltage and capillary temperature were set at 4.50 kv, addition of culture medium (100 lL). The plates were incubated
30.00 V, 23.00 V and 200 °C, respectively, and the quoted m/z val- at 37 °C in a 5% CO2 incubator for 48 h. Upon completion of the
ues are for the major peaks in the isotope distribution. 1H NMR incubation, stock MTT dye solution (20 lL, 5 mg/mL1) was added
and 13C NMR spectra were recorded on a Varian-500 spectrometer to each well. After 4 h, buffer (100 lL) containing dimethylfor-
with DMSO-d6 as solvent and tetramethylsilane (TMS) as an inter- mamide (50%) and sodium dodecyl sulfate (20%) was added to sol-
nal standard at 500 MHz at room temperature. UV–Vis spectra ubilize the MTT formazan. The optical density of each well was
were recorded on a Shimadzu UV-3101PC spectrophotometer measured with a microplate spectrophotometer at a wavelength
and emission spectra were recorded on a Shimadzu RF-4500 lumi- of 490 nm. The 50% inhibitory concentration (IC50) is defined as
nescence spectrometer at room temperature. the concentration to reduce the size of the cell population by
50%. The IC50 values were determined by the percentage of cell via-
2.2. Synthesis of [Ir(ppy)2(DPBD)](PF6) (Ir-1) bility versus concentration on a logarithmic graph and reading off
the concentration at which 50% of cells remain viable relative to
A mixture of cis-[Ir(ppy)2Cl]2 (0.15 g, 0.14 mmol) [26] and DPBD the control. Each experiment was repeated at least three times to
(0.11 g, 0.28 mmol) [27] in a mixture of 42 mL dichloromethane obtain the mean values.
and methanol (VCH2Cl2:VCH3OH = 2:1) was refluxed under argon for
6 h to give a clear red brown solution. Upon cooling, a brown red 2.4. Apoptosis assay by AO/EB staining method
precipitate was obtained by dropwise addition of saturated aque-
ous NH4PF6 solution with stirring at room temperature for 2 h. BEL-7402 cells were seeded onto chamber slides in six-well
The crude product was purified by column chromatography on plates at a density of 2  105 cells per well and incubated for
neutral alumina with a mixture of CH2Cl2–acetone (1:3, v/v) as elu- 24 h. The cells were cultured in RPMI (Roswell Park Memorial
ent. The red band was collected. The solvent was removed under Institute) 1640 supplemented with 10% of fetal bovine serum
reduced pressure and a red powder was obtained. Yield: 76%. Anal. (FBS) and incubated at 37 °C in 5% CO2. The medium was removed
Calc. for C46H32F6N8PIr: C, 53.37; H, 3.12; N, 10.83. Found: C, 53.48; and replaced with medium (final DMSO concentration, 0.05% v/v)
H, 3.34; N, 10.68%. IR (KBr, cm1): 3367m, 3043w, 2038w, 1607m, containing the complex (3.75 and 7.5 lM) for 24 h. The medium
322 Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331

