You are on page 1of 11

Microbial Pathogenesis 146 (2020) 104249

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

Inhibition of biofilm and virulence properties of Pseudomonas aeruginosa by T


sub-inhibitory concentrations of aminoglycosides
Fazlurrahman Khana, Jang-Won Leeb, Aqib Javaidc, Seul-Ki Parka, Young-Mog Kima,b,∗
a
Institute of Food Science, Pukyong National University, Busan, 48513, South Korea
b
Department of Food Science and Technology, Pukyong National University, Busan, 48513, South Korea
c
Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, 201306, UP, India

A R T I C LE I N FO A B S T R A C T

Keywords: Aminoglycosides are a commonly used class of antibiotics; however, their application has been discontinued due
Aminoglycosides to the emergence of multi-drug resistance bacterial strains. In the present study, the subinhibitory concentrations
Antibiofilm (sub-MIC) of several aminoglycosides were determined and tested as an antibiofilm and for their anti-virulence
Anti-virulence properties against Pseudomonas aeruginosa PAO1, which is an opportunistic foodborne pathogen. P. aeruginosa
LasR
PAO1 exhibits multiple mechanisms of resistance, including the formation of biofilm and production of several
Molecular docking
Pseudomonas aeruginosa
virulence factors, against aminoglycoside antibiotics. The sub-MIC of these antibiotics exhibited biofilm in-
hibition of P. aeruginosa in alkaline TSB (pH 7.9). Moreover, various concentrations of these aminoglycosides
also eradicate the mature biofilm of P. aeruginosa. In the presence of sub-MIC of aminoglycosides, the mor-
phological changes of P. aeruginosa were found to change from rod-shaped to the filamentous, elongated, and
streptococcal forms. Similar growth conditions and sub-MIC of aminoglycosides were also found to attenuate
several virulence properties of P. aeruginosa PAO1. Molecular docking studies demonstrate that these ami-
noglycosides possess strong binding properties with the LasR protein, which is a well-characterized quorum-
sensing receptor of P. aeruginosa. The present study suggests a new approach to revitalize aminoglycosides as
antibiofilm and antivirulence drugs to treat infections caused by pathogenic bacteria.

1. Introduction have found that targeting these virulence factors enables the use of
conventional antibiotics and the simultaneous reduction of their se-
Pseudomonas aeruginosa is a pathogenic bacterium that has effec- lective pressure for resistance mutations [14–16]. Thus, attenuation of
tively emerged as a cause of infections in human hosts via its biofilm virulence properties, along with the inhibition of biofilm formation of
lifestyle [1]. Further, it has been identified as a nosocomial biofilm- P. aeruginosa, is a promising therapeutic strategy, as evidenced by
forming pathogen that causes chronic infections in the lungs of patients several previous studies [17,18]. Although several potential drugs that
with cystic fibrosis [2] and is related to infections that are associated act as antibiofilm, antibacterial, and antivirulence agents have been
with otitis and burn wounds [3,4]. The formation of a biofilm an as- either isolated from biological sources or chemically synthesized, more
semblage of bacterial cells that is encased by a self-produced structure drugs need to be discovered [19–27].
that comprises extracellular polymeric substances (EPS), such as exo- Aminoglycosides are a commonly used class of conventional anti-
polysaccharides, protein, and extracellular-DNA (e-DNA) is well-re- biotics against Gram-negative bacteria, along with β-lactams and
cognized as a common resistance property against antibiotics and ad- fluoroquinolones [28]. Aminoglycosides were previously reported to be
verse environmental conditions [5,6]. Biofilm formation by P. limited in their penetration through the biofilm matrix and cell wall
aeruginosa induced major chronic infections in humans and animals and and are encountered by modifying enzymes and efflux pumps [29–33].
constitutes a bacterial mechanism that allows resistance against mul- Furthermore, the misuse and overuse of aminoglycosides over the past
tiple types of conventional antibiotics, including aminoglycosides decades have resulted in resistance development via several means,
[7–9]. In P. aeruginosa, biofilm formation is also associated with the including enzymatic modification (i.e. acetyltransferases, phospho-
production of multiple virulence factors that serve vital roles in bac- transferases, and nucleotidyltransferases), binding target modification,
terial pathogenesis and survival [10–13]. Furthermore, recent studies reduced drug permeability, and efflux system [31,34–36]. Currently,


Corresponding author. Department of Food Science and Technology, Pukyong National University, Busan, 48513, South Korea.
E-mail address: ymkim@pknu.ac.kr (Y.-M. Kim).

https://doi.org/10.1016/j.micpath.2020.104249
Received 30 January 2020; Received in revised form 19 March 2020; Accepted 4 May 2020
Available online 11 May 2020
0882-4010/ © 2020 Elsevier Ltd. All rights reserved.
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 1. Chemical structure of the aminoglycosides tested in the present study.

though fluoroquinolone is commonly used to treat P. aeruginosa, several 2. Materials and methods
reports demonstrate that the P. aeruginosa bacteria exhibit enhanced
biofilm formation and production of virulence factors in the presence of 2.1. Bacterial strains, culture conditions and chemical reagents
the drug [37,38]. Therefore, expanding the variability of drugs options
and targets is highly important for the sustainable use of conventional The human pathogenic bacterium Pseudomonas aeruginosa PAO1
antibiotics in the future [39,40]. KCTC1637 was obtained from Korean Collection for Type Cultures
As such, in the attempts to revitalize drug action, several alternative (KCTC, Daejeon, Korea). The growth condition and culture media for
strategies have been developed over the past few decades [41,42]. One this strain was followed according to our recent report [45,46]. These
of these strategies is to modify the culture environment to become more bacterial cultures were allowed to grow in tryptic soy broth (TSB; Difco
alkaline, which was particularly effective in enabling gentamicin, to- Laboratory Inc., Detroit, MI, USA) with pH of 7.9. The growth condition
bramycin, and streptomycin to exhibit antibiofilm action against P. of these bacterial strains was 35 °C temperature under aerobic condi-
aeruginosa [43,44]. Due to their similarity in structure, several other tion. The aminoglycoside antibiotics such as amikacin, gentamicin,
aminoglycosides [e.g. amikacin, gentamicin, kanamycin, neomycin B, kanamycin, neomycin, netilmicin, and paromomycin aminoglycoside
paromomycin, and netilmicin (chemical structures presented in Fig. 1)] antibiotics were purchase from Sigma-Aldrich Co. (St. Louis, MO, USA).
have been selected in the present study to evaluate their antibiofilm and The stocks of each antibiotic were prepared in acidified water (pH 5.9).
antivirulence activities under an alkaline environment (pH 7.9). Fur-
thermore, in vitro phenotypic studies were also validated by molecular
docking to elucidate quorum-sensing (QS) suppression mechanisms 2.2. Minimum inhibitory concentration (MIC)
using these aminoglycosides. From the present study, considerable in-
sight is gained on (1) changes in biofilm architecture and biofilm- The methodology that was employed to determine the MIC of each
forming cellular morphology upon exposure to drugs, as revealed by aminoglycoside against P. aeruginosa was adapted from a previous re-
microscopic assays, and (2) drug anti-virulent activities by their inter- port [44,47,48]. Briefly, the MIC determination was performed by in-
actions with QS signal molecules (i.e. LasR), as revealed by a molecular cubating the bacterial seed culture (with an initial OD of 0.05) with
docking assay. different concentrations of each antibiotic. The cell culture (240 μl) was
placed in a 96-well microtiter plate, which contained different con-
centrations of aminoglycosides. The plate was kept under shaking
conditions (567 cycles per min) in a microplate reader for 24 h at 35 °C.
At the end of the incubation period, the optical densities of the bacterial
cells were measured using the microplate reader (BioTek, Winooski, VT,
USA) at a wavelength of 600 nm (OD600).

