ArticlePDF Available

Magnetic fields suppress Pseudomonas aeruginosa biofilms and enhance ciprofloxacin activity

Authors:

Abstract and Figures

Due to the refractory nature of pathogenic microbial biofilms, innovative biofilm eradication strategies are constantly being sought. Thus, this study addresses a novel approach to eradicate Pseudomonas aeruginosa biofilms. Magnetic nanoparticles (MNP), ciprofloxacin (Cipro), and magnetic fields were systematically evaluated in vitro for their relative anti-biofilm contributions. Twenty-four-hour biofilms exposed to aerosolized MNPs, Cipro, or a combination of both, were assessed in the presence or absence of magnetic fields (Static one-sided, Static switched, Oscillating, Static + oscillating) using changes in bacterial metabolism, biofilm biomass, and biofilm imaging. The biofilms exposed to magnetic fields alone exhibited significant metabolic and biomass reductions (p < 0.05). When biofilms were treated with a MNP/Cipro combination, the most significant metabolic and biomass reductions were observed when exposed to static switched magnetic fields (p < 0.05). The exposure of P. aeruginosa biofilms to a static switched magnetic field alone, or co-administration with MNP/Cipro/MNP + Cipro appears to be a promising approach to eradicate biofilms of this bacterium.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=gbif20
Download by: [104.249.167.39] Date: 15 December 2015, At: 13:44
Biofouling
The Journal of Bioadhesion and Biofilm Research
ISSN: 0892-7014 (Print) 1029-2454 (Online) Journal homepage: http://www.tandfonline.com/loi/gbif20
Magnetic fields suppress Pseudomonas
aeruginosa biofilms and enhance ciprofloxacin
activity
H.M.H.N. Bandara, D. Nguyen, S. Mogarala, M. Osiñski & H.D.C. Smyth
To cite this article: H.M.H.N. Bandara, D. Nguyen, S. Mogarala, M. Osiñski & H.D.C. Smyth
(2015) Magnetic fields suppress Pseudomonas aeruginosa biofilms and enhance ciprofloxacin
activity, Biofouling, 31:5, 443-457, DOI: 10.1080/08927014.2015.1055326
To link to this article: http://dx.doi.org/10.1080/08927014.2015.1055326
Published online: 23 Jun 2015.
Submit your article to this journal
Article views: 261
View related articles
View Crossmark data
Magnetic elds suppress Pseudomonas aeruginosa biolms and enhance ciprooxacin activity
H.M.H.N. Bandara
a
, D. Nguyen
a
, S. Mogarala
a
, M. Osiñski
b
and H.D.C. Smyth
a
*
a
College of Pharmacy, The University of Texas at Austin, Austin, TX, USA;
b
Department of Electrical & Computer Engineering,
Center for High Technology Materials, The University of New Mexico, Albuquerque, NM, USA
(Received 12 March 2015; accepted 20 May 2015)
Due to the refractory nature of pathogenic microbial biolms, innovative biolm eradication strategies are constantly being
sought. Thus, this study addresses a novel approach to eradicate Pseudomonas aeruginosa biolms. Magnetic nanoparticles
(MNP), ciprooxacin (Cipro), and magnetic elds were systematically evaluated in vitro for their relative anti-biolm con-
tributions. Twenty-four-hour biolms exposed to aerosolized MNPs, Cipro, or a combination of both, were assessed in the
presence or absence of magnetic elds (Static one-sided, Static switched, Oscillating, Static + oscillating) using changes in
bacterial metabolism, biolm biomass, and biolm imaging. The biolms exposed to magnetic elds alone exhibited sig-
nicant metabolic and biomass reductions (p< 0.05). When biolms were treated with a MNP/Cipro combination, the most
signicant metabolic and biomass reductions were observed when exposed to static switched magnetic elds ( p< 0.05).
The exposure of P. aeruginosa biolms to a static switched magnetic eld alone, or co-administration with MNP/Cipro/
MNP + Cipro appears to be a promising approach to eradicate biolms of this bacterium.
Keywords: Pseudomonas aeruginosa; biolms; magnetic elds; ciprooxacin; magnetic nanoparticles; drug delivery
Introduction
In nature, most microorganisms prefer a communal growth
lifestyle rather than surviving as solitary cells or single
species. These microbial communities are termed biolms
(Harding et al. 2009). Biolms are dened as a complex
functional community of one or more species of
microbes encased in an extracellular polymeric network
and attached to one another or to a solid surface
(Samaranayake 2006). These communities are hierarchi-
cally arranged and three dimensionally organized in order
to gain ecological advantages for better survival, when
compared to their planktonic counterparts (Hall-Stoodley
et al. 2004). Gram-positive pathogens such as Staphylo-
coccus epidermidis,Staphylococcus aureus and Strepto-
coccus sp., Gram-negative bacteria such as Pseudomonas
aeruginosa and Enterobacteriaceae, and fungi such as
Candida spp. are frequently associated with biolm
related infections (Lode et al. 2000; Lyczak et al. 2000;
Saltzstein et al. 2007; Romling & Balsalobre 2012). In the
USA, P. aeruginosa is among the 10 most commonly iso-
lated pathogens involved in healthcare-associated infec-
tions and the second most commonly isolated pathogen
associated with ventilator-related pneumonia (Hidron et al.
2008). Moreover, P. aeruginosa biolms cause lung infec-
tions in 95% of adult cystic brosis (CF) patients (Hart &
Winstanley 2002), in addition to chronic wound infections,
catheter associated urinary tract infections, chronic otitis
media, rhinosinusitis and contact lens associated keratitis
(Costerton et al. 1999; Hoiby et al. 2010).
Due to a slow growth rate, altered microbial metabo-
lism, phenotypic changes, oxygen gradients and pH dif-
ferences, extracellular substances and persister cells,
complete eradication of P. aeruginosa biolms with con-
ventional antibiotics is almost impossible (Romling &
Balsalobre 2012). However, it is generally accepted that
superior biolm control can be achieved when the
antibiotic is combined with another anti-biolm agent
(Romling & Balsalobre 2012). For instance, established
P. aeruginosa biolms in CF lungs are treated with an
intensive course of antibiotics (nebulized tobramycin)
and DNases (to disrupt eDNA/extracellular DNA in bio-
lm matrices) (Frederiksen et al. 2006; Lewis 2008;
Hoiby et al. 2011).
Due to the inferior outcomes of existing therapies,
alternative treatment modalities have constantly been
sought to eradicate pathogenic biolms. Early investiga-
tions explored the possibility of using electromagnetic
elds to eliminate P. aeruginosa (Anwar et al. 1992;
Khoury et al. 1992; Benson et al. 1994). However, these
studies were only conducted on planktonic phases ignor-
ing refractory biolms, and also failed to meet expected
outcomes (Grosman et al. 1992; Piatti et al. 2002;
Potenza et al. 2004; Gao et al. 2005; Laszlo & Kutasi
2010). Magnetic nanoparticles (MNP) such as iron
oxides have received much attention in anti-tumor thera-
peutic strategies due to greater biocompatibility, low sys-
temic toxicity, and the ability to release thermal energy
in the presence of oscillating magnetic elds (Johannsen
*Corresponding author. Email: hugh.smyth@austin.utexas.edu
© 2015 Taylor & Francis
Biofouling, 2015
Vol. 31, No. 5, 443457, http://dx.doi.org/10.1080/08927014.2015.1055326
Downloaded by [104.249.167.39] at 13:44 15 December 2015
et al. 2007; Soenen & De Cuyper 2010). However, the
applications of MNPs in biolm elimination are still in
their infancy (Subbiahdoss et al. 2012; Taylor et al.
2012). To illustrate the potential anti-biolm properties
of MNP, the authors have demonstrated the successful
usage of superparamagnetic iron oxide nanoparticles
(SPION) for enhancing drug transport in CF mucus and
alginate gels. When exposed to magnetic elds, SPIONs
demonstrated enhanced penetration in model alginate
biolms (McGill et al. 2009; Armijo, Brandt, Matthew
et al. 2012). Therefore, this emerging treatment strategy,
ie usage of polymeric or inorganic nanoparticles, appears
to be a promising approach to eradicate biolms from
tissues and surrounding surfaces (Francolini & Donelli
2010).
To the authorsknowledge, there are no reports on
the application of various magnetic elds to eliminate
the biolm phase of pathogenic P. aeruginosa, co-ap-
plication of magnetic elds (oscillating, static, and
combinations), or the use of MNPs as an alternative
treatment modality along with the co-delivery of antibi-
otics with MNPs and magnetic elds as a biolm elim-
ination strategy. Thus, the aim of this study was to
investigate the efcacy of various magnetic elds in
eliminating in vitro P. aeruginosa biolms treated with
an aerosolized formulation containing different combina-
tions of MNPs, ciprooxacin (Cipro) and spray dried
lactose (SDL).
Materials and methods
Ciprooxacin
Ciprooxacin hydrochloride USP grade was purchased
from Letco Medical (Decatur, AL, USA, Catalog
No. 690953).
Magnetic nanoparticles
FluidMAG-CMX superparamagentic iron oxide
nanoparticles (SPIONs, 150 nm 40 mg ml
1
) in distilled
water were purchased from Chemicell
®
(Chemicell
GmBH, Berlin, Germany).
Spray-dried lactose
Spray-dried lactose (Super Tab 11SD, monohydrate
lactose USP) was provided by DFE Pharma (Princeton,
NJ, USA).
Microorganisms
Pseudomonas aeruginosa PAO1 (provided by Prof.
Marvin Whitley, Molecular Biosciences, The University
of Texas at Austin, USA) was used throughout the study.
The identity of the bacterium was conrmed with a
commercially available API 20 E kit (Biomérieux, Mercy
IEtoile, France). All isolates were stored in multiple ali-
quots at 20°C, after conrming their purity.
Growth media
Blood agar (Sigma Aldrich, St Louis, MO, USA) and
brain heart infusion (BHI, Sigma Aldrich) solution were
used for culturing P. aeruginosa.
Microbial inocula
Prior to each experiment, P. aeruginosa was subcultured
on blood agar for 18 h at 37°C. A loopful of the over-
night bacterial growth was inoculated into BHI medium
and incubated for 18 h in an orbital shaker (80 rpm) at
37°C. Bacteria were harvested, washed twice in phos-
phate buffered saline (PBS, pH 7.2) and resuspended in
PBS. The concentration of P. aeruginosa was adjusted to
1×10
7
cells ml
1
by ultraviolet-visible spectrophotome-
try and conrmed by hemocytometric counting.
Biolm formation
P. aeruginosa biolms were developed as described by
Bandara, Yau et al. (2010) with the following modica-
tions. Commercially available pre-sterilized, polystyrene,
at bottom 96-well microtiter plates (BD Biosciences,
San Jose, CA, USA) were used. First, 100 μl of a stan-
dard bacterial suspension (10
7
organisms ml
1
) were
prepared and transferred into the wells of a microtiter
plate, and the plate was incubated for 1.5 h (37°C,
75 rpm) to promote microbial adherence to the well
surfaces. After this initial adhesion phase, the bacterial
suspensions were aspirated, and each well was washed
twice with PBS to remove loosely adhering bacterial
cells. A total of 200 μl of BHI were added to each well
and the plate was incubated for 24 h (37°C, 75 rpm).
After incubation, the wells were washed twice with PBS
to eliminate traces of the medium. The effects of various
treatments were studied on these 24 h biolms.
Determination of minimum inhibitory concentration
(MIC)
Planktonic phase
The MIC was determined by a broth microdilution assay
in accordance with the CLSI guidelines (CLSI 2012).
