ArticlePDF Available

Combinations of maggot excretions/secretions and antibiotics are effective against Staphylococcus aureus biofilms and the bacteria derived therefrom

Authors:

Abstract and Figures

Maggots of the blowfly Lucilia sericata are used for the treatment of chronic wounds. Previously we reported that maggot excretions/secretions (ES) break down Staphylococcus aureus biofilms but do not kill the bacteria. As many antibiotics are not effective against biofilms we assessed the effect of combinations of ES and antibiotics on S. aureus biofilms and on the survival of the bacteria released from the biofilms. Effects of ES, antibiotics (vancomycin, daptomycin or clindamycin) and combinations thereof on S. aureus ATCC 29 213 biofilms and bacterial viability were determined using microtitre plates and in vitro killing assays. Vancomycin and daptomycin dose-dependently enhanced biofilm formation, whereas clindamycin reduced S. aureus biofilm size. Adding ES to antibiotic incubations caused a complete biofilm breakdown. After a lag time the bacteria derived from biofilms became susceptible to vancomycin and clindamycin, provided that the medium was refreshed. Daptomycin dose-dependently eliminated the biofilm-derived bacteria immediately. Furthermore, it was significantly more effective against bacteria derived from ES-exposed biofilms than those from control biofilms. ES did not affect the activity of the antibiotics against log-phase S. aureus. Combinations of maggot ES and antibiotics eliminate S. aureus biofilms and the bacteria derived therefrom.
Content may be subject to copyright.
Combinations of maggot excretions/secretions and antibiotics are
effective against Staphylococcus aureus biofilms and the bacteria
derived therefrom
Mariena J. A. van der Plas1, 2, Cheryl Dambrot1, Heleen C. M. Dogterom-Ballering 1, Simone Kruithof 1,
Jaap T. van Dissel 1and Peter H. Nibbering1*
1
Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands;
2
Department of Surgery, Leiden
University Medical Center, Leiden, The Netherlands
*Corresponding author. Tel: þ31-71-526-2204; Fax: þ31-71-526-6758; E-mail: p.h.nibbering@lumc.nl
Received 1 October 2009; returned 20 October 2009; revised 26 January 2010; accepted 26 January 2010
Objectives: Maggots of the blowfly Lucilia sericata are used for the treatment of chronic wounds. Previously we
reported that maggot excretions/secretions (ES) break down Staphylococcus aureus biofilms but do not kill the
bacteria. As many antibiotics are not effective against biofilms we assessed the effect of combinations of ES
and antibiotics on S. aureus biofilms and on the survival of the bacteria released from the biofilms.
Methods: Effects of ES, antibiotics (vancomycin, daptomycin or clindamycin) and combinations thereof on
S. aureus ATCC 29213 biofilms and bacterial viability were determined using microtitre plates and in vitro
killing assays.
Results: Vancomycin and daptomycin dose-dependently enhanced biofilm formation, whereas clindamycin
reduced S. aureus biofilm size. Adding ES to antibiotic incubations caused a complete biofilm breakdown.
After a lag time the bacteria derived from biofilms became susceptible to vancomycin and clindamycin, pro-
vided that the medium was refreshed. Daptomycin dose-dependently eliminated the biofilm-derived bacteria
immediately. Furthermore, it was significantly more effective against bacteria derived from ES-exposed biofilms
than those from control biofilms. ES did not affect the activity of the antibiotics against log-phase S. aureus.
Conclusions: Combinations of maggot ES and antibiotics eliminate S. aureus biofilms and the bacteria derived
therefrom.
Keywords: Lucilia sericata, clindamycin, vancomycin, daptomycin, bacterial killing
Introduction
Chronic wounds are common in patients with vascular insuffi-
ciencies and underlying chronic conditions such as diabetes mel-
litus, as well as in patients suffering from acute, extended
trauma.
1,2
These wounds and associated amputations result in
decreased physical, emotional and social function of patients,
a reduced quality of life and major economic costs for patients,
their families and society as a whole.
3,4
A severe complication
of the healing process is bacterial colonization and subsequent
infection of the wound surface,
57
especially when the bacteria
are residing in biofilms.
8
These latter bacteria exhibit altered
growth characteristics and gene expression profiles as compared
with those present free in the environment, the so-called plank-
tonic bacteria.
9
Importantly, biofilm formation and the conse-
quences thereof for bacterial growth characteristics render
microorganisms resistant to the action of many antibiotics
10,11
as well as cells and effector molecules of the host’s immune
system.
7,12
Bacterial fragments/products released from biofilms
continuously attract host cells to the wound. As phagocytes
cannot ingest the biofilm-associated bacteria and therefore are
unable to eliminate the cause of infection, the subsequent
accumulation of inflammatory cells and enhanced release of
proinflammatory cytokines, proteases and reactive oxygen
species eventually leads to inactivation of growth factors and
tissue destruction,
13,14
thereby contributing to the establishment
and maintenance of chronic wounds.
Sterile larvae—maggots—of the green bottle blowfly Lucilia
sericata are used as a treatment for various types of chronic
wounds.
15 17
Previously we reported the use of maggot
excretions/secretions (ES) to break down Staphylococcus aureus
and Pseudomonas aeruginosa biofilms.
18
However, the bacteria
released from these biofilms were not killed by ES. On the
other hand, many antibiotics cannot break down bacterial
#The Author 2010. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For Permissions, please e-mail: journals.permissions@oxfordjournals.org
J Antimicrob Chemother 2010; 65: 917923
doi:10.1093/jac/dkq042 Advance publication 26 February 2010
917
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
biofilms but effectively kill planktonic bacteria. Therefore, we
assessed the effect of combinations of maggot ES and anti-
biotics on S. aureus biofilms and on the survival of the bacteria
released from these biofilms.
Materials and methods
Maggots and maggot ES
ES of sterile second- and third-instar larvae of L. sericata (a gift from Bio-
Monde GmbH, Barsbu¨ttel, Germany) were collected as described.
19
Larvae were incubated in H
2
O(5mL/larva) for 60 min at ambient temp-
erature in the dark. Next, ES were checked for sterility and stored
at 2208C. Prior to use, ES preparations were pooled and centrifuged
at 1300 gfor 5 min at 48C to remove particulate material. ES protein
concentration was determined using the Pierce BCA Protein Assay kit
according to the manufacturer’s instructions.
Antibiotics
Stock solutions of vancomycin (Pharmachemie B.V., Haarlem, The
Netherlands), daptomycin (Cubicin, Chiron Corporation Limited, Uxbridge,
UK) and clindamycin (Upjohn GmbH, Heppenheim, Germany) were dis-
solved in distilled water to a final concentration of 10 g/L.
S. aureus cultures
S. aureus ATCC 29213 (Manassas, VA, USA) were grown in tryptone soya
broth (TSB) at 378C under vigorous shaking. The MIC values for this
strain are 0.5– 2 mg/L for vancomycin, 0.25– 1 mg/L for daptomycin
and 0.06 –0.25 mg/L for clindamycin.
20
Biofilm assay
Biofilm formation of S. aureus in 96-well polyvinyl chloride (PVC) plates
was conducted as described.
18
In short, bacteria from overnight cultures
were diluted 1:1000 and 5 mL of these bacterial suspensions were added
to each well containing 100 mL of ‘biofilm medium’ consisting of 0.5TSB
supplemented with 0.2% (w/v) glucose. After 24 h, planktonic cells were
removed and 100 mL of biofilm medium with or without antibiotics (1
400 mg/L) and/or ES (20– 200 mg/L) were added to the biofilms. At the
indicated time intervals, planktonic cells were harvested from these
wells and the numbers of viable bacteria were determined microbiologi-
cally using serial dilutions of these suspensions plated six times each
onto agar plates. The lower detection limit of this method is 100 cfu/
well. In addition, after washing the wells with tap water, biofilms were
exposed to a 1% (w/v) Crystal Violet solution for 15 min, washed and
then incubated in absolute ethanol for 15 min to extract the Crystal
Violet retained by the cells. Next, this solution was used to quantify the
amount of biofilm by measuring the absorbance at 590 nm.
