Você está na página 1de 10

Environmental Toxicology and Pharmacology 56 (2017) 383–392

Contents lists available at ScienceDirect

Environmental Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/etap

Research Paper

A zinc Schiff base complex inhibits cancer progression both in vivo and in MARK
vitro by inducing apoptosis
Arpita Banerjeea,c,1, Kaushik Banerjeea,1, Abhinaba Sinhaa, Satyajit Dasa, Saikat Majumderb,

Subrata Majumdarb, Soumitra Kumar Choudhuria,
a
In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
b
Division of Molecular Medicine, Bose Institute, P 1/12, C. I. T. Road, Scheme – VIIM, Kolkata, 700054, West Bengal, India
c
Department of Chemistry, Rammohan College, 102/1, Raja Rammohan Sarani, College Street, Kolkata, West Bengal 700009, India

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer chemotherapy suffers from selectivity and undesired toxicity of the drugs. Since zinc is a biocompatible
Zinc complex tracer element and cysteine derivatives are used in cancer chemoprevention, we intend to develop a complex of
Apoptosis zinc and cysteine-derivatives as potent, non-toxic anticancer agents. Herein, we synthesized and characterized
T-Lymphoblastic leukemia cysteine based ligand, 2-[(2-Hydroxy-3-methoxy-benzylidene)-amino]-3-mercapto-propionic acid and its Zn-
Ehrlich ascites carcinoma
complex, which are found to be non-toxic towards normal human PBMC. Data also revealed that only Zn-
Multidrug resistance
complex exhibited remarkable apoptosis in drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000
cancer cells as assessed by MTT, Cell cycle and AnnexinV binding assay. Moreover, Zn-complex altered ROS and
GSH level of the respective cell lines. Finally, treatment of Zn-complex in Swiss albino mice did not show any
systemic toxicity in preliminary trials in normal mice and remarkably increased the life-span of EAC bearing
mice. In conclusion, the synthesized Zn-complex may be developed for efficacious, multidrug resistance reversal,
non-toxic chemotherapeutic agents in future.

1. Introduction component of various metalloenzymes and transcription factors (Vallee


and Auld, 1989; Emami et al., 2007). Zinc complexes plays vital roles in
The major impediments for the successful treatment of cancer are biological fields as radioprotective agents (Huang et al., 2006), tumor
the cytotoxicity due to lack of selectivity of different antineoplastic photosensitizers (Nakayama et al., 2008), antidiabetic insulin mimetic
agents between healthy and cancer cells as well as development of (Sakurai et al., 2008; Katiyar and Singh, 2008) and antibacterial or
acquired multidrug resistance (MDR) in cancer. Recent studies disclose antimicrobic agents (Katiyar and Singh, 2008; Chohan et al., 2007).
some differences between normal and cancer cells (Dhawan and Divalent zinc [Zn(II)] ions can act as an effective Lewis acid and at the
Chadha, 2010). For example, the relationship between zinc in- same time it has no ligand field stabilization energy and thereby can
sufficiency and cancer has been observed in human, animal and cell easily adapt any coordination geometry to meet the requirement for a
culture studies (Sharif et al., 2012). Zn deficiency cause oxidative DNA particular reaction. Zinc is also significantly nontoxic (Beraldo and
damage and chromosomal breaks in animals (Ho et al., 2003; Oteiza Gambinob, 2004) compared to other metals viz, iron, copper, mercury
et al., 2008; Oteiza et al., 1995; Golub et al., 1985; O’Halloran, 1993) (Vallee and Falchuk, 1993), even at higher doses. Although zinc (II)
and plays cardinal role in DNA synthesis, gene regulation and induction ions possess diverse physiological activities its application as cytotoxic
of apoptosis (Liu et al., 2013; Ott and Gust, 2007). Cancer che- mediator on human cancer cell lines is still obscure (Jiang et al., 2009;
motherapeutic agents primarily cause DNA damage, impede cancer Belicchi et al., 2001; Sheng et al., 2008; Trávníček et al., 2006;
cells proliferation and ultimately induce apoptosis (Zuber et al., 1998; Rodrıguez-Arguelles et al., 2004; Miyamoto et al., 1998). Herein we
Moucheron, 2009; Maheswari et al., 2007; Clarke et al., 1999; Tan report the synthesis and characterization of a novel zinc complex,
et al., 2009; Bazzicalupi et al., 2008; Liu and Wang, 2009). Zinc com- Zn2L2(H2O)2 (henceforth will be mentioned as Zn2L2) and its anti-
plexes cleaves DNA molecule via hydrolytic pathway (Vallee and Auld, proliferative activity against human T lymphoblastic leukemia cell lines
1990; Bae et al., 2003). Zinc is an essential trace element and necessary (in vitro) and against murine Earlich ascites carcinoma cells (in vivo).


Corresponding author.
E-mail address: soumitrag10@gmail.com (S.K. Choudhuri).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.etap.2017.11.004
Received 5 April 2017; Received in revised form 3 November 2017; Accepted 4 November 2017
Available online 06 November 2017
1382-6689/ © 2017 Elsevier B.V. All rights reserved.
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

