ArticlePDF Available

Mechanisms for Rapid Evolution of Carbapenem Resistance in a Clinical Isolate of Pseudomonas aeruginosa

Frontiers
Frontiers in Microbiology
Authors:

Abstract and Figures

Infections by Pseudomonas aeruginosa are difficult to cure due to its high intrinsic and acquired antibiotic resistance. Once colonized the human host, and thanks to antibiotic treatment pressure, P. aeruginosa usually acquires genetic mutations which provide bacteria with antibiotic resistance as well as ability to better adapt to the host environment. Deciphering the evolutionary traits may provide important insights into the development of effective combinatory antibiotic therapy to treat P. aeruginosa infections. In this study, we investigated the molecular mechanisms by which a clinical isolate (ISP50) yields a carbapenem-resistant derivative (IRP41). RNAseq and genomic DNA reference mapping were conducted to compare the transcriptional profiles and in vivo evolutionary trajectories between the two isolates. Our results demonstrated that oprD mutation together with ampC hyper-expression contributed to the increased resistance to carbapenem in the isolate IRP41. Furthermore, a ldcA (PA5198) gene, encoding murein tetrapeptide carboxypeptidase, has been demonstrated for the first time to negatively influence the ampC expression in P. aeruginosa.
Content may be subject to copyright.
fmicb-11-01390 June 18, 2020 Time: 17:19 # 1
ORIGINAL RESEARCH
published: 19 June 2020
doi: 10.3389/fmicb.2020.01390
Edited by:
Yuji Morita,
Meiji Pharmaceutical University, Japan
Reviewed by:
Carlos Juan,
Instituto de Investigación Sanitaria
de Palma (IdISPa), Spain
Erwin Bohn,
University Hospital Tübingen,
Germany
*Correspondence:
Fang Bai
baifang1122@nankai.edu.cn
Yongxin Jin
yxjin@nankai.edu.cn
Specialty section:
This article was submitted to
Antimicrobials, Resistance
and Chemotherapy,
a section of the journal
Frontiers in Microbiology
Received: 31 January 2020
Accepted: 29 May 2020
Published: 19 June 2020
Citation:
Xu C, Wang D, Zhang X, Liu H,
Zhu G, Wang T, Cheng Z, Wu W,
Bai F and Jin Y (2020) Mechanisms
for Rapid Evolution of Carbapenem
Resistance in a Clinical Isolate
of Pseudomonas aeruginosa.
Front. Microbiol. 11:1390.
doi: 10.3389/fmicb.2020.01390
Mechanisms for Rapid Evolution of
Carbapenem Resistance in a Clinical
Isolate of Pseudomonas aeruginosa
Congjuan Xu1, Dan Wang1, Xinxin Zhang1, Huimin Liu2, Guangbo Zhu2, Tong Wang3,
Zhihui Cheng1, Weihui Wu1, Fang Bai1*and Yongxin Jin1*
1State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the
Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China, 2Tianjin Union
Medical Center, Nankai University Affiliated Hospital, Tianjin, China, 3Department of Stomatology, Tianjin First Central
Hospital, Tianjin, China
Infections by Pseudomonas aeruginosa are difficult to cure due to its high intrinsic
and acquired antibiotic resistance. Once colonized the human host, and thanks to
antibiotic treatment pressure, P. aeruginosa usually acquires genetic mutations which
provide bacteria with antibiotic resistance as well as ability to better adapt to the host
environment. Deciphering the evolutionary traits may provide important insights into the
development of effective combinatory antibiotic therapy to treat P. aeruginosa infections.
In this study, we investigated the molecular mechanisms by which a clinical isolate
(ISP50) yields a carbapenem-resistant derivative (IRP41). RNAseq and genomic DNA
reference mapping were conducted to compare the transcriptional profiles and in vivo
evolutionary trajectories between the two isolates. Our results demonstrated that oprD
mutation together with ampC hyper-expression contributed to the increased resistance
to carbapenem in the isolate IRP41. Furthermore, a ldcA (PA5198) gene, encoding
murein tetrapeptide carboxypeptidase, has been demonstrated for the first time to
negatively influence the ampC expression in P. aeruginosa.
Keywords: P. aeruginosa, carbapenem resistance, oprD,ampC,ldcA
INTRODUCTION
Pseudomonas aeruginosa, as an opportunistic human pathogen, is one of the leading causes of
nosocomial infections worldwide (Vincent et al., 1995). Infections by P. aeruginosa are difficult
to treat due to its intrinsic and acquired resistance to a wide range of antibiotics, leaving limited
number of effective antimicrobial agents. Carbapenems are used in clinical practice to treat
P. aeruginosa infections. However, carbapenem resistance of clinical P. aeruginosa isolates has been
increasingly reported (Davies et al., 2011). The mechanisms of carbapenem resistance are usually
multifactorial which include: (i) acquisition of carbapenemase encoding genes through horizontal
gene transfer (Poole, 2011;Potron et al., 2015), (ii) deficiency or repression of the porin (OprD)
for carbapenem (Davies et al., 2011;Poole, 2011), (iii) overexpression of mexAB-oprM efflux pump
(Poole, 2011;Liu et al., 2013;Choudhury et al., 2015), and (iv) overexpression of the chromosomal
gene (ampC) encoding the P. aeruginosa intrinsic cephalosporinase (Poole, 2011;Mirsalehian et al.,
2014). Although these and other studies have described the associated mechanisms of carbapenem
resistance among clinical isolates of P. aeruginosa, there is little information on the detailed
molecular mechanisms leading to the evolutionary dynamics of clinical P. aeruginosa isolates from
carbapenems susceptibility to resistance and the impact of each of these resistance mechanisms.
Frontiers in Microbiology | www.frontiersin.org 1June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 2
Xu et al. Mechanisms of Carbapenem Resistance
In this study, we obtained two P. aeruginosa clinical isolates,
later demonstrated to belong to the same clone, from sputum
samples of the same patient with acute exacerbation of chronic
bronchitis before and after treatment with biapenem. The first
strain was obtained soon after the patient was admitted to the
hospital while the second strain was obtained 4 days after the
antibiotic treatment. The first isolate ISP50 was carbapenem
susceptible, whereas the second one IRP41 was carbapenem
resistant. Therefore, our goal was to decipher the molecular
mechanisms by which the carbapenem resistance had been
evolved so rapidly in the clinical setting. Our experimental results
demonstrated that an oprD null mutation combined with an
elevated ampC expression are the major contributory factors for
the conversion. Furthermore, we have shown for the first time
that LdcA functions as a repressor on the expression of ampC
in P. aeruginosa. These findings provide novel insights into the
regulatory mechanism of ampC expression in P. aeruginosa.
MATERIALS AND METHODS
Basic Characterization of the Bacterial
Strains
Bacterial strains and plasmids used in the study are listed in
Table 1. Carbapenem-susceptible (ISP50) and resistant (IRP41)
P. aeruginosa isolates characterized in this study were obtained
from sputum samples of the same patient with acute exacerbation
of chronic bronchitis before and after treatment with biapenem
for 4 days (dosage at 0.3 g ×2/day) at the Nankai University
Affiliated Hospital, Tianjin, China. The 16S rRNA encoding gene
was amplified (primers listed in Supplementary Table S1) and
sequenced to identify the species of these two isolates (Spilker
et al., 2004). Random amplified polymorphic DNA (RAPD)
typing was carried out using primer 272 as described previously
(Mahenthiralingam et al., 1996). Antibiotic susceptibility test
was conducted using the disk diffusion method according to
the manufacturer’s recommendations, and minimum inhibitory
concentration (MIC) of antibiotics was determined by the
two-fold serial dilution method. Susceptibility was interpreted
according to the Clinical and Laboratory Standards Institute
guidelines (CLSI 2011–2018).
Plasmid Construction
For overexpression of oprD, a 1,802 bp oprD-containing
fragment with its putative Shine-Dalgarno (SD) sequence
was PCR amplified using ISP50 and IRP41 genomic DNA as
templates (the used primers are displayed in Supplementary
Table S1). The PCR products were digested with BamHI
and HindIII, and then ligated into a shuttle vector pUCP24,
resulting in pUCP24-oprDISP50 and pUCP24-oprDIRP41,
respectively. Constructs of pUCP24-ampC, pMMB-ampRISP50,
pMMB-ampRIRP41, pUCP24-ldcAISP50 , pUCP24-ldcAIRP41 and
pUCP24-pbpC were all generated using similar procedures.
For deletion of the ldcA gene, a 836 bp fragment immediately
upstream of the ldcA start codon and a 913 bp fragment
downstream of the ldcA stop codon were PCR amplified,
digested with EcoRI-BamHI and BamHI-HindIII, respectively.
TABLE 1 | Bacterial strains and plasmids used in this study.
Strain or plasmid Description Source of
reference
Strains
DH5αFφ80dlacZ1M15 endA1 recA1
hsdR17(rKmK+)supE44 thi-1 relA1
1(lacZYA-argF)U169 gyrA96 deoR
TransGen
S17-1 RP4-2 Tc::Mu Km::Tn7TprSmrPro Res
Mod+
Dr. Ramphal
ISP50 Clinical isolate from sputum samples of the
a female patient with acute exacerbations
of chronic bronchitis before treatment with
biapenem, IMPs
This study
IRP41 Clinical isolate from sputum samples of the
a female patient with acute exacerbations
of chronic bronchitis after treatment with
Biapenem for 4 days, IMPR
This study
Klebsiella
pneumoniae ATCC
700603
Klebsiella pneumoniae ATCC 700603 Fuxiang
Biotechnology
PA-NK41 KPC2 carbapenemase producing
P. aeruginosa
This study
PA14 Wild type P. aeruginosa strain Liberati et al.
(2006)
PA14pbpC::Tn PA14 with pbpC disrupted by insertion of
Tn
Liberati et al.
(2006)
PAO1 Wild type P. aeruginosa strain Kaufman et al.
(2000)
PAO11ldcA PAO1 with ldcA gene deleted This study
PA141ldcA PA14 with ldcA gene deleted This study
ISP501ldcA ISP50 with ldcA gene deleted This study
PAO11ampR PAO1 with ampR gene deleted This study
Plasmids
pUCP24 Shuttle vector between E. coli and
P. aeruginosa; Gmr
West et al.
(1994)
pMMB67EH-Gm Shuttle vector between E. coli and
P. aeruginosa; Gmr
Long et al.
(2019)
pEX18Tc Gene knockout vector, TcrHoang et al.
(1998)
pUCP24-oprDISP50 oprD gene from ISP50 in pUCP24, GmrThis study
pUCP24-oprDIRP41 oprD gene from IRP41 in pUCP24, GmrThis study
pUCP24-ampC ampC gene from ISP50 in pUCP24, GmrThis study
pUC18T-miniTn7T-
ampC-His
C-terminal His-tagged ampC in
pUC18T-miniTn7T, Tcr
This study
pUCP24-pbpC pbpC gene from ISP50 in pUCP24, GmrThis study
pUCP24-ldcAISP50 ldcA gene from ISP50 in pUCP24, GmrThis study
pUCP24-ldcAIRP41 ldcA gene from IRP41 in pUCP24, GmrThis study
pMMB-ampRISP50 ampR gene from ISP50 in
pMMB67EH-Gm, Gmr
This study
pMMB-ampRIRP41 ampR gene from IRP41 in
pMMB67EH-Gm, Gmr
This study
pEX18-ldcA ldcA gene deletion on pEX18Tc, TcrThis study
pEX18-ampR ampR gene deletion on pEX18Tc, TcrThis study
The two fragments were then ligated into pEX18Tc that was
digested with EcoRI and HindIII, resulting in pEX18-ldcA.
A pEX18-ampR was constructed by similar manipulations.
Gene knockouts in P. aeruginosa were carried out by conjugal
transfer of these plasmids followed by selection for single
Frontiers in Microbiology | www.frontiersin.org 2June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 3
Xu et al. Mechanisms of Carbapenem Resistance
crossovers and then double crossovers as previously described
(Schweizer, 1992).
Multilocus Sequence Typing
Multilocus sequence typing (MLST) was performed following
a previous description (Curran et al., 2004) with minor
modifications to confirm the allelic profiles of the two isolates.
Briefly, genomic DNA was isolated from overnight bacterial
culture with a DNA purification kit (Tiangen Biotech, Beijing,
China) for use as PCR template. The internal fragments of
seven housekeeping genes (acsA,aroE,guaA,mutL,nuoD,ppsA
and trpE) were amplified by PCR and sequenced using primers
described previously (Supplementary Table S1) (Curran et al.,
2004). Gene sequences were then submitted to the P. aeruginosa
MLST database1for assignment of allelic numbers. A sequence
type (ST) was assigned to each isolate by combining the seven
allelic numbers.
