ArticlePDF Available

Ciprofloxacin- and levofloxacin-loaded nanoparticles efficiently suppressed fluoroquinolone resistance and biofilm formation in Acinetobacter baumannii

Authors:

Abstract and Figures

The spread of fluoroquinolone (FQ) resistance in Acinetobacter baumannii represents a critical health threat. This study aims to overcome FQ resistance in A. baumannii via the formulation of polymeric nanoFQs. Herein, 80 A. baumannii isolates were obtained from diverse clinical sources. All A. baumannii isolates showed high resistance to most of the investigated antimicrobials, including ciprofloxacin (CIP) and levofloxacin (LEV) (97.5%). FQ resistance-determining regions of the gyrA and parC genes were the most predominant resistant mechanism, harbored by 69 (86.3%) and 75 (93.8%) of the isolates, respectively. Additionally, plasmid-mediated quinolone resistance genes aac(6′)-Ib and qnrS were detected in 61 (76.3%) and 2 (2.5%) of the 80 isolates, respectively. The CIP- and LEV-loaded poly ε-caprolactone (PCL) nanoparticles, FCIP and FLEV, respectively, showed a 1.5–6- and 6–12-fold decrease in the MIC, respectively, against the tested isolates. Interestingly, the time kill assay demonstrated that MICs of FCIP and FLEV completely killed A. baumannii isolates after 5–6 h of treatment. Furthermore, FCIP and FLEV were found to be efficient in overcoming the FQ resistance mediated by the efflux pumps in A. baumannii isolates as revealed by decreasing the MIC four-fold lower than that of free CIP and LEV, respectively. Moreover, FCIP and FLEV at 1/2 and 1/4 MIC significantly decreased biofilm formation by 47–93% and 69–91%, respectively. These findings suggest that polymeric nanoparticles can restore the effectiveness of FQs and represent a paradigm shift in the fight against A. baumannii isolates.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports
Ciprooxacin‑
and levooxacin‑loaded
nanoparticles eciently
suppressed uoroquinolone
resistance and biolm formation
in Acinetobacter baumannii
Alaa M. Aboelenin
1, Mohammed El‑Mowafy
1, Noha M. Saleh
2, Mona I. Shaaban
1* &
Rasha Barwa
1*
The spread of uoroquinolone (FQ) resistance in Acinetobacter baumannii represents a critical
health threat. This study aims to overcome FQ resistance in A. baumannii via the formulation of
polymeric nanoFQs. Herein, 80 A. baumannii isolates were obtained from diverse clinical sources. All
A. baumannii isolates showed high resistance to most of the investigated antimicrobials, including
ciprooxacin (CIP) and levooxacin (LEV) (97.5%). FQ resistance‑determining regions of the gyrA
and parC genes were the most predominant resistant mechanism, harbored by 69 (86.3%) and 75
(93.8%) of the isolates, respectively. Additionally, plasmid‑mediated quinolone resistance genes
aac(6)-Ib and qnrS were detected in 61 (76.3%) and 2 (2.5%) of the 80 isolates, respectively. The
CIP‑ and LEV‑loaded poly ε‑caprolactone (PCL) nanoparticles, FCIP and FLEV, respectively, showed
a 1.5–6‑ and 6–12‑fold decrease in the MIC, respectively, against the tested isolates. Interestingly,
the time kill assay demonstrated that MICs of FCIP and FLEV completely killed A. baumannii isolates
after 5–6 h of treatment. Furthermore, FCIP and FLEV were found to be ecient in overcoming the FQ
resistance mediated by the eux pumps in A. baumannii isolates as revealed by decreasing the MIC
four‑fold lower than that of free CIP and LEV, respectively. Moreover, FCIP and FLEV at 1/2 and 1/4 MIC
signicantly decreased biolm formation by 47–93% and 69–91%, respectively. These ndings suggest
that polymeric nanoparticles can restore the eectiveness of FQs and represent a paradigm shift in the
ght against A. baumannii isolates.
Acinetobacter baumannii is an aerobic, non-motile Gram-negative coccobacillus that is considered one of
the most hazardous opportunistic pathogens. is organism can get resistance determinants as a result of its
genome plasticity, making the infections it causes dicult to treat1. e World Health Organization (WHO)
has recognized A. baumannii as one of the top three priority pathogens requiring urgent development of
new antimicrobials. Such dangerous pathogens have the potential to cause several diseases, including post-
neurosurgical meningitis, osteomyelitis, lung infections, urinary tract infections, and infections of traumatic
or surgical wounds2.
A. baumannii has developed extraordinary antimicrobial resistance mechanisms, including activated
multidrug eux pumps, increased outer membrane permeability, enzymatic modication of drugs, and target
gene mutation. e combined actions of those mechanisms have led to the development of multiple drug-
resistant (MDR) and extensively drug-resistant (XDR) strains of A. baumannii3. XDR is identied as resistance
to at least one agent in all but bacterial isolates remain susceptible to one or two categories4.
OPEN
1Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, PO Box 35516,
Mansoura, Egypt. 2Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, PO Box 35516,
Mansoura, Egypt. *email: mona_ibrahim@mans.edu.eg; rasha@mans.edu.eg
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
Fluoroquinolones (FQs), such as ciprooxacin (CIP) and levooxacin (LEV), have been widely utilized to
treat A. baumannii infections by inhibiting DNA gyrase and topoisomerase IV. FQ resistance is mostly caused
by chromosomal mutations in the uoroquinolone resistance-determining regions (FQRDRs) of the DNA
gyrase genes (gyrA and gyrB) and/or topoisomerase IV genes (parC and parE), which reduce DNA gyrase or
topoisomerases anity for FQs5. Additionally, mutations in the regulatory genes that manage the expression
of eux pumps and outer membrane proteins (OMPs) are considered an important cause of FQ resistance.
Plasmid-mediated quinolone resistance (PMQR) plays a crucial role in the acquisition of resistance to FQs.
e PMQR genes aid in the selection of mutants with higher levels of resistance rather than conferring FQ
resistance6. ere are dierent types of PMQR determinants; qnr shields FQ targets (bacterial DNA gyrase and
topoisomerase IV) from inhibition. qnr proteins have been identied, including qnrS, qnrA, qnrB, qnrD, qnrC,
qnrVC, and the recently discovered qnrE, with numerous genetic variants7. e second PMQR gene is aac (6)-
Ib. It is an aminoglycoside-modifying enzyme that transfers acetyl groups to some FQs, causing a decrease in
binding to the target site and the elimination of antibacterial eects8. e enhanced eux pumps produced by
plasmid genes for pumps qepA and oqxAB are another important mechanism of PMQR9.
Biolms oer an impenetrable barrier to antibacterial agents, providing the necessary conditions for bacterial
growth and colonization as well as the emergence of serious and health-threatening microbial infections.
Additionally, pathogenic bacteria form biolms that are encased in the exopolysaccharide matrix, and play a
signicant role in pathogenesis, and limit the eectiveness of available treatments10. As a result, antibacterial
therapy requires the search for ecient and biolm-preventing bactericidal drugs11. A smart delivery system has
the potential to improve the bactericidal eectiveness of existing antimicrobials and provide an eective solution
to combat the spread of resistant bacteria12. Developing new generations or derivatives of antimicrobials is a
very expensive investment process that takes a long time to distinguish in pharmaceutical production pipelines.
Nanosize carriers could provide the necessary chemical protection and the ecient delivery of antimicrobial
compounds12. is has drawn attention to their potential for preventing and eradicating biolm development,
and microbial resistance. Along with their ability to increase bacterial uptake, antimicrobial-loaded nanoparticles
have greater penetration power, which would help to prevent the emergence of MDR and XDR. Additionally,
they have greater invivo stability against biodegradation and require low therapeutic doses and less frequent
administration13.
Polymeric nanoparticles (NPs) are solid colloidal nano-based systems ranging in size from 10 to 1000nm.
Biodegradable polymers like alginate, chitosan, and polycaprolactone are commonly utilized for the preparation
of NPs. Poly ε-caprolactone (PCL) is a semicrystalline aliphatic polyester that degrades at a slower rate than
other biodegradable polymers. Such a property can be exploited to deliver antibiotics in a controlled manner
over time14. Unlike other polyesters, PCL degradation products do not elevate the acidity of the surrounding
environment with a minimum impact on homeostasis. PCL was chosen for the preparation of biodegradable NPs
in the current study due to its advantageous biocompatibility, biodegradability, and non-toxicity15.
e aim of this study is the molecular characterization of dierent mechanisms of FQ resistance in A.
baumannii clinical isolates. Additionally, CIP- and LEV-loaded polymeric nanoparticles were formulated using
PCL and further assessed their eectiveness in overcoming FQ resistance and biolm formation in A. baumannii
isolates.
Results
Identication of bacterial isolates
Microscopical characterization and biochemical reactions
In this study, a total of 550 specimens were collected, and 120 isolates were identied as Acinetobacter spp. using
standard microbiological techniques, including Gram staining, colony morphology, and biochemical reactions.
Under the microscope, all 120 isolates of Acinetobacter spp. were seen as Gram-negative coccobacilli. On solid
media, colonies were smooth, occasionally mucoid, and non-lactose fermenters appeared as pale or beige colonies
on MacConkey agar. Metallic reddish colonies were detected using CHROMagar Acinetobacter media aer
overnight incubation at 37°C. Furthermore, isolates of Acinetobacter spp. were positive for catalase and citrate
utilization but were negative for indole, oxidase, methyl red, Voges-Proskauer, and lactose fermentation.
Identication of A. baumannii by PCR
All the microbiologically identied isolates were conrmed to belong to the Acinetobacter genus, as revealed
by the detection of a 425 base pairs (bp) amplicon corresponding to the recA gene (Supplementary Fig.S1).
Furthermore, 80 isolates were identied as A. baumannii species, given the codes Ab1-80, via detection of
the characteristic 208bp fragment of the 16S–23S rRNA gene intergenic spacer (ITS) region in such species
(Supplementary Fig.S1).
e 80 isolates were recovered from blood (n = 28, 35%), sputum (n = 26, 32.5%), wounds (n = 22, 27.5%),
and urine (n = 4, 5%), as shown in Supplementary TableS1.
Determination of antimicrobial susceptibility
We tested the 80 A. baumannii isolates and 2 A. baumannii standard strains, ATCC 19606 and ATCC 17987,
for susceptibility to β-lactams, FQs, aminoglycosides, a sulfa drug, and tetracyclines using the Kirby–Bauer
disc diffusion technique on Mueller–Hinton agar media (Supplementary TableS2). High resistance was
detected in all isolates to most of the investigated antimicrobials (Fig.1). e highest resistance was observed to
ceazidime (100%), cefepime (99%), cefotaxime (99%), tazobactam/piperacillin (99%), sulbactam/ampicillin
(97.5%), imipenem (97.5%), ciprooxacin (97.5%), levooxacin (97.5%), amikacin (95%), gentamicin (92.5%),
and trimethoprim–sulfamethoxazole (83.8%). e lowest level of resistance was observed to doxycycline
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
(55%) and minocycline (48.8%) (Supplementary TableS2). e A. baumannii standard strain ATCC 19606
showed sensitivity to all investigated antimicrobials, excluding ceazidime, cefepime, cefotaxime, tazobactam/
piperacillin, sulbactam/ampicillin, and sulfamethoxazole/trimethoprim, while the standard strain ATCC 17987
showed sensitivity to all except sulfamethoxazole/trimethoprim (Supplementary TableS2).
e results demonstrated that 78 isolates (97.5%) were XDR and FQ-resistant isolates (Supplementary
TableS2). Eight representative XDR and FQ-resistant isolates (Ab29, Ab30, Ab36, Ab60, Ab65, Ab71, Ab72, and
Ab77) were selected to study the eect of the CIP- and LEV-loaded nanopreparations, FCIP, and FLEV, respectively,
on their susceptibility in comparison with free CIP and LEV antimicrobial agents.
Molecular characterization of FQ resistance mechanisms in A. baumannii isolates
FQRDRs and target site mutation
In order to characterize the FQ resistance of the 80 A. baumannii isolates and the 2 A. baumannii standard strains,
the FQRDRs in their genomes were further evaluated. e presence of mutations in gyrA and parC genes, major
FQRDRs, was detected by PCR followed by HinfI digestion, which resulted in successful digestion for the PCR
products with original sequences but not the ones with mutations (Supplementary TableS3). gyrA and parC genes
were detected in A. baumannii with amplicon sizes of 343bp and 327bp, respectively. A total of 69 (86.3%) and 75
(93.8%) of the 80 isolates harbored the gyrA and parC genes, respectively, as shown in Fig.2a. Non-digested PCR
Figure1. Resistance percentage of the 80 recovered A. baumannii isolates to every antimicrobial agent.
