You are on page 1of 10

Materials Science & Engineering C 91 (2018) 372–381

Contents lists available at ScienceDirect

Materials Science & Engineering C


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

Green synthesized silver nanoparticles: Catalytic dye degradation, in vitro T


anticancer activity and in vivo toxicity in rats
Jayachandra Reddy Nakkalaa, Rani Mataa, Kumar Rajab, Varshney Khub Chandrab,

Sudha Rani Sadrasa,
a
Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
b
Department of Veterinary Pathology, Rajiv Gandhi College of Veterinary & Animal Sciences, Kurumbapet, Puducherry, India

A R T I C LE I N FO A B S T R A C T

Keywords: In this study, silver nanoparticles were synthesized (AgNPs) using aqueous rhizome extract of Acorus calamus
Nano silver (ACRE) and evaluated their in vitro anticancer activity and in vivo toxicity in a Wistar rat model. The synthesized
Catalytic activity AgNPs showed good catalytic activity against different organic pollutant dyes. In vitro cytotoxic effects of AgNPs
Anti-cancer activity were assessed in Hep2, COLO 205 and SH-SY5Y cells using MTT assay. Further, the apoptotic changes induced
Bio distribution
by AgNPs in more susceptible Hep2 cells were observed through AO/EB, DCFH-DA, Rhodamine 123, PI/DAPI
Toxicity
staining, oxidative stress markers and Western blotting. In vivo toxicity study revealed substantial alterations in
the levels of serum biochemical markers including AST, ALT, LDH and inflammatory markers such as TNF-α and
IL-6 on day 29 when rats treated with AgNPs as compared to control, however, these levels were restored to
normal at the end of washout period on day 89. No remarkable changes were observed in liver oxidative stress
enzymes. ICP-OES analysis indicated bio-distribution of silver in spleen (5.67 μg/g) and liver (4.98 μg/g) in rats
treated with 10 mg/kg b.w of AgNPs on day 29 and elimination of silver from all organs was observed at the end
of washout period on day 89. Histopathological analysis revealed no significant changes in kidney, spleen, lungs,
heart, testis and brain with 5 and 10 mg/kg b.w of AgNP. However, 10 mg/kg b.w of AgNPs showed moderate
degree of cell swelling and vacuolar degeneration in liver and these alterations were reverted back to normal at
the end of washout period. Findings from this study signify green synthesized AgNPs at low concentrations might
be useful in many ways with ecofriendly nature.

1. Introduction Alternately, the microbial and biological system (green chemistry) ap-
proaches are developed to synthesize NPs without the use of harsh and
In recent years, nanotechnology has fabulous growth due to their expensive toxic chemicals [10,11]. The microorganism based NPs
various applications in several fields such as material science, chem- synthesis is rapidly developed due to their eco-friendly formation of
istry, medicine, bionanotechnology, etc. Nanoparticles (NPs) have high NPs. However, microbial method has disadvantages due to the con-
attention because of their great surface to volume ratio, and tre- sumption of more time for maintaining cultures [12]. Nowadays, the
mendously small size, which leads to difference of physical and che- NPs are synthesized using different plant sources due to their natural
mical properties as compared to same composition of bulk material [1]. availability, rapid formation, and the ecofriendly nature [13]. The
Among the NPs, the researchers have shown more interest in silver NPs AgNPs were synthesized using Acorus calamus rhizome. Acorus calamus
(AgNPs) because of their wide range of applications in antimicrobial, is also called as sweet flag. In Indian Ayurvedic system this herb is used
catalysis, medical, optics and energy. These NPs have been commonly to treat brain disorders, chronic diarrhea, liver troubles, rejuvenator for
synthesized using electrochemical [2], photochemical [3], microwave nervous system, etc. [14]. The rhizome is also used in the preparation of
[4], thermal decomposition [5], chemical reduction [6], and radian modern herbal medicine as it has diuretic, laxative, sedative and car-
assisted process [7] for a long time. minative properties [15].
The AgNPs synthesized from various chemical approaches showed The present study aims to evaluate the catalytic activity of green
toxicity in in vivo studies. The behavior of AgNPs is influenced by the synthesized AgNPs on different organic pollutant dyes, in vitro antic-
size, concentration, coating, and distribution level of the particles [8,9]. ancer activity in cancer cells and in vivo toxicity in a Wistar rat model.


Corresponding author at: Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry 605 014, India.
E-mail addresses: dr.ssrlab@gmail.com, sadrassudha@gmail.com (S.R. Sadras).

https://doi.org/10.1016/j.msec.2018.05.048
Received 13 June 2017; Received in revised form 9 March 2018; Accepted 14 May 2018
0928-4931/ © 2018 Elsevier B.V. All rights reserved.
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

2. Materials and methods rhodamine B (RB), methylene blue (MB), methyl red (MR), methyl or-
ange (MO), cresol red (CRR), acridine orange (AO), eriochrome black T
2.1. Collection and preparation of extracts (EBT), and phenol red (PR). Briefly, the experiment was performed by
mixing 10 mM of 0.3 mL of 4-NP, 1.7 mL of deionized water, 1 mL of
The Acorus calamus rhizome powder was collected from the 150 mM NaBH4 and 15 μg/mL of ACAgNPs. The 10 mM of 30 μL picric
Puducherry local market, Puducherry, India. The powder was authen- acid (PA) or 2, 4, 6-TNT was mixed with 1.97 mL of deionized water,
ticated by Prof. Parthasarathy, Department of Ecology and 1 mL of 150 mM NaBH4 and 15 μg/mL of ACAgNPs. Similarly the
Environmental Science, Pondicherry University, India. The dried 10 mM of 100 μL 3-NP, CBB, CR, EY, RB, MB, MR, MO, CRR, AO, EBT,
powder was mixed with deionized water and the mixture was filtered and PR were individually mixed with 1.9 mL of deionized water, 1 mL
through Whatman No⋅ 1 filter paper. The water from the filtered so- of 150 mM NaBH4 and 15 μg/mL of ACAgNPs. The absorption of the
lution was evaporated using a rotary evaporator followed by lyophilizer mixture was monitored periodically with different time intervals be-
in order to obtain the residue. The phytochemical screening was per- tween the ranges of 200 to 800 nm using UV–visible spectro-
formed using the residue. photometer.

