You are on page 1of 8

Chemico-Biological Interactions 239 (2015) 184–191

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Synthesis, characterization and biological evaluation of Rutin–zinc(II)


flavonoid -metal complex
Norma Estefania Andrades Ikeda a,e,1, Estela Maria Novak b,1, Durvanei Augusto Maria c,1,
Adélia Segin Velosa d, Regina Mara Silva Pereira d,⇑
a
Faculty of Medicine, University of São Paulo, São Paulo, Brazil
b
Fundação Pró-Sangue Hemocentro de São Paulo, São Paulo, Brazil
c
Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, Brazil
d
Department of Pharmacy, Anhanguera University of São Paulo, São Paulo, Brazil
e
Department of Biotechnology, Anhanguera University of São Paulo, São Paulo, Brazil

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

Article history: Synthesis of compounds analogous to natural products from secondary metabolites, such as flavonoids, is
Received 8 December 2014 a promising source of novel drugs. Rutin (quercetin-3-O-rutinoside) is a natural flavone, which has, in its
Received in revised form 19 May 2015 chemical structure, different sites for coordination with transition metals and the complexation with
Accepted 5 June 2015
these metals enhances its biological properties. Rutin–zinc(II), a flavonoid–metal complex, was synthe-
Available online 16 June 2015
sized and characterized by UV–VIS, FT-IR, elemental analysis and 1H NMR. The antioxidant and antitumor
activities, as well as the cytotoxicity and in vivo toxicity of this complex were evaluated and compared
Keywords:
with the free rutin. Rutin–zinc(II) has not shown any cytotoxicity against normal cells (fibroblasts and
Flavonoids
Rutin
HUVECs) or toxicity in BALB/c mice, but has shown antioxidant activity in vitro and cytotoxicity against
Metal complex leukemia (KG1, K562 and Jurkat), multiple myeloma (RPMI8226) and melanoma (B16F10 and SK-Mel-28)
Antitumor cell lines in vitro. In Ehrlich ascites carcinoma model, Rutin–zinc(II) modulated the mitochondrial mem-
brane potential and the expression of genes related to cell cycle progression, angiogenesis and apoptosis.
Ó 2015 Elsevier Ireland Ltd. All rights reserved.

1. Introduction 2. Experimental section

Drug discovery from natural products have brought new clinical 2.1. Synthesis and structural characterization of Rutin–zinc(II)
applications of plant secondary metabolites as templates for complex
synthetic novel drugs [1]. Flavonoids are polyphenolic compounds
found in seeds, fruits, olive oil, tea, and red wine [2–4] that have A solution of [Zn(CH3COO)2]2H2O salt in distilled water was
demonstrated a wide variety of biological actions [5–7]. Rutin slowly added dropwise to a solution of dehydrated rutin
(quercetin-3-O-rutinoside) is a natural flavone, which has, in its (Dimorphandra mollis) in methanol. The mixture was stirred at
chemical structure, different sites for coordination with 90–140 rpm for 24 h at 37–40 °C. The complex was filtered in a
transition metals and the complexation of rutin with these metals vacuum system, washed with methanol and dried at room temper-
enhances its biological properties [8]. It has been reported that ature. Yield: 25%, M.P. 230 °C, Anal. Cal. for found, exp. (calc.)
rutin has several pharmacological properties including antioxidant, C31H48O26Zn2: %C 38.29 (38.62) and %H 4.80 (4.98); IR k (cm-1):
anti-inflammatory, anticarcinogenic, cytoprotective, antiplatelet, 3355(OAH); 1571 (C@O); 1477(C@C); 1270(CAOAC); 804(CAC).
antithrombotic, vasoprotective, and cardioprotective activities NMR1H (d, ppm): 12.85 (brs, 1H, 5-OH); 12.62 (brs, 1H, 7-OH);
[9–11]. 7.50 (brs, 1H, 20 -H); 6.75(brs, 1H, 50 -H); 6.25(d brs, 1H). Melting
points were determined by using a QUIMIS Melting Point
Equipment. The IR spectra were recorded in the solid state in the
range 4000–400 cm1 by using a Nicolet Nexus spectrometer.
⇑ Corresponding author. Tel.: +55 11 2967 9147. The UV–VIS spectra were recorded in aqueous solution (pH 6.5),
E-mail address: rpereira02@hotmail.com (R.M.S. Pereira).
in the range 200–500 nm, by using a Femto 800XI spectrometer.
1
These authors contributed equally to this work. The 1H-NMR spectra were measured at 25 °C in an INOVA –

http://dx.doi.org/10.1016/j.cbi.2015.06.011
0009-2797/Ó 2015 Elsevier Ireland Ltd. All rights reserved.
N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191 185

