Sie sind auf Seite 1von 8

Article

Journal of
Biomedical
Nanotechnology
Copyright © 2013 American Scientific Publishers
All rights reserved Vol. 9, 1–8, 2013
Printed in the United States of America www.aspbs.com/jbn

Antimicrobial Activity of CaO Nanoparticles


Arup Roy1 , Samiran S. Gauri2 , Madhusmita Bhattacharya3 , and Jayanta Bhattacharya1 ∗
1
Department of Mining Engineering, Indian Institute of Technology, Kharagpur 721302, India
2
Department of Biotechnology, Indian Institute of Technology, Kharagpur 721302, India
3
Gitam Institute of Science, Gitam University, Visakhapatnam Campus, Visakhapatnam 530045, India

The high degree of microbial diseases and their multidrug resistant properties make the researchers to develop new
class of antimicrobial agents. A modern and innovative approach of drug development is the use of metallic nanoparti-
cles as new formulations of antimicrobial agents. In this study, microwave irradiated CaO nanoparticles (CaO-NPs) were
used to determine their antimicrobial efficacy against gram negative and gram positive bacteria, as well as pathogenic
yeast. The physiochemical properties of CaO-NPs were characterized by means of X-ray diffraction (XRD) and high res-
olution transmission electron microscopy (HRTEM). The nanoparticles consist of well dispersed agglomerates of grains
with a narrow size distribution of 14–24 nm. The prepared CaO-NPs showed much higher antimicrobial activity against
P seudomonas aeruginosa (ATCC 27853) and Staphylococcus epidermidis (MTCC 435) in comparision to Candida trop-
icalis (NCIM 3110). The minimum inhibitory concentration (MIC) value of CaO-NPs was found within the range of 2–8
mM for all the above tested strains. This bioactive nanoparticle also inhibits the biofilm formation and may have future
applications cheap and non toxic as antimicrobial drug for skin care product development.
KEYWORDS: CaO Nanoparticles, Antimicrobial Activity, Minimum Inhibitory Concentration (MIC), Multidrug Resistant, Biofilm.

INTRODUCTION Over a past few decades, nanoparticles have been exten-


Pseudomonas aeruginosa are the most common Gram- sively applied in various fields of public interest. Nanopar-
negative opportunistic pathogen of humans1 and it causes ticles possess unique physicochemical characteristics, such
ulcer like keratitis, lung infections, cystic fibrosis, noso- as a high ratio of surface area to mass, high reactiv-
comial infection and a wide range of severe and some- ity, and sizes in the range of nanometers (10−9 m).12
times fatal diseases in immunocompromised individuals.2 Nanoparticles have also been successfully used to enhance
Whereas Staphylococcus epidermidis, one of the most the immobilization and activity of catalysts,13 in medical
prevalent cutaneous resident bacteria and resistant to and pharmaceutical nanoengineering for delivery of ther-
many antibiotics and a common opportunistic pathogen of apeutic agents,14 in chronic disease diagnostics, and in
human skin3 which may easily colonize on the indwelling sensors.15 The increasing adaptability of clinical microbial
catheters surface,4 5 prosthetic joints,6 cerebrospinal fluid strains to antimicrobial drug resistance demands highly
effective compounds for the treatment of critical micro-
shunts,7 8 and other medical devices by the production
bial infection. Nanoparticles have demonstrated antimi-
of adhesin like extracellular polysaccharide. Among fungi
crobial activities; the development of novel applications
affecting human health, Candida tropicalis found in nor-
in this field makes them an attractive alternative to con-
mal human mucocutaneous flora, is mainly responsible
ventional dispensesions. Nanoparticles have also been
for septicemia and disseminated candidiasis, especially in
used in clothing and in the food industry to limit bac-
patients with lymphoma, leukemia and diabetes.9–10 Can- terial growth.16–18 For instance, nanoparticles have been
dida tropicalis also shows a high incidence of antifungal examined for their ability to reduce microbial infections
drug resistance.11 in skin19 and burn wounds,20 and also to prevent bac-
terial colonization on various surface devices such as

catheters21 and prostheses.22 Metal oxide nanoparticles
Author to whom correspondence should be addressed.
Email: jayantaism@gmail.com (NPs) are known to possess strong antimicrobial proper-
Received: 23 November 2012 ties. Inorganic metal oxides are being increasingly used for
Revised/Accepted: 4 April 2013 antimicrobial applications. The main advantages of using

J. Biomed. Nanotechnol. 2013, Vol. 9, No. xx 1550-7033/2013/9/001/008 doi:10.1166/jbn.2013.1681 1


Antimicrobial Activity of CaO Nanoparticles Roy et al.