2.2. GC–MS analysis 2.6. Cell culture

The phyto-constituents of ACRE were tested using GC–MS (GC and The Hep2 (human epidermoid carcinoma), COLO 205 (human colon
MS JEOL GC mate supplied with the secondary electron multiplier). The adenocarcinoma) and SH-SY5Y (neuroblastoma) cancer cells were
interpretation of GC–MS spectra was performed by comparing the data cultured in Dulbecco's modified Eagle medium with L-glutamine and
of National Institute Standard and Technology (NIST) database. 1000 mg/L glucose (DMEM) which was supplemented with 10% fetal
bovine serum, streptomycin sulfate (0.1 mg/mL) and penicillin G
2.3. Synthesis of AgNPs (100 units/mL) in the humidified environment consisting of 5% CO2 at
37 °C.
The extract was prepared by mixing the 1 g of the dry powder of
ACRE in100 mL of double distilled water and then kept in boiling water 2.6.1. MTT [(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium
bath for 15 min. The Whatman No⋅ 1 filter paper was used for filtering bromide] assay
the extract which was then stored at 4 °C for further use. The NPs were The anti-proliferative ability of ACAgNPs on Hep2, COLO 205 and
synthesized by mixing different ratios of 1 mM silver nitrate and pre- SH-SY5Y cancer cells was assessed based on the previously reported by
pared extract at room temperature. The color change observed in the MTT assay [16]. The cell viability was determined by the following
mixture is an indicative of formation of NPs. The formation of AgNPs equation and then IC50 values were calculated.
was further confirmed through periodical observation of absorption of
Absorption (test) ∗100
the solution using UV-visible spectroscopy in the range of 300–700 nm. Percentage of viability =
Absorption (control)
The synthesized Acorus calamus silver NPs (ACAgNPs) were collected by
using repeated centrifugation at 18000 rpm for 25 min. The obtained
pellet was further repeatedly washed three times using double distilled 2.6.2. Cytomorphological analysis
water in order to remove the unbounded biomolecules which may be The cultured more susceptible Hep2 cells were seeded
the secondary metabolites or the proteins. The purified AgNPs pellet (1 × 105 cells/well) in a 12 well chamber plate and maintained for 12 h
was dried at room temperature and their characterization was per- in a humidified environment consisting of 5% CO2 at 37 °C. The seeded
formed by using different techniques. cells were then treated with ACAgNPs with their respective IC50 values
and then maintained for 24 h. The cytomorphology of the treated and
2.4. Characterization of AgNPs untreated cells was examined in an OPTIKA (Italia) inverted phase-
contrast microscope.
The formation of ACAgNPs was determined using UV-visible spec-
trophotometer (UV-1700 Shimadzu), with optical density measure- 2.6.3. Acridine orange/ethidium bromide (AO–EB) staining
ments executed at various time intervals between the wavelength The induction of apoptosis by ACAgNPs in Hep2 cells was examined
ranges of 300–700 nm. The baseline was corrected using double dis- using AO/EB double staining technique. The cultured Hep2 cells were
tilled water. The micrograph image and elemental composition of the seeded (1 × 105 cells/well) in a 12 well chamber plate and maintained
ACAgNPs were determined by TEM along with EDAX (JEOL 3010). The for 12 h in a humidified environment consisting of 5% CO2 at 37 °C.
sample was prepared by placing dry powdered AgNPs on the carbon Then the seeded cells were treated with the respective IC50 values of
coated copper grid, which have been dried under a mercury lamp for ACAgNPs and incubated at 37 °C for 24 h. Finally, 50 μL of dye mixture
5 min. The excess powder was removed by using tissue paper. The (AO-EB) was added to the cells and observed under fluorescence mi-
functional groups present in the ACRE and ACAgNPs were identified by croscope (Nikon Eclipse Ti Japan) to detect any apoptotic cell death.
FTIR spectroscopy (Thermo Nicolet Nexus 670 equipped with KBr op-
tics and a DTGS detector). The instrument was operated with a re- 2.6.4. Detection and quantification of intracellular ROS
solution of 4 cm−1 and scanned between the frequency ranges of In this assay, Hep2 cells (1 × 105 cells/well) were plated in a 6 well
500–4000 cm−1. The average hydrodynamic diameter and poly- plate and treated with respective IC50 value of ACAgNPs for 24 h.
dispersity nature of ACAgNPs were determined by the DLS particle size Following treatment, 10 μM of DCFH-DA was added and then kept for
analyzer (ZETA Seizers Nanoseries). The zeta potential of the ACAgNPs 30 min incubation at 37 °C. The treated and untreated cells were ob-
in pure water was further analyzed by using electrophoretic light served under the fluorescence microscopy (Nikon Eclipse Ti Japan) to
scattering at 25 °C in 150 V (Malvern Instruments Nano ZS). detect any ROS production. Similarly, ACAgNPs treated cells were
trypsinized and separately collected in aluminum foil wrapped
2.5. Catalytic activity Eppendorf tubes for ROS quantification. The DCFH-DA (25 μM) solution
was added to cells followed by incubation for 45 min at 37 °C. The in-
The catalytic role of ACAgNPs was assessed using 4-nitrophenol (4- tensity of fluorescence was noted by using the Fluorolog-FL3-11 spec-
NP), 3-nitrophenol (3-NP), 2, 4, 6-trinitrophenol (2, 4, 6-TNT) or picric trofluorometer (HORIBA JobinYvon, NJ, USA) with respective excita-
acid (PA), coomassie brilliant blue (CBB), congo red (CR), eosin Y (EY), tion and emission wavelengths.

373
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 1. (a) UV–visible spectral analysis of ACAgNPs at different time intervals (b) TEM micrograph of ACAgNPs (c) DLS particle size analysis of ACAgNPs (d) Zeta
potential analysis of ACAgNPs.

