You are on page 1of 17

Nanotechnology

PAPER You may also like


- Preparation and characterization of
Green synthesis of Kocuran-functionalized silver graphene nanosheet doped with silver
nanoparticles
glyconanoparticles for use as antibiofilm coatings A H Mohammed and A N Naje

- Increasing the accumulation of aptamer


on silicone urethral catheters AS1411 and verapamil conjugated silver
nanoparticles in tumor cells to enhance
the radiosensitivity of glioma
To cite this article: C Ganesh Kumar and Pombala Sujitha 2014 Nanotechnology 25 325101 Jing Zhao, Dongdong Li, Jun Ma et al.

- Rapid microwave-assisted biosynthesis of


chitooligosaccharide coated silver
nanoparticles: assessments of
antimicrobial activity for paediatric pulp
View the article online for updates and enhancements. therapy
Subhathira Rajasekaran, Sneha S Rao,
Pandurang Appana Dalavi et al.

This content was downloaded from IP address 147.96.58.53 on 26/06/2023 at 11:49


Nanotechnology

Nanotechnology 25 (2014) 325101 (16pp) doi:10.1088/0957-4484/25/32/325101

Green synthesis of Kocuran-functionalized


silver glyconanoparticles for use as
antibiofilm coatings on silicone urethral
catheters
C Ganesh Kumar1,2 and Pombala Sujitha1,2
1
Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Uppal
Road, Hyderabad 500007, India
2
Medicinal Chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Uppal
Road, Hyderabad 500007, India

E-mail: cgkumar@iict.res.in and cgkumar5@gmail.com

Received 7 March 2014, revised 26 May 2014


Accepted for publication 17 June 2014
Published 25 July 2014

Abstract
Microbial infections due to biofilm formation on medical implants are serious complications
arising after surgery which can be prevented by using antimicrobial coatings on biomaterial
surfaces. We developed a simple, rapid and green chemistry approach for synthesis of silver
glyconanoparticles (AgNPs) using Kocuran, an exopolysaccharide produced by Kocuria rosea
strain BS-1. Kocuran-capped AgNPs exhibited a characteristic surface plasmon resonance (SPR)
peak around 435 nm. They were mono-dispersed, spherical with an average particle size of
12 nm. XRD and SAED studies suggested that AgNPs were crystalline in nature. AgNPs had a
zeta potential of −33.9 mV and were anionic charged. They showed colloidal stability at different
pH (6 to 10), temperatures (30 °C to 100 °C), in NaCl, NaNO3 and BSA solutions. Kocuran-
capped AgNPs exhibited effective antimicrobial activity against Staphylococcus aureus and
Escherichia coli and cell death was mainly due to hydroxyl radical induction and depletion of
NADH. They also inhibited the biofilm development by S. aureus and E. coli and confocal
scanning laser microscopic images revealed the damage of intact cell architecture. In vitro
evaluation of Kocuran-capped silver glyconanoparticles on human gingival fibroblasts
demonstrated good cell proliferation as compared to commercial AgNPs suggesting that they are
biocompatible and non-toxic in nature. This is a first report on Kocuran-functionalized AgNPs
exhibiting potential antibacterial and antiadhesive properties for use as antimicrobial coatings
against bacterial adhesion and biofilm formation on silicone urethral catheters.

Keywords: glyconanoparticles, antibiofilm coatings, urethral catheters, Kocuran,


exopolysaccharide
(Some figures may appear in colour only in the online journal)

1. Introduction biofilm formation onto the implant surfaces by opportunistic


microbes cause serious infection and implant failure [1].
Medical devices such as implants, prosthetics, etc are widely Prevention of colonization of these pathogens on medical
used in surgical cases for the replacement of damaged or lost implants is one the major medical issues which adds to
tissues and also finds use in critical care for fluid or gas financial burden since nosocomial infections represents one of
administration using catheters, respectively. Bacterial con- the most serious complications after surgery or critical care. It
tamination during surgery and subsequent adhesion and is reported that more than 1.4 million people worldwide suffer

0957-4484/14/325101+16$33.00 1 © 2014 IOP Publishing Ltd Printed in the UK


Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

from complications of hospital-acquired infections leading to smart nanomaterials for biomedical applications as probes of
a mortality rate of 10–15% [2, 3]. Staphylococcus aureus carbohydrate–carbohydrate interactions and carbohy-
(Gram-positive) and Escherichia coli (Gram-negative) are drate–protein interactions, antiadhesive therapy and biolabels
major causative agents of hospital-acquired infections, parti- [28, 29]. However, some reports indicate that it is difficult to
cularly in immuno-compromised patients, which contribute to produce polysaccharide-based glyconanoparticles with spe-
increased rates of implant-related systemic infections and cific surface functionalities, which is essential in controlling,
mortality [3]. A recent study demonstrated that different S. for example, distribution or cell interactions in vivo [30, 31].
aureus strains are responsible for catheter-related infections Considering the importance of glyconanoparticles from a
and among them 82% are methicillin-resistant strains having biotechnological and biomedical perspective, we report for
numerous genes involved in biofilm formation and bacterial the first time a simple, rapid and greener approach for the
dispersion [4]. synthesis of AgNPs functionalized with Kocuran [32] an
Antimicrobial polymers and coatings containing silver exopolysaccharide (EPS) produced by Kocuria rosea strain
have gained special attention in recent years due to their long- BS-1 with repeating monosaccharide residues of D-glucose,
term biocidal properties, high temperature stability and low D-mannose, D-galactose and D-glucuronic acid having an
volatility [5]. Silver is known to exhibit low toxicity towards average molecular mass of 51.2 kDa, and the application of
mammalian cells [6] and does not easily elicit microbial these Kocuran-capped silver glyconanoparticles (AgNPs) as
resistance [7]. It is reported to cause pits in bacterial cell antimicrobial and antibiofilm coatings on silicone urethral
walls, resulting in increased permeability and cell death [8]. In catheters.
general, the antimicrobial materials and coatings should have
desirable attributes such as potent antibacterial efficacy, low
toxicity, ease of fabrication and environmental safety, to 2. Experimental procedures
qualify them as promising candidates for use in various
applications. Hence, there is a significant interest in the 2.1. Materials
development of silver-based antimicrobial materials and
coatings for application in the health and biomedical devices, All chemicals were of analytical grade and procured from
personal hygiene and food industries. Sigma-Aldrich (St. Louis, MO, USA) unless otherwise stated.
Biodirected synthesis of metal nanoparticles has gained All culture media were purchased from HiMedia Laboratories
renewed attention which involves the use of various bioma- Pvt., Mumbai, India. The commercially available citrate-
terials from plants, bacteria, fungi and yeasts as reducing and synthesized silver nanoparticles (10 ± 2 nm) were obtained
stabilizing agents due to their biocompatibility, low toxicity from Nanocomposix, San Diego, CA, USA. The fluorescent
and eco-friendly nature [9, 10]. Among the metal nano- reporter dye, 3′-(p-hydroxyphenyl) fluorescein (HPF) and
particles, silver nanoparticles have unique optical, electro- LIVE/DEAD® BacLight™ Bacterial Viability kit were pro-
magnetic and physicochemical properties, which are cured from Molecular Probes, Eugene, OR, USA. Thermanox
dependent on their geometrical properties such as size and plastic cover slips were obtained from Nalgene Nunc Inter-
shape and function as bio-nanofactories finding a wide range national, Rochester, NY, USA.
of applications as antibacterial agents [11], and in the medical
sector as biodiagnostics, imaging, drug delivery, bio-labeling 2.2. Kocuran production from K. rosea strain BS-1
and hyperthermia of tumors [12, 13]. The size, shape, stability K. rosea strain BS-1, earlier isolated in our laboratory from a
and bioactivity are dependent on the capping agent used for petroleum contaminated soil sample produced an EPS with
silver nanoparticle synthesis. Silver nanoparticle preparations biosurfactant property [33]. EPS production from strain BS-1
using microbes such as bacteria, yeast, fungi, and actinomy- was achieved in a minimal salts medium containing (per litre):
cetes are well documented [10, 14]. The silver nanoparticle 10 g glucose, 2 g sodium nitrate, 0.5 g NH4SO4, 2.5 g
formation occurs in microorganisms either at intracellular KH2PO4, 2.0 g K2HPO4, 0.2 g MgSO4 and 1 g NaCl, by
[15] or extracellular [16] level. Biomolecules such as nitrate culturing at 35 °C in 1000 ml Erlenmeyer flasks containing
reductase, proteins, amino acids and polysaccharides secreted 250 ml of minimal salts medium (pH 8) with agitation at
by the bacteria act as capping ligands during the silver 180 rpm for 96 h in an Ecotron shaker (Infors AG, Switzer-
nanoparticle synthesis [17]. Some natural products like vita- land). The fermented medium was later subjected to cen-
min E [18], plant extracts [19], honey [20], sweet sorghum trifugation (Sorvall RC 5C Plus; Kendro Lab Products,
syrup [21], etc have also been explored for the synthesis of Ashville, NC, USA) at 10 000 g for 20 min to obtain a cell-
silver nanoparticles. free supernatant [32]. Kocuran (EPS) content in the cell-free
Glyconanoparticles prepared with sugars as capping supernatant was quantified by phenol sulphuric acid
ligands have gained attention in recent years. Recent studies method [34].
have demonstrated the preparation of gold and silver glyco-
nanoparticles functionalized with carbohydrates like glucose 2.3. Preparation and characterization of silver nanoparticles
[22], lactose [23], Ley tetrasaccharide [24], starch [25, 26] and
using Kocuran
rhamnolipids [27]. These carbohydrates possess many
hydroxyl and carbonyl groups which confer the glyconano- Ten milliliter of aqueous silver nitrate (100 mM, Sigma,
particles with unique H-bonding capabilities functioning as USA) was mixed with 90 ml of cell-free supernatant