The rationale behind designing the reported cysteine-based zinc com- 2.3. Preparation of doses for both in vivo and in vitro treatments
plex is multi-dimensional. The thiol group in cysteine can act as a nu-
cleophile in enzymatic reactions. Although the role of different cysteine A stock solution of Zn2L2 (10−2 M) was prepared by dissolving it in
derivatives in chemoprevention and cure has been reported (Lee et al., acid-water just before the in vitro experiments. Then the stock solution
2011; Casini et al., 2002; Li et al., 2008), the role of cysteine as a metal was serially diluted with RPMI medium supplemented with 10% FBS
chelator of a non-toxic metal like zinc has not yet been explored. Schiffs for studies; for in vivo experiments, the complex, Zn2L2 (25 mg) was
bases and their metal complexes as cytotoxic agent have been reported dissolved in 500 μL 1(N) HCl and diluted to 5 mL by adding PBS to
in recent years (Ganguly et al., 2014; Kumar et al., 2010; Tarafder et al., prepare a 5 mg/mL stock solution. This stock solution was serially di-
2000) but cysteine containing Schiff base ligand with zinc has not luted with PBS and was injected intraperitoneally (i.p) to achieve the
previously been reported. desired concentrations. As for vehicle controls, equal volumes of
medium or PBS were added to untreated cells or injected i.p for in vitro
or in vivo experiments respectively.
2. Material and methods
2.4. Animal study
2.1. Chemicals and instruments
Swiss albino mice, originally obtained from National Institute of
Cysteine.HCl, MTT dye [3-(4,5-dimethylthiazol-2-yl)-2,5-diphe- Nutrition (Hyderabad, India) and reared in the institute animal facil-
nyltetrazolium bromide], propidium iodide and Histopaque 1077 were ities, were used for all the experimental purpose with prior approval of
purchased from Sigma Chemical Company (St. Louis, MO). RPMI 1640 the institutional animal ethics committee (IAEC). The experimental
medium and foetal bovine serum were procured from GIBCO Invitrogen protocols (Approval ID: IAEC-1.2/SKC-7/2007/4 dated 22.01.2008)
Corp. (Carlsbad, California). Penicillin and streptomycin were bought described herein were approved by the IAEC (Registration No.: 175/99/
from Himedia (Mumbai, India). FITC-labeled Annexin V and 2′,7′-di- CPCSEA, dated 28.01.2000) in accordance with the ethical guidelines
hydrodichlorofluorescein diacetate (H2-DCFDA) were obtained from laid down by the committee for the purpose of Control and Supervision
Molecular Probes TM, Invitrogen, (Carlsbad, California). o-vanillin of Experiments on Animals (CPCSEA) by the Ministry of Social Justice
(Acros Organics, New Jersey, USA), Zn(CH3COO)2. 2H2O (Merck, and Empowerment, Government of India. Adult female Swiss albino
Mumbai, India), RNase A (Genei, Bangalore, India), were also pur- mice (weighing 18–20 gm, aged between 4 and 6 weeks old) were ac-
chased from the manufacturer. All other reagents were of AR grade and climatized to the experimental room having temperature 25 ± 2 °C,
used without further purification. Elemental analyses (carbon, hy- controlled humidity conditions, and with photo cycle of 12 h light/12 h
drogen, and nitrogen) were performed using a Perkin Elmer 2400 Series dark. The mice were housed in sterile polypropylene cages containing
II CHN analyzer. Infrared spectra (4000–400 cm−1) were recorded at sterile paddy husk as bedding material, fed on autoclaved standard
28 °C on a Perkin-Elmer RX 1 FT spectrophotometer using KBr as mice food pellets and water ad libitum.
medium. UV–vis spectra were recorded in Shimadzu UV 160 A and in
Varian Cary 100 Scan in the range of 800–200 nm. Mass spectrum was 2.5. In vivo cell line, tumor transplantation and experimental protocol
recorded in a Waters Micromass Quattro micro API mass spectrometer
mass spectrometer. Cell survival was evaluated by ELISA reader (Tecan EAC cell was maintained as an ascitic tumor in female Swiss albino
200). The cell cycle analyses and annexin-V binding assay were per- mice. Six groups (each group containing six mice) of animals were
formed using a FACS calibur flow cytometer (Becton Dickinson, USA). taken for animal survival studies. Each mouse was inoculated with 106
Fluorescence was measured in Varian Cary eclipse spectrofluorimeter. EAC cells (six groups) intra-peritoneally. Following 9 days of inoculums
Haematological parameters were analyzed by automated haematology (i.p) Zn2L2 of various doses (20 mg kg−1, 25 mg kg−1, 30 mg kg−1 and
analyzer (Sysmex, KX-21) and haematologic biochemical analyses 50 mg kg−1 of body weight) were injected (i.p) to EAC bearing mice
(blood urea, creatinin, alkaline phosphatase, SGPT, SGOT) were de- while one group kept untreated (control) and another was kept as ve-
termined by automated clinical chemistry analyzer (Olympus, AU400). hicle control. Animals were daily inspected for the assessment of ascitic
load, and body weights were measured. Time of death was recorded for
calculation of mean survival time (MST). The MSTs were recorded for
2.2. Synthesis of the ligand (H2L) and zinc complex (Zn2L2) each group. Mouse survival periods in different groups were also
compared as treated/control (T/C) ratio (percent), i.e., the ratio of the
Ligand (H2L): Cysteine.HCl (1.57 g, 10 m mol) was dissolved in 0.01 survival time (in days) for treated mice to control (untreated) mice.
(M) NaOH. An ethanolic solution of o-vanillin (1.52 g, 10 m mol) was According to standard National Cancer Institute protocols for screening
added slowly drop wise to the cysteine solution. Resulting mixture was new anticancer drugs, it is considered that the increase in survival time
stirred in cold environment (4 °C) and after 30 mins a light precipitate corresponding to T/C ratios around 120% to be “marginal”, T/C ratios
deposited. Again 10 mL ethanol and 0.01(M) NaOH was added to the between 120 and 150% to be “clear” and T/C ratios equal or superior to
mixture and stirred for 2 h. A pale yellow precipitate of 2-[(2-Hydroxy- 150% to be “marked”.
3-methoxy-benzylidene)-amino]-3-mercapto-propionic acid (H2L) was
deposited. The compound filtered and recrystallised from MeOH and 2.6. In vivo toxicity testing
dried in vacuum. Yield 1.1 g, 43.3%; m.p.133 °C; Anal. Found (%); C
51.69; H 4.87; N 5.56. Calcd for C11H13NO4S (%): C 50.74; H 4.92; N To evaluate preliminary sub acute toxicity of Zn2L2, seven experi-
5.65. mental groups (six mice per group) of normal female Swiss albino mice
Zinc complex (Zn2L2): According to the above mentioned method were taken. All the experiments were performed in triplicate. The Zn2L2
H2L was prepared; the pale yellow precipitate was dissolved in NaOH (25 mg) was dissolved in 500 μL ∼ 1(N) HCl and diluted to 5 mL by
(0.01 M) and stirred well until a clear solution was obtained. Aqueous adding PBS to prepare a 5 mg/mL stock solution. This stock solution
solution of Zn(CH3COO)2. 2H2O (2.19 g, 1 m mol) was added in situ and was serially diluted with PBS and was injected intra-peritoneally to
stirred again for 2 h. The resulting zinc complex, Zn2L2(H2O)2 (yellow achieve a final concentration of 20, 25, 30, 50 and 100 mg per kg body
precipitate) was filtered off and washed with EtOH. Yield 3.6 g, weight. During the experimental period, clinical appearance of the mice
73.73%; m.p. > 300 °C; Anal. Found (%);C 41.40; H 3.52; N 4.39; S was daily evaluated. Blood was collected from treated, untreated and
9.95. Calcd for C22H24N2O8S2Zn2: C 41.50; H 3.45; N, 4.40; S 10.06. vehicle control mice via closed cardiac puncture with the help of a 22-
guage hypodermic needle by subxiphoid approach after 14 days of

384
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

treatment to evaluate toxicological parameters. Blood from each group diphenyl-2H-tetrazolium bromide (MTT) for 4 h at 37 °C, 5% CO2
was pooled into separate glass tubes and treated with heparin (antic- (Mossmann, 1983). Cell survival was evaluated by measuring the ab-
oagulant) or left untreated for serum collection. sorbance at 540 nm by ELISA reader. The control value corresponding
to untreated cells was taken as 100% and the viability of treated sam-
2.7. Isolation of bone marrow cells ples were expressed as a percentage of the control. All experiments
were performed in triplicate. The data were used for the construction of
In order to find out preliminary bone marrow toxicity following response curves using the Graph Pad Prism software.
treatment, untreated and Zn2L2 treated female Swiss albino mice were
anaesthetized and the femur bone was cut with the help of a vertebrate
scissor. Bone marrow was flushed with 0.56% KCl solution and cen- 2.11. Cell cycle analysis
trifuged at 3000 rpm for 15 min at 37 °C. Cell viability was tested by
0.4% trypan blue dye solution and cells were counted in a phase con- For the cell cycle analysis cells were seeded in 60 mm tissue culture
trast microscope. The experiment was repeated thrice. The average plate at a density of 5 × 105/plate and allowed to settle for 24 h prior
value of percentage of viable bone marrow cells was calculated from to treatment with complex, Zn2L2. At various time points (12 h, 24 h,
three independent experiments. 48 h, 72 h and 96 h), cells were recovered, washed twice in PBS, fixed
in cold 70% ethanol, and stored at 4 °C until analyzed. Then the cells
2.8. Isolation of spleen cells were washed twice in PBS, incubated for 1 h at room temperature with
250 μg/mL RNase A and for 20 min at 4 °C with 20 μg/mL propidium
Zn2L2 treated and untreated female Swiss mice were anesthetized iodide (PI) in dark. The cell cycle distribution and percentage of
by chloroform and 70% alcohol was sprayed on abdominal region. apoptotic cells were determined using a FACS calibur flow cytometer.
Spleen was removed aseptically and small amount of PBS was injected Ten thousand events were analyzed for each sample. Appropriate gating
to it; a small portion of it was rubbed against an opaque frosted glass was used to select the single-cell population. The same gate was used on
slide. The cell suspension formed is spinned at 1000–1500 rpm for all samples, ensuring that the measurements were made on a standar-
5–10 min. The supernatant was discarded and the cells were washed by dized cell population. The apoptotic cells were considered to constitute
spinning in PBS twice at room temperature. Cell viability was tested by the sub G0/G1 cell population and percentage of cells in each phase of
0.4% trypan blue solution and cells were counted in a phase contrast cell cycle was calculated (Pozarowski and Darzynkiewicz, 2004).
microscope. The experiments were performed in triplicate.