was removed again, and the cells were washed with ice-cold phos- again. The fluorescent dye 20 ,70 -dichlorodihydrofluorescein diac-
phate buffer saline (PBS), and fixed with formalin (4%, w/v). Cell etate (DCHF-DA, 10 lM) was added to the medium to cover the
nuclei were counterstained with acridine orange (AO) and ethid- cells. The treated cells were then washed with cold PBS–EDTA
ium bromide (EB) (AO: 100 mg/mL, EB: 100 mg/mL) for 10 min. twice, collected by trypsinization and centrifugation at 1500 rpm
The cells were observed and imaged under a fluorescence micro- for 5 min, the cell pellets were suspended in PBS–EDTA and imaged
scope (Nikon, Yokohama, Japan) with excitation at 350 nm and under a fluorescent microscope.
emission at 460 nm.
2.8. Localization assay of Ir-1 in the lysosomes
2.5. DNA damage assay
BEL-7402 cells in 12-well plates were cultured overnight. The
DNA damage was investigated by means of comet assay. BEL- cells were treated with the complex (15.0 mM) for 5 h. The lyso-
7402 cells in culture medium were incubated with 3.75 and somes were labeled and tracked using the lysosomotropic probe
7.5 lM of complex for 24 h at 37 °C. The cells were harvested by Lyso Tracker Red (LTR) (50 nM) for 30 min in dark. After the cells
a trypsinization process at 12 and 24 h. A total of 100 lL of 0.5% were washed with PBS twice, the stained cells were analyzed
normal agarose in PBS was dropped gently onto a fully frosted under a fluorescence microscope.
microslide, covered immediately with a coverslip, and then placed
at 4 °C for 10 min. The coverslip was removed after the gel had
2.9. Lysosomal membrane permeability assay
been set. A mixture of 50 lL of the cell suspension (200 cells/lL)
mixed with 50 lL of 1% low melting agarose was preserved at
BEL-7402 cells in 12-well plates were cultured overnight. The
37 °C. A total of 100 lL of this mixture was applied quickly on
cells were treated with the complex (15.0 mM) for 1, 2 and 5 h.
top of the gel, coated over the microslide, covered immediately
After the cells were washed with PBS twice, the cells were stained
with a coverslip, and then placed at 4 °C for 10 min. The coverslip
with acridine orange (AO, 1 mg/mL) for 30 min in dark. The stained
was again removed after the gel had been set. A third coating of
cells were imaged under a fluorescence microscope.
50 lL of 0.5% low melting agarose was placed on the gel and
allowed to place at 4 °C for 15 min. After solidification of the agar-
2.10. Localization assay of the complex in the mitochondria
ose, the coverslips were removed, and the slides were immersed in
an ice-cold lysis solution (2.5 M NaCl, 100 mM EDTA, 10 mM Tris,
BEL-7402 cells were placed in 12-well microassay culture plates
90 mM sodium sarcosinate, NaOH, pH 10, 1% Triton X-100 and
(4  104 cells per well) and grown overnight at 37 °C in a 5% CO2
10% DMSO) and placed in a refrigerator at 4 °C for 2 h. All of the
incubator. 7.5 lM of Ir-1 was added to the wells at 37 °C in a 5%
above operations were performed under low lighting conditions
CO2 incubator for 8 h and further co-incubated with MitoTracker
to avoid additional DNA damage. The slides, after removal from
Deep Red FM (150 nM) at 37 °C for 30 min. Upon completion of
the lysis solution, were placed horizontally in an electrophoresis
the incubation, the wells were washed three times with ice-cold
chamber. The reservoirs were filled with an electrophoresis buffer
PBS. After discarding the culture medium, the cells were imaged
(300 mM NaOH, 1.2 mM EDTA) until the slides were just immersed
under a fluorescence microscope.
in it, and the DNA was allowed to unwind for 30 min in elec-
trophoresis solution. Then the electrophoresis was carried out at
25 V and 300 mA for 20 min. After electrophoresis, the slides were 2.11. Mitochondrial membrane potential assay
removed, washed thrice in a neutralization buffer (400 mM Tris,
HCl, pH 7.5). Cells were stained with 20 lL of EB (20 lgmL1) in BEL-7402 cells were treated for 24 h with different concentra-
the dark for 20 min. The slides were washed in chilled distilled tion of the complex in 12-well plates and were then washed three
water for 10 min to neutralize the excess alkali, air-dried and times with cold PBS. The cells were detached with trypsin–EDTA
scored for comets by fluorescent microscopy. solution. The collected cells were incubated for 20 min with
1 lg/mL of JC-1 (5,50 ,6,60 -Tetrachloro-1,10 ,3,30 -tetraethyl-imidacar-
2.6. Cell cycle arrest by flow cytometry bocyanine iodide) in culture medium at 37 °C in the dark.
Cells were immediately centrifuged to remove the supernatant.
BEL-7402 cells were seeded into six-well plates (Costar, Corning Cell pellets were suspended in PBS and imaged under fluorescence
Corp, New York) at a density of 2  105 cells per well and incubated microscope. The ratio of red/green fluorescence intensity was
for 24 h. The cells were cultured in RPMI 1640 supplemented with determined by flow cytometry.
10% of FBS and incubated at 37 °C in 5% CO2. The medium was
removed and replaced with medium (final DMSO concentration, 2.12. Detection of Ca2+ levels
0.05% v/v) containing complex (7.5 or 15.0 lM). After incubation
for 24 h, the cell layer was trypsinized and washed with cold PBS BEL-7402 cells treated with 15.0 mM of Ir-1 for 2, 4 and 8 h were
and fixed with 70% ethanol. Twenty mL of RNAse (0.2 mg/mL) and incubated with 2.5 lM Fluo-3AM (which can cross the cell mem-
20 mL of propidium iodide (0.02 mg/mL) were added to the cell sus- brane and be cut into Fluo-3 by intracellular esterase) for 30 min
pensions and they were incubated at 37 °C for 30 min. Then the at 37 °C, the cells were washed three times and incubated for
samples were analyzed with a FACSCalibur flow cytometry. The 20 min at 37 °C to ensure that Fluo-3AM (which can specifically
number of cells analyzed for each sample was 10 000. bind to Ca2+ and has a strong fluorescence with an excitation wave-
length of 488 nm) has been completely transformed into Fluo-3 in
2.7. Reactive oxygen species (ROS) detection the cells. Then, the cells were washed two times with PBS and
stained with 5 lgmL1 DAPI solution. Finally, ImageXpress Micro
BEL-7402 cells were seeded into six-well plates at a density of XLS system was used to observe fluorescence, and Multi Wave-
2  105 cells per well and incubated for 24 h. The cells were cul- length Cell Scoring module was used to analyze the data. The inte-
tured in RPMI 1640 supplemented with 10% of FBS and incubated grated intensity/cell which represented the fluorescence intensity
at 37 °C in 5% CO2. The medium was removed and replaced with of each cell was used to measure the levels of Ca2+. The fluores-
medium (final DMSO concentration, 0.05% v/v) containing different cence intensity of each cell was calculated as the total fluorescence
concentration of complex for 24 h. The medium was removed intensity divided by the number of cells.
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 323