2.6.5. Oxidative stress parameters appropriate IC50 concentration of ACAgNPs. The treated cells were kept
The Hep2 cells were cultured in 75 cm2 flasks and treated with for 24 h at 37 °C with 5% CO2. After the treatment, cells were washed
ACAgNPs with IC50 concentration for 24 h. After the treatment, cells using PBS and fixed in 4% paraformaldehyde and then in 70% ethanol.
were washed with PBS and collected in ice-cold PBS at 4 °C. The col- At the end of the experiment, cells were stained using PI or DAPI and
lected cell pellets were then lysed using lysis buffer and centrifuged at observed under fluorescence microscope (Nikon Eclipse Ti Japan) at
10000g for 10 min at 4 °C. The supernatant was then assembled and 20× magnification.
maintained at 4 °C until all the oxidative biomarkers assays were per-
formed. Standard procedures were followed in order to estimate the 2.6.8. Western blot analysis
liver oxidative stress markers, such as LPO [17], SOD [18], catalase In this assay, Hep2 cells were exposed to the different concentration
[19] and GPx [20] and non-enzymatic antioxidant GSH [21]. of ACAgNPs and further incubated at 5% CO2 at 37 °C. After 24 h, the
cells were harvested and lysed with lysis buffer in the presence of
2.6.6. Assessment of mitochondrial membrane potential (MMP or ΔΨm) protease inhibitor cocktail. The collected lysates were then centrifuged
The MMP loss in ACAgNPs treated Hep2 cells were determined by at the rate of 8000 rpm for 10 min at 4 °C and the concentration of
Rhodamine 123 staining. Herein, the Hep2 cells (5 × 105 cells/well) proteins in supernatant was estimated by Lowry method. Equal con-
seeded separately in a six well plate were incubated for 12 h in a hu- centrations of samples were separated by using SDS-PAGE electro-
midified atmosphere having 5% CO2 at 37 °C. Then these cells were phoresis. The separated proteins were transferred onto nitrocellulose
exposed to the IC50 concentration of ACAgNPs for 24 h. Further, the membrane and then the membrane was blocked with 5% non-fat milk
cells were washed using PBS and fixed in 4% paraformaldehyde and solution for 1 h. After incubation, the primary antibody (active caspase
then in 70% of ethanol for 10 min. The 50 μL of rhodamine 123 (10 μg/ 8, 9, 3, pro-lamin, cleaved lamin, cleaved PARP, β-actin) was added and
mL) were added and then kept for 30 min. The excess dye was removed then kept for overnight at 4 °C followed by the addition of enzyme
using PBS and the cells were detected under a fluorescence microscope horseradish peroxidase (HRP) linked secondary antibody for 2 h at
(Nikon Eclipse Ti Japan) at 20× magnification. For quantification, the room temperature (RT). The protein bands were noticed by using
treated and untreated cells were trypsinized and separately collected in 3,3′,5,5′-Tetramethylbenzidine/hydrogen peroxide (TMB/H2O2) solu-
aluminum foiled effendrof tubes. Finally, the rhodamine 123 was added tion or Enhanced chemiluminescence (ECL) and were quantified by
to effendrof tube and kept for 45 min of incubation at 37 °C. The using Image J software.
fluorescence intensity was noted by using a HORIBA JobinYvon, (NJ,
USA) Fluorolog-FL3-11 spectrofluorometer with respective excitation/ 2.7. In vivo studies
emission wavelengths.
2.7.1. Experimental animals
2.6.7. Propidium iodide (PI) and 4′, 6-Diamidino-2-phenylindole The male Wistar rats with body weight 200–230 g (12–14 weeks)
dihydrochloride (DAPI) staining were purchased and maintained in the room temperature (22 ± 2 °C)
The apoptosis associated changes in the cell nucleus like con- with 12:12 h lights and dark cycle, according to the recommendations
densation/fragmentation were identified using PI staining. Herein, the of the Committee of purpose of Control and Supervision of Experiments
cultured Hep2 cells (1 × 105 cells/well) were plated in a six well plate on Animals (CPCSEA), Govt. of India. The rats were kept two weeks for
and kept for 12 h in the incubator. The cells were then treated with an acclimatization and the work was performed with the permission from

374
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 2. (a) Bright field microscopic images of (i) untreated Hep2 cells (ii) ACAgNPs treated Hep2 cells (b) Fluorescent microscopic images of AO/EB staining (i)
untreated Hep2 cells (ii) ACAgNPs treated Hep2 cells (c) Fluorescent microscopic images DCFH-DA staining (i) untreated Hep2 cells (ii) ACAgNPs treated Hep2 cells.

the Institutional Animal Ethical Committee, Pondicherry University 2.7.3. Body weight
(PU/SLS/IAEC/2014/23). Rats were weighed weekly once in order to check the change in
their body weights throughout the experimental period of 28 days.
2.7.2. Experimental procedure
The rats were divided into three groups, each comprising of 6 rats 2.7.4. Serum markers
(n = 6). The total experimental period was 88 days. At the end of experimental period rats were sacrificed and the blood
Group I rats were fed with normal pellet diet (NPD) and they were was collected. The serum was collected immediately from the blood.
considered as a control group; Group II rats were provided with 5 mg/ The serum levels of glucose, inorganic phosphorus (IP), aspartate
kg b.w of ACAgNPs for 28 days orally on alternate days; Group III rats transaminase (AST), alanine transaminase (ALT), lactate dehy-
were treated with 10 mg/kg b.w of ACAgNPs for 28 days orally on al- drogenase (LDH) and alkaline phosphatase (ALP) were determined by
ternate days. using diagnostic kits (AGGAPE-diagnostics). Serum cholesterol was
At the termination of the experimental period, from each group four estimated by CHOD-PAP (Cholesterol Oxidase/Peroxidase Amino
rats were anesthetized with ether and blood was collected in non-he- Phenazone) method (AGGAPE-diagnostics Pvt. Ltd., Kerala, India).
parinized test tubes by cardiac puncture. The samples were centrifuged Serum TG was estimated by Glycerol-3-Phosphate Oxidase – Phenol and
at 2000×g for 10 min in a refrigerated centrifuge and the serum was Amino Phenazone (GPO-PAP) method (AGGAPE-diagnostics Pvt. Ltd.,
collected in clean centrifuge tubes for biochemical analysis. The re- Kerala, India).
maining two rats left for 60 more days (wash out period) to check the
elimination of AgNPs. At 89th day the remaining two rats were an- 2.7.5. Serum inflammatory markers
esthetized and collected serum in a similar way to the above procedure The inflammatory markers of TNF-α and IL-6 were estimated by
for performing experiments. ELISA kits as stated to the supplier's instructions (TNF-α-EASIA and IL-

375
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 3. Oxidant and antioxidant status of Hep2 cells treated with IC50 concentration of ACAgNPs (a) Lipid peroxidase (LPO), (b) Superoxide dismutase (SOD), (c)
Catalase (expressed as μmole of H2O2 degraded/min/mg protein), (d) Glutathione peroxidase (GPx) and (e) Reduced glutathione (GSH). Data is expressed as
mean ± SD of three identical experiments performed in triplicates.