CAZ ceazidime, FEP cefepime, CTX cefotaxime, TZP tazobactam/piperacillin, SAM sulbactam/ampicillin,
IPM imipenem, CIP ciprooxacin, LEV levooxacin, AK amikacin, CN gentamicin, SXT sulfamethoxazole/
trimethoprim, DO doxycycline, MIN minocycline.
Figure2. Distribution of uoroquinolone (FQ) resistance genes among A. baumannii isolates; (a)
uoroquinolone resistance-determining regions (FQRDRs) and (b) plasmid-mediated quinolone resistance
genes (PMQRs).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
products were obtained aer HinfI digestion of gyrA and parC amplicons in all A. baumannii isolates, indicating
mutations in the FQRDR of both genes (Supplementary Fig.S2). In the two standard A. baumannii strains, ATCC
19606 and ATCC17987, obtaining two fragments aer HinfI digestion, at 291 and 52bp conrmed the absence of
mutation in gyrA, while at 206 and 121bp indicated the absence of mutation in parC (Supplementary TableS3).
DNA sequencing of the FQRDRs of gyrA and parC genes in A. baumannii (Ab30, Ab60, and Ab72) isolates
showed gyrA mutations at Ser83 to Leu and parC mutations at Ser80 to Leu. e obtained sequences were depos-
ited in the gene bank with accession numbers OR289925, OR289926, and OR289927.
PMQR genes and eux pump genes
e PMQR genes qnrA, qnrB, qnrS, qnrC, qnrD, and aac(6)-Ib, as well as eux pump-encoding genes oqxAB
and qepA were evaluated via PCR (Supplementary TableS3) using the relevant primers shown in Supplementary
TableS4. e aac(6)-Ib and qnrS genes were detected in A. baumannii with amplicon sizes of 480bp and 427bp,
respectively (Supplementary Fig.S3). Among 80 A. baumannii isolates, the aac(6)-Ib gene was the most detected
PMQR in 61 (76.3%) isolates (Fig.2b). Two A. baumannii isolates (2.5%), coded Ab60 and Ab65, harbored the
qnrS and aac(6)-Ib genes. Nevertheless, the other PMQR- and eux pump-encoding genes, qnrA, qnrB, qnrC,
qnrD, qepA, and oqxAB, were absent in all isolates, as shown in Fig.2b. Additionally, no PMQR genes or eux
pump-encoding genes were detected in the 2 standard A. baumannii strains.
Preparation and physicochemical evaluation of antimicrobial-loaded PCL nanoparticles
e particle size, polydispersity index (PDI), and zeta potential (ZP) are shown in Table1. e determination of
the above parameters was necessary to assess the capability of the preparation method for producing PCL NPs
successfully. e particle size of the prepared NPs was unimodal, ranging from 263.30 ± 2.76 to 271.10 ± 9.14nm
with a very narrow size distribution (PDI ≤ 0.05). e surface of the PCL nanoparticles (NPs) showed a negative
charge of approximately 9mV. e entrapment eciency (EE%), antibiotic loading (AL%), and yield (Y%) of
the NPs are listed in Table1. CIP was entrapped more eciently (54.11%) than LEV (28.14%). Consequently,
the values of AL% and Y% of FCIP (8.64% and 73.65%, respectively) were higher than their corresponding values
for FLEV (5.09% and 64.88%, respectively; Table1).
Solid-state characterization was performed using attenuated total reectance-Fourier transform infrared
spectroscopy (ATR-FTIR) and dierential scanning calorimetry (DSC). ATR-FTIR was conducted to evaluate
any possible chemical interaction between PCL and CIP or LEV. Moreover, the crystallinity or amorphousness
of the developed polymeric matrices of FCIP and FLEV was examined using DSC. e spectra and thermograms of
the investigated samples are shown in Fig.3a,b, respectively. Both CIP and LEV spectra presented characteristic
peaks at 1700 (C=O acid), 1620 (C=O carbonyl), 2850–2930 (aromatic H), and 1510–1530 cm−1 (piperazinyl
group). e PCL spectrum showed characteristic peaks at 1723 (C=O), 1239, 1165 (C–O–C), 2865, and 2944 cm−1
(C–H). e spectra of the binary physical mixtures, PCL/CIP and PCL/LEV illustrated the peaks of the polymer,
while the peaks of the antimicrobial appeared with diminished intensities or even vanished. e spectra of FCIP
and FLEV demonstrated the disappearance of the characteristic peaks of CIP and LEV, respectively.
ermograms of the investigated samples are shown in Fig.3b. e pure CIP was shown to be in crystalline
state, as evidenced by an endothermic peak at 150°C. Additionally, other endothermic peaks of the antimicrobi-
als appeared between 300–320°C. In the thermograms of PCL and binary mixtures (PCL/CIP and PCL/LEV),
an endothermic peak of PCL at approximately 57°C was observed. ermograms of the binary mixtures also
exhibited the melting peaks of the antimicrobial that appeared with reduced intensity. In contrast, the thermo-
grams of FCIP and FLEV demonstrated the amorphousness of their matrices, with no distinctive melting peaks.
e morphology of FCIP, FLE V, and plain NPs was studied using transmission electron microscopy (TEM) as
shown Fig.3c–e, respectively. e results conrmed the formation of perfectly spherical, discrete, and uniform
NPs with a smooth surface. Individual NPs of FCIP and FLEV had a dense core of entrapped antimicrobials embed-
ded in the polymeric matrix of PCL (core–shell architecture) which was not the case for plain NPs (monophasic
polymeric matrix) due to the absence of antimicrobials. Hence, the size of plain NPs was observed to be smaller
than that of the antimicrobial-loaded NPs. However, the actual size of the FCIP and FLEV NPs was consistent with
that measured by dynamic light scattering (DLS) (approximately 270nm).
Table 1. Physicochemical evaluation of CIP- and LEV-loaded nanoparticles. CIP ciprooxacin, LEV
levooxacin, FCIP CIP-loaded nanoparticles, FLEV LEV-loaded nanoparticles, plain NPs plain PCL nanoparticles,
PDI polydispersity index, ZP zeta potential, EE% entrapment eciency, AL% antibiotic loading, Y% yield.
Parameter
Formula
FCIP FLEV Plain NPs
Size (nm) 268.3 ± 4.15 271.1 ± 9.14 263.3 ± 2.76
PDI 0.03 ± 0.01 0.05 ± 0.02 0.03 ± 0.01
ZP (mV) − 8.88 ± 0.5 − 8.67 ± 0.41 − 8.67 ± 0.9
EE% 54.11 ± 2.66 28.14 ± 4.56
AL% 8.64 ± 0.42 5.09 ± 0.83
Y% 73.65 ± 12.68 64.88 ± 11.41 66.7 ± 12.94
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
In vitro release study and kinetic modeling
e invitro release experiment was conducted using the dialysis bag diusion method. is study was performed
to compare the release proles of FCIP and FLEV to the corresponding ones of free CIP and LEV, respectively. e
release behaviors of CIP and LEV are expressed as cumulative percentages released from FCIP and FLEV, as shown
in Fig.3f,g, respectively. Rapid release rates of 94.4 ± 2.5% and 97.86 ± 2% were exhibited by free CIP and LEV
within 6h, respectively. In contrast, the controlled release patterns of CIP and LEV from FCIP and FLEV extended
for 10days to reach 100 ± 0.3% and 87.09 ± 3.3%, respectively, without burst release (Fig.3f,g). e release proles
showed that 20.2% and 20.6% of the loaded-CIP and LEV, respectively, released aer 24h incubation and that
were even much lower than free ones.
e release data were tted to kinetic models. It was found that FCIP and FLEV followed non-Fickian anomalous
diusion with diusion exponent (n) values of 0.72 and 0.52, respectively. On the other hand, the release of the
free antimicrobials followed Case II transport with n values of 1.08 and 0.97, respectively.
Figure3. Solid-state characterization of CIP, LEV, PCL, a physical mixture of PCL and CIP, a physical
mixture of PCL and LEV, FCIP, and FLEV; ATR-FTIR (a) and DSC (b). Morphologies of FCIP (c), FLEV and (d)
Plain NPs (e) by TEM and invitro release of FCIP (f) and FLEV (g). CIP ciprooxacin, LEV levooxacin, PCL
poly ε-caprolactone, FCIP CIP-loaded nanoparticles, FLEV LEV-loaded nanoparticles, plain NPs plain poly
ε-caprolactone nanoparticles, DSC dierential scanning calorimetry, ATR-FTIR attenuated total reectance-
Fourier transform infrared spectroscopy, TEM transmission electron microscopy.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
In vitro antimicrobial activity of the prepared nanoantimicrobials FCIP and FLEV
e minimum inhibitory concentration (MIC) values were measured using the broth microdilution technique
for FCIP and FLEV compared to CIP and LEV, respectively, and displayed in Table2. Eight representative XDR- and
FQ-resistant A. baumannii isolates and 2 A. baumannii standard strains were selected. e MIC values of CIP
ranged from 32 to 128μg/ml, and the MIC values of LEV ranged from 8 to 16μg/ml. e MIC values of FCIP and
FLEV ranged from 10.7 to 21.5 and 0.72 to 1.3, respectively. However, the MIC values of CIP, LEV, FCIP, and FLEV
for A. baumannii standard strains, ATCC 19606 and ATCC 17987 were 0.5 and 0.25μg/ml, respectively. FCIP and
FLEV were more eective than CIP and LEV, due to the increased eciency of CIP and LEV to be delivered into
the bacterial cells (Table2). e MIC of FCIP reduced by 1.5- to 6-fold compared to that of free CIP, while the
MIC of FLEV decreased by 6- to 12-fold compared to that of free LEV. At the same time, the plain NPs revealed no
activity on bacterial growth, indicating that the antimicrobial eects were only obtained from the encapsulated
drug itself (Supplementary TableS5).
Eux pump suppression by FCIP and FLEV
e eux pump inhibitor carbonyl-cyanide-m-chlorophenylhydrazone (CCCP) was used on the 8 selected
representative A. baumannii isolates and 2 standard A. baumannii strains. Only 3 out of 8 A. baumannii
isolates, Ab30, Ab30, and Ab72, of which the eux mechanism was demonstrated to be one of the FQ-resistant
mechanisms were used to study the eect of FCIP and FLEV on eux activity (Supplementary TableS5). at
was conrmed by a four-fold or more reduction in MIC (MIC decrease factor (MDF value of more 4)) in the
case of CIP and LEV, while no change in MIC was observed regarding FCIP and FLEV for the same isolates in the
presence of CCCP (Table3). e plain NPs had no antimicrobial (Supplementary TableS5) when used alone
or in combination with free CIP or LEV with or without CCCP (Supplementary TableS5). e standard A.
baumannii strains, had no eux activity as no change in their MIC of CIP nor LEV with co-treatment with CCCP
(Supplementary TableS5). When we used CCCP with strains (Ab29, Ab36, Ab65, Ab71, and Ab77) which do not
exhibit an eux pump related FQ resistance, the addition of the CCCP did not aect the MIC either with free
Table 2. e MICs for FCIP and FLEV compared to CIP and LEV against uoroquinolone (FQ)-resistant A.
baumannii isolates and standard strains A. baumannii ATCC 19606 and 17978. MIC minimum inhibitory
concentration, CIP ciprooxacin, LEV levooxacin, FCIP CIP-loaded nanoparticles, FLEV LEV-loaded
nanoparticles. *e mean MIC value was reported aer experiments were carried out in duplicate.