2.13. Examination release of cytochrome c by immunofluorescence counted under a light microscope. The mean values from three
independent assays were calculated.
BEL-7402 cells were seeded in a 12-well plate and incubated
overnight. Then cells were treated with 15.0 lM of Ir-1 for 24 h. 2.17. The expression of caspases 3, 8 and Bcl-2 family proteins
Subsequently, the cells were fixed with ice-cold immunol staining
fix solution for 30 min at room temperature. After blocking cells BEL-7402 cells were seeded in 3.5 cm dishes for 24 h and incu-
with immunol staining blocking buffer for 1 h, the cells were trea- bated with 15.0 lM of the complex in the presence of 10% FBS. The
ted with the primary antibody against cytochrome c (1:50 dilution) cells were harvested in lysis buffer. After sonication, the samples
overnight at 4 °C. Next, the plate was washed three times with were centrifuged for 20 min at 13,000 g. The protein concentration
immunol staining wash buffer and probed with Alexa Fluor 488- of the supernatant was determined by BCA (bicinchoninic acid)
Labeled Goat Anti-Mouse IgG (1:500 dilution) in the dark for 1 h assay. Sodium dodecyl sulfate–polyacrylamide gel electrophoresis
at room temperature. Finally, the cells were washed with immunol was done loading equal amount of proteins per lane. Gels were
staining wash buffer three times and the cell nuclei were stained then transferred to poly (vinylidene difluoride) membranes (Milli-
with DAPI. The images were obtained using ImageXpress Micro pore) and blocked with 5% non-fat milk in TBST (20 mM Tris–HCl,
XLS system, and Multi Wavelength Cell Scoring module was used 150 mM NaCl, 0.05% Tween 20, pH 8.0) buffer for 1 h. Then the
to analyze the data. The integrated intensity/cell which represents membranes were incubated with primary antibodies at 1:5,000
the fluorescence intensity of each cell was used to measure the dilutions in 5% non-fat milk overnight at 4 °C, and washed four
release of cyto-c. The fluorescence intensity of each cell was calcu- times with TBST for a total of 30 min. After which the secondary
lated as the total fluorescence intensity divided by the number of antibodies conjugated with horseradish peroxidase at 1:5000 dilu-
cells. tion for 1 h at room temperature and then washed four times with
TBST. The blots were visualized with the Amersham ECL Plus Wes-
2.14. Autophagy induced by complex tern blotting detection reagents according to the manufacturer’s
instructions. To assess the presence of comparable amount of pro-
BEL-7402 cells were seeded onto chamber slides in 12-well teins in each lane, the membranes were stripped finally to detect
plates and incubated for 24 h. The cells were cultured in RPMI the b-actin. The gray values were calculated with BandScan.
1640 supplemented with 10% of FBS and incubated at 37 °C in 5%
CO2. The medium was removed and replaced with medium (final
2.18. Data analysis
DMSO concentration, 0.05% v/v) containing different concentration
of complex for 24 h. The medium was removed again, and the cells
All data were expressed as mean ± SD. Statistical significance
were washed with ice-cold PBS twice. Then the cells were stained
was evaluated using t-tests. Differences were considered to be sig-
with MDC (monodansylcadaverine) solution (50 mM) for 10 min
nificant when the *P value was less than 0.05.
and washed with PBS twice. The cells were observed and imaged
under fluorescence microscope. The effect of the complex on the
expression of LC3 and Beclin-1 proteins was assayed by Western 3. Results and discussion
blot.
3.1. Synthesis and characterization
2.15. The effect of autophagy on cell viability
Ligand DPBD was prepared according to the literature [27]. The
Cell viability was studied using the MTT method. Cells were complex Ir-1 was synthesized by the reaction of cis-[Ir(ppy)2Cl]2
placed in 96-well microassay culture plates (8  104 cells per well) with DPBD in a mixture of dichloromethane and methanol. The
and cultured overnight at 37 °C in a 5% CO2 incubator. The cells crude product was purified by column chromatography on neutral
were pretreated with or without 3-methyladenine (3-MA, 3 mM) alumina. The obtained complex was characterized by elemental
for 3 h, followed by different concentration of Ir-1 for 24 h. After analysis, IR, ESI-MS, 1H NMR and 13C NMR. In the IR spectra, the
incubation, cells were incubated with MTT (0.5 mg/mL) for 4 h at peaks of 3367 cm1 and 3043 cm1 for Ir-1 are assigned the
37 °C. Upon completion of the incubation, 100 lL DMSO was added NAH and CAH stretching vibration. In the ESI-MS spectra, the peak
to solubilize the MTT formazan. The optical density of each well at a m/z value of 889.3 is assigned to the ion peak of [M-PF6]+. In
was then measured with a microplate spectrophotometer at a the 1H NMR spectra of complex, the single peak of 3.47 (s, 4H)
wavelength of 490 nm. The viability (%) of cell growth was calcu- for ANH2 group was observed. The UV–Vis and luminescence of
lated by the formula: (A490 (treatment group)/A490 (control))  100, A490 Ir-1 (5 mM) in PBS solution is shown in Fig. 1a, the maximum
(treatment group) is the mean OD value of cells treated with the various
absorbance of Ir-1 appears at 204 nm, and the complex can emit
ruthenium complexes and A490 (control) is the mean OD value of luminescence in PBS solution at ambient temperature, with a max-
untreated cells. Each experiment was repeated at least three times imum appearing at 576 nm (Fig. 1b). The stability of the complex in
to obtain the mean values. PBS solution was investigated by UV–Vis spectra at 0 and 24 h, no
obvious changes in the absorbance was observed (data no present),
2.16. Matrigel invasion assay indicating that the complex is stable in PBS solution.

The BD Matrigel invasion chamber was used to investigate the 3.2. Cytotoxic activity in vitro
cell invasion according to the manufacturer’s instructions. BEL-
7402 cells (4  104) in serum free media and different concentra- The cell viability of BEL-7402, A549, Eca-109 and normal LO2
tion of the complex were seeded in the top chamber of the two cell induced by the different concentration of complex Ir-1 and
chamber Matrigel system. RPMI-1640 (20% FBS) was added as ligand for 48 h was assayed by MTT method. Cisplatin was used
chemo-attractant into the lower chamber. Cells were allowed to as a positive control. The IC50 values are listed in Table 1. As we
invade for 24 h. After incubation, non-invading cells were removed expect, ligand DPBD has no cytotoxic activity against the selected
from the upper surface and cells on the lower surface were fixed cell lines. However, when ligand bonded metal to form complex,
with 4% paraformaldehyde and stained with 0.1% of crystal violet. the cytotoxicities in vitro are enhanced. The complex shows high
The membranes were photographed and the invading cells were cytotoxic activity toward BEL-7402, A549 and Eca-109 with an
324 Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331

Fig. 1. UV–Vis (a) and luminescence spectra (b) of 5 mM of the complex in PBS solution at room temperature.

Table 1 3.4. DNA damage studies


IC50 values of the complex Ir-1 toward selected cell lines.

Complex BEL-7402 A549 Eca-109 LO2 The apoptosis can also be confirmed by DNA damage. The DNA
QPBD >200 >200 >200 >200
damage was studied by comet assay through the single cell gel
Ir-1 13.8 ± 0.7 14.5 ± 0.6 15.2 ± 2.4 25.0 ± 2.7 electrophoresis. DNA fragmentation is a hallmark of apoptosis,
Cisplatin 11.5 ± 1.3 7.5 ± 1.3 – 9.2 ± 1.4 mitotic catastrophe or both [29,30]. As shown in Fig. 3, in the con-
trol (a), the cells fail to show any comet-like appearance. Exposure
of BEL-7402 cells to 3.75 (b) and 7.5 mM (c) of Ir-1 for 24 h caused a

IC50 value of 13.8 ± 0.7, 14.5 ± 0.6 and 15.2 ± 2.4 mM, respectively.
Interestingly, the complex exhibits relatively low cytotoxic activity
against normal LO2 cells. Comparing the IC50 values, we find that
the complex displays lower inhibitory effect against BEL-7402,
A549 and Eca-109 cell growth than cisplatin and [Ir(ppy)2(BTCP)]+
[25] under the same conditions. Because BEL-7402 cells are sensi-
tive to the complex, this cell was selected for undergoing the fol-
lowing experiments.

3.3. Apoptosis studies with AO/EB methods Fig. 3. Comet assay of BEL-7402 (a) exposure to 3.75 (b) and 7.5 (c) mM of Ir-1 for
24 h. The scale bars for all the figures are 200 .