PAE-MHT Assay for Carbapenemase
Test
The PAE-MHT assay was performed as described previously
(Pasteran et al., 2011) with minor modifications. Briefly, an
inoculum of Klebsiella pneumoniae ATCC 700603 was adjusted
to a 0.5 McFarland turbidity standard (Mcfarland, 1907) followed
by 10-fold dilution with sterile saline, and then inoculated onto
the surfaces of Mueller-Hinton agar (Oxoid) plates (diameter,
10 cm) by swabbing. The plates were allowed to stand at room
temperature. After 10 min, a disk of 10 µg imipenem/meropenem
(Oxoid, United Kingdom) was placed in the center of each
plate. Subsequently, one colony of each P. aeruginosa strain,
grown overnight on LB agar plate, was inoculated onto the
plate in a straight line from the edge of the disk to the
periphery of the plate using a 1-µl loop (BOOPU Biological
Technology, Changzhou, China). Presence of growth of the
K. pneumoniae ATCC 700603 toward imipenem/meropenem
disk was interpreted as carbapenemase positive.
Total RNA Isolation, RT-qPCR and
RNAseq Analysis
Bacterial overnight culture was inoculated into fresh LB medium
(1:50 dilution) and grown to an optical density of 1.0 (wavelength
of 600 nm). Total RNA was extracted using an RNAprep Pure
Cell/Bacteria Kit (Tiangen Biotech, Beijing, China) and reverse
transcribed to cDNA with random primers and PrimeScript
Reverse Transcriptase (Takara). The cDNA was mixed with
indicated primers (Supplementary Table S1) and SYBR premix
Ex Taq II (Takara), and then quantitatively amplified in a
CFX Connect Real-Time system (Bio-Rad, United States).
A 30S ribosomal protein encoding gene rpsL was used as an
internal control.
For RNAseq, total RNA was extracted as described above,
quantified and qualified by Agilent 2100 Bioanalyzer (Agilent
Technologies, Palo Alto, CA, United States), NanoDrop (Thermo
Fisher Scientific Inc.) and 1% agarose gel. Then, the rRNA
1http://pubmlst.org/paeruginosa/
was depleted using Ribo-Zero rRNA Removal Kit (Bacteria,
Illumina). The mRNA was then fragmented and reverse-
transcribed. The double-strand cDNA was purified, ends repaired
and ligated with adaptors. After 11 cycles of PCR amplification,
the PCR products were cleaned, validated with an Agilent 2100
Bioanalyzer (Agilent Technologies, Palo Alto, CA, United States),
and quantified by Qubit 2.0 Fluorometer (Invitrogen, Carlsbad,
CA, United States). The resulting libraries were subjected to
sequencing using an Illumina HiSeq 2500 platform with a 2 ×150
paired-end configuration.
Sequence reads were mapped onto PAO1 reference genome
(NC_002516.2) via software Bowtie2 (v2.1.0). Gene expression
levels were analyzed with HTSeq (v0.6.1p1). Differentially
expressed genes were identified by the DESeq Bioconductor
package, with the fold-change larger than 2 and P-value no more
than 0.05 as cutoff values.
DNA Isolation and Reference Mapping
Bacterial genomic DNA was extracted with DNA purification
kit (Tiangen Biotech, Beijing, China). Fragments smaller than
500 bp were obtained from 200 ng genomic DNA by
sonication (Covaris S220), followed by end treatment and
adaptors ligation. Adaptor-ligated DNA fragments of about
470 bp were recovered using beads and then PCR amplified
for six cycles, and the PCR products were cleaned up using
beads, validated using a Qsep100 (Bioptic, Taiwan, China), and
quantified by Qubit3.0 Fluorometer (Invitrogen, Carlsbad, CA,
United States). Sequencing was carried out using a 2 ×150
paired-end (PE) configuration on an Illumina Hiseq instrument
according to manufacturer’s instructions (Illumina, San Diego,
CA, United States). The data were aligned to the PAO1 reference
genome (NC_002516.2) via software BW2 (version 0.7.12). Single
nucleotide variation (SNV) or InDel mutation were detected
using the software Samtools (version 1.1) and the Unified
Genotyper module from GATK.
PCR and Sequencing of oprD,ampR and
ldcA
The full-length oprD gene with its 105 bp upstream and 209 bp
downstream region was amplified and sequenced using primers
listed in Supplementary Table S1. The oprD gene sequence from
each isolate was aligned with the oprD sequence of the reference
strain PAO12. Analysis of ampR and ldcA were conducted with
similar procedures.
Western Blot Assay
Subcultured samples from equivalent number of bacterial cells
of an optical density of 1.0 (wavelength of 600 nm) were
separated on a 12% SDS-PAGE. The proteins were then
transferred onto a PVDF (polyvinylidene difluoride) membrane
and probed with a mouse monoclonal antibody against His-
tag (Cell Signaling Technology, United States) and the RNA
polymerase alpha subunit RpoA (Biolegend). Signals were
generated using the ECL-Plus kit (Millipore) and detected by a
Biorad imager (ChemiDocXRS).
2www.pseudomonas.com
Frontiers in Microbiology | www.frontiersin.org 3June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 4
Xu et al. Mechanisms of Carbapenem Resistance
Quantification of Biofilm
Biofilm production was measured as described previously (Li
et al., 2013). Overnight culture of each bacterium was diluted
50-fold in LB broth and incubated in each well of a 96-well
plate at 37C. After 24 h, each well was washed with phosphate
buffered saline (PBS, 137 mM NaCl, 10 mM Na2HPO4, 2.7 mM
KCl, pH 7.4) for three times, stained with 0.1% crystal violet, and
then washed three times with PBS. Then, 0.2 ml of decolorizing
solution [methanol (4): acetic acid (1): water (5)] was added
into each well and incubated 10 min at room temperature. Each
sample was then subjected to measurement for OD590 using
Varioskan Flash (Thermo Scientific, Netherlands).
Statistical Analysis
Graphpad software was used to perform the statistical analyses.
The real-time qPCR results were analyzed by Student’s
t-test (two-tailed).
RESULTS
Clinical Isolates ISP50 and IRP41 Belong
to the Same Clone
Sputum samples from a patient with acute exacerbation of
chronic bronchitis were collected before and after treatment
with biapenem for 4 days (dosage at 0.3 g ×2/day). Two
mucoid isolates were selected, one before the biapenem treatment
which was sensitive to imipenem (ISP50), and the other one
after the biapenem treatment which showed resistance against
the imipenem (IRP41). PCR amplification of the 16S rDNAs
as well as their sequence analysis indicated that both of them
were P. aeruginosa (Supplementary Figure S1A). The MLST
analysis of both isolates revealed an allelic profile for acsA,aroE,
guaA,mutL,nuoD,ppsA and trpE as 17, 5, 12, 3, 14, 4, 7,
respectively, corresponding to the same sequence type, ST244.
RAPD typing was further performed on the IRP41 and ISP50
isolates, which also suggested that they were of the same clone
(Supplementary Figure S1B).
Isolate IRP41 Is Resistant to
Carbapenem
Imipenem susceptibilities of IRP41 and ISP50 were found
different based on the preliminary analysis with VITEK
automatic microbe analysis instrument (data not shown).
Therefore, we further examined the imipenem susceptibilities by
both MIC and disk diffusion methods, with the results shown
in Table 2 and Supplementary Figure S2. Initial isolate (ISP50)
was found to be susceptible to the imipenem (53.3 mm/diameter
of inhibition zone), while later isolate (IRP41) was resistant
to the imipenem (10.7 mm/diameter of inhibition zone),
with a 32-fold increase in MIC of imipenem. Although both
strains were susceptible to meropenem (another carbapenem
antibiotic) according to the Clinical and Laboratory Standards
Institute guidelines (CLSI 2011–2018), IRP41 displayed a smaller
diameter of inhibition zone (24 mm) compared to that of ISP50
(44 mm) (Supplementary Figures S2A–D). In addition, MIC
TABLE 2 | MICs (µg/ml) of indicated P. aeruginosa strains.
strains Imipenem
(µg/ml)a
Ampicillin
(µg/ml)
Meropenem
(µg/ml)a
Biapenem
(µg/ml)b
ISP50 0.3125 ND 3.125 0.78125
IRP41 10 ND 6.25 6.25
ISP50/pUCP24 0.3125 ND 3.125 0.78125
IRP41/pUCP24 10 ND 6.25 6.25
IRP41/pUCP24-oprDISP50 0.625 ND 3.125 3.125
IRP41/pUCP24-oprDIRP41 10 ND 6.25 6.25
IRP41/pUCP24-ldcAISP50 5 ND 3.125 3.125
IRP41/pUCP24-ldcAIRP41 10 ND 6.25 6.25
PAO1 1.5625 625 3.125 1.5625
PAO11ldcA 0.78125 1250 1.5625 0.78125
PA14 1.5625 312.5 1.5625 1.5625
PA141ldcA 0.78125 1250 0.3906 0.3906
ND, not determined; aClinical Laboratory Standards Institute (CLSI) susceptibility
breakpoints: imipenem, meropenem 2µg/ml, resistance breakpoints: imipenem,
meropenem 8µg/ml. bno CLSI breakpoint concentrations for biapenem have
been established, but previous studies suggested that MIC 4µg/ml is sensitive
and MIC 16 µg/ml is resistant (Hoban et al., 1993;Hang et al., 2018).
of meropenem/biapenem in IRP41 showed two-fold/eight-fold
increase compared to ISP50 (as displayed in Table 2).
Since the production of horizontally acquired carbapenem-
hydrolyzing enzymes has been defined as a contributory factor in
clinical P. aeruginosa isolates with carbapenem resistance (Potron
et al., 2015), we tested if the IRP41 produced carbapenemase
using a PAE-MHT assay. As the results shown in Supplementary
Figures S2E,F, compared to the PA-NK41, which produces a
KPC2 carbapenemase, no carbapenemase activity was observed
in the IRP41 or ISP50 strain, suggesting that the carbapenem
resistant phenotype of isolate IRP41 is not due to the presence
of any horizontally acquired carbapenemases.
Differential Expression of Genes
Relevant to β-Lactam Resistance
To elucidate the mechanism of reduced susceptibility to
carbapenem in isolate IRP41, we compared the global gene
expression profiles between strains ISP50 and IRP41. Expression
levels of 284 genes were altered comparing the two strains
(Table 3 and Supplementary Table S2). Among them, ampC
gene, encoding the β-lactamase, displayed a 285-fold higher
mRNA level in IRP41 than that in ISP50 (Table 3). To confirm
the up-regulation, we further examined the mRNA levels by real-
time qPCR. As shown in Figure 1A, consistent with the RNAseq
result, the mRNA level of ampC was 413-fold higher in the strain
IRP41 than that in ISP50. To further determine if the increased
ampC expression contributed to the reduced susceptibility to
carbapenem in IRP41, the ampC gene was overexpressed in
the ISP50 strain background and the MIC of carbapenems
was examined. As shown in Supplementary Table S3 and
Supplementary Figure S3A, overexpression of the ampC (1,239-
fold increase compared to that in ISP50/pUCP24) reduced
the susceptibility of ISP50 to imipenem/meropenem/biapenem,
with 16/2/4-fold increase in MIC. These results suggested
that increased expression of the ampC indeed contributed
Frontiers in Microbiology | www.frontiersin.org 4June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 5
Xu et al. Mechanisms of Carbapenem Resistance
TABLE 3 | mRNA levels of genes related to β-lactam resistance and biofilm in
IRP41 compared to those in ISP50.
Gene ID Gene
name
Gene function Fold change
(IRP41/ISP50)
P-value
PA4110 ampC Beta-lactamase 285 6.42E20
PA2233 pslC Biofilm formation protein PslC 30 4.71E09
PA2272 pbpC Penicillin-binding protein 3A 8 1.49E05
PA3719 armR MexR antirepressor ArmR 5 0.001828
PA2234 pslD Biofilm formation protein PslD 11 3.63E06
PA2235 pslE Biofilm formation protein PslE 16 8.15E08
PA2236 pslF Biofilm formation protein PslF 18 9.04E08
PA2237 pslG Biofilm formation protein PslG 8 4.15E05
PA2238 pslH Biofilm formation protein PslH 11 3.93E06
PA2239 pslI Biofilm formation protein PslI 10 7.32E06
PA2240 pslJ Biofilm formation protein PslJ 7 0.000124
PA2241 pslK Biofilm formation protein PslK 5 0.002139
PA2242 pslL Biofilm formation protein PslL 5 0.001121
to the decreased susceptibility of IRP41 to carbapenems, at
least partially.
Besides the ampC, a pbpC gene, encoding penicillin
binding protein 3a, displayed an eight-fold up-regulation in
IRP41 (Table 3). Real-time qPCR confirmed the increased
expression of the pbpC in IRP41 (Figure 1B). However,
overexpression of the pbpC (Supplementary Figure S3B)
in ISP50 (ISP50/pUCP24-pbpC) had no effect on the
bacterial MIC against imipenem/meropenem/biapenem
(Supplementary Table S3), although it restored the MIC of
carbenicillin in PA14pbpC::Tn to that of wild type PA14 strain
(Supplementary Table S3). These results indicated that the
elevated pbpC expression did not contribute to the carbapenem
resistance in IRP41.
Our RNAseq analysis also revealed that armR, encoding MexR
antirepressor (ArmR), was up-regulated five folds in IRP41. It
had been demonstrated that MexR represses the expression of
mexAB-oprM efflux pump (Daigle et al., 2007) whose over-
expression confers meropenem resistance in P. aeruginosa
(Okamoto et al., 2002). However, RNAseq results showed no
significant difference in the transcriptional levels of the mexAB-
oprM between the two strains, and this is confirmed by real-time
qPCR (Figures 1C,D). Further sequence analysis showed that no
mutation happened in the mexR,armR or mexRmexA intergenic
region of the IRP41 and ISP50 strains (data not shown).