inorganic oxides when compared with organic antimicro- Synthesis of CaO Nanoparticles
bial agents are their stability, robustness, and long shelf CaO nanoparticles (CaO-NPs) with narrow size distribu-
life. The considerable antimicrobial activities of inorganic tion were synthesized using microwave irradiation method
metal oxide nanoparticles such as ZnO, MgO, TiO2 , SiO2 with minor modifications.39 Briefly, 0.5 M Ca(NO3 2 ·
and their selective toxicity to biological systems sug- 4H2 O and 0.7 M of NaOH were separately dissolved in
gest their potential application in therapeutics, diagnostics, 50 ml de-ionized water and mixed to form 100 ml mixture
surgical devices and nanomedicine based antimicrobial (solutions). The mixture was stirred for 10 min at room
agents.23–27 The advantages of using these inorganic oxides temperature and it turned into white gel. Then the mix-
nanoparticles as antimicrobial agents are their greater ture was transferred in a 250 mL round bottom flask with
effectiveness on resistant strains of microbial pathogens. condenser attachment kept in a Multi Synth microwave
A higher concentration of smaller particles with a higher refluxing system. Maximum deliverable power output was
surface area gives a better antibacterial behavior,28–30 200 W and was set at a temperature of 160  C for duration
whereas crystalline structure and particle shape have a of 5 min. After microwave processing, the solution was
very small effect.31 A number of mechanisms have also allowed to cool to reach room temperature, naturally. The
been proposed to interpret these experimental observa- resulting precipitate was collected by vacuum filtration and
tions. These include production of active oxygen species washed with de-ionized water and absolute ethanol, and
due to the presence of nanoparticles,30 32 damage of mem- dried in a vacuum at 80  C for 1 h.
brane cell wall through adhesion on the cell membrane,33
penetration through the membrane cell wall30 and cellular Characterization of Synthesized Nanoparticles
internalisation of nanoparticles.34 35 The phase analysis and crystal structure of CaO-NPs was
Although having promising potency towards disease studied by X-ray diffractograms (XRD). The XRD pattern
causing microorganisms of the metal oxide nanoparticles
was recorded using a diffractometer of Philips X-pert Pro
there are a number of literature also available explain-
X-ray diffractometer, with 0.1789 nm Co-K radiation.
ing cytotoxicity and genotoxicity of those nanoparticles.
Particle sizes and morphology were studied with a high
The toxicity of several metal oxide NPs such as TiO2 ,
resolution transmission electron microscope (HRTEM) of
ZnO, and Fe2 O3 has been analyzed using different cell
JEOL (JEM-2100), with an operating voltage 200 kV.
lines. A complete review about this issue has been recently
In sample preparation for HRTEM studies, a small portion
reported.36 At such juncture researchers are interested to
of CaO-NPs were dispersed in acetone and sonicated for
find out new nanomaterials having potential antimicrobial
30 min. Part of this dispersion was dropped over a car-
effects. Although only a few studies have been reported
bon film supported by a copper grid and dried in vacuum
the antibacterial properties of calcium oxide, they show
before imaging.
calcium oxide powder37 38 has a significant promise as
bactericidal agent, for their relatively less toxicity and no
recorded endocrine disruptive effect. Antimicrobial Activity Assay
In this study, a new antimicrobial nanoparticle (CaO- The preliminary antimicrobial susceptibility was tested by
NPs) was synthesized and assessed for antimicrobial activ- using agar-well diffusion method as described by Perez
ities using a panel of bacterial and fungal pathogen. Here, et al.40 Briefly, the bacterial isolates were first grown in
Staphylococcus epidermidis (MTCC 435), Pseudomonas a nutrient broth for 12–18 h before use and standardized
aeruginosa (ATCC 27853) and Candida tropicalis (NCIM to 0.5 McFarland standards (106 cfu ml−1 ). One hundred
3110) are chosen as a representative of Gram negative microliter of the standardized cell suspensions were spread
bacteria, Gram positive bacteria, and fungus, respectively, on a Mueller-Hinton agar (Hi Media) and the agar medium
which are the most causal pathogens for skin diseases. was punched with a six millimeters diameter wells and
filled with different concentration of nanoparticles solu-
tions in equal amounts. The plates are observed for zone
EXPERIMENTAL DETAILS of inhibition after 24 h incubation at 37  C.
Chemicals The Minimum Inhibitory concentration (MIC) and Min-
For the synthesis of CaO-NPs, Ca(NO3 2 · 4H2 O (Merck) imum Bactericidal Concentration (MBC) were determined
and Sodium hydroxide (Merck) and ultra pure Milli-Q by following broth micro-dilution method and agar plating
water were used and; other reagents were obtained from method, respectively according to Clinical and Laboratory
local supplies. Standards Institute (CLSI) guidelines.41 The concentra-
tions of CaO-NPs used for the assay ranged from 0.125 to
Biological Materials 32 mM. MIC value was determined by the microtitre plate
Antimicrobial activity was tested using Staphylococ- dilution method,42 where no visible growth is observed. A
cus epidermidis (MTCC 435) as Gram-positive bacteria, range of CaO concentrations were added to sterile 96-well
Pseudomonas aeruginosa (ATCC 27853) as Gram-negative microtitre plates and the volume was made up to 250 L
bacteria, and Candida tropricalis (NCIM 3110) as fungus. well−1 with Mueller-Hinton broth. Culture of the strain

