Anda di halaman 1dari 8

Colloids and Surfaces B: Biointerfaces 94 (2012) 143150

Contents lists available at SciVerse ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Studies on antibacterial activity of ZnO nanoparticles by ROS induced lipid


peroxidation
R.K. Dutta a, , Bhavani P. Nenavathu a , Mahesh K. Gangishetty a , A.V.R. Reddy b
a
Analytical Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
b
Analytical Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India

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

Article history: Recent studies indicated the role of ROS toward antibacterial activity. In our study we report ROS medi-
Received 6 September 2011 ated membrane lipid oxidation of Escherichia coli treated with ZnO nanoparticles (NPs) as supported
Received in revised form by detection and spectrophotometric measurement of malondialdehyde (MDA) by TBARS (thiobarbi-
16 December 2011
turic acid-reactive species) assay. The antibacterial effects of ZnO NPs were studied by measuring the
Accepted 22 January 2012
growth curve of E. coli, which showed concentration dependent bacteriostatic and bacteriocidal effects
Available online 7 February 2012
of ZnO NPs. The antibacterial effects were characterized by scanning electron microscopy (SEM) and
transmission electron microscopy (TEM). Further, antibacterial effect of ZnO NPs was found to decrease
Keywords:
ZnO nanoparticles
by introducing histidine to the culture medium treated with ZnO NPs. The ROS scavenging action of
Reactive oxygen species histidine was conrmed by treating histidine to the batch of Escherichia coli + ZnO NPs at the end of the
Lipid peroxidation lag phase of the growth curve (Set-I) and during inoculation (Set-II). A moderate bacteriostatic effect
Malondialdehyde (lag in the E. coli growth) was observed in Set-II batch while Set-I showed no bacteriostatic effect. From
Antioxidant these evidences we conrmed that the antibacterial effect of bare as well as TG capped ZnO NPs were
Electron microscopy due to membrane lipid peroxidation caused by the ROS generated during ZnO NPs interaction in culture
medium.
2012 Elsevier B.V. All rights reserved.

1. Introduction in the aqueous suspensions of ZnO NPs by EPR method and also
demonstrated enhanced antibacterial activity of nanocrystalline
Metals and metal oxide nanomaterials e.g., Ag, TiO2 , SiO2 , MgO ZnO attributed due to increased ROS-mediated cell injury [15].
and CaO are getting signicant scientic attention due to their vast In a recent study they have elucidated the role of ROS mediated
existing as well as potential newer applications, especially as bio- antifungal effect of ZnO NPs where ROS levels determined by EPR,
cidal or disinfecting agents [14]. In this regard, ZnO nanoparticles decreased in the presence of histidine [16].
(NPs) were found to be versatile and are considered to be poten- We have carried out a systematic study to elucidate the role
tial next generation materials as biocidal or disinfecting agent, of ROS toward antibacterial activity of ZnO NPs. In a biological
which is attributed to their higher stability than the organic based system, ROS is determined by TBARS method which is based on
disinfectants or anti-microbial agents. Lately, ZnO NPs were stud- formation of a pink colored complex between thiobarbituric acid
ied for their antibacterial [511] and anticancer properties [12]. and malondialdehyde (MDA) that can be quantitatively measured
Various mechanistic approaches were proposed to explain the spectrophotometric method at max = 532 nm [17]. The MDA is pro-
anti-bacterial action of ZnO NPs, including their tentative role in duced during the lipid peroxidation of unsaturated fatty acid by
generating reactive oxygen species (ROS), release of zinc ions, oxygen based free radicals and the measured intensity is related to
cellular membrane dysfunction and nanoparticle internalization. the concentration of generated ROS. The TBARS assay was chosen
Reported evidences have indicated that ROS might be the most to screen and monitor the mechanism of lipid peroxidation which
important cause for antibacterial effect [13]. In this regard, Sawai is a major indicator of oxidative stress [18,19]. Besides, it is rapid,
et al. [14] made a signicant contribution by demonstrating pro- easy to use, inexpensive and sensitive method. Further, this assay
duction of H2 O2 in the aqueous suspension of ZnO NPs. Further, is reported to be useful for measuring antioxidant activity of var-
Lipovsky et al. [8] detected hydroxyl radicals and singlet oxygen ious compounds [20,21]. We have determined MDA levels which
were the measures of ROS generated in the batches of culture media
treated with increasing concentrations of ZnO NPs (bare and thio-
Corresponding author. Tel.: +91 1332 285280; fax: +91 1332 286206. glycerol capped). It has been discussed that the ROS generated in
E-mail address: duttafcy@iitr.ernet.in (R.K. Dutta). the culture media might be due to oxidation of membrane lipids

0927-7765/$ see front matter 2012 Elsevier B.V. All rights reserved.
doi:10.1016/j.colsurfb.2012.01.046
144 R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150

of E. coli leading to their membrane dysfunction and consequently grid and dried at room temperature. The thermogravimetric analy-
leading to bacteriostatic or bactericidal conditions. In order to prove sis of TG capped ZnO nanomaterials was carried out with a heating
the role of ROS, we further demonstrated reduction of antibacterial rate of 5 C min1 from ambient temperature up to 1000 C under
activity of ZnO NPs in the presence of histidine via TBARS studies. a nitrogen ow (200 mL min1 ) using Perkin Elmer Pyris Diamond,
to ensure weight loss in the sample due to thermal degradation of
2. Experimental polymer.