Furthermore, we investigated the effect of antibiotics on bacteria
derived from the biofilms and subsequently transferred to fresh biofilm
medium. For this purpose, the planktonic cells were removed from
24-h-old biofilms, and fresh biofilm medium was added to the wells con-
taining ES (20 –200 mg/L) or H
2
O as a control. After an additional 24 h,
the bacteria released from the biofilms were harvested and 25 mLof
these bacterial suspensions were transferred to wells of a PVC plate con-
taining 75 mL of TSB medium supplemented with antibiotics; the final
concentration of the medium was 0.5TSB and 0.2% glucose. After
3 and 24 h, the numbers of surviving bacteria were determined microbio-
logically as described above.
Concentration– effect relationship for antibiotics
on exponentially growing S. aureus
To further determine the concentration– effect relationship for antibiotics
on planktonic S. aureus in the presence or absence of ES, in vitro killing
assays were conducted as described
21
with minor modifications. Bacteria
in mid-log phase were centrifuged at 2000 gfor 10 min, washed twice
with PBS and resuspended to a concentration of 110
7
bacteria/mL of
biofilm medium supplemented with antibiotics (0.005– 500 mg/L) and/
or ES (20 –200 mg/L). Subsequently, 100 mL aliquots of these bacterial
suspensions were transferred to wells of a 96-well PVC plate and incu-
bated at 378C. After 1, 2 and 3 h, the numbers of surviving bacteria
were determined microbiologically as described above.
Next, the differences between the logarithms (base 10) of the
numbers of cfu in the absence and presence of antibiotics and/or ES
were calculated for each timepoint.
22
For further calculations, the
highest value of the net killing rate during the 3 h of exposure was
used (E
R
). The concentration– effect relationship was established by
using the Hill equation:
ER¼ER;max C=ðEC50 þCÞ
where E
R
,max is the estimated maximal killing rate, Cthe antibiotic con-
centration (mg/L) and EC
50
the estimated antibiotic concentration at
which 50% of the maximal killing is reached. The parameters of this
pharmacodynamic model were calculated in SPSS using non-linear
regression analysis.
Statistical analysis
Statistical analyses were performed using Graphpad Prism version 4.02.
Statistical differences between the values for ES-incubated and
control-incubated bacteria were analysed using a paired t-test. The
level of significance was set at Pvalues ,0.05.
Results
Effect of antibiotics and ES on S. aureus biofilms
The results showed a dose-dependent increase in biofilm size by
vancomycin and daptomycin already within 3 h (Table 1). This
effect persisted over the next 21 h. In contrast, clindamycin
dose-dependently decreased the amount of biofilm; after 3 h
of incubation the biofilm diminished by 28% (Table 1). As
reported previously,
18
within 3 h ES degraded the S. aureus bio-
films completely and this effect was not counteracted by the
antibiotics (data not shown).
Effect of combining ES and antibiotics on the viability
of S. aureus released from biofilms
To find out if the biofilm-derived bacteria are susceptible to anti-
biotics, we incubated the biofilms with various concentrations of
antibiotics and/or ES and determined the number of viable bac-
teria at different intervals.
Preliminary experiments revealed no reduction in the number
of viable bacteria when using 10 mg/L of vancomycin and dap-
tomycin. Vancomycin at concentrations of 50 mg/L slightly but
significantly reduced the number of viable bacteria at 24 h
(Figure 1a), but not after 3 h (data not shown). Daptomycin
dose-dependently reduced the number of biofilm-derived bac-
teria within 3 h; a 3 log reduction was seen for 400 mg/L (data
not shown). This reduction in bacterial numbers continued for
van der Plas et al.
918
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
the next 21 h (Figure 1b). After 3 h of incubation, the number of
viable bacteria was 1 log lower in the presence of clindamycin
compared with control incubations of bacteria derived from
either ES-treated or control biofilms (data not shown). Over the
following 21 h, no increase in bacterial numbers was observed
in the presence of clindamycin (Figure 1c). Furthermore, a dose-
dependent effect of clindamycin was observed at the lowest
concentrations used in the experiments (i.e. 1, 5 and 10 mg/L,
resulting in a reduction in the number of viable bacteria by
53%+9%, 78%+4% and 80%+14%, respectively), whereas
maximal inhibition was reached with clindamycin concentrations
.10 mg/L. Of note, ES (200 mg/L) did not affect the antibacterial
activity of the antibiotics (Figure 1a c); 20 mg/L ES yielded
similar results (data not shown).
Effect of ES and antibiotics on biofilm-derived bacteria
transferred to fresh biofilm medium
As large numbers of bacteria derived from the biofilms remained
viable in the presence of the antibiotics, we considered the possi-
bility that these biofilm-derived bacteria were in a dormant state
making them resistant to these antibiotics. Therefore, bacteria
derived from ES-incubated or control-incubated biofilms were
transferred to fresh biofilm medium supplemented with
antibiotics.
Vancomycin failed to affect the number of viable bacteria at
3 h but induced a 2 log reduction in bacterial counts at 24 h.
This effect was independent of the dose of antibiotics or
whether the biofilms had been exposed to ES (Figure 2a). Dapto-
mycin dose-dependently reduced the number of bacteria within
3 h. Moreover, the bactericidal effect of daptomycin against bac-
teria derived from biofilms exposed to 200 mg/L ES (Figure 2b),
but not to 20 mg/L ES (data not shown), was higher than that
against bacteria from control biofilms. After 24 h, all bacteria
were killed by the various concentrations of daptomycin (data
not shown). Clindamycin prevented an increase in the number
of bacteria at 3 h of incubation (data not shown) and the
number of bacteria remained constant during the following
21 h (Figure 2c); the activity of clindamycin against bacteria
derived from ES-exposed biofilms was similar to that from
control biofilms.
Effect of ES on the concentrationeffect relationship
of antibiotics on exponentially growing S. aureus
To investigate the effect of ES on the activity of the antibiotics, we
determined the killing curves for the various antibiotics using log-
phase bacteria. The results showed a dose-dependent reduction in
the number of viable S. aureus by all three antibiotics; the
maximum effect of daptomycin was higher than that of clindamy-
cin and vancomycin, which were equally effective against the bac-
teria (Figure 3a– c). The estimated EC
50
and E
R
,max values are
given in Table 2. The activity of the antibiotics was not affected
by 20 or 200 mg/L ES. Of note, 500 mg/L daptomycin was suffi-
cient to kill all bacteria within 1 h in four out of five experiments
under all conditions. Furthermore, the maximum effect of clinda-
mycin was observed at 1 mg/L, the maximum effect of vancomy-
cin was observed at 10 mg/L and 500 mg/L daptomycin was
required to reach a maximal effect.
Discussion
The main conclusion from the present study is that combinations
of maggot ES and antibiotics can break down S. aureus biofilms
and subsequently eliminate the bacteria derived therefrom. This
conclusion is based on the following observations. First, ES broke
down established biofilms within 3 h and this effect was not nega-
tively or positively affected by the antibiotics. In the absence of ES,
samples containing vancomycin or daptomycin, antibiotics whose
activity depends on their action on the bacterial cell envelope,
lacked activity against biofilms; similar findings were observed
for the b-lactam antibiotic flucloxacillin (M. J. A. van der Plas,
C. Dambret and P. H. Nibbering, unpublished observations).
In contrast, clindamycin and linezolid (M. J. A. van der Plas,
C. Dambret and P. H. Nibbering, unpublished observations)
decreased the amount of biofilm, albeit that they were unable
to completely eradicate it in the 24 h incubations applied here.