Apoptosis known as a programmed cell death is a fundamental


and inherent biological event for maintaining the balance between
cellular proliferation and death, evasion of apoptosis constitutes a
characteristic feature of human cancers. The morphological
changes of BEL-7402 cells treated by the complex were investi-
gated with acridine orange (AO)/ethidium bromide (EB) double
staining method. It is well known that AO stained viable and apop-
totic cells and emits green fluorescence, EB stained necrotic cells
and emits red fluorescence. As shown in Fig. 2, in the control (a),
the living cells with intact cell nuclei show bright green fluores-
cence. After BEL-7402 cells were incubated with 3.75 (b) and
7.5 mM (c) Ir-1 for 24 h, the apoptotic cells with apoptotic features
such as blebbing, nuclear shrinkage and chromatin condensation
were found, simultaneously, the red necrotic cells were also
observed. The results indicate that complex Ir-1 can induce apop-
tosis in BEL-7402 cells.

Fig. 2. Apoptosis assays of BEL-7402 cells (a) exposure to 3.75 (b) and 7.5 mM (c) of Fig. 4. (A) The cell cycle distribution of BEL-7402 cells (black) exposure to 7.5 (red)
Ir-1 for 24 h and the cell nuclei were stained with AO/EB. The scale bars for all the and 15.0 mM (blue) of Ir-1 for 24 h. (B) The expression of proteins related to the cell
figures are 200 . cycle induced by different concentrations of the complex for 24 h. (Colour online.)
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 325

considerable strand breakage on chromosomal DNA and led to the ent structure of DBPD and BTCP and different cancer cells BEL-7402
appearance of an obscure ‘‘halo” around the nucleus of the BEL- and SGC-7901. The expression of the proteins CDK2 and Cyclin A2
7402 cells. The statistically significant and well-formed comets related to the S phase was assayed by western blot. As shown in
were observed which indicated severe DNA damage. The results Fig. 4B, treatment of BEL-7402 cells with different concentration
of the comet assay suggest that the DNA of a single cell underwent of Ir-1 for 24 h led to a down-regulation in the expression of
degradation as a consequence of direct DNA damage or rapid apop- CDK2 and Cyclin A2 proteins, which further confirms the complex
tosis. Similar results were observed in other metal complexes induces cell cycle arrest at S phase in BEL-7402 cells.
[13,31,32].
3.6. Intracellular reactive oxygen species levels studies
3.5. Cell cycle distribution detection
Many potential anticancer and chemopreventive agents induce
Cell cycle includes three phases: G0/G1, S and G2/M. The drugs apoptosis through mediated-ROS generation [33–35]. To detect
inhibit cancer cell growth through inducing cell cycle arrest at G0/ whether reactive oxygen species (ROS) are involved in apoptotic
G1 or S or G2/M phase. The distribution of BEL-7402 cells in vari- mechanism caused by the complex, intracellular generation of
ous compartments during the cell cycle was analyzed by flow ROS was investigated using DCHF-DA as fluorescence probe.
cytometry in cells stained with propidium iodide. As shown in Finally, this dye is oxidized by intracellular ROS to the highly fluo-
Fig. 4A, in the control, the percentage in the cells at S is 11.06%. rescent compound, namely, 20 ,70 -dichlorofluorescein (DCF). The
Treatment of BEL-7402 cells with 7.5 and 15.0 mM of Ir-1 led to a fluorescent intensity of DCF is proportional to intracellular ROS
large increase of 6.95% and 12.24% in the cells at S phase, which levels. As shown in Fig. 5A, in the control (a) or control + NAC (b,
was accompanied by a relative reduction of 5.53% and 22.75% in 5 mM), no obvious fluorescent points are observed. However, the
the cells at G2/M phase, respectively. These data reveal that the BEL-7402 cells were treated with 3.75 mM Ir-1 (c), 3.75 mM Ir-1
complex inhibits the cell growth of BEL-7402 cells at S phase. In + NAC (d), 7.5 mM Ir-1 (e) and 7.5 mM Ir-1 + NAC (f) for 24 h, a num-
our previous work [25], complex [Ir(ppy)2(BTCP)]PF6 inhibited ber of bright fluorescent points are found, which indicates that the
the cell growth at G0/G1 phase against SGC-7901 cells. Obviously, complex can increase intracellular ROS levels. In the present of
complex [Ir(ppy)2(DPBD)]PF6 and [Ir(ppy)2(BTCP)]PF6 inhibit the NAC, DCF fluorescent intensity decreases. This shows that NAC
cell growth at the different phase. This may be caused by the differ- inhibits the generation of ROS. To quantitatively observe the effect

Fig. 5. (A) Intracellular ROS was detected in BEL-7402 cells (a) exposure to NAC (b), Rosup (c, positive control) and different concentrations of the complex for 24 h, (d)
3.75 mM Ir-1; (e) 3.75 mM Ir-1 + NAC; (f) 7.5 mM Ir-1; (g) 7.5 mM Ir-1 + NAC. (B) The DCF fluorescent intensity was determined after BEL-7402 cells were treated with NAC and
different concentration of Ir-1 for 24 h by ImageXpress Micro XLS system. (C) The superoxide anion level was assayed after BEL-7402 cells (a) were exposed to 3.75 (b) and
7.5 mM (c) of Ir-1 for 24 h and the cells were stained with DHE. (D) DHE fluorescent intensity was determined after BEL-7402 cells were incubated with different
concentrations of the complex for 24 h. (E) The intracellular NO levels were detected after BEL-7402 cells (a) were exposed to 3.75 (b) and 7.5 mM (c) of Ir-1 for 24 h, and the
cells were stained with DAF-FMDA. (F) DAF-FMDA fluorescent intensity was determined after BEL-7402 cells were exposed to different concentrations of the complex for
24 h. *P < 0.05 represents significant differences compared with control. The scale bars for all the figures are 200 .
326 Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331

of concentration of the complex on the ROS levels, the DCF fluores- enhances the intracellular ROS levels with a concentration-depen-
cent intensity was determined and shown in Fig. 5B, the DCF fluo- dent manner.
rescent intensity increases with increasing the concentration of Ir- ROS include superoxide anion, hydroxyl radical and NO, etc. In
1, moreover, in the present of NAC, the DCF fluorescent intensity the assay of intracellular superoxide anion, dihydroethidium
decreases. These data further demonstrate that the complex (DHE) was used as fluorescent probe. After freely passing through

Fig. 6. (A) Location assay of Ir-1 (15.0 mM) in the lysosomes. (B) The lysosomal membrane permeabilization was studied after Ir-1 (15.0 mM) treated BEL-7402 cells (a) for 1
(b), 2 (c) and 5 h (d) and the cells were stained with AO. The scale bars for all the figures are 200 .