6–EASIA-CE, DIAsource Immuno Assays S.A. - Rue du Bosquet, 2 - B- neutral buffered formalin solution. The tissue samples were processed
1348 Louvain-la-Neuve - Belgium). by using paraffin embedding bath system and thin slices were prepared.
The sections were stained with hematoxylin and eosin (H & E) to
2.7.6. Oxidative stress markers estimation analyze any histopathological changes under a light microscope
The liver oxidative stress markers were assessed based on the pre- (Olympus BX40, Japan).
viously proposed standard procedures for LPO [17], SOD [18], catalase
[19], and GPx [20] and non-enzymatic antioxidant, GSH [21]. 2.7.9. Statistical analysis
Statistical analysis was done to the all related experiments among
2.7.7. Distribution of ACAgNPs in different tissues the groups as compared to the control using SPSS software version 20
The inductively coupled plasma optical emission spectrometry (ICP- ANOVA followed Tukey's HSD or by LSD. Student“t” test was used to
OES) technique was used to detect the bio-distribution of ACAgNPs in analyze the statistical significance between the groups. Significance
different freeze dried tissues including liver, kidney, heart, brain, lungs, difference was set at *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001.
spleen and testis on the 29th and 89th day. In this assay, 0.5 g tissue
was weighed and digested in 5 mL of ultra-pure nitric acid for 16 h at 3. Results and discussion
90 °C in a water bath followed by the addition of 1 mL hydrogen per-
oxide solution. Finally, the samples were diluted to a known volume The qualitative phytochemical analysis of ACRE showed the pre-
using Milli-Q water and analyzed by ICP-OES. The bio-distribution of sence of various vital secondary metabolites, such as alkaloids, phenols,
silver in different tissues was expressed as silver quantity/g tissue. flavonoids, terpenoids and cardiac glycosides. The GC–MS analysis
spectrum revealed that ACRE contains the major constituents of glycine
2.7.8. Histopathology (N-phenylacetyl) ethyl ester, penta decanoic acid 14-methyl- methyl
In this procedure, different tissues obtained from kidneys, spleen, ester, 8-octadecanoic acid- methyl ester, isomenthone, 13-hexylox-
liver, brain, heart, lungs, testes were instantaneously fixed in 10% of acyclotridec 10-ene-2-one, somenthone, and phenol 2 (4 (2-

376
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 4. Fluorescent microscopic images of rhodamine 123 staining (i) untreated Hep2 cells (ii) ACAgNPs treated Hep2 cells (b) Fluorescent microscopic images of PI
staining (i) untreated Hep2 cells (ii) ACAgNPs treated Hep2 cells (c) Fluorescent microscopic images of DAPI staining (i) untreated Hep2 cells (ii) ACAgNPs treated
Hep2 cells.

hydroxyethylamino) 2-quinazolinyl (Supplementary data S1). Our results can be corroborated with the earlier report on the formation
of sphere-shaped AgNPs from the Platanus orientalis leaf extract [23].
The average size of the ACAgNPs was found to be 31.83 nm in DLS
3.1. Characterization of AgNPs
particle size analyzer (Fig. 1c). The particle size in DLS appeared bigger
than TEM, because DLS are measured based on the hydrodynamic
The formation of ACAgNPs was observed at room temperature by a
diameter of the particles and provides an intensity weighted average
change in color of the solution from colorless to yellowish brown by
particle size, whereas size obtained from TEM are constructed on the
adding ACRE to 1 mM silver nitrate solution. The color formation might
diameter of dry particle and gives an average particle size [24]. The
be due to the SPR effect and change of Ag+ ions to Ago by the aqueous
zeta potential value of ACAgNPs was −32.3 mV, which indicated that
extract [22]. The aqueous extract contains various secondary metabo-
the synthesized NPs were more stable in nature (Fig.1d). The zeta po-
lites such as alkaloids, phenols, flavonoids, terpenoids and cardiac
tential values of above ± 30 mV can be considered as stable NPs [25].
glycosides, which might be involved in the reduction and capping of
The functional groups present in ACRE which might have involved in
AgNPs. For the optimization process of NPs formation, silver nitrate
the formation of ACAgNPs were determined using FTIR analysis (Sup-
solution was mixed with aqueous plant extract in different ratios, where
plementary data S2). The spectral bands observed in ACRE at 3378,
in optimum formation of NPs was observed in the 5:1 ratio with the λ
2919, 1660, 1400, 1202 cm−1 indicates phenol OeH, alkyl CeH, car-
max at 421 nm. The absorption intensity increased with increasing re-
bonyl C]O, aromatic CeN and CeO or CeOeC respectively. Vibra-
action time. This may be due to the synthesis or increased agglomera-
tional band shifts of 3324, 3234, 1624 and 1373 observed in ACAgNPs
tion of AgNPs. Broadening of peaks was also observed, which indicated
represents aromatic phenol, amine and carbonyl groups of ACRE that
that the synthesized NPs were in poly dispersed nature (Fig. 1a). The
might have involved in the reduction, capping and stabilization of
TEM image revealed spherical shape of ACAgNPs as shown in Fig. 1b.

377
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 5. (a) Western blot analysis shows the expression of β-actin, cleaved caspase 8, caspase 9, caspase 3, Lamin and PARP in Hep2 cells treated with various
concentrations of ACAgNPs for 24 h (b) Densitometric analysis of ACAgNPs treated Hep2 cells. Protein levels were quantified using densitometry analysis and
expressed in relative band intensity. Values represent mean ± SD (n = 4). * indicates value significantly different from the control at P ≤ 0.05.

Table 1
Serum biochemical parameters on 29th (before washout) and 89th day (after washout) rats treated with 5 and 10 mg/kg b.w of ACAgNPs as compared to control rats.
Data is expressed as mean ± SD from four rats in each group on the 29th day and two rats in each group on the 89th day (washout period). (AST- Aspartate
aminotransferase, ALT- Alanine transaminase, LDH-Lactate dehydrogenase, ALP- Alkaline phosphatase (ALP), TG- Triglycerides). * specifies a value significantly
different from the control at P ≤ 0.05.
Parameters Control 5 mg/kg 10 mg/kg Control Washout Washout
5 mg/kg 10 g/kg

Glucose 105.8 ± 6.2 102.8 ± 5.8 93.8 ± 4.0 93.7 ± 4.0 88.2 ± 4.8 87.8 ± 3.5
AST 119.5 ± 3.1 147.7 ± 2.9* 164.3 ± 3.5* 122.9 ± 2.6 120.9 ± 2.6 123.9 ± 3.2
ALT 62.5 ± 4.5 82.7 ± 2.4* 89.1 ± 1.7* 64.5 ± 3.0 68.3 ± 2.9 67.3 ± 0.9
LDH 502 ± 18.5 799.3 ± 12.8* 780 ± 23.9* 459.5 ± 18.5 458.8 ± 9.2 467.2 ± 8.8
ALP 281.4 ± 3.8 280.9 ± 13.8 275.5 ± 17 257.0 ± 7.0 243.4 ± 9.4 246.7 ± 13.1
TG 68.8 ± 4.4 59.1 ± 4.4 72.2 ± 2.2 76.1 ± 3.1 66.2 ± 3.9 89.7 ± 1.4
Cholesterol 71.3 ± 3.3 67.8 ± 2.7 60.6 ± 4.8 83.2 ± 4.6 83.0 ± 4.3 92.1 ± 3.6