2 J. Biomed. Nanotechnol. 9, 1–8, 2013


Roy et al. Antimicrobial Activity of CaO Nanoparticles

was added to the mixtures in such a way that the total CO2 -ethanol exchange in a Critical Point Dryer (Denton
inoculum load was 105 cells/well. Finally, microtitre plates Vacuum, Inc., Cherry Hill, NJ). The cover slips were
were incubated at 37  C for 24 h. Bacterial growth was mounted on SEM stubs with carbon adhesive tabs, then
measured by the optical density at 600 nm using a Mul- sputter-coated with a thin layer of gold using a Scancoat Six
tiskan Spectrum spectrophotometer (model 1500; Thermo Sputter Coater (BOC Edwards, Wilmington, MA). Digital
Scientific, Nyon, Switzerland) and the MIC value of the images of the treated and untreated Pseudomonas aerugi-
CaO-NPs was determined by comparing the cell densi- nosa cells were acquired using a field emission scanning
ties with wells where no CaO-NPs was added as positive electron microscope (FESEM) of Carl Zeiss model Supra-
control and no bacteria with different test concentrations 40 (with an accelerated voltage 10–20 kV) and instrumental
(uninoculated control) were used as negative control to magnifications of 25,000×. Attachment of CaO-NPs over
maintain the sterility as well as blank. Minimum Bacte- the cell surface was studied with in situ EDX in conjunc-
ricidal Concentration (MBC) was determined by colony tion with the FESEM imaging of selective regions.
forming unit (CFU) of each concentration following dilu-
tion plating method.43 44 Determination of all values were
carried out in triplicate and repeated at least four times for RESULTS AND DISCUSSION
accuracy. In case of Candida tropicalis, Sabouraud dex- Characterization of Synthesized Nanoparticals
trose medium was used for all antifungal assays. X-ray diffraction (XRD), measurements were carried out
over the diffraction angle (2) 20 –80 (Fig. 1). Unit cell
Time-Kill Assays parameters were obtained by least-square refinement of the
Time-kill assays were performed to check the efficacy powder XRD data. XRD study revealed that the products
CaO-NPs towards tested pathogens. In this assay, all the are CaO-NPs with lattice constant a = 4801 Å (Fm3m
tested strains were treated with different concentration space group) having nanosized particles; the d-spacing
of CaO-NPs within the range of 0.125–8 mM and their values in the parenthesis indicate respective Miller indices.
growth was monitored at every 1 h time interval. The crystallite size was measured using Scherrer equa-
tion and it was found that the average size (D) of synthe-
Live Microscopy and Biofilm Formation sized CaO-NPs was 16 nm. X-ray diffraction studies shows
The live growth pattern of all the CaO-NPs treated that the microwave assisted synthesized materials were
microorganisms were monitored in microtitre plate by pure monophasic cubic CaO-NPs and the crystal structures
inverted phase contrast microscopy (Olympus 1 × 51). agree well with the corresponding reported JCPDS data
Images were captured by Image ProTM discovery soft- (JCPDS powder diffraction data card no. 77-2376). Line
ware (Olympus). While, the qualitative biofilm forma- broadening of the diffraction peaks is an indication that the
tion assayed for P. aeruginosa was made by fluorescence synthesized materials are in nanometer range. The char-
microscopy. Different concentrations of CaO-NPs were acteristic peaks were higher in intensity and narrower in
used to treat 24 h grown P. aeruginosa in polystyrene plate spectral width, indicating that the products were of good
and again incubated for 48 h at 37  C. After 48 h incu- crystallinity. No peaks corresponding to impurities were
bation, the medium was aspirated off and non-adherent detected, showing that the final products are high quality
cells were removed by washing thoroughly with sterile CaO-NPs.
0.15 M phosphate-buffered saline (PBS) pH 7.2. Bacterial
samples were stained with Syto Green fluorescence stain
according to manufacturer protocol (Invitrogen) and visu-
alized directly in an inverted microscope and photographed
(Olympus).

Examination of Cell Morphology by


Scanning Electron Microscope
Mid-log phase Pseudomonas aeruginosa cultures were
treated with 4 mM CaO-NPs for 12 h. Aliquots of 200 l
treated and untreated cell suspensions were deposited on
glass cover slips. After air drying for 1 h, the cover
slips were fixed with a primary fixative solution contain-
ing 2.5% glutaraldehyde. Subsequently, the fixative solu-
tion was exchanged with 0.1 M imidazole buffer, followed
by dehydration with a series of ethanol solutions (50%,
80% and 100%) with three ethanol changes at each con-
centration. Finally the cover slips were dried with liquid Figure 1. The XRD pattern of synthesized CaO-NPs.

J. Biomed. Nanotechnol. 9, 1–8, 2013 3


Antimicrobial Activity of CaO Nanoparticles Roy et al.