2
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

2.3. Biofilm formation assays 35 °C for 12 h), which are treated with the sub-MIC of each ami-
noglycoside, was evaluated by colorimetric assays, as explained by
Crystal violet staining was used to evaluate biofilm formation, as Essaar et al. [52]. The pink color that is obtained after the extraction
described in previous studies [45,49]. Briefly, the cell culture of the were quantified by OD measurement at 520 nm. The pyoverdine esti-
bacteria with an initial OD of 0.05 was incubated with the sub-MIC of mation was based on the florescence measurement of the cell-free su-
each aminoglycoside in TSB (pH 7.9) media in a 96-well microtiter pernatant, as explained in an earlier study [53]. The cell free super-
plate. After 24 h of incubation at 35 °C, the number of total cells (i.e. natant was obtained from the P. aeruginosa cell culture that was grown
planktonic and biofilm cells) was enumerated by measuring the OD at overnight and treated with the sub-MIC of aminoglycosides and mea-
600 nm. After measuring the OD, the free-floating planktonic cells were sured OD at 405 nm. LasA protease activity of the P. aeruginosa culture
discarded and the adhered cells that were present in the titer wells were that was treated with the sub-MIC of the aminoglycosides was de-
washed three times with distilled water. Furthermore, the adhered cells termined based on the digestion of azocasein, as described earlier [12].
were stained with aqueous crystal violet (0.1%) and incubated for The filtered supernatant was mixed with 0.3% azocasein and incubated
20 min. After staining, the crystal violet solution was discarded from for 4 h at 4 °C to allow the digestion of azocasein. The undigested
each titer well and the stained cells were washed three times with azocasein precipitated with trichloroacetic acid (10%) and was then
distilled water and air dried. Moreover, the dye-bound cells were wa- centrifuged to obtain the cell-free supernatant. Furthermore, the su-
shed three times with distilled water and solubilized using 95% ethanol. pernatant was treated with 1 M NaOH, followed by OD measurement at
The quantitative nature of the stained cells was measured using a mi- 440 nm.
croplate reader (BioTek, Winooski, VT, USA) at 570 nm.
In addition, the eradication effect of different concentrations 2.6. Analysis of flagellar motility
(above-MIC, MIC, and sub-MIC) of aminoglycosides on the mature
biofilm of P. aeruginosa was evaluated, as described in previous studies The impact of the sub-MIC of all aminoglycosides on flagellar-
[45,50]. Firstly, the cell culture (initial OD of 0.05) of P. aeruginosa was mediated motilities (e.g. swimming and swarming) in P. aeruginosa was
placed in the 96-well microtiter plate to allow mature biofilm formation evaluated using a previously described method [12]. The swarming
at 35 °C for 24 h of incubation under non-shaking conditions. After motility was determined on the surface of semi-solid agar media with
discarding the planktonic cells, the established mature biofilms in the the composition of 0.5% casamino acids, 0.5% glucose, and 0.4% Bacto
microtiter plate were washed twice with sterile TSB. The surface-at- Agar, as prepared in the Luria Britani (LB) broth. Similarly, another
tached cells in every well were treated with different concentrations of flagellar motility, i.e. swimming, was also performed in the solid agar
each aminoglycoside and incubated for 12 h at 35 °C. After incubation, media, but with a different medium composition that consisted of 0.2%
the free-floating planktonic cells were removed from each well and casamino acids, 0.3% Bacto Agar, and 30 mM glucose. After auto-
washed three times with distilled water, followed by staining with claving these media, the sub-MIC of each aminoglycoside were added
crystal violet (0.1%) for 20 min. The adhered cells that were stained and poured in Petri dishes to allow solidification. The P. aeruginosa cell
with crystal violet were washed three times with distilled water and air culture (5 μl) that was grown overnight was placed in the center of each
dried. These stained cells were dissolved with 95% ethanol, and their solid agar plate and incubated for 24 h at 35 °C. The swarming and
quantitative values were determined using a plate reader at 570 nm. swimming motilities of the cells on the surface of the agar plate were
Each experiment of the biofilm assay was performed in triplicate. estimated by measuring the diameter (cm) of the traveled cells.

2.4. Analysis of biofilm architecture using a scanning electron microscope 2.7. Molecular docking analysis

Changes in the biofilm architecture and cell morphology of P. aer- AutoDock MGL tools-4.0 was used for performing docking studies as
uginosa in the presence of each aminoglycoside at sub-MICs were vi- it is one of the most frequently used docking tools [54]. Molecular in-
sualized using a scanning electron microscope (SEM) [44,50]. The SEM teractive visualization and analysis of molecular structures were carried
samples of the biofilm cells were prepared according to the aforemen- out in BIOVIA Discovery Studio 2019 [55]. The 3-D structure of LasR
tioned procedure. The cells of P. aeruginosa were allowed to grow on the with 1.4A resolution was retrieved from the protein data bank (ID:
surface of the nylon membrane (0.5 × 0.5 cm) in the presence of the 3ix3). Preprocessing of LasR, such as optimization, energy minimiza-
sub-MIC of aminoglycosides in a 24-well titer plate. The titer plate was tion and addition of hydrogen atoms, was done by SPDBV 4.10 [56].
incubated for 24 h at 35 °C. After incubation, the cells were fixed by This involves removal of all the heteroatoms, its natural ligand i.e. 3-
adding formaldehyde and glutaraldehyde. The free-floating cells that oxo-dodecanoylhomoserine lactone (OdDHL) and coordinates added by
were present in the wells were carefully removed, while the attached SPDBV (Swiss PDB viewer) software in the LasR pdb file. 2D ligand
cells on the nylon membrane were gently washed three times using structures were generated in ChemSketch by using corresponding Inchl
phosphate buffered saline (pH 7.4). After washing, biofilm cells were key values of ligands retrieved from Pubchem. Open Babel software was
dehydrated by successive concentrations of ethanol (50, 70, 80, 95, and used to convert ligand structures from 2D to 3D. During the format
100%) and freeze-dried using a freeze dryer (model no. FD8518, il- conversion, in order to prevent any structural changes in the ligands,
ShinBiobase Co. Ltd., Korea). After fixing on the surface of the SEM the chiral centers of each ligand were selected in their original state.
stubs, coating of the cells was carried out with white gold for 120 s Following active site residues were used for the generation of Grid box
using an ion-sputter (E−1010, Hitachi, Tokyo, Japan) The biofilm cells Leu-36, Tyr-47, Tyr-56, Tyr-64, Asp-73, Thr-75, Ile-52, Ala-50, Gly-38,
were then visualized using the JSM-6490LV microscope (JEOL, Tokyo, Leu-39, Val-76, Cys-79, Trp-88, Tyr-93, Leu-110, Leu-125, Gly-126,
Japan) at a magnification of × 5000 and a voltage of 15 kV. Thr-80, and Ser-125 with coordinates of X:8.833, Y:5.881, and Z:20.282
to specify the possible interaction area of ligands [57,58]. Each ami-
2.5. Analysis of virulence factors noglycoside was docked into LasR protein grid using Lamarckian ge-
netic algorithm. Also, 3-oxo-C12-HSL (OdDHL), a natural ligand of
Quorum sensing (QS) regulates various virulence properties such as LasR, was used as a positive control.
pyocyanin production, pyoverdine production, and protease activity,
within P. aeruginosa and were determined in the presence of the sub- 2.8. Statistical analysis
MIC of aminoglycosides. The procedure used for analysis of virulence
activity was carried out according to previously reported methods Each graph in the manuscript were plotted using GraphPad Prism
[44,51]. Pyocyanin production in the cells of the culture (incubated at 7.0 (GraphPad Software Inc., San Diego, CA). Furthermore, each data

3
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 2. MIC determination of the aminoglycosides against P. aeruginosa PAO1. (A) Bactericidal effect of amikacin and netilmicin and (B) Bactericidal effect of
kanamycin, paromomycin, neomycin and gentamicin.

was also analyzed by performing one-way ANOVA followed by concentration.