500 Mz Spectrophotometer, in DMSO-d6. The chemical shifts are Rutin–zinc(II) administered intraperitoneally (i.p.) for 7, 14 and
referred to DMSO signal at 3.38 ppm. 21 days and observed for 30, 60 and 90 days after treatment.
Toxicology tests were performed with the flavonoid rutin
2.2. Antioxidant activity (5 mg/Kg) in the same conditions. The toxicity was assessed by
body weight measurement, hematological and biochemical analy-
The antioxidant activity of Rutin–zinc(II) was tested by two ses and histological examination of liver and kidneys as indicators.
different methods: superoxide radical scavenger assay and DPPH Mice were electively sacrificed by cervical dislocation on day 30, 60
assay. and 90 and blood samples were collected from retro-orbital plexus.
For histopathological studies, tissues were fixed in 10% neutral buf-
2.2.1. Superoxide radical scavenging assay fered formalin, dehydrated, and embedded in paraffin by routine
The superoxide radicals generated were determined spec- methods and stained with hematoxylin and eosin (H&E).
trophotometrically by monitoring the product of nitroblue
tetrazolium (NBT) [12]. Various concentrations of rutin and 2.5. Antitumor activity evaluation
Rutin–zinc(II) solutions (1, 10 e 100 lM) were added to the reaction
mixture containing: 50 lM NBT, 20 lM phenazine methosulfate, The antitumor activity of the compounds was evaluated in the
156 lM NADH (adenosine 5-trihydrogen diphosphate), in 0.1 M Ehrlich ascites carcinoma (EAC) model. EAC is a spontaneous mur-
phosphate buffer (pH 7.4) making up a final volume of 2.5 mL. ine mammary adenocarcinoma adapted to ascites form and carried
After the incubation of the mixture at 25 °C for 10 min, the in outbred mice by serial intraperitoneal passages. This model has
absorbance was read at 560 nm and compared with the control been used as a transplantable tumor model to investigate the anti-
sample, containing 50 lM NBT. The percentage of superoxide neoplastic effects of many substances or compounds [14,15].
radical scavenging was calculated from the samples optical density.
All the measurements were replicated 3 times. 2.5.1. Experimental design
Mice were inoculated with 1  104 EAC cells into the peritoneal
2.2.2. DPPH assay cavity. The animals were divided into six groups (n = 10) and trea-
The solution contained 1 mL of DPPH (20 lM) and 1 mL of dif- ted with 0.1 mL of the compounds, administered via i.p. route for
ferent concentrations of rutin and Rutin–zinc(II) solution, resulting 14 days. Group A (control group): treated with phosphate-
in a final concentration of DPPH of 10 lM. The mixtures were vig- buffered saline (PBS); Group B: treated with Rutin–zinc(II)
orously mixed and allowed to stand in the dark for 30 min at 25 °C. (5 mg/Kg); Group C: treated with rutin (5 mg/Kg); Group D: trea-
The absorbance of the resulting solutions was measured at 517 nm ted with Rutin–zinc(II) (5 mg/Kg) in combination with paclitaxel
against a blank sample containing only DPPH, the negative control; (15 mg/Kg); Group E: treated with rutin (5 mg/Kg) in combination
rutin and vitamin C served as the positive control [10,11]. All the with paclitaxel (15 mg/Kg); Group F: treated with paclitaxel
measurements were replicated 3 times. (15 mg/Kg). Rutin–zinc(II) and rutin were solubilized as described
above and paclitaxel was diluted in cremophorÒ. In the groups A, B
2.3. Cytotoxicity assay and C, mice were treated daily, in the groups D and E received
Rutin–zinc(II) and rutin daily and paclitaxel on day 4, 8, and 12.
RPMI 8226, KG1, K562, Jurkat and SK-MEL-28 cell lines were Group F received paclitaxel twice a week. The ascitic fluid from
obtained from ATCC (Rockville, MD, USA). The B16F10, the peritoneal cavity of tumor bearing mice was collected to obtain
SK-MEL-28, human fibroblasts and Human Umbilical Vein the tumor cells at the end of the treatment to perform the DNA
Endothelial cells (HUVECs) were provided by Dr. Durvanei fragmentation, mitochondrial membrane potential and angiogene-
Augusto Maria from the Butantan Institute (São Paulo, Brazil). sis, apoptosis and cell cycle-related gene expression analysis of the
The cell lines were routinely cultured in RPMI-1640 medium, con- EAC cells. Hematological analysis was performed to evaluate the
taining 10% inactivated fetal bovine serum (supplemented with effect of the compounds on the EAC bearing mice. The survival
2.0 mM glutamine and 50 lg/mL garamycin) at 37 °C in a humidi- rate was determined by Kaplan–Meier method.
fied incubator with 5% CO2. The cells were treated with Rutin–
zinc(II) and rutin in concentrations ranging from 17.2 lM to 2.5.2. DNA fragmentation analysis
275.6 lM for 24 h and the cell viability was assessed by MTT assay. The ascitic fluid from the peritoneal cavity of tumor bearing
The cell viability was determined by MTT [3-(4,5-dimethylthiazo mice was collected and centrifuged to obtain the tumor cells at
l-2-yl-2,5-diphenyl tetrazolium bromide] method [13]. At the end the end of the treatment to perform the flow cytometry analysis
of the treatment, MTT (0.5 mg/mL) diluted in fresh medium was of DNA fragmentation of the EAC cells. The EAC cells were washed
added. After 4 h of incubation, the blue formazan crystals were dis- with PBS and centrifuged at 1500 rpm at 4 °C for 5 min. Pellets
solved with 100 lL acid-isopropanol and the absorbance was mea- were fixed in 70% ice-cold ethanol overnight. The cells were cen-
sured at 570 nm using a microplate reader (TiterTek Multiskan trifuged, washed with PBS, and incubated in PBS containing
Plus, Flow Laboratories, USA). Experiments were performed in 1 mg/mL RNase for 10 min at room temperature. Samples were
triplicate. stained with 1 mg/mL propidium iodide for 30 min at 4 °C. Data
acquisition was performed by flow cytometry (FACSCalibur) and
2.4. In vivo toxicological studies analyzed by Cell Quest software.