2.9. In vitro cell lines and culture conditions 2.12. Apoptosis assay

The doxorubicin resistant and sensitive human acute T-lympho- Cells (5 × 105) were cultured in 60 mm tissue culture plate and
blastic leukaemia cell lines CEM/ADR5000 and CCRF-CEM respectively allowed to settle for 24 h before treatment with Zn2L2. Cells were then
were kindly provided by Prof. T. Efferth, University of Mainz, Germany. incubated with FITC labeled Annexin V and propidium iodide (PI) at
The doxorubicin resistant CEM/ADR5000 cell line was generated by room temperature for 15 min in the dark and analyzed using a FACS
treating CCRF-CEM cells with doxorubicin doses up to a final con- Calibur flow cytometer (Vermes et al., 1995).
centration of 5000 ng ml−1 doxorubicin (Kimmig et al., 1990). Due to
the doxorubicin selection pressure CEM/ADR5000 specifically over-
expresses P-glycoprotein without concomitant over-expression of MRP1 2.13. Intracellular ROS accumulation study
or BCRP (Gillet et al., 2004). Furthermore, the cross-resistance profile
of CEM/ADR5000 cells to a broad range of established anti-cancer Cells (5 × 105) were cultured in 35 mm tissue culture plate and
drugs and investigative novel compounds have been analyzed (Efferth allowed to settle for 24 h and then the cells were treated with complex
et al., 2008). Therefore, to maintain the doxorubicin resistance property Zn2L2 and incubated at 37 °C, 5% CO2 for 1h–6 h or left untreated. The
in CEM/ADR5000, the cells requires undergoing a continuous selection cells were then washed with PBS and further incubated with H2-DCFDA
pressure of doxorubicin (5000 ng ml−1 doxorubicin) in the growth (2′, 7′ dihydrodichlorofluorescin diacetate) for 30 min at 37 °C in dark.
culture media. Thus, both the cell lines were sub cultured at every 72 h After incubation, cells were washed twice in PBS at room temperature
and CEM/ADR5000 cells were maintained on every 15th day in the for 5 min each time. The fluorescence was measured at excitation and
presence of 5000 ng ml−1 doxorubicin for the period of 72 h. After that, emission wavelengths of the oxidized form at 504 nm and 529 nm re-
the doxorubicin containing medium was replaced with fresh RPMI spectively (Ganguly et al., 2011).
medium and cultured for subsequent generations. In all experiments,
CEM/ADR 5000 cells were used that were cultured without doxorubicin
and doxorubicin treatment of the next immediate generation was also 2.14. Determination of intracellular GSH contents
avoided. These cell lines were cultured in RPMI medium supplemented
with 10% heat inactivated foetal bovine serum (FBS), glutamine Determination of cellular GSH content was performed by a mod-
(0.15%), HEPES (25 mM) 66.67 mg/L penicillin and 100 mg/L strep- ification of a previously reported method (Sedlak and Lindsay, 1968).
tomycin. Cells were grown in plastic tissue culture flasks with a hu- Cells (5 × 105) were cultured (as stated earlier in the section of ‘cell
midified atmosphere of 5% CO2 in incubator at 37 °C. Cells from ex- cycle analysis’) and then Zn2L2 treated and untreated cells were washed
ponentially growing cultures of passage 15 or less were used for all three times with cold washing buffer (0.1 M sodium phosphate, 5 mM
experiments. All experiments were repeated three times. ethylenediaminetetraacetate [EDTA], pH 7.5) and immediately lysed in
100 mL lysis buffer (0.1% Triton-X, 0.1 M sodium phosphate, 5 mM
2.10. Cytotoxicity assay EDTA, pH 7.5). Five minutes later, the cell lysates were acidified with
15 μL of 0.1 N HCl, and protein was precipitated with 15 μL of 50%
For the proliferative assay, 4 × 104 cells were cultured in 96 mi- sulfosalicylic acid. Following centrifugation at 12,000g for 15 min, su-
crowell plates for 24 h at 37 °C, 5% CO2. Then H2L and its Zn2L2 were pernatants were collected for GSH measurement. The supernatants
added in microwells containing the cell at final concentrations of 10−8 were assayed for GSH content by spectrophotometric determination of
M–10−4 M. Following incubation for 48 h and 72 h the cells were in- the reduction of DTNB to 5′- thio-2-nitrobenzonic acid at 412 nm wave
cubated further with 5 mg per ml of 3-(4,5-dimethylthiazol-2-yl)-2,5- length.

385
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

2.15. Peripheral blood mononuclear cell (PBMC) isolation and culture


conditions

Heparinized peripheral blood of human was collected from healthy


volunteers and diluted with equal volume of RPMI 1640. Blood drawn
from healthy volunteers for the isolation of PBMC are done with prior
approval of the Institutional Ethics Committee (IEC). The experimental
protocols [IEC Ref: CNCI/IEC-June-2011(3) dated 24/06/2011] de-
scribed herein were approved by the IEC (certified by the Department
of Clinical Biochemistry, Hospital Unit, Chittaranjan National Cancer
Institute) in accordance with the ethical guidelines laid down by the
committee for the purpose of Control and Supervision of Experiments
with human subjects by the Ministry of Social Justice and
Empowerment, Government of India. Informed consent for participa-
tion in the study was obtained from participants. The participants
stated that, ‘they have been fully explained about the nature and pur-
pose of the study by the supervisor and they agreed upon to participate
in the present study’. They have also been given the opportunity to
question about various aspects of the present study (Volunteer’s profile
given in Supplementary Table 1). Lymphocyte-enriched mononuclear
cells were isolated by Histopaque 1077 density gradient centrifugation
of diluted blood which was washed and finally re-suspended in cold
RPMI 1640 medium and supplemented with 10% heat inactivated FBS.