2.1. Materials 2.4. Antibacterial activity

The wild type Escherichia coli (E. coli K-12 strain MTCC 1302) was The bare as well as TG capped ZnO NPs were assayed for
obtained from IMTECH, Chandigarh, India. Thioglycerol (TG), ZnCl2 , antibacterial activity using growth inhibition studies against gram-
methanol, NaOH, sodium dodecyl sulfate (SDS) and Luria-Bertani negative bacteria E. coli strain K-12. For antibacterial assay, the
(LB) broth media were procured from Himedia, India. Thiobarbitric bacterial colonies were rst grown on solid nutrient agar medium
acid, linolenic acid, histidine, 1,1,3,3 tetramethoxypropane (99% and from the primary cultured agar plates, fresh colonies were
pure) and commercial grade (bare) ZnO nanoparticles (<100 nm inoculated into 100 mL nutrient broth medium comprising of Luria-
in size, 99.99%), were procured from Sigma Aldrich, Germany. All Bertani (LB) media containing 5 g L1 of yeast extract, 10 g L1 of
chemicals and solvents were used without further purication. casein enzymic hydrolysate and 10 g L1 of NaCl in water. The E. coli
growth in the culture media was monitored by measuring its opti-
2.2. Synthesis of TG capped ZnO NPs cal density (O.D) at 600 nm. When the O.D reached to mid log phase
(i.e., O.D = 0.6 units), a 10 L of this culture was retrieved and was
The TG capped ZnO NPs were synthesized by wet-chemical inoculated in 100 mL of freshly prepared nutrient LB broth medium
method using NaOH and ZnCl2 as precursors in methanol and the supplemented with different concentrations of bare ZnO NPs of
pH 7.2 was maintained during the synthesis. 50 mL of 20 mM of concentration ranging between 8 and 45 mg/100 mL of media. Sim-
ZnCl2 solution was prepared in methanol, to which 8.5 L of TG was ilar process was followed for studying the batches treated with
added and stirred for 1 h so that the nal concentration of TG in the TG-capped ZnO NPs of concentration ranging between 8 and 55 mg
solution was 2 mM. About 0.4 g of NaOH pellets were added directly per 100 mL media. Positive controls consisting of NPs in LB medium
to the above mix and the entire solution was stirred for 4 h. The ZnO without inoculum and negative controls consisting of LB medium
NPs were collected after precipitating with n-hexane. The excess TG with only inoculum were used. All the asks were then incubated
was drained out and the NPs after washing with ethanol were kept at 37 C in an orbital shaker at 150 rpm. The bacterial growth was
for overnight drying at 60 C. The TG was used as a stabilizing agent monitored at an interval of 1 h by measuring the O.D of the culture
for controlling the size of the ZnO NPs by preventing agglomeration. media at  = 600 nm.
Powdered nanoparticles were obtained after washing and dry-
ing the precipitate. These nanoparticles were characterized after 2.5. Determination of reactive oxygen species (ROS)
re-dispersing in methanol. The purpose of studying antibacterial
effects of TG capped ZnO NPs was to evaluate whether the capping The ROS generated in the culture media was determined using
agent has any role on antibacterial effect of ZnO NPs. In addition, TBARS assay. An aliquot of 1 mL of culture media was collected
applying capping agent usually results in smaller sized particles to which 200 L of 10% SDS was added and swirled vigorously.
so it would be possible to assess the size dependent antibacterial 2 mL of freshly prepared thiobarbituric acid (TBA) was added to
effect as well. In this regard, it should be mentioned that the con- the above mix and was incubated at 95 C for 60 min. It was
centration of TG used in the synthesis of TG capped ZnO NPs did cooled to room temperature and was centrifuged at 3500 rpm for
not show antibacterial activity as the concentration of TG used was 15 min. The absorption spectrum of the supernatant was recorded
much less than the reported MIC [22]. at max = 532 nm to estimate the formation of TBARS. All anal-
yses were carried out in triplicate. From the TBARS intensities
2.3. Characterization the corresponding levels of MDA were deduced from a calibra-
tion curve plotted between the intensity of TBARS measured at
The X-ray diffraction measurements of commercial ZnO NPs  = 532 nm against different concentrations of MDA synthesized
and TG capped ZnO NPs were performed with powder diffrac- by acidic hydrolysis of 1,1,3,3 tetramethoxypropane (Supporting
tometer (Bruker ARS D8 Advance) operated at 40 kV using graphite information, Fig. S1).
monochromatized Cu K radiation source with a wavelength of
1.54 A in a wide-angle region from 20 to 80 on a 2 scale. The opti- 2.6. Study of antibacterial effect of ZnO NPs treated E. coli in the
cal properties were measured by UVvisible spectrophotometer presence of antioxidants
(Shimadzu, UV-1800) in the range of 200500 nm. The particle size
distribution and morphology of bare (commercial) ZnO nanoparti- Two sets of experiments were carried out to assess the effect
cles and TG capped ZnO NPs were studied using FESEM EDAX (eld of an antioxidant, e.g., histidine as a scavenger of ROS and con-
emission scanning electron microscope coupled to energy disper- sequently reduction of antibacterial activities of ZnO NPs. The
sive X-ray analyzer), FEI-Quanta 200F operated at 20 kV. The sample growth patterns of E. coli treated with commercial ZnO NPs of
preparation for SEM measurement was done by spraying the dis- 35 mg/100 mL media) and TG capped ZnO NPs (55 mg/100 mL
persion of ZnO nanomaterials on a clean glass plate, dried at room broth) were studied in the presence of histidine. The reason for
temperature and coated with a thin layer of Au for electrical con- choosing these concentrations of bare ZnO NPs and TG capped ones
ductivity for incident electrons. The interaction of nanoparticles of was both conditions exhibited similar bacteriostatic growth curves
ZnO with E. coli was imaged by transmission electron microscopy hence expected to generate similar levels of ROS.
(TEM) using FEI Technai-G2 microscope operated at 200 kV. The Set-I: the methodology for cell culture was the same as dis-
samples for TEM analysis were collected at the beginning of station- cussed above i.e., 10 L of K-12 E. coli strain was inoculated in
ary phase and prepared by placing a drop of the culture medium freshly prepared nutrient broth, to which histidine of concentration
containing E. coli treated with ZnO nanoparticles (after suitable ranging between 15 mg and 45 mg/100 mL of culture media was
dilution with buffer solution) on a carbon coated 150 mesh copper introduced after 3 h from the addition of inoculum, corresponding
R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150 145