Secondly, biofilm-derived bacteria became more susceptible to
the action of vancomycin and clindamycin after being
Table 1. Effect of antibiotics on established biofilms of S. aureus
Vancomycin Daptomycin Clindamycin
mg/L 3 h 24 h 3 h 24 h 3 h 24 h
0 0.18+0.02 0.30+0.02 0.21+0.01 0.30+0.02 0.21+0.01 0.29+0.02
1 0.19+0.03 0.26+0.04 0.20+0.03 0.27+0.03 0.20+0.03 0.24+0.04
5 0.21+0.03 0.29+0.05 0.20+0.03 0.26+0.05 0.17+0.03 0.24+0.04
10 0.27+0.04* 0.38+0.06* 0.23+0.04 0.35+0.07 0.17+0.04 0.24+0.04
50 0.26+0.04* 0.36+0.05* 0.26+0.04* 0.45+0.10* 0.16+0.01* 0.24+0.01*
100 0.26+0.04* 0.40+0.03* 0.27+0.01* 0.39+0.03* 0.16+0.01* 0.22+0.01*
200 0.26+0.04* 0.35+0.02* 0.28+0.02* 0.36+0.03* 0.16+0.01* 0.21+0.02*
400 0.22+0.01* 0.43+0.03* 0.33+0.02* 0.38+0.04* 0.15+0.01* 0.21+0.02*
Results (OD
590
) are means+SEM of 4 experiments.
For all samples, the addition of ES resulted in total breakdown of the biofilms (OD ,0.10).
*Values are significantly different from those for control biofilms (P,0.05).
Effect of maggot excretions/secretions and antibiotics on S. aureus
919
JA
C
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
transferred to fresh medium than when the bacteria remained
in the biofilm wells. An explanation for this result could be that
the bacteria derived from biofilms are in a static/dormant state
and therefore less susceptible to antibiotics that solely target
multiplying bacteria.
9
In contrast, daptomycin showed direct
activity against biofilm-derived bacteria whether or not they
were transferred to fresh medium. Moreover, ES (200 mg/L)
enhanced the antibacterial activity of daptomycin, but not of
vancomycin and clindamycin, against biofilm-derived S. aureus
transferred to fresh medium. Although we cannot explain the
latter results, they probably depend on the specific pharmacody-
namic mechanisms of daptomycin. Thirdly, ES did not alter the
activity of the antibiotics against exponentially multiplying
bacteria.
In the interpretation ofthe current findings, the following points
need be considered. First, we performed all experiments with a
single ATCC reference strain of S. aureus. In addition, our preliminary
results show that maggot ES can prevent methicillin-resistant
(a)
cfu/well
108
107
106
105
104
103
102
101
108
107
106
105
104
103
102
101
108
107
106
105
104
103
102
101
0
–+–+–+–+–+
0 50 50 100 100 200 200 400 400
Vancomycin (mg/L)
Daptomycin (mg/L)
(b)
cfu/well
0
–+–+–+–+–+
0 50 50 100 100 200 200 400 400
Clindamycin (mg/L)
(c)
cfu/well
0
–+–+–+–+–+
0 50 50 100 100 200 200 400 400
ES
ES
ES
Figure 1. Effect of antibiotics, ES and combinations thereof on
biofilm-derived S. aureus. Biofilms were washed and then incubated
with increasing concentrations of vancomycin (a), daptomycin (b) or
clindamycin (c) in the absence or presence of 200 mg/L ES for 24 h.
Thereafter, the number of viable bacteria in the medium was
determined microbiologically. Results of 4– 6 experiments are shown,
with lines representing the medians. The values from the samples
containing antibiotics are significantly different from those of the
control wells. Filled circles, no ES; open circles, 200 mg/L ES.
(a)
cfu/well
109
108
107
106
105
104
103
102
101
109
108
107
106
105
104
103
102
101
109
108
107
106
105
104
103
102
101
0
–+–+
*****
–+–+–+
0 50 50 100 100 200 200 400 400
Daptomycin (mg/L)
Vancomycin (mg/L)
(b)
cfu/well
0
–+–+–+–+–+
0 50 50 100 100 200 200 400 400
Clindamycin (mg/L)
(c)
cfu/well
0
–+–+–+–+–+
0 50 50 100 100 200 200 400 400
**
ES
ES
ES
Figure 2. Effect of antibiotics on the numbers of viable S. aureus derived
from ES-exposed and control-incubated biofilms transferred to new
wells. Biofilms were washed and then incubated with ES (200 mg/L) or
not for 24 h and thereafter transferred to wells containing fresh medium
supplemented with various concentrations of vancomycin (a),
daptomycin (b) or clindamycin (c). For vancomycin and clindamycin the
bacteria were recovered after a 24 h incubation and for daptomycin after
3 h. Finally, the number of viable bacteria in the medium was
determined microbiologically. Results of 6– 7 experiments are shown,
with the lines representing the medians. Values are significantly different
from those for bacteria derived from control-incubated biofilms
(*P,0.05 and **P,0.005). Filled circles, no ES; open circles, 200 mg/L ES.
van der Plas et al.
920
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
S. aureus biofilm formation and break down established biofilms
(M. J. A. van der Plas and P. H.Nibbering, unpublished observations).
Nevertheless, we cannot conclude that our findings are generaliz-
able to all S. aureus strains and/or other bacterial species.
However, in agreement with our results, several reports have
described daptomycin as being one of the most effective antibiotics
in the control of biofilm-related S. aureus infections whereas clinda-
mycin and vancomycin were less effective.
23,24
Secondly, the con-
centrations of antibiotics used in the in vitro biofilm assay are
relatively high compared with the free, active antibiotic concen-
trations generally achieved in patients (10–40 mg/L vancomycin,
1–15/20 mg/L daptomycin, 120 mg/L clindamycin). However,
concentrations of antibiotics similar to those used in the current
study can be attained in wounds after topical application. Thirdly,
at their MIC values vancomycin or daptomycin did not affect the
biofilm size, whereas at higher concentrations biofilm formation
was enhanced. It should be realized that MIC concentrations of
antibiotics did not reduce the numberof viable biofilm-derived bac-
teria whereas the higher, biofilm-enhancing concentrations did. In
agreement, supra-MIC concentrations of antibiotics are reported to
be effective against killing of bacteria released from biofilms,
whereas sub-MIC and MIC levels were not.
25
Fourthly, in contrast
to the above-mentioned reports, we did not observe a reduction
in biofilm size when using low levels of antibiotics. The explanation
for these contradictory results could be the method of quantifi-
cation. We used Crystal Violet staining to quantify the amount of
biomass whereas many reports describe the use of redox indicators
to measure the metabolic activity of the bacteria. However,
reduced metabolic activity does not exclude similar or even
increased biomass. In agreement with this, it is reported that
several antibiotics, including vancomycin, reduce the redox poten-
tial of bacteria without reducing the matrix.
26
This may lead to bac-
terial multiplication from the remaining matrix and may even
contribute to the development of resistance against the antibiotics.
Clearly, more research should be done into the effect of antibiotics
on biofilms and the bacteria derived from these structures. Fifthly,
previously we reported that the active molecule in ES is heat
labile.
18
Our recent studies into the effects of various substances
affecting structural features of proteins and inhibiting enzymic
activities indicated that the molecule in ES responsible for
S. aureus biofilm breakdown may be a serine protease (M. J. A.
van der Plas, unpublished observations). Currently, we are purifying
the active molecules from maggot ES by activity-guided chromato-
graphy. However, more research is required before the identity of
the active component of maggot ES is clarified. Obviously, appli-
cation of purified maggot-derived compounds instead of live
maggots will definitely increase the acceptance and use of this
very effective therapy for chronic—non-healing—wounds.
What is the clinical relevance of our findings? The failure of
antibiotics to affect biofilms and the bacteria derived therefrom
parallels their overall lack of activity against bacterial coloniza-
tion and infection of chronic wounds where biofilm formation
may be prominent.