Dunnett's posttest and the results were presented as means ± SD. In other words, the formation of chain, elongation, and filamentous
morphology at a high sub-MIC results in a slightly higher value of
3. Results biofilm formation. The results demonstrate that the sub-MIC of each
aminoglycoside exhibits significant inhibition of biofilm formation. To
3.1. Determining the minimum inhibitory concentrations of aminoglycosides determine the dispersal of the P. aeruginosa mature biofilm by each
aminoglycoside, a mature biofilm was allowed to form for 24 h. The
To determine the MIC and decide the sub-MIC of aminoglycosides, mature biofilm was treated with different concentrations (above-MIC,
an alkaline culture (TSB, pH 7.9) was used to grow P. aeruginosa. MIC, and sub-MIC) of each aminoglycoside and incubated for 12 h to
Different concentration ranges of each aminoglycoside were used to allow eradication. Results indicate that each aminoglycoside exhibits
treat P. aeruginosa, as presented in Fig. 2A and B. Under the afore- significant dispersal of the mature biofilm (Fig. 5). The eradication
mentioned conditions, the MIC of aminoglycosides was identified as process was found to be dependent on concentration, as it is higher at
2 μg/ml for amikacin and 4 μg/ml for netilmicin (Fig. 2A), and 128 μg/ above-MIC levels and lower at sub-MIC levels. The concentration-de-
ml for kanamycin, 128 μg/ml for paromomycin, 16 μg/ml for neo- pendent eradication of biofilm may be due to the electrostatic inter-
mycin, and 8 μg/ml for gentamicin (Fig. 2B). Subsequently, the sub-MIC actions between the positively charged amino group of the aminogly-
levels were chosen based on these results and hover below the MIC. The coside and the negatively charged component of the biofilm matrix.
sub-MIC of each aminoglycoside was used to study the biofilm inhibi-
tion and attenuation of the virulence properties of P. aeruginosa. 3.3. Inhibition of virulence properties

3.2. Biofilm inhibition and mature biofilm dispersion by aminoglycosides Biofilm formation, along with most other virulence properties of P.
aeruginosa that emerge during biofilm formation, are regulated with
The impact of the sub-MIC of each aminoglycoside on biofilm for- involvements by the QS signaling system. Thus, in the present study,
mation in the TSB (pH 7.9) was evaluated. The results demonstrate that along with seeking to understand drug inhibitory activity against the
each aminoglycoside inhibited the formation of biofilm, and inhibition bacterial biofilm, we also sought to examine their effects on pyocyanin
was not a concentration-dependent process (Fig. 3). As observed in production, pyoverdine production, and LasA protease activity.
Fig. 3, the biofilm inhibitory effect varied at certain sub-MIC levels of Pyocyanin and pyoverdine are virulent factors that are produced by P.
each aminoglycoside. However, examination of the biofilm cell and aeruginosa and contribute to bacterial virulence and pathogenesis.
architecture by SEM found that the morphology of the cells changed Results have shown that high sub-MIC levels of all tested antibiotics
with exposure to different sub-MIC levels of the aminoglycosides effectively inhibited the synthesis of pyocyanin, while both high and
(Fig. 4). Furthermore, aminoglycosides at different sub-MIC levels low sub-MIC concentrations of these drugs are effective in inhibiting
caused different forms of morphology of P. aeruginosa. Various changes pyoverdine synthesis (Fig. 6A and B). Similarly, a high sub-MIC of each
in morphology were observed, including chain formation, elongation, aminoglycoside also reduces LasA protease activity significantly
filamentation, and streptococcal form development. For instance, ami- (Fig. 6C). At the initiation of the biofilm formation process, P. aerugi-
kacin at a high sub-MIC (1 μg/ml) shows filamentation (Fig. 4). Kana- nosa chemotactically moved to the biotic or abiotic surfaces and be-
mycin also induced a slight elongation of the cells at a high sub-MIC came sessile in nature. In the present study, the flagellar motility, i.e.
(64 μg/ml), whereas at a low sub-MIC (4 μg/ml), the cell completely swarming and swimming, was monitored in the presence of each ami-
changed to the streptococcal form. In the presence of netilmicin ami- noglycoside at their sub-MIC levels. Each aminoglycoside at a high sub-
noglycoside, the cells were found to change to the streptococcal form at MIC significantly attenuated swarming motility compared to in the
all tested sub-MIC levels. In the presence of a high sub-MIC (8 μg/ml) control group (Fig. 7). A similar finding was obtained regarding
for neomycin, the cellular morphology constituted the chain form, swimming motility inhibition by all aminoglycosides, in which high
whereas at a low sub-MIC (0.5 μg/ml), all bacterial cells were in the sub-MIC levels reduced bacterial motility (Fig. 8). Overall, both the
streptococcal form. In the presence of paromomycin, the streptococcal swimming and swarming motilities of P. aeruginosa were more effec-
form was found at a low sub-MIC (32 μg/ml). Interestingly, upon ap- tively inhibited by the high sub-MIC of all tested aminoglycosides.
plication of gentamicin at a high sub-MIC (1 μg/ml), the cells were
highly elongated; however, at a low concentration (0.25 μg/ml), only 3.4. Interaction of aminoglycosides with the LasR amino acid residue
rod-shaped morphology was observed. Thus, based on the SEM ana-
lysis, it is concluded that the value of biofilm formation is changed due The in-vitro anti-quorum-sensing-mediated inhibition of virulence
to changes in cellular morphology, and thus is independent of properties of P. aeruginosa was further validated using a molecular

4
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 3. Effects on biofilm formation and cell growth of P. aeruginosa PAO1 in the presence of sub-MIC of aminoglycosides. (A) Amikacin, (B) Gentamicin, (C)
Kanamycin, (D) Neomycin, (E) Netilmicin and (F) Paromomycin. *P < 0.05 and **P < 0.01 were accepted as statistically significant and ns indicated non-
significance.

Fig. 4. Microscopic visualization of biofilm architecture and cell morphology by SEM in the presence of different sub-MIC of aminoglycosides.

5
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Residues, such as Tyr-56, Trp-60, Asp-73, and Ser-129, were found


to be common in most interactions, which is consistent with the re-
sidues that are involved in the binding of OdDHL. Most compounds
interacted with LasR via a common residue, i.e. Trp-60, which seems to
be a potential drug target. However, this finding requires further ex-
perimental validation. Apart from hydrogen bonding, most compounds
exhibited different types of stacking interactions with the residues of
the active site of LasR, especially with aromatic amino acids, such as
Tyr-47 and Tyr-64. Further, this may be yet another reason for the large
negative values of the binding energy. The 2D- and 3D-docked con-
formation of each aminoglycoside into the active site of LasR, along
with electrostatic interactions, such as H-bonds, van der Waals forces,
Pi-Pi stacking, Pi-alkyl stacking, and amide-Pi stacking, are represented
in Fig. 9.
Based on the docking studies, the aminoglycosides can be concluded
to interact with the QS receptor LasR. Based on the molecular docking
studies, these aminoglycosides can be used as potential antibiofilm and
virulence-attenuating drugs against P. aeruginosa.