All animal experiments were performed with the approval of 2.5.3. Mitochondrial membrane potential (Dwm) analysis
the Institutional Animal Care and Use Committee of the Butantan The mitochondrial membrane potential (Dwm) was assessed in
Institute (Permit number: n°611/09). BALB/c female mice, EAC cells by flow cytometry using Rhodamine 123 (Invitrogen), a
8-week-old, weighing 20–22 g were maintained in a 12 h fluorescent lipophilic cationic dye which preferentially enters into
light-and-dark cycle, at a temperature of 22 ± 2 °C and were fed mitochondria due to the highly negative mitochondrial membrane
with standard pellet chow, with free access to water. The Rutin– potential. Depolarization of DWm results in the loss of
zinc(II) complex was solubilized in deionized water and rutin Rhodamine-123 from the mitochondria and a decrease in intracel-
was solubilized in propylene glycol and diluted in deionized water. lular fluorescence [16]. The cells were collected from ascitic fluid of
The animals were divided in three groups and treated with tumor bearing mice, washed with PBS and centrifuged at 1500 rpm
186 N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191

at 4 °C for 5 min and incubated in PBS containing 5 mg/mL by 1H NMR. The proton signals of Rutin–zinc(II) shifted to lower
Rhodamine 123 for 10 min at 37 °C, in a humidified incubator with frequencies relative to the free flavonoid. The coordination with
5% CO2 for 30 min. Data acquisition was performed by flow cytom- the Zn(II) ion decreases the electron density of the flavonoid
etry (FACSCalibur) and analyzed by Cell Quest software. around 5-OH, probably due to the interaction with Zn(II) (a Lewis
acid). The 1H NMR signal assigned to 3’ and 4’ hydroxyl in Rutin–
2.5.4. qRT-PCR analysis of angiogenesis, apoptosis and cell cycle- zinc (II) becomes broader when compared with free rutin. These
related gene expression data are in agreement with the IR and the elemental analyses data
Total RNA was isolated from EAC cells using TRIzolÒ reagent demonstrating metallic ion coordination in positions 4, 5 and 30 , 40 ,
(Invitrogen) according to the protocol supplied by the manufac- and four water molecule are coordinated in each ion (Fig. 1).
turer. Reverse transcription reaction was carried out using 1 lg
of RNAse-free DNase (Qiagen)-treated total RNA following the pro- 3.2. Antioxidant activity
tocol of SuperScript™ III, First-Strand Synthesis SuperMix for
RT-PCR (Invitrogen). The primer sequences of VEGF, Cyclin D1, Superoxide radicals generated from dissolved oxygen by
Caspase-3 Caspase-8 and GAPDH and cycling conditions used were PMS-NADH coupling can be measured by their ability to reduce
previously described [17–20]. Quantitative PCR assays were car- NBT. As shown in Fig. 3, in a concentration of 10 lM Rutin–
ried out by using Rotor Gene RG-3000 rotor (Corbett Research, zinc(II) showed better effect than rutin. However, in a concentra-
AUS) with PlatinumÒ SYBRÒ Green qPCR Super Mix-UDG reagent tion of 100 lM Rutin–zinc(II) superoxide radical scavenging was
(Invitrogen). GAPDH was used as a reference for relative quantifi- 25% higher than the rutin scavenging ability. The percentage inhi-
cation and data were analyzed according to the Livak method [21]. bition of Rutin–zinc(II) and rutin at 100 lM was 70%, and 55%,
respectively. DPPH assay showed that Rutin–zinc(II) was more
2.6. Statistical analyses effective than the free flavonoid rutin, which inhibited 80% at
30.4 lM in comparison with free rutin, which inhibited 45% in
Statistical analyses were performed by using One-way Analysis the same concentration. The Rutin–zinc(II) effect is comparable
of Variance (ANOVA) and Tukey–Kramer post hoc test for multiple with vitamin C (Fig. 2).
comparisons. Statistical significance was set at p < 0.05.
3.3. Cytotoxicity
3. Results
The cytotoxic effect of Rutin–zinc(II) in comparison with free
3.1. Structural characterization rutin was determined by MTT assay. Rutin–zinc(II) showed cyto-
toxicity against all tumor cell lines tested after 24 h of treatment,
The complex Rutin–zinc(II) was obtained when a solution of but, in fibroblasts and HUVECs normal cell lines, no cytotoxic
[Zn(CH3COO)2].2H2O salt in distilled water was slowly added drop- effects were observed. The human acute myeloid leukemia cell line
wise to a solution of dehydrated rutin in methanol. A new com- (KG1) showed higher sensitivity to the complex with an IC50 of
pound Rutin–zinc(II) was characterized by UV–VIS and FT-IR 91.4 lM than RPMI8226 multiple myeloma cell line (IC50 of
spectroscopy. Comparing the electronic spectra (200–500 nm) of 196.6 lM) as shown in Table 1. The flavonoid rutin showed no
rutin and Rutin–zinc(II), a band shift at 353 nm to 393 nm (band cytotoxicity against tumor and normal cell lines tested.
I of rutin) and another at 259 nm to 269 nm (band II of rutin) were
observed (Fig. 1). These shifts suggest that two metal ions may be 3.4. Toxicity of Rutin–zinc(II) in mice
occupying the coordination sites 4, 5 and 40 , 50 (Fig. 1). The IR spec-
trum of Rutin–zinc(II) showed a band shift in 1655 Rutin–zinc(II) Subchronic toxicity tests were performed to determine Rutin–
to 1627 cm1 indicating an interaction of the Zn(II) ion via the car- zinc(II) toxicity. No behavioral changes or physical signs of pain,
bonyl group, in position 4 (Fig. 1). In comparison with the rutin fre- discomfort or neurological impairment such as pilo-erection,
quencies of mOH, mC@C, mCAOAC and mCAC bands (3351, 1597, 1295 e vocalization, motility alterations, dyspnea, cyanosis and convul-
808 cm1, respectively), the complex Rutin–zinc(II) shows bands sions were observed in the mice, and all groups of treatment
in 3322, 1571, 1272 e 805 cm1, respectively, shifting to lower fre- showed progressive weight gain throughout the experiment in
quencies, indicating also the Zn(II) ion coordination at position 30 comparison with the control group (Table 2). The effect of
and 40 as well. The complexation of Zn(II) to rutin was also verified 5 mg/Kg Rutin–zinc(II) and rutin on red blood cells, white blood