7,8,10,27
Therefore, biofilm matrices and the
associated bacteria have to be targeted simultaneously to eradi-
cate chronic infections. Previously we found that maggot ES
break down biofilms of S. aureus
18
without killing the released
bacteria. Here we report that the released bacteria become sus-
ceptible to the actions of antibiotics that fail to affect
biofilm-associated microorganisms when they start multiplying.
Therefore, combinations of maggot ES and antibiotics would
ensure complete breakdown of the biofilms, thereby preventing
bacterial re-growth from the remaining matrix, and prompt anti-
biotic action against the bacteria released from the biofilms.
Additionally, these bacteria will be subjected to the effector
7
(a)
X/ = no ES
= 20 mg/L ES
= 200 mg/L ES
5
6
4
ER
3
2
1
0
0.001 0.01 0.1 1
Vancomycin (mg/L)
10 100 1000
0.0001 0.001 0.01 0.1 1 10 100
/
/
8
7
(b)
X/ = no ES
= 20 mg/L ES
= 200 mg/L ES
5
6
4
ER
3
2
1
0
0.1 1 10
Daptomycin (mg/L)
100 1000 10000
/
/
7
(c)
X/ = no ES
= 20 mg/L ES
= 200 mg/L ES
5
6
4
ER
3
2
1
0
Clindamycin (mg/L)
/
/
Figure 3. Effect of ES on the concentration–effect relationships of the
antibiotics against log-phase S. aureus. Bacteria were incubated with
increasing concentrations of vancomycin (a), daptomycin (b) or
clindamycin (c) in the presence or absence of ES (20–200 mg/L).
Results of 6– 8 experiments are shown. The dose effect relationships
were calculated from these data using the Hill equation.
Table 2. Pharmacodynamic parameters of the antibiotics and ES
Vancomycin Daptomycin Clindamycin
ES
(mg/L) E
R
,max/h
EC
50
(mg/L) E
R
,max/h
EC
50
(mg/L) E
R
,max/h
EC
50
(mg/L)
0 1.65 0.167 5.41 46.24 1.64 0.036
20 1.77 0.080 7.25 57.99 1.57 0.025
200 1.98 0.110 5.77 39.30 2.30 0.024
Effect of maggot excretions/secretions and antibiotics on S. aureus
921
JA
C
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
mechanisms of the immune system and ingestion by
maggots.
28,29
Thus, addition of maggots or maggot ES to anti-
biotics may become a promising approach for the treatment of
chronically colonized/infected surfaces of unresponsive wounds.
In this respect, it should be realized that some current treatment
modalities, where maggots apparently are used as a replace-
ment for instead of as an adjunct to antibiotics, often overesti-
mate bacterial killing by ES when applied in therapeutically
relevant amounts.
18
Of note, antibiotics including vancomycin
and clindamycin have no detrimental effects on maggot
growth and survival.
30
Based on our results and other
reports,
23,24
daptomycin and ES combined appear particularly
promising for the treatment of biofilm-related S. aureus wound
infections. Daptomycin, in contrast to vancomycin and cationic
antimicrobial peptides, kills bacteria without inducing bacterial
lysis.
31 33
As chronic wounds often are characterized by pro-
longed and dysregulated inflammatory responses,
13,34 36
decreased bacterial lysis may reduce proinflammatory responses
to bacterial products by immune cells, thereby contributing to
the healing process.
32
Acknowledgements
We thank Emile F. Schippers for technical assistance.
Funding
This study was supported by internal funding.
Transparency declarations
None to declare.
References
1Bartus CL, Margolis DJ. Reducing the incidence of foot ulceration and
amputation in diabetes. Curr Diab Rep 2004; 4: 413– 8.
2Ferrell BA, Josephson K, Norvid P et al. Pressure ulcers among patients
admitted to home care. J Am Geriatr Soc 2000; 48: 1042– 7.
3Boulton AJ, Vileikyte L, Ragnarson-Tennvall G et al. The global burden
of diabetic foot disease. Lancet 2005; 366: 1719– 24.
4Peters EJ, Childs MR, Wunderlich RP et al. Functional status of persons
with diabetes-related lower-extremity amputations. Diabetes Care 2001;
24: 1799– 804.
5Gjodsbol K, Christensen JJ, Karlsmark T et al. Multiple bacterial species
reside in chronic wounds: a longitudinal study. Int Wound J 2006; 3:
225–31.
6Harrison-Balestra C, Cazzaniga AL, Davis SC et al. A wound-isolated
Pseudomonas aeruginosa grows a biofilm in vitro within 10 hours and is
visualized by light microscopy. Dermatol Surg 2003; 29: 631– 5.
7Davis SC, Martinez L, Kirsner R. The diabetic foot: the importance of
biofilms and wound bed preparation. Curr Diab Rep 2006; 6: 439– 45.
8Edwards R, Harding KG. Bacteria and wound healing. Curr Opin Infect
Dis 2004; 17:916.
9Stoodley P, Sauer K, Davies DG et al. Biofilms as complex differentiated
communities. Annu Rev Microbiol 2002; 56: 187– 209.
10 Sheldon AT Jr. Antibiotic resistance: a survival strategy. Clin Lab Sci
2005; 18: 170–80.
11 Gilbert P, Allison DG, McBain AJ. Biofilms in vitro and in vivo: do
singular mechanisms imply cross-resistance? Symp Ser Soc Appl
Microbiol 2002; 98S– 110S.
12 Leid JG, Shirtliff ME, Costerton JW et al. Human leukocytes adhere to,
penetrate, and respond to Staphylococcus aureus biofilms. Infect Immun
2002; 70: 6339– 45.
13 Lobmann R, Schultz G, Lehnert H. Proteases and the diabetic foot
syndrome: mechanisms and therapeutic implications. Diabetes Care
2005; 28: 461–71.
14 Wagner C, Kaksa A, Muller W et al. Polymorphonuclear neutrophils in
posttraumatic osteomyelitis: cells recovered from the inflamed site lack
chemotactic activity but generate superoxides. Shock 2004; 22: 108– 15.
15 Mumcuoglu KY, Ingber A, Gilead L et al. Maggot therapy for the
treatment of diabetic foot ulcers. Diabetes Care 1998; 21: 2030– 1.
16 Sherman RA, Wyle FA, Vulpe M et al. The utility of maggot therapy for
treating chronic wounds. Am J Trop Med Hyg 1993; Suppl: 266.
17 Stoddard SR, Sherman RA, Mason BE et al. Maggot debridement
therapy. An alternative treatment for nonhealing ulcers. J Am Podiatr
Med Assoc 1995; 85: 218–21.
18 van der Plas MJA, Jukema GN, Wai SW et al. Maggot excretions/
secretions are differentially effective against biofilms of Staphylococcus
aureus and Pseudomonas aeruginosa.J Antimicrob Chemother 2008;
61: 117–22.
19 van der Plas MJA, van der Does AM, Baldry M et al. Maggot excretions/
secretions inhibit multiple neutrophil pro-inflammatory responses.
Microbes Infect 2007; 9: 507– 14.
20 Clinical and Laboratory Standards Institute. Performance Standards
for Antimicrobial Susceptibility Testing: Fifteenth Informational
Supplement M100-S15. CLSI, Wayne, PA, USA 2005.
21 Nibbering PH, Ravensbergen E, Welling MM et al. Human lactoferrin
and peptides derived from its N terminus are highly effective against
infections with antibiotic-resistant bacteria. Infect Immun 2001; 69:
1469– 76.
22 van Ogtrop ML, Mattie H, Guiot HFL et al. Comparative study of the
effects of four cephalosporins against Escherichia coli in vitro and in
vivo. Antimicrob Agents Chemother 1990; 34: 1932– 7.
23 Flemming K, Klingenberg C, Cavanagh JP et al. High in vitro
antimicrobial activity of synthetic antimicrobial peptidomimetics against
staphylococcal biofilms. J Antimicrob Chemother 2009; 63:13645.