IRP41 Encodes an Inactive OprD
To further elucidate the molecular mechanisms of the decreased
susceptibility to carbapenem in IRP41, genome reference
mapping was performed to identify the accumulated mutations
in the genomes of IRP41 and ISP50 in comparison with PAO1
reference genome (see text footnote 2). Between strains ISP50
and IRP41, there are 84 frameshifts (deletion/insertion) and 377
nonsynonymous single nucleotide variations (SNV) (including
17 early stops) (Supplementary Table S4). Among them, a
G831A substitution in the oprD gene resulted in a premature
termination of the OprD in the IRP41 strain. To confirm the
result, oprD genes were PCR amplified from the genomic DNA
of strains ISP50 and IRP41. Sequence analysis of the amplicons
revealed that oprD in ISP50 was the same as that of PAO1,
while the oprD from IRP41 showed a G831A substitution, which
confirmed the reference mapping results. To assess if the G831A
substitution in oprD contributed to the reduced susceptibility of
the IRP41 to carbapenems, the oprD gene from both ISP50 and
IRP41 were expressed in the IRP41 strain background. As can
be observed in Table 2, introduction of the oprDISP50 reduced
the MIC of imipenem/meropenem/biapenem in IRP41, with
16/2/2-fold decrease, while the oprDIRP41 did not. These results
indicated that the G831A substitution in oprD indeed disrupted
its function and lead to the reduced susceptibility to carbapenem
in strain IRP41.
Mechanisms of ampC Hyper-Expression
in IRP41
AmpR is a transcriptional regulator encoded immediately
upstream of the ampC, regulating the ampC expression (Kong
et al., 2005). The genome reference mapping result revealed that
the ampR sequence of IRP41 is identical to that of PAO1, while
in strain ISP50 an “A” was substituted by “G” at 821th position of
the ampR gene, resulting in an E274G substitution. These results
were further confirmed by sequencing of the PCR amplicons. To
test if the nonsynonymous mutation in ampR of ISP50 resulted in
a lower level of ampC expression, both ampR genes from IRP41
and ISP50 were cloned and expressed in the background of ISP50.
As shown in Supplementary Figure S3C and Supplementary
Table S3, expression of the ampRISP50 conferred ISP50 strain a
higher ampC mRNA level and increased MIC against ampicillin
compared to that of ampRIRP41, although it had no observable
influence on the MIC of imipenem/meropenem/biapenem.
The ampRISP50 and ampRIRP41 were further introduced
into PAO11ampR, and their effects on MICs to imipenem,
meropenem, biapenem and ampicillin in PAO11ampR were
examined. As shown in Supplementary Table S3, both
ampRISP50 and ampRIRP41 restored the MIC against ampicillin
and biapenem in PAO11ampR to that in wild type PAO1
strain, while in contrast to ampRIRP41, the ampRISP50 conferred
PAO11ampR a repeatable two-fold increase in MIC against
imipenem compared to that of wild type PAO1. Unexpectedly, no
influence on MIC of meropenem was observed in PAO11ampR.
These results suggested that A821G SNV of ampR in fact confers
slightly higher ampC expression-activating capacity than the
wild type AmpR, thus resulting in higher, rather than lower
ampC in ISP50.
ldcA (PA5198) encodes a cytosolic LD-carboxypeptidase
which removes the terminal amino acid D-alanine from the
tetra peptide of the peptidoglycan (Korza and Bochtler, 2005).
Our genome reference mapping and PCR amplicons sequencing
results revealed that a C was deleted at 445 bp of the ldcA gene
in strain IRP41, leading to a frameshift mutation after R149 of
the LdcA. To test if this frameshift mutation in ldcA contributed
to the elevated expression of the ampC in IRP41, we expressed
the ldcAIRP41 or ldcAISP50 (same as ldcA of PAO1) in the IRP41
background. As shown in Figure 2A, the relative mRNA level of
Frontiers in Microbiology | www.frontiersin.org 5June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 6
Xu et al. Mechanisms of Carbapenem Resistance
FIGURE 1 | Relative mRNA levels of indicated genes in indicated strains. (A) Relative mRNA levels of ampC in indicated strains. (B) Relative mRNA levels of pbpC in
indicated strains. (C) Relative mRNA levels of armR in ISP50 and IRP41. (D) Relative mRNA levels of mexB in ISP50 and IRP41. Total RNA was isolated from
indicated strains at OD600 of 1.0, and the relative mRNA levels of indicated genes were determined by real-time qPCR using rpsL as an internal control. ns, not
significant, ****P<0.0001, by Student’s t-test.
ampC in IRP41/pUCP24-ldcAISP50, but not in IRP41/pUCP24-
ldcAIRP41, was restored to that in ISP50/pUCP24, and the
MIC to imipenem/meropenem/biapenem in IRP41/pUCP24-
ldcAISP50 was decreased by two folds (Table 2). To confirm the
expression level of ampC gene, a C-terminal His-tagged ampC
driven by its native promoter was integrated into the genomes
of ISP50 and IRP41. As shown in Figure 2B, the AmpC-His level
was much higher in IRP41 than that in ISP50, and overexpression
of ldcAISP50, but not ldcAIRP41 , reduced the amount of AmpC-
His in the IRP41 to almost that of the ISP50. These results
demonstrated that frameshift-mutation of the ldcA in IRP41
contributes to the increased expression of the ampC in IRP41.
To further confirm the effect of ldcA on ampC repression,
the whole ldcA open reading frame was deleted in PA14 and
PAO1 backgrounds, resulting in PA141ldcA and PAO11ldcA,
respectively. Similar to previous report in E. coli (Templin et al.,
1999), both of the ldcA deletion mutants showed a reduced
growth rate and proneness to lysis during stationary growth
phase (Supplementary Figure S4). Interestingly, the expression
levels of ampC displayed 2.6-fold and 4.0-fold increase in
the 1ldcA mutants compared to their parental strains PAO1
and PA14, respectively (Figure 3A). Accordingly, the MIC of
ampicillin in PAO11ldcA and PA141ldcA displayed a two-fold
and four-fold increase compared to their respective parental
strains (Table 2). We further deleted the ldcA gene from the ISP50
strain. Consistent with the above observations, the expression
level of ampC in the ISP501ldcA increased 1.8-fold compared
to that in the ISP50 strain (Figure 3B). The difference in fold
increase of ampC mRNA levels between ISP50 and IRP41 (413
folds) vs. between ISP50 and ISP501ldcA (1.8 folds) suggested
that other factors also contributed to the increased expression of
the ampC in IRP41.
In addition, we also examined the effect of ldcA mutation on
MICs against piperacillin, ceftazidime, aztreonam, ciprofloxacin
and tobramycin. Except for a two-fold decrease against
ceftazidime and meropenem in PAO11ldcA, no MICs change
was observed for piperacillin, aztreonam, ciprofloxacin and
tobramycin in ldcA mutants (Table 4). However, we didn’t
obtain the IRP411ldcA, although deletion of ldcA in IRP41
strain had been tried for several times. We assume that it may
Frontiers in Microbiology | www.frontiersin.org 6June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 7
Xu et al. Mechanisms of Carbapenem Resistance
FIGURE 2 | Mechanism of increased mRNA levels of ampC in IRP41. (A) Relative mRNA levels of ampC gene in indicated strains. Total RNA was isolated from
indicated strains at OD600 of 1.0, and the relative mRNA levels of ampC were determined by real-time qPCR using rpsL as an internal control. ns, not significant,
***P<0.001, by Student’s t-test. (B) Levels of AmpC-His in indicated strains detected with Western blot assay. Strains with an ampC-His in their chromosomes
were cultured to OD600 of 1.0. Samples from equal number of bacterial cells were loaded onto 12% SDS-PAGE gels and probed with an antibody against His-tag or
RpoA. The RNA polymerase alpha subunit RpoA served as a control.
FIGURE 3 | Relative mRNA levels of ampC gene in indicated strains (A,B). Total RNA was isolated from indicated strains at OD600 of 1.0, and the relative mRNA
levels of ampC gene were determined by real-time qPCR using rpsL as an internal control. **P<0.01, ***P<0.001, ****P<0.0001, by student’s t-test.
be due to presence of some unknown factors to prevent the
recombination in IRP41.
Since AmpR is the regulator of ampC, and its 274th
amino acid is different between IRP41 and ISP50, we
further compared the ampC expression levels and the MIC
to ampicillin/imipenem/meropenem/biapenem between
ISP501ldcA expressing AmpRISP50 and AmpRIRP41. As
shown in Supplementary Figure S3D, consistent with that
in the ISP50 strain, expression of AmpRISP50 in ISP501ldcA
(ISP501ldcA/pMMB-ampRISP50) resulted in a higher mRNA
level of ampC compared to that of ISP501ldcA/pMMB-
ampRIRP41, as well as a two-fold increase in MIC of ampicillin
(Supplementary Table S3). Similar to that in ISP50 strain,
overexpression of both AmpRIRP41 and AmpRISP50 showed no
influence on MIC of imipenem/meropenem/biapenem in the
ISP501ldcA strain. Again, these are consistent with the earlier
observation that the AmpR point mutation in ISP50 confers a
higher transcriptional activator activity on ampC.
Frontiers in Microbiology | www.frontiersin.org 7June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 8
Xu et al. Mechanisms of Carbapenem Resistance
Furthermore, as the expression level of pbpC is much higher
in IRP41 than that in ISP50, we overexpressed pbpC in the
ISP501ldcA background to see if the elevated pbpC expression
had any effect on the ampC mRNA levels. However, the mRNA
level of ampC was not affected by the overexpression of pbpC
in ISP501ldcA (data not shown), consistent with such null effect
observed in the ISP50 background (Supplementary Table S3 and
Supplementary Figure S3E).
IRP41 Is a Better Biofilm Former Than
ISP50
Our RNA-seq result also revealed an increased expression
of pslD-pslL operon (Table 3) which is involved in biofilm
formation. Accordingly, we examined and compared the biofilm
formation between IRP41 and ISP50 strains. As the results
shown in Figure 4, the IRP41 strain produced more biofilm than
the ISP50 strain.
DISCUSSION
In this study, we examined MICs of imipenem, meropenem and
biapenem, three carbapenem antibiotics, for all the strains in
Table 2 and Supplementary Table S3. Compared to meropenem,
the contribution of AmpC overproduction and OprD deficiency
to biapenem susceptibility in IRP41 resembles to that of
imipenem. The difference among the susceptibility to imipenem,
meropenem and biapenem between ISP50 and IRP41 may reflect
their difference in penetration rate, stability to the hydrolysis
by β-lactamase, PBP protein binding profile (Yang et al.,
1995;Bonfiglio et al., 2002), β-lactamase deactivating capability
and export by the efflux pumps (Chen and Livermore, 1994;
Okamoto et al., 2002).
AmpC expression is regulated by AmpR, a LysR family
transcriptional regulator (Kong et al., 2005). Previous studies
reported that ampR gene point mutations were associated with
increased ampC expression in both Enterobacter cloacae and
P. aeruginosa clinical isolates (Kuga et al., 2000;Bagge et al.,
2002). Our study presents E274G substitution in AmpR actually
lead to a higher level of ampC expression. Besides the expression
of ampC, AmpR regulates expression of hundreds of genes
involved in diverse pathways such as physiological processes
and metabolism (Balasubramanian et al., 2015). It has been
demonstrated that ampR mutant displays an impaired growth
under iron-limiting conditions and sensitive to many agents
that affect cell growth (Balasubramanian et al., 2014, 2015).
Thus, it is possible that restoration to wild type AmpR from
E274G substituted AmpR of ISP50 provided the IRP41 strain
TABLE 4 | MICs (µg/ml) of indicated P. aeruginosa strains.
Strains TAZ (µg/ml) Cipro (µg/ml) Tob (µg/ml) Pip (µg/ml) AZT (µg/ml) Mem (µg/ml)
PAO1/pUCP24 3.125 0.3125 8 31.25 1.953125 3.125
PAO11ldcA/pUCP24 1.5625 0.3125 8 31.25 1.953125 1.5625
PAO11ldcA/pUCP24-ldcA 3.125 0.3125 8 31.25 1.953125 3.125
ISP50/pUCP24 3.125 0.625 16 62.5 7.8125 3.125
ISP501ldcA/pUCP24 3.125 0.625 16 62.5 7.8125 3.125
ISP501ldcA/pUCP24-ldcA 3.125 0.625 16 62.5 7.8125 3.125
TAZ, ceftazidime; Cipro, ciprofloxacin; Tob, tobramycin; Pip, piperacillin; AZT, aztreonam; Mem, meropenem.
FIGURE 4 | Biofilm formation by ISP50 and IRP41 strains. Overnight cultures of ISP50 and IRP41 were 50-fold diluted in LB and incubated in each well of a 96-well
plate for 24 h at 37C. Then the biofilm was stained with 0.1% crystal violet (A), dissolved by 0.2 ml of decolorizing solution and subjected to measurement at OD590
(B). ****P<0.0001 by Student’s t-test.