Table 2
Serum oxidative stress markers, Lipid peroxidase (LPO), Superoxide dismutase (SOD), Catalase (CAT-expressed as μmole of H2O2 degraded/min/mg protein),
Glutathione peroxidase (GPx), Reduced glutathione (GSH) and inflammatory markers like TNF-α – Tumor necrosis factor- α, IL-6 Interleukin-6 of rats treated with 5
and 10 mg/Kg b.w of ACAgNPs as compared to control on day 29th and 89th (Washout period). Data is expressed as mean ± SD from four rats in each group on 29th
day and two rats in each group on 89th day (washout period). *designates value significantly different from the control at P ≤ 0.05.
Parameters Control 5 mg/kg b.w 10 mg/kg b.w Control Washout Washout
5 mg/kg b.w 10 mg/kg

LPO 17.6 ± 0.57 17.0 ± 1.0 16.7 ± 0.7 17.6 ± 0.5 17.2 ± 1.0 17.8 ± 0.7
SOD 29.3 ± 3.9 25.9 ± 2.3 27.4 ± 1.6 29.3 ± 3.9 27.0 ± 3.9 29.1 ± 2.2.
CAT 212.2 ± 22.7 208.1 ± 14.1 197.1 ± 7.4 122 ± 22.7 224.9 ± 11.5 205.7 ± 9.9
GPx 27.6 ± 2.5 25.6 ± 2.0 26.3 ± 2.6 27.6 ± 2.5 25.6 ± 2.5 24.6 ± 2.5
GSH 44.9 ± 1.4 53.9 ± 3.0 52.3 ± 1.7 44.9 ± 1.4 49.8 ± 0.7 44.5 ± 1.7
TNF-α 6.5 ± 0.75 68 ± 0.3* 74.5 ± 1.08* 6.5 ± 0.75 9.5 ± 0.7 9.0 ± 0.7
(pg/ml)
IL-6 77 ± 1.4 213 ± 7.0* 330.5 ± 3.5* 64.5 ± 1.0 70 ± 0.7 71 ± 1.0
(pg/ml)

ACAgNPs. The GC–MS analysis showed the presence of bioorganic redox potential among the donor and the acceptor could deter the
compounds in ACRE such as glycine (N-phenylacetyl) ethyl ester, 13- passage of electrons between them [26,27]. Silver, platinum and gold
hexyloxacyclotridec 10-ene-2-one, somenthone, and phenol 2 (4 (2- NPs acts as an effective catalyst based on the intermediate redox po-
hydroxyethylamino) 2-quinazolinyl, which supports the functional tential value between the donor and the acceptor may aid in the
groups revealed in FTIR spectrum that are involved in the NPs forma- transfer of electrons, which could cause an electron relay system [28].
tion and capping. In aqueous solution the different organic dyes such as 4-NP, 3-NP, PA or
2,4,6-TNP, CBB, CR, EY, RB, MB, MR, MO, CRR, AO, EBT, and PR
3.2. Catalytic activity showed absorption maximum respectively at 318 nm, 331 nm, 357 nm,
588 nm, 495 nm, 530 nm, 554 nm, 664 nm, 522 nm, 464 nm, 440 nm,
The catalytic dye degradation relays on the transfer of electrons 490 am 545 nm and 426 nm. The dyes absorption maxima was not al-
between the donor and the acceptor. However, large differences in tered after addition NaBH4, might be due to the presence of high kinetic

378
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

Fig. 6. H&E stained histopathological images of rats (a) liver (b) kidney (c) spleen (d) brain (e) heart (f) lungs (H&E × 200), (a) (i) Control (ii) ACAgNPs (5 mg/kg
b.w) (iii) ACAgNPs (10 mg/kg b.w) (iv) Washout ACAgNPs (5 mg/kg b.w) (v) Washout ACAgNPs (10 mg/kg b.w).

barrier between the reciprocally deterring negative ions BH4− and respectively for Hep2, COLO 205 and SH-SY5Y cells (Supplementary
organic dyes. The 4-NP, 3-NP, PA or 2,4,6-TNP, CBB, CR, EY, RB, MB, data S4). Based on these results more susceptible Hep2 cells were se-
MR, MO, CRR, AO, EBT, and PR organic dyes absorption intensity was lected due to their low IC50 values for further studies to assess the mode
decreased respectively within 8, 9, 16, 5, 10, 8, 3, 12, 15, 3, 6, 60, 16, of action of ACAgNPs. Cyto-morphological analysis of Hep2 cells
and 18 min by the addition of ACAgNPs. The time dependent de- showed distinctive features of cell death, including rounding up of
gradation/reduction of organic pollutant dyes spectral peaks in the nuclei and shrinkage of cells following treatment with IC50 concentra-
presence and absence of ACAgNPs was showed in supplementary data tions of ACAgNPs (Fig. 2a). Hep2 cells treated with ACAgNPs exhibited
S3. orange colored apoptotic bodies an indication of late apoptosis
(Fig. 2b). Similar observations were reported in a previous study where
3.3. Anticancer activity Chitosan nanocarrier AgNPs induced late apoptosis in human colon
cancer cells as (HT-29 cells) [29]. A majority of anticancer drugs trig-
Results obtained from the MTT assay indicated decreased cell via- gers apoptotic cell death in cancer cells by altering the levels of oxi-
bility in all the cancer cell lines treated with ACAgNPs. The IC50 values dants/anti-oxidant status. Hence, ROS levels in Hep2 cells treated with
of ACAgNPs were found to be 29.5 μg/mL, 99.4 μg/mL and 149.4 μg/ ACAgNPs were detected by DCFH-DA staining. The increased intensity
mL for 24 h, while 16.4 μg/mL, 73 μg/mL and 97.2 μg/mL for 48 h of green fluorescence was observed in ACAgNPs treated Hep2 cells as