Briey, bacterial suspensions (5 × 10
5
cells ml
1
) were
treated with the antibiotic in a concentration gradient
(two-fold dilutions) and incubated in a 96-well microtiter
plate for 24 h at 35°C. At the end of this incubation, the
optical density of the bacterial growth was observed
444 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
by unaided eye. The MIC is dened as the lowest
concentration of antimicrobial agent that completely
inhibits growth of the organism, ie P. aeruginosa grown in
the microtiter wells, compared to the solvent control as
detected by the unaided eye. The assay was performed as
quadruplicates three separate times.
Biolm phase
P. aeruginosa biolms were developed in sterile 96-well
plates (BD Biosciences) as described above. Biolms
were washed twice with PBS and ciprooxacin was
administered in a concentration gradient (two-fold dilu-
tion). The plates were incubated for 24 h at 37°C and
80 rpm.
At the end of the incubation period, an XTT
reduction assay (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-
2H-tetrazolium-5-carboxanilide) was performed to quan-
tify the viability of the biolms. The lowest concentration
of the antibiotic at which the bacterial cells demonstrate
80% viability (as measured by XTT readings) when
compared to the solvent control, is considered to be the
minimum biolm inhibitory concentration (MBC) of
the antibiotic against P. aeruginosa. The assay was
performed as quadruplicates three separate times.
Spray drying of formulations
The spray-dried lactose particles loaded with ciprooxa-
cin, MNP, or both, were prepared using a Buchi
®
B-290
mini spray drier (Buchi, Flawil, Switzerland). The two
formulations with MNPs were prepared in 1 g batches
(ie formulations of SDL + MNP and SDL + MNP +
Cipro) and the other two formulations without MNPs
were prepared in 2.5 g batches (ie formulations of SDL
and SDL + Cipro).
Cipro and MNP were mixed at 5% and 1% (w/w),
respectively, in lactose to prepare dry formulations. SDL,
SDL + Cipro, SDL + MNP, and SDL + MNP + Cipro
formulations were prepared by spray drying. The follow-
ing parameters were used in the spray drying process:
feed solutions consisted of 2.5% (w/v) lactose in distilled
water, inlet temperature 150°C, feed rate 1.8 ml min
1
,
airow rate (N
2
/qFlow) 357 l h
1
,N
2
pressure 80psi, and
aspiration rate 35 m
3
h
1
. Sixty-one percent, 67% and
77% of SDL + Cipro, SDL + MNP, and SDL + MNP +
Cipro respectively, were recovered after spray drying.
Exposure of biolms to magnetic elds
Biolm formation and treatment
P. aeruginosa biolms were grown for 24 h in sterile
6-well plates and tissue culture treated Petri dishes ( for
subsequent magneTherm (nanoTherics Ltd, Newcastle-
under-Lyme, UK) treatment) as described above.
Twenty-four-hour biolms were washed twice with sterile
PBS and the physical mixtures of SDL, SDL + Cipro,
SDL + MNP, SDL + MNP + Cipro were applied to the
biolm using an aerosol dosing method (Figure 1). A dry
powder insufator (Model DP-4 M, Penn-Century Inc.,
Wyndmoor, PA, USA) was used. The insufator was
loaded with known weights of spray-dried formulations
and the compound was delivered to a plate. The loaded
formulations were weighed and the amounts of Cipro
delivered were calculated. Subsequently, the initial load-
ing weight of the formulation, and the number of deliver-
ies needed in order to deposit a desired concentration of
Cipro, were calculated. Final Cipro deposition was esti-
mated to be 1 μgml
1
. Although P. aeruginosa is known
not to metabolize SDL, it is commonly used as an excipi-
ent in inhalational drug formulations (Hugh & Leifson
1953; McGowan 2006; Ungaro et al. 2012); therefore,
SDL was used as the control. Subsequently, the plates
were exposed to one of four different magnetic eld treat-
ments. Static magnetic eld treatments involved exposing
the 24 h biolm, with or without spray dried particles, to
magnetic elds from the bottom of a 6-well plate for 6 h
using molybdenum magnets. The strength of the magnetic
eld was measured by a Gaussmeter (FW BELL 5180
Gauss/Tesla meter, Stanford Magnets, Irvine, CA, USA).
Figure 1. The method of biolm treatment with different
particles. A dry powder insufator (Model DP-4 M) was used
and the nal Cipro deposition was estimated to be 1 μgml
1
.
Biofouling 445
Downloaded by [104.249.167.39] at 13:44 15 December 2015
The well was positioned over the magnet and the probe
was placed on the oor of the well when taking the mea-
surements. The magnetic eld strength at the biolm was
4.44 kG (Figure 2A). Switched static magnetic eld treat-
ment involved exposing the 24 h biolm, with or without
spray dried particles, to magnetic elds from the bottom
of the 6-well plates for 30 min (the magnetic eld strength
was 4.44 kG) followed by exposing from the top of the 6-
well plate for 30 min (the magnet was positioned over the
lid of the well and the probe was placed at a similar dis-
tance between the lid and the oor of the well when tak-
ing the measurements; the magnetic eld strength was
0.12 kG) for a total exposure period of 6 h (Figure 2B).
The oscillating magnetic eld treatment involved expos-
ing the 24 h biolm, with or without spray-dried particles,
to magnetic elds generated by magneTherm (biolms in
Petri dishes, 17 turn coil of 474 kHz nominal frequency,
current supply: 20 V, 4.3 A, magneTherm), for 30 min
(Figure 2C). The static plus oscillating (Static + oscillat-
ing) magnetic eld treatment involved exposing the 24 h
biolm, with or without spray-dried particles, to static
magnetic elds as mentioned above for 1 h followed by a
30 min exposure to oscillating magnetic elds (Figure 2D).
After the magnetic eld treatments, biolms were incu-
bated in an 80% humidied incubator for 24 h at 37°C
(Figure 2D). At the end of the incubation period, the bio-
lms were washed twice with PBS; XTT reduction assays
were performed to quantify the viability of biolms by
means of measuring metabolic activity, and crystal violet
assays were used to quantify biolm biomass.
XTT reduction assay
At the end of the incubation of both test and control
biolms, a standard XTT reduction assay was per-
formed as described by Bandara, Lam et al. (2010)to
measure the viability of biolms by means of bacterial
cell metabolic activity. In brief, commercially available
XTT powder (Sigma Aldrich) was dissolved in PBS to
anal concentration of 1 mg ml
1
. Then the solution
was lter-sterilized (0.22 μm pore size lter) and stored
at 70°C. Freshly prepared 0.4 mM menadione solution
was used for the XTT reduction assay. XTT solution
was thawed and mixed with menadione solution at 20:1
(v/v) immediately before the assay. Thereafter, PBS:
XTT:menadione in 79:20:1 proportions were added to
each culture dish containing biolms and incubated in
the dark for 5 h at 37°C. The color changes were mea-
sured with a microtiter plate reader (Innite M200
microplate reader, TECAN US Inc., Morrisville, NC,
USA) at 492 nm. All assays were carried out in
quadruplicates on three different occasions.
Crystal violet (CV) assay
At the end of the incubation of both test and control bio-
lms, CV assays were performed to quantify biolm bio-
mass. Biolms were carefully washed twice with PBS
and stained with a 1% CV solution for 15 min at 25°C
without shaking. Wells were carefully washed three
times with PBS to remove excess stain and air dried at
Figure 2. Methods of exposure of P. aeruginosa biolm to different magnetic elds. (A) Static one sided magnetic eld exposure;
magnets were placed under the 6-well plates for 6 h. (B) Static switched magnetic eld exposure; the locations of the magnets were
switched from top to bottom and vice versa every 30 min for 6 h. (C) Oscillating magnetic eld exposure; biolms were exposed to
oscillating magnetic elds for 30 min using magneTherm. (D) Static + oscillating magnetic eld exposure; biolms were exposed to
static magnetic eld as shown in (A) for 1 h followed by oscillating magnetic elds for 30 min using magneTherm.
446 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
room temperature. Thirty percent acetic acid was added
to the wells containing stained biolms before incubation
for 20 min at 25°C. The solution was transferred to a
new well plate and the optical density was measured at
570 nm. All assays were carried out in quadruplicates on
three different occasions.
Confocal laser scanning microscopy (CLSM)
Biolms were prepared on sterile coverslips placed in
commercially available sterile at bottom six well plates
(Nunclon, Nunc, Thermo Fisher Scientic, Waltham, MA,
USA) as described above. Pre-formed 24 h biolms were
exposed to magnetic elds and incubated for another 24 h
at 37°C in a humidied incubator. At the end of incuba-
tion, the prewashed coverslips were stained with Live and
Dead stain (Live/Dead BacLight Bacterial Viability kit,
Invitrogen, Eugene, OR, USA) (Bandara, Yau et al. 2010).
Component A (cyto 9, excitation/emission; 480/500 nm)
and Component B (propidium iodide, excitation/emission;
490/635 nm) were mixed (1:1) and 1 μl of dye mixture
was added to the well containing 1 ml of PBS and the
coverslip. The plate was incubated in the dark at room
temperature for 15 min. The biolm was then analyzed by
uorescent microscopy using a confocal laser scanning
microscope (Leica TCS SP5 II upright microscope, Leica,
Wetzlar, Germany). For each coverslip, nine microscope
elds (three vertical elds × three horizontal elds) were
imaged and in the experiment and images were repeated
on two different occasions.
Statistical analysis
Statistical analysis was performed using SPSS software
(version 16.0). A MannWhitney U-test was performed to
compare the signicance of differences between the corre-
sponding control and test samples of the P. aeruginosa
biolms and to compare the signicance of differences
between test samples of the P. aeruginosa biolms under
different treatment conditions. A p-value < 0.05 was
considered statistically signicant.
Results
The MIC of ciprooxacin was 0.125 μgml
1
for plank-
tonic P. aeruginosa and the MBC was 16 μgml
1
for
P. aeruginosa biolms formed in 24 h.
The effects of magnetic elds on P. aeruginosa biolms
treated with control particles
Biolm metabolism (XTT reduction assay )
When the biolms were treated with SDL control parti-
cles and with various magnetic elds as mentioned
above, all the test samples exposed to magnetic elds
exhibited a signicant reduction in metabolic activity
when compared to the untreated (ie magnetic eld free)
biolm controls (p< 0.05, Figure 3A). Comparing the
different magnetic eld treatments to each other, there
were no signicant differences in the mean XTT
readings.
Biolm biomass (CV assay )
When the biolms were treated with SDL and exposed
to one of the four different magnetic elds, all treatments
showed a signicant reduction in biomass compared to
the no magnetic eld control (p< 0.05, Figure 3B). Bio-
lms exposed to static switched, oscillating and static +
oscillating magnetic elds showed a signicantly lower
biomass compared to the biolms exposed to a static
magnetic eld (p< 0.05, Figure 3B). Biolms exposed
to a static switched magnetic eld had a signicantly
lower biomass compared with those exposed to the oscil-
lating and static + oscillating magnetic elds (p< 0.05,
Figure 3B).
CLSM
The control biolm (Figure 3C) that was not exposed to
magnetic elds demonstrated a dense, spatially oriented
and conuent biolm and a typical live to dead ratio for
a 24 h biolm. The biolms exposed to static magnetic
elds exhibited a signicantly lower quantity of bacterial
cells and there was no organized structure (Figure 3D).
However, the apparent ratio of live to dead cells did not
show signicant differences compared to the control
(Figure 3D). Similar reductions in bacterial count were
observed in the biolms exposed to static switched mag-
netic elds (Figure 3E). However, remnants of the bio-
lm structure were preserved, and isolated islands were
observed. The live/dead cell ratio in the biolms exposed
to static switched magnetic elds appeared to be similar
to the control (Figure 3E).