2
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

containing Kocuran suspension (10 mg ml−1), and kept for room temperature for 60 days; the stored glyconanoparticles
continuous stirring at 200 rpm for 15 min at 50 °C. Sample were redispersed in water by sonication to regenerate a sol
aliquots (2 ml) were withdrawn at periodic intervals and followed by UV-visible spectroscopic analysis on differ-
monitored for the bioreduction of silver ions. UV–visible ent days.
spectra of these sample aliquots were recorded as a function
of reaction time from 300 to 600 nm on a UV–visible double
beam spectrophotometer (Lambda 25, PerkinElmer, Shelton, 2.5. Antimicrobial activity of Kocuran-capped silver
CT, USA) at room temperature (28 °C). The nanoparticles glyconanoparticles
were washed with distilled water, subjected to centrifugation
(Sorvall RC 5C Plus, Kendro Lab Products, Ashville, NC, Antimicrobial activity of Kocuran-capped AgNPs in com-
USA) at 9700 rpm for 30 min, concentrated and later stored at parison with commercially available citrate-synthesized silver
4 °C for further use. The synthesized glyconanoparticles were nanoparticles (10 ± 2 nm) was determined using the microtiter
further characterized using FT-IR, TEM, EDAX, XRD and broth dilution method [16]. The pathogenic bacterial strains
zeta potential analysis. The FT-IR spectrum of the AgNPs in used were Bacillus subtilis MTCC 121, S. aureus MLS 16
the form of KBr pellets was recorded on Thermo-Nicolet MTCC 2940, S. aureus MTCC 96, Micrococcus luteus
Nexus 670 spectrometer at a resolution of 4 cm−1 in wave- MTCC 2470, E. coli MTCC 739, Klebsiella planticola
number region of 400–4000 cm−1. Morphological analysis of MTCC 530 and Pseudomonas aeruginosa MTCC 2453,
silver nanoparticle formation was confirmed using transmis- along with drug resistant bacterial strains such as E. coli
sion electron microscope (TEM). The powdered AgNPs were ATCC 35218, S. aureus subsp. aureus ATCC 29213 and
re-dispersed in deionized water by ultrasonication for 5 min Enterococcus faecalis ATCC 29212. The different bacterial
and then subjected to TEM analysis by drying the nano- strains (107 cfu ml−1 cells) were inoculated in 100 μl of
particle dispersion on amorphous carbon-coated copper grids Muller–Hinton broth along with different concentrations of
at 37 °C. Kocuran-capped AgNPs were imaged using FEI
silver glyconanoparticles. After 24 h incubation, 40 μl of p-
Tecnai G2 S-Twin TEM instrument operated at an accel-
iodonitrotetrazolium (INT, Sigma) dye (0.02%, 20 mg INT
erating voltage of 300 kV. The energy dispersive x-ray
dissolved in 100 ml of 40% dimethylformamide) was added
(EDAX) spectroscopy was performed on a Scanning Electron
Microscope (Hitachi S-520) equipped with an EDAX detector to each well and incubated for 2 h. The reduction of p-iodo-
(Oxford LINK-ISIS 300) for elemental composition analysis nitrotetrazolium was spectroscopically measured at 450 nm
and EDAX spectrum was measured at 10 kV accelerating using TRIAD multimode reader (Dynex Technologies,
voltage. The x-ray diffraction (XRD) pattern of AgNPs was Chantilly, VA) to determine the minimum inhibitory con-
recorded employing a XPert PRO P Analytical PW 3040/60 centration (MIC) values. All the results were represented as
(PANalytical BV, The Netherlands) x-ray diffractometer the average of three independent experiments.
operated at a voltage of 40 kV and current of 30mA using
CuKα radiation (λ = 0.154 056 nm) and the scan parameters
set were scan rate 1.2° per minute and scan range 2.6. Growth analysis of S. aureus and E. coli
2θ = 10°–80°. The powdered AgNPs dispersed in deionized
The growth rate profile of S. aureus and E. coli in the pre-
water by ultrasonication were also subjected to dynamic light
sence of Kocuran-capped AgNPs at different concentrations
scattering (DLS) measurements at 25 °C on a Zetasizer Nano
ZS (Malvern Instruments, Worcestershire, UK) instrument was determined by measuring the optical density at 600 nm.
equipped with a He–Ne laser operating at 632.8 nm and a The optical density of 0.1 at 600 nm corresponded to a con-
scattering detector at 173°. The nanoparticle charge quantified centration of 108 cfu ml−1. The bacteria (OD600 of 0.1) were
as zeta potential was determined using the same instrument cultured in Mueller-Hinton broth in the absence and presence
at 25 °C. of Kocuran-capped AgNPs at different concentrations (0.1, 1,
5, and 10 μg ml−1); incubated at 37 °C for 24 h and the cell
viability (%) was assessed based on the colony forming units
2.4. Stability studies of Kocuran-capped silver
(cfu ml−1) measurements. The results are represented as the
glyconanoparticles
means of triplicate experiments.
Temperature and pH stability of Kocuran-capped AgNPs was
evaluated in the temperature range of 30 to 100 °C and
pH range of 6 to 10. Further, the stability was assessed in 2.7. TEM studies
distilled water, NaCl (different concentrations from 10, 50,
100, 150 and 200 mM), NaNO3 (different concentrations The bacterial cultures were centrifuged at 5000 g and washed
from 10, 50, 100, 150 and 200 mM), and in BSA solution thrice with 0.1 M HEPES buffer and then fixed overnight with
(different concentrations ranging from 0.05, 0.1, 0.2, 0.3, 0.4 1% (v v−1) osmium tetroxide in HEPES buffer followed by
and 0.5%). In all the cases, the stability of silver glycona- sequential gradient dehydration from 10% to 100% (v v−1)
noparticles was monitored by measuring the absorbance ethyl alcohol and then TEM images were captured on FEI
changes using UV-visible absorption spectroscopy. The sta- Tecnai G2 S-Twin TEM (FEI Company, Hillsboro, OR)
bility of Kocuran-capped AgNPs was monitored by storage at operated at an accelerating voltage of 300 kV.