2.16. Statistical analysis Fig. 1. Plausible structure of the ligand H2L and its zinc complex Zn2L2(H2O)2.

All data reported in this communication are the arithmetic mean (Supplementary Figure S6). The plausible structure of the ligand and its
from three independent experiments performed in triplicate zinc complex is given in Fig. 1.
mean ± S.D. unless stated otherwise. The unpaired Student’s t-test was
used to evaluate the significance of differences between groups, ac-
cepting P < 0.05 as a level of significance. Data analyses were per- 3.2. 1H NMR spectrum
formed using the Prism software (GraphPad, San Diego, CA).
The ligand, 2-[(2-Hydroxy-3-methoxy-benzylidene)-amino]-3-mer-
3. Results capto-propionic acid (H2L) contains only 12 protons [3 non-equivalent
aromatic protons; 2 equivalent methelene protons; 3 non-equivalent
3.1. Chemical characterization methoxy protons; 2 equivalent acetylene protons; 1 phenolic-OH
proton; 1 thiol proton & 1 carboxylic proton] (Fig. 1). However, proton
The Schiff-base ligand (H2L) and its Zn2L2 were synthesized in situ NMR peaks for the ligand records only 12 protons instead of 13 protons
and characterized through routine physicochemical techniques. UV [3 non-equivalent aromatic protons; 2 methelene protons; 3 non-
spectral bands of ligand H2L (Supplementary Figure S1) and zinc equivalent methoxy protons; 2 equivalent HCN protons; 1 phenolic-OH
complex Zn2L2 (Supplementary Figure S2) was recorded in dimethyl proton & 1 thiol proton] (Supplementary Figure S7). The carboxylic
sulpoxide (DMSO) and acid-water respectively. No absorbance in the acid proton remains unascertained. Proton NMR peak of the ligand in
visible region was observed for both H2L and Zn2L2. Important UV DMSO-d6 recorded at δ 6.70–6.95 (s, 3H) for 3 non-equivalent aromatic
bands were observed at 236, 255, 279 and 338 nm. The λ values in the protons. Two equivalent eCH2 protons recorded at δ 3.55 (s, 2H). Three
UV region were due to π-π or n-π* transition of electrons in the com- non-equivalent eOCH3 protons recorded at 3.10–3.20 (m, 3H). One
pound. For Zn2L2 UV bands were observed at 270 nm (π → π*) and 340 phenolic-OH proton recorded at δ 5.64 (s, H). Furthermore, two
(S → Zn) nm and shifted to towards higher wave length i.e lower energy equivalent HCN protons recorded at δ 2.50–2.57 (s, 2H) and one thiol
indicating complex formation. The sharp peak in the IR spectrum of the (eSH) proton recorded at δ 2.26 (m, H) following experimental ana-
ligand H2L at 1637 cm−1 (Supplementary Figure S3) is characteristic of lysis. On the other hand, in the spectrum of the ligand, we could not
the azomethine functionality and the band at 1484 cm−1 is due to ascertain the carboxylic acid (normal range δ 10.1–11.3) proton peaks.
skeletal vibration of the aromatic moiety. A broad weak band at However, based on other elemental analysis (Mass, IR etc.), we have
2550 cm−1 corroborates with the presence of thiol group. A band at established the presence of eCOOH group in the ligand. Proton NMR
1279 cm−1 is observed due to stretching of phenolic CeO bond. In the peak of Zinc complex of the ligand (Zn2L2) in DMSO-d6 (Supplementary
IR spectrum of Zn2L2 (Supplementary Figure S4) the νC]N character- Figure S8) recorded at δ 6.3–6.53 (m, 3H) for 3 non-equivalent aro-
istic stretching band of the ligand at 1637 cm−1 shifted to a lower matic protons. Two equivalents eCH2 protons recorded at δ 3.43.
1626 cm−1 possibly due to zinc complex formation. Skeletal vibrations eOCH3 protons recorded at δ 3.6–3.8 (m, 3H). Furthermore, the HCN
observed at1445 cm−1. The broad weak band due to presence of thiol protons in the complex recorded at δ 2.45–2.53 (s, 2H) and carboxylic
group disappeared in complex may be indicative of coordination of acid proton at δ 10.26 (m, H). In NMR spectrum of the complex we
thiolate sulphur with zinc. Band observed at 3390 cm−1 indicates the observe a shift of electron density from the ligand to metal moiety.
existence of water molecule in the complex (Nakamoto, 2009). Mass Aromatic protons shift from the range of δ 6.70–6.95 in the ligand to δ
spectral study of H2L dissolved in DMSO showed formation of peak at 6.30–6.53 in the complex. Methelene protons shift from δ 3.55 in the
256.3 (calculated m/z: 256 amu) which corroborate well with the re- ligand to δ 3.43 in the complex. This is an indication of considerable
spective mono-positive species of formulation [LH]+ (Supplementary drift of electrons from two neighbouring groups viz. eN]C and
Figure S5). The Mass spectral study of Zn2L2 was performed in acid- eCOOH to the metal moiety. The shielding of the aromatic and me-
water. Formation of the peak at 637 (calculated m/z: 636 amu), cor- thelene protons indicates the coordination of the ligand to metal
roborate well with the mono-positive species of formulation [M+1] + moiety, i.e. formation of Zn2L2 complex. However, the methoxy protons

386
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

(eOCH3) in the ligand shifted to lower field; from ligand 3.10–3.20 to compound concentration (M). The same MTT experiment was also
3.6–3.8 in the complex due to deshielding of protons. This indicates the performed on normal human PBMC taking the same concentration and
participation of (CH3eC]Ne) group in coordination with zinc atom. time range. The data indicates that H2L and Zn2L2 did not impart any
Probably methoxy (eOCH3) protons were not pulled from ligand to significant toxicity on the normal cells at 10−4M or lower doses but
metal moiety. Furthermore, data revealed that one phenolic-OH proton only Zn2L2 (and not H2L) induces death to the malignant cells (Fig. 2).
peak and one thiol (eSH) proton peak of the ligand are disappeared in For this reason, only the Zn2L2 was used for biological evaluation. IC50
the complex. This indicates the involvement of phenolic-OH group and values for CEM/ADR5000 and CCRF-CEM were found to be 5 × 10−5M
eSH group in the bond formation with metal moiety. In addition to the and 10−4M respectively and were used for further in vitro studies. It is
DMSO-d6 solvent peak, another peak at 1.89 was observed. This free interesting to note that Zn2L2 induces death to the resistant cell line at a
proton at 1.89 (d ppm) may be assigned as protons peak of water of lower concentration.
crystallization.
On the basis of these assignments we possibly can infer that the 3.4. Cell cycle analysis
phenolic-OH group and eSH group are involved in bond formation with
metal moiety as a part of a rigid cyclic system and one water molecule Flow cytometric assay using propidium iodide (PI) has been widely
attached to the rigid structure of Zn2L2 in such a manner that the used for the evaluation of apoptosis in different experimental models.
protons of the water molecule remain a bit shielded. The flow cytometric assay of PI stained CCRF-CEM and CEM/ADR5000
cell lines was performed to assess the effect of the zinc complex on the
3.3. Cytotoxicity (MTT) assay DNA quantification to identify the cell cycle position. Both cell lines
were treated with concentration corresponds to IC50 values of Zn2L2.
Cytotoxicity of the ligand H2L and its zinc complex Zn2L2 on human The percentage of cells in each phase of the cell cycle was determined.
acute T-lymphoblastic leukaemia cell lines (CEM/ADR5000 and CCRF- It was found that in CCRF-CEM (Fig. 3) the counts of sub diploidal (sub
CEM) was performed by using MTT-assay (Fig. 2). Both H2L and Zn2L2 G1/G0; M1) cells increase (1.53%, 7.73%, 8.69%, 8.39% and 15.43%
were added in microwells containing the cell culture to attain final for 12 h, 24 h, 48 h, 72 h and 96 h respectively) with respect to un-
concentrations of 10−8M–10−4M. At different hours (48 h and 72 h) treated control (0.55%). The increase in sub G0/G1 (M1) population
the cell survival was evaluated by measuring the absorbance in the represents cells with significant DNA fragmentation pointed towards a
visible region. The IC50 (i.e., the concentration of the complex that re- late apoptotic stage. Flow cytometry of CEM/ADR5000 (Fig. 4) re-
stricts cell growth to 50%, compared with that of the control experi- vealed an enhanced number of cells in sub G1/G0 population in a time
ment) values for CEM/ADR5000 and CCRF-CEM were calculated from dependent manner (5.65%, 8.28%, 22.79%, 41.67% and 27.88% for
the curves constructed by plotting the cell survival (%) versus the 12 h, 24 h, 48 h, 72 h and 96 h respectively) with respect to untreated

Fig. 2. Cytotoxic effects of ligang H2L and omplex Zn2L2 on (a) CCRF-CEM, (b) CEM/ADR5000 and (c) human PBMC cell lines. Dose response curves for H2L and Zn2L2 were assessed by
MTT assay. Cells were seeded into 96-well plates (4 × 104 cells/well) and allowed to incubate overnight at 37 °C in a 5% CO2 incubator. On next day, cells were treated with increasing
concentrations of H2L and Zn2L2 and incubated for 48 and/72 h. Results were expressed as percentage viability of untreated cells. Value represents the mean ± SD of three independent
experiments with four replicates in each. *P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test.