Exposure to an oscillating magnetic eld caused
complete disruption of the biolm (Figure 3F). There
was no structured biolm observed in any of the
CLSM images. Instead, scattered bacterial cells were
visible in the microscope with apparently higher pro-
portions of dead cells compared to the control biolm
(Figure 3F). The biolm treated with a static + oscillat-
ing magnetic eld also exhibited a signicantly dis-
rupted biolm architecture and haphazardly distributed
bacterial cells (Figure 3G). No comparisons were made
between the XTT and microscope ndings due to the
qualitative and denitive nature of the CLSM imaging
assessment.
Biofouling 447
Downloaded by [104.249.167.39] at 13:44 15 December 2015
(A) (B)
(C) (D) (E)
(G)(F)
Figure 3. The effects of various magnetic elds on P. aeruginosa biolms. (A) The effect of various magnetic elds on SDL-treated
biolms XTT reduction assay ndings; note the signicant reduction in biolm metabolism (mean XTT values) when exposed to
any of the magnetic elds compared to the unexposed control. (B) The effect of various magnetic elds on SDL-treated biolms
CV assay ndings; note that the most signicant reduction in the biomass (mean OD) was when exposed to static switched magnetic
elds. * Indicates signicant changes and p< 0.05 is considered to be statistically signicant (n= 12). (CG) CLSM images of
P. aeruginosa biolms exposed to different magnetic elds (stained using the LIVE/DEAD BacLight bacterial viability kit; Invitro-
gen); live cells are stained green and dead cells red. (C) Undisturbed control. (D) Exposed to static one sided magnetic elds. (E)
Exposed to static alternating magnetic elds. (F) Exposed to oscillating magnetic elds. (G) Exposed to static and oscillating magnetic
elds. Note the signicant reduction in the cellular content, stratied architecture and the lower live: dead cell ratio and lack of extra-
cellular components in the test biolms (D, E, F and G) compared to the three dimensionally arranged and dense biolm controls
with substantial extracellular materials.
448 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
The effects of various magnetic elds on P. aeruginosa
biolms co-treated with magnetic nanoparticles
Biolm metabolism (XTT reduction assay )
When SDL + MNP-treated biolms were exposed to the
aforementioned different magnetic elds, all test treat-
ments (except those exposed to oscillating magnetic
elds) showed a signicant suppression of biolm meta-
bolism (p< 0.05) compared to an untreated biolm con-
trol (Figure 4A). Biolms that were exposed to static
switched magnetic elds demonstrated the lowest meta-
bolic activity, and had a signicantly lowered metabolic
activity compared to the biolms exposed to the other
magnetic eld treatments (p< 0.05, Figure 4A).
Biolm biomass (CV assay )
Biolms treated with SDL + MNP and exposed to static
switched magnetic elds and oscillating magnetic elds
showed a signicant reduction in their biomass compared
to the controls and compared to the other magnetic eld
treatments (p< 0.05, Figure 4B). Conversely, biolms
exposed to static magnetic elds showed a signicant
increase in biomass compared to the SDL + MNP-treated
controls (p< 0.05, Figure 4B).
The effects of various magnetic elds on P. aeruginosa
biolms co-treated with ciprooxacin
Biolm metabolism (XTT reduction assay )
When P. aeruginosa biolms were treated with SDL +
Cipro and exposed to the aforementioned different mag-
netic elds, all the test biolms except the one exposed
to the static + oscillating magnetic elds exhibited a
signicant reduction in metabolism compared to the
untreated biolm controls (p< 0.05, Figure 5A).
Biolm biomass (CV assay )
Biolms that were co-treated with Cipro and exposed to
static switched, oscillating, and static + oscillating mag-
netic elds exhibited a signicantly lower biomass when
compared to the controls and to the static eld treatment
(p< 0.05, Figure 5B).
The effects of various magnetic elds on P. aeruginosa
biolms co-treated with both magnetic nanoparticles
and ciprooxacin
Biolm metabolism (XTT reduction assay )
When P. aeruginosa biolms were treated with SDL +
MNP + Cipro and exposed to static switched magnetic
elds, a signicant decrease in metabolism was observed
compared to the untreated biolm controls and the other
magnetic eld treatments (p< 0.05, Figure 6A). In con-
trast, biolms treated with SDL + MNP + Cipro, and
exposed to other magnetic elds, did not signicantly
differ from the controls.
Biolm biomass (CV assay )
The outcome of the exposure of SDL + MNP + Cipro-
treated biolms to different magnetic elds was com-
pared. All the biolms exposed to magnetic elds showed
a signicantly reduced biomass compared to the unex-
posed sample (p< 0.05, Figure 6B). When compared
Figure 4. The effects of various magnetic elds on P. aeruginosa biolms treated with magnetic nanoparticles. (A) The effect of
various magnetic elds on SDL + MNP-treated biolms XTT reduction assay ndings; note the most signicant reduction of bio-
lm metabolism (mean XTT values) when exposed to static switched the magnetic elds compared to the unexposed control and
other exposed biolms. (B) The effect of various magnetic elds on SDL + MNP-treated biolms CV assay ndings; note that the
most signicant reduction in the biomass (mean OD) was when exposed to static switched magnetic elds. * Indicates signicant
changes and p< 0.05 is considered statistically signicant (n= 12).
Biofouling 449
Downloaded by [104.249.167.39] at 13:44 15 December 2015
with the biolm exposed to a static magnetic eld, the
remaining three biolms exposed to different magnetic
elds showed a signicant reduction in overall biomass
(p< 0.05, Figure 6B). There were no signicant changes
among other comparisons.
The effects of various particle treatments on P.
aeruginosa biolms
Biolm metabolism (XTT reduction assay )
When considering the different particle treatments alone
(without magnetic exposure), only SDL + MNP + Cipro-
treated biolms showed a signicantly lower metabolic
activity compared to the SDL-treated control (p< 0.05,
Figure 7A).
In contrast, when different particle treatments were
exposed to one of the magnetic elds, there were no
signicant changes observed in any tested biolms
compared to their respective controls.
Biolm biomass (CV assay )
A signicant reduction in the biomass of all three test P.
aeruginosa biolms (treated with SDL + MNP, SDL +
Cipro and SDL + MNP + Cipro) that were not exposed
to test magnetic elds was observed compared to biolm
treated with SDL (p< 0.05, Figure 7B).
Figure 5. The effects of various magnetic elds on P. aeruginosa biolms treated with ciprooxacin. (A) The effect of various mag-
netic elds on SDL + Cipro-treated biolms XTT reduction assay ndings; note the most signicant reduction of biolm metabolism
(mean XTT values) when exposed to static switched the magnetic elds compared to the unexposed control and other exposed bio-
lms. (B) The effect of various magnetic elds on SDL + Cipro-treated biolms CV assay ndings; note that the most signicant
reduction of the biomass (mean OD) was when exposed to static switched magnetic elds. * Indicates signicant changes and p< 0.05
is considered statistically signicant (n= 12).
Figure 6. The effects of various magnetic elds on P. aeruginosa biolms treated with magnetic nanoparticles and ciprooxacin.
(A) The effect of various magnetic elds on SDL + MNP + Cipro-treated biolms XTT reduction assay ndings; note the most sig-
nicant reduction of biolm metabolism (mean XTT values) when exposed to static switched magnetic elds compared to the unex-
posed control and other exposed biolms. (B) The effect of various magnetic elds on SDL + MNP + Cipro-treated biolms CV
assay ndings; note that the most signicant reduction in the biomass (mean OD) was when exposed to static switched magnetic
elds. * Indicates signicant changes and p< 0.05 is considered statistically signicant (n= 12).
450 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
Discussion
Due to the refractory nature of pathogenic biolms,
many different biolm eradication strategies and new
therapeutic agents have been investigated. Some strate-
gies have used chemical approaches such as anti-
adhesins/polymeric coatings (Cassinelli et al. 2000; van
de Belt et al. 2001; von Bismarck et al. 2001; John
et al. 2007; Johansson et al. 2008), quorum sensing
inhibitors (Francolini et al. 2004; Cirioni et al. 2006;
Antoniani et al. 2010), biosynthesis inhibitors (Ueda
et al. 2009), biolm dispersal agents (Raad et al. 2007;
Kim et al. 2008; Barraud et al. 2009; Cai et al. 2009;
Davies & Marques 2009) and natural products
(Bjarnsholt et al. 2007; Kalishwaralal et al. 2010;
Kostenko et al. 2010; Wu et al. 2011), among others.
Physical methods have also been investigated, including
ultrasound waves (Ensing et al. 2005), low frequency
vibrations (Bandara et al. 2014) and other methods (Di
Poto et al. 2009).
Unlike previous reports, the current study sought to
evaluate the potential for eradication of biolms using
a combination of chemical and physical approaches.
Nanoparticles, a test antibacterial compound, and mag-
netic elds were systematically evaluated to assess the
relative anti-biolm contributions of each approach and
combinations thereof. The following discussion is orga-
nized in increasing order of complexity with the initial
focus on the separate treatments followed by discus-
sion of the treatment combinations. Interestingly, the
most effective treatment conditions observed in the
present study were obtained using magnetic elds
alone.
Magnetic elds alone disrupt P. aeruginosa biolms
Signicant reductions in biolm metabolic activity and
biomass were observed when different magnetic elds
were applied in the absence of nanoparticle treatments.
The highest suppression of viability and biomass was
observed in the biolm exposed to a static switched
eld. The reduction in cell viability must be attributed to
bacterial cell death and the reduction in biomass must be
related to both bacterial death and loss of the extracellu-
lar matrix of the biolm. Furthermore, ultrastructural
views conrmed the severe destructive effects of mag-
netic elds on the architecture of P. aeruginosa biolms.
Thus, the magnetic elds appear to negatively affect the
structure, cell viability and extracellular materials (either
their synthesis or direct disruption) of mature P. aerugi-
nosa biolms.
There are several early reports on the effects of mag-
netic elds on bacterial growth (Kohno et al. 2000; Piatti
et al. 2002; Potenza et al. 2004; Gao et al. 2005; Laszlo
& Kutasi 2010). Most previous studies were conducted
using static magnetic elds ranging from 30 mT to 14.1
T, and exposing from 30 min to six days, using a variety
of microbes and growth conditions. After the exposure
to static magnetic elds, E. coli,Staphylococcus aureus,
S. mutans,Rubus fruticosus,Shewanella oneidensis,Sac-
charomyces cerevisiae,Bacillus circulans,Micrococcus
luteus,Pseudomonas uorescens,Salmonella enteritidis,
and Serratia marcescens did not exhibit any signicant
changes in their growth (Grosman et al. 1992; Piatti
et al. 2002; Potenza et al. 2004; Gao et al. 2005; Laszlo
& Kutasi 2010). The present results, in comparison,
demonstrated that static magnetic elds had a signicant
Figure 7. The effects of various particle treatments on P. aeruginosa biolms. (A) Particle treatment of biolms with no magnetic
exposure biolms XTT reduction assay ndings; note the signicantly low metabolic activity of the biolm treated with SDL +
MNP + Cipro compared to the SDL-treated biolm. (B) Particle treatment of biolms with no magnetic exposure CV assay nd-
ings; note the signicantly low biomass of the biolm treated with all particle combinations compared to the SDL-treated biolm. *
Indicates signicant changes and p< 0.05 is considered statistically signicant (n= 12).
Biofouling 451
Downloaded by [104.249.167.39] at 13:44 15 December 2015
inhibitory effect on P. aeruginosa biolm viability and
biomass. In line with the present results, Piatti et al.
(2002) also noted that there was a signicant growth
inhibition of S. marcescens and Hordeum vulgare when
exposed to static magnetic elds for 24 h. It should be
noted that previously reported studies were conducted in
planktonic bacterial cultures, thus the extrapolation of
these ndings to a biolm environment must be made
with caution.