379
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

compared to untreated cells, which suggest increased ROS levels in caspases- 8, 9, 3, lamin and PARP respectively 3.8, 1.9, 4.1, 1.1, and 2.5
treated cells (Fig. 2c). after 24 h in Hep2 cells treated with ACAgNPs (45 μg/mL) as compared
The spectrofluorometric analysis carried out to quantify ROS levels to control cells (Fig. 5b). These findings reveal that both intrinsic and
in ACAgNPs treated Hep2 cells; there was a 66% increase in the in- extrinsic apoptotic pathways are involved in cell death of Hep2 cells
tensity of green fluorescence (Supplementary data S5). In order to as- when treated with ACAgNPs.
sess the oxidative stress mediated damage, the levels of oxidant markers
including LPO and anti-oxidant enzymes- SOD, GSH, GPx, and catalase 3.4. In-vivo toxicity
were measured in Hep2 cells treated with ACAgNPs. Results indicated
1.2 fold increase of MDA level in Hep2 cells after treatment with The small quantity of silver is commonly nonhazardous [40].
ACAgNPs as compared to control cells. Decreased activity of natural However, the toxicity of AgNPs is most controversial till date due to its
defense enzymes like SOD, GPx, catalase and depletion of non-enzy- possible risk when used in biological system. It is necessary to evaluate
matic GSH levels were also observed, respectively 2.7, 2.5, 1.1 and 0.53 the in vivo toxicity of green synthesized AgNPs in animal models for
folds in ACAgNPs treated cells as compared to control (Fig. 3). Similar further successful use in the future applications in the field of biology
observations have been reported earlier in zinc oxide NPs treated and medicine.
HepG2 cells where induction of apoptosis was mediated through the
alteration of the oxidant/anti-oxidant balance [30]. 3.4.1. Body weight
Interruption in mitochondrial membrane potential (MMP) is one of Body weight of the rats treated with ACAgNPs was recorded at
the most basic intracellular actions after the stimulation of apoptosis various time intervals during the experimental period. It was observed
[31]. It is well known that mitochondria is the main site for production that the body weight of treated rats was similar to that of control group
of ROS inside the cell and any condition causing depletion of anti- (Supplementary data S7). Similar findings have been reported with
oxidants and excess formation of ROS trigger mitochondrial damage. PVP-coated AgNPs and silver ions [41] which showed no significant
The increase in ROS levels has been shown to cause loss of MMP, which changes in body weight.
is a main characteristic of apoptosis. In this study, MMP was de-
termined using Rhodamine 123 staining. Rhodamine 123 is a lipophilic 3.4.2. Biochemical parameters
cationic dye which can easily pass into the intact mitochondria and The serum biochemical parameters such as glucose, inorganic
markedly accumulate in the inner mitochondrial membrane and exhibit phosphorous, SGOT, SGPT, ALP, LDH, cholesterol, triglycerides and
high green fluorescence. ACAgNPs treated Hep2 cells exhibited less anti-oxidant enzymes like LPO, SOD, CAT, GPx, and GSH were analyzed
green fluorescence intensity indicating loss of MMP while the untreated at the end of the experimental (29th day) and washout period (89th
cells exhibited high green fluorescence implying healthy mitochondria day). A significant increase in the serum AST, ALT, and LDH levels were
(Fig. 4a). The spectrofluorometric analysis indicated that the ACAgNPs observed in ACAgNPs treated rats on day 29th as compared to control
treated Hep2 cells showed 28% decrease in the fluorescent intensity as group rats but no significant difference was observed in between 5 and
compared to untreated cells (Supplementary data S6). In acute myeloid 10 mg/kg b.w treated group rats. However, increased AST, ALT, and
leukemia cells, treatment with PVP-coated AgNPs induced generation LDH levels on day 29th were reverted to normal levels after washout
of ROS and subsequent loss of MMP [32]. To further investigate the period (89th day) (Table 1). In a previous study, liver markers including
apoptosis-associated nuclear changes in Hep2 cells treated with AST, ALT, and ALP levels were found to be negatively altered in rats
ACAgNPs, PI and DAPI staining was performed to assess changes such treated with 10 mg/kg b.w of Ag/Au nanocomposites [42].
as nuclear condensation and fragmentation. The ACAgNPs treated Hep2 The ACAgNPs treated rats did not show any significant alteration in
cells showed condensation/fragmentation of nucleus by staining with other biochemical parameters such as glucose, inorganic phosphorous,
PI or DAPI (Fig. 4b–c), indicating apoptotic changes in treated cells and cholesterol, triglycerides, and anti-oxidant enzymes like LPO, SOD,
normal smooth nuclei in untreated control cells. CAT, GPx, and GSH as compared to control rats (Table 2). Previous
The induction of apoptosis generally occurs through two major study showed that the GPx, and SOD levels were decreased, while total
signaling pathways namely extrinsic and intrinsic that is regulated anti-oxidant capacity (TAC) levels were positively altered in Wistar rats
through caspase 8 and caspase 9 respectively [33]. The extrinsic administrated (intra peritoneal) with 5 mg/kg b.w of AgNPs [43].
pathway mediated apoptosis is initiated by binding of membrane re-
ceptor to an extracellular ligand such as FAS, which progress to bind 3.4.3. Inflammatory markers
adapter proteins connected death domain (FADD) in the intracellular The levels of inflammatory cytokines, TNF–α and IL-6 in ACAgNPs
receptor part and this death inducing signaling complex (DISC) recruit treated rats were found increased as compared to control rats on day
procaspase 8 and initiate activation of caspase 8 [34]. Once caspase 8 is 29. There were no significant differences observed in the levels of
activated, it activates the signaling cascade and consequent cell death TNF–α between 5 and 10 mg/kg b.w of ACAgNPs treated rats. On the
by triggering downstream execution caspases, such as caspase 3 [35]. other hand the IL-6 levels were significantly differed between 5 and
On the other hand, intrinsic pathway is initiated by the accumulation of 10 mg/kg b.w of ACAgNPs treated rats. However, these levels were
ROS, which in turn lead to failure in continuing the MMP and release of restored to normal after washout period on day 89 (Table 2). Previous
cytochrome C [36]. The released cytosolic cytochrome C combines with report also indicated alterations in the levels of inflammatory markers
pro-caspase 9, dATP and apoptotic protease activating factor 1 (Apaf-1) when treated with NPs [44,45].
to form apoptosome complex [37]. The apoptosome complex activates
caspase 3 through the downstream signaling cascade and the activated 3.4.4. ICP-OES analysis
caspase 3 cleaves over hundred substrates, including lamins, PARP, and ICP-OES analysis was used to detect the distribution of AgNPs in
plentiful DNA associated proteins that identify and triggers signals for different vital organs such as liver, kidney, spleen, lungs, heart, testis
DNA strand break [38]. The active caspase 3 cleaves PARP, and the and brain at the end of the experimental period on day 29 and at the
cleaved product of PARP is a hallmark of apoptosis [39]. To reveal the end of the washout period on day 89. ACAgNPs were not detected in the
molecular mechanism involved in induction of apoptosis by ACAgNPs liver, kidney, spleen, lungs, heart, testis and brain in rats treated at
in treated Hep2 cells expression of caspase 8, caspase 9, caspase 3, 5 mg/kg b.w concentration. On the other hand, at 10 mg/kg b.w con-
lamin and PARP were determined. It was observed that A549 and Hep2 centration, ACAgNPs were found in liver and spleen with concentration
cells showed increased expression of active caspases- 8, 9, and 3, lamin of 5.67 μg/g in spleen and 4.98 μg/g in liver (Supplementary data S8).
and PARP with increasing concentration of ACAgNPs (Fig. 5a). Densi- However, it was found that silver was completely eliminated from the
tometry analysis exhibited fold increase in the expression of active liver and spleen at the end of washout period. The major silver