Frontiers in Microbiology | www.frontiersin.org 8June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 9
Xu et al. Mechanisms of Carbapenem Resistance
a better adaption to the host environment. Also, an increased
expression of the pbpC is neither responsible for the higher
ampC expression nor the decreased susceptibility to carbapenem
in IRP41 strain. The regulatory mechanism for the elevated
pbpC expression in IRP41 and its functional roles remain to
be investigated.
AmpD, a cytosolic peptidoglycan amidase, cleaves the peptide
chain from N-acetyl-anhydromuramic acid peptides which are
the inducer molecules for the ampC expression via binding to
the AmpR (Langaee et al., 1998;Lister et al., 2009;Dhar et al.,
2018). Mutations in AmpD and its two homologous proteins,
AmpDh2 and AmpDh3, contribute to a stepwise de-repression
of the ampC in the wild-type strain PAO1 (Juan et al., 2006).
Similarly, ampD mutations leading to ampC de-repression have
been reported in clinical isolates (Schmidtke and Hanson, 2008).
However, some AmpC over-expressing P. aeruginosa strains do
not exhibit mutation in ampD,ampDh2,ampDh3,ampR, or the
ampRampC intergenic region (Wolter et al., 2007;Schmidtke
and Hanson, 2008), suggesting that other undiscovered factors
or pathways likely contribute to the upregulation of ampC
expression. Besides, mutational inactivation of dacB, encoding
penicillin-binding protein 4 (PBP4), has been reported to
trigger a stable AmpC overproduction in P. aeruginosa
(Moya et al., 2009). Mutation of the lytic transglycosylases
encoding genes sltB1 and mltB1 in PAO1 resulted in a stable
AmpC hyperproduction in the presence of β-lactam antibiotic
(Cavallari et al., 2013;Juan et al., 2017). Inactivation of
mpl (encoding cytosolic UDP-N-acetylmuramate:L-alanyl-γ-D-
glutamyl-meso-diaminopimelate ligase) and nuoN (encoding
NADH dehydrogenase I chain N) led to an increase in the
expression of ampC (Tsutsumi et al., 2013;Juan et al., 2017). In
the two isolates used in this study, no mutation was found in
the above genes, except for the AmpR with E274G substitution
in ISP50 which had no effect on the increased ampC expression
in IRP41. These suggested the presence of unknown molecular
basis driving AmpC hyperproduction in the IRP41 strain.
In the present study, inactivation of the ldcA contributes to
the elevated expression of the ampC in P. aeruginosa. LdcA,
encoding an LD carboxypeptidase, cleaves the D-alanine from
N-acetyl-anhydromuramic acid tetrapeptides (anhNAM-P4) in
P. aeruginosa (Korza and Bochtler, 2005), generating N-acetyl-
anhydromuramic acid tripeptide (anhNAM-P3). Functional loss
of the LdcA leads to the accumulation of anhNAM-P4, thus our
observation suggests that the anhNAM-P3 is less potent inducer
for the ampC expression than the anhNAM-P4. This is consistent
with a previous study in which the anhNAM-P4 was shown
to be a critical activator ligand for β-lactamase expression in
Stenotrophomonas maltophilia (Huang et al., 2017). Results from
previous studies implied anhNAM-P5 as the genuine AmpR-
binding signal (Dietz et al., 1997;Lee et al., 2016) and further
suggested that only muropeptides containing a terminal d-Ala-
d-Ala motif (i.e., muropentapeptides) is capable of binding to
the AmpR for ampC induction (Vadlamani et al., 2015;Dik
et al., 2017). However, in a most recent report, anhNAM-
P3 was demonstrated to function as an ampC inducer, albeit
much less potent than the anhNAM-P5 (Torrens et al., 2019).
As these studies were carried out under different conditions
(with or without antibiotic induction) using different mutant
backgrounds, it is possible that different signaling pathways may
have been involved in the AmpR mediated regulation of the
ampC in the clinical strains analyzed in this study.
This is the first report linking ldcA to the regulation of ampC
expression in P. aeruginosa. Interestingly, compared to the 400-
fold increase of ampC mRNA in IRP41, the 1ldcA mutants
of PA14, PAO1 and ISP50 showed only 1.8–4 folds increases
in the ampC mRNA compared to their parental strains. Such
minor changes in the ampC expression, in addition to the growth
defect of the ldcA mutant at stationary phase, may explain why
LdcA was not identified as a repressor for the ampC in previous
studies. One may argue that the observed 2–4 folds increase in
MIC to ampicillin may be due to the reduced growth of the
1ldcA mutant. However, we do not feel this is the case because
the proneness to lysis happens during the stationary growth
phase (OD600 about 2.0). In the case of IRP41, we postulate
that there are other factors contributing to the increased ampC
expression in addition to the ldcA. The mechanism underlying
this observation is yet to be elucidated.
OprD has been reported to be the most prevalent cause of
imipenem resistance among clinical isolates of P. aeruginosa.
Insertion of various IS elements and point mutations with
premature stops on the oprD gene had been associated
with the carbapenem resistance among clinical isolates of
P. aeruginosa (Hirabayashi et al., 2017). In this study, a G831A
substitution in oprD resulted in loss of the OprD function.
With the OprD inactivation and elevated ampC expression,
the clinical isolate IRP41 displayed resistance to carbapenem.
It has been demonstrated that derepression of AmpC in
PAO1 has no obvious impact on the MIC of imipenem (Juan
et al., 2006). While in another study, the loss of AmpC
from PAO1 displayed a four-fold increase in susceptibility to
imipenem (Masuda et al., 1999). Derepression of AmpC was
associated with 4–8/8–64-fold increase in MIC of imipenem in
an OprD+/OprDbackground, respectively (Livermore, 1992;
Mushtaq et al., 2004). And also, mutational variants of the
AmpC cephalosporinase provide P. aeruginosa with imipenem
resistance (Rodriguez-Martinez et al., 2009). In our study,
overexpression of ampC conferred ISP50 a 16-fold increase in
MIC of imipenem. However, both AmpC and OprD are of PAO1-
type in ISP50, which suggests that other unknown factors may
contribute to the change in susceptibility to imipenem by the
overexpression of AmpC.
MexAB-OprM is able to expel a wide variety of antibiotics
(Kohler et al., 1999;Lister et al., 2009) and its expression is
repressed by MexR (Poole et al., 1996). A 53-amino-acid long
antirepressor, ArmR, could interact with and inhibit the MexR
function, upregulating the mexAB-oprM expression (Wilke et al.,
2008). However, in the present study, increased expression of
armR in IRP41 did not cause altered expression level of mexAB-
oprM. In fact, mexAB-oprM is expressed and contributes to
antimicrobial resistance in wild type Pseudomonas aeruginosa,
it is more difficult to detect mexAB-oprM overexpression in
mutant cells than the other efflux pump encoding genes (e.g.,
mexCD-oprJ,mexEF-oprN). In addition, previous study has
revealed MexR is a redox regulator which senses oxidative
Frontiers in Microbiology | www.frontiersin.org 9June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 10
Xu et al. Mechanisms of Carbapenem Resistance
stress inside bacterial cells to regulate mexAB-oprM expression
(Chen et al., 2008). nalD encodes a second repressor of the
mexAB-oprM (Morita et al., 2006). Mutations in nalD resulted in
increased MexAB-OprM expression in lab and clinical isolates of
P. aeruginosa (Sobel et al., 2005). However, no mutation occurred
in the nalD gene of the IRP41 or ISP50 isolates. The mechanisms
underlying these observations are under active investigation.
It has been demonstrated that nalC (PA3721), encoding a
TetR family repressor, negatively regulated armR, and S127P
substitution in NalC impaired its armR-repressing capacity (Cao
et al., 2004). Compared to ISP50, no mutations were observed
in the intergenic region between nalC and PA3720, while a
S127P substitution in NalC happened in IRP41 (Supplementary
Table S4), which may be the cause to result in the increased armR
transcriptional level in IRP41.
IRP41 is a better biofilm former than ISP50. However, the
MICs against ciprofloxacin and levofloxacin in IRP41 are the
same as ISP50 (data not shown), and the MICs against gentamicin
and tobramycin display a two-fold decrease in IRP41 than that
in ISP50 (data not shown). A previous study had shown that the
Psl did not affect bacterial MIC to biapenem for planktonic cells
(Murakami et al., 2017). Therefore, we assumed that the increased
biofilm production did not contribute to the increased MIC to
carbapenem in IRP41.
It is not necessary for clinical isolates processing a major
resistance determinant against a certain antibiotic category
to accumulate the other resistance mechanisms. However,
the concomitant presence of multiple carbapenem resistance
mechanisms has been observed in P. aeruginosa (Meletis et al.,
2014). In our study, the overproduction of AmpC β-lactamase
together with OprD deficiency leads imipenem-susceptible ISP50
to imipenem-resistant IRP41 derivative, as well as a reduced
susceptibility to meropenem and biapenem.
DATA AVAILABILITY STATEMENT
The datasets generated for this study can be found in NCBI, under
accession PRJNA635437.
ETHICS STATEMENT
We have a waiver from the ethics committee, exempting this
study from the requirement to have ethics approval and written
informed as the clinical strains used in the study come from
the routine procedures of the clinical laboratory rather than the
clinical trials.
AUTHOR CONTRIBUTIONS
YJ and FB conceived and designed the experiments. CX, DW,
XZ, HL, GZ, and TW performed the experiments. WW, FB, ZC,
and YJ analyzed the data. YJ wrote the manuscript. All authors
contributed to the article and approved the submitted version.
FUNDING
This work was supported by the National Science Foundation
of China (31600110, 31870130, 31670130, 81670766, and
31970680), Science and Technology Committee of Tianjin
(17JCQNJC09200 and 19JCYBJC24700), National Key Research
and Development Project of China (2017YFE0125600), Science
and Technology Program of Sichuan Province (2018JZ0069), and
the Fundamental Research Funds for the Central Universities,
Nankai University (63191121, 63191117, and 63191521).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: https://www.frontiersin.org/articles/10.3389/fmicb.
2020.01390/full#supplementary-material
FIGURE S1 | PCR results of ISP50 and IRP41 strains. (A) 16S rDNA gene
amplification; (B) RAPD typing.
FIGURE S2 | Carbapenem susceptibility and carbapenemase production of
indicated strains. (A,B) Imipenem inhibition zones of indicated strains on a 6-cm
disk. (C,D) Meropenem inhibition zones of indicated strains on a 6-cm disk. (E,F)
PAE-MHT assay using K. pneumoniae ATCC 700603 as indicator.
Carbapenemase production test on imipenem (E) and meropenem (F) on a 10-cm
disk, 0: ISP50, 1: IRP41, 2: PA-NK41, a producer of KPC2 carbapenemase.
FIGURE S3 | Relative mRNA levels of indicated genes in indicated strains.
(A,C–E) Relative mRNA levels of ampC in indicated strains. (B) Relative mRNA
levels of pbpC in indicated strains. Total RNA was isolated from indicated strains
at OD600 of 1.0, and the relative mRNA levels of ampC or pbpC gene were
determined by real-time qPCR using rpsL as an internal control. ns, not significant,
P<0.05, ∗∗ P<0.01, ∗ ∗ ∗ P<0.001, ∗ ∗ ∗ ∗ P<0.0001, by Student’s t-test.
FIGURE S4 | Growth curves of indicated strains.
TABLE S1 | Primers used in this study.
TABLE S2 | Transcriptome analysis: differentially expressed genes by RNAseq
without those listed in Table 2.
TABLE S3 | MICs (µg/ml) of indicated P. aeruginosa strains.
TABLE S4 | SNVs identified by genome reference mapping with elimination of
SNVs with synonymous mutation and the same mutation between
ISP50 and IRP41.
REFERENCES
Bagge, N., Ciofu, O., Hentzer, M., Campbell, J. I., Givskov, M., and Hoiby,
N. (2002). Constitutive high expression of chromosomal beta-lactamase in
Pseudomonas aeruginosa caused by a new insertion sequence (IS1669) located
in ampD. Antimicrob. Agents Chemother. 46, 3406–3411. doi: 10.1128/aac.46.
11.3406-3411.2002
Balasubramanian, D., Kumari, H., Jaric, M., Fernandez, M., Turner, K. H., Dove,
S. L., et al. (2014). Deep sequencing analyses expands the Pseudomonas
aeruginosa AmpR regulon to include small RNA-mediated regulation of iron
acquisition, heat shock and oxidative stress response. Nucleic Acids Res. 42,
979–998. doi: 10.1093/nar/gkt942
Balasubramanian, D., Kumari, H., and Mathee, K. (2015). Pseudomonas aeruginosa
AmpR: an acute-chronic switch regulator. Pathog. Dis. 73, 1–14. doi: 10.1111/
2049-632x.12208
Bonfiglio, G., Russo, G., and Nicoletti, G. (2002). Recent developments in
carbapenems. Expert Opin. Investig. Drugs 11, 529–544. doi: 10.1517/13543784.