24 Smith K, Perez A, Ramage G et al. Comparison of biofilm-associated
cell survival following in vitro exposure of meticillin-resistant
Staphylococcus aureus biofilms to the antibiotics clindamycin,
daptomycin, linezolid, tigecycline and vancomycin. Int J Antimicrob
Agents 2009; 33: 374–8.
25 Desrosiers M, Bendouah Z, Barbeau J. Effectiveness of topical
antibiotics on Staphylococcus aureus biofilm in vitro. Am J Rhinol 2007;
21: 149–53.
26 Tote K, Berghe DV, Deschacht M et al. Inhibitory efficacy of various
antibiotics on matrix and viable mass of Staphylococcus aureus and
Pseudomonas aeruginosa biofilms. Int J Antimicrob Agents 2009; 33:
525–31.
27 Howell-Jones RS, Wilson MJ, Hill KE et al. A review of the microbiology,
antibiotic usage and resistance in chronic skin wounds. J Antimicrob
Chemother 2005; 55: 143– 9.
28 Robinson W, Norwood VH. The role of surgical maggots in the
disinfection of osteomyelitis and other infected wounds. J Bone Joint
Surg Am 1933; 15: 409–12.
29 Mumcuoglu KY, Miller J, Mumcuoglu M et al. Destruction of bacteria in
the digestive tract of the maggot of Lucilia sericata (Diptera:
Calliphoridae). J Med Entomol 2001; 38: 161– 6.
van der Plas et al.
922
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
30 Sherman RA, Wyle FA, Thrupp L. Effects of seven antibiotics on the
growth and development of Phaenicia sericata (Diptera: Calliphoridae)
larvae. J Med Entomol 1995; 32: 646– 9.
31 Alder J. The use of daptomycin for Staphylococcus aureus infections in
critical care medicine. Crit Care Clin 2008; 24: 349–63.
32 English BK, Maryniw EM, Talati AJ et al. Diminished macrophage
inflammatory response to Staphylococcus aureus isolates exposed to
daptomycin versus vancomycin or oxacillin. Antimicrob Agents
Chemother 2006; 50: 2225– 7.
33 Wale LJ, Shelton AP, Greenwood D. Scanning electronmicroscopy of
Staphylococcus aureus and Enterococcus faecalis exposed to
daptomycin. J Med Microbiol 1989; 30:459.
34 Rosner K, Ross C, Karlsmark T et al. Immunohistochemical
characterization of the cutaneous cellular infiltrate in different areas of
chronic leg ulcers. APMIS 1995; 103: 293– 9.
35 Loots MA, Lamme EN, Zeegelaar J et al. Differences in
cellular infiltrate and extracellular matrix of chronic diabetic and
venous ulcers versus acute wounds. J Invest Dermatol 1998; 111:
850–7.
36 Wetzler C, Kampfer H, Stallmeyer B et al. Large and sustained
induction of chemokines during impaired wound healing in the
genetically diabetic mouse: prolonged persistence of neutrophils and
macrophages during the late phase of repair. J Invest Dermatol 2000;
115: 245– 53.
Effect of maggot excretions/secretions and antibiotics on S. aureus
923
JA
C
by guest on December 28, 2015http://jac.oxfordjournals.org/Downloaded from
... Compounds such as serine proteases, chymotrypsin, fatty acid and phormicin C are derived from maggot extract and have been introduced as anti-biofilm compounds. These compounds have also negative effects on the metabolic activity of bacterial cells, membrane, cell wall which lead to cell death (10,(37)(38)(39). Based on TTC method, it was found that the synergistic effect of FLIP7 present in the maggot extract of Calliphora vicina along with antibiotics (meropenem, ampicillin, amikacin, kanamycin) inhibits metabolic activity of S. aureus and P. aeroginosa (35). ...
... Although gentamycin has more effective on S. aureus than larvae extract (33), our data showed that minimum inhibitory dose of antibiotic (in planktonic mode) coupled with larvae extract can prevent bacterial biofilm formation. Previous studies have shown that Lucilia sericata and Calliphora vicina maggot extract work synergistically with antibiotics as interfered from the reductions in crystal violet binding to S. aureus, P. aeroginosa , and E. coli biofilms (35,38). The use of the antibiotic ciprofloxacin at levels below the MIC and the combination of daptomycin and ES have also eradicated the antibiotic resistant infection (38,41). ...
... Previous studies have shown that Lucilia sericata and Calliphora vicina maggot extract work synergistically with antibiotics as interfered from the reductions in crystal violet binding to S. aureus, P. aeroginosa , and E. coli biofilms (35,38). The use of the antibiotic ciprofloxacin at levels below the MIC and the combination of daptomycin and ES have also eradicated the antibiotic resistant infection (38,41). ...
Article
Background: This study explores the impact of Lucilia sericata maggots on the development and eradication of biofilms created by the pathogenic bacteria, Staphylococcus aureus and Pseudomonas aeruginosa. Methods: We assessed the influence of Lucilia sericata maggot extract on the viability of planktonic bacteria, the formation and disruption of biofilms, bacterial metabolic activity. Also the effect of simultaneous ES-antibiotic treatment in biofilm elimination was investigated. Additionally, the expression levels of genes associated with biofilm formation, namely LasI, psLA, agrA, and icaD was studied. Results: The results showed that ES can reduce the viability of planktonic S. aureus, significantly. Furthermore, ES of larvae fed on S. aureus-infected meat displayed the most substantial inhibition of biofilm formation (62.11% and 75.04% inhibition for S. aureus and P. aeruginosa, respectively). A similar trend was observed in biofilm destruction, with values of 56.67% and 68.50% inhibition for S. aureus and P. aeruginosa, respectively. The simultaneous application of ES of larvae that fed on S. aureus-infected meat and the minimum inhibitory concentration (MIC) of gentamicin resulted in 100% inhibition of biofilm formation by S. aureus. Notably, the group treated with ES of larvae fed on S. aureus-infected meat exhibited the most significant reduction in metabolic activity, with values of 95.03% and 68.25% for S. aureus and P. aeruginosa, respectively. The expression of LAsI and pslA genes in P. aeruginosa and the expression of agrA and icaD genes in S. aureus has decreased Conclusion: The findings of this study demonstrate that maggot extract has not only impacted the formation, but also eliminated the biofilms of S. aureus and P. aeruginosa.
... Of the 35 remaining papers, 23 were excluded through full-text screening as they failed to meet the inclusion criteria (see PRISMA flowchart Figure 1 for details). A total of 12 articles were included in this review [82][83][84][85][86][87][88][89][90][91][92][93]. ...
... Eighty-three percent (10/12) of the studies analyzed were in vitro assessments of the effects of larvae/larval secretions on biofilm [84][85][86][87][88][89][90][91]93,94], with 17% (2/12) of studies being ex vivo in nature, one using a porcine-skin-based model [92] and the other a humanskin-based model [95]. In 67% (8/12) of studies, a passive secretion collection strategy was utilized, whereby live larvae were immersed in a suitable diluent, such as sterile H 2 O or 0.9% phosphate-buffered saline (PBS) solution [84][85][86][87][88][89][90]93], whereas 17% (2/12) utilized an extraction process that involved the milling and further processing of dried L. sericata larvae [94,95], 8% (1/12) utilized a recombinant enzyme derived from L. sericata secretions [91], and 8% (1/12) applied live larvae directly to their biofilm model [92]. ...