3
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

2.8. Measurement of hydroxyl radical formation in S. aureus 2.11. Biofilm assay in S. aureus and E. coli
and E. coli
The effect of Kocuran-capped AgNPs on the biofilm formation
The induction of the formation of hydroxyl radicals in S. of S. aureus and E. coli cultures was assessed by crystal violet
aureus and E. coli cultures in the presence of Kocuran-capped staining [37]. The cultures were initially incubated in 96-well
AgNPs was measured using the fluorescent reporter dye, 3′- microtiter plates at 37 °C for 12 h. The cultures were treated
(p-hydroxyphenyl) fluorescein (HPF). The bacterial cultures with Kocuran-capped AgNPs at different concentrations (0.1,
(OD600 of 0.1) were treated with Kocuran-capped AgNPs 1, 5 and 10 μg ml−1) and re-incubated at 37 °C for 24 h. The
(5 μg ml−1) and incubated at 37 °C for 3 h. Samples were biofilm formation was assessed by staining with 0.1% crystal
collected just before the addition of AgNPs and at periodic violet for 5 min and the dye was solubilized with 150 μl of
intervals of 1 h, washed with distilled water and suspended in isopropanol-0.04 N HCl and 50 μl of 0.25% SDS and the plates
PBS (pH 7.2). The hydroxyl radical formation was detected were spectroscopically measured at 590 nm. The percentage of
biofilm formation was calculated from the equation: [1 − (ODT/
using 5 mM of HPF by measuring the mean fluorescence on
ODC)] × 100 where ODT is the optical density of the cultures
FACS Caliber flow cytometer (BD Biosciences, Mountain
treated with Kocuran-capped AgNPs and ODC is the optical
View, CA, USA).
density of untreated cultures. All the results were represented
as the average of three independent experiments.
2.9. Measurement of protein leakage in S. aureus and E. coli
2.12. Confocal laser scanning microscopy
The protein leakage from S. aureus and E. coli cells was
The biofilm formation was assessed using LIVE/DEAD®
determined by treatment of the bacterial cultures (OD600 of
BacLight™ Bacterial Viability kit and visualized by confocal
0.1) with Kocuran-capped AgNPs at a concentration of laser scanning microscopy. Briefly, the cells of S. aureus and
5 μg ml−1 and incubated at 37 °C for 4 h. Sample aliquots E. coli (OD600 = 0.01) were cultured on Thermanox plastic
(1 ml) were collected periodically at hourly intervals and cover slips placed in the wells of 24-well plates containing
centrifuged at 5000 g for 30 min at 4 °C. The protein content Muller–Hinton broth for 12 h at 37 °C. Later, they were
in the cell-free supernatant was quantified using Bradford’s treated with Kocuran-capped AgNPs at different concentra-
assay [35]. The untreated cultures were used as controls and tions (0.1, 1, 5 and 10 μg ml−1) for 24 h. The cover slips were
all the experiments were run in triplicates. washed with PBS and the bacterial biofilms were stained with
1 ml of 0.6% Live/Dead BacLight stain following the staining
kit instructions. The stained biofilms were visualized by
2.10. Measurement of NAD+ and NADH concentration in S. Olympus FluoView 500 confocal laser microscope (Olympus
aureus and E. coli Optical, Tokyo, Japan) using an argon ion laser at
480–490 nm for excitation and a 500–635 nm band pass filter
The changes in NAD+ and NADH concentration in S. aureus
for emission. CLSM images were processed by Olympus
and E. coli cultures in the presence of Kocuran-capped FluoView 500 software and the scale bar for each image
AgNPs was monitored by NAD cycling assay [36]. The S. is 10 μm.
aureus and E. coli cultures were treated with silver glyco-
nanoparticles (5 μg ml−1) and incubated at 37 °C. Sample 2.13. Biofilm inhibition assay on silicone urethral catheters
aliquots (5 ml) were collected at periodic hourly intervals for
6 h, centrifuged at 10 000 g at 4 °C for 10 min and the cell Silicone urethral catheters were coated with the Kocuran-
pellets were collected. To the ice-cold cell pellets, 75 μl of capped AgNPs according to a simple dispersion method. A
0.2 M NaOH was added to extract NADH and 75 μl of 0.2 M sterile silicone urethral catheter (Unomedical A/S, Birkerod,
HCl was added for NAD+ extraction. The samples were then Denmark) was cut into 4 cm long pieces and these pieces
heated at 100 °C for 10 min and centrifuged at 10 000 g for were placed in test tubes incubated in the colloidal solution of
5 min. The NAD+ cycling assay was performed in a 96-well Kocuran-capped AgNPs (100 μg) for 24 h at 37 °C. The
catheters were later completely dried at 50 °C to form an
microtiter plate. The reaction mixture contained 30 μl of 1 M
antimicrobial coating. The time-dependent performance of the
Bicine buffer (pH 8.0), 75 μl sample, 75 μl neutralizing buffer
antimicrobial coating of Kocuran-capped silver glyconano-
(0.1 M HCl for NADH, or 0.1 M NaOH for NAD+), 30 μl of
particles on silicone urinary catheters was evaluated against
phenazine methosulfate, 30 μl of 3-(4, 5–dimethylthiazol-2- uropathogenic strains of S. aureus and E. coli which were
yl)-2,5-diphenyltetrazolium bromide (MTT), 30 μl of 100% grown overnight in test tubes with sterile silicone urethral
ethanol and 30 μl of 40 mM EDTA. After incubation at 30 °C catheters coated with and without Kocuran-capped AgNPs
for 3 min, 6 μl of yeast alcohol dehydrogenase I (ADH-I, (100 μg) and the tubes were then incubated for 24, 48, 72 and
Sigma) at a concentration of 500 U ml−1 was added and the 96 h at 37 °C. The cultures were removed and the catheters
rate of MTT reduction was recorded by measuring the were washed with distilled water. After washing, 3 ml of
absorbance at 570 nm. The rate of MTT reduction is pro- crystal violet (0.1%) was added to the catheters and stained
portional to the concentration of NAD+ or NADH in the for 10 min. The stained biofilms were rinsed with distilled
sample. water (3×) and allowed to dry at room temperature for 15 min

4
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

before examination. The dye was solubilized with 150 μl of that would have an influence on the size, shape or stability.
isopropanol-0.04 N HCl and 50 μl of 0.25% SDS and the Based on the optimization studies, the optimal concentrations
collected dye solution was read at 590 nm on a UV–visible to be used were Kocuran (10 mg ml−1) and AgNO3 (100 mM).
double beam spectrophotometer (Lambda 25, PerkinElmer, The bioreduction of silver ions was monitored by UV-visible
Shelton, CT, USA). The results were expressed in percent spectroscopy and the spectra for the reaction of cell-free
inhibition of growth of free-floating organisms or biofilm supernatant with silver nitrate were recorded at room tem-
formation in each test tube relative to the average growth or perature (28 °C) as a function of time (supplementary figure
biofilm formation in control tubes (tubes with untreated S3). The reaction due to the reduction of silver ions was evi-
catheters). The results were represented as the means of tri- denced by the development of yellowish brown color. This
plicate experiments. High resolution-scanning electron could be attributed to the direct redox reaction between
microscope (HR-SEM) images were also recorded using a Kocuran and silver nitrate, since there was no addition of any
Hitachi S-4700 Scanning Electron Microscope (Hitachi High reducing agent in the reaction mixture. The nanoparticles
Technologies, Pleasanton, CA) to determine the surface exhibited an absorption peak around 435 nm after 6 min of
topography of biofilm formation on untreated and treated reaction, which is a characteristic SPR band of silver nano-
catheters coated with Kocuran-capped AgNPs. particles possibly due to the excitation of longitudinal plasmon
vibrations in silver nanoparticles in the solution [39, 40]. It was
2.14. In vitro biocompatibility studies of Kocuran-capped silver also observed that the absorption peak intensity increased as a
glyconanoparticles function of time and varied with increased Kocuran con-
centration (data not shown). This variation can be attributed to
The in vitro biocompatibility of Kocuran-capped silver gly- the difference in particle size and shape of the synthesized
conanoparticles was evaluated on human gingival fibroblast nanoparticles [41, 42]. Further, the increased intensity and
(HGF) cell line procured from ScienCell Research Laboratories sharpness in the SPR band of excitation spectra indicated that
(Carlsbad, CA, USA) using 3-(4,5-dimethylthiazol-2yl)-2,5- the silver nanoparticles were effectively capped and/or stabi-
diphenyl tetrazolium bromide (MTT) reduction method [38]. lized with Kocuran and were non-agglomerated. In the case of
The HGF cell line was grown in DMEM supplemented with polysaccharide-based nanoparticle synthesis, it was demon-
10% fetal bovine serum and 100 IU ml−1 penicillin-strepto- strated that the sugar moieties of the EPS molecules mainly
mycin and incubated at 37 °C in 5% CO2 humidified atmo- contribute to the reducing effect and can also act as effective
sphere. For assessing the in vitro biocompatibility, the HGF colloidal stabilizers that can be explored for green synthesis of
were cultured at a cell density of 5 × 103 cells well−1 in 96 well nanoparticles [43].
plates for 12 h and then treated with Kocuran, Kocuran-capped The proposed mechanism of reduction in the case of
silver glyconanoparticles and commercially available citrate- Kocuran can be explained as follows: Kocuran is an acidic
synthesized AgNP at various concentrations (1–200 μg ml−1). tetrapolysaccharide with repeating sugar residues of glucose,
The cells were incubated at 37 °C for 48 h and then 20 μl of galactose, mannose (sugar content 91%) and glucuronic acid
MTT solution (5 mg ml−1 in PBS) was added to each well and content (9%) [32]. The hydroxyl and carboxylic groups of the
again incubated for 4 h at 37 °C. The purple colored formazan sugar moieties present in this biopolymer assists in the
crystals formed were dissolved in 150 μl well−1 of DMSO and complexation of silver ions. The silver ions oxidize the
the cell proliferation (%) was measured at 570 nm in a hydroxyl groups to carbonyl groups and during this process
microplate reader (BioRad Laboratories, Hercules, CA, USA). the silver ions are reduced to elemental silver. In addition to
The results are represented as the means of triplicate experi- this inherent oxidation, the dissolved air may also cause
ments run in four replicates. oxidation of the existing hydroxyl groups to carbonyl groups
such as aldehydes and carboxylates. These potential reducing
2.15. Statistical analysis aldehyde groups along with the other existing carbonyl
Statistical analysis was performed using GraphPad PRISM groups reduce more and more of silver ions to elemental
software version 3.0 (GraphPad Software, La Jolla, CA, silver. In this process, the nanoparticles are probably capped
USA). Data are expressed as the mean ± S.D of three inde- and stabilized by the EPS molecules. Since these carbohy-
pendent experiments. All experimental data were compared drate polymers are very complex, it is expected that more than
using Student’s t-test. In all comparisons, p < 0.05 was con- one mechanism is involved in the complexation and sub-
sidered statistically significant. sequent reduction of silver ions by the Kocuran poly-
saccharide. Complexation of silver ions by hydroxyl groups
and its subsequent reduction by aldehyde groups was earlier
3. Results and discussion reported in the case of starch-capped silver nanoparticles [44].
In the case of silver nanoparticles produced using gum Aca-
cia, the carboxylate groups were involved in the complexa-
3.1. Synthesis and characterization of Kocuran-capped silver
glyconanoparticles
tion of silver ions and its subsequent reduction by hydroxyl
groups [45].
Preliminary optimization experiments were carried out to The FT-IR spectra revealed that the surface functional
determine the optimal concentrations of Kocuran (supple- groups present on the Kocuran-capped AgNPs and that of
mentary figure S1) and silver nitrate (supplementary figure S2) pure Kocuran. The absorption peak position observed at

5
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 2. XRD spectrum of Kocuran-capped silver


glyconanoparticles.