Isolate code
MIC* (μg/ml)
Fold decrease in MIC
MIC* (μg/ml)
Fold decrease in MICCIP FCIP LEV FLEV
Ab29 32 10.7 3 8 0.72 11
Ab30 32 10.7 3 8 0.72 11
Ab36 32 21.5 1.5 8 0.72 11
Ab60 128 21.5 6 16 1.3 12
Ab65 64 21.5 3 16 1.3 12
Ab71 64 21.5 3 8 1.3 6
Ab72 64 21.5 3 8 1.3 6
Ab77 64 21.5 3 8 1.3 6
ATCC 19606 0.5 0.5 1 0.5 0.5 1
ATCC 17987 0.25 0.25 1 0.25 0.25 1
Table 3. Eect of CCCP on the MICs of FCIP, CIP, FLEV and LEV against three representative XDR isolates
of A. baumannii. MIC minimum inhibitory concentration, CIP ciprooxacin, LEV levooxacin, FCIP CIP-
loaded nanoparticles, FLEV LEV-loaded nanoparticles, MDF MIC decrease factor, CCCP carbonyl-cyanide-m-
chlorophenylhydrazone. *e mean MIC value was reported aer experiments were carried out in duplicate.
Isolate code
MIC* (μg/ml)
MDF
MIC (μg/ml)
MDFCIP CIP + CCCP FCIP FCIP + CCCP
Ab30 32 8 4 10.7 10.7 1
Ab60 128 16 8 21.5 21.5 1
Ab72 64 16 4 21.5 21.5 1
Isolate code
MIC* (μg/ml)
MDF
MIC (μg/ml)
MDFLEV LEV + CCCP FLEV FLEV + CCCP
Ab30 8 2 4 0.72 0.72 1
Ab60 16 4 4 0.17 0.17 1
Ab72 8 2 4 1.3 1.3 1
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
or encapsulated FQ (Supplementary TableS5). erefore, the increased killing activity of the PCL nanoparticle-
encapsulated CIP and LEV is associated with pump inhibition.
Time killing assay
Incubation of A. baumannii isolates Ab30, Ab60, and Ab72 with the MICs of FCIP or FLEV caused a signicant
drop (P < 0.05) in bacterial growth within 2–3h of incubation and complete bacterial killing aer 5–6h, as shown
in Fig.4. Free LEV reduced the viable bacterial count with 10% inhibition aer 24h incubation at the same
concentrations while free CIP showed no eect upon treatment. Treating A. baumannii Ab30, Ab60 and Ab72
isolates with the MIC of FCIP caused a signicant decrease (P < 0.05) in bacterial growth within 2h of incubation
and complete bacterial killing aer 6h in the Ab30 isolate (Fig.4a) and aer 5h in the Ab60 and Ab72 isolates,
Figure4. Time kill assay of treated A. baumannii isolates with the MIC of FCIP and the MIC of FLEV compared
to CIP and LEV over a period of 24h; A. baumannii isolate Ab30 treated with (a) FCIP and CIP and (b) FLEV
and LEV, A. baumannii isolate Ab60 treated with (c) FCIP and CIP and (d) FLEV and LEV and A. baumannii
isolate Ab72 treated with (e) FCIP and CIP and (f) FLEV and LEV. Experiments represent three replicates and
are expressed as the mean ± SD. Statistical signicance was assessed by the one-way ANOVA test: P < 0.05
was considered signicant (*P < 0.05). MIC minimum inhibitory concentration, CIP ciprooxacin, LEV
levooxacin, FCIP CIP-loaded nanoparticles, FLEV LEV-loaded nanoparticles, plain NPs plain poly ε-caprolactone
nanoparticles.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
as shown in Fig.4c,e, respectively. Similarly, the count of such A. baumannii isolates signicantly reduced aer
2h of incubation with FLEV (P < 0.05), complete bacterial killing aer 6h in the Ab30 and Ab60 isolates (Fig.4b,d)
and aer 5h in the Ab72 isolate, as shown in Fig.4f. e plain NPs had no killing activity as there was no eect
on the viable count of such isolates aer 24h (Fig.4).
Biolm elimination by FCIP and FLEV
e 80 clinical isolates of A. baumannii were classied as strong producers (62.5%, n = 50), moderate producers
(21.3%, n = 17) and weak producers (13.8%, n = 11) based on their ability to form biolms. Only 2.5% (n = 2) of
these isolates failed to form biolms (Supplementary TableS6). e 2 standard A. baumannii strains, ATCC 19606
and ATCC17987, were strong biolm producers (Supplementary TableS6). e results of biolm adherence of 80
A. baumannii isolates are summarized in Supplementary TableS6. Among the 78 FQ-resistant isolates (97.5%), 50
(64.1%) were strong, 16 (20.5%) were moderate, 10 (12.8%) were weak and 2 (2.6%) were non-biolm forming,
as shown in Fig.5a.
Subinhibitory concentrations of FCIP and FLEV (1/2 and 1/4 MIC) led to a signicant dose-dependent decrease
in biolm formation in the selected strong biolm-forming isolates (Ab30, Ab60, and Ab72) and standard A. bau-
mannii strain ATCC19606 compared to cultures treated with CIP and LEV or treated with plain NPs (Fig.5b,c,
respectively). FCIP was associated with 60–93% and 47–80% inhibition in biolm formation using 1/2 and 1/4
MIC, respectively, in the investigated isolates. Additionally, 1/2 and 1/4 MIC of FLEV caused 83–91% and 69–89%
inhibition, respectively, of biolm formation in such isolates. e greatest reduction (93%) in biolm formation
was achieved using 1/2 MIC FCIP on the Ab72 isolate (Fig.5b). On the other hand, CIP and LEV were found to
be associated with 2–10% and 3–11% inhibition, respectively, when used at their subinhibitory concentrations
(1/2 and 1/4 MIC), which indicates that there is no eect of either 1/2 or 1/4 MIC of CIP or LEV on biolm
formation (Fig.5b,c).
Discussion
A. baumannii is one of the deadliest and most contagious Gram-negative bacteria that has an improved capac-
ity to escape human immune responses and resist several types of antimicrobials, resulting in potentially fatal
pneumonia and bacteremia1. FQs have been found to be eective against A. baumannii isolates over the past
40years, although resistance to these antimicrobial agents has emerged quickly2. ere have been several reports
of the spread of FQ resistance among A. baumannii isolates worldwide16,17.
In the present study, 80 isolates of A. baumannii were obtained from diverse clinical sources. Among the
eighty conrmed A. baumannii isolates, resistance to the investigated antimicrobial agents was highly prevalent
(Fig.1). Most of the isolates were XDR (97.5%), while 97.5% of the isolates were resistant to both LEV and CIP.
On the other hand, 37 out of 80 isolates were sensitive to minocycline, with the highest frequency (46.3%).
A single point mutation in DNA gyrase is required for A. baumannii to be resistant to FQs; nevertheless,
simultaneous mutations in the FQRDR areas of the gyrA and parC genes are anticipated to dramatically increase
the level of FQ resistance. Many reports have shown that FQ resistance in A. baumannii is related to spontane-
ous mutations in the FQRDRs of the gyrA and parC genes18,19. e A. baumannii isolates were analyzed for their
FQRDRs using PCR, followed by HinfI digestion. Out of the 80 isolates in this study, 69 (86.3%) and 75 (93.8%)
carried the mutations in the gyrA (343bp) and parC (327bp) genes, respectively (Fig.2a). In A. baumannii
(Ab30, Ab60, and Ab72) isolates, DNA sequencing of the FQRDRs of the gyrA and parC genes revealed gyrA
mutations at Ser83 to Leu and parC mutations at Ser80 to Leu. All CIP- and LEV-resistant isolates were found
to harbor simultaneous mutations in the FQRDRs of both the gyrA and parC genes. It is suggested that these
two mutations are associated with CIP- and LEV-resistance, as previously mentioned in several reports2022.
PMQR plays a signicant role in the development of resistance to FQ and may be a factor in the rise in spon-
taneous FQRDR mutations6. e three most well-known mechanisms of resistance to FQs related to PMQR
involve protecting the binding site in DNA-gyrase (qnr gene)7, altering the drug enzymatically (aac(6)-Ib gene)8,
and expelling the agent by eux pumps (oqxAB and qepA genes)9. Numerous investigations have revealed that
FQ-resistant isolates of A. baumannii lack qnrA, qnrB, and qnrS genes23,24. Regarding PMQR genes in this study,
the aac(6)-Ib gene (76.3%) was prevalent among the A. baumannii isolates, and two isolates (2.5%) harbored
the qnrS and aac(6)-Ib genes. However, such isolates lacked other PMQR genes, qnrA, qnrB, qnrC, qnrD, qepA
and oqxAB, as shown in Fig.2b.
FQ-resistant A. baumannii causes serious public and nosocomial infections16. erefore, this study aims
to restore the antimicrobial properties of FQs to alter this problem, and polymeric nanoparticles would be the
methodology to achieve that. e double emulsication process is the method of choice for encapsulating hydro-
philic drugs inside NPs. Although the EE% of the hydrophilic water-soluble drugs is usually low, adjusting the
pH values of the internal and external aqueous phases could be applied to enhance the EE%. As FQ contains one
carboxylic group and three basic nitrogen sites, its solubility is pH-dependent. At pH 3 of the internal aqueous
phase (W1), the soluble cationic species of CIP and LEV predominated. On the other hand, neutral pH stimu-
lates the formation of the zwitterionic least soluble species in the external aqueous phase (W2, pH 7.5). Such
a preferential solubility pattern could secure ecient loading of the antimicrobials and prevent their diusion
out to the external aqueous phase during the formation of NPs. e higher EE% of FCIP could be attributed to
the lower solubility of CIP than LEV at pH 7.5 (W2). Hence, more LEV molecules might prefer their existence
in the external aqueous phase rather than entrapping inside the core of the FLEV25. e negative surface charge
of the NPs can be attributed to the ionization of the surface free carboxylic groups of PCL (matrix polymer) in
their aqueous dispersion26.
e characteristic peaks of CIP, LEV, and PCL in their individual ATR-FTIR spectra in Fig.3a agree with
those reported by other investigators27,28. Nevertheless, the diminished intensities of the antimicrobial peaks in
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
the binary physical mixture spectra could be related to the dilution of CIP and LEV with PCL. In the ATR-FTIR
spectra of FCIP and FLEV, the absence of antimicrobial characteristic peaks indicated their entrapment within
the cores of the NPs. However, the bands that appeared in the range of 2800–3500 cm−1 were attributed to the
characteristic absorption peaks of the hydroxyl groups in polyvinyl alcohol (PVA) (the NP stabilizer).
In the DSC thermogram, the endothermic peaks of the antimicrobials that appeared between 300 and 320°C
in Fig.3b could be ascribed to the melting of CIP and LEV29. e reduction in the intensities of the melting peaks
of the antimicrobials exhibited in the thermograms of the binary mixtures could be attributed to the dilution
action of PCL. e absence of melting peaks in the thermograms of FCIP and FLEV indicated the amorphousness
of the polymeric matrices of the prepared NPs. e ndings of the solid-state characterization were consistent
Figure5. Biolm formation/inhibition assay; (a) Distribution of FQ-resistant A. baumannii isolates and
standard strains A. baumannii ATCC 19606 and 17978 according to biolm formation. (b,c) e impact
of subinhibitory concentrations (1/2 and 1/4 MIC) of FCIP and FLEV compared to CIP and LEV on biolm
formation of three representative isolates of A. baumannii that were both XDR and strong biolm producers
and standard A. baumannii strain ATCC19606: (b) e eect of FCIP and CIP. (c) e eect of FLEV and LEV.
Control: Culture treated with plain NPs. Experiments represent four replicates and are expressed as the
mean ± SD. Statistical signicance was assessed by the one-way ANOVA test: P < 0.01 was considered signicant
(**P < 0.01). MIC minimum inhibitory concentration, CIP ciprooxacin, LEV levooxacin, FCIP CIP-loaded
nanoparticles, FLEV LEV-loaded nanoparticles, plain NPs plain poly ε-caprolactone nanoparticles.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
with each other. e spherical core–shell morphology of FCIP and FLEV could secure the stealth of antimicrobials.
Moreover, the spherical geometry of the NPs could increase bacterial uptake by inuencing the contact area with
the cell membrane receptors more than rod-shaped particles30.