11.4.529
Frontiers in Microbiology | www.frontiersin.org 10 June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 11
Xu et al. Mechanisms of Carbapenem Resistance
Cao, L., Srikumar, R., and Poole, K. (2004). MexAB-OprM hyperexpression
in NalC-type multidrug-resistant Pseudomonas aeruginosa: identification and
characterization of the nalC gene encoding a repressor of PA3720-PA3719. Mol.
Microbiol. 53, 1423–1436. doi: 10.1111/j.1365-2958.2004.04210.x
Cavallari, J. F., Lamers, R. P., Scheurwater, E. M., Matos, A. L., and Burrows,
L. L. (2013). Changes to its peptidoglycan-remodeling enzyme repertoire
modulate beta-lactam resistance in Pseudomonas aeruginosa.Antimicrob.
Agents Chemother. 57, 3078–3084. doi: 10.1128/aac.00268-13
Chen, H., Hu, J., Chen, P. R., Lan, L., Li, Z., Hicks, L. M., et al. (2008). The
Pseudomonas aeruginosa multidrug efflux regulator MexR uses an oxidation-
sensing mechanism. Proc. Natl. Acad. Sci. U.S.A. 105, 13586–13591. doi: 10.
1073/pnas.0803391105
Chen, H. Y., and Livermore, D. M. (1994). In-vitro activity of biapenem, compared
with imipenem and meropenem, against Pseudomonas aeruginosa strains and
mutants with known resistance mechanisms. J. Antimicrob. Chemother. 33,
949–958. doi: 10.1093/jac/33.5.949
Choudhury, D., Das Talukdar, A., Dutta Choudhury, M., Maurya, A. P., Paul,
D., Dhar Chanda, D., et al. (2015). Transcriptional analysis of MexAB-OprM
efflux pumps system of Pseudomonas aeruginosa and its role in carbapenem
resistance in a tertiary referral hospital in India. PLoS One 10:e0133842. doi:
10.1371/journal.pone.0133842
Curran, B., Jonas, D., Grundmann, H., Pitt, T., and Dowson, C. G. (2004).
Development of a multilocus sequence typing scheme for the opportunistic
pathogen Pseudomonas aeruginosa.J. Clin. Microbiol. 42, 5644–5649. doi: 10.
1128/jcm.42.12.5644-5649.2004
Daigle, D. M., Cao, L., Fraud, S., Wilke, M. S., Pacey, A., Klinoski, R., et al.
(2007). Protein modulator of multidrug efflux gene expression in Pseudomonas
aeruginosa.J. Bacteriol. 189, 5441–5451. doi: 10.1128/jb.00543-07
Davies, T. A., Marie Queenan, A., Morrow, B. J., Shang, W., Amsler, K., He,
W., et al. (2011). Longitudinal survey of carbapenem resistance and resistance
mechanisms in Enterobacteriaceae and non-fermenters from the USA in 2007-
09. J. Antimicrob. Chemother. 66, 2298–2307. doi: 10.1093/jac/dkr290
Dhar, S., Kumari, H., Balasubramanian, D., and Mathee, K. (2018). Cell-wall
recycling and synthesis in Escherichia coli and Pseudomonas aeruginosa - their
role in the development of resistance. J. Med. Microbiol. 67, 1–21. doi: 10.1099/
jmm.0.000636
Dietz, H., Pfeifle, D., and Wiedemann, B. (1997). The signal molecule for
beta-lactamase induction in Enterobacter cloacae is the anhydromuramyl-
pentapeptide. Antimicrob. Agents Chemother. 41, 2113–2120. doi: 10.1128/aac.
41.10.2113
Dik, D. A., Dominguez-Gil, T., Lee, M., Hesek, D., Byun, B., Fishovitz, J., et al.
(2017). Muropeptide binding and the X-ray structure of the effector domain of
the transcriptional regulator AmpR of Pseudomonas aeruginosa.J. Am. Chem.
Soc. 139, 1448–1451. doi: 10.1021/jacs.6b12819
Hang, Y., Chen, Y., Xue, L., Sun, S., Liu, L., Gao, J., et al. (2018). Evaluating
biapenem dosage regimens in intensive care unit patients with Pseudomonas
aeruginosa infections: a pharmacokinetic/pharmacodynamic analysis using
Monte Carlo simulation. Int. J. Antimicrob. Agents 51, 484–487. doi: 10.1016/
j.ijantimicag.2017.07.005
Hirabayashi, A., Kato, D., Tomita, Y., Iguchi, M., Yamada, K., Kouyama, Y.,
et al. (2017). Risk factors for and role of OprD protein in increasing minimal
inhibitory concentrations of carbapenems in clinical isolates of Pseudomonas
aeruginosa.J. Med. Microbiol. 66, 1562–1572. doi: 10.1099/jmm.0.000601
Hoang, T. T., Karkhoff-Schweizer, R. R., Kutchma, A. J., and Schweizer, H. P.
(1998). A broad-host-range Flp-FRT recombination system for site-specific
excision of chromosomally-located DNA sequences: application for isolation
of unmarked Pseudomonas aeruginosa mutants. Gene 212, 77–86. doi: 10.1016/
s0378-1119(98)00130- 9
Hoban, D. J., Jones, R. N., Yamane, N., Frei, R., Trilla, A., and Pignatari, A. C.
(1993). In vitro activity of three carbapenem antibiotics. Comparative studies
with biapenem (L-627), imipenem, and meropenem against aerobic pathogens
isolated worldwide. Diagn. Microbiol. Infect. Dis. 17, 299–305. doi: 10.1016/
0732-8893(93)90039- a
Huang, Y. W., Wang, Y., Lin, Y., Lin, C., Lin, Y. T., Hsu, C. C., et al. (2017). Impacts
of penicillin binding protein 2 Inactivation on beta-Lactamase expression and
muropeptide profile in Stenotrophomonas maltophilia. mSystems 2:17. doi: 10.
1128/mSystems.00077-17
Juan, C., Moya, B., Perez, J. L., and Oliver, A. (2006). Stepwise upregulation of the
Pseudomonas aeruginosa chromosomal cephalosporinase conferring high-level
beta-lactam resistance involves three AmpD homologues. Antimicrob. Agents
Chemother. 50, 1780–1787. doi: 10.1128/aac.50.5.1780-1787.2006
Juan, C., Torrens, G., Gonzalez-Nicolau, M., and Oliver, A. (2017). Diversity
and regulation of intrinsic beta-lactamases from non-fermenting and other
Gram-negative opportunistic pathogens. FEMS Microbiol. Rev. 41, 781–815.
doi: 10.1093/femsre/fux043
Kaufman, M. R., Jia, J., Zeng, L., Ha, U., Chow, M., and Jin, S. (2000). Pseudomonas
aeruginosa mediated apoptosis requires the ADP-ribosylating activity of
exoS. Microbiology 146(Pt 10), 2531–2541. doi: 10.1099/00221287-146-10-
2531
Kohler, T., Michea-Hamzehpour, M., Epp, S. F., and Pechere, J. C. (1999).
Carbapenem activities against Pseudomonas aeruginosa: respective
contributions of OprD and efflux systems. Antimicrob. Agents Chemother.
43, 424–427. doi: 10.1128/aac.43.2.424
Kong, K. F., Jayawardena, S. R., Indulkar, S. D., Del Puerto, A., Koh, C. L., Hoiby,
N., et al. (2005). Pseudomonas aeruginosa AmpR is a global transcriptional
factor that regulates expression of AmpC and PoxB beta-lactamases, proteases,
quorum sensing, and other virulence factors. Antimicrob. Agents Chemother. 49,
4567–4575. doi: 10.1128/aac.49.11.4567-4575.2005
Korza, H. J., and Bochtler, M. (2005). Pseudomonas aeruginosa LD-
carboxypeptidase, a serine peptidase with a Ser-His-Glu triad and a nucleophilic
elbow. J. Biol. Chem. 280, 40802–40812. doi: 10.1074/jbc.M506328200
Kuga, A., Okamoto, R., and Inoue, M. (2000). ampR gene mutations that greatly
increase class C beta-lactamase activity in Enterobacter cloacae.Antimicrob.
Agents Chemother. 44, 561–567. doi: 10.1128/aac.44.3.561-567.2000
Langaee, T. Y., Dargis, M., and Huletsky, A. (1998). An ampD gene in Pseudomonas
aeruginosa encodes a negative regulator of AmpC beta-lactamase expression.
Antimicrob. Agents Chemother. 42, 3296–3300. doi: 10.1128/aac.42.12.3296
Lee, M., Dhar, S., De Benedetti, S., Hesek, D., Boggess, B., Blazquez, B., et al.
(2016). Muropeptides in Pseudomonas aeruginosa and their role as elicitors of
beta-lactam-antibiotic resistance. Angew. Chem. Int. Ed. Engl. 55, 6882–6886.
doi: 10.1002/anie.201601693
Li, K., Xu, C., Jin, Y., Sun, Z., Liu, C., Shi, J., et al. (2013). SuhB is a regulator
of multiple virulence genes and essential for pathogenesis of Pseudomonas
aeruginosa.mBio 4:e419-13. doi: 10.1128/mBio.00419-13
Liberati, N. T., Urbach, J. M., Miyata, S., Lee, D. G., Drenkard, E., Wu, G.,
et al. (2006). An ordered, nonredundant library of Pseudomonas aeruginosa
strain PA14 transposon insertion mutants. Proc. Natl. Acad. Sci. U.S.A. 103,
2833–2838. doi: 10.1073/pnas.0511100103
Lister, P. D., Wolter, D. J., and Hanson, N. D. (2009). Antibacterial-
resistant Pseudomonas aeruginosa: clinical impact and complex regulation
of chromosomally encoded resistance mechanisms. Clin. Microbiol. Rev. 22,
582–610. doi: 10.1128/cmr.00040-09
Liu, Y., Li, X. Y., Wan, L. G., Jiang, W. Y., Li, F. Q., and Yang, J. H. (2013). Efflux
system overexpression and decreased OprD contribute to the carbapenem
resistance among extended-spectrum beta-lactamase-producing Pseudomonas
aeruginosa isolates from a Chinese university hospital. Microb. Drug Resist. 19,
463–468. doi: 10.1089/mdr.2013.0010
Livermore, D. M. (1992). Interplay of impermeability and chromosomal beta-
lactamase activity in imipenem-resistant Pseudomonas aeruginosa.Antimicrob.
Agents Chemother. 36, 2046–2048. doi: 10.1128/aac.36.9.2046
Long, Y., Fu, W., Li, S., Ren, H., Li, M., Liu, C., et al. (2019). Identification of novel
genes that promote persister formation by repressing transcription and cell
division in Pseudomonas aeruginosa.J. Antimicrob. Chemother. 74, 2575–2587.
doi: 10.1093/jac/dkz214
Mahenthiralingam, E., Campbell, M. E., Foster, J., Lam, J. S., and Speert,
D. P. (1996). Random amplified polymorphic DNA typing of Pseudomonas
aeruginosa isolates recovered from patients with cystic fibrosis. J. Clin.
Microbiol. 34, 1129–1135. doi: 10.1128/jcm.34.5.1129-1135.1996
Masuda, N., Gotoh, N., Ishii, C., Sakagawa, E., Ohya, S., and Nishino, T. (1999).
Interplay between chromosomal beta-lactamase and the MexAB-OprM efflux
system in intrinsic resistance to beta-lactams in Pseudomonas aeruginosa.
Antimicrob. Agents Chemother. 43, 400–402. doi: 10.1128/aac.43.2.400
Mcfarland, J. (1907). The nephelometer: an instrument for estimating the
number of bacteria in suspensions used for calculating the opsonic index
and for vaccines. J. Am. Med. Assoc. 49, 1176–1178. doi: 10.1001/jama.1907.
25320140022001f
Meletis, G., Vavatsi, N., Exindari, M., Protonotariou, E., Sianou, E., Haitoglou,
C., et al. (2014). Accumulation of carbapenem resistance mechanisms in
Frontiers in Microbiology | www.frontiersin.org 11 June 2020 | Volume 11 | Article 1390
fmicb-11-01390 June 18, 2020 Time: 17:19 # 12
Xu et al. Mechanisms of Carbapenem Resistance
VIM-2-producing Pseudomonas aeruginosa under selective pressure. Eur. J.
Clin. Microbiol. Infect. Dis. 33, 253–258. doi: 10.1007/s10096-013-1952- 3
Mirsalehian, A., Kalantar-Neyestanaki, D., Nourijelyani, K., Asadollahi, K.,
Taherikalani, M., Emaneini, M., et al. (2014). Detection of AmpC-beta-
lactamases producing isolates among carbapenem resistant P. aeruginosa
isolated from burn patient. Iran J. Microbiol. 6, 306–310.
Morita, Y., Cao, L., Gould, V. C., Avison, M. B., and Poole, K. (2006). nalD
encodes a second repressor of the mexAB-oprM multidrug efflux operon
of Pseudomonas aeruginosa.J. Bacteriol. 188, 8649–8654. doi: 10.1128/jb.013
42-06
Moya, B., Dotsch, A., Juan, C., Blazquez, J., Zamorano, L., Haussler, S., et al. (2009).
Beta-lactam resistance response triggered by inactivation of a nonessential
penicillin-binding protein. PLoS Pathog. 5:e1000353. doi: 10.1371/journal.ppat.