Figure 1. Transmission electron micrograph of Kocuran-capped


silver glyconanoparticles dispersed on an amorphous carbon-coated the state of the nanoparticle surface and predicting its long-
copper grid. Scale bar corresponds to 20 nm. Inset: selected electron term stability. Kocuran-capped AgNPs were stable and
area diffraction (SAED) pattern of silver glyconanoparticles. showed no agglomeration which was supported by the zeta
potential value of −33.9 mV and were anionic charged (sup-
3429 cm−1 corresponded to the hydroxyl stretch, while the plementary figure S6). It was reported that nanoparticles with
absorption peaks observed at 2924 cm−1 represented the C–H zeta potential values greater than +30 mV or lower than
stretching. The peaks at 1073 and 693 cm−1 were attributed to −30 mV typically have high degrees of stability. However, the
C–O–C and C–O-P rings of polysaccharides (supplementary dispersions with a low zeta potential will eventually aggregate
figure S4(a)). After the bioreduction of silver nitrate with EPS due to van der Waals interparticle attraction [48]. The EDAX
sugars in the solution phase, the created peak at 1636 cm−1 is spectral analysis revealed the elemental composition profile of
characteristic to the binding of the –C = O functional moiety the synthesized Kocuran-capped AgNPs, indicating the pre-
and the shift in the peak at 1032 towards a lower frequency as sence of strong elemental silver signals at 3 keV (supple-
compared to the peak positioned at 1073 cm−1 for Kocuran is mentary figure S7). Furthermore, the XRD spectrum of the
attributed to the binding of C-O-C and C-C-H groups of the Kocuran-capped AgNPs (figure 2) showed four main char-
Kocuran EPS with silver nanoparticles. Further, the broad acteristic Bragg diffraction peaks for silver nanocrystallites at
peak at 635 cm−1 is related to the silver nanoparticles bonding 2θ values of 40.1, 45.1, 65.0, and 82.1 which corresponded to
with oxygen from the hydroxyl groups of the sugar residues the main (111), (200), (220) and (311) crystallographic planes
(supplementary figure S4(b)). Based on these observations, of the face-centered cubic (fcc) silver crystals, which con-
the FT-IR spectra clearly suggests that Kocuran molecules firmed the SAED results. The size, shape, stability and
capped on the silver nanoparticles and the hydroxyl groups bioactivity are mainly dependent on the type of the capping
played a key role in reducing and stabilizing the silver ligand used for nanoparticle synthesis. Different natural
nanoparticles. Our results corroborates with the observations polysaccharides such as starch [44], gum acacia [45], chitosan
of the earlier reports [46, 47]. [49], and gum ghatti (Anogeissus latifolia) [50] have been
To gain a detailed understanding of the size and shape of earlier explored for the synthesis of silver nanoparticles.
silver glyconanoparticles, the surface morphology of the Recent reports suggests that diverse polysaccharides of
Kocuran-capped AgNPs was examined by TEM. The TEM microbial origin such as carboxymethylated curdlan and
image (figure 1) clearly revealed that the Kocuran-capped fucoidan biopolymers [51], sulphated polysaccharides iso-
AgNPs were found to be non-agglomerated, predominantly lated from marine red algae, Porphyra vietnamensis [52],
were spherical in shape and mono-dispersed. The average exopolysaccharides from Lactobacillus rhamnosus [53] and
particle size and size distribution (polydispersity index) of the B. subtilis [47] have gained renewed interest as promising
AgNPs were determined by DLS analysis and the average capping ligands for the biosynthesis of AgNPs for different
size of nanoparticles computed from the histogram was found therapeutic applications.
to be 12 nm with a narrow size distribution (supplementary
figure S5). The electrostatic repulsion between the nano-
3.2. Stability analysis of Kocuran-capped silver
particles depends on the zeta potential value (measured in
glyconanoparticles
mV). It is observed that the higher the zeta potential, the
stronger the repulsion which results in a more stable system It was earlier reported that the instability of silver nano-
[48]. Therefore, the zeta potential value aids in understanding particles at different physical and chemical conditions leads to

6
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

the dissociation of ions, formation of aggregates and increase Table 1. Antimicrobial activities of Kocuran, Kocuran-capped silver
in the particle size that affects the electromagnetic properties, glyconanoparticles and commercially available silver nanoparticles.
mobility and bioavailability of silver nanoparticles [54]. Minimum inhibitory concentration (MIC,
Considering these facts, the effect of pH, temperature, solu- μg ml−1)
tions of NaCl, NaNO3 and BSA on the stability of Kocuran-
Kocuran-capped
capped AgNPs was studied by measuring the change in the Bacterial silver glyconano- Silver nano-
SPR absorption band using UV-visible spectroscopy. The strainsa Kocuran particles particlesb
Kocuran-capped AgNPs were stable at a pH range of 7 to 10
(supplementary figure S8(a)) and temperature range of 30 °C Staphylococcus 150 4.68 9.37
to 100 °C (supplementary figure S8(b)). In both the cases, aureus
there was no significant change observed in their absorption MTCC 96
S. aureus 150 9.37 18.75
spectrum. In addition, the Kocuran-capped AgNPs exhibited
MLS16
good stability when dispersed in electrolyte solutions of NaCl MTCC 2940
and NaNO3 at concentrations of 10, 50, 100 and 150 mM Bacillus subtilis 75 18.75 18.75
(supplementary figures S8(c), (d)). However, at 200 mM MTCC 121
concentration of NaCl and NaNO3, there was a slight decrease Micrococcus 75 37.5 75
in the absorption intensity. The AgNPs also showed similar luteus
absorption spectrum in BSA solutions of 0.05, 0.1, 0.2 and MTCC 2470
0.3%, while a slight decrease in the absorption intensity was Klebsiella plan- 150 18.74 18.74
observed in the case of 0.4 and 0.5% BSA solutions (sup- ticola
plementary figure S8(e)). The slight decrease in the absorp- MTCC 530
tion intensity at higher concentration (>200 mM) for Escherichia coli 75 4.68 9.37
MTCC 739
electrolyte solutions like NaCl and NaNO3, and BSA solution
Pseudomonas 150 37.5 75
concentrations (0.4% and 0.5%) is mainly due to the aggre- aeruginosa
gation of nanoparticles [55]. These stability studies clearly MTCC 2453
demonstrate that the Kocuran-capped AgNPs under different E. coli ATCC 300 37.5 75
conditions of pH, temperature, NaCl, NaNO3 and BSA 35218
solutions qualifies it as a promising candidate for various S. aureus subsp. 300 18.74 75
biological and therapeutic applications. The stability of the aureus ATCC
Kocuran-capped AgNPs was evaluated by UV-visible spec- 29213
troscopy and the respective absorption spectra were recorded Enterococcus 300 18.74 18.74
after 1 h, 5 days, 10 days, 15 days, 30 days, 45 days and 60 faecalis
days of storage at room temperature (supplementary figure S8 ATCC 29212
(f)). The UV-visible spectra showed that the Kocuran capped a
MTCC = Microbial type culture collection, CSIR-Institute of Microbial
AgNPs exhibited an intense SPR peak around 450 nm. As the Technology, Chandigarh, India; ATCC = American type culture collection,
time progressed from 1 to 30 days, similar plasmon resonance Manassas, VA, USA.
b
bands were observed with no change in the position of SPR, Commercial AgNP (citrate-reduced) procured from NanoComposix, San
Diego, CA, USA.
except for a decrease in the absorption intensity. After 30th
day, a shift in the position of SPR peak was observed with a
decrease in intensity, suggesting that the stability of nano-
particles decreased after 30 days. 18.74 and 18.74 μg ml−1, while the commercial AgNP
showed MIC values of 75, 75 and 18.74 μg ml−1. In com-
parison to commercial AgNP, the Kocuran-capped silver
3.3. Antimicrobial activity studies of Kocuran-capped silver
glyconanoparticles exhibited good antimicrobial activity
glyconanoparticles
which qualifies them as a promising antimicrobial agent
Kocuran-capped AgNPs exhibited good antibacterial activity against multidrug resistant bacterial pathogens.
against both Gram-positive and Gram-negative bacterial The MIC value of a particular antimicrobial agent is an
strains. The minimum inhibitory concentrations observed important quantitative parameter dependent on the microbial
against different bacterial pathogens and drug resistant strains strain. Different factors such as particle size, shape, crystal-
are shown in table 1. Kocuran-capped AgNPs exhibited linity, surface chemistry, pH, ionic strength, divalent cations,
promising antimicrobial activity against S. aureus MTCC 96 ligands and macromolecules were demonstrated to affect the
and E. coli MTCC 739 with MIC values of 4.68 μg ml−1, and antimicrobial activity [56]. In the present study, Kocuran and
S. aureus MLS16 MTCC 2940 (MIC value of 9.37 μg ml−1), the corresponding Kocuran-capped silver glyconanoparticles
while moderate antimicrobial activity was observed against exhibited different MIC values with regard to different bac-
M. luteus MTCC 2470 and P. aeruginosa MTCC 2453 with terial strains. This may be due to the fact that when Kocuran
MIC values of 37.5 μg ml−1. The MIC values for the drug and the corresponding Kocuran-capped silver glyconano-
resistant strains such as E. coli ATCC 35218, S. aureus subsp. particles interacted with the test strains; the bacterial strains
aureus ATCC 29213 and E. faecalis ATCC 29212 were 37.5, counteract the effect by secreting secondary metabolites into