For invitro release, the sustained performance of FCIP and FLEV in Fig.3f,g, respectively, might be attributed
to the eective entrapment of antimicrobials within their cores. is conclusion agreed with the morphological
results, where the compact polymeric coat around the antimicrobial-loaded core likely prolonged antimicrobial
release. If the antimicrobial was not eciently entrapped, an unfavorable initial release (burst release) would
have occurred, which was the case with free CIP and LEV. Furthermore, the slow dissolution of the hydrophobic
PCL shell of FCIP and FLEV via the hydrolysis of ester bonds followed by pore creation would further prolong
the release of the entrapped antimicrobial, which extended for 10days in our study. is nding could oer the
method for the production of antimicrobial with long-lasting activities31.
e release data tted to the kinetic modeling revealed that the release of FCIP and FLEV was ruled mainly by
the diusion mechanism, which ensured a sustained release of the antimicrobials, as observed in their release
patterns. Alternatively, the release of free antimicrobials followed Case II transport (zero-order kinetics), which
means that the dissolution rather than diusion was the release limiting step32. e latter nding could be
attributed to formation of zwitterionic least soluble species of CIP and LEV at pH 7.4 of the release medium33.
ese ndings indicate that not only the release pattern but also the release kinetics could be modied by the
incorporation of CIP and LEV in PCL-based NPs; FCIP and FLEV, respectively.
Numerous studies have indicated improved antibacterial drug activity when entrapped in polymeric
nanoparticles31,34. ese results can be attributed to a variety of variables, such as improved drug delivery to the
site of action, increased drug stability when encapsulated into nanoparticles, and easier drug penetration into
bacterial cells35. In this study, the potency of CIP and LEV loaded into PCL nanoparticles against FQ-resistant
A. baumannii strains was increased compared to that of free CIP and LEV. Similarly, a CIP polymer-lipid hybrid
nanoformulation with greater antibacterial activity against a clinical E. coli isolate has been detected36.
Indeed, the features of FQs with low molecular weight and zwitterion composition are primarily responsible
for their ability to pass through the membrane of Gram-negative bacteria37. Additionally, the bacteria have
developed resistance to CIP and LEV due to chromosomal mutations that alter the target enzymes. FQ resist-
ance may occur by increasing eux or decreasing uptake, leading to reduced drug accumulation. Moreover,
plasmid-acquired resistance genes can produce proteins protecting bacterial molecules, antimicrobial metaboliz-
ing enzymes, or drug eux pumps5,6,9. e MICs of FCIP and FLEV against the investigated A. baumannii isolates
decreased by 1.5–6- and 6–12-fold, respectively, by encapsulating the drugs into nanoparticles (Table2). Such
a decrease in the MIC levels in our study may be due to enhanced drug penetration by nanoparticles into the
bacterial cell, which prevented bacterial development38. is behavior can be explained by the fact that NPs
can act as carriers for antimicrobials, eectively concealing them, enhancing their penetration of bacterial cell
walls, and helping to overcome resistance mechanisms of XDR bacteria39. erefore, the increased membrane
permeability obtained by nanosize-encapsulated CIP and LEV may account for the observed improvement in
the antibacterial action of FCIP and FLEV. Additionally, the prepared particle size aects membrane permeability40;
therefore, FCIP (268.3 ± 4.15nm) and FLEV (271.1 ± 9.14nm) nanoparticles were able to penetrate bacterial cells.
e nanosize and charge of the CIP and LEV formulations, as well as the bacterial hydrophobic anity for the
PCL polymer, which aids in rapid permeation across the bacterial outer membrane, may be the cause of the
instantaneous microbial killing induced by FCIP and FLEV41.
FQ resistance in A. baumannii isolates may be inuenced by eux-based systems42. CCCP increases the sensi-
tivity of several MDR bacteria, including A. baumannii, to various antimicrobials by inhibiting eux pumps43,44.
A four-fold or greater decline in MIC when CCCP was added to CIP and LEV served as evidence of its signi-
cance in increasing CIP and LEV resistance in some isolates45. e MICs of FCIP and FLEV nanopreparations were
not changed for the same isolates by co-treated CCCP. We thought that the reason for such a result is that the
nanoprepared antimicrobials totally inhibited the eux pump activity, so that the CCCP had no longer eect
on the MIC value when added to the culture (Table3). e eux-resistant mechanism was defeated by FCIP and
FLEV, as evidenced by the fact that their MIC was four times lower than that of free CIP and LEV, respectively,
on A. baumannii isolates with eux pump activity. NPs can bypass eux pumps by acting as a Trojan horse,
delivering antimicrobials, or by interacting with eux pumps to create irreversible blockage31,46. is notion is
strongly supported by the result that MIC for XDR strains without drug eux pumps was not aected by co-
treatment of CCCP with LEV and CIP, either, free or encapsulated into NPs. Similarly, zinc oxide nanoparticles
were demonstrated to have a unique eux pump inhibitory action on S. aureus eux pumps47. Additionally,
azithromycin poly lactic-co-glycolic acid nanoparticles (AZI-PLGA NPs) found to eectively counter the eux-
resistant mechanism exhibited by AZI-resistant bacteria. is was evidenced by a fourfold decrease inthe MIC
of NPs compared to free AZI48.
e rate of bacterial killing aer antimicrobial treatment is critical for preventing the emergence of antimi-
crobial resistance34. e killing activity of CIP and LEV was tremendously enhanced by PCL coating, as revealed
by the complete killing of the bacterial cells aer 5–6h of treatment with FCIP and FLEV compared to CIP and
LEV, respectively (Fig.4). On the other hand, aer 24h of incubation at the same concentrations, LEV slightly
(10%) decreased the viable bacterial count.
Biolm is a dispersed microbial growth that is challenging to penetrate and becomes resistant to conventional
treatment10. A variety of tactics have been studied to improve antibiolm activity, particularly in relation to
biolms that develop on medical devices that have been implanted. In the present study, 50 of 78 FQ-resistant
A. baumannii clinical isolates were strong biolm producers (Fig.5a). Subinhibitory concentrations of FCIP
and FLEV nanopreparations (1/2 and 1/4 MIC) signicantly reduced biolm formation by 47–93% and 69–91%
in strong biolm-forming isolates, whereas CIP and LEV, at their subinhibitory concentrations, aect biolm
formation by 10–17% (Fig.5b,c). Antimicrobial agents’ penetration and eectiveness are improved by the
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
formulation of nanotherapy, which enhances the solubility and minimises the agglomeration of antimicrobials.
Nanoformulations of CIP and LEV were found to reduce their particle size and increase their antimicrobial-loaded
penetration. Moreover, the hydrophobic properties of PCL chains, which speed up the antimicrobials penetration
and cause the bacterial cell wall to burst, inhibit the growth of biolms and stop microbial colonization49. At the
same instance, the eect of metallic nanoparticles on A. baumannii biolms was demonstrated, as they inhibited
the biolm of A. baumannii by 88%50. Curcumin NPs, aluminium oxide NPs, silver NPs and other nanoparticles
were also found to suppress the growth of A. baumannii biolms5153.
In conclusion, this is the rst report to study the inuence of CIP- and LEV-loaded nanoparticles on FQ
resistance and biolm inhibition ofclinical A. baumannii isolates in Egypt. e CIP- and LEV-loaded nano-
particles were found to be highly eective in killing A. baumannii and inhibiting biolm formation. Moreover,
it was demonstrated that encapsulation of CIP or LEV within NPs was a promising strategy to overcoming
eux-resistant mechanism towards FQs and improve their antibacterial eect. In future work, FQ-loaded PCL
nanoparticles will be investigated for ecacy invivo using adequate animal models.
Methods
Materials
Poly ε-caprolactone (PCL, Mn 80,000kDa), polyvinyl alcohol (PVA, Mw 14kDa), and methylene chloride (Mw
84.93) were purchased from Aldrich-Sigma Chemical Company, USA. Pharmaceutical grades of ciprooxacin
HCl (CIP) and levooxacin HCl (LEV) were kindly presented from EPICO and AMOUN pharmaceutical com-
panies in Egypt, respectively. In the nanopreparations procedures, deionized water (Millipore®, 18.2Mcm) was
used as the source of water.
Bacterial isolates
A. baumannii isolates from clinical specimens were obtained from the Central Microbiology Laboratory of
Mansoura University Hospital (MUH), Egypt, between December 2018 and November 2019. ese isolates
were obtained from diverse clinical specimens from patients in the intensive care unit (ICU), including wounds,
sputum, urine, and blood, according to hospital records. is work complies with the ethical guidelines of the
Research Ethics Committee in the Faculty of Pharmacy, Mansoura University, Egypt (Permit Number: 2022-193).
A. baumannii isolates were puried from the obtained specimens according to standard microbiological
culture techniques. All specimens were streaked on the chromogenic culture media Acinetobacter (CHRO-
Magar Acinetobacter Media, Paris, France) and incubated at 37°C for 48h. Isolates were also identied as the
Acinetobacter genus according to standard microbiological techniques, including colony morphology, Gram
stain, and biochemical reactions54. e standard strains, A. baumannii ATCC 19606 and 17978, were used as
positive controls.
Rapid extraction of genomic DNA, PCR conditions, and purication of PCR products
e genomic DNA of A. baumannii isolates was prepared by suspending fresh colonies in 100µl of distilled water,
followed by heating at 95°C for 10min. e bacterial suspension was centrifuged at 12,000rpm for 5min, and
the clear supernatant was transferred to a new tube and stored at − 20°C.
Unless otherwise specied, Dream Taq polymerase (Fermentas) was used for all routine PCRs. A reaction
mixture (25μl) containing 0.5μl of each primer (10μM), 12.5μl Dream Taq Green PCR Master Mix (2 ×), 1μl
of extracted DNA, and 9.5μl nuclease-free water was prepared. All PCRs were carried out under the following
conditions: primary denaturation at 95°C for 5min, followed by 35 cycles of denaturation at 95°C for 30s,
annealing for 30s at the temperature specic for each primer pair (Supplementary TableS4), and extension at
72°C for 1min, followed by one cycle of nal extension at 72°C for 10min. e PCR products were analyzed
by agarose gel electrophoresis (1.5% w/v agarose gel) and visualized by a UV transilluminator aer ethidium
bromide staining.
In the case of experiments that required purication of PCR products, digestion of PCR products, and
sequencing, a PCR purication kit (ermo, USA, Catalog number: K0701) was utilized according to the manu-
facturer’s directions.
Molecular identication
e isolates were conrmed as A. baumannii using a one-tube multiplex PCR method of the recA gene (charac-
teristic of the Acinetobacter genus) and ITS region (specic for A. baumannii spp.) by the primers listed in Sup-
plementary TableS455. e target amplicons of the recA gene and ITS region were 425 and 208bp, respectively.
e standard strains, A. baumannii ATCC 19606 and 17978, were used as positive controls.
Detection of antimicrobial susceptibility
e antimicrobial susceptibility prole was determined using the Kirby–Bauer disc diusion technique on Muel-
ler–Hinton agar media. e following antimicrobial discs (Oxoid, UK) were used to dene resistance proles
among A. baumannii clinical isolates: ceazidime (CAZ, 30µg), cefepime (FEP, 30µg), cefotaxime (CTX, 30µg),
piperacillin-tazobactam (TZP, 100μg/10µg), ampicillin-sulbactam (SAM, 10μg/10μg), imipenem (IPM, 10μg),
ciprooxacin (CIP, 5μg), levooxacin (LEV, 5μg), amikacin (AK, 30µg), gentamicin (CN, 10μg), trimetho-
prim–sulfamethoxazole (SXT, 1.25μg/23.75μg), doxycycline (DO, 30µg) and minocycline (MIN, 30µg). e
inhibition zone diameter was determined and interpreted consistent with the recommendations of the Clinical
and Laboratory Standards Institute guidelines56.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
Molecular characterization of FQ resistance mechanisms in A. baumannii isolates
e FQRDRs of gyrA and parC, besides the PMQR genes qnrA, qnrB, qnrS, qnrC, qnrD, and aac(6)-Ib and the
eux pump-encoding genes oqxAB and qepA, were amplied via conventional PCR using the relevant primers
shown in Supplementary TableS4.
e PCR of the FQRDRs of gyrA and parC was performed using the specic primers listed in Supplementary
TableS4. e purication of amplicons was carried out using a PCR purication kit (ermo, USA, catalog
number: K0701). e puried amplicons were digested with the HinfI digestion enzyme (ermo Scientic,
USA) according to the manufacturer’s protocol. e digested PCR products were then separated by agarose gel
electrophoresis (1.5% w/v agarose gel). e separated fragments were analyzed for mutations in the FQRDRs20,21.