4. Discussion

Currently, alternative strategies have considered revitalizing these


drugs using their sub-MIC levels, shifting to other targets that are clo-
sely associated with biofilm formation, which includes the quorum-
sensing system and their down-regulated virulence factors, and using
various drug combinations (i.e. combinatory strategy) [7,15,59]. The
present study was designed to revitalize aminoglycosides as antibiofilm
and virulence-attenuating drugs against P. aeruginosa. Furthermore, due
to previous evidence that suggests the supportive role of an alkaline pH
(pH 7.9) of the culture environment in antibiofilm activity of ami-
noglycosides (e.g. gentamicin, tobramycin, and streptomycin) [44,60],
this condition was also applied in the present study. Before testing the
biofilm inhibition and virulence-attenuating properties against P. aer-
uginosa with these aminoglycosides, their MIC and sub-MIC values have
been determined using the alkaline pH (7.9). Results show that each
aminoglycoside exhibits different MIC values, which is in accord with
previous findings on tobramycin and streptomycin [44]. All the tested
aminoglycosides at their sub-MIC exhibited significant biofilm inhibi-
tion, although the inhibition was not dependent on concentration. This
effect is in accord with the biofilm inhibition properties of P. aeruginosa
by tobramycin and tetracycline in alkaline media, as recently reported
[44]. The biofilm inhibition properties by sub-MIC of aminoglycosides
were also visualized by SEM analysis, in which results demonstrate that
cells are less dense on the surface of the nylon membrane compared to
the control (Fig. 4). Interestingly, the morphology of the observed cells
was found to vary from rod-shaped to the filamentous, elongated, and
Fig. 5. Dispersion of established 24-h mature biofilm of P. aeruginosa PAO1 by streptococcal forms. Kanamycin and netilmicin caused P. aeruginosa
various concentrations of aminoglycosides. (A) Amikacin and netilmicin, (B) biofilm cells to change to streptococcal cell shapes, whereas gentamicin
Gentamicin and neomycin, and (C) Kanamycin and paromomycin. *P < 0.05
caused the bacterial cells to elongate at high sub-MIC levels (1 μg/ml)
and **P < 0.01 were accepted as statistically significant and ns indicated non-
and no changes at lower sub-MIC levels (0.25 μg/ml). Previous studies
significance.
have reported that the cell elongation process of P. aeruginosa occurs in
anaerobiosis and nitric oxide (NO) presence primarily to utilize the
docking strategy. Results from the molecular docking of aminoglyco- limited oxygen source for proceeding cell growth [61–63]. The fila-
sides with LasR, amikacin, gentamicin, kanamycin, netilmicin, strep- mentation and streptococcal development of the P. aeruginosa cell has
tomycin, and tobramycin revealed greater binding efficacy with the also been previously reported by Waisbren et al. [64] in the presence of
active site that is present in LasR. A relatively higher score than that of amikacin that is either treated singly or in combination with other
OdDHL was observed (Table 1). However, neomycin and paromomycin antibiotics. Thus, the present study suggests that the cell undergoes
exhibited slightly lower binding energy than OdDHL. The probable morphology changes to cope with the effect of the sub-MIC of ami-
reason for this binding efficiency difference involves the improved noglycosides; as such, due to morphology changes, biofilm inhibition is
hydrogen bonding and the presence of hydrophobic interactions of not dependent on concentration (Fig. 3). A recent report showed that
these compounds with residues that are present in the active site of the formation of the filamentous morphology of Vibrio cholerae cells
LasR. Residues of LasR, including Tyr-56, Trp-60, Tyr-64, Asp-73, Arg- enables chitin surface attachment and dense biofilm architecture for-
61, Ser-129, Cys-79, Gly-126, Thr-75, Val-76, Thr-115, Trp-88, Ala-105, mation [65]. Thus, the concentration-independent nature of biofilm
Leu-125, Leu-40, Leu-39, Asp-65, and Glu-48, have been shown to be inhibition by these aminoglycosides may relate to the high amount of
involved in forming hydrogen bonds with different aminoglycosides dye that binds to the elongated and filamentous cells, as compared to
(Table 1). the rod and streptococci forms of the cells. Overall, this indicates that

6
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 6. Inhibitory effects of aminoglycosides at sub-MIC levels on the production of virulence factors of P. aeruginosa PAO1. (A) Pyocyanin production, (B)
Pyoverdine production and (C) LasA protease activity. The production of each virulence factor in the presence of aminoglycosides are represented a relative value
with respect to the control. All the experiments were performed in triplicates. *P < 0.05 and **P < 0.01 were accepted as statistically significant and ns indicated
non-significance.

Fig. 7. Inhibition of swarming motility of P. aeruginosa PAO1 by aminoglycosides at sub-MIC levels reflected by diameter value (cm) of swarming motility on agar
plates. A representative image of each assay is presented and each experiment was carried out three times. **P < 0.01 was considered as statistically significant.

7
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 8. Inhibition of swimming motility of P. aeruginosa PAO1 by aminoglycosides at sub-MIC levels reflected by diameter value (cm) of swimming motility on agar
plates. A representative image of each assay is presented and each experiment was carried out three times. **P < 0.01 was considered as statistically significant and
ns indicated non-significance.

Table 1
Interaction of different aminoglycosides with the LasR QS receptor of P. aeruginosa.
Target (LasR)
Ligand
Minimum binding energy (Kcal/ Hydrogen-bond(s) with Hydrophobic Interaction(s) with
mol)

3-Oxo-C12-HSL (OdDHL) −9.91 Tyr-56, Trp-60, Tyr-64, Asp-73, Arg-61, Ser- Tyr-47, Val-76, Cys-79, Leu-125
129
Gentamicin −12.25 Thr-75, Trp-60, Cys-79, Gly-126 Tyr-47, Gly-38, Tyr-64, Leu-40, Ala-50, Val-76, Gly-38, Leu-125,
Leu-36,
Tobramycin −10.47 Ser-129, Thr-75, Trp-60, Arg-61, Val-76 Gly-38, Leu-36, Ala-127, Tyr-64, Ala-50, Ile-52
Neomycin −4.66 Tyr-56, Trp-60, Tyr-64, Thr-115, Trp-88, Val-76, Leu-40, Ala-50, Leu-125, Ala-127
Ala-105
Kanamycin −10.21 Ser-129, Thr-75, Thr-115, Leu-125, Trp-60 Tyr-64, Leu-36,
Netilmicin −12.11 Ser-129, Thr-115, Thr-75 Tyr-64, Tyr-56, Trp-88, Leu-36
Paromomycin −6.36 Tyr-56, Trp-60, Tyr-64, Thr-115, Trp-88, Val-76, Ala-127, Leu-40, Ala-50
Tyr-93
Amikacin −15.24 Trp-60, Thr-75, Thr-115, Leu-40, Leu-39 Tyr-64,Leu-36, Ile-52,
Streptomycin −9.41 Tyr-47, Trp-60, Asp-65, Glu-48, Thr-75 Trp-88, Val-76, Leu-36, Ala-127, Ala-50, Tyr-64, Ser-129, Ile-52