As yet, few mechanisms have been identied by
which magnetic elds may inuence microorganism via-
bility and growth. However, magnetic eld effects are
likely dependent upon many variables including species,
strain, exposure time, the strength of the magnetic eld
and the growth environment (Bajpai & Basu 2013). For
instance, to elicit growth inhibition using static magnetic
elds in S. aureus,S. mutans and E. coli they must be
grown under anaerobic conditions (Kohno et al. 2000).
The present authors did not study the effects of the vari-
ous factors that inuence the effects of magnetic elds
on biolms, such as magnetic eld strength or time of
eld application. Thus, the discrepancies between the
present study and previous reports may be due to differ-
ences in any of the aforementioned conditions. In addi-
tion, magnetic elds may interact with iron related
cellular processes (Schwartz et al. 1989; Stepanian et al.
2000; Zhang et al. 2002). P. aeruginosa also depends on
iron for virulence, alginate production, mucoid pheno-
type change and biolm formation (Banin et al. 2005;
Wiens et al. 2014). Although the exact mechanism is yet
to be elucidated, the reduction in the biolm biomass
and the viability of P. aeruginosa in the current study
may also be related to the blockage of iron acquisition
enzymes, as shown previously in E. coli K-12 and
Mycobacterium tuberculosis (Schwartz et al. 1989;
Stepanian et al. 2000; Zhang et al. 2002).
Although there are no reports of the usage of oscillat-
ing magnetic elds in controlling animal pathogens,
these have been suggested as a disinfection method in
agricultural products and food due to their signicant
inhibitory effects on plant bacterial pathogens (Lipiec
et al. 2004). Interestingly, the current study also clearly
indicates that P. aeruginosa biolms can be signicantly
disrupted through exposure to oscillating magnetic elds,
as well as by a combination of static and oscillating
magnetic elds. The aforementioned agricultural studies
were not conducted in biolm environments, thus direct
comparisons between these studies cannot be made.
There are no reports to the authorsknowledge that
describe the potential mechanism by which magnetic
elds control P. aeruginosa biolms. According to an
investigation of global gene expression conducted by
Sandvik (2013), extremely low-frequency magnetic elds
up-regulated transposase activity, membrane transport
processes, and signal transduction systems in a frequency-
dependent manner, suggesting that magnetic elds
induced changes in ion transport in P. aeruginosa.
MNPs alone do not affect the viability of P. aeruginosa
biolms
The major advantage of using nanoparticles is that the
target area can be precisely located and the subsequent
release of the drug can be pre-planned (Francolini &
Donelli 2010). MNPs can be precisely controlled by a
magnetic eld and driven to the specic location in the
body for the desired action (Corchero & Villaverde
2009, Xie et al. 2009). Previously, MNPs have been suc-
cessfully used in targeting cancer cells, imaging and drug
delivery (Gindy & Prudhomme 2009).
Recent studies have suggested that MNP may possess
potential antimicrobial properties depending on the
nanoparticle type, dose and the pathogen (Taylor & Webster
2009). For instance, iron oxide with silver nanoparticles
showed good antimicrobial properties against E. coli,S. epi-
dermidis,Bacillus subtilis and MRSA (methicillin resistant
S. aureus)biolms (Kvitek & Soukupova 2009; Durmus &
Webste r 2013) but not against genetically engineered P.
aeruginosa PTSOX4 (Kafayati et al. 2012). The antimicro-
bial activity of nanoparticles in previous studies was sug-
gested to be due to their high surface-area-to-volume ratio,
which may be accompanied by the enhanced synthesis of
reactive oxygen species (ROS) and free radicals (Nel et al.
2009;Allaker2010;Mahmoudietal.2011). Thus,
nanoparticles may interact with microbial membranes and
kill them by damaging the microbial structure (Nel et al.
2009;Allaker2010;Mahmoudietal.2011). The disruption
of cell membrane and cytoplasmic leakage in response to
chitosan nanoparticles has been previously observed by
atomic force microscopy (Qi et al. 2004)
Consistent with the aforementioned previous reports
on P. aeruginosa PTSOX4, it is also reported here that
MNP alone had no signicant effects on the metabolism
of P. aeruginosa biolms. However, a signicant
reduction in the biomass of the biolm was observed
(Figure 4). The differential effect of MNP on biolm
viability and biomass suggests that the dose of MNP
needed to induce bacterial lysis may be higher than that
needed to disrupt the extra cellular matrix. In addition,
there are concerns about the use of MNP in biolm elim-
ination, as the exposure of P. aeruginosa biolms to
SPIONs at concentrations up to 200 μgml
1
resulted in
an increase in biolm biomass and cell density (Haney
et al. 2012). Similar positive effects were not observed
in the present study, most likely because MNP concen-
trations that were estimated to be several-fold lower than
200 μgml
1
were used. Nevertheless, as described in the
current literature, the usage of MNPs has been mainly
452 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
investigated in planktonic microorganisms and the possi-
ble prevention of biolm formation (Taylor & Webster
2009; Anghel et al. 2013; Wang et al. 2013). Thus, more
studies are necessary to explore the properties of MNPs
for use in the eradication of existing biolms.
MNPs signicantly disrupt P. aeruginosa biolms in
the presence of static switched magnetic elds
The uptake of nanoparticles by biolms is usually low
due to their poor diffusion and penetration properties
(Fabrega et al. 2009; Liu et al. 2009; Gunawan et al.
2011; Raghupathi et al. 2011, Cui et al. 2012). How-
ever, nanoparticles that possess magnetic properties, eg
SPIONs, chitosan coated SPIONs and silver nanoparti-
cles, have been shown to overcome this limitation. In the
presence of magnetic elds, these nanoparticles can be
targeted to the infection site, and disrupt and eradicate
pathogenic biolms (Lee et al. 2005; McGill et al. 2009;
Armijo, Brandt, Matthew et al. 2012; Armijo, Brandt,
Rivera et al. 2012; Chen et al. 2012). For instance,
Durmus and Webster (2013) reported that the exposure
of SPIONs to static magnetic elds further increased
their antibacterial properties against MRSA compared to
unexposed controls (Durmus & Webster 2013). Similarly,
the exposure of P. aeruginosa biolms treated with
SPIONs to static switched magnetic elds caused a sig-
nicant reduction in biolm metabolism and biomass.
The use of static one sided magnetic exposure failed to
elicit similar results, and displayed an increase in the
biomass possibly due to the additive effect of MNP mass
to the existing biolm mass. This suggests that static
switched magnetic elds may be more effective in direct-
ing MNPs to a target site and in destroying established
biolms when compared to static magnetic elds. The
latter nding further conrms the results obtained by
Park et al. (2011). They also reported > 4 log inactiva-
tion of P. aeruginosa PA01 biolm after treatment for
8 min with SPIONs and a magnetic eld. The treatment
caused a substantial disintegration of the bacterial cell
membrane in the biolm and was suggested to be purely
due to the thermal effect generated by MNPs and the
magnetic eld (Park et al. 2011).
Exposure of Pseudomonas uorescens to iron oxide
magnetic nanoparticles and an oscillating magnetic eld
of 873 kHz/100 Oe (Oersteds) resulted in a complete
eradication of planktonic bacteria and a signicant
reduction in biolm viability at 8 min exposure, at 55°C
(Rodrigues et al. 2012). However, the complete eradica-
tion of a biolm was not seen even after 17 min expo-
sure at 60°C (Rodrigues et al. 2012). In contrast, when
P. aeruginosa biolms were treated with MNP and
exposed to oscillating magnetic elds, no signicant
changes in biolm viability were observed. However,
biomass was signicantly reduced. The difference in the
outcome could be due to a lower dose of MNP and/or
magnetic eld and/or the temperature used. Nevertheless,
the present ndings again suggest that the dose of MNP
needed for bacterial cell lysis may be higher than that
needed for matrix disruption.
Ciprooxacin signicantly eliminates P. aeruginosa
biolms in the presence of static switched magnetic
elds
Early research conducted in the 1990s suggested the
possibility of using magnetic elds as a treatment option
to improve the efciency of antimicrobials on biolms
formed on various medical devices (Khoury et al. 1992;
Benson et al. 1994). Benson et al. (1994) reported that
the application of static magnetic elds (510 G),
together with gentamycin, signicantly reduced the
P. aeruginosa counts on various biomaterial surfaces.
Thus they suggested that static magnetic elds may
enhance the activity of gentamycin against P. aeruginosa
biolms by forming radical intermediates and the
up-regulation of the bacterial membrane protein, porins
(Benson et al. 1994). In another study, cephalothin-
resistant P. aeruginosa exhibited re-sensitization to
cephalothin after exposure to a static electromagnetic
eld (0.35 Tesla of electromagnetic eld for 15 min)
(Samarbaf-Zadeh et al. 2006). Similarly, in the present
study, ciprooxacin-treated P. aeruginosa biolms
exposed to static switched magnetic elds exhibited the
most signicant inhibitory effects on biolm metabolism
and biomass. Static one sided and oscillating magnetic
eld treatment regimens also exhibited less-signicant
ndings. It can be speculated that static switched mag-
netic elds disrupt the biolm matrix more efciently
than static one sided magnetic elds, providing cipro-
oxacin with better access to deeper layers of the bio-
lm. Further investigations are necessary to explain the
mechanism of bacterial killing and biolm reduction
mediated by ciprooxacin with assistance from static
switched magnetic elds.
Magnetic nanoparticles assist ciprooxacin in biolm
killing
In the latter part of the study P. aeruginosa biolms were
treated with MNP, ciprooxacin and magnetic elds. To
the authorsknowledge, there are no reports in the litera-
ture of simultaneous application of all three treatment
modalities to bacterial biolms. However, several studies
have reported that co-administration of antibiotics with
nanoparticles resulted in better inhibitory effects on bio-
lms. For instance, Cotar et al. (2013) reported that
MNPs conjugated to polymixin B, streptomycin and cefo-
taxime exhibited a dose-dependent signicant suppression
of biolm formation by P. aeruginosa and E. coli as
Biofouling 453
Downloaded by [104.249.167.39] at 13:44 15 December 2015
estimated by the total biolm biomass. Armijo, Brandt,
Rivera et al. (2012) suggested that when administered
together, SPIONs capped with PEG (polyethylene glycol)
may contribute to a better distribution of antibiotics in
P. aeruginosa biolms in cystic brosis lungs, due to
their superior penetrating properties. Furthermore, Au and
Ag nanoparticles functionalized with ampicillin were also
shown to be effective broad-spectrum bactericides against
both Gram-negative and Gram-positive bacteria (Brown
et al. 2012). The present results also conrm the supe-
rior inhibitory effects of MNP and antibiotics on
P. aeruginosa biolms. SDL + MNP + Cipro-treated
P. aeruginosa biolms, even without exposure to mag-
netic elds, showed signicantly lower metabolism and
biomass compared to SDL-treated control biolms.
However, when biolms were treated with MNP and
ciprooxacin individually (SDL + MNP and SDL +
Cipro), no signicant results were observed (despite a
signicant reduction in their biomass). This clearly indi-
cates that the combined treatment of MNP and ciproox-
acin is more effective for eradicating P. aeruginosa
biolms than their individual effects.
Interestingly, when the combined treatment of SDL +
MNP + Cipro was exposed to magnetic elds, a static
switched magnetic eld was the only magnetic exposure
that could exert a signicant reduction in biolm meta-
bolism compared to the unexposed controls, despite the
signicant reduction in biomass in all biolms except the
one exposed to static magnetic elds. Hence, when all
different treatment situations are considered, the best bio-
lm suppression was observed with a static switched
magnetic eld exposure.
Conclusions
In summary, it is reported here, for the rst time, that
mere exposure of biolms to magnetic elds resulted in
the signicant destruction and killing of P. aeruginosa
biolms. Static switched magnetic elds demonstrated
superior anti-biolm properties when applied alone or
when co-applied with MNP, Cipro or MNP + Cipro.