387
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

Fig. 3. Cell cycle distribution of CCRF-CEM cells after treatment. CCRF-CEM cells treated with Zn2L2 (IC50) for indicated times were harvested and fixed in 70% ethanol. After staining
with propidium iodide they were analyzed using a flow cytometer.

control (2.26%). A sudden reduction in sub G1/G0 population at 96 h Zn2L2 may have the ability to suppress proliferation of drug resistance
may be attributed to formation cell debris (due to large number of cell cancer cells by blocking cell cycle progression and causing apoptosis.
death) that did not fall within the gated population. Moreover a sig-
nificant increase in number of cells in the G1/G0 (M2) phase of the cell
3.5. AnnexinV-FITC binding assay
cycle from 40.74% (untreated) to 47.24% (12 h) and eventually 54.13%
(24 h) was noted which is indicative of G1/S arrest of cell cycle. Thus
To further confirm and evaluate the induction of apoptosis, we

Fig. 4. Cell cycle distribution of CEM/ADR5000 cells after treatment. CEM/ADR5000 cells treated with Zn2L2 (IC50) for indicated times were harvested and fixed in 70% ethanol. After
staining with propidium iodide they were analyzed using a flow cytometer.

388
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

Fig. 5. Cell cycle distribution of (A) CCRF-CEM and (B) CEM/ADR5000 cells following treatment. Cells were treated with Zn2L2 (IC50) for indicated times were harvested and fixed in
70% ethanol. After staining with propidium iodide they were analyzed using a flow cytometer.

stained cells with annexin V-FITC and PI. Flow cytometric analysis of 3.8. In vivo toxicity
dual stained (annexin V-FITC and PI) CCRF-CEM (Fig. 5a) treated with
Zn2L2 at different time intervals (12 h, 24 h and 48 h) exhibited a A standardized study on the sub acute systemic toxicity (14 days) of
progressive decrease (13.10%, 10.31% and 7.89%) in the annexin V- Zn2L2 was performed on female Swiss albino mice taking as a model
FITC positive population of cells (untreated control;2.32%) and also system. In preliminary trials the complex administrated at different
showed a gradual increase in dual positive population (86.90%, doses intra-peritoneally in mice. Results indicated that the animals at
89.64%, 92.01% for 12 h, 24 h and 48 h respectively) as compared to different dose groups (up to 100 mg kg−1 of body weight) did not ex-
untreated control (0.83%). These results suggest that the complex set hibit any pathological changes as compared to untreated (control)
CCRF-CEM cells to death through induction of apoptosis. Same ex- group of animals. Any changes in biochemical parameters such as, le-
periment when performed on the resistant counterpart CEM/ADR5000 vels of serum glutamate ortho-transferase (SGOT), serum glutamine
(Fig. 5b) showed same trend that is, annexin V-FITC positive population pyruvate transaminase (SGPT), alkaline phosphatase (ALP) (indicative
diminishes gradually (53.02%, 42.56%, 28.55% for 12 h, 24 h and 48 h of hepatocellular injury) or creatinine and urea (indicative of impaired
respectively) and there is a steady raise in dual positive population renal function) are indicative of hepatic and/or renal toxicity. No
(44.59%, 55.49%, 72.85% for 12 h, 24 h and 48 h respectively) with mortality or toxic symptoms were observed in treated groups with re-
compared to untreated control (1.45%). Thus the raise in the above said spect to control groups of animals. No significant differences were ob-
population depicted that Zn2L2 induces apoptosis also to CEM/ served in the body weight gain/loss pattern, organ weight, hematolo-
ADR5000 cells. In both cases the drop in annexin-FITC positivity value gical (Table 1) or biochemical parameters (Table 2). Additionally there
indicates that the cells were entered into late apoptotic stage. was no change in spleen and bone marrow cell count after the treatment
for 14 days (Table 3). The results suggested that the selected doses of
3.6. Intracellular ROS accumulation study Zn2L2 at concentrations of 20 mg kg−1 bw, 25 mg kg−1 bw,
30 mg kg−1 bw, 50 mg kg−1 bw and 100 mg kg−1 bw are non-toxic.
In cancer cells excess ROS generation can serve as an endogenous Although the 100 mg kg−1 bw concentration is non toxic we did not
source of DNA-damaging agents that can promote apoptosis. This ob- proceed beyond 50 mg kg−1 bw as at 100 mg kg−1 bw the drug solu-
servation encouraged us to enquire whether the intra cellular ROS tion became acidic.
generation is involved in Zn2L2 mediated apoptosis. We measured in- Zn complex caused an increase in life span of Ehrlich ascites carcinoma
tracellular ROS (viz, hydroxidyl, peroxyl) using oxidation sensitive bearing mice. The potential of any anti-cancer drug can be judged by its
fluorescence dye DCFDA in CCRF-CEM and CEM/ADR 5000 cell lines ability to increase life span. For this purpose female Swiss albino mice
(Fig. 6). The synthesized zinc complex is capable of producing ROS in were inoculated through intra peritoneal route with the Ehrlich ascites
cancerous cells. It was found that the complex increases ROS within the carcinoma (EAC). 10 days after development of tumor, the animals
cells. Accumulation of ROS in experimental cell lines was observed and were treated with 20 mg kg−1 of bw, 25 mg kg−1 of bw, 30 mg kg−1 of
sustained up to the 6th h as compared to untreated control. The results bw and 50 mg kg−1 of bw of Zn2L2 and animals were monitored daily
suggested that these elevated levels of ROS (at a given experimental for any discomfort or pain. The results presented in Table 4 showed that
time frame) may be involved in promotion of apoptosis. Zn2L2 is able to enhance the survival of each group of treated mice
compared to untreated controls. But among the given doses only
30 mg kg−1 of bw showed an increase in T/C values upto 150%.
3.7. Determination of intracellular GSH content
Moreover, total volume of ascitic fluid and total packed cell volumes
were also reduced when measured following 14 days treatment
We measured intracellular GSH in CCRF-CEM and CEM/ADR 5000
(Table 4).
cell lines, following drug treatment (Fig. 7). Zn2L2 increases GSH level
significantly within the CEM/ADR 5000 cells. In its sensitive counter-
part CCRF-CEM it down regulates GSH level in early hours whereas in 4. Discussion
late hours non-significant increase of GSH level is observed (up to the
6th h) as compared to untreated control. The present work is based on our proposition that a coordination

389
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

Fig. 6. Measurement of Intra-cellular reactive oxygen species (ROS) generation treated with Zn2L2 on (A) CEM/ADR5000 and (B) CCRF-CEM cells. Cells were either kept untreated or
treated with Zn2L2 (IC50) and intra cellular ROS generation was measured [in terms of peroxide using dichlorofluorescein diacetate (DCF-DA)], at different time intervals. Data are
expressed as a percentage of the control and are presented as mean ± SD from 3 independent experiments. Differences between the control and treated cells are significant at
*P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test.

Fig. 7. Measurement of Intra-cellular glutathione (GSH) generation of (A) CEM/ADR5000 and (B) CCRF-CEM cells after treatment with Zn2L2. Cells were either kept untreated or treated
with Zn2L2 (IC50) and intra cellular GSH generation was measured at indicated time points. Data are expressed as a percentage of the control and are presented as mean ± SD from 3
independent experiments. Differences between the control and treated cells are significant at *P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test.