to the end of the lag phase of the growth curve. The delayed addition more for higher concentrations of ZnO NPs. Signicant delay (2 h)
of histidine was to ensure the ROS generation. in the onset of the growth was observed for the batch treated with
Set-II: histidine concentration of 45 mg/100 mL was added at 35 mg of bare ZnO NPs/100 mL culture media and its corresponding
the time of addition of inoculum E. coli + ZnO NPs (35 mg/100 mL inhibition of growth was estimated to be 80% with respect to the
media) to conrm the role of ROS in the antibacterial action of ZnO control batch. Complete inhibition of the bacterial growth, i.e., bac-
NPs. Respective growth curves and the MDA levels were measured teriocidal condition was observed for the batch treated with 45 mg
for both the sets of experiments. of bare ZnO NPs/100 mL media, corresponding to minimum bac-
tericidal concentration. It may be surmised that the antibacterial
effect of ZnO NPs were dependent on the concentration ZnO NPs
3. Results and discussion
treated in the culture media and was in good agreement with the
previous reports [411].
The bare ZnO NPs (commercially procured) showed X-ray
Similarly, the growth patterns of the K-12 strain of E. coli treated
diffraction peaks at (1 0 0), (1 0 1), (1 0 2), (1 1 0), (1 0 3) and (1 1 2)
with the TG capped ZnO NPs of concentrations ranging between
planes (Supporting information, Fig. S2) corresponded to hexagonal
8 mg and 55 mg in 100 mL of LB media were assayed (Fig. 1b). The
wurtzite phase of ZnO, JCPDS card No: 5-0664. The crystallite size
batches treated with 8 mg and 15 mg of the TG capped ZnO NPs
(D) was calculated from Debye Scherrer formula D = 0.9/ cos();
were similar to that of the control while the minimum inhibitory
where  is the wavelength of X-ray source, is full-width at half
concentration for the capped NPs was found to be 25 mg/100 mL
maximum (FWHM) in radians and  is Braggs diffraction angle.
culture media. The TG capped ZnO NPs also showed bacteriostatic
The morphology and size distribution of the bare ZnO NPs were
condition at higher concentrations of treated TG capped ZnO NPs.
also conrmed from FESEM studies where the energy dispersive
Unlike bare ZnO NPs, the bacteriocidal condition for TG capped ZnO
X-ray analysis (EDAX) of these NPs exhibited characteristic X-ray
NPs was not observed even at a concentration of 55 mg/100 mL
peaks of Zn and O (Fig. S3a). The size of these particles appeared to
culture media. The batch treated with 55 mg of TG capped ZnO
be less than 100 nm and was in good agreement with the reported
NPs/100 mL media exhibited 80% of inhibition of the growth with
average size by Sigma Aldrich.
respect to the control batch. Interestingly the inhibition pattern
The thioglycerol (TG) capped ZnO NPs were successfully syn-
was similar to the batch of 35 mg of bare ZnO NPs/100 mL.
thesized as evidenced from its XRD pattern, which matched with
From these studies, it was evident that the bare ZnO NPs showed
those of bare ZnO NPs (Supporting information, Fig. S2). The peaks
higher antibacterial activity than the TG capped ones. Notably, the
were broad and the crystallite size (D) of these TG capped nanopar-
bare ZnO NPs were several folds bigger in size than the TG capped
ticles was measured to be 4 nm. The broad diffraction peaks
ZnO NPs. Therefore our studies contradicted to the earlier reports
observed for TG capped ZnO NPs could be due to various reasons,
that the smaller sized ZnO NPs exhibit more antibacterial effect
namely, small particle sizes together with strain due to defects in
[11]. It is therefore interesting to study the mechanism underly-
the nanomaterials. Zak et al. [23] used modeled study to explain
ing the antibacterial effect due to both capped as well as bare ZnO
the broadening of XRD peak of ZnO NPs. The scanning electron
NPs. In this regard, several mechanisms have been proposed by
microscopy study of the TG capped ZnO nanoparticles showed sig-
different researchers, which can be broadly classied into three