Thus, upon further optimization, a combined therapy of
antibiotics with MNPs and exposure to magnetic elds
can be used as a promising novel therapeutic approach/
treatment strategy for removal of pathological biolms.
Further studies are necessary to evaluate the efcacy of
these magnetic elds in the management of biolm-as-
sociated infections in soft tissues as well as on medical
devices.
The results also indicate that the combination ther-
apy of MNP and ciprooxacin demonstrates superior
properties in eliminating biolms compared to their indi-
vidual effects. In the spectrum of the dosage used,
although MNP itself did not elicit signicant effects in
respect of bacterial killing, it did reduce the biolm
mass. Exposure to magnetic elds did not show any
added advantage when treated with MNP. Hence, a
longer exposure period with stronger magnetic elds
and a superior safety margin may be required to trigger
a signicant effect.
Acknowledgements
The authors would like to thank Dr Kristin Fathe from the
University of Texas at Austin for editorial assistance.
Conict of interest disclosure statement
No potential conict of interest was reported by the authors.
References
Allaker RP. 2010. The use of nanoparticles to control oral bio-
lm formation. J Dent Res. 89:11751186.
Anghel I, Grumezescu AM, Holban AM, Ficai A, Anghel AG,
Chiriuc MC. 2013. Biohybrid nanostructured iron oxide
nanoparticles and Satureja hortensis to prevent fungal bio-
lm development. Int J Mol Sci. 14:1811018123.
Antoniani D, Bocci P, Maciag A, Raffaelli N, Landini P. 2010.
Monitoring of diguanylate cyclase activity and of cyclic-
di-GMP biosynthesis by whole-cell assays suitable for
high-throughput screening of biolm inhibitors. Appl
Microbiol Biotechnol. 85:10951104.
Anwar H, Strap JL, Costerton JW. 1992. Establishment of
aging biolms: possible mechanism of bacterial resistance
to antimicrobial therapy. Antimicrob Agents Chemother.
36:13471351.
Armijo LM, Brandt YI, Matthew D, Yadav S, Maestas S,
Rivera AC, Cook NC, Withers NJ, Smolyakov GA,
Adolphi NL, et al. 2012. Iron oxide nanocrystals for mag-
netic hyperthermia applications. Nanomaterials. 2:134146.
Armijo LM, Brandt YI, Rivera AC, Cook NC, Plumley JB,
Withers NJ, Kopciuch M, Smolyakov GA, Huber DL,
Smyth HDC, et al. 2012. Multifunctional superparamag-
netic nanoparticles for enhanced drug transport in cystic
brosis. Nanosystems in Engineering and Medicine Pro-
ceedings of the SPIE. 8548:12. doi: 10.1117/12.943621.
Bajpai I, Basu B. 2013. Strategies to prevent bacterial adhesion
on biomaterials. In Ramalingam M, Wang X, Chen G, Ma P,
Cui F-Z, editors. Biomimetics: advancing nanobiomaterials
and tissue engineering. Chapter 7. Hoboken (NJ): Wiley;
p. 163202.
Bandara HM, Harb A, Kolacny D Jr, Martins P, Smyth HD.
2014. Sound waves effectively assist Tobramycin in
elimination of Pseudomonas aeruginosa biolms in vitro.
AAPS PharmSciTech. 15:16441654.
Bandara HM, Lam OL, Watt RM, Jin LJ, Samaranayake LP.
2010. Bacterial lipopolysaccharides variably modulate
in vitro biolm formation of Candida species. J Med
Microbiol. 59:12251234.
Bandara HM, Yau JY, Watt RM, Jin LJ, Samaranayake LP.
2010. Pseudomonas aeruginosa inhibits in-vitro Candida
biolm development. BMC Microbiol. 10:125133.
Banin E, Vasil ML, Greenberg EP. 2005. Iron and Pseu-
domonas aeruginosa biolm formation. Proc Natl Acad Sci
U S A. 102:1107611081.
Barraud N, Schleheck D, Klebensberger J, Webb JS, Hassett
DJ, Rice SA, Kjelleberg S. 2009. Nitric oxide signaling in
454 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
Pseudomonas aeruginosa biolms mediates phosphodi-
esterase activity, decreased cyclic di-GMP levels, and
enhanced dispersal. J Bacteriol. 191:73337342.
Benson DE, Grissom CB, Burns GL, Mohammad SF. 1994.
Magnetic eld enhancement of antibiotic activity in
biolm forming Pseudomonas aeruginosa. ASAIO J. 40:
M371M376.
Bjarnsholt T, Kirketerp-Moller K, Kristiansen S, Phipps R,
Nielsen AK, Jensen PO, Hoiby N, Givskov M. 2007. Silver
against Pseudomonas aeruginosa biolms. APMIS.
115:921928.
Brown AN, Smith K, Samuels TA, Lu J, Obare SO, Scott ME.
2012. Nanoparticles functionalized with ampicillin
destroy multiple-antibiotic-resistant isolates of Pseu-
domonas aeruginosa and Enterobacter aerogenes and
methicillin-resistant Staphylococcus aureus. Appl Environ
Microbiol. 78:27682774.
Cai Y, Fan Y, Wang R, An MM, Liang BB. 2009. Synergistic
effects of aminoglycosides and fosfomycin on Pseu-
domonas aeruginosa in vitro and biolm infections in a rat
model. J Antimicrob Chemother. 64:563566.
Cassinelli C, Morra M, Pavesio A, Renier D. 2000. Evaluation
of interfacial properties of hyaluronan coated poly(methyl-
methacrylate) intraocular lenses. J Biomater Sci Polym
Ed.11:961977.
Chen T, Wang R, Xu LQ, Neoh KG, Kang ET. 2012.
Carboxymethyl chitosan-functionalized magnetic nanoparti-
cles for disruption of biolms of Staphylococcus aureus
and Escherichia coli. Ind Eng Chem Res. 51:1316413172.
Cirioni O, Giacometti A, Ghiselli R, DellAcqua G, Orlando F,
Mocchegiani F, Silvestri C, Licci A, Saba V, Scalise G,
et al. 2006. RNAIII-Inhibiting peptide signicantly reduces
bacterial load and enhances the effect of antibiotics in the
treatment of central venous catheterassociated Staphylo-
coccus aureus infections. J Infect Dis. 15:180186.
CLSI. 2012. Methods for dilution antimicrobial susceptibility
tests for bacteria that grow aerobically; approved standard.
9th ed. Wayne (PA): Clinical and Laboratory Standards
Institute [CLSI].
Corchero JL, Villaverde A. 2009. Biomedical applications of dis-
tally controlled magnetic nanoparticles. Trends Biotechnol.
27:468476.
Costerton JW, Stewart PS, Greenberg EP. 1999. Bacterial bio-
lms: a common cause of persistent infections. Science.
284:13181322.
Cotar AI, Grumezescu AM, Andronescu E, Voicu G, Ficai A,
Ou KL, Huang KS, Chiriuc MC. 2013. Nanotechnological
solution for improving the antibiotic efciency against bio-
lms developed by Gram-negative bacterial strains. Lett
Appl NanobioSci. 2:97104.
Cui Y, Zhao Y, Tian Y, Zhang W, Lu X, Jiang X. 2012.
The molecular mechanism of action of bactericidal
gold nanoparticles on Escherichia coli. Biomaterials.
33:23272333.
Davies DG, Marques CN. 2009. A fatty acid messenger is
responsible for inducing dispersion in microbial biolms. J
Bacteriol. 191:13931403.
Di Poto A, Sbarra MS, Provenza G, Visai L, Speziale P.
2009. The effect of photodynamic treatment com-
bined with antibiotic action or host defence mechanisms
on Staphylococcus aureus biolms. Biomaterials. 30:
31583166.
Durmus NG, Webster TJ. 2013. Eradicating antibiotic-resistant
biolms with silver-conjugated superparamagnetic iron
oxide nanoparticles. Adv Healthc Mater. 2:165171.
Ensing GT, Roeder BL, Nelson JL, Van Horn JR, Van der Mei
HC, Busscher HJ, Pitt WG. 2005. Effect of pulsed ultra-
sound in combination with gentamicin on bacterial viability
in biolms on bone cements in vivo. J Appl Microbiol.
99:443448.
Fabrega J, Renshaw JC, Lead JR. 2009. Interactions of silver
nanoparticles with Pseudomonas putida biolms. Environ
Sci Technol. 43:90049009.
Francolini I, Donelli G. 2010. Prevention and control of bio-
lm-based medical-device-related infections. FEMS Immu-
nol Med Microbiol. 59:227238.
Francolini I, Norris P, Piozzi A, Donelli G, Stoodley P. 2004.
Usnic acid, a natural antimicrobial agent able to inhibit
bacterial biolm formation on polymer surfaces. Antimi-
crob Agents Chemother. 48:43604365.
Frederiksen B, Pressler T, Hansen A, Koch C, Hoiby N. 2006.
Effect of aerosolized rhDNase (Pulmozyme) on pulmonary
colonization in patients with cystic brosis. Acta Paediatr.
95:10701074.
Gao W, Liu Y, Zhou J, Pan H. 2005. Effects of a strong static
magnetic eld on bacterium Shewanella oneidensis: an
assessment by using whole genome microarray. Bioelectro-
magnetics. 26:558563.
Gindy ME, Prudhomme RK. 2009. Multifunctional nanoparti-
cles for imaging, delivery and targeting in cancer therapy.
Expert Opin Drug Deliv. 6:865878.
Grosman Z, Kolar M, Tesarikova E. 1992. Effects of static
magnetic eld on some pathogenic microorganisms. Acta
Univ Palacki Olomuc Fac Med. 134:79.
Gunawan P, Guan C, Song X, Zhang Q, Leong SS, Tang C,
Chen Y, Chan-Park MB, Chang MW, Wang K, et al. 2011.
Hollow ber membrane decorated with Ag/MWNTs:
toward effective water disinfection and biofouling control.
ACS Nano. 5:1003310040.
Hall-Stoodley L, Costerton JW, Stoodley P. 2004. Bacterial bio-
lms: from the natural environment to infectious diseases.
Nat Rev Microbiol. 2:95108.
Haney C, Rowe JJ, Robinson JB. 2012. Spions increase biolm
formation by Pseudomonas aeruginosa. J Biomater
Nanobiotechnol. 03:508518.
Harding MW, Marques LL, Howard RJ, Olson ME. 2009. Can
lamentous fungi form biolms? Trends Microbiol.
17:475480.
Hart CA, Winstanley C. 2002. Persistent and aggressive bacte-
ria in the lungs of cystic brosis children. Br Med Bull.
61:8196.
Hidron AI, Edwards JR, Patel J, Horan TC, Sievert DM,
Pollock DA, Fridkin SK. 2008. NHSN annual update:
antimicrobial-resistant pathogens associated with health-
care-associated infections: annual summary of data reported
to the national healthcare safety network at the centers for
disease control and prevention, 20062007. Infect Control
Hosp Epidemiol. 29:9961011.
Hoiby N, Ciofu O, Bjarnsholt T. 2010. Pseudomonas
aeruginosa biolms in cystic brosis. Future Microbiol.
5:16631674.
Hoiby N, Ciofu O, Johansen HK, Song ZJ, Moser C, Jensen
PO, Molin S, Givskov M, Tolker-Nielsen T, Bjarnsholt T.
2011. The clinical impact of bacterial biolms. Int J Oral
Sci. 3:5565.
Hugh R, Leifson E. 1953. The taxonomic signicance of fer-
mentative versus oxidative metabolism of carbohydrates by
various Gram negative bacteria. J Bacteriol. 66:2426.