... Eighty-three percent (10/12) of the studies analyzed were in vitro assessments of the effects of larvae/larval secretions on biofilm [84][85][86][87][88][89][90][91]93,94], with 17% (2/12) of studies being ex vivo in nature, one using a porcine-skin-based model [92] and the other a humanskin-based model [95]. In 67% (8/12) of studies, a passive secretion collection strategy was utilized, whereby live larvae were immersed in a suitable diluent, such as sterile H 2 O or 0.9% phosphate-buffered saline (PBS) solution [84][85][86][87][88][89][90]93], whereas 17% (2/12) utilized an extraction process that involved the milling and further processing of dried L. sericata larvae [94,95], 8% (1/12) utilized a recombinant enzyme derived from L. sericata secretions [91], and 8% (1/12) applied live larvae directly to their biofilm model [92]. For those studies that did not use a larval-derived recombinant (11/12), 25% (3/12) did not state the instar of larvae used for their extraction or treatment protocol [89,92,94], 25% (3/12) employed 3rd-instar larvae [85,93,95], 17% (2/12) used instar 1 and 3 larvae [86,88], and 25% (3/12) collected secretions from instar 2 and 3 larvae [84,87,90]. ...
Article
Full-text available
Chronic wounds present a global healthcare challenge and are increasing in prevalence, with bacterial biofilms being the primary roadblock to healing in most cases. A systematic review of the to-date knowledge on larval therapy’s interaction with chronic-wound biofilm is presented here. The findings detail how larval therapy—the controlled application of necrophagous blowfly larvae—acts on biofilms produced by chronic-wound-relevant bacteria through their principle pharmacological mode of action: the secretion and excretion of biologically active substances into the wound bed. A total of 12 inclusion-criteria-meeting publications were identified following the application of a PRISMA-guided methodology for a systematic review. The findings of these publications were qualitatively analyzed to provide a summary of the prevailing understanding of larval therapy’s effects on bacterial biofilm. A further review assessed the quality of the existing evidence to identify knowledge gaps and suggest ways these may be bridged. In summary, larval therapy has a seemingly unarguable ability to inhibit and degrade bacterial biofilms associated with impaired wound healing. However, further research is needed to clarify and standardize the methodological approach in this area of investigation. Such research may lead to the clinical application of larval therapy or derivative treatments for the management of chronic-wound biofilms and improve patient healing outcomes at a time when alternative therapies are desperately needed.
... Inadequate setup of experimental groups, for example, can generate data with high bias and little scientific benefit. [30,38,39] Chrysomya albiceps [40], Chrysomya megacephala (Fabricius) [40,41], Chrysomya putoria (Wiedemann) [40], Chrysomya rufifacies (Macquart) [42], Co. macellaria [42,43], L. cuprina [31,[44][45][46], L. eximia [32], L. sericata [33,34,39,[47][48][49], Musca domestica Linnaeus [50], P. terraenovae [35], S. magellanica [35,37,39,49,51,52] Action against bacterial biofilm Ca. vicina [53], L. sericata [54][55][56][57] Synergism between larval excretions/ secretions and antibiotics L. cuprina [45], L. sericata [56,58] 11. Bioprospecting and Testing of New Fly Species for Maggot Therapy ...
... Inadequate setup of experimental groups, for example, can generate data with high bias and little scientific benefit. [30,38,39] Chrysomya albiceps [40], Chrysomya megacephala (Fabricius) [40,41], Chrysomya putoria (Wiedemann) [40], Chrysomya rufifacies (Macquart) [42], Co. macellaria [42,43], L. cuprina [31,[44][45][46], L. eximia [32], L. sericata [33,34,39,[47][48][49], Musca domestica Linnaeus [50], P. terraenovae [35], S. magellanica [35,37,39,49,51,52] Action against bacterial biofilm Ca. vicina [53], L. sericata [54][55][56][57] Synergism between larval excretions/ secretions and antibiotics L. cuprina [45], L. sericata [56,58] 11. Bioprospecting and Testing of New Fly Species for Maggot Therapy ...
... Studies on biofilms have shown that, although maggot ES can degrade and disintegrate biofilms of different species, the bacteria released by these biofilms are not destroyed [176]. This was investigated further by Van der Plas et al. who indicated that biofilms resisted antibiotics alone, but the combination of antibiotics and maggot secretion (daptomycin, vancomycin, or clindamycin) caused the destruction of S. aureus biofilms and the eradication of the bacteria found in the biofilms [181]. This introduces a strategic approach to the treatment of biofilm-associated infections, whereby the use of a combination of maggot ES and antibiotics could generate more successful therapeutic outcomes than the use of either one of these methods alone [182]. ...
Article
Full-text available
Microbial pathogens and their virulence factors like biofilms are one of the major factors which influence the disease process and its outcomes. Biofilms are a complex microbial network that is produced by bacteria on any devices and/or biotic surfaces to escape harsh environmental conditions and antimicrobial effects. Due to the natural protective nature of biofilms and the associated multidrug resistance issues, researchers evaluated several natural anti-biofilm agents, including bacteriophages and their derivatives, honey, plant extracts, and surfactants for better destruction of biofilm and planktonic cells. This review discusses some of these natural agents that are being put into practice to prevent biofilm formation. In addition, we highlight bacterial biofilm formation and the mechanism of resistance to antibiotics.
... Maggots exert many effects that may be beneficial for wound healing. Earlier maggot excretions/secretions (ES) have been implicated in the breakdown of bacterial biofilms of Staphylococcus aureus and RELATIVE ABUNDANCE AND ANTIMICROBIAL ACTIVITIES Pseudomonas aeruginosa [9], [10]. Furthermore, maggots ingest and subsequently kill bacteria in their digestive tract [11], although quorum sensing regulated virulence factors from P. aeruginosa may be toxic to maggots [12]. ...
Article
Full-text available
Maggots have long been used as a traditional way of cleansing and healing gangrenous wounds. Blowflies' abundance and antibacterial activities were determined using baited traps and agar cup plate technique. The family calliphoridae was the most abundant in all the sites sampled. The total of 5755 calliphorid flies captured belonged to two genera, Lucinia robineau (81.49%) and Chrysomya megacephala (18.51%). There was a significant difference in their abundance and a positive correlation between abundance and the two species with relative humidity and rainfall and a negative correlation with temperature. The isolates from infected wounds and whitlows were Pseudomonas earoginosa, Staphylococus aureus, Klebsiella pneumoniae and Staphylococcus epidermidis. Antibacterial susceptibility screening showed that, the maggot saliva/excretion was able to inhibit the growth. The zones of inhibition recorded was Pseudomonas aeruginosa was 24mm while Ampiclox (control) 37mm; Staphylococcus aureus was 20mm while Ampiclox (control) 31mm; Klebsiella pneumoniae was 27mm while Ampiclox (control) 39mm and Staphyloccocus epidermidis was 24mm while Ampiclox (control) 28mm. The result of the antibacterial susceptibility screening also revealed that, the greatest effect of the maggot saliva/excretion was obtained against K. pneumoniae while the lowest was obtained against S. aureus. Minimum Inhibitory Concentration (MIC) was between 40mg/ml and 60mg/ml while Minimum Bactericidal Concentration (MBC) screening showed that Pseudomonas earoginosa, Staphylococus aureus, Klebsiella pneumoniae, Staphyloccocus epidermidis have the same bactericidal concentrations of 60mg/ml each while Ampiclox (control) had 40mg/ml.
... A common observation from biofilm investigations is that whilst maggot excretions and secretions are able to degrade and break down the biofilm of various species, the bacteria which are released from these biofilms are not destroyed [27,31,54,55]. This was explored further by a group of researchers who noted that biofilms resisted antibiotics alone, but found that combining a treatment of maggot secretion and antibiotics (vancomycin, daptomycin or clindamycin) resulted in both the break-down of S. aureus biofilm and the elimination of the resulting bacteria [62]. This introduces a promising approach to the treatment of biofilm-infected chronic wounds, whereby use of a combination of maggot excretions and secretions and antibiotics could result in a more successful treatment than the use of a single method alone. ...