Table 1
Effect of Zn2L2 on haematological parameters of normal female Swiss albino mice.

Parameters Untreated Vehicle Control 20 mg/Kg 25 mg/Kg 30 mg/Kg 50 mg/Kg 100 mg/Kg

3
WBC (x10 /μL) 6.1 ± 0.7 6.9 ± 0.77ns 6.5 ± 0.72ns 8.1 ± 0.8ns 14.0 ± 3.0* 6.2 ± 0.7ns 8.5 ± 0.82ns
RBC (x106/μL) 10.3 ± 1.1 9.5 ± 0.85ns 9.92 ± 0.89ns 9.91 ± 0.89ns 10.41 ± 1.2ns 9.08 ± 0.9ns 9.56 ± 1.0ns
HGB (g/dL) 15.8 ± 3.1 14.4 ± 3.0ns 14.4 ± 2.9ns 13.5 ± 2.8ns 15.5 ± 2.9ns 14.6 ± 2.6ns 14.6 ± 2.7ns
HCT (%) 52.1 ± 10.2 48.6 ± 8.8ns 50.3 ± 9.7ns 48.1 ± 9.8ns 55.0 ± 11.2ns 47.5 ± 8.8ns 49.5 ± 9.3ns
MCV (fL) 50.8 ± 9.9 51.2 ± 10.0ns 50.7 ± 9.9ns 48.5 ± 9.1ns 52.8 ± 10.4ns 52.3 ± 10.1ns 51.8 ± 10.2ns
MCH (pg) 15.4 ± 2.9 15.2 ± 3.0ns 14.5 ± 2.4ns 13.6 ± 2.0ns 14.9 ± 2.7ns 16.1 ± 3.5ns 15.3 ± 2.9ns
PLT (x103/μL) 897 ± 89 954 ± 93* 868 ± 52.2ns 1057 ± 112* 656 ± 72** 350 ± 31*** 1262 ± 132***
LYM# (x103/μL) 5.2 ± 0.52 5.6 ± 0.53ns 5.8 ± 0.56ns 6.9 ± 0.78ns 8.6 ± 0.86* 5.5 ± 0.56ns 7.0 ± 0.72ns
RDW (fL) 29.9 ± 5.7 27.4 ± 5.2ns 28.7 ± 5.6ns 31.6 ± 6.0ns 29.1 ± 5.5ns 27.3 ± 5.1ns 28.3 ± 5.4ns
PDW (fL) 7.4 ± 0.8 8.0 ± 0.86ns 6.8 ± 0.75ns 7.5 ± 0.83ns 7.4 ± 0.80ns 7.2 ± 0.82ns 7.2 ± 0.83ns
MPV (fL) 6.1 ± 0.62 6.6 ± 0.68ns 6.0 ± 0.65ns 6.2 ± 0.70ns 6.5 ± 0.68ns 6.3 ± 0.64ns 6.3 ± 0.67ns
P-LCR (%) 2.7 ± 0.3 4.7 ± 0.57* 4.0 ± 0.51* 3.3 ± 0.43ns 5.9 ± 0.60* 5.2 ± 0.56* 4.6 ± 0.54*

Data are expressed as a percentage of the control and are presented as mean ± SD from 3 independent experiments. Differences between the control and treated cells are significant at
*P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test, ns: not significant.

Table 2
Effect of Zn2L2 on biochemical parameters (hepatic and renal functions) of normal female Swiss albino mice.

Parameters Untreated Vehicle Control 20 mg/Kg 25 mg/Kg 30 mg/Kg 50 mg/Kg 100 mg/Kg

Urea (mg/dL) 75 ± 6.9 84 ± 8.0ns 62 ± 5.8ns 60 ± 5.8* 41 ± 3.8* 82 ± 7.9ns 108 ± 11.0*
Creatinine (mg/dL) 0.3 ± 0.02 0.4 ± 0.05ns 0.3 ± 0.04ns 0.4 ± 0.03ns 0.4 ± 0.03ns 0.3 ± 0.02ns 0.4 ± 0.032ns
Alkaline Phosphatase (U/L) 179 ± 18.8 282 ± 29.3* 299 ± 30.1** 324 ± 31.4** 217 ± 22.5* 140 ± 13.7ns 200 ± 19.8*
SGOT (U/L) 212 ± 22.4 261 ± 25.8* 164 ± 17.5ns 206 ± 19.2ns 278 ± 26.9* 176 ± 16.6* 57 ± 6.2***
SGPT (U/L) 75 ± 7.8 100 ± 10.7* 71 ± 6.9ns 80 ± 7.8ns 76 ± 7.4ns 259 ± 24.7*** 79 ± 7.7ns

Data are expressed as a percentage of the control and are presented as mean ± SD from 3 independent experiments. Differences between the control and treated cells are significant at
*P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test, ns: not significant.

390
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

Table 3
Effect of Zn2L2 on spleen and bone marrow count of normal female Swiss albino mice.

Parameters Untreated Vehicle Control 20 mg/Kg 25 mg/Kg 30 mg/Kg 50 mg/Kg 100 mg/Kg

Spleen Cells (% of Viable Cells) 93.7 ± 1.56 93.9 ± 2.6ns 93.1 ± 2.6ns 93.9 ± 2.58ns 93.3 ± 2.6ns 94.5 ± 2.52ns 94.4 ± 2.5ns
Bone Cells (% of Viable Cells) 97.7 ± 2.3 97.9 ± 2.1ns 97.8 ± 3.4ns 98.0 ± 2.23ns 98.1 ± 3.21ns 98.2 ± 3.15ns 98.1 ± 3.2ns

Data are expressed as a percentage of the control and are presented as mean ± SD from 3 independent experiments. Differences between the control and treated cells are significant at
*P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test, ns: not significant.

Table 4
Effect of Zn2L2 on survival of EAC bearing female Swiss albino mice.

Parameters Untreated 20 mg/Kg 25 mg/Kg 30 mg/Kg 50 mg/Kg

Total volume (mL) 11.04 ± 1.1 9.01 ± 0.80ns 7.99 ± 0.81* 8.97 ± 0.74** 13.2 ± 0.12ns
PCV (mL) 6.5 ± 0.56 5.33 ± 0.46ns 4.73 ± 0.41* 5.30 ± 0.53** 7.81 ± 0.82*
MST (d) 28 38* 37* 42** 26ns
T/C (%) 100 136* 132* 150** 92.9ns

Data are expressed as a percentage of the control and are presented as mean ± SD from 3 independent experiments. Differences between the control and treated cells are significant at
*P < 0.05, **P < 0.01, ***P < 0.001, by unpaired Student’s t-test, ns: not significant.