Figure 2. The HRTEM micrograph of synthesized CaO-NPs with inset showing lattice spacing and particles size distribution
histogram in the inset.

Morphological characterization was carried out by minimum bactericidal concentration (MBC). Table I shows
HRTEM, which is a direct way that provides visual comparative study of MIC and MBC values of different
demonstration to estimate particle size exactly. Figure 2 microbes. The MIC values of CaO-NPs were found to
shows HRTEM micrograph of the synthesized CaO-NPs.
HRTEM micrograph of the sample indicates that the sam-
Table I. Antimicrobial activity of CaO nanopartocles against
ple was well dispersive nanoparticles. Inset in Figure 2 three different microbes.
shows the lattice fringe of 0.2403 nm corresponds to the
(200) plane of CaO-NPs. The image shows an abundance Antimicrobial activity (mM)
of particles whose size distribution is given by the his- Name of organism MIC MBC
togram shown in the inset of Figure 2, the histogram was
S. epidermidis 2 4
obtained by analyzing several frames of similar bright P. aeruginosa 4 8
field images using IMAGE J software. HRTEM shows C. tropicalis 8 8
microstructure of the nanomaterials having cubic shapes
which is similar with X-ray diffraction study. The particles
have an average size of ∼18 nm, which is in close agree- Table II. Comparative table of minimum bactericidal concen-
ment with an average D-value 16 nm determined from the trations (MBCs) of some metals and metal oxides nanoparti-
XRD peak broadening. In this present work minor modifi- cles against P. aeruginosa.
cation in the synthesis process of CaO-NPs produces nar- Name of
row size distribution and smaller sized nanoparticles than Sl. no. Nanopartical MBC (mM) References
earlier report.39 1 Ag 011 [45]
2 Cu 7869 [44]
Antimicrobial Assay 3 Cu2 O 1747 [44]
The antibacterial effect of CaO-NPs against Staphylo- 4 CuO 6285 [44]
5 ZnO 6141 [46]
coccus epidermidis, Pseudomonas aeruginosa and Can-
6 TiO2 10016 [46]
dida tropicalis growth was verified by two established 7 CaO 8 In this study
measures: minimum inhibitory concentration (MIC) and

4 J. Biomed. Nanotechnol. 9, 1–8, 2013


Roy et al. Antimicrobial Activity of CaO Nanoparticles

Jones et al.48 reported MIC of ZnO was 5 mM, whereas


the studied CaO-NPs has much lower MIC value of only
2 mM against Staphylococcus epidermidis. The higher
antimicrobial activity may be due to high surface area
(96.52 m2 /g) and small particle sizes (∼18 nm) of synthe-
sized CaO-NPs and thus can cause the bacteria to coagu-
late in nanoparticle suspensions.

Time-Kill Assays
All the microorganisms were challenged with CaO-NPs
for a period of 4 h. The antimicrobial effect was shown
to be dose-dependent and rapid (Fig. 4) for all the tested
strains. Microbial reduction was significant and reached
near 100% in case of Staphylococcus epidermidis within 2
h and Pseudomonas aeruginosa within 3 h. In case of Can-
Figure 3. Growth pattern of three tested organisms in pres- dida tropicalis the reduction was 97% at 3 h incubation.
ence of different concentrations of CaO-NPs. Culture medium For Staphylococcus epidermidis, 4 and 8 mM of CaO-NPs
was supplemented with various concentrations (0–32 mM) of
were the lowest most effective concentrations. For Pseu-
nanoparticles. Bacterial growth was measured at 600 nm after
24 h at 30 ± 2  C. The bacterial growth OD was calculated by domonas aeruginosa, 8 mM at 3 h and 4 mM at 4 h of
deduction of blank from test. Data are the mean of triplicate CaO-NPs concentrations were found to be the most effec-
experiments ± S.E. tive dose. In case of Candida tropicalis, the most effective
lowest antimicrobial CaO-NPs concentration was found at
be 2, 4 and 8 mM for Staphylococcus epidermidis, Pseu- 8 mM at 4 h.
domonas aeruginosa and Candida tropicalis respectively
(Fig. 3 and Fig. S2). The MBC result showed that the Microscopic Observations
cells treated with 4, 8 and 8 mM of the nanoparticles for
Figure 5 shows fluorescence micrograph of inhibitory
Staphylococcus epidermidis, Pseudomonas aeruginosa and
effect of CaO-NPs on biofilm formation against Pseu-
Candida tropicalis were no longer culturable, suggesting
domonas aeruginosa. Complete (Fig. 5(C)) biofilm forma-
a lethal effect of CaO-NPs. The selected CaO-NPs were
tion was observed in case of 4 mM concentration of CaO-
effective in killing a range of bacterial pathogens com-
NPs. Effects of CaO-NPs on Pseudomonas aeruginosa cell
mon in hospital-acquired infections. The bactericidal con-
centration was quite lower for Staphylococcus epidermidis morphology were also examined by FESEM micrograph.
(4 mM) in comparision to gram negative Pseudomonas Pseudomonas aeruginosa (rod shaped bacteria) show their
aeruginosa (8 mM) and unicellular yeast Candida trop- unique shapes in untreated control sample (Fig. 5(D)).
icalis (8 mM). It has been suggested that the greater Although the cells of P. aeruginosa were still present (Fig.
amount of negatively charged peptidoglycans would make 5(E)), most strains of Pseudomonas aeruginosa were dam-
Gram-positive bacteria more susceptible to such positively aged and extensively disappeared by addition of CaO-NPs
charged antimicrobials.45 However, in comparison to some solution. The cell surface of Pseudomonas aeruginosa was
other reported metal or metal oxide nanoparticles, the covered with substance resulted from the cell disruption
studied CaO-NPs needed much lower bactericidal concen- after the CaO-NPs treatment shown in (Fig. 5(F)). The
trations for Pseudomonas aeruginosa as shown in Table II. presence of CaO-NPs on the bacterial cell surface was