Johansson EM, Crusz SA, Kolomiets E, Buts L, Kadam RU,
Cacciarini M, Bartels KM, Diggle SP, Camara M, Williams
Biofouling 455
Downloaded by [104.249.167.39] at 13:44 15 December 2015
P, et al. 2008. Inhibition and dispersion of Pseudomonas
aeruginosa biolms by glycopeptide dendrimers targeting
the fucose-specic lectin LecB. Chem Biol. 15:12491257.
Johannsen M, Gneveckow U, Taymoorian K, Thiesen B,
Waldofner N, Scholz R, Jung K, Jordan A, Wust P,
Loening SA. 2007. Morbidity and quality of life during
thermotherapy using magnetic nanoparticles in locally
recurrent prostate cancer: results of a prospective phase I
trial. Int J Hyperthermia. 23:315323.
John T, Rajpurkar A, Smith G, Fairfax M, Triest J. 2007. An-
tibiotic pretreatment of hydrogel ureteral stent. J Endourol.
21:12111216.
Kafayati ME, Raheb J, Angazi MT, Alizadeh S, Bardania H.
2012. The effect of magnetic Fe3O4 nanoparticles on the
growth of genetically manipulated bacterium, Pseudomonas
aeruginosa (PTSOX4). Iran J Biotechnol. 11:4146.
Kalishwaralal K, BarathManiKanth S, Pandian SR, Deepak V,
Gurunathan S. 2010. Silver nanoparticles impede the bio-
lm formation by Pseudomonas aeruginosa and Staphylo-
coccus epidermidis. Colloids Surf B. 79:340344.
Khoury AE, Lam K, Ellis B, Costerton JW. 1992. Prevention
and control of bacterial infections associated with medical
devices. ASAIO J. 38:M174M178.
Kim J, Pitts B, Stewart PS, Camper A, Yoon J. 2008. Compar-
ison of the antimicrobial effects of chlorine, silver ion, and
tobramycin on biolm. Antimicrob Agents Chemother.
52:14461453.
Kohno M, Yamazaki M, Kimura II, Wada M. 2000. Effect of
static magnetic elds on bacteria: Streptococcus mutans, Sta-
phylococcus aureus, and Escherichia coli. Pathophysiology.
7:143148.
Kostenko V, Lyczak J, Turner K, Martinuzzi RJ. 2010. Impact
of silver-containing wound dressings on bacterial biolm
viability and susceptibility to antibiotics during prolonged
treatment. Antimicrob Agents Chemother. 54:51205131.
Kvitek L, Soukupova J. 2009. Comment on Preparation and
antibacterial activity of Fe3O4@Ag nanoparticles.
Nanotechnology. 14:028001.
Laszlo J, Kutasi J. 2010. Static magnetic eld exposure fails to
affect the viability of different bacteria strains. Bioelectro-
magnetics. 31:220225.
Lee D, Cohen RE, Rubner MF. 2005. Antibacterial properties
of Ag nanoparticle loaded multilayers and formation of
magnetically directed antibacterial microparticles. Lang-
muir. 21:96519659.
Lewis K. 2008. Multidrug tolerance of biolms and persister
cells. Curr Top Microbiol Immunol. 322:107131.
Lipiec J, Janas P, Barabasz W. 2004. Effect of oscillating mag-
netic eld pulses on the survival of selected microorgan-
isms. Int Agrophysics. 18:325328.
Liu S, Wei L, Hao L, Fang N, Chang MW, Xu R, Yang Y,
Chen Y. 2009. Sharper and faster nano dartskill more
bacteria: a study of antibacterial activity of individually dis-
persed pristine single-walled carbon nanotube. ACS Nano.
3:38913902.
Lode H, Raffenberg M, Erbes R, Geerdes-Fenge H, Mauch H.
2000. Nosocomial pneumonia: epidemiology, pathogenesis,
diagnosis, treatment and prevention. Curr Opin Infect Dis.
13:377384.
Lyczak JB, Cannon CL, Pier GB. 2000. Establishment of Pseu-
domonas aeruginosa infection: lessons from a versatile
opportunist. Microbes Infect. 2:10511060.
Mahmoudi M, Lynch I, Ejtehadi MR, Monopoli MP, Bombelli
FB, Laurent S. 2011. Proteinnanoparticle interactions:
opportunities and challenges. Chem Rev. 111:56105637.
McGill SL, Cuylear C, Adolphi NL, Osinski M, Smyth HDC.
2009. Enhanced drug transport through alginate biolms
using magnetic nanoparticles. SPIE international sympo-
sium on biomedical optics BiOS. doi: 10.1117/12.816830.
McGowan JE Jr. 2006. Resistance in nonfermenting Gram-
negative bacteria: multidrug resistance to the maximum.
Am J Infect Control. 34:S29-37; discussion S64-73.
Nel AE, Madler L, Velegol D, Xia T, Hoek EM, Somasundaran
P, Klaessig F, Castranova V, Thompson M. 2009. Under-
standing biophysicochemical interactions at the nano-bio
interface. Nat Mater. 8:543557.
Park H, Park HJ, Kim JA, Lee SH, Kim JH, Yoon J, Park TH.
2011. Inactivation of Pseudomonas aeruginosa PA01 bio-
lms by hyperthermia using superparamagnetic nanoparti-
cles. J Microbiol Methods. 84:4145.
Piatti E, Albertini MC, Baffone W, Fraternale D, Citterio B,
Piacentini MP, Dacha M, Vetrano F, Accorsi A. 2002. An-
tibacterial effect of a magnetic eld on Serratia marcescens
and related virulence to Hordeum vulgare and Rubus
fruticosus callus cells. Comp Biochem Physiol B Biochem
Mol Biol. 132:359365.
Potenza L, Ubaldi L, De Sanctis R, De Bellis R, Cucchiarini L,
Dacha M. 2004. Effects of a static magnetic eld on cell
growth and gene expression in Escherichia coli. Mutat
Res. 561:5362.
Qi L, Xu Z, Jiang X, Hu C, Zou X. 2004. Preparation and
antibacterial activity of chitosan nanoparticles. Carbohydr
Res. 339:26932700.
Raad I, Hanna H, Jiang Y, Dvorak T, Reitzel R, Chaiban G,
Sherertz R, Hachem R. 2007. Comparative activities of
daptomycin, linezolid, and tigecycline against catheter-
related methicillin-resistant Staphylococcus bacteremic iso-
lates embedded in biolm. Antimicrob Agents Chemother.
51:16561660.
Raghupathi KR, Koodali RT, Manna AC. 2011. Size-dependent
bacterial growth inhibition and mechanism of antibacterial
activity of zinc oxide nanoparticles. Langmuir. 27:
40204028.
Rodrigues D, Bañobre-López M, Espiña B, Rivas J, Azeredo J
2012. Control of planktonic bacterial cells and biolms
through magnetic hyperthermia. Paper presented at:
Biolms 5 - International Conference Paris, France. 142
143. Available from: http://repositorium.sdum.uminho.pt/bit
stream/1822/23908/5/205.pdf.
Romling U, Balsalobre C. 2012. Biolm infections, their resili-
ence to therapy and innovative treatment strategies. J Intern
Med. 272:541561.
Saltzstein D, Wachs B, Perroncel R, Benson A, Herrington J,
Haverstock D, Pertel P. 2007. Complicated urinary tract
infections treated with extended-release ciprooxacin with
emphasis on Pseudomonas aeruginosa. J Chemother.
19:694702.
Samaranayake LP. 2006. Essential microbiology for dentistry.
Edinburgh: Churchill Livingstone.
Samarbaf-Zadeh AR, Moosavi R, Tahmasbi-Birgani MJ, Darki
H. 2006. The effect of static electromagnetic eld on
cephalothin-resistant Pseudomonas aeroginosa. Jundishapur
J Natural Pharm Prod. 1:1317.
Sandvik EL. 2013. Electric current and magnetic eld effects
on bacterial biolms [PhD Thesis]. Bozeman: Montana
State University.
Schwartz E, Svejnochova M, Siposova E. 1989. Different
effects of a weak static and a weak rotating magnetic eld
on the growth of mycobacteria. Bratisl Lek Listy. 90:
787792.
456 H.M.H.N. Bandara et al.
Downloaded by [104.249.167.39] at 13:44 15 December 2015
Soenen SJ, De Cuyper M. 2010. Assessing iron oxide
nanoparticle toxicity in vitro: current status and future pro-
spects. Nanomedicine (Lond). 5:12611275.
Stepanian RS, Barsegian AA, Alaverdian ZhR, Oganesian GG,
Markosian LS, Airapetian SN. 2000. The effect of mag-
netic elds on the growth and division of the lon mutant of
Escherichia coli K-12. Radiats Biol Radioecol. 40:
319322.
Subbiahdoss G, ShariS, Grijpma DW, Laurent S, van der
Mei HC, Mahmoudi M, Busscher HJ. 2012. Magnetic tar-
geting of surface-modied superparamagnetic iron oxide
nanoparticles yields antibacterial efcacy against biolms
of gentamicin-resistant staphylococci. Acta Biomater.
8:20472055.
Taylor EN, Kummer KM, Durmus NG, Leuba K, Tarquinio
KM, Webster TJ. 2012. Superparamagnetic iron oxide
nanoparticles (SPION) for the treatment of antibiotic-resis-
tant biolms. Small. 8:30163027.
Taylor EN, Webster TJ. 2009. The use of superparamagnetic
nanoparticles for prosthetic biolm prevention. Int J
Nanomedicine. 4:145152.
Ueda A, Attila C, Whiteley M, Wood TK. 2009. Uracil
inuences quorum sensing and biolm formation in
Pseudomonas aeruginosa and uorouracil is an antagonist.
Microb Biotechnol. 2:6274.
Ungaro F, dAngelo I, Coletta C, dEmmanuele di Villa Bianca
R, Sorrentino R, Perfetto B, Tufano MA, Miro A, La
Rotonda MI, Quaglia F. 2012. Dry powders based on
PLGA nanoparticles for pulmonary delivery of antibiotics:
modulation of encapsulation efciency, release rate and
lung deposition pattern by hydrophilic polymers. J Control
Release. 10:149159.
van de Belt H, Neut D, Schenk W, van Horn JR, van der Mei
HC, Busscher HJ. 2001. Infection of orthopedic implants
and the use of antibiotic-loaded bone cements. A review
Acta Orthop Scand. 72:557571.
von Bismarck P, Schneppenheim R, Schumacher U. 2001. Suc-
cessful treatment of Pseudomonas aeruginosa respiratory
tract infection with a sugar solution-a case report on a lec-
tin based therapeutic principle. Klin Padiatr. 213:285287.
Wang Q, Perez JM, Webster TJ. 2013. Inhibited growth of
Pseudomonas aeruginosa by dextran- and polyacrylic acid-
coated ceria nanoparticles. Int J Nanome. 8:33953399.
Wiens JR, Vasil AI, Schurr MJ, Vasil ML. 2014. Iron-regulated
expression of alginate production, mucoid phenotype, and
biolm formation by Pseudomonas aeruginosa. MBio. 5:
e0101001013.
Wu H, Lee B, Yang L, Wang H, Givskov M, Molin S, Hoiby
N, Song Z. 2011. Effects of ginseng on Pseudomonas
aeruginosa motility and biolm formation. FEMS Immunol
Med Microbiol. 62:4956.
Xie J, Huang J, Li X, Sun S, Chen X. 2009. Iron oxide
nanoparticle platform for biomedical applications. Curr
Med Chem. 16:12781294.
Zhang S, Wei W, Zhang J, Mao Y, Liu S. 2002. Effect of static
magnetic eld on growth of Escherichia coli and relative
response model of series piezoelectric quartz crystal.
Analyst. 127:373377.