the morphological changes of the bacterial biofilm cells can allow un- present study, the modified pH of the culture environment may have
derstanding the multi-drug resistance of the P. aeruginosa biofilm. facilitated drug-eradicating activity. As P. aeruginosa is a nosocomial
In addition to the various levels of inhibiting biofilm formation, the pathogen that colonizes on several types of surfaces (e.g. medical de-
sub-MIC levels of all tested antibiotics exerted eradication action to- vices and food processing equipment) to cause infections, an antibiotic
wards the pre-established mature P. aeruginosa biofilm. In the present effectively exhibiting both biofilm inhibition and pre-existing mature
study, such action was most profound by gentamicin and neomycin. biofilm eradication against the bacteria is considered highly potential
The dispersal efficiency of the mature biofilm of P. aeruginosa by these [68–70].
aminoglycosides was found to be similar to those of streptomycin and For the present study, the sub-MIC levels of several aminoglycoside
tobramycin, as reported recently [44]. Furthermore, several previous drugs were evaluated for their actions against P. aeruginosa virulence
studies have reported several modifications of these antibiotics, such as factors, which included QS-regulated pyocyanin, pyoverdine, and LasA
featuring conjugation onto nanocarriers or combination with other protease enzymes. In P. aeruginosa, pyocyanin plays a crucial role in
drugs, have successfully eradicated the mature biofilm [66,67]. In the causing cystic fibrosis and lung infections [71–73]; whereas pyoverdine

8
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

Fig. 9. Aminoglycosides docked in different binding pockets of LasR of P. aeruginosa. (A) 2D interaction of amikacin, gentamicin, kanamycin, neomycin and
netilmicin with the LasR active site, (B) 3D interaction of amikacin, gentamicin, kanamycin, neomycin and netilmicin with the LasR active site, (C) 2D interaction of
paromomycin, streptomycin, tobramycin and N-3-(oxododecanoyl)-L-homoserine lactone (OdDHL) with the LasR active site and (D) 3D interaction of paromomycin,
streptomycin, tobramycin and N-3-(oxododecanoyl)-L-homoserine lactone (OdDHL) with the LasR active site.

is produced for bacterial metabolism in an iron-limited environment protease enzymes, previous studies demonstrate that LasR is also in-
[46,74,75]. Protease enzymes, such as protease A (LasA), elastase B volved in regulating the rhl system, biofilm formation, and the pro-
(LasB), and alkaline enzyme, are crucially utilized for bacterial colo- duction of other virulence factors (e.g. pyocyanin and rhamnolipid)
nization and host cell damage [76–78]. Overall, results have shown that [90,91]. Thus, in the present study, we have further performed in silico
the inhibitory action of sub-MIC levels of tested antibiotics was most docking analysis to evaluate molecular interactions between QS-asso-
effective against pyoverdine production, pyocyanin production, and ciated LasR proteins and different aminoglycosides. Docking results
LasA protease activity of P. aeruginosa. indicate that the aminoglycosides with inhibition against pyocyanin
Motility is also considered among the virulence properties of P. and LasA protease also exhibit high affinity binding to the active site of
aeruginosa [79]. The surface movements, such as swimming and LasR. The mode of this binding was found to involve hydrogen bonds
swarming by flagella, play a determining role in the bacterial initial and hydrophobic interactions, which is similar to several antibiofilm
attachment and colonization processes [79–81]. In the present study, agents that were previously reported (e.g. vitexin and 6-gingerol)
the treatments of various antibiotics were found to cause significantly [24,90]. Overall, it can be inferred that the antibiofilm activities of
different inhibitory effects on P. aeruginosa motility. For both the these antibiotics are derived from targeting the QS regulatory protein
swimming and swarming movements, the high sub-MIC levels of all LasR. Thus, based on the present findings, modifying the culture en-
tested antibiotics were more effective than the lower ones. vironment and concentration optimization results in the revitalization
It is well noted that biofilm establishment and virulence properties of antibiotics as an antibiofilm and antivirulence agent to treat biofilm-
are majorly regulated by the following two QS cell-to-cell commu- forming pathogenic bacteria.
nication systems in P. aeruginosa: the las and rhl systems [82–84]. The
former system (i.e. las system), via interactions between the transcrip- 5. Conclusion
tional regulator LasR and 3O–C12-HSL diffusible signal molecules that
are encoded by lasI cognate acyl homoserine lactone synthase, is re- In the present study we selected several aminoglycosides and per-
sponsible for the activation of lasA, lasB, and lasI, which encode for formed biofilm inhibition related assays using their sub-MIC levels and
various protease enzymes, such as LasA protease, LasB elastase, and alkaline (pH 7.9) culture environment. Each aminoglycoside was found
alkaline protease [85,86]. As aforementioned, these enzymes play an to exhibit different inhibition level towards P. aeruginosa biofilm for-
important role in P. aeruginosa pathogenesis [87–89]. In addition to mation, as well as different eradicating activity to the bacterial pre-