natures of agglomeration of spherical shaped nanoparticles and
categories [13]: (a) physical processes, comprising of adsorption
their EDAX analysis exhibited the characteristic K and L X-rays of
of ZnO NPs on the surface of the E. coli cells; (b) biological pro-
zinc, K X-ray of oxygen (Supporting information, Fig. S3b).
cesses leading to internalization of NPs in the E. coli, via channels
The TG capped ZnO NPs showed a blue shift with excitonic peak
or proteins at the cell wall and (c) chemical phenomena result-
measured at 310 nm (Supporting information, Fig. S4a) as com-
ing in production of reactive species. Further, antibacterial effect
pared to the UVvisible absorption band measured at 370 nm for
of ZnO NPs due to combination of these phenomena could not
commercial ZnO NPs (Fig. S4b). The corresponding band gap for
however be ruled out. In order to understand the antibacterial
TG capped ZnO NPs was calculated to be 4.0 eV, which is typically
effect, it is important to realize the structure of an E. coli (gram
due to particles of sizes less than 5 nm of ZnO NPs. The absorp-
negative bacteria). It comprises of cell wall with three layered mem-
tion peak was quite sharp, indicating monodispersed nature of the
brane structure as shown in a schematic diagram (Fig. 2). The outer
ZnO NPs. The TG capping on ZnO NPs were further conrmed from
layer consists of membrane proteins, porins, phospholipids and
thermogravimetric analysis which showed 23% mass loss due to
lipopolysaccharides. The middle layer consists of peptidoglycan.
degradation of thioglycerol capping and also removal of absorbed
The inner cytoplasmic membrane consists of 40% phospholipids
water (Supporting information, Fig. S5) and was in good agreement
and 60% protein. The gram negative cell wall of E. coli is due to nega-
with the reported literature [24]. From thermal analysis, it was
tively charged lipopolysaccharides which might allow the ZnO NPs
deduced that the thioglycerol concentration in the TG-capped NPs
to interact electrostatically [25]. A major functional consequence of
was 0.46 mM, which was much lower than the reported minimum
interaction of these ZnO NPs could lead to breakdown of the mem-
inhibitory concentration (2 mM) of TG for E. coli growth.
brane barrier of E. coli, which could accompany dissipation of ion
gradients and might eventually lead to cell swelling and lysis [26].
3.1. Evaluation of antibacterial effect of ZnO NPs In addition, ZnO NPs are reported to be internalized via the usual
transport channels and the internalized NPs might interact with the
The growth patterns of the K-12 strain of E. coli treated with cellular constituents to cause antibacterial effect. Brayner et al. [5]
the bare (commercial) ZnO NPs of concentrations ranging between Huang et al. [7] and Nair et al. [12] have demonstrated using TEM
8 mg and 45 mg in 100 mL of LB media were assayed (Fig. 1a). The data that ZnO NPs of sizes 11 nm and 60 nm were internalized in
growth curve of the batch treated with 8 mg of bare ZnO NPs was E. coli and corroborated these results with alteration of morphology
similar to that of the control batch. The minimum inhibitory con- and damage of bacterial cell. However, the mode of internaliza-
centration (MIC) for bare ZnO NPs was found to be 15 mg/100 mL tion of such large particles were non-trivial as for gram-negative
of culture media. The inhibition of bacterial growth was found to prokaryotic cells, e.g., E. coli, the average maximum size range for
increase with the concentrations of ZnO NPs treated in culture non-specic uptake of large molecules is of the order of 1.82 nm
media. The antibacterial effect of these bare ZnO NPs was also evi- via an OmpF porins [27,28]. Our TEM studies showed adherence
dent from the delay in the onset of the logarithmic growth phase, of bare ZnO NPs (Fig. 3a) and TG capped ZnO NPs (Fig. 3b) on the
referred to as bacteriostatic condition. The delay was found to be surface of E. coli. The adherence of NPs on the surface of the E. coli
146 R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150