Fig. 1. Spectrum of UV–VIS in methanol solution of 50 lM of rutin and Rutin–zinc(II) (A) and schematic structure of Rutin–zinc(II) (B).
N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191 187

A 80
B 100

superoxide scavenger (%)


80

DPPH inhibition (%)


60

60
40

40

20
20

0 0
1 μM 10 μM 100 μM 1 μM 10 μM 100 μM Vitamin C Rutin Rutin-zinc(II)
Rutin Rutin-zinc(II)

Fig. 2. Superoxide radical scavenging activity of rutin and Rutin–zinc(II) at 1, 10 and 100 lM (A). DPPH inhibition from vitamin C, rutin and Rutin–zinc(II) at 30.4 lM (B).

Fig. 3. Histological analysis of liver (a), kidney (b) and lung (c) of BALB-c mice treated with Rutin–zinc(II) for 21 days and sacrificed 90 days after treatment. H&E stain (x400).

Table 1 3.5. The combination of rutin and Rutin–zinc(II) with paclitaxel


Determination of Rutin–zinc(II) IC50 by MTT assay. enhanced DNA fragmentation
Cell lines Rutin–zinc(II)
IC50 (lM) The effects of Rutin–zinc(II), rutin, paclitaxel and combinations
B16F10 160.7 on DNA fragmentation in EAC cells, in vivo, were investigated. The
SK-MEL 28 194.0 percentages of DNA fragmentation of the untreated control group
KG1 91.4 (16.6 ± 3.7%), Rutin–zinc(II) (31.5 ± 5.7%), rutin (16.9 ± 8.8%), pacli-
RPMI 8226 196.6 taxel (36.0 ± 1.8%), rutin in combination with paclitaxel
JURKAT 150.2
K562 173.2
(63.4 ± 3.5%) and Rutin–zinc(II) in combination with paclitaxel
Fibroblasts >275.6 (60.7 ± 8.1%) demonstrates that the combination with Rutin–
HUVECs >275.6 zinc(II) and rutin significantly enhanced paclitaxel antitumor activ-
ity (Fig. 4).
cells, platelets and liver enzymes was assessed after 7, 14 and
21 days of treatment and 30,60 and 90 days after treatment. An 3.6. Loss of mitochondrial membrane potential (Dwm) in EAC cells
increase in the number of platelets, red blood cells and white blood
cells was observed after 21 days of treatment in the groups treated The changes in the mitochondrial membrane potential (Dwm)
with rutin and Rutin–zinc(II) (Table 2). However, 90 days after was assessed by flow cytometry using Rhodamine 123 fluorescent
treatment all parameters returned to normal levels. No significant probe. The Fig. 5 shows the percentages of Dwm in the EAC cells in
changes in ALT and AST levels were observed (Table 2). all groups of treatment: rutin (30.0 ± 8.0%), Rutin–zinc(II)
Histopathological examinations of the tissues indicated that there (8.7 ± 6.3%), paclitaxel (2.4 ± 1.2%), rutin in combiation with pacli-
were no detectable abnormalities and alterations in the micro- taxel (2.7 ± 0.3%) and Rutin–zinc(II) in combination with paclitaxel
scopic examination of the liver, kidney and lungs in comparison (2.8 ± 0.8%) and untreated control group (86.7 ± 8.3%). The results
to the control (Fig. 3). demonstrate a significant loss of mitochondrial membrane

Table 2
Effect of intraperitoneal administration of rutin and Rutin–zinc(II) at the dose of 5 mg/Kg on hematological parameters, liver enzymes and body weight in BALB/c mice.

Groups RBC (103/mm3) WBC (103/mm3) PLT (103/mm3) ALT (U/mL) AST (U/mL) Weight (g)
Control 9.05 ± 0.66 4.91 ± 0.79 687.5 ± 97.08 19.4 ± 1.7 17.6 ± 1.7 24.34 ± 0.70
Rutin 13.13 ± 1.31⁄⁄ 9.95 ± 1.63⁄⁄⁄ 2595 ± 432.60⁄⁄⁄ 19.2 ± 1.5 16.1 ± 0.4 22.48 ± 1.15
Rutin–zinc(II) 12.48 ± 1.74⁄ 8.32 ± 0.58⁄⁄ 2036 ± 94.54⁄ 19.4 ± 1.2 17.2 ± 1.0 23.39 ± 1.32

Data showed as mean ± SD, n = 10. ***p < 0.0001, **p < 0.001, *p < 0.05, compared with untreated control (ANOVA and Tukey–Kramer post-hoc test) after 21 days of treatment.
RBC: red blood cell, WBC: white blood cell, PLT: platelets, AST: aspartate aminotransferase, ALT: alanine amionotransferase.
188 N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191