Article
Objective: Maggot therapy (MT) or larval debridement therapy is a recognised, effective but underutilised treatment for the management of hard-to-heal wounds and infected ulcers. It is available on NHS prescription in the UK, where wound management is predominantly nurse-led. Anecdotal reports and published literature suggest that nurses may be reluctant to utilise the therapy. The aim of this study was to evaluate the feelings and opinions of nurses regarding the use of MT. Method: The first stage of this mixed-methods study was a focus group held to discuss MT and opinions of specialist nurse clinicians. Next, an anonymised web-based online survey was launched through the Nursing Times journal and distributed through social media targeting all nurses. Finally, in-depth interviews were held with specialist and generalist nurses. Results: Awareness of MT among all nurses was extremely high. A breakdown of results showed that MT was much more highly regarded by wound specialist nurses than non-wound specialist nurses. The latter exhibited a greater level of reluctance to administer the therapy, with almost one-third of these nurses surveyed saying they found maggots disgusting and that the idea of MT made their skin crawl. In-depth interviews revealed that a lack of knowledge about MT was a prime concern. Conclusion: Wound specialist nurses are more likely to embrace MT than non-wound nurse specialists, who report a varying degree of wariness to MT. Our study highlights a need for better education and training in MT for all nurses, to address issues with acceptance and willingness to treat or help treat patients with hard-to-heal wounds which are suitable for MT.
Article
Purpose In the skin, Lucilia sericata maggot excretions/secretions (ES) accelerate wound healing and limit inflammation. This study aimed to determine whether ES have similar beneficial effects at the ocular surface. Methods Human corneal epithelial cells (HCEC) were cultured with ES and cell viability was determined by the MTT assay. Additionally, mRNA expression of growth factors, antimicrobial peptides (AMPs) and cytokines was assessed by qPCR. ES ability to modulate TLR-induced IL-6 and IL-8 expression was determined by qPCR and ELISA. ES potential to promote corneal healing was evaluated in vitro by a migration assay in HCEC, and in vivo using a mouse model. Results ES did not impair HCEC viability up to 25μg/ml. Among the factors evaluated, only hBD-2 was upregulated (2.5-fold) by 1.5μg/ml ES after 6hrs (P = 0.04). In HCEC, ES reduced Poly I:C-induced IL-6 and IL-8 mRNA (P ≤ 0.001) and protein (P ≤ 0.0001) expression. A similar effect was observed with Flagellin (TLR5 agonist) but it was less robust for FSL-1 (TLR2/6 agonist) and Pam3CSK4 (TLR1/2 agonist). The greatest in vitro migration effect was observed with 6.2μg/ml ES after 44hrs where gap area compared to vehicle was 53.3 ± 3.7% vs. 72.6 ± 5.4% (P = 0.001). In the mouse model, the maximum healing effect was present with 1.5μg/ml ES after 12hrs with a wound area of 19.0 ± 2.7% vs. 60.1 ± 21.6% (P = 0.003) or 77% reduction of the wound area compared to the negative control. Conclusions ES significantly reduce in vitro TLR-induced production of inflammatory cytokines and promote corneal wound healing.
Article
Full-text available
The aim of the study was to investigate the antimicrobial effect of different antibiotics and synthetic antimicrobial peptidomimetics (SAMPs) on staphylococcal biofilms. Biofilms of six staphylococcal strains (two Staphylococcus haemolyticus, two Staphylococcus epidermidis and two Staphylococcus aureus isolates) were grown for 24 h in microtitre plates. They were washed and treated for 24 h with different concentrations of linezolid, tetracycline, rifampicin and vancomycin and four different SAMPs. After treatment, the redox indicator Alamar Blue was used to quantify metabolic activity of bacteria in biofilms, and confocal laser scanning microscopy with LIVE/DEAD staining was used to further elucidate any effects. At MIC levels, rifampicin and tetracycline showed a marked reduction of metabolic activity in the S. epidermidis and S. haemolyticus biofilm. Linezolid had a moderate effect and vancomycin had a poor effect. MIC x10 and MIC x100 improved the antimicrobial activity of all antibiotics, especially vancomycin. However, metabolic activity was not completely suppressed in strong biofilm-producing strains. At MIC x10, the three most effective SAMPs (Ltx5, Ltx9 and Ltx10) were able to completely eliminate metabolic activity in the S. epidermidis and S. haemolyticus biofilms, which was also confirmed by complete cell death using confocal laser scanning microscopy investigations. Although none of the Ltx SAMPs could fully suppress metabolic activity in the S. aureus biofilm, their effect was superior to all tested antibiotics. SAMPs had superior antimicrobial activity in staphylococcal biofilms compared with conventional antibiotics and are potential new therapeutic agents for biofilm-associated infections.
Article
Full-text available
A thigh muscle infection induced with Escherichia coli in irradiated mice was used as a model to compare the in vivo pharmacodynamics of the antibacterial effect of four cephalosporins (i.e., cefepime, ceftriaxone, ceftazidime, and cefoperazone) with the in vitro antibacterial pharmacodynamics of these drugs. The following in vitro pharmacodynamic parameters were determined: the maximum effect as a measure for efficacy, the 50% effective concentration as a parameter for potency, and the slope of the concentration-effect relationship. For analysis of the in vivo antibacterial pharmacodynamics, the same parameters were applied for the dose instead of the concentration. For the detection of a relationship between concentration and antibacterial effect in vivo, we determined the pharmacokinetics of the four cephalosporins in the plasma of mice. The results showed that, in general, there is a direct relationship between the in vivo and in vitro pharmacodynamics of these cephalosporins. The maximum effects of cefepime, ceftazidime, and cefoperazone were approximately similar in vivo and in vitro. The sequence of potency of these drugs was, in descending order, cefepime, ceftazidime, and cefoperazone. Ceftriaxone differed from the other three cephalosporins in that it displayed unexpected in vivo pharmacodynamics. Ceftriaxone was just as efficacious as the other three in vitro, but its maximum effect in vivo was much lower. This relatively low maximum effect of ceftriaxone in vivo was not explained by the pharmacokinetic characteristics of the drug. From the present results it can be concluded that the in vitro efficacy of cephalosporins does not necessarily have a predictive value for the in vivo efficacy.
Article
BACKGROUND: In chronic wounds, biofilms probably play a vital role in protecting bacteria from host defenses and antimicrobial medications by creating a barrier of exopolysaccharide that is difficult for the immune system and antibiotics to penetrate. A biofilm consists of an exopolysaccharide matrix that is produced and secreted by certain species of bacteria. OBJECTIVE: The purpose of this study was to visualize and time the progressing growth of a biofilm by a wound-isolated Pseudomonas aeruginosa. METHODS: P. aeruginosa that was initially isolated from a human burn wound was allowed to grow a biofilm in vitro. We used a modified Congo red staining technique to demonstrate the sequential development of a mature biofilm as examined by light microscopy. RESULTS: We show that the exopolysaccharide of the developing biofilm is visible in just 5 hours after inoculation and has the characteristics of a mature biofilm by 10 hours. CONCLUSION: The rapidity of biofilm growth suggests that bacteria in wounds possess the capacity of producing this shield against antibiotics and immune effector cells early in the infection process. Therefore, efforts to prevent or slow the proliferation of bacteria and biofilms should occur soon after a wound is created. Additionally, this staining technique can be used to demonstrate the ability of agents to slow biofilm growth or to interrupt formed biofilm and may be useful in future studies of chronically infected wounds. A BIOFILM is produced by certain species of bacteria and consists of an exopolysaccharide (EPS) matrix, which is composed of very long, thin molecular chains that can associate in a number of different ways. 1 Biofilms can act as protective barriers for bacteria by shielding them from antimicrobial medications and the immune system, and they appear to be a vital component in resistant bacterial colonization. The biofilm forms when planktonic bacteria attach themselves to a surface and begin to secrete their EPS. The EPS from many nearby bacteria can convalesce and form a thick slime layer, or biofilm, that shelters the bacteria from host defenses and antibiotics (Figure 1). Figure 1Image Tools Biofilms have been found to exist in many diseases, most notably cystic fibrosis that is chronically infected with a biofilm-secreting Pseudomonas aeruginosa. 2 Biofilms have also been demonstrated in urinary tract infections, 3,4 chronic ear infections, 5–7 and peptic ulcers. 8 The presence of biofilms in chronic wounds has been hypothesized by investigators. 9 Serralta et al. 10 used bacteria retrieval results to suggest that P. aeruginosa forms a biofilm in vivo by 72 hours after inoculation. Using a well-established porcine model, 11–16 partial-thickness wounds were inoculated with P. aeruginosa, and the bacteria were quantified after either scrubbing or flushing the wound with saline. The scrub technique yielded more bacteria, indicating bacterial wound attachment and biofilm formation. Investigators examined the wounds for biofilm only at 72 hours and therefore did not pinpoint the exact time necessary for bacteria to form biofilms in vivo within that time frame. The goals of this study were to visualize a growing biofilm in vitro from a wound-isolated P. aeruginosa using a modified staining technique and to determine the specific time period needed to form a mature biofilm from a bacterial concentration of 105 colony forming units—a quantity that is often used to define the presence of infection.
