• Aucun résultat trouvé

Carbapenemase and virulence factors of Enterobacteriaceae in North Lebanon between 2008 and 2012: evolution via endemic spread of OXA-48

N/A
N/A
Protected

Academic year: 2021

Partager "Carbapenemase and virulence factors of Enterobacteriaceae in North Lebanon between 2008 and 2012: evolution via endemic spread of OXA-48"

Copied!
7
0
0

Texte intégral

(1)

Carbapenemase and virulence factors of Enterobacteriaceae in

North Lebanon between 2008 and 2012: evolution via endemic

spread of OXA-48

R. Beyrouthy

1–4

, F. Robin

1,2,4

, F. Dabboussi

3

, H. Mallat

3

, M. Hamze´

3

and R. Bonnet

1,2,4

*

1

Clermont Universite´, UMR 1071 Inserm/Universite´ d’Auvergne, 63000 Clermont-Ferrand, France;

2

INRA, USC 2018,

63000 Clermont-Ferrand, France;

3

Health and Environment Microbiology Laboratory, AZM Research Centre of Biotechnology,

Doctoral School of Sciences and Technology, and Faculty of Public Health, Lebanese University, Tripoli, Lebanon;

4

Centre Hospitalier Universitaire, 63000 Clermont-Ferrand, France

*Corresponding author. CHU, Laboratoire de Bacteriologie, 58 rue Montalembert, 63000 Clermont-Ferrand, France. Tel:+33-(0)4-73-754-920; Fax:+33-(0)4-73-754-922; E-mail: rbonnet@chu-clermontferrand.fr

Received 4 January 2014; returned 21 February 2014; revised 24 April 2014; accepted 30 April 2014

Objectives: To investigate the resistance to carbapenems in Enterobacteriaceae and the underlying resistance

mechanisms in North Lebanon between 2008 and 2012.

Methods: A total of 2767 Enterobacteriaceae isolates recovered from clinical samples collected in Nini Hospital

(North Lebanon) were screened for a decrease in susceptibility or resistance to ertapenem (MIC .0.25 mg/L).

Enterobacteriaceae were similarly screened from 183 faecal samples obtained from non-hospitalized patients.

The bacterial isolates were assigned to clonal lineages by PFGE and multilocus sequence typing. Carbapenemase

genes, their genetic environment and virulence genes were characterized by molecular approaches.

Results: The rate of Enterobacteriaceae exhibiting a decrease in susceptibility or resistance to ertapenem

increased from 0.4% in 2008 –10 to 1.6% in 2012 for the clinical isolates recovered from hospitalized patients.

Of these isolates, scattered among seven enterobacterial species, 88% produced OXA-48 carbapenemase.

However, Escherichia coli represented 73% of the OXA-48-producing Enterobacteriaceae collected in 2012 at

this hospital. During the faecal carriage study performed in non-hospitalized patients, E. coli was the only

species producing OXA-48. The bla

OXA-48

gene was mainly found within Tn1999.2-type transposons inserted

into E. coli chromosomes or in

50, 62 or 85 kb plasmids. The plasmids and chromosomal inserts were related

to pOXA-48a. Molecular typing of the isolates revealed clonal diversity of E. coli and Klebsiella pneumoniae

pro-ducing OXA-48. OXA-48 was observed in all major E. coli phylogroups, including D and B2, and isolates harbouring

virulence genes of extra-intestinal pathogenic E. coli. Although not belonging to highly virulent capsular

geno-types, the OXA-48-producing K. pneumoniae harboured genes associated with virulence or host colonization.

Conclusions: Horizontal transfer of related plasmids has facilitated the spread of the bla

OXA-48

gene into several

Enterobacteriaceae species, including virulent E. coli. Their clonal diversity and the presence of faecal carriers in

the community suggest an endemic spread of OXA-48.

Keywords: OXA-48, Escherichia coli, Klebsiella pneumoniae

Introduction

Carbapenems are broad-spectrum antibiotics that constitute the

last-line therapeutic option available to treat infections caused

by multidrug-resistant Enterobacteriaceae.

1

However, due to

the emerging resistance to carbapenems worldwide, the

anti-microbial activity of these drugs is no longer guaranteed.

2

The

most concerning mechanism of resistance is the acquisition of

carbapenem-hydrolysing b-lactamases. The most prevalent

carbapenemases are Ambler class A enzymes such as KPC-type

b

-lactamases, class B enzymes (such as VIM-, IMP- and

NDM-type metallo-b-lactamases) and class D enzymes (such as

OXA-48).

3

OXA-48 was initially identified from a Klebsiella pneumoniae

isolate in Turkey

4

and then spread to various Enterobacteriaceae

species, especially throughout the southern Mediterranean area

and in Europe.