7
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

the medium which alter the pH and ionic strength. These 10 μg ml−1 showed a significant decrease in the cell viability
variations influence the interaction and binding of nano- (cfu ml−1) and the growth was completely inhibited after 2 h
particles to the bacterial cell and also the aggregation states of of post-treatment period, in the case of both S. aureus and
silver nanoparticles. The microbial strains under these con- E. coli cultures. This observation was further confirmed by
ditions exhibited differences in the vulnerability to silver TEM analysis (figure 3), which revealed that in the case of
nanoparticles and hence the differences in MIC values were both S. aureus and E. coli cultures treated with Kocuran-
observed against the tested bacterial strains. capped AgNPs (5 μg ml−1), the intracellular localization of
Further, the antimicrobial activity effect exhibited by AgNPs was observed which exerted the possible bactericidal
Kocuran-capped silver nanoparticles is additive since the action.
silver nanoparticles synthesis is without the addition of any
external reducing agent and Kocuran itself acted as a capping
3.5. Effect of Kocuran-capped silver glyconanoparticles on
ligand causing the reduction and stabilization of the silver
hydroxyl radical production in S. aureus and E. coli
nanoparticles. In order to prove that Kocuran-capped glyco-
nanoparticles contributed to the antimicrobial activity, The ability of Kocuran-capped AgNPs to induce reactive
experiments were performed in comparison with commer- oxygen species (ROS) production was monitored by mea-
cially available citrate-synthesized silver nanoparticles. The suring the production of hydroxyl radicals. The hydroxyl
results suggest that Kocuran-capped glyconanoparticles radical production in S. aureus MTCC 96 and E. coli MTCC
showed good antimicrobial activity as compared to citrate- 739 in the presence of silver glyconanoparticles at a lethal
synthesized silver nanoparticles. It was earlier reported that concentration (5 μg ml−1) was measured as a function of time
EPS capped AgNPs exhibited efficient bactericidal activity using 3′-(p-hydroxyphenyl) fluorescein (HPF) fluorescence.
and higher adherence towards the bacterial cell surface. The In the case of untreated cultures of S. aureus and E. coli, there
physical interaction between the nanoparticles and the bac- was no significant increase in the hydroxyl radical production
terial cell wall surface was found to be the key factor for cell over the post-treatment period as indicated by a constant HPF
damage [47]. Similarly, in the present study, the Kocuran fluorescence (figures 4(a), (c)). When S. aureus and E. coli
polysaccharide capped on the AgNPs might have contributed cultures were treated with Kocuran-capped AgNPs, there was
to the better interaction with the bacterial cell wall and also an increase in the HPF fluorescence during the post-treatment
enhanced the effective anchoring of AgNPs on the cell sur- period indicating the generation of hydroxyl radicals
face. The AgNP anchored to the bacterial cell surface serve as (figures 4(b), (d)). This steady increase in the hydroxyl radi-
reservoirs for silver ions and the release of these silver ions cals in both S. aureus and E. coli is lethal to the bacterial cells
causes cell death. which results in a decreased absorbance (confirmed by growth
In the present study, S. aureus MTCC 96 and E. coli curve analysis). This suggests the probable adverse role of
MTCC 739 strains exhibited promising antimicrobial activity ROS in the primary generation of hydroxyl radicals by
and they were selected for further studies as representative Kocuran-capped AgNPs which results in lethal and lytic
Gram-positive and Gram-negative bacterial strains, which are effects in the case of both S. aureus and E. coli cells.
the most common pathogenic bacteria. It was earlier reported
that silver and nanosilver exhibited effective antimicrobial
3.6. Effect of Kocuran-capped silver glyconanoparticles on
activity against different pathogenic species such as E. coli, B.
microbial electron transport system
subtilis, Vibrio cholerae, S. aureus, P. aeruginosa and
Streptococcus pyrogens [57]. The mechanism of antibacterial The effect of Kocuran-capped AgNPs on the microbial elec-
effect of nanosilver is complex and the cell death is majorly tron transport chain was assessed by measuring the change in
caused by direct damage of the cell membrane, uncoupling the levels of NAD+/NADH ratios through NAD cycling
oxidative phosphorylation, induced free radical formation and assay. The change in the ratios of NAD+ and NADH (NAD+/
interfering with the respiratory chain, electron chain system, NADH) in S. aureus MTCC 96 and E. coli MTCC 739 during
proteins, membrane-bound enzymes and DNA [56]. the post-treatment of Kocuran-capped AgNPs is shown in
figures 5(a), (b). After 1 h of post-treatment with AgNPs
3.4. Effect of Kocuran-capped silver glyconanoparticles on the
(5 μg ml−1), a predominant increase in NAD+/NADH ratios to
2.26 and 2.83 was observed in the case of both S. aureus and
growth of S. aureus and E. coli
E. coli, respectively, while in the case of untreated cultures,
The effect of Kocuran-capped AgNPs at concentrations of the NAD+/NADH ratios remained at a constant level of 0.47
0.1, 1, 5 and 10 μg ml−1 on the growth of S. aureus MTCC 96 and 0.32 for both S. aureus and E. coli cultures, respectively.
and E. coli MTCC 739 was assessed as a function of time and Kocuran-capped AgNPs affected the microbial electron
the growth curves are represented as supplementary figures transport system as indicated by the increase in NAD+/NADH
S9(a), (b). The untreated control group of S. aureus and ratios which is due to increased oxidation of NADH through
E. coli exhibited normal growth pattern as observed by a the electron transport chain. Further, the hyperactivation of
steady increase in the optical density at 600 nm over the electron transport chain and rapid depletion of reducing
incubation time. When treated with 0.1 μg ml−1 of AgNPs equivalents resulted in superoxide production [58, 59]. It was
there was no significant change in the growth for both S. established that the superoxide causes damage to the iron-
aureus and E. coli cultures, while treatments with 5 and sulphur clusters of the redox system making it susceptible to

8
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 3. TEM analysis of (a) S. aureus MTCC 96 and (b) E. coli MTCC 739 treated with Kocuran-capped silver glyconanoparticles. The
organisms were cultured in Muller–Hinton broth and treated with Kocuran-capped silver glyconanoparticles (5 μg ml−1) and further
incubated at 37 °C for 24 h. The intracellular localization of Kocuran-capped silver glyconanoparticles was observed through TEM images
(A1 and A2 of S. aureus MTCC 96, and B1 and B2 of E. coli MTCC 739). Scale bar of A1 and B1 is 1 μm, while A2 and B2 are magnified
images of A1 and B1 at scale bar of 200 nm.

Fenton reaction. These ferrous ions are oxidized through the the viability of S. aureus and E. coli cells. It was earlier
Fenton reaction forming hydroxyl radicals [60]. reported that hydroxyl radicals are extremely toxic and exert
deleterious effect on proteins, DNA, lipids and other bio-
molecules. They react with the membrane lipids to cause lipid
3.7. Effect of Kocuran-capped silver glyconanoparticles on peroxidation and increased membrane permeability that
protein leakage in S. aureus and E. coli results in protein leakage and subsequent cellular death [61].
The bactericidal action of Kocuran-capped AgNPs causing
protein leakage was monitored by measuring the release of
3.8. Antibiofilm activity of Kocuran-capped silver
protein content from the bacterial cells during the post-treat-
glyconanoparticles
ment period. The extent of protein leakage in S. aureus
MTCC 96 and E. coli MTCC 739 cells when treated with Kocuran-capped AgNPs effectively inhibited the biofilm
Kocuran-capped AgNPs at 5 μg ml−1 is depicted in supple- formation by S. aureus MTCC 96 and E. coli MTCC 739 in a
mentary figures S10(a), (b). After 1 h of post-treatment, there concentration-dependent manner (figure 6). Kocuran-capped
was no significant increase in the protein leakage in the case AgNPs at 1 μg ml−1 concentration reduced the preformed
of both S. aureus and E. coli cells as compared to untreated biofilm formation by S. aureus and E. coli to 61.44% and
cultures. After 2 h of post-treatment, the protein leakage 58.16%, respectively. When the biofilms were treated with
increased from 27.9 μg ml−1 to 45.7 μg ml−1 in the case of S. 5 μg ml−1 of AgNPs, the biofilm formation was 20.08% and
aureus, while the protein leakage in the case of E. coli 18.17% as compared to the untreated cultures. In order to
increased from 21.1 μg ml−1 to 50.18 μg ml−1. However, the check the antibiofilm activity, the biofilms were stained with
untreated S. aureus and E. coli cultures showed no increase in LIVE/DEAD BacLight bacterial viability kit and the confocal
protein leakage. This increased protein leakage from the laser scanning microscope (CLSM) images showed a dose-
bacterial cultures upon treatment to lethal doses of Kocuran- dependent inhibitory effect of Kocuran-capped AgNPs on the
capped silver glyconanoparticles correlates to the decline in biofilm formation by S. aureus MTCC 96 (figure 7) and

9
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 4. Effect of Kocuran-capped silver glyconanoparticles on hydroxyl radical production in S. aureus MTCC 96 and E. coli MTCC 739.
The hydroxyl radical production was measured using the HPF fluorescence as a function of time in (a) S. aureus MTCC 96 without Kocuran-
capped silver glyconanoparticles treatment (control). (b) S. aureus MTCC 96 treated with Kocuran-capped silver glyconanoparticles. (c)
E. coli MTCC 739 without Kocuran-capped silver glyconanoparticles treatment (control). (d) E. coli MTCC 739 treated with Kocuran-
capped silver glyconanoparticles. The S. aureus MTCC 96 and E. coli MTCC 739 bacterial cells were cultured in Muller–Hinton broth in the
presence of Kocuran-capped silver glyconanoparticles (5 μg ml−1) and incubated at 37 °C for 24 h and the mean HPF fluorescence was
measured. The S. aureus MTCC 96 and E. coli MTCC 739 cultures without treatment of Kocuran-capped silver glyconanoparticles were run
in parallel as controls. The peaks represent hydroxyl radical formation at 1 h (black line), 2 h (blue line), 3 h (green line) and 4 h (red line)
after post-treatment, respectively.