Article
In vitro biofilm-producing capacity in isolates of Staphylococcus aureus and Pseudomonas aeruginosa collected from the sinus cavities after endoscopic sinus surgery (ESS) are associated with a poor outcome in patients with chronic rhinosinusitis (CRS). However, conventional oral antibiotic therapy is frequently ineffective in eradicating bacteria in the biofilm form. Increasing the concentration of antibiotics may offer a means of countering this resistance. The aim of this study was to determine the in vitro activity of moxifloxacin (MOXI) against S. aureus in biofilm form (recovered from patients with CRS at least 1 year post-ESS). This study was performed in a research microbiology laboratory, where five isolates of S. aureus with known biofilm-forming capacity were cultured in Tryptic Soy Broth 0.5% glucose in 96-well plates at 37 degrees C for 24 hours. After visual confirmation of biofilm formation, plates were incubated in phosphate-buffered saline (PBS) or with MOXI at concentrations of 0.1x, Ix, 100x, and 1000x minimal inhibitory concentration (MIC) for an additional 24 hours. Biofilm from 3 wells of each concentration were collected and sonicated and the number of viable bacteria was determined by serial dilution and plating. After incubation, the number of viable bacteria was similar for nontreated and MOXI-treated biofilms at MIC and sub-MIC levels. However, MOXI at 1000x (0.1-0.2 mg/mL) gave a 2 to 2.5 log reduction in number of viable bacteria. In vitro results show that increased concentrations of antibiotics, easily attainable in topical solutions, are effective in killing bacteria in bacterial biofilms. This suggests a role for topical antibiotic therapies in the treatment of biofilm infections.
Article
Diabetes is a common disease that is associated with numerous complications, including foot ulceration and amputation. In diabetic patients, the incidence of foot ulcers ranges from 1.0% to 4.1%, and the incidence of lower-extremity amputations ranges from 2.1 to 13.7 per 1000. Risk factors for developing foot ulcers and subsequent amputation include neuropathy, peripheral vascular disease, and trauma. To reduce these complications, several preventive strategies have been devised, from reducing risk factors to improving treatment and management.
Article
In diabetic patients, wound healing is impaired. We studied the pathogenesis behind this clinical observation by characterizing the pattern of deposition of extracellular matrix (ECM) molecules and the cellular infiltrate in chronic (>8 wk) diabetic wounds, compared with chronic venous ulcers and an acute wound healing model. Punch biopsies were obtained from the chronic ulcer margins and control samples were collected from upper leg skin 5, 19, 28 d and 12 and 18 mo postwounding (p.w.). T cells, B cells, plasma cells, granulocytes and macrophages, and the ECM molecules fibronectin (FN), chondroitin sulfate (CS), and tenascin (TN) were visualized using immunohistochemical techniques. Expression of FN, CS, and TN was detected in dermal tissue early in normal wound healing (5-19 d p.w.). Abundant staining was seen 3 mo p.w., returning to prewounding levels after 12-18 mo p.w. In the dermis of chronic diabetic and venous ulcers with a duration of 12 mo or more, a prolonged presence of these ECM molecules was noted. Compared with normal wound healing: (i) the CD4/CD8 ratio in chronic wounds was significantly lower (p < 0.0027) due to a relatively lower number of CD4+ T cells; (ii) a significantly higher number of macrophages was present in the edge of both type of chronic ulcers (p < 0.001 versus day 29 p.w.); and (iii) more B cells and plasma cells were detected in both type of chronic wounds compared with any day in the acute wound healing model (p < 0.04 for CD20+ and p < 0.01 for CD79a+ cells). These data indicate that important differences exist in the cellular infiltrate and ECM expression patterns of acute, healing versus chronic wounds, which may be related to the nonhealing status of chronic wounds.
Article
Both bacteria and the matrix are essential for the development of biofilms. Antimicrobials should therefore be tested against both components. The aim of this study was to determine the structure-activity relationships of different antibiotics against biofilm-forming Staphylococcus aureus and Pseudomonas aeruginosa strains using in vitro biofilm discriminatory assays. Only four of twelve antibiotics showed efficacy against S. aureus biofilms. Rifampicin had a 50% inhibitory activity both against the matrix and bacteria at 16 x the minimum bactericidal concentration (MBC). Polymyxin B killed nearly all bacteria at 8 x MBC, but left the matrix undisturbed. Both P. aeruginosa biofilms responded differently to antibiotic treatment. Rifampicin showed the greatest activity, with 100% killing of microorganisms combined with 91% destruction of the matrix at the MBC. In conclusion, rifampicin showed the highest activity on biofilm matrix and bacteria in S. aureus and P. aeruginosa biofilms. Our results also indicated that biofilm viable mass was more susceptible to treatment than the biofilm matrix, which is mainly responsible for biofilm persistence. Future research should specifically focus on compounds destroying the matrix that can be used as an adjunct to antibiotic therapy.
Article
The efficacy of commonly used antistaphylococcal antimicrobials (clindamycin, linezolid and vancomycin) and recently developed antibiotics (daptomycin and tigecycline) was compared against clinical isolates of meticillin-resistant Staphylococcus aureus (MRSA). Minimum inhibitory concentrations (MICs), minimum bactericidal concentrations, time-kill kinetics and biofilm-associated cell survival were examined for 12 clinical isolates of MRSA treated with each antibiotic. The MIC ranges for daptomycin, linezolid, tigecycline, clindamycin and vancomycin were 0.06-0.25, 1-2, 0.06, 0.125-1024 and 0.5-1 microg/mL, respectively. Daptomycin and vancomycin were bactericidal following 6h of incubation with planktonic cells, whilst clindamycin, linezolid and tigecycline were bacteriostatic. None of the antibiotics killed 100% of biofilm-associated cells. Mean cell survival in biofilms treated with clindamycin, daptomycin, linezolid, tigecycline and vancomycin was 62%, 4%, 45%, 43% and 19%, respectively. Although all antibiotics were effective against planktonic staphylococcal populations, vancomycin and daptomycin possessed superior activity against biofilm-associated cells.
Article
The novel lipopeptide antibiotic, daptomycin, at a concentration of 8 mg/L, caused gross morphological changes in both a methicillin-sensitive and a methicillin-resistant strain of Staphylococcus aureus and in a strain of Enterococcus faecalis. The earliest (after 1 h) surface lesion observed was the appearance of boss-like processes randomly distributed on the cell surface. Later, grossly deformed bacteria were seen and in two of the three bacteria prolonged exposure led to degeneration of the cells into an amorphous syncytial mass. Omission of calcium (which is known to potentiate the activity of daptomycin) from the culture medium did not affect the morphological response to an inhibitory concentration of the antibiotic.