Figure 4. Reduction of microbial population with time when treated with different concentrations of CaO-NPs (0.125, 0.25, 0.5,
1, 2, 4 and 8 mM). Results are given as percentage of control (microorganisms are in nanoparticle-free phosphate-buffered
saline). Standard error bars are shown. Three replicate experiments on three separate occasions were performed.

J. Biomed. Nanotechnol. 9, 1–8, 2013 5


Antimicrobial Activity of CaO Nanoparticles Roy et al.

Figure 5. Fluorescence microscopy images of P. aeruginosa biofilm formation inhibition (A) Control cells without CaO-NPs,
(B) treated with 2 mM CaO-NPs (C) treated with 4 mM CaO-NPs. P. aeruginosa culture was stained with Syto Green flouroscence
stain (invitrogen) Scale bar: 20 m.; FESEM images of P. aeruginosa: (D) without nanoparticles; (E) with 4 mM CaO-NPs (F) mag-
nified images of Images showed the nanoparticles were adhere to bacterial cell surface and ruptured the cell membrane. All the
images were taken at 400× magnification.

supported by EDX analysis in Figure S1 (in supporting When compared to normal untreated microbial population,
materials). CaO-NPs treated microbial population showed up to
Bacterial growth inhibitory effects of CaO-NPs were 95 fold destruction (Fig. 6) in their cell structure.
studied by phase contrast and fluorescence microscopy. Figure 6(A) showed that, after addition of 2 mM

Figure 6. Phase contrast microscopy image (400× magnifications) of S. epidermidis (A), P. aeruginosa (B) and C. tropicalis (C)
treated with 0 mM, 2 mM and 4 mM CaO-NPs nanoparticles after 24 h incubation at 37  C. Scale bar: 20 m.