Fig. 7. (A) Location assay of Ir-1 in the mitochondria. (B) The mitochondrial membrane potential was determined under fluorescent microscope while BEL-7402 cells (a) were
treated with cccp (b, positive control), 3.75 (c) and 7.5 mM (d) of Ir-1 for 24 h. (C) The ratio of red/green fluorescence intensity was determined by flow cytometry while BEL-
7402 cells were incubated with NAC, 7.5 mM Ir-1, 7.5 mM Ir-1 + NAC, 15.0 mM Ir-1 and 15.0 mM Ir-1 + NAC for 24 h. *P < 0.05 represents significant differences compared with
control. The scale bars for all the figures are 200 .
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 327

plasma membrane, nonfluorescent DHE is oxidized by ROS to lular fluorescence, which merged very well with the red fluores-
ethidium, which intercalates with DNA and stains nuclei bright cence emitted from the lysosomal tracker, demonstrating that
red fluorescent. As shown in Fig. 5C, after treatment of BEL-7402 the complex locates at the lysosomes. Lysosomal membrane per-
cells (a) with 3.75 (b) and 7.5 mM (c) of Ir-1 for 24 h, the more meabilization can cause the release of lysosomal proteases, e.g.,
bright red DHE fluorescent intensity was found, which indicated cathepsin B, from lysosomes to cytosol to promote apoptosis
that the complex can enhance the superoxide anion levels. To [42]. The lysosomal membrane permeabilization induced by the
investigate the effect of concentration of Ir-1 on the intracellular complex was studied using AO as fluorescent probe. AO is a lysoso-
superoxide anion levels, DHF fluorescent intensity was detected. motropic metachromatic fluorochrome. When excited with blue
As shown in Fig. 5D, in the control, the DHE fluorescent intensity light, AO emits red fluorescence at high concentrations (when it
is 19.40. After BEL-7402 cell was exposed to 3.75, 7.5 and is present in lysosomes) and green fluorescence at low concentra-
15.0 mM of Ir-1 for 24 h, the DHE fluorescent intensity increased, tions (when it is present in the cytosol and the nucleus) [43]. As
which showed that the complex increases intracellular superoxide shown in Fig. 6B, after treatment of BEL-7402 cells (a) with
anion in a concentration-dependent manner. 15.0 mM of Ir-1 for 1 (b), 2 (c) and 5 h (d), the lysosomal permeabil-
The intracellular NO levels were also determined using 3- itztion can be observed, as indicated by increased numbers of ‘‘pale
amino-4-aminomethyl-20 ,70 -difluorescein diacetate (DAF-FMDA) cells,” i.e., cells with reduced numbers of AO-accumulating lyso-
as a fluorescent indicator of intracellular NO [36,37]. As shown in somes. These results demonstrated that Ir-1 can induce lysosomal
Fig. 5E, treatment of BEL-7402 cells (a) with 3.75 (b) and 7.5 mM membrane permeabilization in BEL-7402 cells.
(c) of Ir-1 for 24 h led to an increase of DAF-FMDA green fluores-
cence. The DAF-FMDA green fluorescent intensity was detected 3.8. Location assay of the complex and the changes of mitochondrial
using ImageXpress Micro XLS system and Multi Wavelength Cell membrane potential
Scoring module. The DAF-FMDA fluorescent intensity increases
with increasing concentration of Ir-1, which suggests that the com- The complex can enhance the intracellular ROS level, this
plex can enhance the intracellular NO levels (Fig. 5F). prompts us to evaluate the changes of mitochondrial membrane
potential. Mitochondria play a key role in the intrinsic apoptosis
3.7. Location and lysosomal membrane permeabilization (LMP) pathway, and loss of the mitochondrial membrane potential
(DWm) appears to be a universal event during the intrinsic apop-
Lysosomes, a kind of spherical vesicles containing various tosis pathway [44]. To investigate the location of the complex in
enzymes that are able to break down almost all kinds of biomole- the mitochondria, Mito TrackerÒ Deep Red FM (ThermoFisher,
cules, including proteins, nucleic acids, lipids, carbohydrates, and 100 nM) was used as red fluorescent probe. As shown in
cellular debris, act as the waste disposal system of eukaryotes
through degradation of unwanted molecules in the cytoplasm
[38–40]. The location of the complex in the lysosomes was inves-
tigated using LysoTracker Red as fluorescent probe. It is well
known that LysoTracker Red, a fluorescent weak base, could accu-
mulate in acidic organelles and successfully stain lysosomes in liv-
ing cells [41]. After BEL-7402 cells were treated with 15.0 mM of Ir-
1 for 5 h, the cells were imaged under fluorescent microscope. As
shown in Fig. 6A, the lysosomes were stained red fluorescence,
BEL-7402 cells incubated with Ir-1 showed bright-green intracel-

Fig. 8. (A) Intracellular Ca2+ levels were assayed after BEL-7402 cells (a) were Fig. 9. (A) The release of cytochrome c was assayed after BEL-7402 cells (a) were
exposed to 15.0 mM of Ir-1 for 2 (b), 4 (c) and 8 h (d). (B) The integrated fluorescent exposed to 7.5 mM of Ir-1 for 24 h. (B) The integrated fluorescent intensity/cell was
intensity/cell was determined after BEL-7402 cells were treated 15.0 mM of Ir-1 for determined after different concentration of Ir-1 treated BEL-7402 cells for 12 and
2, 4 and 8 h. *P < 0.05 represents significant differences compared with control. The 24 h. *P < 0.05 represents significant differences compared with control. The scale
scale bars for all the figures are 200 . bars for all the figures are 200 .
328 Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331