1000353
Murakami, K., Ono, T., Viducic, D., Somiya, Y., Kariyama, R., Hori, K., et al. (2017).
Role of psl Genes in Antibiotic Tolerance of Adherent Pseudomonas aeruginosa.
Antimicrob. Agents Chemother. 61:e002587-16. doi: 10.1128/aac.02587-16
Mushtaq, S., Ge, Y., and Livermore, D. M. (2004). Doripenem versus
Pseudomonas aeruginosa in vitro: activity against characterized isolates,
mutants, and transconjugants and resistance selection potential. Antimicrob.
Agents Chemother. 48, 3086–3092. doi: 10.1128/aac.48.8.3086-3092.2004
Okamoto, K., Gotoh, N., and Nishino, T. (2002). Alterations of susceptibility
of Pseudomonas aeruginosa by overproduction of multidrug efflux systems,
MexAB-OprM, MexCD-OprJ, and MexXY/OprM to carbapenems: substrate
specificities of the efflux systems. J. Infect. Chemother. 8, 371–373. doi: 10.1007/
s10156-002- 0193-7
Pasteran, F., Veliz, O., Rapoport, M., Guerriero, L., and Corso, A. (2011). Sensitive
and specific modified Hodge test for KPC and metallo-beta- lactamase detection
in Pseudomonas aeruginosa by use of a novel indicator strain, Klebsiella
pneumoniae ATCC 700603. J. Clin. Microbiol. 49, 4301–4303. doi: 10.1128/jcm.
05602-11
Poole, K. (2011). Pseudomonas aeruginosa: resistance to the max. Front. Microbiol.
2:65. doi: 10.3389/fmicb.2011.00065
Poole, K., Tetro, K., Zhao, Q., Neshat, S., Heinrichs, D. E., and Bianco, N.
(1996). Expression of the multidrug resistance operon mexA-mexB-oprM in
Pseudomonas aeruginosa: mexR encodes a regulator of operon expression.
Antimicrob. Agents Chemother. 40, 2021–2028. doi: 10.1128/aac.40.9.
2021
Potron, A., Poirel, L., and Nordmann, P. (2015). Emerging broad-spectrum
resistance in Pseudomonas aeruginosa and Acinetobacter baumannii:
mechanisms and epidemiology. Int. J. Antimicrob. Agents 45, 568–585.
doi: 10.1016/j.ijantimicag.2015.03.001
Rodriguez-Martinez, J. M., Poirel, L., and Nordmann, P. (2009). Extended-
spectrum cephalosporinases in Pseudomonas aeruginosa.Antimicrob. Agents
Chemother. 53, 1766–1771. doi: 10.1128/aac.01410-08
Schmidtke, A. J., and Hanson, N. D. (2008). Role of ampD homologs in
overproduction of AmpC in clinical isolates of Pseudomonas aeruginosa.
Antimicrob. Agents Chemother. 52, 3922–3927. doi: 10.1128/aac.00341-08
Schweizer, H. P. (1992). Allelic exchange in Pseudomonas aeruginosa using novel
ColE1-type vectors and a family of cassettes containing a portable oriT and the
counter-selectable Bacillus subtilis sacB marker. Mol. Microbiol. 6, 1195–1204.
doi: 10.1111/j.1365-2958.1992.tb01558.x
Sobel, M. L., Hocquet, D., Cao, L., Plesiat, P., and Poole, K. (2005). Mutations
in PA3574 (nalD) lead to increased MexAB-OprM expression and multidrug
resistance in laboratory and clinical isolates of Pseudomonas aeruginosa.
Antimicrob. Agents Chemother. 49, 1782–1786. doi: 10.1128/aac.49.5.1782-
1786.2005
Spilker, T., Coenye, T., Vandamme, P., and LiPuma, J. J. (2004). PCR-based assay
for differentiation of Pseudomonas aeruginosa from other Pseudomonas species
recovered from cystic fibrosis patients. J. Clin. Microbiol. 42, 2074–2079. doi:
10.1128/jcm.42.5.2074-2079.2004
Templin, M. F., Ursinus, A., and Holtje, J. V. (1999). A defect in cell wall recycling
triggers autolysis during the stationary growth phase of Escherichia coli.EMBO
J. 18, 4108–4117. doi: 10.1093/emboj/18.15.4108
Torrens, G., Hernandez, S. B., Ayala, J. A., Moya, B., Juan, C., Cava, F., et al.
(2019). Regulation of AmpC-Driven beta-Lactam resistance in Pseudomonas
aeruginosa: different pathways, different signaling. mSystems 4:e00524-19. doi:
10.1128/mSystems.00524-19
Tsutsumi, Y., Tomita, H., and Tanimoto, K. (2013). Identification of novel genes
responsible for overexpression of ampC in Pseudomonas aeruginosa PAO1.
Antimicrob. Agents Chemother. 57, 5987–5993. doi: 10.1128/aac.01291-13
Vadlamani, G., Thomas, M. D., Patel, T. R., Donald, L. J., Reeve, T. M., Stetefeld,
J., et al. (2015). The beta-lactamase gene regulator AmpR is a tetramer
that recognizes and binds the D-Ala-D-Ala motif of its repressor UDP-N-
acetylmuramic acid (MurNAc)-pentapeptide. J. Biol. Chem. 290, 2630–2643.
doi: 10.1074/jbc.M114.618199
Vincent, J. L., Bihari, D. J., Suter, P. M., Bruining, H. A., White, J., Nicolas-Chanoin,
M. H., et al. (1995). The prevalence of nosocomial infection in intensive care
units in Europe. results of the european prevalence of infection in intensive
care (EPIC) study. EPIC international advisory committee. JAMA 274, 639–644.
doi: 10.1001/jama.274.8.639
West, S. E., Schweizer, H. P., Dall, C., Sample, A. K., and Runyen-Janecky, L. J.
(1994). Construction of improved Escherichia-Pseudomonas shuttle vectors
derived from pUC18/19 and sequence of the region required for their
replication in Pseudomonas aeruginosa.Gene 148, 81–86. doi: 10.1016/0378-
1119(94)90237-2
Wilke, M. S., Heller, M., Creagh, A. L., Haynes, C. A., McIntosh, L. P., Poole,
K., et al. (2008). The crystal structure of MexR from Pseudomonas aeruginosa
in complex with its antirepressor ArmR. Proc. Natl. Acad. Sci. U.S.A. 105,
14832–14837. doi: 10.1073/pnas.0805489105
Wolter, D. J., Schmidtke, A. J., Hanson, N. D., and Lister, P. D. (2007).
Increased expression of ampC in Pseudomonas aeruginosa mutants selected
with ciprofloxacin. Antimicrob. Agents Chemother. 51, 2997–3000. doi: 10.1128/
aac.00111-07
Yang, Y., Bhachech, N., and Bush, K. (1995). Biochemical comparison of
imipenem, meropenem and biapenem: permeability, binding to penicillin-
binding proteins, and stability to hydrolysis by beta-lactamases. J. Antimicrob.
Chemother. 35, 75–84. doi: 10.1093/jac/35.1.75
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2020 Xu, Wang, Zhang, Liu, Zhu, Wang, Cheng, Wu, Bai and Jin.
This is an open-access article distributed under the terms of the Creative Commons
Attribution License (CC BY). The use, distribution or reproduction in other forums
is permitted, provided the original author(s) and the copyright owner(s) are credited
and that the original publication in this journal is cited, in accordance with accepted
academic practice. No use, distribution or reproduction is permitted which does not
comply with these terms.
Frontiers in Microbiology | www.frontiersin.org 12 June 2020 | Volume 11 | Article 1390
... However, the development of resistance to β-lactams limits their effectiveness. Several important resistance mechanisms have been reported including: (i) overproduction of the chromosomally encoded β-lactamase (AmpC) [4]; (ii) loss/reduced production of the outer membrane porin OprD [5,6]; (iii) expression of multidrug efflux pumps, such as MexAB-OprM, MexCD-OprJ, and MexXY [4,6]; and (iv) acquisition of the extended-spectrum β-lactamase via horizontal gene transfer [7]. Of note, these resistance mechanisms have been found to contribute to the antibiotic resistance of P. aeruginosa in the clinical setting. ...
... Of note, these resistance mechanisms have been found to contribute to the antibiotic resistance of P. aeruginosa in the clinical setting. Importantly, via characterization of clinical isolates from a single patient, evolutionary trajectories and molecular mechanisms for antibiotic resistance have been well established in P. aeruginosa [5,[8][9][10]. However, ...
... Genomic DNAs of R1 and S2 were extracted from overnight cultures of bacterial cells using DNA purification kit (Tiangen Biotec, Beijing, China). Genomic DNA reference mapping was carried out by GENEWIZ Life Sciences (Suzhou, China) as described in our previous studies [5,16]. The raw sequence data have been deposited in the NCBI with an accessible number PRJNA863030 (www.ncbi.nlm.nih.gov/sra/?term=PRJNA863030). ...
Article
Full-text available
Pseudomonas aeruginosa is an important nosocomial pathogen which frequently becomes resistant to most antibiotics used in chemotherapy, resulting in treatment failure among infected individuals. Although the evolutionary trajectory and molecular mechanisms for becoming β-lactam resistant have been well established for P. aeruginosa, the molecular basis of reversion from β-lactam resistant to susceptible is largely unexplored. In this study, we investigated the molecular mechanisms by which a ceftazidime-resistant clinical strain is converted to a ceftazidime-susceptible isolate under the clinical setting. RNA sequencing and genomic DNA reference mapping were conducted to compare the transcriptional profiles and chromosomal mutations between these two isolates. Our results demonstrate that a gain-of-function mutation in ampD, via deletion of a 53 bp duplicated nucleotide sequence, is the contributory factor for the conversion. Furthermore, we show for the first time that AmpD is involved in intraspecies competitiveness in P. aeruginosa. We also found that AmpD is not responsible for phenotypic changes between R1 and S2, including growth rate, motilities, pyocyanin, rhamnolipid, and biofilm production. This finding provides novel insights into the alteration of β-lactam sensitivity in P. aeruginosa under the clinical setting.
... Several mechanism including AmpC overproduction, the production of carbapenemases, the overactivity of e ux pumps, and the loss or downregulation of outer membrane porins such as OprD are involved to carbapenem resistance. On the other hand, according to some reports carbapenem resistance Gramnegative bacteria are usually resistance to many classes of antibiotic agents such as uoroquinolones and aminoglycosides [24]. During to past decade one of the main mechanisms of resistance among gram-negative bacilli which could be couple with carbapenemase is novel plasmid-mediated aminoglycoside resistance mechanism caused by 16S rRNA methylase [25]. ...
Preprint
Full-text available
Background: Carbapenem-resistant Pseudomonas aeruginosa (CRPA) is the one of the main Gram-negative bacterium cause of infections in hospital settings and the spread of them is significant challenge to public health. Methods: In this study, a total of 30 non-duplicate isolates of CRPA were collected. Antibacterial susceptibility of bacteria to antibiotic agents and AmpC overproducer isolates were determined. Minimal biofilm inhibitory concentration (MBIC) of isolates to cefepime (FEP), imipenem (IPM), ceftazidime (CAZ), and meropenem (MEM) were evaluated with/without cloxacillin (CLX). The carbapenemase and 16S rRNA methylase genes were identified by PCR and the transcription levels of oprD, ampC, and mexA genes were determined by quantitative real-time PCR. ERIC-PCR was used to detect genetic relationships among the isolates. Results: All isolates were resistance to IPM, MEM, CAZ, FEP, CIP, GEN, TOB and strong biofilm producer. The resistance genes including blaNDM, blaIMP, blaVIM, blaSIM, blaGES, and armA were detected in 21 (70%), 6(20%), 3 (10%), 2 (6.6%), 1 (3.3%), and 56.6% of the isolates, respectively. CLX at 250 and 500 µg/mL significantly reduced the level of MIC to MEM, IPM, CAZ, and FEP and at 2000 µg/mL significantly reduced the level of MBIC to MEM, IPM, CAZ, and FEP. In all of isolates the transcription levels of oprD were significantly downregulated as well as were showed significantly increasing for ampC and mexA. ERIC-PCR typing results divided 30 isolates into four clusters. Conclusion: In this study we reported the spread of different clone of CRPA harboring co-existence various carbapenemase genes with armA 16S rRNA methylase for the first time in Kerman, Iran. Also, our isolates had a combination of resistance mechanisms to carbapenems as well as biofilm formation along with resistance to aminoglycosides, the further spread of which could cause serious challenge in hospital settings. Therefore, serious monitoring is necessary to reduce their prevalence in our hospital.
... Previous studies from Pakistan described ST3493, ST3494, ST3472, ST3489, ST3491, ST3492 and ST664 among P. aeruginosa isolates [93,109]. However, global data showed a diverse range of STs [40,102,125,126]. In our study, two high-risk clones ST235 and ST277 were identified. ...