6 J. Biomed. Nanotechnol. 9, 1–8, 2013


Roy et al. Antimicrobial Activity of CaO Nanoparticles

concentration of CaO-NPs in Staphylococcus epidermidis, promising antibacterial agents. The effect was dose depen-
cells became irregular in size, shape and clamping also dent and found more pronounced against gram positive
occurs. Whereas, after addition of 4 mM of CaO-NPs, organisms than gram negative ones. Effect of contact time
few cells were observed but their shapes were irregular with CaO-NPs and microorganisms also showed promis-
in nature and most of the cells were found lysed. Figure ing result. CaO-NPs showed antimicrobial effect towards
6(B) shows microscopic image of Pseudomonas aerugi- test organisms Staphylococcus epidermidis, Pseudomonas
nosa after addition of CaO-NPs. Most of the cells were aeruginosa and Candida tropicalis in decreasing order.
elongated and made up of irregular shape after addition
of 2 mM solution of CaO-NPs, and after the addition of Acknowledgments: The authors would like to
4 mM of CaO-NPs a very few well structured cells were acknowledge the financial support provided by the “Coun-
found. Most of them were spherical in shape and lost cell cil of Scientific and Industrial Research (CSIR), India” to
structure integrity. Similar results were also observed in the first author to carry out the work.
case of Candida tropicalis, where fungi lost their morpho-
logical integrity (Fig. 6(C)). REFERENCES
The optical inspection of bacterial specimens and bio- 1. E. Vanhaecke, J. P. Remon, M. Moors, F. Raes, D. D. Rudder, and
logical samples in general in their indigenous states is nor- A. V. Peteghem, Kinetics of pseudomonas aeruginosa adhesion to
mally performed by phase contrast microscopy where the 304 and 316-L stainless steel: Role of cell surface hydrophobicity.
image contrast is obtained by converting the phase vari- Appl. Environ. Microbiol. 56, 788 (1990).
2. S. Cappello and S. P. P. Guglielmino, Effects of growth temperature
ation of the transparent biological object into the ampli- on polystyrene adhesion of Pseudomonas aeruginosa ATCC 27853.
tude variation. All the microbial strains were found to lose Braz. J. Microbiol. 37, 205 (2006).
their cell structure integrity after treatment with CaO-NPs. 3. Z. Gao, C. H. Tseng, Z. Pei, and M. J. Blaser, Molecular analysis
Spherical shape was observed due to inhibition of cell divi- of human forearm superficial skin bacterial biota. Proc. Natl. Acad.
Sci. USA 104, 2927 (2007).
sion. CaO-NPs appear to cause immediate and widespread
4. G. L. Archer, Antimicrobial susceptibility and selection of resistance
change in cell morphology after addition. The disruption among Staphylococcus epidermidis isolates recovered from patients
of cellular membrane was well supported by the image with infections of indwelling foreign devices. Antimicrob. Agents
of scanning electron microscopy, where the membrane Chemother. 14, 353 (1978).
damage was clearly visible after treatment with CaO-NPs 5. J. W. Bender and W. T. Hughes, Fatal Staphylococcus epider-
midis sepsis following bone marrow transplantation. Johns Hopkins
(Fig. 5). The result shows good agreement with previ- Med. J. 146, 13 (1980).
ously reported literature.32 37 49 50 Sawai et al.32 reported 6. L. E. Blouse, G. D. Lathrop, L. W. Kolonel, and R. M. Brockett,
that slurry of powder oxides like MgO and CaO generate Epidemiologic features and phage types associated with nosocomial
superoxide (O− 2 ) and have antibacterial as well as anti-
infections caused by Staphylococcus epidermidis. Zentralbl Bakte-
fungal activity. Though the generation of reactive oxygen riol Parasitenk Infektionsk Hyg Abt 1 Orig Reihe A 241, 119 (1978).
7. R. P. Callaghan, S. J. Cohen, and G. T. Stewart, Septicaemia due
species (ROS) in the presence of CaO-NPs was not verified to colonization of Spitz–Holter valves by staphylococci. Br. Med. J.
in our experiments, there exist a good number of citations 1, 860 (1961).
claiming generation of supper oxide (O2− ) from the CaO 8. S. C. Schoenbaum, P. Gardner, and J. Shillito, Infection of cere-
surface.49–52 The studies point the role of active oxygen brospinal fluid shunts: Epidemiology, clinical manifestations and
therapy. J. Infect. Dis. 131, 543 (1975).
as one primary mechanism of antimicrobial activity that is
9. W. G. Powderly, K. H. Mayer, and J. R. Perfect, Diagnosis and
also, among other things, influenced by the increase of treatment of oropharingeal candidiasis in patients infected with HIV:
pH in the medium. Sawai et al.49 50 reported that in addi- A critical reassessment. AIDS Res. Hum. Retroviruses 15, 1405
tion to high pH another factor, active oxygen also helps (1999).
in antibacterial activity of CaO and MgO powder. ROS 10. J. W. Rippon, Medical Mycology, 3rd edn. WB Saunders Co,
Philadelphia, USA (1988).
such as HO•2 , • O−2 , and H2 O2 generated from the surface of 11. D. Law, C. B. Moore, L. A. Joseph, M. G. Keaney, and D. W.
CaO-NPs, react and break the structure of polyunsaturated Denning, High incidence of antifungal drug resistance in Candida
phospholipids in the microbial cell membrane. In addition, tropicalis. Int. J. Antimicrob. Agents 7, 241 (1996).
the alkaline nature of the CaO-NPs could help metal oxide 12. J. S. Kim, E. Kuk, K. N. Yu, J. H. Kim, S. J. Park, H. J. Lee,
nanopartcles to be surrounded with OH− layers.53 S. H. Kim, Y. K. Park, Y. H. Park, C. Y. Hwang, Y. K. Kim, Y. S.
Lee, D. H. Jeong, and M. H. Cho, Antimicrobial effects of silver
nanoparticles. Nanomed. Nanotechnol. Biol. Med. 3, 95 (2007).
13. P. Wang, Nanoscale biocatalyst systems. Curr. Opinion. Biotechnol.
CONCLUSION
17, 574 (2006).
In summary, the broad-spectrum antimicrobial efficiency 14. L. Zhang, F. X. Gu, J. M. Chan, A. Z. Wang, R. S. Langer, and O. C.
of CaO-NPs was demonstrated. Minor modified method Farokhzad, Nanoparticles in medicine: Therapeutic applications and
for synthesis CaO-NPs has prominent control on size developments. Clin. Pharmacol. Ther. 83, 761 (2008).
(∼18 nm) and narrow size distribution (14–24 nm). 15. B. Hong, J. Kai, Y. Ren, J. Han, Z. Zou, C. H. Ahn, and K. A.
Kang, Part VII-Nano-Bio Technology-30 Highly sensitive rapid, reli-
Obtained values of MIC for Staphylococcus epider- able, and automatic cardiovascular disease diagnosis with nanopar-
midis, Pseudomonas aeruginosa and Candida tropicalis ticle fluorescence enhancer and MEMS. Adv. Exp. Med. Biol. 614,
showed excellent antibacterial activity and can be used as 265 (2008).