Fig. 7A, in the control, the mitochondria are stained in bright red. 3.9. Detection of intracellular Ca2+ levels
After BEL-7402 cells were exposed to 7.5 mM of Ir-1 for 8 h, the
complex emits green fluorescence. The complete merge of the Calcium is a prevalent signaling molecule in plentiful metabolic
red and the green fluorescence indicates that the complex enters processes and cellular signal responses, which can induce cell
into the mitochondria. The changes in the mitochondrial mem- death if maintained incessantly at high concentration [45]. In the
brane potential were also assayed under a fluorescent microscope assay of intracellular Ca2+ levels, Fluo-3MA was used as fluorescent
using JC-1 as fluorescent probe. It is well known that JC-1 emits probe, which combines with Ca2+ to be transferred into Fluo-3. The
red fluorescence corresponding to high mitochondrial membrane intracellular Ca2+ level in BEL-7402 cells induced by the complex
potential, and JC-1 emits green fluorescence corresponding to low was determined under ImageXpress Micro XLS system and
mitochondrial membrane potential. As shown in Fig. 7B, in the Multi Wavelength Cell Scoring module. As shown in Fig. 8A, in
control (a), JC-1 emits red fluorescence. After the treatment of the control (a), weak fluorescent points were found. 7.5 mM of Ir-
BEL-7402 cells with CCCP (b, carbonylcyanide-m-chlorophenylhy- 1 treated BEL-7402 cells at 2 (b), 4 (c) and 8 h, a number of bright
drazone, positive control), 3.75 mM (c) and 7.5 mM (d) for 24 h, JC- green fluorescent points were observed. The results show that the
1 emits bright green fluorescence. The changes from red to green complex can induce an increase of intracellular Ca2+ levels.
fluorescence suggest that the complex induces a decrease in the To investigate the effect of exposure time on the intracellular
mitochondrial membrane potential. To further explore the rela- Ca2+ levels, the Fluo-3 fluorescent intensity was determined. Seen
tionship between mitochondrial membrane potential and ROS, from Fig. 8B, we found that the Fluo-3 fluorescent intensity
the ratio of red/green fluorescent intensity was determined by increases with increasing the exposed time of BEL-7402 cells to
flow cytometry. As shown in Fig. 7C, in the control (a) or con- the complex.
trol + NAC (b), the ratios of red/green fluorescence are 2.10 and
1.97, respectively. Treatment of BEL-7402 cells with 7.5 mM Ir-1 3.10. Release of cytochrome c studies
(c), 7.5 mM Ir-1 + NAC (d), 15.0 mM Ir-1 (e) and 15.0 mM Ir-1
+ NAC (d) for 24 h, the ratios of red/green fluorescent intensity Cytochrome c (cyt-c) can induce cell apoptosis by activating
are 0.69, 1.07, 0.33 and 0.44, respectively. The results reveal that caspases proteases, which is released from the mitochondrial
NAC inhibits the changes of mitochondrial membrane potential, membrane space by reason of the mitochondrial dysfunction
namely, ROS can increase the changes of mitochondrial mem- [46]. As shown in Fig. 9A, there was no obvious green fluorescence
brane potential. Furthermore, the complex shows a concentra- of cytochrome c was observed in the control cells. After BEL-7402
tion-dependent manner to induce a decrease in the cells were treated with 7.5 mM of Ir-1 for 24 h, a number of green
mitochondrial membrane potential. fluorescent points were found, which indicated that the release

Fig. 10. (A) Autophagy was assayed after BEL-7402 cells (a) were treated with 3.75 (b), 7.5 (c) and 15.0 mM (d) of Ir-1 for 24 h. (B) The assay of LC3 and beclin-1 protein
expression induced by 15.0 mM of the complex by western blot. (C) Effect of 3-MA and NAC on the viability of BEL-7402 cells exposed to different concentrations of Ir-1, Ir-1
+ 3-MA and Ir-1 + NAC for 24 h. The scale bars for all the figures are 200 .
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 329

of cytochrome c was occurred. The integrated intensity/cell was 3.12. Cell invasion studies
obtained by Multi Wavelength Cell Scoring module. As shown in
Fig. 9B, the integrated intensity/cell increases with increasing the Since metastasis poses a foremost threat for cancer deaths,
time of BEL-7402 exposed to Ir-1. The results show that the com- inhibiting metastasis is an urgent therapeutic need [49]. To inves-
plex can release cyt-c from the mitochondria with a time-depen- tigate the effect of the complex on inhibiting migration, the tran-
dent manner. swell invasion assay in BEL-7402 cells was treated by Matrigel
invasion assay. As shown in Fig. 11A, BEL-7402 cells (a) were
exposed to 3.75 (b), 7.5 (c) and 15.0 mM (d) for 24 h, the number
3.11. Autophagy induced by the complex of cell invasion decreases gradually. Thus, we conclude that the
complex can effectively inhibit the cell invasion. The percentage
Autophagy is a highly conserved degradation pathway for bulk of different concentration of Ir-1 inhibiting cell invasion was calcu-
cellular components; it includes macroautophagy, microautophagy lated and shown in Fig. 11B, when the concentration of Ir-1 is
and chaperone-mediated autophagy [47]. Autophagy is closely 15.0 mM, the percentage of Ir-1 inhibiting cell invasion reaches
involved in the etiology of many important human diseases,
including cancer, neurodegenerative diseases and metabolic disor-
ders [48]. To evaluate the efficiency of the complex on autophagy,
the autophagy was performed under a fluorescent microscope
using MDC as fluorescent probe. As shown in Fig. 10A, there was
no obvious fluorescent points were observed in the control (a).
Exposure of BEL-7402 cells to 3.75 (b), 7.5 (c) and 15.0 mM (d) of
Ir-1 for 24 h, the MDC labeling green fluorescent points in the cyto-
plasm were discovered, which suggests that the autophagic vac-
uoles were formed. The results show that the complex can
induce autophagy. The up-regulation of Beclin-1 and the conver-
sion of the soluble form of LC3 (LC3-I) to the lipidated and
autophagosome-associated form (LC3-II) are hallmarks of autop-
hagy. As shown in Fig. 10B, the up-regulation of Beclin-1 protein
and the conversion of LC3-1 to LC3-II compared to the control were
observed, which further shows that the complex can induce autop-
hagy. In addition, the relationship among cell viability, autophagy
and ROS was evaluated by MTT method. As shown in Fig. 10C, in
the presence of 3-MA (6 mM, an inhibitor to inhibit autophagy)
and NAC (5 mM, an inhibitor to inhibit the generation of ROS),
the cell viability decreases or increases, respectively, which indi- Fig. 12. Western blot analysis of PARP, caspase 3, Bcl-2, Bad and Bax in BEL-7402
cells treated with NAC, 15.0 mM of Ir-1 and 15.0 mM of Ir-1 + NAC for 24 h. b-actin
cates that autophagy causes less cell death and ROS induce more
was used as internal control.
cell death.