Regarding gyrA, a single undigested PCR fragment (343bp) demonstrates the presence of a mutation at Ser83,
whereas two fragments, indicating digestion, at 291 and 52bp conrm the absence of mutation. In the case of
parC, a single undigested PCR band (327bp) conrms the presence of a mutation at Ser80, while digestion
generating two fragments at 206 and 121bp indicates the absence of a mutation.
ree representative XDR isolates, Ab30, Ab60, and Ab72, were selected for sequencing of the FQRDRs in
both the gyrA and parC genes. e target sequences in the gyrA and parC genes were amplied using Phusion
High-Fidelity DNA Polymerase (ermo Scientic, USA) and the specied primers in Supplementary TableS4.
PCRs were done according to the manufacturer’s instructions. e puried PCR products were sent to Sigma
Scientic Service Technical Support Company in Cairo, Egypt for sequencing by an Applied Biosystems 3500 XL
Genetic Analyzer and PCR primers specic for each gene. FinchTV program was used to analyze and visualize
chromatograms.
Preparation of NPs
e double emulsion/solvent evaporation method was used to prepare NPs. e ingredients of the prepared
NPs are shown in Supplementary TableS7. CIP or LEV was dissolved in sterile deionized water to form W1 at
a concentration of 2% w/v. PCL was dissolved in methylene chloride at a concentration of 15mg/ml to serve
as the organic phase (O). e pH of W1 was adjusted to 3, and then 1ml of it was emulsied in 10ml of O. A
primary emulsion (W1/O) was formed via ultrasonication for 1min at 100% amplitude in pulse mode (2s on
and 1s o) (Sonics Vibra Cell, Sonic & Materials, INC, USA) in an ice bath. W1/O was added to 200ml of a pH-
adjusted aqueous solution of 0.5% w/v PVA (W2, pH 7.5). is mixture was rapidly sonicated for 3min under
the above conditions to make a double emulsion (W1/O/W2). Aer solvent evaporation, the NPs were isolated
by centrifugation (Benchtop Centrifuge, Sigma Laborzentrifugen, Germany) at 10,000rpm for 1h, washed,
and centrifuged at the same speed for 30min to remove a clear supernatant. e NP pellets were dispersed in
deionized water then freeze-dried at − 80°C using a Freeze Dryer (SIM FD8-8T, SIM International, USA). e
lyophilized NPs were collected and refrigerated at 4°C for further evaluation. Plain NPs were prepared by the
same procedure as that for drug-free W1 at pH 3.
Physicochemical evaluation of the prepared NPs
A particle size analyzer (Malvern Instruments Ltd., England) equipped with DLS was used to determine the size
and surface charge, or ZP, of the NPs. e EE% of NPs was determined using an indirect method. Briey, the
concentration of unentrapped antimicrobials in the supernatants recovered aer centrifugation was measured
and subtracted from that of the total antimicrobials. Spectrophotometric measurements of CIP and LEV were
conducted using the supernatant of the plain NP as a blank at 275 and 278nm, respectively (UV–VIS Spectro
double beam, Labomed Inc., USA). e weight of lyophilized NPs was determined to calculate the AL% and Y%
of NPs. EE%, AL%, and Y% were calculated using the following equations:
Spectral analysis of PCL, pure CIP, pure LEV, and binary physical mixtures of each antimicrobial with PCL,
FCIP, and FLEV was conducted using ATR-FTIR (ermo Fisher Scientic, Inc., Waltham, MA, USA). DSC was
used to assess the crystallinity of the abovementioned samples (DSC, Pyris 6 DSC, Perkin Elmer, USA). e
morphology of the NPs was determined by TEM (TEM, JEOL 1010; JEOL Ltd, Tokyo, Japan). Briey, a 200-mesh
copper grid coated with carbon was placed with an aqueous drop of the NP dispersion, and any surplus liquid
was absorbed using lter paper. Subsequently, the samples were dried at room temperature so that they could
be observed under a 200kV voltage.
In vitro release study and kinetic modeling
e invitro release of CIP and LEV from the NPs was evaluated by the dialysis bag diusion method. e FCIP
or FLEV (equivalent to 2.3mg) was suspended in 1ml of deionized water and placed in pre-equilibrated dialysis
bags (Dialysis Sacks, Avg. Flat Width 35mm, MWCO 12kDa, Sigma-Aldrich). Each dialysis bag was immersed
in a beaker containing 100ml of phosphate buer (pH 7.4) to represent the release medium and maintained at
EE
%=
Total Antibiotic
Free Antibiotic
Total Antibiotic
×
100
AL
%=
Total Antibiotic
Free Antibiotic
Wt of NPs
×
100
Y
%=
Wt of NPs
Wt of Antibiotic
+
PCL
×
100
Content courtesy of Springer Nature, terms of use apply. Rights reserved
13
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
37 ± 0.5°C in a shaking incubator at 100rpm (GFL Gesellscha für Labortechnik, Burgwedel, Germany). Every
predetermined time interval, 2ml aliquots were sampled and replaced with an equal fresh volume. e samples
were then ltered (0.45μm), appropriately diluted, and analyzed using spectrophotometry at 275 and 278nm
for CIP and LEV, respectively. Each experiment was done in triplicate, and the cumulative released percentage
of the antimicrobial was calculated at every time interval using preconstructed calibration curves. To explain the
release mechanisms of CIP and LEV, the release data were kinetically analyzed using the zero-order, rst-order,
Higuchi diusion mechanism, and Korsmeyer-Peppas model. e model with the highest correlation coecient
(r2) was the one that described the release mechanism.
Antimicrobial evaluation of the prepared NPs
e activity of FCIP and FLEV against eight representative XDR-resistant isolates was compared to CIP and
LEV, respectively. e microtiter plate assay method was used to determine MICs of CIP, LEV and their
nanopreparations56. Muller-Hinton broth medium (100μl) was pipetted into sterile microtiter plate wells. CIP,
FCIP, LEV, and FLEV were prepared as two-fold serial dilutions in Muller-Hinton broth medium ranging from 1024
to 0.5μg/ml. Inoculation of all dilutions was made with overnight cultures of the isolates at a nal inoculum of
5 × 105CFU/ml. Positive (culture only) and negative (medium only) controls were performed in all experiments.
Under the same conditions, the MICs of plain NPs were also determined. e plates were incubated at 37°C for
24h. Each experiment was performed in duplicate.
Eect of FCIP and FLEV on eux activity
To study the eect of nanoformulated FQs on FQ resistance via an eux pump mechanism, the MICs of CIP,
FCIP, LEV and FLEV were determined. e eux pump inhibitor CCCP was administered at a subinhibitory
concentration (20μg/ml) to each well. e wells were inoculated with diluted culture (5 × 105CFU/ml) of XDR
A. baumannii isolates (Ab30, Ab60, and Ab72) where the eux mechanism was demonstrated to be one of the
FQ-resistant mechanisms. Furthermore, the eect of CCCP on the MICs of plain NPs, CIP, and LEV against
representative FQ-resistant and XDR A. baumannii isolates and 2 standard strains A. baumannii was investigated.
A positive control for each isolate was included to investigate the viability of dierent isolates in the presence
of CCCP alone. e MDF was determined for each isolate in duplicate. Inhibition of the eux pump by CCCP
was deemed to have a considerable eect when the MDF value was 4 or above57.
Antimicrobial killing assay
e killing rate of FQ-resistant bacteria by FCIP and FLEV was determined and compared to CIP- and LEV-free
antimicrobials, respectively. e investigated XDR A. baumannii isolates (Ab30, Ab60, and Ab72) were propa-
gated until the bacterial count reached 5 × 106CFU/ml. e investigated isolates were treated with FCIP and FLEV
at the MIC and incubated at 37°C. Samples were collected at 0, 1, 2, 3, 4, 6, 10, and 24h, and each sample was
ten-fold serially diluted to determine the viable bacterial count. In the same instance, bacterial counts of cultures
treated with CIP and LEV were also performed under the same conditions. As a control, bacterial growth with-
out free antimicrobials (CIP or LEV) or nanoformulated antimicrobials (FCIP or FLEV) and plain NPs were also
investigated. e surface drop method was used in triplicate to calculate the number of bacteria that recovered
over time following treatment58. Following antimicrobial treatment, the number of recovered cells was plotted
against the CFU/ml over time. Each experiment was carried out in triplicate.
Eect of FCIP and FLEV on biolm formation
e capacity of biolm formation among 80 A. baumannii isolatesand2 standard A. baumannii strains, ATCC
19606 and ATCC17987, was assessed invitro using 96-well microtiter plates as previously mentioned59. e
formed biolm was stained with 1% w/v crystal violet followed by solubilization using glacial acetic acid (33%
v/v). e solubilized biolm was measured using an ELx808TM Absorbance Microplate Reader (BioTek Instru-
ments Inc., Winooski, VT) at OD490nm. A negative control of the medium was included in each experiment. e
mean OD490nm of each bacterial isolate from four independent experiments was calculated to assess the ability
of A. baumannii isolates to produce biolms. Biolm formation by the A. baumannii isolate was repeated in
quadruplicate.
To determine the biolm formation capacity of A. baumannii isolates, the cut-o optical density (ODc)
was established as three standard deviations above the mean OD of the inoculum-free negative control. Strains
were classied as follows: non-biolm producer (N) if OD ODc, weak biolm producer (W) if ODc < ODW ≤ 2
ODc, moderate biolm producer (M) if 2 ODc < ODM 4 ODc, and strong biolm producer (S) if ODS > 4 ODc.
ree representative strong biolm-forming, FQ-resistant, and XDR isolates (Ab30, Ab60, and Ab72) were
selected to assess the eect of FCIP and FLEV on biolm formation. Subinhibitory concentrations for CIP, FCIP,
LEV, and FLEV (1/2 and 1/4 MIC) were incorporated during bacterial incubation in 96-well plates60. Controls of
medium only or cultures without drugs were included in each experiment. e biolm-forming reference strain
of A. baumannii ATCC 19606 was used as a positive control.
Statistical data analysis
Statistical analysis involved calculating the mean and standard deviation of the values. All microbiological assays
were performed in duplicate except the antimicrobial killing assay, and the capacity for biolm formation was
repeated in triplicate and quadruplicate, respectively. A one-way ANOVA test was calculated, with the signi-
cance value set at P < 0.05 or P < 0.01 using the GraphPad Prism soware package (version 8.3.0).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
14
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
Ethics approval
e Research Ethics Committee at the Faculty of Pharmacy at Mansoura University in Egypt (Permit Number:
2022-193) has approved this work as ethically compliant.
Data availability
All data generated or analyzed during this study are included in this published article [and its supplementary
information le].
Received: 23 July 2023; Accepted: 31 January 2024
References
1. Harding, C. M., Hennon, S. W. & Feldman, M. F. Uncovering the mechanisms of Acinetobacter baumannii virulence. Nat. Rev.
Microbiol. 16, 91–102. https:// doi. org/ 10. 1038/ nrmic ro. 2017. 148 (2018).
2. Mohammed, M. A., Salim, M. T. A., Anwer, B. E., Aboshanab, K. M. & Aboulwafa, M. M. Impact of target site mutations and
plasmid associated resistance genes acquisition on resistance of Acinetobacter baumannii to uoroquinolones. Sci. Rep. 11, 20136.
https:// doi. org/ 10. 1038/ s41598- 021- 99230-y (2021).
3. Vrancianu, C. O., Gheorghe, I., Czobor, I. B. & Chiriuc, M. C. Antibiotic resistance proles, molecular mechanisms and innovative
treatment strategies of Acinetobacter baumannii. Microorganisms https:// doi. org/ 10. 3390/ micro organ isms8 060935 (2020).
4. Magiorakos, A. P. et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: An international expert
proposal for interim standard denitions for acquired resistance. Clin. Microbiol. Infect. 18, 268–281. https:// doi. org/ 10. 1111/j.
1469- 0691. 2011. 03570.x (2012).
5. Aldred, K. J., Kerns, R. J. & Oshero, N. Mechanism of quinolone action and resistance. Biochemistry 53, 1565–1574. https:// doi.
org/ 10. 1021/ bi500 0564 (2014).
6. Venkataramana, G. P., Lalitha, A. K. V., Mariappan, S. & Sekar, U. Plasmid-mediated uoroquinolone resistance in Pseudomonas
aeruginosa and Acinetobacter baumannii. J. Lab. Physicians 14, 271–277. https:// doi. org/ 10. 1055/s- 0042- 17426 36 (2022).