E. coli MTCC 739 (figure 8). The LIVE/DEAD BacLight kit biofilm and caused bacterial cell death. These results reflect
is used to monitor the viability of bacterial populations as a the strong inhibitory effect of Kocuran-capped AgNPs on
function of the membrane integrity of the cells which uses biofilm formation by S. aureus and E. coli.
appropriate mixtures of SYTO 9, a green fluorescent nucleic
acid dye, and propidium iodide, a red fluorescent nucleic acid
3.9. Antibiofilm property of Kocuran-capped silver
dye. The SYTO 9 is permeable to live bacterial cells having
glyconanoparticle coating on silicone urethral catheters
intact cell membranes and stains them green, while the pro-
pidium iodide stains red the bacteria having damaged cell The proposed mechanism of Kocuran-capped silver glyco-
membranes that are considered to be dead or in dying state. nanoparticle-coating on the silicone urethral catheters can be
Under CSLM, the images are thus observed as green and red explained as follows: surface charge and hydrophobicity are
fluorescent images for live and dead bacteria, respectively. the two properties that play a critical role in determining the
The untreated S. aureus and E. coli cells showed rapid biofilm surface chemistry of the coating material that will influence
formation. There was no significant change in the biofilm the bacterial attachment and biofilm formation [62]. The
formation by S. aureus and E. coli when they were treated Kocuran-capped silver glyconanoparticles are negatively
with 0.1 μg ml−1 of Kocuran-capped AgNPs. Further, when charged based on the zeta potential value and the coatings on
treated with 1 μg ml−1 of AgNPs, the intact architecture in S. the hydrophobic silicone urethral catheters are due to elec-
aureus and E. coli was changed. The treatment with 5 and trostatic interactions which in turn repel bacterial adhesion
10 μg ml−1 of AgNPs damaged the structural integrity of the due to electrostatic repulsion between the often negatively

10
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

charged bacterial surface and the negatively charged glyco-


nanoparticle coated catheter surface. Further, the time-
dependent performance evaluation of Kocuran-capped silver
glyconanoparticle-based coating on silicone urethral catheters
showed inhibition in the biofilm formation by S. aureus and
E. coli after 24, 48, 72 and 96 h incubation as revealed by the
crystal violet assay and illustrated in figures 9 and 10. The
antimicrobial effect on biofilm formation by Kocuran-capped
silver glyconanoparticle-based coating on silicone urethral
catheters as a function of time showed that the biofilm for-
mation in Kocuran-capped AgNPs-coated catheters was 9.1%
and 10.7%, while in the case of uncoated catheters was
89.99% and 93.24%, respectively, after 24 h incubation, as
quantified by crystal violet assay. Moreover, the Kocuran-
capped silver glyconanoparticles-coated catheters showed
reduced biofilm formation of 13.78% and 14.29% in S. aur-
eus and E. coli, respectively, even after 96 h of incubation,
while the uncoated catheters showed 99% biofilm formation
in the case of both S. aureus and E. coli (supplementary
figures S11(a), (b)). Thus, the catheters functionalized with
Kocuran-capped silver glyconanoparticles acted as anti-
microbial coatings which were effective in inhibiting the
bacterial adhesion and preventing the biofilm formation by
uropathogenic bacterial strains like S. aureus MTCC 96 and
E. coli MTCC 739.
Figure 5. Effect of Kocuran-capped silver glyconanoparticles on The HR-SEM analysis to reveal the surface topography
NAD+/NADH ratio in S. aureus MTCC 96 and E. coli MTCC 739. confirmed that the AgNPs-coated catheters showed inhibition
The NAD+/NADH ratios were measured as a function of time in (a)
S. aureus MTCC 96 and (b) E. coli MTCC 739 in the presence of of biofilm formation by S. aureus and E. coli (figure 11),
Kocuran-capped silver glyconanoparticles. The S. aureus MTCC 96 while the biofilm formation was profuse in the case of
and E. coli MTCC 739 were cultured in Muller–Hinton broth in the uncoated catheters (figure 12). Intensive care units are the
presence of Kocuran-capped silver glyconanoparticles (5 μg ml−1) common sites for transmission of nosocomial bacterial
and incubated at 37 °C for 24 h and the levels of NAD+/NADH infections which result in serious complications after surgery
ratios were measured by NAD cycling assay. The untreated group
represents the S. aureus MTCC 96 and E. coli MTCC 739 cultures [63]. The inner and outer abiotic surfaces of several biome-
that did not receive treatment with Kocuran-capped silver glycona- dical devices such as stents, heart valves, vascular grafts and
noparticles. All the experiments were repeated three times and the catheters are frequently colonized with bacteria through
bars represent the means ± S.D. bacterial adhesion and biofilm formation resulting in these
infections [64], which is a widely growing problem in health
care system. It was earlier demonstrated that the coating of
AgNPs on polyurethane and plastic catheters exhibited anti-
bacterial and antiadhesive properties against pathogenic
strains of E. coli, Enterococci, P. aeruginosa and Staphylo-
coccus [65]. In the present study, the Kocuran-capped AgNPs
offered homogenous coating on the urethral catheters, which
effectively prevented the bacterial adhesion and biofilm for-
mation by S. aureus and E. coli. The results obtained in our
study clearly demonstrate the potential use of Kocuran-cap-
ped AgNPs on the surface coatings of the medical devices for
preventing bacterial infections.

Figure 6. Biofilm formation by S. aureus MTCC 96 and E. coli 3.10. In vitro biocompatibility studies of Kocuran-capped silver
MTCC 739 treated with Kocuran-capped silver glyconanoparticles. glyconanoparticles
The S. aureus MTCC 96 and E. coli MTCC 739 were cultured in
Muller–Hinton broth for 12 h at 37 °C and then treated with 0.1, 1, 5 In the biodirected synthetic approach, the polysaccharide-
and 10 μg ml−1 of Kocuran-capped silver glyconanoparticles for mediated synthesis and subsequent surface chemistry changes
24 h. The biofilm formation was demonstrated using crystal violet
assay. The untreated group represents the S. aureus MTCC 96 and of silver nanoparticles would increase the biocompatibility of
E. coli MTCC 739 cultures that did not receive treatment with silver nanoparticles. The results of MTT assay are shown in
Kocuran-capped silver glyconanoparticles. All the experiments were figure 13. The HGF cells incubated with Kocuran-capped
repeated three times and the bars represent the means ± S.D. silver glyconanoparticles maintained higher average cell

11
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 7. Confocal laser scanning microscope (CLSM) analysis of biofilm formation by S. aureus MTCC 96 cells treated with Kocuran-
capped silver glyconanoparticles. The CLSM images of biofilm formation in S. aureus MTCC 96 (a) without treatment (control); (b) treated
with Kocuran-capped silver glyconanoparticles, 0.1 μg ml−1; (c) treated with Kocuran-capped silver glyconanoparticles, 1 μg ml−1; (d) treated
with Kocuran-capped silver glyconanoparticles, 5 μg ml−1; and (e) treated with Kocuran-capped silver glyconanoparticles, 10 μg ml−1. The S.
aureus MTCC 96 cells were cultured in Muller–Hinton broth on Thermanox plastic cover slips at 37 °C for 24 h and then treated with 0.1, 1,
5 and 10 μg ml−1 of Kocuran-capped silver glyconanoparticles. The biofilm formation was observed by CLSM after staining with Live/Dead
staining kit. The untreated S. aureus MTCC 96 represents the group that did not receive any treatment. The scale bars are 10 μm.