complex of zinc may be less toxic and potent anticancer agent if a 5. Conclusions
suitable cysteine-based moiety can be attached to zinc atom [34–36].
Under this backdrop we selected the non-toxic ligand, 2-[(2-Hydroxy-3- This newly synthesized Zn-complex appears to be a promising
methoxy-benzylidene)-amino]-3-mercapto-propionic acid that does not chemotherapeutic agent that specifically targets malignant cells and
shows any significant cytotoxicity towards malignant cells. The Schiff- overcomes multidrug resistance in cancer without inducing any sig-
base ligand and its zinc complex have been synthesized as well as nificant undesired side effects in normal cells as well as in normal mice
characterized through details spectroscopic studies from which we model.
found that the assigned ligand to metal charge transfer (S → Zn LMCT)
band may be responsible for the yellow colour of the complex Conflicts of interest
(Supplementary Figure S1–S8). Their anti cancer properties have been
evaluated. Cytotoxicity assay (MTT assay) of H2L and Zn2L2 on both None.
CCRF-CEM and CEM/ADR 5000 cell lines shows that Zn2L2 but not H2L
has the anti-proliferative effect against these malignant cell lines. Zn2L2 Acknowledgements
proves to be non toxic (MTT assay) towards non-malignant PBMC.
Zn2L2 is found to be more effective at a comparatively lower con- This work was supported by grants from Council of scientific and
centration on CEM/ADR 5000 and therefore can be used as potent re- industrial research, India (Award No. 09/030/(0061)/2011 EMR-I) and
sistant modifying agent (RMA). Cell cycle analysis and annexin V Indian Council of Medical Research (ICMR), New Delhi, India [Award
binding assays revealed that Zn2L2 induces apoptosis in both the cell No. 74/10/2014-PERS. (EMS)] for financial support. The funder had no
lines. In CEM/ADR5000, the Zn2L2 induces apoptosis via G1/S block as role in the study design, data collection and analysis, decision to pub-
revealed by Cell cycle analysis. Zn2L2 significantly alters cellular ROS lish or preparation of the manuscript.
and GSH level in CCRF-CEM and CEM/ADR 5000 with respect to un-
treated control. Therefore we assume that in CCRF-CEM apoptosis is Appendix A. Supplementary data
triggered by generation of oxidative stress that elevates the cellular ROS
level and simultaneously depletes the cellular GSH level. In doxorubicin Supplementary data associated with this article can be found, in the
resistant CEM/ADR5000, Zn2L2 increases both cellular ROS and GSH. online version, at http://dx.doi.org/10.1016/j.etap.2017.11.004.
The elevated ROS level subsequently causes oxidative stress and en-
hances GSH level, which consequently sensitizes the cells towards drug References
and ultimately induces apoptosis (Chen et al., 2008). Finally the results
of in vivo sub acute systemic toxicity studies performed on female Swiss Bae, K.H., Kwon, Y.D., Shin, H.C., Hwang, M.S., Ryu, E.H., Park, K.S., Yang, H.Y., Lee,
albino mice suggests that Zn2L2 does not impart any significant toxic D.K., Lee, Y., Park, J., Kwon, H.S., Kim, H.W., Yeh, B.I., Lee, H.W., Sohn, S.H., Yoon,
J., Seol, W., Kim, J.S., 2003. Human zinc fingers as building blocks in the con-
symptoms up to a dose of 100 mg/kg of body weight. It is also worth struction of artificial transcription factors. Nat. Biotechnol. 21, 275–280.
noting that the Zn2L2 at a dose of 30 mg kg−1, increase the life span of Bazzicalupi, C., Bencini, A., Bianchi, A., Biver, T., Boggioni, A., Bonacchi, S., Danesi, A.,
Ehrlich ascites carcinoma (EAC) bearing mice up to T/C (%) value Giorgi, C., Gratteri, P., Ingrain, A.M., Secco, F., Sissi, C., Valtancoli, B., Venturini, M.,
2008. DNA binding by a new metallointercalator that contains a proflavine group
150% when compared to untreated control. All these doses showed bearing a hanging chelating unit. Chemistry 14, 184–196.
“marked activity” according to standard National Cancer Institute Belicchi, M.F., Bisceglie, F., Pelosi, G., Tarasconia, P., Albertini, R., Pinelli, S., 2001. New
protocols for screening new anticancer drugs. Moreover, Zn2L2 treat- methyl pyruvate thiosemicarbazones and their copperand zinc complexes: synthesis,
characterization, X-ray structure and biological activity. J. Inorg. Biochem. 287,
ment up to 50 mg/kg of body weight exhibited no significant toxicity on 137–147.
normal female Swiss albino mice. Furthermore, we are trying to deci- Beraldo, H., Gambinob, D., 2004. The wide pharmacological versatility of semi-
pher the underlying molecular mechanisms of Zn2L2-induced apoptosis. carbazones, thiosemicarba-zones and their metal complexes. Mini Rev. Med. Chem. 4,
31–39.
Additionally, detailed animal experimentation on the reported complex
Casini, A., Scozzafava, A., Supuran, C.T., 2002. Cysteine-modifying agents: a possible
will provide clinically relevant information in developing possible po- approach for effective anticancer and antiviral drugs. Environ. Health Perspect. 110,
tent anticancer agent. 801–806.
Chen, H.H., Song, I.S., Hossain, A., Choi, M.K., Yamane, Y., Liang, Z.D., Lu, J., Wu, L.Y.,
Siddik, Z.H., Klomp, L.W., Savaraj, N., Kuo, M.T., 2008. Elevated glutathione levels
confer cellular sensitization to cisplatin toxicity by up-regulation of copper trans-
porter hCtr1. Mol. Pharmacol. 74, 697–704.