(a) Control
8mg of ZnO NPs per 100mL CM
(b)
Control
15mg of ZnO NPs per 100mL CM 15mg of TG-ZnO NPs per 100mL CM
25mg of ZnO NPs per 100mL CM 25mg of TG-ZnO NPs per 100mL CM
30mg of ZnO NPs per 100mL CM
45mg of TG-ZnO NPs per 100mL CM
35mg of ZnO NPs per 100mL CM
55mg of TG-ZnO NPs per 100mL CM
45mg of ZnO NPs per 100mL CM
2.0
2.0
1.6
1.6

O.D 600 nm
O.D600 nm

1.2 1.2

0.8 0.8

0.4 0.4

0.0
0.0
0 2 4 6 8 10 12 0 4 8 12
Time (h) Time (h)

Fig. 1. Growth patterns of E. coli treated with different concentrations of (a) bare ZnO NPs; (b) TG capped ZnO NPs (CM, culture media). The results correspond to mean SD
(SD, standard deviation; n = 3).

might lead to physical blockages of the transport channels of the cell


membranes, due to which transportation of cellular nutrients from HO2 + e + H+ H2 O2
media were restricted, resulting into cell starvation and eventually
cell mortality. Deformation of cellular morphology, cell swelling The hydroxyl radicals generated in suspensions of ZnO NPs in
and membrane rupture were observed in our SEM study (Fig. 3c) culture media could extract the hydrogen atom from allylic position
showing ZnO particles on the surface of the cell wall by EDAX anal- of the unsaturated fatty acid. The allyl free radical that is formed
ysis. Further, TEM study of ZnO NPs treated with E. coli also revealed might react with the oxygen molecule to form lipid peroxide rad-
deformation of cellular morphology, cell swelling and membrane ical (LOO ), which could undergo rearrangement to form MDA, as
rupture (Fig. 3d). It may be remarked here that the damages of cell schematically shown in (Supporting information, Fig. S6). The MDA
membrane might have occurred due to oxidation of lipid mem- binds with thiobarbitiric acid to form a complex referred to as
brane in the cell wall which resulted in porous cell membrane and TBARS which corresponds to a characteristic UVvisible absorption
facilitated internalization of ZnO NPs in cells. peak at max = 532 nm and hence can be determined by spectropho-
The oxidation of membrane lipids could be triggered by reactive tometric method. The intensity of the absorption at max = 532 nm
oxygen species like H2 O2 [4] or highly reactive hydroxyl radical is directly related to the amount of MDA formed, which is equiv-
and singlet oxygen [15], which were reported to be generated in alent of ROS generated during the interaction of ZnO NPs in the
ZnO nanoparticles suspended in water. The mechanism for the pro- culture media.
duction of ROS (H2 O2 and OH ) was attributed to light induced Compared to the control batch (i.e., without ZnO NPs in culture
effect. Notably, ordinary room light with a total light intensity of media), the MDA equivalents for the bare as well as TG capped
10 W/cm1 was sufcient for generating ROS [15]. The mecha- ZnO NPs treated batches were higher (Fig. 4a) and increased lin-
nism of light induced generation of ROS could be given as follows early (R2 = 0.94) with increasing concentrations of bare ZnO NPs
[29], and TG capped ZnO NPs (Fig. 4b). This indicated increase in the ROS
generation due to higher concentrations of ZnO NPs in the culture
ZnO + h ZnO + e + h+ ; h+ + H2 O OH + H+
media. It was noted that the MDA equivalent (ROS equivalence)
was about 36% higher in the batch treated with 45 mg of bare ZnO
e + O2 O2 NPs in 100 mL of culture media (which corresponded to 100% inhi-
bition of E. coli growth, i.e., bactericidal) as compared to the control
batch. The batches treated with 35 mg, 25 mg and 15 mg of bare
O2 + H+ HO2 ZnO NPs per 100 mL media, which showed bacteriostatic condition,

Porins Lipopolysachcharides

Membrane Phospholipid
Protein

Outer membrane
ZnONPs C (8nm)

Peptidoglycan

Cytoplasmic
Cytoplasm membrane

Fig. 2. Schematic structure of E. coli showing potential interaction with ZnO NPs.
R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150 147