5,6

OXA-48-encoding genes are primarily found in

K. pneumoniae and, to a lesser extent, in Escherichia coli and

#The Author 2014. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com

(2)

Enterobacter spp.

6

Their emergence is mediated by the rapid

spread of broad host-range conjugative plasmids harbouring

the bla

OXA-48

gene located within a Tn1999-type composite

transposon.

7–9

The bla

OXA-48

gene was essentially observed

in the IncL/M plasmids pOXA-48a and derivatives (62 kb),

7,8

pKPoxa-48N2 (160 kb) characterized from a K. pneumoniae strain

isolated in France

8

and pJEG011 (72 kb) characterized from a

K. pneumoniae strain isolated in Australia.

9

In addition to often being multiresistant, K. pneumoniae is an

opportunistic pathogen and can give rise to severe diseases

responsible for community- and hospital-acquired infections. Its

virulence trait is due to the presence of the K1 and K2 capsular

ser-otypes, lipopolysaccharide synthesis, iron-scavenging systems

and adhesins and its ability to form biofilms.

10

E. coli organisms

include commensal isolates belonging to the E. coli phylogroups

A and B1 and virulent isolates, including intestinal pathogenic

E. coli and extra-intestinal pathogenic E. coli (ExPEC).

11

The latter

belong to the B2 and D E. coli phylogroups and have acquired

pathogenicity islands encoding virulence factors.

12

Although

the association of virulence with resistance was observed in

an OXA-48-producing E. coli isolate,

13

the virulence traits of

carbapenemase-producing bacteria remain largely unknown.

The aim of this work was to investigate the evolution of

the resistance to carbapenems in Enterobacteriaceae isolated

in North Lebanon between 2008 and 2012, the underlying

resistance mechanisms and the genetic background and

viru-lence factors of these bacteria.

Materials and methods

Bacteria

A total of 2767 Enterobacteriaceae strains isolated from clinical samples in Nini Hospital, Lebanon, between January 2008 and December 2012 were screened for reduced susceptibility or resistance to ertapenem (MIC .0.25 mg/L). Nini Hospital has 125 general-care beds and 12 intensive-care beds. The annual number of hospital admissions is18000 and the hospital serves an area population of1000000. Twenty-four non-redundant clinical strains were recovered from hospitalized patients. None of the patients included in the study had travelled recently.

In parallel, Enterobacteriaceae exhibiting decreased susceptibility or resistance to ertapenem (MIC .0.25 mg/L) were collected from 183 faecal samples obtained in December 2012 from healthy children who had not been admitted to hospital and had not taken antibiotics in the previous 6 months. Non-redundant intestinal Enterobacteriaceae were recovered by spreading the corresponding faecal samples on MacConkey agar plates (bioMe´rieux, Marcy-l’E´toile, France) supplemented with ertapenem (0.5 mg/L) or cefotaxime (4 mg/L).

The strains were identified using the MALDI-TOF mass spectrometry system VITEK MS (bioMe´rieux). Rifampicin-resistant E. coli C600 was used for mating-out assays and E. coli DH5a for electroporation.

Determination of genetic relatedness and the

E. coli phylogroup

PFGE was performed using a CHEF-DR III apparatus (Bio-Rad Laboratories, Hercules, CA, USA) according to the manufacturer’s recommendations. Pulsotypes were interpreted according to criteria previously defined by Tenover et al.14Allelic profiles and sequence types (STs) were assigned using the multilocus sequence typing (MLST) scheme described by Diancourt et al.15 (http://www.pasteur.fr/recherche/genopole/PF8/mlst/ Kpneumoniae.html) and Achtman (http://mlst.warwick.ac.uk/mlst/dbs/Ecoli)

for K. pneumoniae and E. coli isolates, respectively. E. coli phylogenetic groups were assigned by pentaplex PCR.16

Antibiogram and MIC determination

Antibiotic susceptibility was assessed by the disc diffusion method and the determination of MICs by a broth microdilution method according to the guidelines of EUCAST (http://www.eucast.org/). Antimicrobial agents were obtained from Sigma Chemical Co. (St Louis, MO, USA) or directly from manufacturing companies. Carbapenemase production was detected by a modified Hodge test using combined disc tests of ertapenem with and without cloxacillin and the Mastdiscs ID inhibitor combination disc method (Mast Diagnostics, Bootle, UK).17,18Extended-spectrum b-lactamase (ESBL) production was detected by both the combination disc test and the double-disc synergy test according to the guidelines of EUCAST (http://www.eucast.org/).