viabilities as compared to the commercial AgNP up to a been thoroughly characterized using several spectroscopic
concentration of 100 μg ml−1. However, both types of Ag techniques. They were also assessed for their morphology,
nanoparticles significantly reduced the cell viability after 24 h stability, antibacterial and antibiofilm activities, induction of
of incubation at a concentration range of 100–200 μg ml−1. In intracellular ROS, depletion of NADH equivalents, and pro-
addition, the biocompatibility of the polysaccharide tein leakage. Kocuran-capped AgNPs were spherical with
(Kocuran) alone tested at concentrations ranging from narrow size distribution and were found to be stable and
1–200 μg ml−1 showed no significant difference in the cell showed no aggregation under various conditions of pH,
proliferation as compared to the untreated cells. In light of the temperature, and in solutions of NaCl, NaNO3 and BSA. The
above, the MTT cell proliferation results demonstrated good synthesized Kocuran-capped AgNPs exhibited pronounced
in vitro biocompatibility of the Kocuran-capped silver gly- in vitro antibacterial and antiadhesive activities against the S.
conanoparticles in human gingival fibroblasts as compared to aureus and E. coli strains in a concentration-dependent
the commercial AgNPs suggesting that they are biocompa-
manner. The bactericidal activity studies revealed that the
tible and non-toxic in nature and find suitable application in
formation of bactericidal ROS, in particular hydroxyl radicals
urethral catheters.
in both S. aureus and E. coli, which contributed to the bac-
tericidal action of Kocuran-capped AgNPs. These hydroxyl
radicals subsequently activated the downstream signaling
4. Conclusions pathways resulting in the depletion of reducing equivalents
and bacterial cell death. The enhanced antibacterial activity of
We have developed a simple, rapid, clean and green chem- Kocuran-capped AgNPs may be attributed to the surface
istry approach for the synthesis of biocompatible silver gly- functionalization with the EPS sugar residues, which facil-
conanoparticles using Kocuran EPS which acts as both a itates the distribution and penetration of glyconanoparticles
capping and reducing agent. The biosynthesized AgNPs have into the cell wall, and the structure-activity relationship. In

12
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 8. Confocal laser scanning microscope (CLSM) analysis of biofilm formation by E. coli MTCC 739 cells treated with Kocuran-capped
silver glyconanoparticles. The CLSM images of biofilm formation in E. coli MTCC 739 (a) without treatment (control); (b) treated with
Kocuran-capped silver glyconanoparticles, 0.1 μg ml−1; (c) treated with Kocuran-capped silver glyconanoparticles, 1 μg ml−1; (d) treated with
Kocuran-capped silver glyconanoparticles, 5 μg ml−1; and (e) treated with Kocuran-capped silver glyconanoparticles, 10 μg ml−1. The E. coli
MTCC 739 cells were cultured in Muller–Hinton broth on Thermanox plastic cover slips at 37 °C for 24 h and then treated with 0.1, 1, 5 and
10 μg ml−1 of Kocuran-capped silver nanoparticles. The biofilm formation was observed by CLSM after staining with Live/Dead staining kit.
The untreated E. coli MTCC 739 represents the group that did not receive any treatment. The scale bars are 10 μm.

addition, the in vitro biofilm assay and confocal microscopic


examination evidenced the efficacy of Kocuran-capped
AgNPs to inhibit the biofilm formation by S. aureus and
E. coli. The in vitro biocompatibility evaluation of the
Kocuran-capped silver glyconanoparticles on human gingival
fibroblasts demonstrated good cell proliferation as compared
to the commercial AgNPs suggesting that they are bio-
compatible and non-toxic in nature. Our data also suggests
that the Kocuran-capped AgNPs are hydrophilic, stable and
biodegradable nanomaterials which find application as coat-
ings on biomedical implants due to their intrinsic antibacterial
and antibiofilm properties and may help to prevent infections
and biofilm formation by drug-resistant pathogens as well as a
model system for studying various carbohydrate-carbohy-
drate, carbohydrate-protein and carbohydrate-cell
Figure 9. Kocuran-capped silver glyconanoparticles coating inhibits biointeractions.
biofilm formation on silicone urethral catheters. The organisms were
cultured overnight with sterile silicone urethral catheters coated with
(+) and without (−) Kocuran-capped silver glyconanoparticles. The Acknowledgments
biofilm formation by (a) S. aureus MTCC 96, (b) E. coli MTCC 739,
and (c) untreated catheter (control) after 24 h incubation was
demonstrated using crystal violet staining. The authors acknowledge the financial assistance provided to
Pombala Sujitha in the form of Senior Research Fellowship

13
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

Figure 10. Performance evaluation of Kocuran-capped silver glyconanoparticle-coating on silicone urethral catheters as a function of time.
The organisms were cultured overnight with sterile silicone urethral catheters coated with (+) and without (−) Kocuran-capped silver
glyconanoparticles. The biofilm formation by (a) S. aureus MTCC 96, and (b) E. coli MTCC 739 after 48, 72 and 96 h incubation was
demonstrated using crystal violet staining.

Figure 11. SEM images of Kocuran-capped silver glyconanoparticles coated urethral catheter. The surface was colonized with (a) S. aureus
MTCC 96 and (b) E. coli MTCC 739 after 48 h of incubation.

Figure 12. SEM images of uncoated catheter. The surface was colonized with (a) S. aureus MTCC 96 and (b) E. coli MTCC 739 after 48 h of
incubation.

14
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

[12] Rosi N L and Mirkin C A 2005 Nanostructures in


biodiagnostics Chem. Rev. 105 1547–62
[13] Chen X and Schluesener H J 2008 Nano-silver: a nanoproduct
in medical application Toxicol. Lett. 176 1–12
[14] Mandal D, Bolander M E, Mukhopadhyay D, Sarkar G and
Mukherjee P 2006 The use of microorganisms for the
formation of metal nanoparticles and their application Appl.
Microbiol. Biotechnol. 69 485–92
[15] Mukherjee P et al 2001 Fungus-mediated synthesis of silver
nanoparticles and their immobilization in the mycelial
matrix a novel biological approach to nanoparticle synthesis
Nano Lett. 1 515–9
[16] Kumar C G and Mamidyala S K 2011 Extracellular synthesis
of silver nanoparticles using culture supernatant of
Pseudomonas aeruginosa Colloids Surf. B 84 462–6
[17] Tran Q H, Nguyen V Q and Le A-T 2013 Silver nanoparticles:
synthesis, properties, toxicology, applications and
Figure 13. In vitro biocompatibility studies of Kocuran-capped silver perspectives Adv. Nat. Sci.: Nanosci. Nanotechnol. 4
glyconanoparticles on human gingival fibroblasts (HGF). The 033001
cultured HGFs were treated with Kocuran, Kocuran-capped silver [18] Zhang L, Shen Y H, Xie A J, Li S K, Jin B K and Zhang Q F
glyconanoparticles and commercially available citrate-synthesized 2006 One-step synthesis of monodisperse silver
AgNP at various concentrations (1–200 μg ml−1), incubated at 37 °C nanoparticles beneath vitamin E Langmuir monolayers
for 48 h and measured for cell proliferation (%) using MTT assay. J. Phys. Chem. B 110 6615–20
[19] Kumar V and Yadav S K 2009 Plant mediated synthesis of
silver and gold nanoparticles and their applications J. Chem.
by Council of Scientific and Industrial Research (CSIR), New Technol. Biotechnol. 84 151–7
Delhi, India. [20] Philip D 2010 Honey mediated green synthesis of silver
nanoparticles Spectrochim. Acta A 75 1078–81
[21] Kumar C G, Mamidyala S K, Reddy M N and Reddy B V S
2012 Silver glyconanoparticles functionalized with sugars of
sweet sorghum syrup as an antimicrobial agent Process
References Biochem. 47 1488–95
[22] Barrientos A G, De la Fuente J M, Rojas T C,
Fernández A and Penades S 2003 Gold glyconanoparticles:
[1] Costerton J W, Montanaro L and Arciola C R 2005 Biofilm in synthetic polyvalent ligands mimicking glycocalyx-like
implant infections: its production and regulation Int. J. Artif. surfaces as tools for glycobiological studies Chem. Eur. J.
Organs 28 1062–8 9 1909–21
[2] World Health Organization 2002 Prevention of hospital- [23] De la Fuente J M, Barrientos A G, Rojas T C, Rojo J, Canada J,
acquired infections A Practical Guide 2nd edn Available Fernandez A and Penades S 2001 Gold glyconanoparticles
from: www.who.int/csr/resources/publications/drugresist/ as water-soluble polyvalent models to study carbohydrate
whocdscsreph200212.pdf (Accessed 1 January 2014) interactions Angew. Chem. Int. Ed. 40 2257–61
[3] Tiwari P and Rohit M 2013 Assessment of costs associated [24] De Paz J L, Ojeda R, Barrientos A G, Penades S and
with hospital-acquired infections in a private tertiary care Martin-Lomas M 2005 Synthesis of a Lex
hospital in India Value Health Reg. Issues 2 87–91 neoglycoconjugate and Ley-functionalized gold
[4] Paniagua-Contreras G, Sainz-Espunes T, Monroy-Perez E, glyconanoparticles Tetrahedron: Asymmetry 16 149–58
Rodriguez-Moctezuma J R, Arenas-Aranda D, [25] Raveendran P, Fu J and Wallen S L 2003 Completely ‘green’
Negrete-Abascal E and Vaca S 2012 Virulence markers in synthesis and stabilization of metal nanoparticle J. Am.
Staphylococcus aureus strains isolated from hemodialysis Chem. Soc. 125 13940–1
catheters of mexican patients Adv. Microbiol. 2 476–87 [26] Raveendran P, Fu J and Wallen S L 2006 A simple and ‘green’
[5] Kumar R and Münstedt H 2005 Silver ion release from method for the synthesis of Au, Ag, and Au–Ag alloy
antimicrobial polyamide/silver composites Biomaterials 26 nanoparticles Green Chem. 8 34–8
2081–8 [27] Kumar C G, Mamidyala S K, Das B, Sridhar B,
[6] Hollinger M A 1996 Toxicological aspects of topical silver Sarala Devi G and Karuna M S L 2010 Synthesis of
pharmaceuticals Crit. Rev. Toxicol. 26 255–60 biosurfactant-based silver nanoparticles with purified
[7] Brady M J, Lisay C M, Yurkovetskiy A V and Sawan S P 2003 rhamnolipids isolated from Pseudomonas aeruginosa BS-
Persistent silver disinfectant for the environmental control of 161R J. Microbiol. Biotechnol. 20 1061–8
pathogenic bacteria Am. J. Infect. Control 31 208–14 [28] De la Fuente J M and Penades S 2005 Glyco-quantum dots: a
[8] Sondi I and Salopek-Sondi B 2004 Silver nanoparticles as new luminescent system with multivalent carbohydrate
antimicrobial agent: a case study on E. coli as a model for display Tetrahedron: Asymmetry 16 387–91
gram-negative bacteria J. Colloid Interface Sci. 275 177–82 [29] De la Fuente J M and Penades S 2006 Glyconanoparticles:
[9] Rai M, Yadav A and Gade A 2008 Current trends in types, synthesis and applications in glycoscience,
phytosynthesis of metal nanoparticles Crit. Rev. Biotechnol. biomedicine and material science Biochim. Biophys. Acta
28 277–84 1760 636–51
[10] Li X, Xu H, Chen Z-S and Chen G 2011 Biosynthesis of [30] Lemarchand C, Gref R and Couvreur P 2004 Polysaccharide-
nanoparticles by microorganisms and their applications decorated nanoparticles Eur. J. Pharm. Biopharm. 58
J. Nanomater. 2011 270974 327–41
[11] Sharma V K, Yngard R A and Lin Y 2009 Silver nanoparticles: [31] Liu Z, Jiao Y, Wang Y, Zhou C and Zhang Z 2008
green synthesis and their antimicrobial activities Adv. Polysaccharides-based nanoparticles as drug delivery
Colloid Interface Sci. 145 83–96 systems Adv. Drug Delivery Rev. 60 1650–62