9
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

established mature biofilm. The sub-MICs of aminoglycosides also PloS One 12 (2017) e0176883.
showed anti-virulence activity against the bacterial motility, as well as [13] O. Orgad, Y. Oren, S.L. Walker, M. Herzberg, The role of alginate in Pseudomonas
aeruginosa EPS adherence, viscoelastic properties and cell attachment, Biofouling 27
the production of pyocyanin, pyoverdine and LasA protease enzymes, (2011) 787–798.
which is likely to derive from their binding with QS regulatory mole- [14] J. Munguia, V. Nizet, Pharmacological targeting of the host-pathogen interaction:
cule. For future studies, it is suggested that the effect of the tested an- alternatives to classical antibiotics to combat drug-resistant superbugs, Trends
Pharmacol. Sci. 38 (2017) 473–488.
tibiotics on other virulence factors should be exploited. In addition, [15] O. Fleitas Martinez, M.H. Cardoso, S.M. Ribeiro, O.L. Franco, Recent advances in
molecular studies on the drug binding with different QS associated anti-virulence therapeutic strategies with a focus on dismantling bacterial mem-
proteins are required to further confirm the mechanism of antibiofilm brane microdomains, toxin neutralization, quorum-sensing interference and biofilm
inhibition, Front Cell Infect Microbiol 9 (2019) 74.
activities of these aminoglycosides at sub-MIC level. [16] P.F. Vale, L. McNally, A. Doeschl-Wilson, K.C. King, R. Popat, M.R. Domingo-
Sananes, et al., Beyond killing: can we find new ways to manage infection? Evol
Compliance with ethical standards Med Public Health 2016 (2016) 148–157.
[17] S. Wagner, R. Sommer, S. Hinsberger, C. Lu, R.W. Hartmann, M. Empting, et al.,
Novel strategies for the treatment of Pseudomonas aeruginosa infections, J. Med.
This article does not contain any studies with human participants or Chem. 59 (2016) 5929–5969.
animals. [18] V.C. Scoffone, G. Trespidi, L.R. Chiarelli, G. Barbieri, S. Buroni, Quorum sensing as
antivirulence target in cystic fibrosis pathogens, Int. J. Mol. Sci. 20 (2019).
[19] M. Mohammadi, F. Masoumipour, M. Hassanshahian, T. Jafarinasab, Study the
Funding information antibacterial and antibiofilm activity of Carum copticum against antibiotic-resistant
bacteria in planktonic and biofilm forms, Microb. Pathog. 129 (2019) 99–105.
This work was supported by Basic Science Research Program [20] Z. Mohsenipour, M. Hassanshahian, Antibacterial activity of Euphorbia hebecarpa
alcoholic extracts against six human pathogenic bacteria in planktonic and biofilm
through the National Research Foundation of Korea funded by the forms, Jundishapur J. Microbiol. 9 (2016) e34701-e.
Ministry of Education (NRF-2019R1A2C1087156). [21] I. Sadeghian, M. Hassanshahian, S. Sadeghian, S. Jamali, Antimicrobial effects of
Quercus brantii fruits on bacterial pathogens, Jundishapur J. Microbiol. 5 (2012)
465–469.
Author statement [22] M. Artini, A. Patsilinakos, R. Papa, M. Božović, M. Sabatino, S. Garzoli, et al.,
Antimicrobial and antibiofilm activity and machine learning classification analysis
Fazlurrahman Khan: Conceptualization, Investigation, of essential oils from different mediterranean plants against Pseudomonas aerugi-
nosa, Molecules 23 (2018) 482.
Methodology, Validation, Writing and Editing. [23] R. Ragno, R. Papa, A. Patsilinakos, G. Vrenna, S. Garzoli, V. Tuccio, et al., Essential
Jang-Won Lee: Methodology. oils against bacterial isolates from cystic fibrosis patients by means of antimicrobial
Aqib Javaid: Methodology, Validation and Writing. and unsupervised machine learning approaches, Sci. Rep. 10 (2020) 2653.
[24] M.C. Das, P. Sandhu, P. Gupta, P. Rudrapaul, U.C. De, P. Tribedi, et al., Attenuation
Seul-Ki Park: Methodology.
of Pseudomonas aeruginosa biofilm formation by Vitexin: a combinatorial study with
Young-Mog Kim: Conceptualization, Funding acquisition, Editing azithromycin and gentamicin, Sci. Rep. 6 (2016) 23347.
and Supervision. [25] F. Khan, D.T.N. Pham, S.F. Oloketuyi, P. Manivasagan, J. Oh, Y.-M. Kim, Chitosan
and their derivatives: antibiofilm drugs against pathogenic bacteria, Colloids Surf. B
Biointerfaces 185 (2020) 110627.
Declaration of competing interest [26] F. Khan, S.F. Oloketuyi, Y.-M. Kim, Diversity of bacteria and bacterial products as
antibiofilm and antiquorum sensing drugs against pathogenic bacteria, Curr. Drug
Targets 20 (2019) 1156–1179.
The authors declare that they have no conflict of interest. [27] M. Mulat, A. Pandita, F. Khan, Medicinal plant compounds for combating the multi-
drug resistant pathogenic bacteria: a review, Curr. Pharmaceut. Biotechnol. 20
Acknowledgement (2019) 183–196.
[28] Y. Doi, J.I. Wachino, Y. Arakawa, Aminoglycoside resistance: the emergence of
acquired 16S ribosomal RNA methyltransferases, Infect. Dis. Clin. 30 (2016)
Author would like to thank Dung Thuy Nguyen Pham for her 523–537.
technical help during the experiment and language editing. [29] W. Khan, S.P. Bernier, S.L. Kuchma, J.H. Hammond, F. Hasan, G.A. O'Toole,
Aminoglycoside resistance of Pseudomonas aeruginosa biofilms modulated by ex-
tracellular polysaccharide, Int. Microbiol. 13 (2010) 207–212.
References [30] M. Wilton, L. Charron-Mazenod, R. Moore, S. Lewenza, Extracellular DNA acidifies
biofilms and induces aminoglycoside resistance in Pseudomonas aeruginosa,
Antimicrob. Agents Chemother. 60 (2016) 544–553.
[1] Z. Pang, R. Raudonis, B.R. Glick, T.J. Lin, Z. Cheng, Antibiotic resistance in
[31] M.S. Ramirez, M.E. Tolmasky, Aminoglycoside modifying enzymes, Drug Resist.
Pseudomonas aeruginosa: mechanisms and alternative therapeutic strategies,
Updates 13 (2010) 151–171.
Biotechnol. Adv. 37 (2019) 177–192.
[32] M.S. Ramirez, N. Nikolaidis, M.E. Tolmasky, Rise and dissemination of aminogly-
[2] S. Rajan, L. Saiman, Pulmonary infections in patients with cystic fibrosis, Semin.
coside resistance: the aac(6')-Ib paradigm, Front. Microbiol. 4 (2013) 121.
Respir. Infect. 17 (2002) 47–56.
[33] J.T. Jo, F.S. Brinkman, R.E. Hancock, Aminoglycoside efflux in Pseudomonas aeru-
[3] D. Davies, Understanding biofilm resistance to antibacterial agents, Nat. Rev. Drug
ginosa: involvement of novel outer membrane proteins, Antimicrob. Agents
Discov. 2 (2003) 114–122.
Chemother. 47 (2003) 1101–1111.
[4] C.A. Fux, J.W. Costerton, P.S. Stewart, P. Stoodley, Survival strategies of infectious
[34] K. Shi, S.J. Caldwell, D.H. Fong, A.M. Berghuis, Prospects for circumventing ami-
biofilms, Trends Microbiol. 13 (2005) 34–40.
noglycoside kinase mediated antibiotic resistance, Front Cell Infect Microbiol 3
[5] E. Maunders, M. Welch, Matrix exopolysaccharides; the sticky side of biofilm for-
(2013) 22.
mation, FEMS Microbiol. Lett. 364 (2017).
[35] L. Hidalgo, K.L. Hopkins, B. Gutierrez, C.M. Ovejero, S. Shukla, S. Douthwaite,
[6] L.C. Powell, M.F. Pritchard, E.L. Ferguson, K.A. Powell, S.U. Patel, P.D. Rye, et al.,
et al., Association of the novel aminoglycoside resistance determinant RmtF with
Targeted disruption of the extracellular polymeric network of Pseudomonas aerugi-
NDM carbapenemase in Enterobacteriaceae isolated in India and the UK, J.
nosa biofilms by alginate oligosaccharides, NPJ Biofilms Microbiomes 4 (2018) 13.
Antimicrob. Chemother. 68 (2013) 1543–1550.
[7] O. Ciofu, T. Tolker-Nielsen, Tolerance and resistance of Pseudomonas aeruginosa
[36] S. Garneau-Tsodikova, K.J. Labby, Mechanisms of resistance to aminoglycoside
biofilms to antimicrobial agents-how P. aeruginosa can escape antibiotics, Front.
antibiotics: overview and perspectives, Medchemcomm 7 (2016) 11–27.
Microbiol. 10 (2019) 913.
[37] Y. Morita, J. Tomida, Y. Kawamura, Responses of Pseudomonas aeruginosa to anti-
[8] L.R. Mulcahy, V.M. Isabella, K. Lewis, Pseudomonas aeruginosa biofilms in disease,
microbials, Front. Microbiol. 4 (2014) 422.
Microb. Ecol. 68 (2014) 1–12.
[38] K. Poole, Pseudomonas aeruginosa: resistance to the max, Front. Microbiol. 2
[9] K. Lee, S.S. Yoon, Pseudomonas aeruginosa biofilm, a programmed bacterial life for
(2011) 65.
fitness, J. Microbiol. Biotechnol. 27 (2017) 1053–1064.
[39] C. Ghosh, P. Sarkar, R. Issa, J. Haldar, Alternatives to conventional antibiotics in the
[10] L.A. Meirelles, D.K. Newman, Both toxic and beneficial effects of pyocyanin con-
era of antimicrobial resistance, Trends Microbiol. 27 (2019) 323–338.
tribute to the lifecycle of Pseudomonas aeruginosa, Mol. Microbiol. 110 (2018)
[40] A.R. Hauser, J. Mecsas, D.T. Moir, Beyond antibiotics: new therapeutic approaches
995–1010.
for bacterial infections, Clin. Infect. Dis. 63 (2016) 89–95.
[11] J. Poppe, J. Reichelt, W. Blankenfeldt, Pseudomonas aeruginosa pyoverdine ma-
[41] F. Khan, D.T.N. Pham, Y.-M. Kim, Alternative strategies for the application of
turation enzyme PvdP has a noncanonical domain architecture and affords insight
aminoglycoside antibiotics against the biofilm-forming human pathogenic bacteria,
into a new subclass of tyrosinases, J. Biol. Chem. 293 (2018) 14926–14936.
Appl. Microbiol. Biotechnol. 104 (2020) 1955–1976.
[12] J. Luo, B. Dong, K. Wang, S. Cai, T. Liu, X. Cheng, et al., Baicalin inhibits biofilm
[42] F. Khan, D.T. Nguyen Pham, S.F. Oloketuyi, Y.-M. Kim, Antibiotics and their dif-
formation, attenuates the quorum sensing-controlled virulence and enhances
ferent application strategies in controlling the biofilm forming pathogenic bacteria,
Pseudomonas aeruginosa clearance in a mouse peritoneal implant infection model,
Curr. Pharmaceut. Biotechnol. 21 (2019) 270–286.