A Control
B C Paclitaxel
Control
Rutin Rutin -zinc(II) Rutin-zinc(II)+Paclitaxel
Rutin +Paclitaxel

80
D
***
***

60
EAC cells (%)

40 ***
***

20

0
1

1
S

.
N /M

N /M

S
N /M

N /M

N /M

N /M

.
ag

ag

ag

ag

ag

ag
G

G
2

2
0/

0/

0/

0/

0/

0/
fr

fr

fr

fr

fr

fr
G

G
G

G
A

A
D

D
Control Rutin Rutin-Zn(II) Paclitaxel Rutin+paclitaxel Ru-Zn(II)+paclitaxel
(untreated)

Fig. 4. DNA fragmentation analysis (A, B, C and D) of EAC cells collected from tumor bearing mice treated with rutin, Rutin–zinc(II), paclitaxel, paclitaxel associated with rutin
and Rutin–zinc(II) in comparison with untreated control group. ANOVA (p < 0.0001), ***p < 0.0001 (Tukey–Kramer post-hoc test).

Control
B Rutin
A 100 Rutin-Zn(II)
mitochondrial membrane potential-ΔΨm (%)

80

60 Control
Paclitaxel
C Rutin+paclitaxel
Rutin-Zn(II)+paclitacel
40 ***

20
***
*** *** ***
0
Control Rutin Rutin-Zn(II) Paclitaxel Rutin+paclitaxel Rutin-Zn(II)+paclitaxel
(untreated)

Fig. 5. Mitochondrial membrane potential analysis (A, B and C) of EAC cells collected from tumor bearing mice treated with rutin, Rutin–zinc(II), paclitaxel, paclitaxel
associated with rutin and Rutin–zinc(II) in comparison with untreated control group. ANOVA (p < 0.0001), ***p < 0.0001 (Tukey–Kramer post-hoc test).

potential in the ECA cells of the groups treated with paclitaxel and the tested compounds significantly down-regulated VEGF and
combinations in comparison with the single flavonoid rutin and Cyclin D1 and up-regulated Caspase-3 and Caspase-8 gene expres-
Rutin–zinc(II) (see Table 3). sion in comparison with untreated control group.

3.7. Angiogenesis, apoptosis and cell cycle-related gene expression 4. Discussion


analysis
Natural products like flavonoids represent an attractive source
The expression of VEGF, Cyclin D1, Caspase-3 Caspase-8 genes of varied structures that exhibit potent biological activities and
in EAC cells was assessed by qRT-PCR. Data presented in Fig. 6 desirable pharmacological profiles [22,23]. Furthermore, flavo-
shows the effect of the different treatments in EAC in vivo. All of noids can form complexes with transition metal ions and these
N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191 189

Table 3 concentrations and experimental conditions. The flavonoid


Hematological parameters of EAC bearing mice. metal-complex Rutin–zinc(II) was synthesized and characterized
Groups RBC (103/ WBC (103/ PLT (103/mm3) by UV–VIS, FT-IR spectroscopy and 1H NMR indicating that the
mm3) mm3) Zn(II) ion is coordinated to the benzopyrane ring of rutin (positions
Control (normal) 8.87 ± 0.85 3.91 ± 0.45 687.5 ± 97.0 4 and 5), and to the aromatic ring (30 and 40 positions). Rutin–
Control (untreated) 4.00 ± 0.55 5.37 ± 0.35 967.0 ± 90.5 zinc(II) has shown cytotoxicity against leukemia, multiple mye-
Rutin 6.87 ± 0.30⁄⁄⁄a 8.42 ± 0.43⁄⁄⁄a 984.0 ± 25.97 loma and melanoma cell lines, but did not show any cytotoxicity
Rutin–Zn(II) 6.26 ± 0.5⁄⁄⁄a 7.10 ± 0.18⁄⁄⁄a 924.0 ± 63.69
Paclitaxel 2.99 ± 0.24 2.97 ± 0.12 594.0 ± 30,51
against normal cells in vitro. However, at the same concentrations,
Rutin + paclitaxel 2.12 ± 0.15 2.10 ± 0.17⁄⁄b 766.0 ± 34.0⁄b free rutin did not show cytotoxicity against normal and cancer
Rutin– 5.97 ± 0.38⁄⁄⁄b 3.57 ± 0.33 1286 ± 73.15⁄⁄⁄b cells. Regarding the cytotoxicity of rutin against the cancer cells
Zn(II)+paclitaxel evaluated in this work, there are no reports of cytotoxicity against
The data were expressed as mean ± SD, n = 10. ***p < 0.0001, **p < 0.001, *p < 0.05, RPMI 8226 and KG1 cell line and the flavonoid showed no cytotox-
compared with (a) untreated control group and (b) paclitaxel treated group icity against SK-ML-28 cell line [25]. Rutin from Pupalia lappacea
(ANOVA and Tukey–Kramer post-hoc test). extract showed cytotoxicity against a K562 cell line [26] and the
flavonoid isolated from Leonurus heterophyllus showed no cytotox-
icity against a K562 cell line [27], wolfberry fractions containing
complexes possess higher scavenging and anti-inflammatory activ- rutin showed cytotoxicity against Jurkat cell line [28]. Rutin
ities and cytoprotective effects than the parent flavonoid [24]. The demonstrated antimetastatic activity in vivo in B16F10 melanoma
synthesis of a new Zn(II) complex bearing a rutin ligand started [29]. Matsuo et al. [30] evaluated the cytotoxicity of various flavo-
from the premise that this polyphenol demonstrated anticancer noids, including rutin, against normal cells. The results demon-
activity in several works carried out with different rutin sources, strated that rutin did not show cytotoxicity against HUVEC