Fig. 3. (a) TEM images showing adherence of bare ZnO NPs on the surface of E. coli, (b) TEM images of TG capped ZnO NPs treated E. coli, (c) SEM image showing deformed
E. coli engulfed by bare ZnO NPs, supported by EDAX spectrum showing Zn L-X-ray, and (d) TEM image of E. coli showing cell swelling and cell wall deformation due to
interaction of ZnO NPs.

corresponded to generation of about 23.6%, 16% and 5.5% of MDA formation as evident from the increasing order of equivalent MDA
equivalent respectively. Similarly, with respect to the control batch, in the culture medium. Our results were in good agreement with
the relative amounts of MDA equivalent in the batches treated with a recent report on toxicity of ZnO nanoparticles on gram positive
25 mg, 45 mg and 55 mg of TG capped ZnO NPs were 6.9%, 13.3% bacteria and cancer cells by apoptosis by lipid peroxidation [30].
and 24%, respectively. The measured MDA equivalent, which was
equivalent to the generation of ROS, correlated with the growth 3.2. Histidine inhibits the antibacterial effect caused by ZnO NPs
patterns of bare and TG capped ZnO NPs treated E. coli. From these
observation it may be remarked that the inhibition of the growth In order to conrm the ROS mediated antibacterial effect of
curve due to the increase in the ZnO NPs were attributable to ROS ZnO NPs the growth patterns of E. coli treated with ZnO NPs in
148 R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150

(a) Bare ZnO NPs (b) Bare ZnO NPs


40 40

MDA equivalents (nmol/mL)


TG-ZnO NPs
TG-ZnO NPs

MDA (nmol/mL)
35
35

30

30
25

25
0 15 25 35 45 -- 0 10 20 30 40 50
Concentration of ZnO NPs Concentration of ZnO NPs
(mg/100mL) (mg/100mL)

Fig. 4. Showing increase in the MDA equivalents (nmol/mL) with increase in the concentrations of (a) bare ZnO NPs/100 mL of culture media and TG capped ZnO NPs/100 mL
culture media and (b) linear correlation between MDA formed and concentrations of bare and TG capped ZnO NPs in the culture media. The results correspond to mean SD
(SD, standard deviation; n = 3).

Control
45mg Histidine
(a) 25mg Histidine (b)
15mg Histidine
MDA equivalents (nmol/mL)
50
2.0 Without Histidine
1.6 45
bare ZnO NPs
1.2 40
O.D600nm

0.8 35

0.4
30
0.0
0 2 4 6 8 10 12 0 15 25 45 -- --
Time (h) Concentration of Histidine
(mg/100mL)

Control
45mg Histidine
(c) 35mg Histidine (d)
15mg Histidine 55
MDA equivalents (nmol/mL)

2.0
Without Histidine
50
1.5 TG capped ZnO NPs
45
O.D600 nm

1.0 40

35
0.5
30
0.0
0 4 8 12 0 15 25 45 -- --
Time (h) Concentration of Histidine
(mg/100mL)

Fig. 5. Growth curve of batches of E. coli treated with 35 mg of bare ZnO NPs/100 mL media and supplemented with different concentrations of histidine added at the end of
the lag phase of the growth prole, (b) showing decrease in MDA equivalents (nmol/mL) with the increase in histidine concentrations, (c) growth curve of batches of E. coli
treated with 55 mg TG capped ZnO NPs/100 mL media and supplemented with different concentrations of histidine at the end of lag phase of the growth prole, and (d)
showing decrease in MDA equivalents (nmol/mL) with the increase in histidine concentrations. The values for each measurement correspond to mean SD (SD, standard
deviation, n = 3).
R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150 149

the presence of antioxidant were studied. It was thought that the Control
ROS generated due to the interaction ZnO NPs with E. coli could 45mg Histidine (Set II)
be scavenged by the antioxidants. In this regard, we studied the
2.0 45mg Histidine (Set I)
effect of histidine, which is a well known scavenger of hydroxyl
radicals and singlet oxygen [15], toward inhibition of antibacterial
activity of ZnO nanoparticles. Firstly, it was important to conrm 1.6
that histidine did not exhibit antibacterial activity. In order to ver-
ify this, control experiments were carried out where the culture
1.2