Fig. 11. (A) Microscope images of invading BEL-7402 cells (a) that have migrated through the Matrigel induced by 3.75 (b), 7.5 (c) and 15.0 mM (d) of the Ir-1 for 24 h. (B) The
percentage of inhibiting cell invasion of BEL-7402 cells induced by different concentration of Ir-1 for 24 h. *P < 0.05 represents significant differences compared with control.
The scale bars for all the figures are 200 .
330 Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331

Fig. 13. The molecular mechanism of the complex induced apoptosis in BEL-7402 cells.

68.1%. It is clear that the complex inhibited cell invasion with a cell invasion and the percentage of Ir-1 (15.0 mM) inhibiting cell
concentration-dependent manner. invasion reaches 68.1%. The complex can induce autophagy and
autophagy prevents cell death, whereas ROS cause more cell death
3.13. The expression assays of the proteins related to apoptosis compared to the control. The western blot reveals that the complex
up-regulates caspase 3, Bad and Bax expression, and down-regu-
PARP (poly (ADP ribose) polymerase), an enzyme involved in lates Bcl-2 proteins expression. In summary, the complex induces
DNA repair, is a preferential substrate for caspase-3 [50]. PARP apoptosis through the following two pathways (Fig. 13): (I) the
plays an active role in key biological processes, PARP cleavage complex induces DNA damage, then the complex induces cell cycle
is a hallmark of apoptosis [51]. Caspases are known to mediate arrest at S phase, finally, the complex causes apoptosis. (II) The
the apoptotic pathway, caspase 3 mediates intrinsic pathway complex induces apoptosis through ROS-mediated lysosome–mi-
[52-54]. PARP protein cleavage in the complex-treated BEL- tochondrial dysfunction pathway.
7402 cells was investigated. As shown in Fig. 12, BEL-7402 cells
were incubated with 15.0 mM of Ir-1 for 24 h, the complex treat- Acknowledgements
ment caused cleavage of PARP, 116 KD into 89 KD fragment was
observed. Treatment of BEL-7402 cells with 15.0 mM of Ir-1 for This work was supported by the National Nature Science Foun-
24 h resulted in a significant increase in the levels of caspase 3 dation of China (No 21877018) and the Natural Science Foundation
expression compared to the control. On the other hand, Bcl-2 of Guangdong Province (No. 2016A030313728).
family proteins play an important role in regulating apoptosis.
Treatment of BEL-7402 cells with the complex led to down-regu- References
lation in Bcl-2 protein expression, and up-regulation of the
expression of Bad and Bax proteins. In addition, in the presence [1] A. Kumar, R.K. Gupta, R.P. Paitandi, K.B. Singh, S.K. Trigun, M.S. Hundal, D.S.
Pandey, J. Organomet. Chem. 801 (2016) 68.
of NAC (NAC + Ir-1), the expression of PARP, caspase 3, Bcl-2,
[2] L. Kelland, Nat. Rev. Cancer 7 (2007) 573.
Bad and Bax was inhibited, which exhibits that ROS enhance [3] E.R. Jamieson, S.J. Lippard, Chem. Rev. 99 (1999) 2467.
apoptotic efficiency. [4] D. Wang, S.J. Lippard, Nat. Rev. Drug Discov. 4 (2005) 307.
[5] E. Wong, C.M. Giandomenico, Chem. Rev. 99 (1999) 2451.
[6] P.J. Loehrer, S.D. Williams, L.H. Einhorn, J. Natl. Cancer Inst. 80 (1988) 1373.
4. Conclusions [7] F.N. Akladios, S.D. Andrew, C.J. Parkinson, J. Biol. Inorg. Chem. 21 (2016) 407.
[8] E.J. Gao, N. Sun, S.Z. Zhang, Y.Q. Ding, X. Qiu, Y. Zhang, M.C. Zhu, Eur. J. Med.
Chem. 121 (2016) 1.
Complex Ir-1 was synthesized and characterized. The assays of [9] W.X. Chen, X.D. Song, S.F. He, J. Sun, J.X. Chen, T. Wu, Z.W. Mao, J. Inorg.
cytotoxic activity in vitro show that the complex displays moder- Biochem. 164 (2016) 91.
[10] A. Pastuszko, K. Majchrzak, M. Czyz, B. Kupcewicz, E. Budzisz, J. Inorg.
ate cytotoxicity against the selected cancer cell lines. Morphologi- Biochem. 159 (2016) 133.
cal changes demonstrate that Ir-1 can induce apoptosis in BEL- [11] C. Zhang, B.J. Han, C.C. Zeng, S.H. Lai, W. Li, B. Tang, D. Wan, G.B. Jiang, Y.J. Liu, J.
7402 cells. Additionally, the complex increases intracellular ROS Inorg. Biochem. 157 (2016) 62.
[12] C.C. Zeng, S.H. Lai, J.H. Yao, C. Zhang, H. Yin, W. Li, B.J. Han, Y.J. Liu, Eur. J. Med.
and Ca2+ levels and induces cytochrome c release. Location studies
Chem. 122 (2016) 118.
show that complex locates at the lysosomes. After inducing lysoso- [13] N.A. Vyas, S.N. Ramteke, A.S. Kumbhar, P.P. Kulkarni, V. Jani, U.B. Sonawane, R.
mal membrane permeabilization, the complex enters into the R. Joshi, B. Joshi, A. Erxleben, Eur. J. Med. Chem. 121 (2016) 793.
mitochondria and causes a decrease in the mitochondrial mem- [14] C. Qian, J.Q. Wang, C.L. Song, L.L. Wang, L.N. Ji, H. Chao, Metallomics 5 (2013)
844.
brane potential. The cell cycle arrest shows that the complex inhi- [15] M. Tian, J.J. Li, S.M. Zhao, L.H. Guo, X.D. He, D.L. Kong, H.R. Zhang, Z. Liu, Chem.
bits cell growth at S phase. The complex can effectively inhibit the Commun. 53 (2017) 12810.
Y.-J. Wang et al. / Polyhedron 156 (2018) 320–331 331