Biofouling 457
Downloaded by [104.249.167.39] at 13:44 15 December 2015
... Picoli et al. found low biofilm production by E. coli isolate (56.3%) due to Bv melittin [87]. The effect of BV is to eliminate biofilm in Gram-positive and Gram-negative bacteria [88,89]. Furthermore, αFe 2 O 3 NPs have a less significant antibacterial activity than its significant anti-biofilm potential as it facilitates the delivery of different antimicrobial agents conjugated with it into the biofilm by its magnetic field. ...
... Furthermore, αFe 2 O 3 NPs have a less significant antibacterial activity than its significant anti-biofilm potential as it facilitates the delivery of different antimicrobial agents conjugated with it into the biofilm by its magnetic field. [88,90,91]. Bare ZnO NFs against the E7 isolate showed inhibition in biofilm formation by 25.13 ± 1.42, which is similar to the findings of [92]. ...
... Picoli et al. found low biofilm production by E. coli isolate (56.3%) due to Bv melittin [87]. The effect of BV is to eliminate biofilm in Gram-positive and Gram-negative bacteria [88,89]. Furthermore, αFe 2 O 3 NPs have a less significant antibacterial activity than its significant anti-biofilm potential as it facilitates the delivery of different antimicrobial agents conjugated with it into the biofilm by its magnetic field. ...
... Furthermore, αFe 2 O 3 NPs have a less significant antibacterial activity than its significant anti-biofilm potential as it facilitates the delivery of different antimicrobial agents conjugated with it into the biofilm by its magnetic field. [88,90,91]. Bare ZnO NFs against the E7 isolate showed inhibition in biofilm formation by 25.13 ± 1.42, which is similar to the findings of [92]. ...
Article
This study targeted developing a novel Zinc oxide with alpha hematite nanoflowers (NFs)-loaded bee venom (Bv) (Bv-ZnO@αFe2O3 NFs) as a bio-natural product from bees to combine both the advantages of combination magnetic properties and the antimicrobial and anti-biofilm properties on isolated coliform bacteria from the effluent of wastewater treatment plants. About 24 isolates of treated wastewater isolates were multidrug resistant (MDR). The phylogenetic grouping of Escherichia coli (E. coli) and Klebsiella pneumonia (K. pneumonia) showed that the largest group was Group A, followed by Group B2 and Group B1. Fourier transform infrared (FTIR), The X-ray diffraction (XRD), and scanning electron microscopy-energy dispersive X-ray analysis (SEM− EDX) validated the coating operation’s contact with Bv onto ZnO@αFe2O3 NFs. According to high-resolution transmission electron microscopy (TEM) and selected area electron diffraction (SAED), pattern analyses for prepared nanoformulations exhibited a spherical shape of αFe2O3 (~9–15 nm), and floral needle shapes with uniform distribution of size with aggregation of ZnOαFe2O3 and Bv-ZnO@αFe2O3 NFs around (~100–200 nm). The toxicity of Bv-ZnO@αFe2O3 NFs was comparable up to 125 µg mL−1, when it reached 64.79% (IC50, 107.18 µg mL−1). The antibacterial activity showed different zones of inhibition against different isolates. The biofilm inhibitory activity of NPs and NFs showed a highly significant reduction (p < 0.001) in treated biofilms with ZnO@αFe2O3 and Bv-ZnO@αFe2O3. In essence, ZnO@αFe2O3 and Bv-ZnO@αFe2O3 NFs are promising antimicrobials for inhibiting the growth and biofilm of MDR E. coli and K. pneumonia isolates, thereby, biocontrol of wastewater.
... Magnetic field exposure can significantly remove biofilms. For instance, Bandara et al. (2015) reported a significant reduction (between 27 and 93%) in the biomass of P. aeruginosa 24 h biofilms when treated with the magnetic field of different modalities (i.e. static one-sided, static switched, alternating, Static + alternating magnetic field) for respective times compared to untreated. ...
... The exact mechanism of action of magnetic fields on biofilm is not yet clear. However, based on the microscopy images it seems that magnetic fields were able to ravage the biofilms, particularly, by negatively affecting the EPS matrix (Bandara et al., 2015;Chopra et al., 2017;Pinto et al., 2020). Magnetic fields in conjunction with magnetic nanoparticles have been used to increase the anti-biofilm activity (Quan et al., 2019). ...
Article
In the food industry, equipment surfaces with microbial entities can easily contaminate the food being processed, thereby leading to significant economic problems and even public health issues. To ensure the hygiene of materials in contact with food, cleaning processes are commonly implemented. Conventional strategies used for biofilm removal are energy and water intensive processes. Besides, they also use a substantial amount of chemicals. However, despite the strong enforcement of the conventional cleaning processes, each year, alarming statistics are reported by sanitary organizations, emphasizing the need of improving the safety and hygiene of equipment surfaces. Therefore, the investigation of strategies to effectively remove biofilm biomass is becoming essential. This literature review aims to describe the effectiveness of different physical strategies for biofilm biomass removal. Along with conventional physical methods (conventional CIP, water jet, and static foam cleaning) widely used in the food industry, other promising methods such as pulsed flow, bubble flow, foam flow, ice pigging, snow CO2, electromagnetic methods, plasma, and air jet were reported. Further, the principles, cleaning mechanisms, important process parameters, as well as, limitations associated with these methods have been discussed. All of these methods have shown a promising ability to remove biofilm, while some of them like foam flow and pulsed flow were found to be eco-friendly as they reduced environmental impacts based on LCA analyses.
... [42][43][44][45][46] Non-thermal mechanisms by which magnetic nanoparticles subjected to an oscillating magnetic field enhance antibiotic effectiveness have also been proposed via mechanical disruption of the biofilm matrix. [47][48][49] Reviews of recent work on use of innovative strategies by which nanoparticles can be used to disrupt biofilms, including effects of oscillating magnetic fields, were given by Pinto et al., 50 Quan et al., 49 and Sahli et al. 51 The current study derives an analytical solution for a viscoelastic hydrogel (matrix) immersed in water in the presence of superparamagnetic nanoparticles exposed to an oscillating magnetic field directed tangent to the hydrogel surface. This problem is important for prediction of limiting displacement and stresses required for the mechanical disruption of a biofilm matrix under a sufficiently strong oscillating magnetic field, as well as for characterization of biofilm matrix mechanical response under weaker magnetic forcing. ...
Article
A study was conducted of the effect of superparamagnetic nanoparticles on a hydrogel in the presence of an oscillating magnetic field directed tangent to the hydrogel surface. The oscillating magnetic field causes the particles to oscillate laterally in the hydrogel, with some of the particles adhering to the hydrogel matrix and other particles moving freely through the hydrogel pore spaces. The analysis was performed for a three-phase matrix-water-particles model, in which the solvent (water) and hydrogel matrix are interacting continua and the particles are a discrete phase. The study examined the effect of fluid elasticity on wave propagation due to the no-slip boundary condition acting under the transversely oscillating magnetic field. A memory effect within the fluid results in a deviation of the minimum and maximum shear rates observed in one half of the oscillation period from those observed in the other half of the oscillation period. The behavior of the hydrogel with different values of the governing dimensionless parameters was assessed. The matrix Reynolds number, the Deborah number, and the ratio of matrix relaxation to retardation times were all observed to have significant influence on the hydrogel viscoelastic response and on the wave propagation within the hydrogel. The phase difference between the water and matrix oscillations is strongly influenced by the phase interaction force coefficient, the Deborah number, and the ratio of free to captured particles. The system is found to approach an asymptotic state at a high Deborah number, which is independent of the value of the Deborah number.
... Magnetic fields have potential applications in medicine as a means of sterilization (high-frequency fields) [73], as well as being a mechanism to increase the sensitivity of pathogens to antibiotics [52,74] and remove bacterial biofilms from surgical instruments [61]. ...
Article
Full-text available
Despite the growing prevalence of using living organisms in industry, the control of biotechnological processes remains highly complex and constitutes one of the foremost challenges in these applications. The usage of electromagnetic fields offers a great opportunity to control various biotechnological processes by alternating growth and cell metabolism without influencing the characteristics of the cultivation medium or the products of the biotechnological process. The investigation of electromagnetic field applications across various industries, including food production, medicine, and pollutant mitigation, has yielded substantial insights. We used the scientific databases PubMed and ScienceDirect to select 103 experimental and theoretical articles that included original results suitable for further investigation. This type of search was repeated with every new relevant article iteratively until no new articles could be detected. Notably, even weak, low-frequency magnetic fields can accelerate the growth of certain organisms, further stabilize the bacterial community in activated sludge within wastewater treatment plants, enhance the fermentation capabilities of both yeast and bacteria, enhance metal bioleaching by the activation of bacterial metabolism, or improve the metal tolerance of plants during the phytoremediation process. Moreover, magnetic fields exhibit a promising sustainable possibility for the better control of biotechnological processes, thus making these processes more competitive compared with the currently used long-term unsustainable extraction of metals. Although with these interesting results, these examples represent highly exceptional applications. Despite these examples, the overall application potential of magnetic fields remains largely unexplored and unknown.
Article
Full-text available
Even though the bioeffects of electromagnetic radiation (EMR) have been extensively investigated during the past several decades, our understandings of the bioeffects of EMR and the mechanisms of the interactions between the biological systems and the EMRs are still far from satisfactory. In this article, we introduce and summarize the consensus, controversy, limitations, and unsolved issues. The published works have investigated the EMR effects on different biological systems including humans, animals, cells, and biochemical reactions. Alternative methodologies also include dielectric spectroscopy, detection of bioelectromagnetic emissions, and theoretical predictions. In many studies, the thermal effects of the EMR are not properly controlled or considered. The frequency of the EMR investigated is limited to the commonly used bands, particularly the frequencies of the power line and the wireless communications; far fewer studies were performed for other EMR frequencies. In addition, the bioeffects of the complex EM environment were rarely discussed. In summary, our understanding of the bioeffects of the EMR is quite restrictive and further investigations are needed to answer the unsolved questions.
Article
Methicillin-Resistant Staphylococcus aureus (MRSA) biofilms are among the most dangerous infections on medical implants, typically requiring surgical explantation and replacement. This study investigated the thermal susceptibility of MRSA biofilms to thermal shocks from 60 to 80 °C for 1-30 min as well as the effect of various antibiotics (most notably methicillin) on thermal mitigation. Pre- and post-shock exposure to three different classes of antibiotics (ciprofloxacin, tobramycin, and methicillin) at concentrations ranging from 0.25 to 128 μg mL-1 were investigated. MRSA biofilms exhibited thermal susceptibility comparable to other common nosocomial pathogens, such as Pseudomonas aeruginosa, though with greater variability. Exposure to antibiotics of any class significantly decreased the degree of thermal shock required for reliable mitigation, including at subclinical concentration. These combined treatments reduced biofilm population more than the sum of thermal and chemical treatments alone, demonstrating synergism, while also indicating a critical population drop of ∼4.5 log10 beyond which the biofilms typically became non-viable.
Article
Full-text available
Limited research has suggested iron oxide nanoparticles (FeNP) have an inhibitory effect against several different genera of bacteria: Staphylococcus, Bacillus and Pseudomonas spp. In this study we looked at the effect of three different sets of Fe3O4 nanoparticles (FeNPs) on the development of Pseudomonas aeruginosa PAO1 biofilms. Two of the tested NPs were SPIONs (Superparamagnetic Iron Oxide Nanoparticles). Exposure of cells to the SPIONs at concentrations up to 200 μg/ml resulted in an increase in biofilm biomass by 16 h under static conditions and a corresponding increase in cell density in the bulk liquid. In contrast, these biofilms had decreased levels of extracellular DNA (eDNA). Fe(II) levels in the supernatants of biofilms formed in the presence of FeNPs exceeded 100 μM compared with 20 μM in control media without cells. Spent cell supernatants had little effect on Fe(II) levels. Cells also had an effect on the aggregation behavior of these nanoparticles. SPIONs incubated with cells exhibited a decrease in the number and size of FeNP aggregates visible using light microscopy. SPIONs resuspended in fresh media or spent culture supernatants formed large aggregates visible in the light microscope upon exposure to a supermagnet; and could be pelleted magnetically in microtitre plate wells. In contrast, SPION FeNPs incubated with cells were unaffected by exposure to the supermagnet and could not be pelleted. The results of this study indicate a need to reconsider the effects of FeNPs on bacterial growth and biofilm formation and the effect the bacterial cells may have on the use and recovery of SPIONs.