O.D600nm
media without ZnO NPs, were treated with 45 mg/100 mL of histi-
dine and compared with the batch without hisitidine (as given in
Fig. S7). It was observed that the growth curve of E. coli for both the 0.8
cases were similar, which conrmed that histidine does not exhibit
antibacterial activity. 0.4
Furthermore, the inhibition of the bacterial growths which were
treated with 35 mg/100 mL ZnO NPs, were reduced as the histidine
concentration was increased from 15 mg/100 mL to 45 mg/100 mL 0.0
of culture medium (Fig. 5a). This was substantiated by decrease
0 2 4 6 8 10 12
in the MDA equivalents with the increase in the histidine con-
centration (Fig. 5b). Similar activity of histidine toward inhibition Time (h)
of antibacterial effect of TG capped ZnO NPs and correspond- Fig. 6. Growth curve of the batch of E. coli treated with 35 mg of bare ZnO
ing decrease in ROS formation was also observed (Fig. 5c and d, NPs/100 mL culture media and supplemented with histidine at the end of the lag
respectively). The decrease in MDA equivalents due to addition of phase, i.e., after 3 h (Set-I) and at the time of addition of inoculum (Set-II). The values
increasing concentration of histidine, indicated scavenging of ROS for each measurement correspond to mean SD (SD, standard deviation, n = 3).
generated during interaction of ZnO NPs in culture media.
To further ascertain the effect of histidine toward scavenging
culture media, and bacteriocidal (no growth of E. coli) condition
ROS generated during interaction of ZnO NPs in culture media,
for the batch treated with 45 mg/100 mL of culture media. While
two sets of experiments were carried out on batches treated with
for TG capped ZnO NPs, bacteriostatic condition prevailed up to
35 mg/100 mL media where 45 mg of histidine was added to the
NP concentration of 55 mg/100 mL which corresponded to about
culture media after 3 h of inoculum corresponding to the end of
80% inhibition of growth and was similar to the batch treated with
the lag phase of the E. coli growth curve (Set-I) and at the time
35 mg/100 mL of bare ZnO NPs. The lesser antibacterial effect of TG
of inoculation (Set-II). The delay in supplementing histidine was
capped NPs was attributed to the capping agent. Further, the role
done to facilitate the production of ROS in the culture media so that
of ROS toward their antibacterial effect of ZnO NPs was conrmed
the generated ROS can be scavenged by the supplemented antiox-
by observing a reduction in the inhibition of bacterial growth due
idants. In the Set-I, the delay in the onset of logarithmic phase of
to external supplementation of histidine in the culture media. The
the growth curve was diminutive. The inhibition of the growth was
corresponding MDA levels were also reduced. This was attributed to
only 10% of the control batch as compared to the 80% inhibition
ROS scavenging action of histidine. These results strongly suggested
observed in the case where histidine was not supplemented (as
that the antibacterial effect of ZnO NPs were due to membrane lipid
shown in Fig. 5a and c). Such signicant reduction of antibacterial
oxidation caused by ROS generated during ZnO NPs interaction with
effect of ZnO NPs could be attributed to ROS scavenging action of
E. coli. The outcome the membrane lipid oxidation perhaps led to
histidine. On the other hand, the batch treated with 35 mg/100 mL
cell wall dysfunctioning and rupture.
of ZnO NPs and histdine together (Set-II), exhibited similar growth
curve to that of the control samples (Fig. 6). Though at such con-
centration of ZnO NPs, the ROS formation was expected, but the Acknowledgments
presence of histidine was sufcient to scavenge the generated ROS.
It was noted from Fig. 5b and d, that histidine was not capable to This work has been carried out under the BRNS project
scavenge all the generated ROS in the culture media. This could be 2007/37/47/BRNS and authors thank BRNS for the support. Mr.
attributed due to its scavenging action of only hydroxyl radical and Mahesh K Gangishetty is thankful to BRNS, India for awarding fel-
singlet oxygen [15], while hydrogen peroxide which is a byproduct lowship. Mr. N. Bhavani Prasad is thankful to MHRD, Govt of India
of cellular metabolism might not be scavenged by histidine. Overall, for awarding senior research fellowship. We would like to thank
accumulated evidences strongly supported the role of ROS toward Mr. Khushwant Singh, one of our research group members and Dr.
antibacterial activity which was most likely due to membrane lipid R. Acharya, Bhabha Atomic Research Centre, Mumbai for their keen
peroxidation. interest in this work. We thank Institute Instrumentation Centre of
IIT Roorkee for allowing us to use the instrumental facilities.

4. Conclusions Appendix A. Supplementary data

Production of reactive oxygen species (ROS) during interaction Supplementary data associated with this article can be found in
of ZnO NPs with E. coli was conrmed to be the key phenomenon the online version, at doi:10.1016/j.colsurfb.2012.01.046.
for antibacterial effect of ZnO NPs. The generated ROS in the cul-
ture media was capable to cause oxidation of lipid membrane in
References
the cell wall of E. coli, as evident from the determination of mal-
ondialdehyde, which is a by-product of lipid peroxidation reaction [1] J.J. Antony, P. Sivalingam, D. Siva, S. Kamalakkannan, K. Anbarasu, R. Sukirtha,
of unsaturated fatty acid. The malondialdehyde levels were higher M. Krishnan, S. Achiraman, Colloids Surf. B 88 (2011) 134140.
for the batches treated with higher concentrations of ZnO NPs, and [2] L. Shtykova, C. Fant, P. Handa, A. Larsson, K. Berntsson, H. Blanck, R. Simonsson,
M. Nydn, H.I. Hrelind, Prog. Org. Coat. 64 (2009) 2026.
it was correlated with bacteriostatic conditions for the batches [3] S. Makhluf, R. Dror, Y. Nitzan, Y. Abramovich, R. Jelinek, A. Gedanken, Adv. Funct.
treated with ZnO NPs of concentration up to 35 mg/100 mL of Mater. 15 (2005) 17081715.
150 R.K. Dutta et al. / Colloids and Surfaces B: Biointerfaces 94 (2012) 143150