7. Saki, M. et al. Occurrence of plasmid-mediated quinolone resistance genes in Pseudomonas aeruginosa strains isolated from clinical
specimens in southwest Iran: A multicentral study. Sci. Rep. 12, 2296. https:// doi. org/ 10. 1038/ s41598- 022- 06128-4 (2022).
8. Bush, N. G., Diez-Santos, I., Abbott, L. R. & Maxwell, A. Quinolones: Mechanism, lethality and their contributions to antibiotic
resistance. Molecules 25, 5662 (2020).
9. Dehnamaki, M., Ghane, M. & Babaeekhou, L. Detection of OqxAB and QepA eux pumps and their association with antibiotic
resistance in Klebsiella pneumoniae isolated from urinary tract infection. Int. J. Infect . 7, e107397. https:// doi. org/ 10. 5812/ iji. 107397
(2020).
10. Mohammed, E.-M., Abdelaziz, E. & Mona, S. In Microorganisms, Ch. 10 (eds Miroslav, B. et al.) (IntechOpen, 2019).
11. Aboelenin, A. M., Hassan, R. & Abdelmegeed, E. S. e eect of EDTA in combination with some antibiotics against clinical isolates
of gram negative bacteria in Mansoura, Egypt. Microb Pathog 154, 104840. https:// doi. org/ 10. 1016/j. micpa th. 2021. 104840 (2021).
12. Wnorowska, U. et al. Nanoantibiotics containing membrane-active human cathelicidin LL-37 or synthetic ceragenins attached to
the surface of magnetic nanoparticles as novel and innovative therapeutic tools: Current status and potential future applications.
J. Nanobiotechnol. 18, 3. https:// doi. org/ 10. 1186/ s12951- 019- 0566-z (2020).
13. Makabenta, J. M. V. et al. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat. Rev. Microbiol. 19,
23–36. https:// doi. org/ 10. 1038/ s41579- 020- 0420-1 (2021).
14. Mahmoud, B. S. & McConville, C. Box–Behnken design of experiments of polycaprolactone nanoparticles loaded with irinotecan
hydrochloride. Pharmaceutics 15, 1271 (2023).
15. Łukasiewicz, S., Mikołajczyk, A., Błasiak, E., Fic, E. & Dziedzicka-Wasylewska, M. Polycaprolactone nanoparticles as promising
candidates for nanocarriers in novel nanomedicines. Pharmaceutics 13, 191 (2021).
16. Hong, C. K., Kim, J. & Kim, G. Characteristics of quinolone resistance in multidrug-resistant Acinetobacter baumannii strains
isolated from general hospitals. Jundishapur J. Microbiol. 14, e115128. https:// doi. org/ 10. 5812/ jjm. 115128 (2021).
17. Moosavian, M. et al. Antimicrobial resistance patterns and their encoding genes among clinical isolates of Acinetobacter baumannii
in Ahvaz, Southwest Iran. MethodsX 7, 101031. https:// doi. org/ 10. 1016/j. mex. 2020. 101031 (2020).
18. Valentine, S. C. et al. Phenotypic and molecular characterization of Acinetobacter baumannii clinical isolates from nosocomial
outbreaks in Los Angeles County, California. J. Clin. Microbiol. 46, 2499–2507. https:// doi. org/ 10. 1128/ jcm. 00367- 08 (2008).
19. Wisplingho, H. et al. Mutations in gyrA and parC associated with resistance to uoroquinolones in epidemiologically dened
clinical strains of Acinetobacter baumannii. J. Antimicrob. Chemother. 51, 177–180. https:// doi. org/ 10. 1093/ jac/ dkf254 (2003).
20. Vila, J., Ruiz, J., Goñi, P., Marcos, A. & Jimenez de Anta, T. Mutation in the gyrA gene of quinolone-resistant clinical isolates of
Acinetobacter baumannii. Antimicrob. Agents Chemother. 39, 1201–1203. https:// doi. org/ 10. 1128/ aac. 39.5. 1201 (1995).
21. Vila, J., Ruiz, J., Goñi, P. & Jimenez de Anta, T. Quinolone-resistance mutations in the topoisomerase IV parC gene of Acinetobacter
baumannii. J. Antimicrob. Chemother. 39, 757–762. https:// doi. org/ 10. 1093/ jac/ 39.6. 757 (1997).
22. Ostrer, L., Khodursky, R. F., Johnson, J. R., Hiasa, H. & Khodursky, A. Analysis of mutational patterns in quinolone resistance-
determining regions of GyrA and ParC of clinical isolates. Int. J. Antimicrob. Agents 53, 318–324. https:// doi. org/ 10. 1016/j. ijant
imicag. 2018. 12. 004 (2019).
23. Higuchi, S., Shikata, M., Chiba, M., Hoshino, K. & Gotoh, N. Characteristics of antibiotic resistance and sequence type of
Acinetobacter baumannii clinical isolates in Japan and the antibacterial activity of DS-8587. J. Infect. Chemother. 20, 256–261.
https:// doi. org/ 10. 1016/j. jiac. 2013. 12. 001 (2014).
24. Bakour, S. et al. Antibiotic resistance determinants of multidrug-resistant Acinetobacter baumannii clinical isolates in Algeria.
Diagn. Microbiol. Infect. Dis. 76, 529–531. https:// doi. org/ 10. 1016/j. diagm icrob io. 2013. 04. 009 (2013).
25. Roca Jalil, M. E., Baschini, M. & Sapag, K. Removal of ciprooxacin from aqueous solutions using pillared clays. Materials (Basel)
https:// doi. org/ 10. 3390/ ma101 21345 (2017).
26. Badri, W. et al. Polycaprolactone based nanoparticles loaded with indomethacin for anti-inammatory therapy: From preparation
to exvivo study. Pharm. Res. 34, 1773–1783. https:// doi. org/ 10. 1007/ s11095- 017- 2166-7 (2017).
27. Gaspar, M. C., Pais, A., Sousa, J. J. S., Brillaut, J. & Olivier, J. C. Development of levooxacin-loaded PLGA microspheres of suitable
properties for sustained pulmonary release. Int. J. Pharm. 556, 117–124. https:// doi. org/ 10. 1016/j. ijpha rm. 2018. 12. 005 (2019).
28. Gupta, H. et al. Biodegradable levooxacin nanoparticles for sustained ocular drug delivery. J. Drug Target. 19, 409–417. https://
doi. org/ 10. 3109/ 10611 86x. 2010. 504268 (2011).
29. Akdag Cayli, Y. et al. Dry powders for the inhalation of ciprooxacin or levooxacin combined with a mucolytic agent for cystic
brosis patients. Drug Dev. Ind. Pharm. 43, 1378–1389. https:// doi. org/ 10. 1080/ 03639 045. 2017. 13189 02 (2017).
30. Sousa de Almeida, M. et al. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chem. Soc.
Rev. 50, 5397–5434. https:// doi. org/ 10. 1039/ D0CS0 1127D (2021).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
15
Vol.:(0123456789)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
31. Wang, L., Hu, C. & Shao, L. e antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J.
Nanomed. 12, 1227–1249. https:// doi. org/ 10. 2147/ ijn. S1219 56 (2017).
32. Costa, P. & Sousa Lobo, J. M. Modeling and comparison of dissolution proles. Eur. J. Pharm. Sci. 13, 123–133. https:// doi. org/ 10.
1016/ S0928- 0987(01) 00095-1 (2001).
33. Uhljar, L. et al. In vitro drug release, permeability, and structural test of ciprooxacin-loaded nanobers. Pharmaceutics https://
doi. org/ 10. 3390/ pharm aceut ics13 040556 (2021).
34. Shaaban, M. I., Shaker, M. A. & Mady, F. M. Imipenem/cilastatin encapsulated polymeric nanoparticles for destroying carbapenem-
resistant bacterial isolates. J. Nanobiotechnol. 15, 29. https:// doi. org/ 10. 1186/ s12951- 017- 0262-9 (2017).
35. Khan, I., Saeed, K. & Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 12, 908–931. https:// doi. org/
10. 1016/j. arabjc. 2017. 05. 011 (2019).
36. Omer, M. E. et al. Novel self-assembled polycaprolactone-lipid hybrid nanoparticles enhance the antibacterial activity of
ciprooxacin. SLAS Technol. 25, 598–607. https:// doi. org/ 10. 1177/ 24726 30320 943126 (2020).
37. Cramariuc, O. et al. Mechanism for translocation of uoroquinolones across lipid membranes. Biochim. Biophys. Acta 1818,
2563–2571. https:// doi. org/ 10. 1016/j. bbamem. 2012. 05. 027 (2012).
38. Yeh, Y.-C., Huang, T.-H., Yang, S.-C., Chen, C.-C. & Fang, J.-Y. Nano-based drug delivery or targeting to eradicate bacteria for
infection mitigation: A review of recent advances. Front. Chem. https:// doi. org/ 10. 3389/ fchem. 2020. 00286 (2020).
39. Lin, M. et al. Synergistic eect of co-delivering ciprooxacin and tetracycline hydrochloride for promoted wound healing by
utilizing coaxial PCL/gelatin nanober membrane. Int. J. Mol. Sci. 23, 1895 (2022).
40. Samiei, M., Farjami, A., Dizaj, S. M. & Lotpour, F. Nanoparticles for antimicrobial purposes in Endodontics: A systematic review
of in vitro studies. Mater. Sci. Eng. C 58, 1269–1278. https:// doi. org/ 10. 1016/j. msec. 2015. 08. 070 (2016).
41. Baptista, P. V. et al. Nano-strategies to ght multidrug resistant bacteria—“A Battle of the Titans”. Front. Microbiol. https:// doi. org/
10. 3389/ fmicb. 2018. 01441 (2018).
42. Huang, L. et al. Bacterial multidrug eux pumps at the frontline of antimicrobial resistance: An overview. Antibiotics 11, 520
(2022).
43. Sanchez-Carbonel, A. et al. e eect of the eux pump inhibitor Carbonyl Cyanide m-Chlorophenylhydrazone (CCCP) on the
susceptibility to imipenem and cefepime in clinical strains of Acinetobacter baumannii. PLoSOne 16, e0259915. https:// doi. org/
10. 1371/ journ al. pone. 02599 15 (2021).
44. Moazzen, Z., Eslami, G., Hashemi, A. & Youse Nojookambari, N. Eux pump inhibitor carbonyl cyanide 3-chlorophenylhydrazone
(CCCP) eect on the minimum inhibitory concentration of ciprooxacin in Acinetobacter baumannii strains. Arch. Clin. Infect.
Dis. 13, e59644. https:// doi. org/ 10. 5812/ archc id. 59644 (2018).
45. Blanco, P. et al. Bacterial multidrug eux pumps: Much more than antibiotic resistance determinants. Microorganisms ht tps:// doi.
org/ 10. 3390/ micro organ isms4 010014 (2016).
46. Modi, S. K., Gaur, S., Sengupta, M. & Singh, M. S. Mechanistic insights into nanoparticle surface-bacterial membrane interactions
in overcoming antibiotic resistance. Front. Microbiol. https:// doi. org/ 10. 3389/ fmicb. 2023. 11355 79 (2023).
47. Banoee, M. et al. ZnO nanoparticles enhanced antibacterial activity of ciprooxacin against Staphylococcus aureus and Escherichia
coli. J. Biomed. Mater. Res. B Appl. Biomater. 93, 557–561. https:// doi. org/ 10. 1002/ jbm.b. 31615 (2010).
48. Abo-Zeid, Y., Amer, A., Bakkar, M. R., El-Houssieny, B. & Sakran, W. Antimicrobial activity of azithromycin encapsulated into
PLGA NPs: A potential strategy to overcome eux resistance. Antibiotics (Basel) https:// doi. org/ 10. 3390/ antib iotic s1111 1623
(2022).
49. Cheow, W. S. & Hadinoto, K. Antibiotic polymeric nanoparticles for biolm-associated infection therapy. Methods Mol. Biol. 1147,
227–238. https:// doi. org/ 10. 1007/ 978-1- 4939- 0467-9_ 16 (2014).
50. Salunke, G. R. et al. Rapid ecient synthesis and characterization of silver, gold, and bimetallic nanoparticles from the medicinal
plant Plumbago zeylanica and their application in biolm control. Int. J. Nanomed. 9, 2635–2653. https:// doi. o r g/ 10. 2147/ i jn. S59834
(2014).