380
J.R. Nakkala et al. Materials Science & Engineering C 91 (2018) 372–381

deposition in the liver may be due to its detoxification function in the triangular gold nanoprisms, Nat. Mater. 3 (2004) 482–488.
biological systems. Previous studies also report that the accumulation of [13] R. Konwarh, B. Gogoi, R. Philip, M.A. Laskar, N. Karak, Biomimetic preparation of
polymer-supported free radical scavenging, cytocompatible and antimicrobial “green”
silver may due to the formation of thiol‑silver complexes in liver be- silver nanoparticles using aqueous extract of Citrus sinensis peel, Colloids Surf. B:
cause the sulfur containing groups commonly present in liver, which Biointerfaces 84 (2011) 338–345.
[14] R. Singh, P.K. Sharma, R. Malviya, Pharmacological properties and ayurvedic value of
have high affinity to silver [46,47]. Indian buch plant (Acorus calamus): a short review, Adv, Biol. Res. 5 (2011) 145–154.
[15] Gualtiero Simonetti, Stanley Schuler (Ed.), Simon & Schuster's Guide to Herbs and Spices,
3.4.5. Histopathology Simon & Schuster, Inc, 1990(ISBN 0-671-73489-X).
[16] J.R. Nakkala, M. Rani, S.S. Rani, Biological activities of green silver nanoparticles syn-
The rats treated with 5 mg/kg b.w of ACAgNPs exhibited normal thesized with Acorous calamus rhizome extract, Eur. J. Med. Chem. 85 (2014) 784–794.
hepatic parenchyma whereas 10 mg/kg b.w treated rats exhibited his- [17] H. Ohkawa, N. Ohishi, K. Yagi, Assay for lipid peroxides in animal tissues by thiobarbi-
turic acid reaction, Anal. Biochem. 95 (1979) 351–358.
topathological alterations like moderate degree of cell swelling and
[18] S. Marklund, G. Marklund, Involvement of the superoxide anion radical in the auto-
vacuolar degeneration. However, these histopathological alterations xidation of pyrogallol and a convenient assay for superoxide dismutase, Eur. J. Biochem.
were not observed in rats after washout period. The other vital organs 47 (1974) 69–74.
[19] A.K. Sinha, Colorimetric assay of catalase, Anal. Biochem. 47 (1972) 389–394.
such as kidney, spleen, lungs, heart, testis and brain did not show any [20] J.T. Rotruck, A.L. Pope, H.E. Ganther, A.B. Swanson, D.G. Hateman, W.G. Hoekstra,
histopathological changes by treatment with 5 and 10 mg/kg b.w of Selenium: biochemical role as a component of glutathione peroxidase, Science 179 (1973)
ACAgNPs (Fig. 6). 588–590.
[21] G. Ellman, Tissue sulfhydryl groups, Arch. Biochem. Biophys. 82 (1959) 70–77.
[22] P. Mulvaney, Surface plasmon spectroscopy of nanosized metal particles, Langmuir 12
4. Conclusion (1996) 788–800.
[23] A.Y. Obaid, S.A. Al-Thabaiti, E.H. El-Mossalamy, L.M. Al-Harbi, Z. Khan, Extracellular
bio-synthesis of silver nanoparticles, Arab. J. Chem. 10 (2017) 226–231.
The green based silver NPs showed good in vitro catalytic and anti- [24] M. Ahamed, R. Posgai, T.J. Gorey, M. Nielsen, S.M. Hussain, J.J. Rowe, Silver nano-
cancer activities. The in vivo data suggest that the NPs alter some of the particles induced heat shock protein 70, oxidative stress and apoptosis in Drosophila
melanogaster, Toxicol. Appl. Pharmacol. 242 (2010) 263–269.
serum biochemical markers in treated rats on day 29 but the levels were [25] C. Jacobs, R.H. Müller, Production and characterization of a budesonide nanosuspension
restored to normal after washout period on day 89. Complete elim- for pulmonary administration, Pharm. Res. 19 (2002) 189–194.
[26] G. Wilkinson, R.D. Gillard, J.A. Mccleverty, Comprehensive Co-Ordination Chemistry,
ination of silver from rats was observed on day 89. This study provides
Pergamon Press, Oxford, 1987.
the preliminary guidance towards the development of multifaceted [27] R.G. Compton, Comprehensive Chemical Kinetics, Elsevier, New York, 1989.
applications of green synthesized AgNPs. [28] K. Mallick, M. Witcomb, M. Scurrell, Silver nanoparticle catalysed redox reaction: an
electron relay effect, Mater. Chem. Phys. 97 (2006) 283–287.
[29] P. Sanpui, A. Chattopadhyay, S.S. Ghosh, Induction of apoptosis in cancer cells at low
Acknowledgements silver nanoparticle concentrations using chitosan nanocarrier, ACS Appl. Mater.
Interfaces 3 (2011) 218–228.
[30] M.J. Akhtar, M. Ahamed, S. Kumar, M.M. Khan, J. Ahmad, S.A. Alrokayan, Zinc oxide
The authors acknowledge the DST, Government of India, New Delhi, nanoparticles selectively induce apoptosis in human cancer cells through reactive oxygen
for financial support in the form of DST-FIST. The authors thank DBT- species, Int. J. Nanomedicine 7 (2012) 845–857.
[31] F. Qi, A. Li, L. Zhao, H. Xu, H. Inagaki, D. Wang, X. Cui, B. Gao, N. Kokudo, M. Nakata,
IPLS (BUILDER) program for Fluorescence microscope facility and the
W. Tang, Cinobufacini, an aqueous extract from Bufo bufo gargarizans Cantor, induces
Central Instrumentation Facility (CIF), Pondicherry University. The apoptosis through a mitochondria-mediated pathway in human hepatocellular carcinoma
authors also express sincere thanks to IIT Madras and ILS, Ahmadabad cells, J. Ethnopharmacol. 128 (2010) 654–661.
[32] D. Guo, L. Zhu, Z. Huang, H. Zhou, Y. Ge, W. Ma, J. Wu, X. Zhang, X. Zhou, Y. Zhang,
University for instrument facility. The first author (JR Nakkala) conveys Y. Zhao, N. Gu, Anti-leukemia activity of PVP-coated silver nanoparticles via generation