15
Nanotechnology 25 (2014) 325101 C G Kumar and P Sujitha

[32] Kumar C G and Sujitha P 2013 Kocuran, an exopolysaccharide [49] Tran H V, Tran L D, Ba C T, Vu H D, Nguyen T N,
isolated from Kocuria rosea strain BS-1 and evaluation of its Pham D G and Nguyen P X 2010 Synthesis,
in vitro immunosuppression activities Enzyme Microb. characterization, antibacterial and antiproliferative activities
Technol. 55 113–20 of monodisperse chitosan-based silver nanoparticles
[33] Kumar C G, Mamidyala S K, Sujitha P, Muluka H and Colloids Surf. A 360 32–40
Akkenapally S 2012 Evaluation of critical nutritional [50] Kora A J, Beedu S R and Jayaraman A 2012 Size-controlled
parameters and their significance in the production of green synthesis of silver nanoparticles mediated by gum
rhamnolipid biosurfactants from Pseudomonas aeruginosa ghatti (Anogeissus latifolia) and its biological activity Org.
BS-161R Biotechnol. Progr. 28 1507–16 Med. Chem. Lett. 2 17
[34] Chaplin M F and Kennedy J F 1986 Carbohydrate Analysis: A [51] Leung T C Y, Wong C K and Xie Y 2010 Green synthesis of
Practical Approach (Washington, DC: IRL Press) silver nanoparticles using biopolymers, carboxymethylated
pp 129–36 —curdlan and fucoidan Mater. Chem. Phys. 121 402–5
[35] Bradford M M 1976 Rapid and sensitive method for the [52] Venkatpurwar V and Pokharkar V 2011 Green synthesis of
quantitation of microgram quantities of protein utilizing silver nanoparticles using marine polysaccharide: study of
the principle of protein-dye binding Anal. Biochem. 72 in-vitro antibacterial activity Mater. Lett. 65 999–1002
248–54 [53] Kanmani P and Lim S T 2013 Synthesis and structural
[36] Leonardo M R, Dailly Y and Clark D P 1996 Role of NAD in characterization of silver nanoparticles using bacterial
regulating the adhE gene of Escherichia coli J. Bacteriol. exopolysaccharide and its antimicrobial activity against food
178 6013–8 and multidrug resistant pathogens Process Biochem. 48
[37] Jackson D W, Suzuki K, Oakford L, Simecka J W, 1099–106
Hart M E and Romeo T 2002 Biofilm formation and [54] Jiang J, Oberdorster G and Biswas P 2009 Characterization of
dispersal under the influence of the global regulator CsrA of size, surface charge, and agglomeration state of nanoparticle
Escherichia coli J. Bacteriol. 184 290–301 dispersions for toxicological studies J. Nanopart. Res. 11
[38] Mosmann T 1983 Rapid colorimetric assay for cellular growth 77–89
and survival: application to proliferation and cytotoxicity [55] El Badawy A M, Luxton T P, Silva R G, Scheckel K G and
assays J. Immunol. Methods 65 55–63 Suidan M T 2010 Impact of environmental conditions (pH,
[39] Mulvaney P 1996 Surface plasmon spectroscopy of nanosized ionic strength, and electrolyte type) on the surface charge
metal nanoparticles Langmuir 12 788–800 and aggregation of silver nanoparticles suspensions Environ.
[40] Mock J J, Barbic M, Smith D R, Shultz D A and Shultz S 2002 Sci. Technol. 44 1260–6
Shape effect in plasmon resonance of individual colloidal [56] Marambio-Jones C and Hoek E M V 2010 A review of the
silver nanoparticles J. Chem. Phys. 116 6755–9 antibacterial effects of silver nanomaterials and potential
[41] Chung Y C, Chen I-H and Chen C-J 2008 The surface implications for human health and the environment
modification of silver nanoparticles by phosphoryl disulfides J. Nanopart. Res. 12 1531–51
for improved biocompatibility and intracellular uptake [57] Cho K H, Park J E, Osaka T and Park S G 2005 The study of
Biomaterials 29 1807–16 antimicrobial activity and preservative effects of nanosilver
[42] Ramirez M, Bashir S, Luo Z and Liu J L 2009 Green synthesis ingredient Electrochim. Acta 51 956–60
and characterization of polymer-stabilized silver [58] Imlay J A 2006 Iron-sulphur clusters and the problem with
nanoparticles Colloids. Surf. B 73 185–91 oxygen Mol. Microbiol. 59 1073–82
[43] Liu J C, Qin G, Raveendran P and Ikushima P 2006 Facile [59] Keyer K and Imlay J A 1996 Superoxide accelerates DNA
green synthesis, characterization, and catalytic function of β- damage by elevating free-iron levels Proc. Natl. Acad. Sci.
D-glucose-stabilized Au nanocrystals Chem. Eur. J. 12 USA 93 13635–40
2131–8 [60] Hantke K 2001 Iron and metal regulation in bacteria Curr.
[44] Vigneshwaran N, Nachane R P, Balasubramanya R H and Opin. Microbiol. 4 172–7
Varadarajan P V 2006 A novel one-pot ‘green’ synthesis of [61] Farr S B and Kogoma T 1991 Oxidative stress responses in
stable silver nanoparticles using soluble starch Carbohydr. Escherichia coli and Salmonella typhimurium Microbiol.
Res. 341 2012–8 Rev. 55 561–85
[45] Mohan Y M, Raju K M, Sambasivudu K, Singh S and [62] Rzhepishevska O, Hakobyan S, Ruhal R, Gautrot J,
Sreedhar B 2007 Preparation of acacia stabilized silver Barbero D and Ramstedt M 2013 The surface charge of anti-
nanoparticle: a green approach J. Appl. Polym. Sci. 106 bacterial coatings alters motility and biofilm architecture
375–81 Biomater. Sci. 1 589–602
[46] Omoike A and Chorover J 2004 Spectroscopic study of [63] Vincent J L, Bihari D and Sutar P M 1995 The prevalence of
extracellular polymeric substances from Bacillus subtilis: nosocomial infection in intensive care units in Europe: the
aqueous chemistry and adsorption effects results of the EPIC study J. Am. Med. Assoc. 274 639–44
Biomacromolecules 5 1219–30 [64] Costerton J W, Stewart P S and Greenberg E P 1999 Bacterial
[47] Selvakumar R, Aravind S, Anuradha M A and biofilms: a common cause of persistent infections Science
Balachandran Y L 2013 A facile synthesis of silver 284 1318–22
nanoparticle with SERS and antimicrobial activity using [65] Monteiro D R, Gorup L F, Takamiya A S, Ruvollo A C,
Bacillus subtilis exopolysaccharides J. Exp. Nanosci. de Camargo E R and Barbosa D B 2009 The growing
doi:10.1080/17458080.2013.778425 importance of materials that prevent microbial adhesion:
[48] Hunter R J 1981 Zeta Potential in Colloid Science, Principles antimicrobial effect of medical devices containing silver Int.
and Applications (London: Academic Press) pp 17–21 J. Antimicrob. Agents 34 103–10

16

You might also like