[16] J.J. Cao, C.P. Tan, M.H. Chen, N. Wu, D.Y. Yao, X.G. Liu, L.N. Ji, Z.W. Mao, Chem. [33] G.B. Jiang, J.H. Yao, J. Wang, W. Li, B.J. Han, Y.Y. Xie, G.J. Lin, H.L. Huang, Y.J. Liu,
Sci. 8 (2017) 631. New J. Chem. 38 (2014) 2554.
[17] Y. Hisamatsu, N. Suzuki, A.A. Masum, A. Shibuya, R. Abe, A. Sato, S.I. Tanuma, S. [34] W. Li, B.J. Han, J.H. Yao, G.B. Jiang, Y.J. Liu, RSC Adv. 5 (2015) 24534.
Aoki, Bioconjugate Chem. 28 (2017) 507. [35] K.E. Prosser, S.W. Chang, F. Saraci, P.H. Le, C.J. Walsby, J. Inorg. Biochem. 167
[18] J.S.Y. Lau, P.K. Lee, K.H.K. Tsang, C.H.C. Ng, Y.W. Lam, S.H. Cheng, K.K.W. Lo, (2017) 89.
Inorg. Chem. 48 (2009) 708. [36] H. Kojima, Y. Urano, K. Kikuchi, T. Higuchi, Y. Hirata, T. Nagano, Angew. Chem.
[19] L. Tabrizi, H. Chiniforoshan, Dalton Trans. 46 (2017) 2339. Int. Ed. Engl. 38 (1999) 3209.
[20] S.K. Tripathy, U. De, N. Dehury, P. Laha, M.K. Panda, H.S. Kim, S. Patra, Dalton [37] X. Zheng, G. Ning, D. Hong, M. Zhang, Mol. Biol. 279 (2004) 69.
Trans. 45 (2016) 15122. [38] C. Settembre, A. Fraldi, D.L. Medina, A. Ballabio, Nat. Rev. Mol. Cell. Biol. 14
[21] C.L. Wang, J.F. Liu, Z.Z. Tian, M. Tian, L.J. Tian, W.Q. Zhao, Z. Liu, Dalton Trans. (2013) 283.
46 (2017) 6870. [39] C. Hiebel, C. Behl, Life. Sci 138 (2015) 3.
[22] J.J. Li, L.H. Guo, Z.Z. Tian, M. Tian, S.M. Zhang, K. Xu, Y.C. Qian, Z. Liu, Dalton [40] D.A. Salazar, A. Rodríguez-López, A. Herren~o, H. Barbosa, J. Herrera, A. Ardila,
Trans. 46 (2017) 15520. G.E. Barreto, J. González, C.J. Alméciga-Díaz, Mol. Genet. Metab. 117 (2016)
[23] R.P. Paitandi, S. Mukhopadhyay, R.S. Singh, V. Sharma, S.M. Mobin, D.S. Pandey, 129.
Inorg. Chem. 56 (2017) 12232. [41] B. Sun, J. Liu, Y. Gao, H.B. Zhen, L. Li, Q.W. Hu, H.Q. Yuan, H.X. Lou, Eur. J. Med.
[24] C. Zhang, S.H. Lai, H.H. Yang, D.G. Xing, C.C. Zeng, B. Tang, D. Wan, Y.J. Liu, RSC Chem. 136 (2017) 603.
Adv. 7 (2017) 17752. [42] L. Galluzzi, J.M. Bravo-San Pedro, G. Kroemer, Nat. Cell Biol. 16 (2014) 728.
[25] C. Zhang, S.H. Lai, C.C. Zeng, B. Tang, D. Wan, D.G. Xing, Y.J. Liu, J. Biol. Inorg. [43] P. Boya, G. Kroemer, Oncogene 27 (2008) 6434.
Chem. 21 (2016) 1047. [44] C.B. Zhao, W.J. Gao, T.S. Chen, Apoptosis 19 (2014) 668.
[26] S. Sprouse, K.A. King, P.J. Spellane, R.J. Watts, J. Am. Chem. Soc. 106 (1984) [45] M.J. Berridge, M.D. Bootman, H.L. Roderick, Nat. Rev. Mol. Cell Biol. 4 (2003)
6647. 517.
[27] A.K. Bilakhiya, B. Tyagi, P. Paul, Inorg. Chem. 41 (2002) 3830. [46] X. Liu, C.N. Kim, J. Yang, R. Jemmerson, X. Wang, Cell 86 (1996) 147.
[28] T. Mosmann, J. Immunol. Methods 65 (1983) 55. [47] N. Mizushima, M. Komatsu, Cell 147 (2011) 728.
[29] R.R. Tice, E. Agurell, D. Anderson, B. Burlinson, A. Hartmann, H. Kobayashi, Y. [48] A.J. Meijer, P. Codogno, Crit. Rev. Clin. Lab. Sci. 46 (2009) 210.
Miyamae, E. Rojas, J.C. Ryu, Y.F. Sasaki, Environ. Mol. Mutagen. 35 (2000) 206. [49] A.F. Chambers, A.C. Groom, I.C. MacDonald, Nat. Rev. Cancer 2 (2002) 563.
[30] C. Alapetite, T. Wachter, E. Sage, E. Moustacchi, Int. J. Radiat. Biol. 69 (1996) [50] F. Malik, A. Kumar, S. Bhushan, S. Khan, A. Bhatia, K.A. Suri, G.N. Qazi, J. Singh,
359. Apoptosis 12 (2007) 2115.
[31] D. Wan, B. Tang, Y.J. Wang, B.H. Guo, H. Yin, Q.Y. Yi, Y.J. Liu, Eur. J. Med. Chem. [51] P.J. Duriez, G.M. Shah, Biochem. Cell Biol. 75 (1997) 337.
139 (2017) 180. [52] G.S. Salvesen, V.M. Dixit, Cell 91 (1997) 443.
[32] J.F. Kou, C. Qian, J.Q. Wang, X. Chen, L.L. Wang, H. Chao, L.N. Ji, J. Biol. Inorg. [53] N.A. Thornberry, Y. Lazebnik, Science 281 (1998) 1312.
Chem. 17 (2012) 81. [54] C.L. Hsu, Y.S. Yu, G.C. Yen, J. Agric. Food Chem. 58 (2010) 2201.

Das könnte Ihnen auch gefallen