51. Muzammil, S. et al. Aluminium oxide nanoparticles inhibit EPS production, adhesion and biolm formation by multidrug resistant
Acinetobacter baumannii. Biofouling 36, 492–504. https:// doi. org/ 10. 1080/ 08927 014. 2020. 17768 56 (2020).
52. Hosseini, A., Nejadsattari, T. & Zargar, M. In vitro anti-biolm activity of curcumin nanoparticles in Acinetobacter baumannii: A
culture-based and molecular approach. Arch. Clin. Infect. Dis. 14, e83263. https:// doi. org/ 10. 5812/ archc id. 83263 (2019).
53. Hetta, H. F. et al. Antibiolm and antivirulence potential of silver nanoparticles against multidrug-resistant Acinetobacter
baumannii. Sci. Rep. 11, 10751. https:// doi. org/ 10. 1038/ s41598- 021- 90208-4 (2021).
54. Gupta, N., Gandham, N., Jadhav, S. & Mishra, R. N. Isolation and identication of Acinetobacter species with special reference to
antibiotic resistance. J. Nat. Sci. Biol. Med. 6, 159–162. https:// doi. org/ 10. 4103/ 0976- 9668. 149116 (2015).
55. Chen, T. L. et al. Comparison of one-tube multiplex PCR, automated ribotyping and intergenic spacer (ITS) sequencing for rapid
identication of Acinetobacter baumannii. Clin. Microbiol. Infect. 13, 801–806. https:// doi. org/ 10. 1111/j. 1469- 0691. 2007. 01744.x
(2007).
56. CLSI. Performance Standards for Antimicrobial Susceptibility Testing, 30th edn CLSI Supplement M100 (Clinical and Laboratory
Standards Institute, Wayne, PA, 2020).
57. Huguet, A., Pensec, J. & Soumet, C. Resistance in Escherichia coli: Variable contribution of eux pumps with respect to dierent
uoroquinolones. J. Appl. Microbiol. 114, 1294–1299. https:// doi. org/ 10. 1111/ jam. 12156 (2013).
58. Barbosa, H. R. et al. Counting of viable cluster-forming and non cluster-forming bacteria: A comparison between the drop and
the spread methods. J. Microbiol. Methods 22, 39–50. https:// doi. org/ 10. 1016/ 0167- 7012(94) 00062-C (1995).
59. Stepanovic, S., Vukovic, D., Dakic, I., Savic, B. & Svabic-Vlahovic, M. A modied microtiter-plate test for quantication of
staphylococcal biolm formation. J. Microbiol. Methods 40, 175–179. https:// doi. org/ 10. 1016/ s0167- 7012(00) 00122-6 (2000).
60. omas, R., Nair, A. P., Kr, S., Mathew, J. & Ek, R. Antibacterial activity and synergistic eect of biosynthesized AgNPs with
antibiotics against multidrug-resistant biolm-forming coagulase-negative Staphylococci isolated from clinical samples. Appl.
Biochem. Biotechnol. 173, 449–460. https:// doi. org/ 10. 1007/ s12010- 014- 0852-z (2014).
Acknowledgements
Sincere gratitude is extended to Mansoura University’s clinical laboratory for donating clinical isolates.
Author contributions
A.A.: Methodology, data analysis and writing the manuscript. M.E.: Participation in data analysis and writing
of the manuscript. N.S.: Participation in the preparation, evaluation of nanoparticles, data analysis and writing
of manuscript. M.S.: Suggestion the idea of the research, supervision, troubleshooting and revision of the
manuscript. R.B.: Supervision the research and revision of the manuscript. All authors read and approved the
nal manuscript.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
16
Vol:.(1234567890)
Scientic Reports | (2024) 14:3125 | https://doi.org/10.1038/s41598-024-53441-1
www.nature.com/scientificreports/
Funding
Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in
cooperation with e Egyptian Knowledge Bank (EKB).
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 024- 53441-1.
Correspondence and requests for materials should be addressed to M.I.S.orR.B.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the articles Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2024
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Chitosan, a natural polymer derived from chitin, possesses notable biological properties such as biocompatibility and the ability to penetrate cells. Its use enables the formulation of PNPs with potential for targeted drug delivery to specific tissues, along with its capability to interact with biological molecules, making it an appealing choice for biomedical applications (Iqbal et al. 2015;López-Royo et al. 2021;Akel et al. 2021;Alenazi et al. 2022;Aboelenin et al. 2024). ...
... Its unique ability to efficiently encapsulate molecules with antibacterial activity, regardless of their hydrophilic, lipophilic, or amphiphilic nature, underscores its fundamental utility for targeted therapeutic strategies against bacterial infections. The versatility of this approach, combined with its simplicity and cost-effectiveness, not only enables the creation of multifunctional systems but also opens up promising avenues for combating antibioticresistant bacteria and other infectious pathogens in healthcare applications (Heidari et al. 2022;Aboelenin et al. 2024;Majid et al. 2024). ...
Article
Full-text available
The design, development, and obtaining of nanostructured materials, such as polymeric nanoparticles, have garnered interest due to loading therapeutic agents and its broad applicability. Polymeric nanoparticle synthesis employs advanced techniques such as the double emulsion approach and the pH-driven method, allowing the efficient incorporation of active compounds into these matrices. These loading methods ensure compound stability within the polymeric structure and enable control of the release of therapeutic agents. The ability of loaded polymeric nanoparticles to transport and release therapeutic agents on target manner represents a significant advancement in the quest for effective therapeutic solutions. Amid escalating concerns regarding antimicrobial resistance, interventions using polymeric nanostructures stand out for the possibility of carrying antimicrobial agents and enhancing antibacterial action against antibiotic-resistant bacteria, making a new therapeutic approach or complement to conventional treatments. In this sense, the capability of these polymeric nanoparticles to act against Escherichia coli underscores their relevance in controlling bacterial infections. This mini-review provides a comprehensive synthesis of promising techniques for loading therapeutic agents into polymeric nanoparticles highlighting methodologies and their implications, addressing prospects of combating bacterial infections caused by E. coli. Key points • The double emulsion method provides control over size and release of bioactives. • The pH-driven method improves the solubility, stability, and release of active. • The methods increase the antibacterial action of those encapsulated in PNPs. Graphical abstract
... Several factors contribute to the emergence of AMR, including genetic mutations; incomplete antibiotic treatment, which allow pathogens to survive and develop resistance; as well as the overuse and misuse of antibiotics [8]. Biofilms also create the ideal environment for bacterial colonization and growth and the emergence of potentially fatal microbial infections [9]. These biofilms not only significantly contribute to the progression of diseases but also pose a formidable challenge in treatment efficacy. ...
Article
Full-text available
Antimicrobial resistance (AMR), caused by microbial infections, has become a major contributor to morbid rates of mortality worldwide and a serious threat to public health. The exponential increase in resistant pathogen strains including Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) poses significant hurdles in the health sector due to their greater resistance to traditional treatments and medicines. Efforts to tackle infectious diseases caused by resistant microbes have prompted the development of novel antibacterial agents. Herein, we present selenium and copper oxide monometallic nanoparticles (Se-MMNPs and CuO-MMNPs), characterized using various techniques and evaluated for their antibacterial potential via disc diffusion, determination of minimum inhibitory concentration (MIC), antibiofilm, and killing kinetic action. Dynamic light scattering (DLS), scanning electron microscopy (SEM/EDX), and X-ray diffraction (XRD) techniques confirmed the size-distribution, spherical-shape, stability, elemental composition, and structural aspects of the synthesized nanoparticles. The MIC values of Se-MMNPs and CuO-MMNPs against S. aureus and E. coli were determined to be 125 μg/mL and 100 μg/mL, respectively. Time–kill kinetics studies revealed that CuO-MMNPs efficiently mitigate the growth of S. aureus and E. coli within 3 and 3.5 h while Se-MMNPs took 4 and 5 h, respectively. Moreover, CuO-MMNPs demonstrated better inhibition compared to Se-MMNPs. Overall, the proposed materials exhibited promising antibacterial activity against S. aureus and E. coli pathogens.
Article
Full-text available
Background: The Box-Behnken design of experiments (BBD) is a statistical modelling technique that allows the determination of the significant factors in developing nanoparticles (NPs) using a limited number of runs. It also allows the prediction of the best levels of variables to obtain the desired characteristics (size, charge, and encapsulation efficiency) of the NPs. The aim of this study was to examine the effect of the independent variables (amount of polymer and drug, and surfactant concentration) and their interaction on the characteristics of the irinotecan hydrochloride (IRH)-loaded polycaprolactone (PCL) NPs and to determine the most optimum conditions for producing the desired NPs. Methods: The development of the NPs was carried out by a double emulsion solvent evaporation technique with yield enhancement. The NPs data were fitted in Minitab software to obtain the best fit model. Results: By using BBD, the most optimum conditions for producing the smallest size, highest magnitude of charge, and highest EE% of PCL NPs were predicted to be achieved by using 61.02 mg PCL, 9 mg IRH, and 4.82% PVA, which would yield 203.01 nm, -15.81 mV, and 82.35% EE. Conclusion: The analysis by BBD highlighted that the model was a good fit to the data, confirming the suitability of the design of the experiments.
Article
Full-text available
Antimicrobial Resistance (AMR) raises a serious concern as it contributes to the global mortality by 5 million deaths per year. The overall impact pertaining to significant membrane changes, through broad spectrum drugs have rendered the bacteria resistant over the years. The economic expenditure due to increasing drug resistance poses a global burden on healthcare community and must be dealt with immediate effect. Nanoparticles (NP) have demonstrated inherent therapeutic potential or can serve as nanocarriers of antibiotics against multidrug resistant (MDR) pathogens. These carriers can mask the antibiotics and help evade the resistance mechanism of the bacteria. The targeted delivery can be fine-tuned through surface functionalization of Nanocarriers using aptamers, antibodies etc. This review covers various molecular mechanisms acquired by resistant bacteria towards membrane modification. Mechanistic insight on 'NP surface-bacterial membrane' interactions are crucial in deciding the role of NP as therapeutic. Finally, we highlight the potential accessible membrane targets for designing smart surface-functionalized nanocarriers which can act as bacteria-targeted robots over the existing clinically available antibiotics. As the bacterial strains around us continue to evolve into resistant versions, nanomedicine can offer promising and alternative tools in overcoming AMR.
Article
Full-text available
Antimicrobial resistance represents a public health problem with a major negative impact on health and socioeconomic development, and is one of the biggest threats in the modern era. This requires the discovery of new approaches to control microbial infections. Nanomedicine could be one of the promising strategies to improve the treatment of microbial infections. Polymer nanoparticles (PNPs) were reported to overcome the efflux-resistant mechanism toward chemotherapeutic agents. However, to the best of our knowledge, no studies were performed to explore their ability to overcome the efflux-resistant mechanism in bacteria. In the current study, azithromycin (AZI), a macrolide antibiotic, was encapsulated into a biocompatible polymer, poly (lactic-co-glycolic acid) (PLGA) using the nano-precipitation method. The effect of the drug to polymer ratio, surfactant, and pH of the aqueous medium on particle size and drug loading percentage (DL%) were investigated in order to maximize the DL% and control the size of NPs to be around 100 nm. The antibacterial activity of AZI-PLGA NPs was investigated against AZI-resistant bacteria; Methicillin-resistant Staphylococcus aureus (MRSA) and Enterococcus faecalis (E. faecalis), where the efflux mechanism was demonstrated to be one of the resistant mechanisms. AZI-PLGA NPs were safer than free AZI, as revealed from the cytotoxicity test, and were able to overcome the efflux-resistant mechanism, as revealed by decreasing the MIC of AZI-PLGA NPs by four times than free AZI. The MIC value reduced from 256 to 64 µg/mL and from >1000 to 256 µg/mL for MRSA and E. faecalis, respectively. Therefore, encapsulation of AZI into PNPs was shown to be a promising strategy to overcome the efflux-resistant mechanism towards AZI and improve its antibacterial effect. However, future investigations are necessary to explore the effect (if any) of particle size, surface charge, and material composition of PNPs on antibacterial activity. Moreover, it is essential to ascertain the safety profiles of these PNPs, the possibility of their large-scale manufacture, and if this concept could be extended to other antibiotics.