1.0
A 1.0 B Cyclin D1 relative gene expression
VEGF relative gene expression

0.8
0.8

0.6
0.6
***

0.4
0.4 ***

0.2 0.2

0.0 0.0
in

l
)

II)

l
tin

l
d)

II)

el
l

xe

xe

xe
ed
xe

xe

ut

c(
ax
c(
te

ita
ita

ita
at
Ru

ita
i ta

in
R
in
ea

it

re

cl

cl
Z

ac
cl

cl
-z

ac

a-
nt
tr

Pa
Pa
a

+p
+p
tin
un

in
(u
+p
+p

ut

in

II)
Ru
l(

ol
tin

II)

ut

n(
ro

tr
n(

-Z
R
Ru

on
nt

-Z

in
C
Co

tin

ut
R
Ru

C 1.0
***
D
1.0
Caspase-8 relative gene expression
Caspase-3 relative gene expression

***
0.8 0.8

0.6 0.6

0.4 0.4

0.2 0.2

0.0 0.0
in

l
)

el
l

in

l
d)

el
l
xe

xe
ed

II

xe

xe
II
ax
ut

c(

ax
ut

c(
te
ita
ita
at

ita
ita
in
R

lit

in
ea

lit
re

cl

l
Z

cl

l
ac
ac

ac
ac
tr
a-
nt

a-

a
un
+p

P
+p
in

+p

P
+p
(u

in
ut

in

II )

l(

ut

in

II)
ol

ut

n(

ro

ut

n(
tr

-Z
R

t
on

-Z
R
on
in

in
C

C
ut

ut
R

Fig. 6. VEGF (A), Cyclin D1 (B), Caspase-3 (C) and Caspase-8 (D) gene expression analysis of EAC cells collected from tumor bearing mice treated with with rutin, Rutin–
zinc(II), paclitaxel, paclitaxel in combination with rutin and Rutin–zinc(II). ANOVA (p < 0.0001), ***p < 0.0001 (Tukey–Kramer post-hoc test).
190 N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191