10
F. Khan, et al. Microbial Pathogenesis 146 (2020) 104249

[43] D. Lebeaux, A. Chauhan, S. Letoffe, F. Fischer, H. de Reuse, C. Beloin, et al., pH- [66] H. Ren, J. Wu, A. Colletta, M.E. Meyerhoff, C. Xi, Efficient eradication of mature
Mediated potentiation of aminoglycosides kills bacterial persisters and eradicates in Pseudomonas aeruginosa biofilm via controlled delivery of nitric oxide combined
vivo biofilms, J. Infect. Dis. 210 (2014) 1357–1366. with antimicrobial peptide and antibiotics, Front. Microbiol. 7 (2016) 1260.
[44] F. Khan, J.-W. Lee, D.T.N. Pham, J.-H. Lee, H.-W. Kim, Y.-K. Kim, et al., [67] A. Zhang, H. Mu, W. Zhang, G. Cui, J. Zhu, J. Duan, Chitosan coupling makes
Streptomycin mediated biofilm inhibition and suppression of virulence properties in microbial biofilms susceptible to antibiotics, Sci. Rep. 3 (2013) 3364.
Pseudomonas aeruginosa PAO1, Appl. Microbiol. Biotechnol. 104 (2020) 799–816. [68] P. Gupta, A. Sarkar, P. Sandhu, A. Daware, M.C. Das, Y. Akhter, et al., Potentiation
[45] F. Khan, P. Manivasagan, D.T.N. Pham, J. Oh, S.K. Kim, Y.M. Kim, Antibiofilm and of antibiotic against Pseudomonas aeruginosa biofilm: a study with plumbagin and
antivirulence properties of chitosan-polypyrrole nanocomposites to Pseudomonas gentamicin, J. Appl. Microbiol. 123 (2017) 246–261.
aeruginosa, Microb. Pathog. 128 (2019) 363–373. [69] P. Gupta, S. Chhibber, K. Harjai, Subinhibitory concentration of ciprofloxacin tar-
[46] D.T.N. Pham, F. Khan, T.T.V. Phan, S.K. Park, P. Manivasagan, J. Oh, et al., Biofilm gets quorum sensing system of Pseudomonas aeruginosa causing inhibition of biofilm
inhibition, modulation of virulence and motility properties by FeOOH nanoparticle formation & reduction of virulence, Indian J. Med. Res. 143 (2016) 643–651.
in Pseudomonas aeruginosa, Braz. J. Microbiol. 50 (2019) 791–805. [70] H. Saini, S. Chhibber, K. Harjai, Azithromycin and ciprofloxacin: a possible sy-
[47] X. Zhu, D. Liu, A.K. Singh, R. Drolia, X. Bai, S. Tenguria, et al., Tunicamycin nergistic combination against Pseudomonas aeruginosa biofilm-associated urinary
mediated inhibition of wall teichoic acid affects Staphylococcus aureus and Listeria tract infections, Int. J. Antimicrob. Agents 45 (2015) 359–367.
monocytogenes cell morphology, biofilm formation and virulence, Front. Microbiol. [71] G.W. Lau, D.J. Hassett, H. Ran, F. Kong, The role of pyocyanin in Pseudomonas
9 (2018) 1352. aeruginosa infection, Trends Mol. Med. 10 (2004) 599–606.
[48] F. Khan, J.-W. Lee, P. Manivasagan, D.T.N. Pham, J. Oh, Y.-M. Kim, Synthesis and [72] B. Rada, T.L. Leto, Pyocyanin effects on respiratory epithelium: relevance in
characterization of chitosan oligosaccharide-capped gold nanoparticles as an ef- Pseudomonas aeruginosa airway infections, Trends Microbiol. 21 (2013) 73–81.
fective antibiofilm drug against the Pseudomonas aeruginosa PAO1, Microb. Pathog. [73] C.C. Caldwell, Y. Chen, H.S. Goetzmann, Y. Hao, M.T. Borchers, D.J. Hassett, et al.,
135 (2019) 103623. Pseudomonas aeruginosa exotoxin pyocyanin causes cystic fibrosis airway patho-
[49] R. Papa, L. Selan, E. Parrilli, M. Tilotta, F. Sannino, G. Feller, et al., Anti-biofilm genesis, Am. J. Pathol. 175 (2009) 2473–2488.
activities from marine cold adapted bacteria against staphylococci and Pseudomonas [74] I.J. Schalk, L. Guillon, Pyoverdine biosynthesis and secretion in Pseudomonas aer-
aeruginosa, Front. Microbiol. 6 (2015) 1333. uginosa: implications for metal homeostasis, Environ. Microbiol. 15 (2013)
[50] F. Khan, P. Manivasagan, J.W. Lee, D.T.N. Pham, J. Oh, Y.M. Kim, Fucoidan-sta- 1661–1673.
bilized gold nanoparticle-mediated biofilm inhibition, attenuation of virulence and [75] D.R. Kirienko, D. Kang, N.V. Kirienko, Novel pyoverdine inhibitors mitigate
motility properties in Pseudomonas aeruginosa PAO1, Mar. Drugs 17 (2019). Pseudomonas aeruginosa pathogenesis, Front. Microbiol. 9 (2018) 3317.
[51] J.H. Lee, Y.G. Kim, M.H. Cho, J.A. Kim, J. Lee, 7-Fluoroindole as an antivirulence [76] E. Kessler, M. Safrin, J.K. Gustin, D.E. Ohman, Elastase and the LasA protease of
compound against Pseudomonas aeruginosa, FEMS Microbiol. Lett. 329 (2012) Pseudomonas aeruginosa are secreted with their propeptides, J. Biol. Chem. 273
36–44. (1998) 30225–30231.
[52] D.W. Essar, L. Eberly, A. Hadero, I.P. Crawford, Identification and characterization [77] G. Doring, H.J. Obernesser, K. Botzenhart, B. Flehmig, N. Hoiby, A. Hofmann,
of genes for a second anthranilate synthase in Pseudomonas aeruginosa: inter- Proteases of Pseudomonas aeruginosa in patients with cystic fibrosis, J. Infect. Dis.
changeability of the two anthranilate synthases and evolutionary implications, J. 147 (1983) 744–750.
Bacteriol. 172 (1990) 884–900. [78] S.S. Twining, S.E. Kirschner, L.A. Mahnke, D.W. Frank, Effect of Pseudomonas aer-
[53] A. Stintzi, K. Evans, J.M. Meyer, K. Poole, Quorum-sensing and siderophore bio- uginosa elastase, alkaline protease, and exotoxin A on corneal proteinases and