J. Biomed. Nanotechnol. 9, 1–8, 2013 7


Antimicrobial Activity of CaO Nanoparticles Roy et al.

16. C. F. Chau, S. H. Wu, and G. C. Yen, The development of regula- 35. Z. Xiu, Q. B. Zhang, H. L. Puppala, V. L. Colvin, and P. J. J.
tions for food nanotechnology. Trends in Food Sci. Technol. 18, 269 Alvarez, Negligible particle-specific antibacterial activity of silver
(2007). nanoparticles, Nano Lett. 12, 4271 (2012).
17. N. Vigneshwaran, A. A. Kathe, P. V. Varadarajan, R. P. Nachane, 36. N. Singh, B. Manshian, G. J. Jenkins, S. M. Grif?ths, P. M. Williams,
and R. Balasubramanya, Functional finishing of cotton fabrics using T. G. Maffeis, and C. J. Wright, Nanogenotoxicology: The DNA
silver nanoparticles. J. Nanosci. Nanotechnol. 7, 1893 (2007). damaging potential of engineered nanomaterials. Biomaterials 30,
18. J. M. Conlon, J. Kolodziejek, and N. Nowotny, Antimicrobial pep- 3891 (2009).
tides from ranid frogs: Taxonomic and phylogenetic markers and a 37. J. Sawai, H. Igarashi, A. Hashimoto, T. Kokugan, and M. Shimizu,
potential source of new therapeutic agents. Biochim. Biophys. Acta Evaluation of growth inhibitory effect of ceramics powder slurry on
1696, 1 (2004). bacteria by conductance method. J. Chem. Eng. Jpn. 28, 288 (1995).
19. J. E. Paddle-Ledinek, Z. Nasa, and H. J. Cleland, Effect of different 38. J. Sawai and T. Yoshikaw, Quantitative evaluation of antifungal activ-
wound dressings on cell viability and proliferation. Plastic Recon- ity of metallic oxide powders (MgO, CaO and ZnO) by an indirect
struct. Surg. 117, 110S (2006). conductimetric assay. J. Appl. Microbiol. 96, 803 (2004).
20. E. Ulkur, O. Oncul, H. Karagoz, E. Yeniz, and B. Celikoz, Compar- 39. A. Roy and J. Bhattacharya, Microwave-assisted synthesis and char-
ison of silver-coated dressing (Acticoat), chlorhexidine acetate 0.5% acterization of CaO nanoparticles. Int. J. Nanosci. 10, 413 (2011).
(Bactigrass), and fusidic acid 2% (Fucidin) for topical antibacterial 40. C. Perez, M. Paul, and P. Bazerque, An antibiotic assay by the agar
effect in methicillin-resistant Staphylococci-contaminated, full-skin well diffusion method. Acta Bio. Med. Exp. 15, 113 (1990).
thickness rat burn wounds. Burns 31, 874 (2005). 41. CLSI, Clinical and Laboratory Standards Institute, Performance stan-
21. U. Samuel and J. P. Guggenbichler, Prevention of catheter-related dards for antimicrobial susceptibility testing. Seventeenth informa-
infections: the potential of a new nano-silver impregnated catheter. tional supplement. Document M100-S17 Wayne, PA (2007).
Int. J. Antimicrob. Agents 23, S75 (2004). 42. A. P. Wang, Y. P. Su, S. Wang, M. Q. Shen, F. Chen, M. Chen, X. Z.
22. G. Gosheger, J. Hardes, H. Ahrens, A. Streitburger, H. Buerger, Ran, T. M. Chen, and J. P. Wang, Antibacterial activity and mecha-
M. Erren, A. Gunsel, F. H. Kemper, W. Winkelmann, and C. von nism of recombinant human -defensin 5 against clinical antibioti-
Eiff, Silver-coated megaendoprostheses in a rabbit model—an anal- cresistant strains, Afr. J. Microbiol. Res. 4, 626 (2010).
ysis of the infection rate and toxicological side effects. Biomaterials 43. A. Giacometti, O. Cirioni, R. Ghiselli, L. Goffi, F. Mocchegiani,
25, 5547 (2004). A. Riva, G. Scalise, and V. Saba, Polycationic peptides as prophy-
23. J. Mohsen and B. Zahra, Protein nanoparticle: A unique system as lactic agents against methicillin-susceptible or methicillin-resistant
drug delivery vehicles. Afr. J. Biotechnol. 7, 4926 (2008). Staphylococcus epidermidis vascular graft infection. Antimicrob.
24. K. Sobha, K. Surendranath, V. Meena, K. T. Jwala, N. Swetha, and Agents Chemother. 44, 3306 (2000).
K. S. M. Latha, Emerging trends in nanobiotechnology. J. Biotech. 44. P. H. S. Kwakman, A. A. Velde, C. M. J. E. Vandenbroucke-Grauls,