(200 lM) and fibroblasts (500 lM), which corroborates our find- 5. Conclusion
ings [30]. The cytotoxic activity showed by Rutin–zinc(II) was also
observed in a Quercetin–zinc(II) complex that has the property to In the present study, the results demonstrated that the newly
intercalate into C–G DNA sequences suggesting metal ion coordi- synthesized molecule Rutin–zinc(II) is more efficient as an antiox-
nation with oxygen atom in the phosphodiester backbone resulting idant agent than free rutin. Moreover, Rutin–zinc (II) has not
in cleavage of DNA in vitro [31]. Transition metal complexes like shown any cytotoxicity against normal cells or toxicity in BALB/c
platinum complexes have been utilized in cancer treatment based mice, but has shown antioxidant activity and cytotoxicity against
on their successful antitumor activity derived from their ability to cancer cell lines in vitro and synergistic antitumor activity prevent-
form covalent adducts with DNA by ligand exchange [32]. DNA is ing side effects of chemotherapy. The data obtained from the UV–
generally the major biological target of platinum compounds and VIS, FT-IR, mass spectrometry and NMR studies indicate that Zn(II)
their cytotoxicity is correlated with the formation of is coordinated to rutin in the benzopyrane ring (positions 4 and 5)
1,2-intrastrand adducts between the N7 atoms of two adjacent and in the aromatic ring (30 and 40 positions). The increase in bio-
purine residues [33]. The in vivo studies in EAC bearing mice logical activity of Rutin–zinc(II) complex could be associated with
demonstrated that in comparison with rutin, Rutin–zinc(II) signif- the coordination of Zn(II) in those positions.
icantly induced DNA fragmentation in tumor cells. However, in
combination with paclitaxel, both rutin and Rutin–zinc(II) signifi- Conflict of interest
cantly enhanced DNA fragmentation. Paclitaxel is a diterpenoid
isolated from Taxus brevifolia, used clinically for the treatment of The authors declare that there is no conflict of interests regard-
ovarian and breast cancer and primarily kills cancer cells via ing the publication of this paper.
microtubule stabilization [34,35]. The loss of mitochondrial mem-
brane potential can activate caspase signaling pathways triggering
Transparency Document
the release of cytochrome c and other apoptotic factors to the cyto-
plasm [36]. Although the treatment with rutin and Rutin–zinc(II)
The Transparency document associated with this article can be
significantly decreased the mitochondrial membrane potential
found in the online version.
the effects of the combination with paclitaxel were the strongest.
The treatments demonstrated antiproliferative and antiangiogenic
activity modulating genes related to cell cycle progression, angio-
genesis and apoptosis with decreased cyclin D1 and VEGF expres- Acknowledgements
sion and increased expression of initiator and executor caspases-8
and -3. However, the combination of Rutin–zinc(II) with paclitaxel Financial support from FAPESP is gratefully acknowledged.
showed synergistic antitumor activity, preventing anemia and
myelossuppression, undesirable side effects of cancer and References
chemotherapy, and in addition increased lifespan of EAC bearing
mice as observed in Fig. 7. Metal–flavonoid chelates, like com- [1] M.J. Balunas, A.D. Kinghorn, Drug discovery from medicinal plants, Life Sci. 78
plexes of rutin with Fe(II), Fe(III), Cu(II) and Zn(II), play an impor- (5) (2005) 431–441.
[2] E. Middleton, The effects of plant flavonoids on mammalian cells: implications
tant role in protecting biological macromolecules, such as proteins, for inflammation, heart disease, and cancer, Pharmacol. Rev. 52 (4) (2000)
lipids and DNA, from oxidative stress that contribute to cancer, 673–751.
atherosclerosis, cardiovascular diseases, aging and inflammatory [3] R.J. Nijveldt, E. van Nood, D.E. van Hoorn, P.G. Boelens, K. van Norren, P.A. van
Leeuwen, Flavonoids: a review of probable mechanisms of action and potential
diseases [37,38]. Within the same in situ environment, malignancy applications, Am. J. Clin. Nutr. 74 (2001) 418–425.
develops under conditions in which the malignant cells are suscep- [4] S. Martens, A. Mithöfer, Flavones and flavone synthases, Phytochemistry 20
tible to the potential cytotoxic effects of the cellular zinc levels that (2005) 2399–2407.
[5] L. Le Marchand, Cancer preventive effects of flavonoids:a review, Biomed.
exist in the normal cells. Sliwinsky and colleagues proposed that Pharmacother. 56 (2002) 296–301.
the mechanism of such action of zinc involves enhanced expres- [6] C. Kandaswami, L.T. Lee, P.P. Lee, J.J. Hwang, F.C. Ke, Y.T. Huang, M.T. Lee, The
sion of anti-apoptotic Bcl-2 and Bcl-XL proteins and increased antitumor activities of flavonoids, In Vivo 5 (2005) 895–909.
[7] M.K. Chahar, N. Sharma, M.P. Dobhal, Y.C. Joshi, Flavonoids: a versatile source
expression of proteins of mitogenic signaling pathways, including
of anticancer drugs, Pharmacog. Rev. 9 (2001) 1–12.
p53, AKT and MAP kinases [39]. The results demonstrated that [8] I.B. Afanas’ev, E.A. Ostrakhovitch, E.V. Mikhal’chik, G.A. Ibragimova, L.G.
the flavonoid metal-complex Rutin–zinc(II) could be a potential Korkina, Enhancement of antioxidant and anti-inflammatory activities of
chemopreventive and therapeutic anticancer agent. bioflavonoid rutin by complexation with transition metals, Biochem.
Pharmacol. 61 (6) (2001) 677–684.
[9] Y. Yang, H. Lin, K. Su, C. Chen, Y. Yu, C. Lin, S. Yu, H. Yan, K. Su, Y.S. Chen, ‘‘Rutin,
a flavonoid that is a main component of saussurea involucrata, attenuates the
senescence effect in d-Galactose aging mouse model, Evid. Based Complement.
Altern. Med. 2012 (2012) 1–10.
Control (untreated)
Rutin [10] J.C.T. Carvalho, J.A.A. Sertie, M.V.J. Barbosa, Anti-inflammatory activity of the
Rutin-Zn(II) crude extract from the fruits of Pterodon emarginatus Vog, J. Ethnopharmacol.
100 Rutin+paclitaxel 64 (2) (1999) 127–133.
Rutin-Zn(II)+paclitaxel
[11] L. Yu, K. Zhou, J.W. Parry, Inhibitory effects of wheat bran extracts on human
80 Paclitaxel
LDL oxidation and free radicals, LWT Swiss Soc. Food Sci. Technol. 38 (5)
Percent survival

(2005) 463–470.
60 [12] M. Nikishimi, N. Rao, K. Yagi, The occurrence of superoxide anion in the
reaction of reduced phenazine methosulfate and molecular oxygen, Biochem.
40 Biophys. Res. Commun. 46 (2) (1972) 849–854.
[13] T. Mossman, Rapid colorimetric assay for cellular growth and survival:
20 application to proliferative and citotoxicity assays, J. Immunol. Methods 65
(1–2) (1983) 55–63.
0 [14] A.M. Boldi, Libraries from natural product-like scaffolds, Curr. Opin. Chem.
0 2 4 6 8 10 12 14 16 18 20 22 24 Biol. 8 (3) (2004) 281–286.
Time (days) [15] S. Shang, D.S. Tan, Advancing chemistry and biology through diversity-
oriented synthesis of natural product-like libraries, Curr. Opin. Chem. Biol. 9
Fig. 7. Kaplan–Meier survival curve for EAC bearing mice treated with rutin, Rutin– (3) (2005) 248–258.
zinc(II), paclitaxel, paclitaxel associated with rutin and Rutin–zinc(II). [16] V.A. Kostyuk, A.I. Potapovich, T.V. Kostyuk, M.G. Cherian, Metal complexes of
p = 0.0043(Log-Rank test). dietary flavonoids: evaluation of radical scavenger properties and protective
N.E.A. Ikeda et al. / Chemico-Biological Interactions 239 (2015) 184–191 191