Article
Full-text available
Background: Magnetite (Fe3O4) nanoparticles are currently one of the important and acceptable magnetic nanoparticles for biomedical applications. To use magnetite nanoparticles for bacteria cell separation, the surface of nanoparticles would be modified for immobilizing of nanoparticles on the surface of bacteria. Functionalization of magnetite nanoparticles is performed by different surfactants such as glycine or oleic acid to attach on the bacteria cell surface simultaneously. The magnetic nanoparticles have very low toxicity on the living cells. There are some studies on evaluating the toxicity of magnetite nanoparticles on eukaryote cells, which their results showed negligible toxicity in eukaryote cells of the modified magnetite nanoparticles with different surfactants. But the toxicity of magnetite nanoparticles on bacteria cells is not reported. Objectives: in this study, the effect of the magnetic nanoparticles iron oxide (Fe3O4) on the growth rate of the genetically engineered Pseudomonas aeruginosa (PTSOX4) cells in different media with different magnetic nanoparticles concentration have been investigated. Materials and Methods: In this study, the genetically manipulated bacterial cells, Pseudomonas aeruginosa (PTSOX4), were coated with magnetic Fe3O4 nanoparticles to evaluate the toxicity effect of these nanoparticles on the growth rate of this strain in Laurial Bertany (LB) and Basal Salt media (BSM) separately. In addition the minimal inhibitory concentration (MIC) and the minimal bactericidal concentration (MBC) tests of these nanoparticles were examined. Results: A low concentration of nanoparticles has little toxicity effect on the cell growth in this bacterium. Maximal level of the growth obtained in the late stationary phase, using a concentration of 500 ppm or more of Fe3O4nanoparticles, but a high concentration of these nanoparticles, more than 1000 PPM, resulted in reducing the cell growth rate. However, there was not a considerable lethal effect on the cell viability. Moreover, using a high nanoparticle concentration leads to a high level of bacterial cell coating due to more contact of the nanoparticles to bacterial cell surface. Conclusions: It is concluded that magnetite nanoparticles have negligible toxicity on the living bacteria cells and they are so applicable in different parts of biotechnology fields. © 2013, National Institute of Genetic Engineering and Biotechnology; Published by Kowsar Corp.
Article
Full-text available
Unlabelled: Pseudomonas aeruginosa strains of non-cystic fibrosis (non-CF) origin do not produce significant amounts of extracellular alginate and are nonmucoid. In CF, such isolates can become mucoid through mutation of one of the genes (mucA, mucB, mucC, or mucD) that produce regulatory factors that sequester AlgU, required for increased expression of alginate genes. Mutation of the muc genes in the nonmucoid PAO1, PA14, PAKS-1, and Ps388 strains led to increased levels of extracellular alginate and an obvious mucoid phenotype, but only under iron-limiting growth conditions (≤5 µM), not under iron-replete conditions (≥10 µM). In contrast, >50% of P. aeruginosa isolates from chronic CF pulmonary infections expressed increased levels of alginate and mucoidy both under iron-limiting and iron-replete conditions (i.e., iron-constitutive phenotype). No single iron regulatory factor (e.g., Fur, PvdS) was associated with this loss of iron-regulated alginate expression and mucoidy in these CF isolates. However, the loss of only pyoverdine production, or its uptake, abrogated the ability of P. aeruginosa to produce a robust biofilm that represents the Psl-type of biofilm. In contrast, we show that mutation of the pyoverdine and pyochelin biosynthesis genes and the pyoverdine receptor (FpvA) lead to iron-constitutive expression of the key alginate biosynthesis gene, algD, and an explicitly mucoid phenotype in both iron-limiting and iron-replete conditions. These data indicate that alginate production and mucoidy, in contrast to other types of biofilms produced by P. aeruginosa, are substantially enhanced under iron limitation. These results also have compelling implications in relation to the use of iron chelators in the treatment of P. aeruginosa CF infections. Importance: Pseudomonas aeruginosa is a leading model for the investigation of biofilms. While data have been generated about the role of iron in alginate-independent (Psl/Pel) biofilm development, there is a paucity of data regarding the role of iron in alginate production and its associated mucoid phenotype. We demonstrate that biologically relevant levels of iron that exist in the airway mucus of cystic fibrosis (CF) patients have a substantial influence on production of alginate and the overt mucoid phenotype, pathognomonic of P. aeruginosa infections in CF. Mucoid mutants of non-CF P. aeruginosa isolates are mucoid only under iron limitation and do not express increased levels of alginate under iron-replete growth conditions. However, a significant number of long-term CF isolates lost their iron-regulated expression of increased alginate production and mucoidy and became iron constitutive for these properties. In contrast to the formation of Psl-type biofilms, increasing iron limitation ultimately leads to an iron-constitutive expression of alginate and mucoidy.
Article
Full-text available
Ceria (CeO2) nanoparticles have been widely studied for numerous applications, but only a few recent studies have investigated their potential applications in medicine. Moreover, there have been almost no studies focusing on their possible antibacterial properties, despite the fact that such nanoparticles may reduce reactive oxygen species. In this study, we coated CeO2 nanoparticles with dextran or polyacrylic acid (PAA) because of their enhanced biocompatibility properties, minimized toxicity, and reduced clearance by the immune system. For the first time, the coated CeO2 nanoparticles were tested in bacterial assays involving Pseudomonas aeruginosa, one of the most significant bacteria responsible for infecting numerous medical devices. The results showed that CeO2 nanoparticles with either coating significantly inhibited the growth of Pseudomonas aeruginosa, by up to 55.14%, after 24 hours compared with controls (no particles). The inhibition of bacterial growth was concentration dependent. In summary, this study revealed, for the first time, that the characterized dextran- and PAA-coated CeO2 nanoparticles could be potential novel materials for numerous antibacterial applications.
Article
Full-text available
Cutaneous wounds are often superinfected during the healing process and this leads to prolonged convalescence and discomfort. Usage of suitable wound dressings is very important for an appropriate wound care leading to a correct healing. The aim of this study was to demonstrate the influence of a nano-coated wound dressing (WD) on Candida albicans colonization rate and biofilm formation. The modified WD was achieved by submerging the dressing pieces into a nanofluid composed of functionalized magnetite nanoparticles and Satureja hortensis (SO) essential oil (EO). Chemical composition of the EO was established by GC-MS. The fabricated nanostructure was characterized by X-ray Diffraction (XRD), Transmission Electron Microscopy (TEM), Differential Thermal Analysis (DTA) and Fourier Transform-Infrared Spectroscopy (FT-IR). The analysis of the colonized surfaces using (Scanning Electron Microscopy) SEM revealed that C. albicans adherence and subsequent biofilm development are strongly inhibited on the surface of wound dressing fibers coated with the obtained nanofluid, comparing with regular uncoated materials. The results were also confirmed by the assay of the viable fungal cells embedded in the biofilm. Our data demonstrate that the obtained phytonanocoating improve the resistance of wound dressing surface to C. albicans colonization, which is often an etiological cause of local infections, impairing the appropriate wound healing.
Chapter
Introduction Characteristics of Prokaryotic Cells Summary
Article
Bacteria that attach to surfaces aggregate in a hydrated polymeric matrix of their own synthesis to form biofilms. Formation of these sessile communities and their inherent resistance to antimicrobial agents are at the root of many persistent and chronic bacterial infections. Studies of biofilms have revealed differentiated, structured groups of cells with community properties. Recent advances in our understanding of the genetic and molecular basis of bacterial community behavior point to therapeutic targets that may provide a means for the control of biofilm infections.
Article
Microbial biofilms are highly refractory to antimicrobials. The aim of this study was to investigate the use of low-frequency vibration therapy (20-20 kHz) on antibiotic-mediated Pseudomonas aeruginosa biofilm eradication. In screening studies, low-frequency vibrations were applied on model biofilm compositions to identify conditions in which surface standing waves were observed. Alginate surface tension and viscosity were also measured. The effect of vibration on P. aeruginosa biofilms was studied using a standard biofilm assay. Subminimal inhibitory concentrations (sub-MIC) of tobramycin (5 μg/ml) were added to biofilms 3 h prior, during, and immediately after vibration and quantitatively assessed by (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) reduction assay (XTT) and, qualitatively, by confocal laser scanning microscopy (CLSM). The standing waves occurred at frequencies <1,000 Hz. Biofilms vibrated without sub-MIC tobramycin showed a significantly reduced metabolism compared to untreated controls (p < 0.05). Biofilms treated with tobramycin and vibrated simultaneously (450, 530, 610, and 650 Hz), or vibrated (450 and 650 Hz) then treated with tobramycin subsequently, or vibrated (610Hz, 650Hz) after 3 h of tobramycin treatment showed significantly lower metabolism compared to P. aeruginosa biofilm treated with tobramycin alone (p < 0.05). CLSM imaging further confirmed these findings. Low frequency vibrations assisted tobramycin in killing P. aeruginosa biofilms at sub-MIC. Thus, sound waves together with antibiotics are a promising approach in eliminating pathogenic biofilms.
Article
Bacteria in biofilms are much more resistant to antibiotics and microbicides as compared to their planktonic stage. Thus, to achieve the same antibacterial efficacy, a much higher dose of antibiotics is required for biofilm bacteria. However, the widespread use of antibiotics has been recognized as the main cause for the emergence of antibiotic-resistant microbial species, which has now become a major public health crisis globally. In this work, we present an efficient nonantibiotic-based strategy for disrupting biofilms using carboxymethyl chitosan (CMCS) coated on magnetic iron oxide nanoparticles (CMCS-MNPs). CMCS-MNPs demonstrate strong bactericidal activities against both Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli) planktonic cells. More than 99% S. aureus and E. coli planktonic cells were killed after incubation with CMCS-MNPs for 10 and 5 h, respectively. In the presence of a magnetic field (MF), CMCS-MNPs can effectively penetrate into both S. aureus and E. coli biofilms, resulting in a reduction of viable cells counts by 84% and 95%, respectively, after 48 h incubation, as compared to the control experiment without CMCS-MNPs or CMCS. CMCS-MNPs are noncytotoxic toward mammalian cells and can potentially be a useful antimicrobial agent to eliminate both planktonic and biofilm bacteria.
Article
Iron oxide colloidal nanoparticles (ferrofluids) are investigated for application in the treatment of cystic fibrosis lung infections, the leading cause of mortality in cystic fibrosis patients. We investigate the use of iron oxide nanoparticles to increase the effectiveness of administering antibiotics through aerosol inhalation using two mechanisms: directed particle movement in the presence of an inhomogeneous static external magnetic field and magnetic hyperthermia. Magnetic hyperthermia is an effective method for decreasing the viscosity of the mucus and biofilm, thereby enhancing drug, immune cell, and antibody penetration to the affected area. Iron oxide nanoparticles of various sizes and morphologies were synthesized and tested for specific losses (heating power). Nanoparticles in the superparamagnetic to ferromagnetic size range exhibited excellent heating power. Additionally, iron oxide / zinc selenide core/shell nanoparticles were prepared, in order to enable imaging of the iron oxide nanoparticles. We also report on synthesis and characterization of MnSe/ZnSeS alloyed quantum dots.