Article
Full-text available
Carbapenem-resistant Pseudomonas aeruginosa (P. aeruginosa) strains have become a global threat due to their remarkable capability to survive and disseminate successfully by the acquisition of resistance genes. As a result, the treatment strategies have been severely compromised. Due to the insufficient available data regarding P. aeruginosa resistance from Pakistan, we aimed to investigate the resistance mechanisms of 249 P. aeruginosa strains by antimicrobial susceptibility testing, polymerase chain reaction for the detection of carbapenemases, aminoglycoside resistance genes, extended-spectrum beta-lactamases (ESBLs), sequence typing and plasmid typing. Furthermore, we tested silver nanoparticles (AgNPs) to evaluate their in vitro sensitivity against antimicrobial-resistant P. aeruginosa strains. We observed higher resistance against antimicrobials in the general surgery ward, general medicine ward and wound samples. Phenotypic carbapenemase-producer strains comprised 80.7% (201/249) with 89.0% (179/201) demonstrating genes encoding carbapenemases: blaNDM-1 (32.96%), blaOXA48 (37.43%), blaIMP (7.26%), blaVIM (5.03%), blaKPC-2 (1.12%), blaNDM-1/blaOXA48 (13.97%), blaOXA-48/blaVIM (1.68%) and blaVIM/blaIMP (0.56%). Aminoglycoside-modifying enzyme genes and 16S rRNA methylase variants were detected in 43.8% (109/249) strains: aac(6′)-lb (12.8%), aac(3)-lla (12.0%), rmtB (21.1%), rmtC (11.0%), armA (12.8%), rmtD (4.6%), rmtF (6.4%), rmtB/aac(3)-lla (8.2%), rmtB/aac(6′)-lla (7.3%) and rmtB/armA (3.6%). In total, 43.0% (77/179) of the strains coharbored carbapenemases and aminoglycoside resistance genes with 83.1% resistant to at least 1 agent in 3 or more classes and 16.9% resistant to every class of antimicrobials tested. Thirteen sequence types (STs) were identified: ST235, ST277, ST234, ST170, ST381, ST175, ST1455, ST1963, ST313, ST207, ST664, ST357 and ST348. Plasmid replicon types IncFI, IncFII, IncA/C, IncL/M, IncN, IncX, IncR and IncFIIK and MOB types F11, F12, H121, P131 and P3 were detected. Meropenem/AgNPs and Amikacin/AgNPs showed enhanced antibacterial activity. We reported the coexistence of carbapenemases and aminoglycoside resistance genes among carbapenem-resistant P. aeruginosa with diverse clonal lineages from Pakistan. Furthermore, we highlighted AgNP’s potential role in handling future antimicrobial resistance concerns.
... Whereas the most common strategies for resistance to colistin are the alterations of the bacterial outer membrane via modifying the structure of its lipopolysaccharide and reducing its negative charge, in addition to overexpression of the efflux-pump regulators [23]. Resistance to carbapenems is usually multifactorial where it includes the acquisition of carbapenemase encoding genes through horizontal gene transfer, downregulation of the porin (OprD) for carbapenem as well as overexpression of mexAB-oprM efflux pump [24]. It was also reported that P. aeruginosa resistance to aminoglycosides is correlated to the production of aminoglycoside-modifying enzymes as well as efflux mechanisms of resistance [25]. ...
Article
Full-text available
Background Carbapenem-resistant Pseudomonas aeruginosa (CRPA) represents an escalating healthcare hazard with high mortality worldwide, especially in presence of biofilm. The current study aimed to evaluate the anti-biofilm potentials of ceftazidime, colistin, gentamicin, and meropenem alone and in combinations against biofilm-forming CRPA. Methods Biofilm killing and checkerboard assay were performed to detect the effectiveness of combined antibiotics against biofilms and planktonic cells, respectively. The bacterial bioburden retrieved from the established biofilms following treatment with combined antibiotics was utilized to construct a three-dimensional response surface plot. A sigmoidal maximum effect model was applied to determine the pharmacodynamic parameters (maximal effect, median effective concentration, and Hill factor) of each antibiotic to create a mathematical three-dimensional response surface plot. Results Data revealed statistically significant (p < 0.05) superior anti-biofilm potential in the case of colistin followed by a lower effect in the case of gentamicin and meropenem, while ceftazidime exhibited the least anti-biofilm activity. The fractional inhibitory concentration index (FICI ≤ 0.5) indicated synergism following treatment with the combined antibiotics. An elevated anti-biofilm activity was recorded in the case of gentamicin/meropenem compared to ceftazidime/colistin. Synergistic anti-biofilm potentials were also detected via the simulated pharmacodynamic modeling, with higher anti-biofilm activity in the case of the in vitro observation compared to the simulated anti-biofilm profile. Conclusions The present study highlighted the synergistic potentials of the tested antibiotic combinations against P. aeruginosa biofilms and the importance of the mathematical pharmacodynamic modeling in investigating the efficacy of antibiotics in combination as an effective strategy for successful antibiotic therapy to tackle the extensively growing resistance to the currently available antibiotics.
... Moreover, a little downregulation of oprD and obvious up-regulation of MexX/ MexY was observed in PAO1 wild-types, PAO1ΔlasI and PAO1ΔrhlI treated with 2 mg/l of AMK ( Figure 4E). These results could be related to the mechanism of P. aeruginosa resistance to antibiotics (Mesaros et al., 2007;Moradali et al., 2017;Xu et al., 2020). However, the expression of OprD and efflux systems showed no significant difference between wild-type and lasI or rhlI deficient strain ( Figure 4E). ...
Article
Full-text available
The prevalence and genetic mechanism of antibiotic heteroresistance (HR) have attracted significant research attention recently. However, non-genetic mechanism of HR has not been adequately explored. The present study aimed to evaluate the role of quorum sensing (QS), an important mechanism of behavioral coordination in different subpopulations and consequent heteroresistance. First, the prevalence of HR to 7 antibiotics was investigated in 170 clinical isolates of P. aeruginosa using population analysis profiles. The results showed that P. aeruginosa was significantly heteroresistant to meropenem (MEM), amikacin (AMK), ciprofloxacin (CIP), and ceftazidime (CAZ). The observed HR was correlated with down-regulation of QS associated genes lasI and rhlI. Further, loss-of-function analysis results showed that reduced expression of lasI and rhlI enhanced HR of P. aeruginosa to MEM, AMK, CIP, and CAZ. Conversely, overexpression of these genes or treatment with 3-oxo-C12-HSL/C4-HSL lowered HR of P. aeruginosa to the four antibiotics. Additionally, although downregulation of oprD and upregulation of efflux-associated genes was evident in heteroresistant subpopulations, their expression was not regulated by LasI and RhlI. Moreover, fitness cost measurements disclosed higher growth rates of PAO1ΔlasI and PAO1ΔrhlI in the presence of sub-MIC antibiotic as compared with that of wild-type PAO1. Our data suggest that under temporary antibiotic pressure, downregulation of QS might result in less fitness cost and promote HR of P. aeruginosa.
... Another study demonstrated that AmpC overexpression along with OprD deficiency altered the resistance phenotype of an isolate susceptible to imipenem, making it resistant, as well as reducing its susceptibility to meropenem and biapenem (Xu et al., 2020). ...
Article
Full-text available
Carbapenem-resistance is a great challenge for antimicrobial therapy in Pseudomonas aeruginosa multidrug-resistant infections, as it reduces therapeutic options. This study investigated carbapenem-resistance mechanisms in six strains of non-carbapenemase-producing P. aeruginosa. Minimal inhibitory concentrations for imipenem and meropenem were determined by epsilometric test and broth microdilution. Mutations in the oprD gene were investigated by PCR, followed by sequencing. Transcriptional levels of oprD, ampC, and efflux pumps genes were analysed through RT-qPCR. Detection of efflux and AmpC activity was assessed by MIC reduction in the presence of the inhibitors: PAβN and cloxacillin, respectively. Resistant strains showed moderate levels of resistance for the evaluated carbapenems. Sequencing of oprD gene revealed similar mutation patterns in strains of the same Sequence Type -ST. A premature stop codon was detected only in the resistant strains of ST2236. Indel mutations were found in the oprD gene of ST2237 strains. Failure to detect oprD transcripts by RT-qPCR further confirms the absence of porin on ST2237 strains. ST2236 strains showed low transcriptional levels for oprD. MexXY-OprM was the only efflux system overexpressed in resistant strains. However, no efflux and AmpC activity was detected. High transcriptional levels of ampC were found in 50% of non-induced resistant strains. All imipem-induced resistant strains showed an increase in ampC expression (>102 - 103 X). It was concluded that the reduction and/or loss of OprD associated with AmpC overexpression is probably the main carbapenem-resistance mechanism in the evaluated strains.
Article
Full-text available
Purpose The emergence of carbapenem-resistant P. aeruginosa (CRPA) harbouring acquired carbapenemase genes (blaVIM, blaIMP and blaNDM) has become a global public health threat. Three CRPA isolates included in the study had an extensively drug-resistant phenotype with susceptibility to colistin only and were positive for the blaNDM-1 gene. The current study aimed to investigate the genomic epidemiology and molecular characteristics of the blaNDM-1-positive CRPA isolates collected from the Gauteng region, South Africa. Methods Short read whole genome sequencing (WGS) was performed to determine sequence types (STs), genetic relatedness, resistome, virulome and the genetic environment of the blaNDM-1 gene. Results The WGS and phylogenetic analyses revealed that the study isolates belonged to an international high-risk clone ST773 and belonged to the same clade with eight blaNDM-1-positive ST773 isolates from Hungary, India, Nigeria, South Korea and USA. The study isolates harboured a wide repertoire of intrinsic and acquired antibiotic resistance genes (ARGs) related with mobile genetic elements, porins and efflux pumps, as well as virulence factor genes. The clade-specific ARGs (blaNDM-1, floR2/cmlA9, rmtB4, tetG) were found in a putative integrative and conjugative element (ICE) region similar to ICE6660-like. Conclusion As ICE carrying the blaNDM-1 gene can easily spread to other P. aeruginosa isolates and other Gram-negative bacteria, the findings in this study highlight the need for appropriate management strategies and active surveillance of CRPA isolates in the Gauteng region, South Africa.
Preprint
In the US, carbapenem resistance in Pseudomonas aeruginosa is strongly linked to the regulation of chromosomal resistance determinants, AmpC and OprD. The β-lactamase AmpC requires overexpression and genetic modifications to be capable of inhibiting imipenem activity. The outer membrane porin OprD can be downregulated or undergo genetic modifications that strongly correlate with imipenem non-susceptibility. Co-administration of imipenem and the β-lactamase inhibitor, relebactam, can lower imipenem MICs and restore susceptibility. However, how this occurs in P. aeruginosa isolates that do not overproduce AmpC or produce a functional OprD for imipenem entry is not understood. Therefore, we investigated whether imipenem could enter P. aeruginosa in the absence of OprD and whether any of the chromosomal β-lactamases (AmpC, OXA-51, PIB-1) contributed to imipenem and/or imipenem/relebactam non-susceptibility. This investigation evaluated 17 imipenem non-susceptible clinical isolates and 3 laboratory strains of PAO1, two of which were porin deletion mutants for either oprD or opdP . Expression of OXA-50 and PIB-1 RNA was similar to PAO1. However, all 20 isolates exhibited AmpC induction under sub-lethal exposure to imipenem. This occurred in the absence of detectable OprD protein in 18 isolates. Collectively, our data identify that OprD is not the only channel required for imipenem entry and that in many isolates the restored susceptibility to imipenem by imipenem/relebactam was due to the interaction of relebactam on the overexpression of AmpC due to imipenem induction.
Article
Pseudomonas is a bacterial genus, with a bona fide environmental habitat that comprises different species, some of them causing diseases in humans and plants, as well as some strains with biotechnological potential. Amongst them, Pseudomonas aeruginosa is currently one of the most important nosocomial pathogens. In addition, this microorganism is a prevalent cause of chronic infections in cystic fibrosis patients and in people suffering from chronic obstructive pulmonary disease. The success of P. aeruginosa in colonising different habitats largely relies on its metabolic versatility and robustness. Besides, this bacterial pathogen harbours in its core genome a large set of virulence determinants that allows it to colonise/infect a variety of hosts, from unicellular organisms to humans. Nevertheless, these are not just the only conditions needed for infecting patients at hospitals. Taking into consideration that infected patients are regularly under antibiotic treatment, the ability to avoid antibiotics' action is also needed. In this sense, P. aeruginosa displays a characteristic low susceptibility to several antibiotics currently used in therapy. This is due to the reduced permeability of its cellular envelopes and the presence in its genome of an arrangement of genes encoding multidrug efflux pumps and antibiotic-inactivating enzymes that contribute to its resilience to antibiotics. Besides intrinsic resistance, P. aeruginosa is able to evolve towards antibiotic resistance through mutations (particularly relevant in the case of chronic infections) and via acquisition of antibiotic resistance genes. It is worth mentioning that acquired resistance is not the only venue that P. aeruginosa has for avoiding the action of antibiotics. Transient resistance can also confer this phenotype. Indeed, the induction of the expression of intrinsic resistance genes by conditions or compounds that P. aeruginosa could face during infection can compromise the effectiveness of antibiotics for treating such infections. In addition, tolerant cells able to survive during the exposure to bactericidal antibiotics without an increase in their antibiotic resistance phenotype are found as well in these patients, and they are the prelude of the evolution towards antibiotic resistance. Finally, P. aeruginosa biofilms, frequently encountered in the lungs of cystic fibrosis patients, in prostheses, or in catheters, present low antibiotic susceptibility and are associated with recalcitrance and disease worsening.