Molecular identification of b-lactamases and additional

associated resistance genes

b-Lactamase-encoding genes (blaTEM, blaCTX-MblaKPC, blaNDM, blaVIMand

blaOXA-48) were detected by PCR amplification (Table S1, available as

Supplementary data at JAC Online). The genetic environment of the blaOXA-48gene was further investigated by PCR and sequencing using

primers specific for the insertion sequence IS1999 and the blaOXA-48

gene to map the composite transposon Tn1999 and its derivatives Tn1999.2, Tn1999.3 and Tn1999.4.

Analysis of plasmids and chromosome

Transferability of the blaOXA-48 gene was studied by a mating-out

assay. When these experiments failed, plasmid DNA was extracted with NucleoBondwXtra (Macherey-Nagel, Germany) and transferred by

electro-poration into a bacterial recipient. Selection was performed on agar plates supplemented with ticarcillin (32 mg/L) and rifampicin (300 mg/L) for the mating-out assay and ticarcillin (32 mg/L) for electroporation. The plasmid content of the bacteria and the size of plasmids were determined using plas-mid DNA extracted by the method of Kado and Liu19with the plasmids Rsa (39 kb), TP114 (61 kb), pCFF04 (85 kb) and pCFF14 (180 kb) as standards.

PCR-based replicon typing (PBRT) was used to identify plasmid incom-patibility groups in transconjugants or transformants.20The repA, traU and parA genes of the pOXA-48a plasmid were detected by PCR, as previously described.8

Chromosome analysis

The location of the blaOXA-48gene was investigated by PFGE using the

endonuclease I-CeuI and hybridizations with probes specific for the 16S rRNA gene, the blaOXA-48gene and IS1999, as previously described.21

The chromosomal insertion of the blaOXA-48gene was further mapped

by PCR using blaOXA-48-containing DNA fragments purified from PFGE

gels and primers specific for pOXA-48a, as previously described.7

Virulence factor-encoding genes and

pathogenicity islands

K. pneumoniae virulence genes (allS, cf29a, kfu, magA, mrkD, rmpA, uge, ureA, wabG, wzy and wzx) and the polyketide synthetase (pks) gen-omic island were screened by PCR, as previously described.10,22,23 Similarly, virulence factors encoded by the E. coli isolates were detected by PCR targeting the genes encoding adhesins (papC, papG alleles, sfa and hra), siderophores (fyA, iroN and ear), cyclomodulins (pks, cnf and hlyA), malx and pathogenicity islands (IJ96, IIJ96, ICFT073, IICFT073,

I536, II536, III536, IV536and VI536), as previously described.24,25

Beyrouthy et al.

(3)

Biofilm formation

Biofilm production was assessed using the microtitre plate assay, as described by the modified protocol of O’Toole and Kolter.26The F plasmid-bearing Escherichia coli TG1 strain27was used as a positive control.

Results

Decrease in susceptibility to ertapenem

in hospitalized patients

In total, 2767 Enterobacteriaceae isolates were recovered from

clinical samples at Nini Hospital, Tripoli, Lebanon, between 2008

and 2012. These isolates mainly belonged to the species E. coli

(n ¼ 2284, 82.6%), K. pneumoniae (n ¼ 230, 8.3%), Enterobacter

cloacae (n¼ 10, 0.3%), Enterobacter aerogenes (n ¼ 6, 0.2%) and

Proteus mirabilis (n ¼ 106, 3.8%). Twenty-four non-redundant

isolates exhibiting resistance or reduced susceptibility to

ertape-nem (MIC .0.25 mg/L) were recovered between 2008 and

2012. The isolates belonged to seven bacterial species and were

predominantly collected from urine (n ¼ 14) and pus (n ¼ 6)

(Table

1

). The rate of isolates exhibiting reduced susceptibility or

resistance to ertapenem increased from 0.4% (n ¼ 6/1385) in

2008 – 10 to 0.9% (n ¼ 6/650) in 2011 and 1.6% (n ¼ 12/732)

in 2012.

Intestinal carriage in the community

In parallel, the faecal carriage of Enterobacteriaceae

exhibit-ing resistance or reduced susceptibility to ertapenem (MIC

Table 1. Phenotypic and genotypic characteristics of OXA-48-producing isolates

Isolate

STa(E. coli

phylogroup)

Isolation site (year)

OXA-48 genetic environment

Other b-lactamases

MIC (mg/L)

transposon parAb repAb traUb

genetic

support ETP IPM MEM CTX CAZ

E. coli

EC8 131 (B2) urine (2012) DTn1999.2c + + + chromd 2 1.0 0.5 0.12 0.25 0.5

EC37 88 (A) stoole(2012) DTn1999.2c + 2 2 chromd 2 2 2 0.5 0.5 0.5

EC49 38 (D) stoole(2012) NTf + 2 2 chromd CTX-M-24, TEM-1 1 0.5 0.12 256 1

EC119 3877 (A) stoole(2012) Tn1999.2 + + + 62 kb 2 1 0.5 0.25 0.06 0.12

EC253 617 (A) urine (2012) Tn1999.2 + + + 62 kb CTX-M-15, OXA-1, TEM-1 2 1 0.25 128 16