391
A. Banerjee et al. Environmental Toxicology and Pharmacology 56 (2017) 383–392

Chohan, Z.H., Arif, M., Sarfraz, M., 2007. Metal-based antibacterial and antifungal amino Miyamoto, D., Endo, N., Oku, N., Arima, Y., Suzuki, T., Suzuki, Y., 1998. Beta-thujaplicin
acid derived Schiff bases: their synthesis: characterization and in vitro biological zinc chelate induces apoptosis in mouse high metastatic melanoma B16BL6 cells.
activity. Appl. Organomet. Chem. 21, 294–302. Biol. Pharm. Bull. 21, 1258–1262.
Clarke, M.J., Zhu, F., Frasca, D.R., 1999. Non-platinum chemotherapeutic metallo- Mossmann, T., 1983. Rapid colorimetric assay for cellular growth and survival: applica-
pharmaceuticals. Chem. Rev. 99, 2511–2534. tion to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63.
Dhawan, D.K., Chadha, V.D., 2010. Zinc: a promising agent in dietary chemoprevention Moucheron, C., 2009. From cisplatin to photoreactive Ru complexes: targeting DNA for
of cancer. Indian J. Med. Res. 132, 676–682. biomedical applications. New J. Chem. 33, 235–245.
Efferth, T., Konkimalla, V.B., Wang, Y., Sauerbrey, A., Meinhardt, S., Zintl, F., Mattern, J., Nakamoto, K., 2009. Infrared and Raman Spectra of Inorganic and Coordination
Volm, M., 2008. Prediction of broad spectrum resistance of tumors towards antic- Compounds, Part B; Applications in Coordination, Organometallic, and Bioinorganic
ancer drugs. Clin. Cancer Res. 14, 2405–2412. Chemistry. Wiley, New Jersey.
Emami, S., Hosseinimehr, S.J., Taghdisi, S.M., Akhlaghpoor, S., 2007. Kojic acid and its Nakayama, A., Hiromura, M., Adachi, Y., Sakurai, H., 2008. Molecular mechanism of
manganese and zinc complexes as potential radioprotective agents. Bioorg. Med. antidiabetic zinc-allixin complexes: regulations of glucose utilization and lipid me-
Chem. Lett. 17, 45–48. tabolism. J. Biol. Inorg. Chem. 13, 675–684.
Ganguly, A., Basu, S., Banerjee, K., Chakraborty, P., Sarkar, A., Chatterjee, M., Choudhuri, O’Halloran, T.V., 1993. Transition metals in control of gene expression. Science 261,
S.K., 2011. Redox active copper chelate overcomes multidrug resistance in T-lym- 715–725.
phoblastic leukemia cell by triggering apoptosis. Mol. BioSyst. 7, 1701–1712. Oteiza, P.I., Olin, K.L., Fraga, C.G., Keen, C.L., 1995. Zinc deficiency causes oxidative
Ganguly, A., Chakraborty, P., Banerjee, K., Choudhuri, S.K., 2014. The role of a Schiff damage to proteins, lipids and DNA in rat testes. J. Nutr. 125, 823–829.
base scaffold, N-(2-hydroxy acetophenone) glycinate-in overcoming multidrug re- Oteiza, P.I., Clegg, M.S., Zago, M.P., Keen, C.L., 2008. Zinc deficiency induces oxidative
sistance in cancer. Eur. J. Pharm. Sci. 51, 96–109. stress and AP-1 activation in 3T3 cells. Free Radic. Biol. Med. 28, 1091–1099.
Gillet, J.P., Efferth, T., Steinbach, D., Hamels, J., de Longueville, F., Bertholet, V., Ott, I., Gust, R., 2007. Non platinum metal complexes as anti-cancer drugs. Arch. Pharm.
Remacle, J., 2004. Microarray-based detection of multi-drug resistance in human (Weinheim) 340, 117–126.
tumor cells by expression profiling of ATP-binding cassette transporter genes. Cancer Pozarowski, P., Darzynkiewicz, Z., 2004. Analysis of cell cycle by flow cytometry.
Res. 64, 8987–8993. Methods Mol. Biol. 281, 301–311.
Golub, M.S., Gershwin, M.E., Hurley, L.S., Hendrickx, A.G., Saito, W.Y., 1985. Studies of Rodrıguez-Arguelles, M.C., Belicchi, M.E., Bisceglie, F., Pelizzi, C., Pelosi, G., Pinelli, S.,
marginal zinc deprivation in rhesus monkeys: infant behavior. Am. J. Clin. Nutr. 42, Sassi, M., 2004. Synthesis, characterization and biological activity of Ni, Cu and Zn
1229–1239. complexes of isatin hydrazones. J. Inorg. Biochem. 98, 313–321.
Ho, E., Courtemanche, C., Ames, B.N., 2003. Zinc deficiency induces oxidative DNA da- Sakurai, H., Yoshikawa, Y., Yasui, H., 2008. Current state for the development of me-
mage and increases p53 expression in human lung fibroblasts. J. Nutr. 133, tallopharmaceutics and anti-diabetic metal complexes. Chem. Soc. Rev. 37,
2543–2548. 2383–2392.
Huang, Q., Pan, Z., Wang, P., Chen, Z., Zhang, X., Xu, H., 2006. Zinc(II) andcopper(II) Sedlak, J., Lindsay, R.H., 1968. Estimation of total, protein-bound, and nonprotein sulf-
complexes of beta-substituted hydroxylporphyrins as tumor photosensitizers. Bioorg. hydryl groups in tissue with Ellman’s reagent. Anal. Biochem. 25, 192–205.
Med. Chem. Lett. 16, 3030–3033. Sharif, R., Thomas, P., Zalewski, P., Fenech, M., 2012. Zinc deficiency or excess within the
Jiang, Q., Zhu, J., Zhang, Y., Xiao, N., Guo, Z., 2009. DNA binding property, nuclease physiological range increases genome instability and cytotoxicity respectively, in
activity and cytotoxicity of Zn(II) complexes of terpyridine derivatives. Biometals 22, human oral keratinocyte cells. Genes Nutr. 7, 139–154.
297–305. Sheng, X., Guo, X., Lu, X.M., Lu, G.Y., Shao, Y., Liu, F., Xu, Q., 2008. DNA binding,
Katiyar, A., Singh, V.P., 2008. Synthesis, structural studies and bio-activity of Mn(II), Co cleavage, and cytotoxic activity of the preorganized dinuclear zinc(II) complex of
(II), Ni(II), Cu(II) and Zn(II) complexes withp-amino acetophenone salicyloyl hy- triazacyclononane derivatives. Bioconjug. Chem. 19, 490–498.
drazone. J. Coord. Chem. 61, 3200–3212. Tan, J., Wang, B., Zhu, L., 2009. DNA binding, cytotoxicity, apoptotic inducing activity,
Kimmig, A., Gekeler, V., Neumann, M., Frese, G., Handgretinger, R., Kardos, G., Diddens, and molecular modeling study of quercetin zinc (II) complex. Bioorg. Med. Chem. 17,
H., Niethammer, D., 1990. Susceptibility of multidrug-resistant human leukemia cell 614–620.
lines to human interleukin 2-activated killer cells. Cancer Res. 50, 6793–6799. Tarafder, M.T.H., Ali, M.A., Wee, D.J., Azahari, K., Silong, S., Crouse, K.A., 2000.
Kumar, K.S., Ganguly, S., Veerasamy, R., De Clercq, E., 2010. Synthesis, antiviral activity Complexes of a tridentate ONS Schiff base. Synth. Biol. Prop. Trans. Met. Chem. 25,
and cytotoxicity evaluation of Schiff bases of some 2-phenyl quinazoline-4(3)H-ones. 456–460.
Eur. J. Med. Chem. 45, 5474–5479. Trávníček, Z., Kryštof, V., Šipl, M., 2006. Zinc(II) complexes with potent cyclin-dependent
Lee, Y., Kim, H., Lee, J., Kim, K., 2011. Anticancer activity of S-allylmercapto-L-cysteine kinase inhibitors derived from 6-benzylaminopurine. J. Inorg. Biochem. 100,
on implanted tumor of human gastric cancer cell. Biol. Pharm. Bull. 34, 677–681. 214–225.
Li, J.-H., Wang, J.-T., Mao, J.-W., Ji, L.-N., 2008. Two novel zinc complexes of dis- Vallee, B.L., Auld, D.S., 1989. Short and long spacer sequences and other structural fea-
ubstituted 2, 2’-bipyridine with ammonium groups as artificial nucleases. Inorg. tures of zinc binding sites in zinc enzymes. FEBS Lett. 257, 138–140.
Chem. Commun. 11, 865–868. Vallee, B.L., Auld, D.S., 1990. Zinc coordination, function, and structure of zinc enzymes
Liu, C., Wang, L., 2009. DNA hydrolytic cleavage catalyzed by synthetic multinuclear and other proteins. Biochemistry 29, 5647–5659.
metallonucleases. Dalton Trans. 38, 227–239. Vallee, B.L., Falchuk, K.H., 1993. The biochemical basis of zinc physiology. Physiol. Rev.
Liu, S., Cao, W., Yu, L., Zheng, W., Li, L., Fan, C., Chen, T., 2013. Zinc(II) complexes 73, 79–118.
containing bis-benzimidazole derivatives as a new class of apoptosis inducers that Vermes, I., Haanen, C., Steffens-Nakken, H., Reutelingsperger, C., 1995. A novel assay for
trigger DNA damage-mediated p53 phosphorylation in cancer cells. Dalton Trans. 42, apoptosis. Flow cytometric detection of phosphatidylserine expression on early
5932–5940. apoptotic cells using fluorescein labelled Annexin V. J. Immunol. Methods 184,
Maheswari, P.U., Barends, S., Ozalp-Yaman, S., de Hoog, P., Casellas, H., Teat, S.J., 39–51.
Massera, C., Lutz, M., Spek, A.L., van Wezel, G.P., Gamez, P., Reedijk, J., 2007. Zuber, G., Quada, J.C., Hecht, S.M., 1998. Sequence selective cleavage of a DNA octa-
Unique ligand-based oxidative DNA cleavage by zinc(II) complexes of hpyramol and nucleotide by chlorinated bithiazoles and bleomycins. J. Am. Chem. Soc. 120,
hpyrimol. Chemistry 13, 5213–5222. 9368–9369.

392

Você também pode gostar