Article
Full-text available
The extensive use of β-lactam antibiotics and the bacterial adaptive capacity have led to the apparently unstoppable increase of antimicrobial resistance, one of the current major global health challenges. In the leading nosocomial pathogen Pseudomonas aeruginosa , the mutation-driven AmpC β-lactamase hyperproduction stands out as the main resistance mechanism, but the molecular cues enabling this system have remained elusive until now. Here, we provide for the first time direct and quantitative information about the soluble cell wall-derived fragments accounting for the different levels and pathways of AmpC hyperproduction. Based on these results, we propose a hierarchical model of signals which ultimately govern ampC hyperexpression and resistance.
Article
Full-text available
Objectives: Bacterial persisters are a small subpopulation of cells that are highly tolerant of antibiotics and contribute to chronic and recalcitrant infections. Global gene expression in Pseudomonas aeruginosa persister cells and genes contributing to persister formation remain largely unknown. The objective of this study was to examine the gene expression profiles of the persister cells and those that regained growth in fresh medium, as well as to identify novel genes related to persister formation. Methods: P. aeruginosa persister cells and those that regrew in fresh medium were collected and subjected to RNA sequencing analysis. Genes up-regulated in the persister cells were overexpressed to evaluate their roles in persister formation. The functions of the persister-contributing genes were assessed with pulse-chase assay, affinity chromatography, fluorescence and electron microscopy, as well as a light-scattering assay. Results: An operon containing PA2282-PA2287 was up-regulated in the persister cells and down-regulated in the regrowing cells. PA2285 and PA2287 play key roles in persister formation. PA2285 and PA2287 were found to bind to RpoC and FtsZ, which are involved in transcription and cell division, respectively. Pulse-chase assays demonstrated inhibitory effects of PA2285 and PA2287 on the overall transcription. Meanwhile, light-scattering and microscopy assays demonstrated that PA2285 and PA2287 interfere with cell division by inhibiting FtsZ aggregation. PA2285 and PA2287 are conserved in pseudomonads and their homologous genes in Pseudomonas putida contribute to persister formation. Conclusions: PA2285 and PA2287 are novel bifunctional proteins that contribute to persister formation in P. aeruginosa.
Article
Full-text available
The bacterial cell-wall that forms a protective layer over the inner membrane is called the murein sacculus - a tightly cross-linked peptidoglycan mesh unique to bacteria. Cell-wall synthesis and recycling are critical cellular processes essential for cell growth, elongation and division. Both de novo synthesis and recycling involve an array of enzymes across all cellular compartments, namely the outer membrane, periplasm, inner membrane and cytoplasm. Due to the exclusivity of peptidoglycan in the bacterial cell-wall, these players are the target of choice for many antibacterial agents. Our current understanding of cell-wall biochemistry and biogenesis in Gram-negative organisms stems mostly from studies of Escherichia coli. An incomplete knowledge on these processes exists for the opportunistic Gram-negative pathogen, Pseudomonas aeruginosa. In this review, cell-wall synthesis and recycling in the various cellular compartments are compared and contrasted between E. coli and P. aeruginosa. Despite the fact that there is a remarkable similarity of these processes between the two bacterial species, crucial differences alter their resistance to β-lactams, fluoroquinolones and aminoglycosides. One of the common mediators underlying resistance is the amp system whose mechanism of action is closely associated with the cell-wall recycling pathway. The activation of amp genes results in expression of AmpC β-lactamase through its cognate regulator AmpR which further regulates multi-drug resistance. In addition, other cell-wall recycling enzymes also contribute to antibiotic resistance. This comprehensive summary of the information should spawn new ideas on how to effectively target cell-wall processes to combat the growing resistance to existing antibiotics.
Article
Full-text available
This review deeply addresses for the first time the diversity, regulation and mechanisms leading to mutational overexpression of intrinsic β-lactamases from non-fermenting and other non-Enterobacteriaceae Gram-negative opportunistic pathogens. After a general overview of the intrinsic β-lactamases described so far in these microorganisms, including circa. 60 species and 100 different enzymes, we review the wide array of regulatory pathways of these β-lactamases. They include diverse LysR-type regulators, which control the expression of β-lactamases from relevant nosocomial pathogens such as Pseudomonas aeruginosa or Stenothrophomonas maltophilia or two-component regulators, with special relevance in Aeromonas spp., along with other pathways. Likewise, the multiple mutational mechanisms leading to β-lactamase overexpression and β-lactam resistance development, including AmpD (N-acetyl-muramyl-L-alanine amidase), DacB (PBP4), MrcA (PPBP1A) and other PBPs, BlrAB (two-component regulator) or several lytic transglycosylases among others, are also described. Moreover, we address the growing evidence of a major interplay between β-lactamase regulation, peptidoglycan metabolism and virulence. Finally, we analyse recent works showing that blocking of peptidoglycan recycling (such as inhibition of NagZ or AmpG) might be useful to prevent and revert β-lactam resistance. Altogether, the provided information and the identified gaps should be valuable for guiding future strategies for combating multidrug-resistant Gram-negative pathogens.
Article
Full-text available
Purpose: This study examined the risk factors for, and molecular mechanisms underlying, the increase in carbapenem minimum inhibitory concentrations (MICs) in clinical isolates of Pseudomonas aeruginosa. Methodology: Consecutive clinical isolates of P. aeruginosa were collected. The MicroScan WalkAway system detected more than fourfold increases in the MICs of carbapenems in P. aeruginosa isolates serially recovered from some patients during their clinical course. The clinical risk factors associated with this increase were examined by multiple logistic regression analysis. Western blot analysis and nucleotide sequencing of the oprD gene of 19 clonally related and paired P. aeruginosa isolates from the same patients were undertaken to examine the mechanisms underlying the increase in MICs. Results: The results showed that prior use of carbapenems (OR, 2.799; 95 % CI, 1.088-7.200; P=0.033) and the use of ventilators or tracheostomies (OR, 2.648; 95 % CI, 1.051-6.671; P=0.039) were risk factors for increased carbapenem MICs. Analysis of the underlying mechanisms revealed that loss of functional OprD protein due to mutation of the oprD gene tended to occur in P. aeruginosa isolates with imipenem MICs of more than 8 µg ml(-1); a reduction in OprD expression was observed in P. aeruginosa isolates with imipenem MICs of 4 or 8 µg ml(-1). This difference in the resistance mechanism was not correlated with the MICs of meropenem. Conclusion: This difference in the resistance mechanism of P. aeruginosa indicates a critical breakpoint at an imipenem MIC of 8 µg ml(-1), in accordance with EUCAST criteria. Reducing carbapenem use will prevent P. aeruginosa clinical isolates from developing resistance to carbapenems.
Article
Full-text available
Inducible expression of chromosomally encoded β-lactamase(s) is a key mechanism for β-lactam resistance in Enterobacter cloacae , Citrobacter freundii , Pseudomonas aeruginosa , and Stenotrophomonas maltophilia . The muropeptides produced during the peptidoglycan recycling pathway act as activator ligands for β-lactamase(s) induction. The muropeptides 1,6-anhydromuramyl pentapeptide and 1,6-anhydromuramyl tripeptide are the known activator ligands for ampC β-lactamase expression in E. cloacae . Here, we dissected the type of muropepetides for L1/L2 β-lactamase expression in an mrdA deletion mutant of S. maltophilia . Distinct from the findings with the ampC system, 1,6-anhydromuramyl tetrapeptide is the candidate for ΔmrdA -mediated β-lactamase expression in S. maltophilia . Our work extends the understanding of β-lactamase induction and provides valuable information for combating the occurrence of β-lactam resistance.
Article
Full-text available
Bacteria attached to a surface are generally more tolerant to antibiotics than their planktonic counterparts, even without the formation of a biofilm. The mechanism of antibiotic tolerance in biofilm communities is multifactorial, and the genetic backgound underlying this antibiotic tolerance has not yet been fully elucidated. Using transposon mutagenesis, we isolated a mutant with reduced tolerance to biapenem (relative to the wild-type) from adherent cells. Sequencing analysis revealed a mutation in the pslL gene, which is a part of the polysaccharide biosynthesis operon. P. aeruginosa PAO1Δ pslBCD mutant demonstrated a 100-fold lower survival during exposure to biapenem in planktonic and biofilm cells, with a similar phenotype observed in mouse infection model and clinical strains. Transcriptional analysis of adherent cells revealed increased expression of both pslA and pelA, which are directly regulated by bis-(3′, 5′)-cyclic-di-guanidine monophosphate (c-di-GMP). Inactivation of wspF resulted in a significantly increased tolerance to biapenem due to increased production of c-di-GMP. The loss of pslBCD in the Δ wspF mutant background abolished the biapenem-tolerant phenotype of the Δ wspF mutant, underscoring the importance of psl in biapenem tolerance. Overexpression of PA2133, which can catalyze the degradation of c-di-GMP, led to significant reduction in biapenem tolerance in adherent cells, indicating that c-di-GMP is essential in mediating the tolerant effect. The effect of pslBCD on antibiotic tolerance was evident, with 50- and 200-fold lower survival in the presence of ofloxacin and tobramycin, respectively. We speculate that the psl genes, which are activated by surface adherence through elevated intracellular c-di-GMP levels, confer tolerance to antimicrobials.
Article
Full-text available
A complex link between cell-wall recycling/repair and the manifestation of resistance to β-lactam antibiotics in many Enterobacteriaceae and Pseudomonas aeruginosa exists. This process is mediated by specific cell-wall-derived muropeptide products. These muropeptides are internalized into the cytoplasm and bind to the transcriptional regulator AmpR, which controls the cytoplasmic events that lead to expression of β-lactamase, an antibiotic-resistance determinant. The effector-binding domain (EBD) of AmpR was purified to homogeneity. We document that the EBD exists exclusively as a dimer even at a concentration as low as 1 μM. The EBD binds to the suppressor ligand UDP-N-acetyl-β-D-muramyl-L-Ala-γ-D-Glu-meso-DAP-D-Ala-D-Ala (4) and binds to two activator muropeptides, N-acetyl-β-D-glucosamine-(1→4)-1,6-anhydro-N-acetyl-β-D-muramyl-L-Ala-γ-D-Glu-meso-DAP-D-Ala-D-Ala (1c) and 1,6-anhydro-N-acetyl-β-D-muramyl-L-Ala-γ-D-Glu-meso-DAP-D-Ala-D-Ala (2c) as assessed by non-denaturing mass spectrometry. The EBD does not bind to 1,6-anhydro-N-acetyl-β-D-muramyl-L-Ala-γ-D-Glu-meso-DAP (2a). This binding selectivity revises the dogma in the field. The crystal structure of the EBD dimer was solved to 2.2 Å resolution. The EBD crystallizes in a "closed" conformation, in contrast to the "open" structure required to bind the muropeptides. Structural issues of this ligand recognition are addressed by molecular dynamics simulations, which reveal significant differences among the complexes with the effector molecules.
Article
Full-text available
Muropeptides are a group of bacterial natural products generated from the cell wall in the course of its turnover. These compounds are cell-wall recycling intermediates and are also involved in signaling within the bacterium. However, the identity of these signaling molecules remains elusive. The identification and characterization of 20 muropeptides from Pseudomonas aeruginosa is described. The least abundant of these metabolites is present at 100 and the most abundant at 55,000 molecules per bacterium. Analysis of these muropeptides under conditions of induction of resistance to a β-lactam antibiotic identified two signaling muropeptides (N-acetylglucosamine-1,6-anhydro-N-acetylmuramyl pentapeptide and 1,6-anhydro-N-acetylmuramyl pentapeptide). Authentic synthetic samples of these metabolites were shown to activate expression of β-lactamase in the absence of any β-lactam antibiotic, thus indicating that they serve as chemical signals in this complex biochemical pathway.
Article
This study sought to both identify optimal dosage regimens and estimate pharmacokinetic/pharmacodynamics characteristics of short-infusion (SI) versus extended-infusion (EI) for the use of biapenem against Pseudomonas aeruginosa infections in Chinese Intensive Care patients. A total of 85 strains of P. aeruginosa were collected, and the minimum inhibitory concentration (MIC) of biapenem was measured by the serial two-fold agar dilution method. We designed four frequently used clinical regimens: biapenem 300mg I.V. q12h, q8h, and q6h, and 600mg q12h. The Monte Carlo Simulation (MCS) was performed using previously published pharmacokinetics data to calculate the probability of target attainment (PTA) and the cumulative fraction of response (CFR) of these regimens as a SI (0.5h) and an EI (1h, 2h, 3h, 4h). For a target of 40%fT >MIC (serum drug concentration remains above the MIC for a dosing period), all regimens did not achieve any CFRs >90% for P. aeruginosa, multidrug resistant (MDR)-PA and even non MDR-PA. The traditional biapenem SI regimens most commonly seen in clinical practice were insufficient in treating both MDR and non-MDR P. aeruginosa in ICU patients. However, biapenem 600mg q12h over 2-4h EI regimens could obtain CFR >90% with 20%fT>MIC. Clinical trials should aim to validate the potentially greater PK/PD index with administering higher doses with more frequency and longer extended infusions.