Mol. Bio. Rev. 5, 001 (2010). S. J. H. Deventer, and S. A. J. Zaat, Treatment and prevention
25. K. A. Laura, Y. L. Delina, and J. J. A. Pedro, Comparative eco- of Staphylococcus epidermidis experimental biomaterial-associated
toxicity of nanoscale TiO2 , SiO2 , and ZnO water suspensions. infection by bactericidal peptide 2. Antimicrob. Agents Chemother.
J. Water Res. 40, 3527 (2006). 50, 3977 (2006).
26. J. Sawai, Quantitative evaluation of antibacterial activities of metal- 45. G. Ren, D. Hu, E. W. C. Cheng, M. A. Vargas-Reus, P. Reip, and
lic oxide powders ZnO, MgO and CaO by conductimetric assay. R. P. Allaker, Characterisation of copper oxide nanoparticles for
J. Microb. Meth. 54, 177 (2003). antimicrobial applications. Int. J. Antimicrob. Agents 33, 587 (2009).
27. K. M. Reddy, F. Kevin, B. Jason, G. W. Denise, H. Cory, and 46. J. Jain, S. Arora, J. Rajwade, M. Jyutika, P. Omray, S. Khandelwal,
P. Alex, Selective toxicity of zinc oxide nanoparticles to prokaryotic and M. K. Paknikar, Silver nanoparticles in therapeutics: devel-
and eukaryotic systems. J. Appl. Phys. Lett. 90, 1 (2007). opment of an antimicrobial gel formulation for topical use. Mol.
28. J. Sawai, H. Igarashi, A. Hashimoto, T. Kokugan, and M. Shimizu, Pharm. 6, 1388 (2009).
Effect of particle size and heating temperature of ceramic powders 47. V. S. Desai and M. Kowshik, Antimicrbial activity of titanium diox-
on antibacterial activity of their slurries. J. Chem. Eng. Jpn. 29, 251 ides nanopartical synthesized by sol-gel techniques. Res. J. Micro-
(1996). biol. 4, 97 (2009).
29. O. Yamamoto, Influence of particle size on the antibacterial activity 48. N. Jones, B. Ray, T. K. Ranjit, and A. C. Manna, Antibacterial
of zinc oxide. Int. J. Inorg. Mater. 3, 643 (2001). activity of ZnO nanoparticle suspensions on a broad spectrum of
30. S. Makhluf, R. Dror, Y. Nitzan, Y. Abramovich, R. Jelinek, and microorganisms. FEMS Microbiol. Lett. 279, 71 (2008).
A. Gedanken, Microwave-assisted synthesis of nanocrystalline MgO 49. J. Sawai, H. Kojima, H. Igarashi, A. Hashimoto, S. Shoji, T. Sawaki,
and its use s a bacteriocide. Adv. Funct. Mater. 15, 1708 (2005). A. Hakoda, E. Kawada, T. Kokugan, and M. Shimizu, Antibacterial
31. O. Yamamoto, M. Hotta, J. Sawai, T. Sasamoto, and H. Kojima, characteristics of magnesium oxide powder. World J. Microb. Biot.
Influence of powder characteristic of ZnO on antibacterial activity- 16, 187 (2000).
effect of specific surface area. J. Ceram. Soc. Jpn. 106, 1007 (1998). 50. J. Sawai, S. Shiga, and H. Kojima, Kinetic analysis of death
32. J. Sawai, E. Kawada, F. Kanou, H. Igarashi, A. Hashimoto, of bacteria in CaO powder slurry. Int. Biodeter. Biodegr. 47, 23
T. Kokugan, and M. Shimizu, Detection of active oxygen generated (2001).
from ceramic powders having antibacterial activity. J. Chem. Eng. 51. O. Yamamoto, T. Shimura, J. Sawai, H. Kojima and T. Sasamoto,
Jpn. 29, 627 (1996). effect of CaO doping on antibacterial activity of ZnO powders.
33. P. K. Stoimenov, R. L. Klinger, G. L. Marchin, and K. J. Klabunde, J. Ceram. Soc. Japan 108, 156 (2000).
Metal oxide nanoparticles as bactericidal agents. Langmuir 18, 6679 52. J. Sawai, Quantitative evaluation of antibactrerial activities of metal-
(2002). lic oxide powders (MgO, CaO and ZnO) by conductimetric assay.
34. R. Brayner, R. Ferrari-Lliou, N. Brivois, S. Djediat, M. F. Benedetti, J. Appl. Microbiol. 89, 397 (2000).
and F. Fievet, Toxicological impact studies based on Escherichia coli 53. K. Sugiyama, T. Suzuki, and T. Satoh, Bactericidal activity of
bacteria in ultrafine ZnO nanoparticles colloidal medium. Nano Lett. silicate-containing hydroxyapatite. J. Antibact. Antifung. Agents 23,
6, 866 (2006). 67 (1995).

8 J. Biomed. Nanotechnol. 9, 1–8, 2013

Das könnte Ihnen auch gefallen