synthesis in Pseudomonas aeruginosa: lasR/lasI mutants exhibit reduced pyoverdine proteins, Invest. Ophthalmol. Vis. Sci. 34 (1993) 2699–2712.
biosynthesis, FEMS Microbiol. Lett. 166 (1998) 341–345. [79] D. Drake, T.C. Montie, Flagella, motility and invasive virulence of Pseudomonas
[54] G.M. Morris, R. Huey, A.J. Olson, Using AutoDock for ligand-receptor docking, Curr aeruginosa, J. Gen. Microbiol. 134 (1988) 43–52.
Protoc Bioinformatics (2008) (Chapter 8):Unit 8 14. [80] J.W. Newman, R.V. Floyd, J.L. Fothergill, The contribution of Pseudomonas aeru-
[55] D.S. BIOVIA, Discovery Studio Modeling Environment, Dassault Systèmes, San ginosa virulence factors and host factors in the establishment of urinary tract in-
Diego, 2016 Release 2017 ed. fections, FEMS Microbiol. Lett. (2017) 364.
[56] M.U. Johansson, V. Zoete, O. Michielin, N. Guex, Defining and searching for [81] T.S. Murray, M. Ledizet, B.I. Kazmierczak, Swarming motility, secretion of type 3
structural motifs using DeepView/Swiss-PdbViewer, BMC Bioinf. 13 (2012) 173. effectors and biofilm formation phenotypes exhibited within a large cohort of
[57] P. Parasuraman, B. Devadatha, V.V. Sarma, S. Ranganathan, D.R. Ampasala, Pseudomonas aeruginosa clinical isolates, J. Med. Microbiol. 59 (2010) 511–520.
B. Siddhardha, Anti-quorum sensing and antibiofilm activities of Blastobotrys [82] J. Lee, L. Zhang, The hierarchy quorum sensing network in Pseudomonas aeruginosa,
parvus PPR3 against Pseudomonas aeruginosa PAO1, Microb. Pathog. 138 (2020) Protein Cell 6 (2015) 26–41.
103811. [83] I. Castillo-Juarez, T. Maeda, E.A. Mandujano-Tinoco, M. Tomas, B. Perez-Eretza,
[58] M. Kalia, P.K. Singh, V.K. Yadav, B.S. Yadav, D. Sharma, S.S. Narvi, et al., Structure S.J. Garcia-Contreras, et al., Role of quorum sensing in bacterial infections, World J
based virtual screening for identification of potential quorum sensing inhibitors Clin Cases 3 (2015) 575–598.
against LasR master regulator in Pseudomonas aeruginosa, Microb. Pathog. 107 [84] M. Kostylev, D.Y. Kim, N.E. Smalley, I. Salukhe, E.P. Greenberg, A.A. Dandekar,
(2017) 136–143. Evolution of the Pseudomonas aeruginosa quorum-sensing hierarchy, Proc. Natl.
[59] L. Lu, W. Hu, Z. Tian, D. Yuan, G. Yi, Y. Zhou, et al., Developing natural products as Acad. Sci. U. S. A. 116 (2019) 7027–7032.
potential anti-biofilm agents, Chin. Med. 14 (2019) 11. [85] K. Papenfort, B.L. Bassler, Quorum sensing signal-response systems in Gram-nega-
[60] M.J. Henry-Stanley, D.J. Hess, C.L. Wells, Aminoglycoside inhibition of tive bacteria, Nat. Rev. Microbiol. 14 (2016) 576–588.
Staphylococcus aureus biofilm formation is nutrient dependent, J. Med. Microbiol. [86] M. Traidej, M.E. Marquart, A.R. Caballero, B.A. Thibodeaux, R.J. O'Callaghan,
63 (2014) 861–869. Identification of the active site residues of Pseudomonas aeruginosa protease IV.
[61] L.R. Hoffman, A.R. Richardson, L.S. Houston, H.D. Kulasekara, W. Martens- Importance of enzyme activity in autoprocessing and activation, J. Biol. Chem. 278
Habbena, M. Klausen, et al., Nutrient availability as a mechanism for selection of (2003) 2549–2553.
antibiotic tolerant Pseudomonas aeruginosa within the CF airway, PLoS Pathog. 6 [87] S.J. Park, S.K. Kim, Y.I. So, H.Y. Park, X.H. Li, D.H. Yeom, et al., Protease IV, a
(2010) e1000712. quorum sensing-dependent protease of Pseudomonas aeruginosa modulates insect
[62] S.S. Yoon, R.F. Hennigan, G.M. Hilliard, U.A. Ochsner, K. Parvatiyar, M.C. Kamani, innate immunity, Mol. Microbiol. 94 (2014) 1298–1314.
et al., Pseudomonas aeruginosa anaerobic respiration in biofilms: relationships to [88] L.L. Blackwood, R.M. Stone, B.H. Iglewski, J.E. Pennington, Evaluation of
cystic fibrosis pathogenesis, Dev. Cell 3 (2002) 593–603. Pseudomonas aeruginosa exotoxin A and elastase as virulence factors in acute lung
[63] M.Y. Yoon, K.M. Lee, Y. Park, S.S. Yoon, Contribution of cell elongation to the infection, Infect. Immun. 39 (1983) 198–201.
biofilm formation of Pseudomonas aeruginosa during anaerobic respiration, PloS One [89] K. Vadakkan, A.A. Choudhury, R. Gunasekaran, J. Hemapriya, S. Vijayanand,
6 (2011) e16105. Quorum sensing intervened bacterial signaling: pursuit of its cognizance and re-
[64] S.J. Waisbren, D.J. Hurley, B.A. Waisbren, Morphological expressions of antibiotic pression, J Genet Eng Biotechnol 16 (2018) 239–252.
synergism against Pseudomonas aeruginosa as observed by scanning electron mi- [90] H.S. Kim, S.H. Lee, Y. Byun, H.D. Park, 6-Gingerol reduces Pseudomonas aeruginosa
croscopy, Antimicrob. Agents Chemother. 18 (1980) 969–975. biofilm formation and virulence via quorum sensing inhibition, Sci. Rep. 5 (2015)
[65] B.R. Wucher, T.M. Bartlett, M. Hoyos, K. Papenfort, A. Persat, C.D. Nadell, Vibrio 8656.
cholerae filamentation promotes chitin surface attachment at the expense of com- [91] V. Venturi, Regulation of quorum sensing in Pseudomonas, FEMS Microbiol. Rev. 30
petition in biofilms, Proc. Natl. Acad. Sci. U. S. A. 116 (2019) 14216–14221. (2006) 274–291.

11

You might also like