activity against oxidative stress in vivo, Cell. Mol. Biol. (Noisy-le-Grand) 53 (1) [28] L.G. Menon, R. Kuttan, G. Kuttan, Inhibition of lung metastasis in mice induced
(2007) 62–69. by B16F10 melanoma cells by polyphenolic compounds, Cancer Lett. 95 (1–2)
[17] S.R. Maxwell, Prospects for the use of antioxidant therapies, Drugs 49 (3) (1995) 221–225.
(1995) 345–361. [29] C. Martínez Conesa, V. Vicente Ortega, M.J. Yáñez Gascón, M. Alcaraz Baños, M.
[18] A. Braca, C. Sortino, M. Politi, I. Morelli, J. Mendez, Antioxidant activity of Canteras Jordana, O. Benavente-García, J. Castillo, Treatment of metastatic
flavonoids from Licania licaniaeflora, J. Ethnopharmacol. 79 (3) (2002) 379– melanoma B16F10 by the flavonoids tangeretin, rutin, and diosmin, J. Agric.
381. Food Chem. 53 (17) (2005) 6791–6797.
[19] D. Malesev, V. Kuntic, Investigation of metal–flavonoid chelates and the [30] M. Matsuo, N. Sasaki, K. Saga, T. Kaneko, Cytotoxicity of Flavonoids toward
determination of flavonoids via metal-flavonoid complexing reactions, J. Cultured Normal Human Cells, Biol. Pharm. Bull. 28 (2) (2005) 253–259.
Serbian Chem. Soc. 72 (10) (2007) 921–939. [31] F. Pierard, A. Kirsch-De Mesmaeker, Bifunctional transition metal complexes as
[20] J. Tan, B. Wang, L. Zhu, DNA binding, cytotoxicity, apoptotic inducing activity, nucleic acid photoprobes and photoreagents, Inorg. Chem. Commun. 9 (2006)
and molecular modeling study of quercetin zinc(II) complex, Bioorg. Med. 111–126.
Chem. 17 (2009) 614–620. [32] F. Zobi, B. Spingler, T. Fox, R. Alberto, Toward novel DNA binding metal
[21] S. Yamasaki, K. Sakata-Sogawa, A. Hasegawa, T. Suzuki, K. Kabu, E. Sato, T. complexes: structure and basic kinetic data of
Kurosaki, S. Yamashita, M. Tokunaga, K. Nishida, T. Hirano, Zinc is a novel [M(9MeG)2(CH3OH)(CO)3]+(M=99Tc, Re), Inorg. Chem. 42 (9) (2006) 2818–
intracellular second messenger, J. Cell Biol. 177 (4) (2007) 637–645. 2820.
[22] M. Murakami, T. Hirano, Intracellular zinc homeostasis and zinc signaling, [33] Y. Wang, Z. Yang, Synthesis, characterization and DNA-binding properties of
Cancer Sci. 99 (8) (2008) 1515–1522. three 3d transition metal complexes of the Schiff base derived from
[23] T. Sliwinski, A. Czechowska, M. Kolodziejczak, J. Jajte, M. Wisniewska- diethenetriamine with PMBP, Transition Met. Chem. 30 (2005) 902–906.
Jarosinska, J. Blasiak, Zinc salts differentially modulate DNA damage in [34] M. Ceruti, P. Crosasso, P. Brusa, S. Arpicco, F. Dosio, L. Cattel, Preparation,
normal and cancer cells, Cell Biol. Int. 33 (4) (2009) 542–547. characterization, cytotoxicity and pharmacokinetics of liposomes containing
[24] L.C. Costello, R.B. Franklin, Cytotoxic/tumor suppressor role of zinc for the water-soluble prodrugs of paclitaxel, Control Rel. 63 (2000) 141–153.
treatment of cancer: an enigma and an opportunity, Expert Rev. Anticancer [35] S. Chen, X. Chen, Y. Xiu, K. Sun, Z. Zong, Y. Zhao, MicroRNA 490–3P enhances
Ther. 12 (1) (2012) 121–128. the drug-resistance of human ovarian cancer cells, J. Ova. Res. 7 (1) (2014) 84.
[25] S.B. Ere, C. Karaalp, E. Bedir, H. Kaehlig, S. Glasl, S. Khan, L. Krenn, Secondary [36] V. Gogvadze, S. Orrenius, B. Zhivotovsky, Multiple pathways of cytochrome c
metabolites of Centaurea calolepis and evaluation of cnicin for anti- release from mitochondria in apoptosis, Biochim. Biophys. Acta 757 (2006)
inflammatory, antioxidant, and cytotoxic activities, Pharm. Biol. 49 (8) 639–647.
(2011) 840–849. [37] S.R. Maxwell, Prospects for the use of antioxidant therapies, Drugs 49 (1995)
[26] A. Ravi, M. Alvala, V. Sam, A.M. Kalle, V.K. Irlapati, B.M. Reddy, Anticancer 345–361.
activity of Pupalia lappacea on chronic myeloid leukemia K562 cells, in: Daru: J. [38] A. Braca, C. Sortino, M. Politi, I. Morelli, J. Mendez, Antioxidant activity of
Facul. Pharm. 20 (1) (2012) 86. flavonoids from Licania licaniaeflora, J. Ethnopharmacol. 79 (2002) 379–381.
[27] Y. Cong, J. Guo, T. Wang, M. Li, K. Li, J. Wang, Q. Li.Chemical constituents and [39] T. Sliwinski, A. Czechowska, M. Kolodziejczak, J. Jajte, M. Wisniewska-
antitumor activity on leukemia K562 cell of Leonurus heterophyllus, Zhongguo Jarosinska, J. Blasiak, Zinc salts differentially modulate DNA damage in
Zhong Yao Za Zhi 34 (14) (2009) 1816–1818. normal and cancer cells, Cell Biol. Int. 33 (2009) 542–547.

You might also like