special thanks to CSIR, India for the financial assistance in the form of reactive oxygen species and release of silver ions, Biomaterials 34 (2013) 7884–7894.
junior research fellowship (JRF) and senior research fellowship (SRF). [33] K.K. Hui, A.K. Kanungo, A.J. Elia, J.T. Henderson, Caspase-3 deficiency reveals a phy-
siologic role for Smac/DIABLO in regulating programmed cell death, Cell Death Differ. 18
(2011) 1780–1790.
Appendix A. Supplementary data [34] Y. Dai, T.S. Lawrence, L. Xu, Overcoming cancer therapy resistance by targeting inhibitors
of apoptosis proteins and nuclear factorkappa B, Am. J. Transl. Res. 1 (2009) 1–15.
[35] X. Jiang, X. Wang, Cytochrome c-mediated apoptosis, Annu. Rev. Biochem. 73 (2004)
Supplementary data to this article can be found online at https:// 87–106.
doi.org/10.1016/j.msec.2018.05.048. [36] X. Wang, The expanding role of mitochondria in apoptosis, Genes Dev. 15 (2003)
2922–2933.
[37] A. Thorburn, Death receptor-induced cell killing, Cell. Signal. 16 (2004) 139–144.
References [38] A. Nawab, M. Yunus, A.A. Mahdi, S. Gupta, Evaluation of anti-cancer properties of
medicinal plants from the Indian sub-continent, Mol. Cell Pharmacol. 3 (2011) 21–29.
[39] H.L. Yang, S.C. Chen, C.S. Chen, S.Y. Wang, Y.C. Hseu, Alpinia pricei rhizome extracts
[1] M. Brust, D.J. Schiffrin, D. Bethell, C.J. Kiely, Adv. Mater. 7 (1995) 795. induce apoptosis of human carcinoma KB cells via a mitochondria-dependent apoptotic
[2] L. Rodriguez-Sanchez, M.C. Blanco, M.A. Lopez-Quintela, Electrochemical synthesis of pathway, Food Chem. Toxicol. 46 (2008) 3318–3324.
silver nanoparticles, J. Phys. Chem. B 104 (2000) 9683–9688. [40] M.A.M. Khan, S. Kumar, M. Ahamed, S.A. Alrokayan, M.S. AlSalhi, Structural and thermal
[3] K. Mallick, M.J. Witcomb, M.S. Scurrell, Polymer stabilized silver nanoparticles: a pho- studies of silver nanoparticles and electrical transport study of their thin films, Nanoscale
tochemical synthesis route, J. Mater. Sci. 39 (2004) 4459–4463. Res. Lett. 6 (2011) 434.
[4] H. Yin, T. Yamamoto, Y. Wada, S. Yanagida, Large-scale and size-controlled synthesis of [41] M. Van der zande, R.J. Vandebriel, E. Van Doren, E. Kramer, Z. Herrera Rivera,
silver nanoparticles under microwave irradiation, Mater. Chem. Phys. 83 (2004) 66–70. C.S. Serrano-Rojero, E.R. Gremmer, J. Mast, R.J. Peters, P.C. Hollman, P.J. Hendriksen,
[5] S. Navaladian, B. Viswanathan, R.P. Viswanath, T.K. Varadarajan, Thermal decomposi- H.J. Marvin, A.A. Peijnenburg, H. Bouwmeester, Distribution, elimination, and toxicity of
tion as route for silver nanoparticles, Nanoscale Res. Lett. 2 (2007) 44–48. silver nanoparticles and silver ions in rats after 28-day oral exposure, ACS Nano 6 (2012)
[6] K. Shameli, M.B. Ahmad, W.M.Z.W. Yunus, N.A. Ibrahim, M. Darroudi, Synthesis and 7427–7442.
characterization of silver/talc nanocomposites using the wet chemical reduction method, [42] F.A. Sulaiman, M.A. Akanji, H.O.B. Oloyede, A.A. Sulaiman, A. Olatunde, E.B. Joel,
Int. J. Nanomedicine 5 (2010) 743–751. T.H. Adewale, H.A. Adeboye, S.O. Idris, A.L. Quadri, R.A. Oyegoke, O.S. Adeyemi, Oral
[7] K. Shameli, M. Ahmad, W.M.Z.W. Yunus, A. Rustaiyan, N.A. Ibrahim, M. Zargar, exposure to silver/gold nanoparticles: status of rat lipid profile, serum metabolites and
Y. Abdollahi, Green synthesis of silver/montmorillonite/chitosan bionanocomposites tissue morphology, J. Med. Sci. 15 (2015) 71–79.
using the UV irradiation method and evaluation of antibacterial activity, Int. J. [43] A. Ranjbar, Z. Ataie, F. Khajavi, H. Ghasemi, Effects of silver nanoparticle (Ag NP) on
Nanomedicine 5 (2010) 875–887. oxidative stress biomarkers in rat, Nanomed. J. 1 (2014) 205–210.
[8] J. Liu, D.A. Sonshine, S. Shervani, R.H. Hurt, Controlled release of biologically active [44] J.D. Byrne, J.A. Baugh, The significance of nanoparticles in particle-induced pulmonary
silver from nanosilver surfaces, ACS Nano 4 (2010) 6903–6913. fibrosis, McGill, J. Med. 11 (2008) 43–50.
[9] S. Kittler, C. Greulich, J. Diendorf, M. Koller, M. Epple, Toxicity of silver nanoparticles [45] C. Monteiller, L. Tran, W. MacNee, S. Faux, A. Jones, B. Miller, K. Donaldson, The pro-
increases during storage because of slow dissolution under release of silver ions, Chem. inflammatory effects of low-toxicity low-solubility particles, nanoparticles and fine par-
Mater. 22 (2010) 4548–4554. ticles, on epithelial cells in vitro: the role of surface area, Occup. Environ. Med. 64 (2007)
[10] B. Ankamwar, C. Damle, A. Ahmad, M. Sastry, Biosynthesis of gold and silver nano- 609–615.
particles using Emblica officinalis fruit extract, their phase transfer and transmetallation [46] J. Liu, D.A. Sonshine, S. Shervani, R.H. Hurt, Controlled release of biologically active
in an organic solution, J. Nanosci. Nanotechnol. 5 (2005) 1665–1671. silver from nanosilver surfaces, ACS Nano 4 (2010) 6903–6913.
[11] J. Huang, Q. Li, D. Sun, Y. Lu, Y. Su, X. Yang, H. Wang, Y. Wang, W. Shao, H. Ning, [47] D.K. Tiwari, T. Jin, J. Behari, Dose-dependent in-vivo toxicity assessment of silver na-
J. Hong, C. Chen, Biosynthesis of silver and gold nanoparticles by novel sundried noparticle in Wistar rats, Toxicol. Mech. Methods 21 (2011) 13–24.
Cinnamomum camphora leaf, Nanotechnology 18 (2007) 105104–105115.
[12] S.S. Shankar, A. Rai, B. Ankamwar, A. Singh, A. Ahmad, M. Sastry, Biological synthesis of

381

You might also like