[4] O. Yamamoto, M. Komatsu, J. Sawai, Z. Nakagawa, J. Mater. Sci. Mater. Med. 15 [16] A. Lipovsky, Y. Nitzan, A. Gedanken, R. Lubart, Nanotechnology 22 (2011)
(2004) 847851. 105101105107.
[5] R. Brayner, R. FerrariIliou, N. Brivois, S. Djediat, M.F. Benedetti, Fievet, Nano [17] L.T. Rael, G.W. Thomas, M.L. Craun, C.G. Curtis, R. Bar-Or, D. Bar-Or., J. Biochem.
Lett. 6 (2006) 866870. Mol. Biol. 37 (2004) 749752.
[6] K.M. Reddy, K. Feris, J. Bell, D.G. Wingett, C. Hanley, A. Punnoose, Appl. Phys. [18] K. Yagi, Free Radical Antioxid. Protoc. 108 (1998) 101106.
Lett. 90 (2007) 21390212139023. [19] G. Lefevre, Ann. Biol. Clin. 56 (1998) 305319.
[7] Z. Huang, X. Zheng, D. Yan, G. Yin, X. Liao, Y. Kang, Y. Yao, D. Huang, B. Hao, [20] C. Villa, Gac. Med. Mex. 136 (2000) 249256.
Langmuir 24 (2008) 41404144. [21] A. Hunnisett, Biol. Trace Elem. Res. 47 (1995) 125132.
[8] A. Lipovsky, Z. Tzitrinovich, H. Friedmann, G. Applerot, A. Gedanken, R. Lubart, [22] K.K. Jensen, G.T. Javor, Chemotherapy 24 (1981) 860867.
J. Phys. Chem. C 113 (2009) 1599716001. [23] A.K. Zak, W.H.A. Majid, M.E. Abrishami, R. Youse, Solid State Sci. 13 (2011)
[9] R.K. Dutta, P.K. Sharma, R. Bhargava, N. Kumar, A.C. Pandey, J. Phys. Chem. B 251256.
114 (2010) 55945599. [24] A.K. Singh, V. Vishwanth, V.C. Janu, J. Lumin. 129 (2008) 874878.
[10] R. Wahab, A. Mishra, S.I. Yun, Y.S. Kim, H.S. Shin, Appl. Microbiol. Biotechnol. [25] K. Feris, C. Otto, J. Tinker, D. Wingett, A. Punnoose, A. Thurber, M.
87 (2010) 19171925. Kongara, M. Sabetian, B. Quinn, C. Hanna, D. Pink, Langmuir 26 (2010)
[11] K.R. Raghupathi, R.T. Koodali, A.C. Manna, Langmuir 27 (2011) 40204028. 44294436.
[12] S.K. Nair, A. Sasidharan, V.V. Divya Rani, D. Menon, S. Nair, K. Manzoor, S. Raina, [26] R.W. Titball, A.K. Basak, in: J.E. Alouf, M.R. Popoff (Eds.), A Comprehensive
J. Mater. Sci. Mater. Med. 20 (2009) 235241. Sourcebook of Bacterial Protein Toxins, 3rd Ed., Academic Press, London, 2006,
[13] L. Zhang, Y. Jiang, Y. Ding, N. Daskalakis, L. Jeuken, M. Povey, A.J. ONeill, D.W. pp. 516534.
York, J. Nanopart. Res. 12 (2009) 16251636. [27] R. Koebnik, K.P. Locher, P.V. Gelder, Mol. Microbiol. 37 (2000) 239259.
[14] J. Sawai, E. Kawada, F. Kanou, H. Igarashi, A. Hashimoto, T. Kokugan, M. Shimizu, [28] G.E. Schulz, Curr. Opin. Struct. Biol. 10 (2000) 443447.
J. Chem. Eng. 29 (1996) 627633. [29] N. Padmavathy, R. Vijayaraghavan, Sci. Technol. Adv. Mater. 9 (2008) 37.
[15] G. Applerot, A. Lipovsky, R. Dror, N. Perkas, Y. Nitzan, R. Lubart, A. Gedanken, [30] M. Premanathan, K. Karthikeyan, K. Jeyasubramanian, G. Manivannan,
Adv. Funct. Mater. 19 (2009) 842852. Nanomedicine 7 (2011) 184192.

Anda mungkin juga menyukai