Article
Full-text available
Multidrug efflux pumps function at the frontline to protect bacteria against antimicrobials by decreasing the intracellular concentration of drugs. This protective barrier consists of a series of transporter proteins, which are located in the bacterial cell membrane and periplasm and remove diverse extraneous substrates, including antimicrobials, organic solvents, toxic heavy metals, etc., from bacterial cells. This review systematically and comprehensively summarizes the functions of multiple efflux pumps families and discusses their potential applications. The biological functions of efflux pumps including their promotion of multidrug resistance, biofilm formation, quorum sensing, and survival and pathogenicity of bacteria are elucidated. The potential applications of efflux pump-related genes/proteins for the detection of antibiotic residues and antimicrobial resistance are also analyzed. Last but not least, efflux pump inhibitors, especially those of plant origin, are discussed.
Article
Full-text available
Combining multiple drugs or biologically active substances for wound healing could not only resist the formation of multidrug resistant pathogens, but also achieve better therapeutic effects. Herein, the hydrophobic fluoroquinolone antibiotic ciprofloxacin (CIP) and the hydrophilic broad-spectrum antibiotic tetracycline hydrochloride (TH) were introduced into the coaxial polycaprolactone/gelatin (PCL/GEL) nanofiber mat with CIP loaded into the PCL (core layer) and TH loaded into the GEL (shell layer), developing antibacterial wound dressing with the co-delivering of the two antibiotics (PCL-CIP/GEL-TH). The nanostructure, physical properties, drug release, antibacterial property, and in vitro cytotoxicity were investigated accordingly. The results revealed that the CIP shows a long-lasting release of five days, reaching the releasing rate of 80.71%, while the cumulative drug release of TH reached 83.51% with a rapid release behavior of 12 h. The in vitro antibacterial activity demonstrated that the coaxial nanofiber mesh possesses strong antibacterial activity against E. coli and S. aureus. In addition, the coaxial mats showed superior biocompatibility toward human skin fibroblast cells (hSFCs). This study indicates that the developed PCL-CIP/GEL-TH nanofiber membranes hold enormous potential as wound dressing materials.
Article
Full-text available
This study aimed to assess the presence of qnrA, qnrB, qnrC, qnrD, qnrS, qepA, and aac(6′)-Ib-cr determinants as well as quinolone resistance pattern of clinical isolates of P. aeruginosa in Ahvaz, southwest Iran. A total of 185 clinical isolates of P. aeruginosa were collected from 5 university-affiliated hospitals in Ahvaz, southwest Iran. The disk diffusion method was applied to assess the quinolone resistance pattern. The presence of qnrA, qnrB, qnrC, qnrD, qnrS, qepA, and aac(6′)-Ib-cr genes was investigated by the polymerase chain reaction (PCR) method. Overall, 120 (64.9%) isolates were non-susceptible to quinolones. The most and the less quinolone resistance rates were observed against ciprofloxacin (59.4%) and ofloxacin (45.9%), respectively. The prevalence rates of qnr genes were as follows: qnrA (25.8%), qnrB (29.2%), and qnrS (20.8%). The qnrB gene was the most common type of qnr genes. The qnr genes were occurred in 37.5% (n = 45/120) of quinolne-resistant isolates, simultaneously. The qnrC, qnrD, qepA, and aac(6′)-Ib-cr genes were not recognized in any isolates. In conclusion, the ofloxacin was the most effective quinolone. This study was the first to shed light on the prevalence of PMQR genes among P. aeruginosa isolates in southwest Iran.
Article
Full-text available
Introduction Pseudomonas aeruginosa and Acinetobacter baumannii are important pathogens in health care–associated infections. Fluoroquinolone resistance has emerged in these pathogens. In this study, we aimed to determine the occurrence of plasmid-mediated quinolone resistance (PMQR) determinants (qnrA, qnrB, qnrS, aac(6′)-Ib-cr, oqxAB, and qepA) by polymerase chain reaction (PCR) and the transmissibility of plasmid-borne resistance determinants in clinical isolates of P. aeruginosa and A. baumannii. Materials and Methods The study included P. aeruginosa (85) and A. baumannii (45) which were nonduplicate, clinically significant, and ciprofloxacin resistant. Antibiotic susceptibility testing was done by disk diffusion method for other antimicrobial agents, namely amikacin, ceftazidime, piperacillin/tazobactam, ofloxacin, levofloxacin, and imipenem. Minimum inhibitory concentration of ciprofloxacin was determined. Efflux pump activity was evaluated using carbonyl-cyanide m-chlorophenylhydrazone (CCCP). The presence of PMQR genes was screened by PCR amplification. Transferability of PMQR genes was determined by conjugation experiment, and plasmid-based replicon typing was performed. Results Resistance to other classes of antimicrobial agents was as follows: ceftazidime (86.9%), piperacillin/tazobactam (73.8%), imipenem (69.2%), and amikacin (63.8%). The minimal inhibitory concentration (MIC)50 and MIC90 for ciprofloxacin were 64 and greater than or equal to 256 µg/mL, respectively. There was a reduction in MIC for 37 (28.4%) isolates with CCCP. In P. aeruginosa, 12 (14.1%) isolates harbored qnrB, 12 (14.1%) qnrS, 9 (10.5%) both qnrB and qnrS, 66 (77.6%) aac(6′)-Ib-cr, and 3 (3.5%) oqxAB gene. In A. baumannii, qnrB was detected in 2 (4.4%), 1 (2.2%) harbored both the qnrA and qnrS, 1 isolate harbored qnrB and qnrS, 21 (46.6%) aac(6′)-Ib-cr, and 1 (2.2%) isolate harbored oqxAB gene. Notably, qepA gene was not detected in any of the study isolates. Conjugation experiments revealed that 12 (9.2%) were transferable. Of the transconjugants, seven (58.3%) belonged to IncFII type plasmid replicon, followed by four (33.3%) IncA/C and one (8.3%) IncFIC type. Conclusion The plasmid-mediated resistance aac(6′)-Ib-cr gene is primarily responsible for mediating fluoroquinolone resistance in clinical isolates of P. aeruginosa and A. baumannii. The predominant plasmid type is IncFII.
Article
Full-text available
Introduction In the last years the rapid expansion of multidrug-resistant A . baumannii strains have become a major health problem. Efflux pumps are a group of transport proteins that contribute to the development of antibiotic resistance. The aim of this study was to evaluate the effect of the efflux pump inhibitor carbonyl cyanide 3-chlorophenylhydrazone (CCCP) on the antimicrobial action of imipenem and cefepime on clinical strains of A . baumannii . Materials and methods A total of 49 non-duplicate clinical samples were collected during January through December of 2018 from patients hospitalized in the Hospital Regional Docente de Cajamarca . Of the 49 samples obtained, the confirmatory identification of A . baumannii was performed on 47 samples by molecular methods. The amplification of the bla OXA-51-like gene was carried out by polymerase chain reaction (PCR). The determination of the minimum inhibitory concentration (MIC) was calculated using the microdilution method in culture broth. The susceptibility to both antibiotics (cefepime and imipenem) was evaluated in the presence and absence of the inhibitor carbonyl cyanide 3-chlorophenylhydrazone (CCCP). Results A total of 47 strains of A . baumannii were isolated: 97.87% (46/47) were resistant to Imipenem, 2.13% (1/47) of them were classified as intermediate and none of these strains were susceptible. On the other hand, 51.06% (24/47) of isolates were resistant to cefepime; 19.15% (9/47) intermediate and 29.79% (14/47) susceptible. We considered a significant difference in antibiotic susceptibility if the MIC changed at least 4 dilutions, after the addition of the inhibitor. In the case of CCCP in addition to imipenem, 2.1% (1/47) had a significant change of 4 or more reductions in MIC, 59.6% (28/47) achieved a change equal or less than 3 dilutions and 17.0% (8/47) did not have any change. In the case of CCCP with cefepime the percentage of strains with the significant change of MIC was 8.5% (4/47). On the other hand, 53.2% (24/47) presented a reduction equal or less than 3 dilutions and 12.8% (6/47) did not show changes. Conclusion In conclusion, our results demonstrate that the use of CCCP may improve the antibiotic effect of imipenem and cefepime on clinical strains of A . baumannii . The relevance of this study is that it provides evidence that this efflux pump inhibitor may be an alternative treatment against multidrug-resistant A . baumannii .
Article
Full-text available
Among bacterial species implicated in hospital-acquired infections are the emerging Pan-Drug Resistant (PDR) and Extensively Drug-Resistant (XDR) Acinetobacter (A.) baumannii strains as they are difficult to eradicate. From 1600 clinical specimens, only 100 A. baumannii isolates could be recovered. A high prevalence of ≥ 78% resistant isolates was recorded for the recovered isolates against a total of 19 tested antimicrobial agents. These isolates could be divided into 12 profiles according to the number of antimicrobial agents to which they were resistant. The isolates were assorted as XDR (68; 68%), Multi-Drug Resistant (MDR: 30; 30%), and PDR (2; 2%). Genotypically, the isolates showed three major clusters with similarities ranging from 10.5 to 97.8% as revealed by ERIC-PCR technique. As a resistance mechanism to fluoroquinolones (FQs), target site mutation analyses in gyrA and parC genes amplified from twelve selected A. baumannii isolates and subjected to sequencing showed 12 profiles. The selected isolates included two CIP-susceptible ones, these showed the wild-type profile of being have no mutations. For the ten selected CIP-resistant isolates, 9 of them (9/10; 90%) had 1 gyrA/1 parC mutations (Ser 81 → Leu mutation for gyrA gene and Ser 84 → Leu mutation for parC gene). The remaining CIP-resistant isolate (1/10; 10%) had 0 gyrA/1 parC mutation (Ser 84 → Leu mutation for parC gene). Detection of plasmid-associated resistance genes revealed that the 86 ciprofloxacin-resistant isolates carry qnrA (66.27%; 57/86), qnrS (70.93%; 61/86), aac (6')-Ib-cr (52.32%; 45/86), oqxA (73.25%; 63/86) and oqxB (39.53%; 34/86), while qepA and qnrB were undetected in these isolates. Different isolates were selected from profiles 1, 2, and 3 and qnrS, acc(6,)-ib-cr, oqxA, and oqxB genes harbored by these isolates were amplified and sequenced. The BLAST results revealed that the oqxA and oqxB sequences were not identified previously in A. baumannii but they were identified in Klebsiella aerogenes strain NCTC9793 and Klebsiella pneumoniae, respectively. On the other hand, the sequence of qnrS, and acc(6,)-ib-cr showed homology to those of A. baumannii. MDR, XDR, and PDR A. baumannii isolates are becoming prevalent in certain hospitals. Chromosomal mutations in the sequences of GyrA and ParC encoding genes and acquisition of PAFQR encoding genes (up to five genes per isolate) are demonstrated to be resistance mechanisms exhibited by fluoroquinolones resistant A. baumannii isolates. It is advisable to monitor the antimicrobial resistance profiles of pathogens causing nosocomial infections and properly apply and update antibiotic stewardship in hospitals and outpatients to control infectious diseases and prevent development of the microbial resistance to antimicrobial agents.
Article
Full-text available
Background: Acinetobacter baumannii is the causative agent in various types of hospital-acquired infections, including respiratory, urinary tract, and wound infections. Objectives: This study investigated the primary mechanisms underlying quinolone resistance in A. baumannii strains, isolated from samples collected from general hospitals. Methods: Ninety-eight strains of A. baumannii were isolated from clinical specimens from general hospitals from 2017 – 2019. Antimicrobial susceptibility, efflux pump inhibition tests, multilocus sequence typing (MLST), and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analyses were conducted on 64 strains, and the blaoxa-51-like gene sequence was detected. Results: In the antimicrobial susceptibility test, 78.1% (n = 50) of the strains exhibited resistance to ciprofloxacin, a quinolone antibiotic, and 57.8% (n = 37) strains were multidrug resistant (MDR). For 18 strains, the minimum inhibitory concentration of ciprofloxacin reduced in presence of an efflux pump inhibitor. Sequence analysis revealed that in 50 strains of A. baumannii, the codon for serine (TCA) in gyrA was replaced by that for leucine (TTA), whereas in 43 strains, the codon for serine (TCG) in parC was replaced by that for leucine (TTG). Multilocus sequence typing analysis confirmed 18 sequence types, and allelic number analysis showed the presence of nine gyrB alleles, with gyrB3 showing the highest frequency (62.5%). Conclusions: The findings of this study will be useful in improving treatment efficiency and preventing the spread of A. baumannii (both MDR and non-MDR strains).