EC254 38 (D) urine (2012) DTn1999.2c + 2 2 chromd CTX-M-24, TEM-1 1 0.5 0.25 16 1

EC260 1711 (B1) urine (2012) Tn1999.2 + + + 62 kbg TEM-1 0.5 0.25 0.25 0.06 0.06

EC264 227 (A) urine (2012) DTn1999.2c + 2 2 chromd TEM-1 0.5 0.25 0.12 0.06 0.06

EC265 38 (D) pus (2012) DTn1999.2c + 2 2 chromd CTX-M-24, TEM-1 1 0.5 0.12 128 1

EC267 88 (A) sputum (2012) DTn1999.2c + 2 2 chromd CTX-M-15, TEM-1 2 1 0.25 16 16

EC269 617 (A) urine (2012) Tn1999.2 + + + 62 kbg CTX-M-15, TEM-1 2 1 0.25 512 128

EC15 127 (B2) sputum (2011) DTn1999.2c + 2 2 chromd 2 0.5 0.25 0.25 0.06 0.06

K. pneumoniae KP78 1157 blood (2012) Tn1999.2 + + + 62 kbg SHV-1 0.5 1 0.25 2 2 KP87 1156 pus (2012) Tn1999 + + + 62 kbg CTX-M-15, SHV-1, TEM-1 0.5 1 0.25 64 32 KP34 45 urine (2011) Tn1999.2 + + + 62 kbg SHV-1, TEM-1 1 1 0.25 0.06 0.06 KP9 866 urine (2009) Tn1999.2 + + + 62 kbg SHV-1 8 8 4 32 16 KP26 866 urine (2008) Tn1999.2 + + + 62 kbg SHV-1 32 64 8 16 1 KP112 866 pus (2008) Tn1999.2 + + + 85 kbh SHV-1 8 8 4 8 0.5

E. aerogenes EA2 — pus (2012) Tn1999.2 + + + 62 kbg 2 1 0.5 0.25 32 64

C. koseri CK8 — urine (2011) Tn1999.2 + + + 62 kbg CTX-M-15, TEM-1 2 1 0.25 256 32

R. planticola RA35 — pus (2011) Tn1999.2 + + + 62 kbg 2 2 1 2 0.5 1

C. freundii CF29 — urine (2010) Tn1999.2 + + + 50 kbi TEM-1 0.5 0.5 0.5 0.25 0.25

E. cloacae ECL6 — urine (2010) Tn1999.2 + + + 62 kbg 2 2 0.5 0.25 1 0.5

E. cloacae ECL8 — urine (2009) Tn1999.2 + + + 62 kbg 2 8 8 4 128 32

ETP, ertapenem; IPM, imipenem; MEM, meropenem; CTX, cefotaxime; CAZ, ceftazidime.

aSTs for E. coli and K. pneumoniae isolates. bPCR for genes specific for the pOXA-48a backbone. c

Incomplete structure of Tn1999.2.

dChromosome-mediated OXA-48. e

Faecal carriage in children having no known contact with the hospital.

fNon-typeable Tn1999-type environment. g62 kb plasmid encoding OXA-48. h

85 kb plasmid encoding OXA-48.

i50 kb plasmid encoding OXA-48.

OXA-48 carbapenemase in North Lebanon

JAC

(4)

.0.25 mg/L) was screened in 2012 from samples obtained from

183 healthy children in the community. The children were enrolled

in two schools with different socio-economic levels. School 1

(n ¼ 120) was a welfare school catering for children of poor

families. School 2 (n ¼ 87) was a private institution, with pupils

that were drawn from upper-class families. Three E. coli isolates

exhibiting resistance to ertapenem (n ¼ 3/183, 1.5%) were

recovered from children (9, 11 and 12 years old) enrolled in

three different classes at school 1.

Phenotypic and molecular identification of b-lactamases

PCR and sequencing confirmed the presence of the bla

OXA-48

gene

in 88% of the clinical isolates (n ¼21/24) and in the three faecal

E. coli isolates from healthy children (Table

1

). No other gene

encoding carbapenemase was detected in the isolates, including

those devoid of the bla

OXA-48

gene (Enterobacter spp., n¼ 3/24). In

addition, the ESBL synergy test was positive for eight

OXA-48-producing isolates. PCR and sequencing showed the presence of

the ESBL-encoding genes bla

CTX-M-15

and bla

CTX-M-24

in five and

three isolates, respectively (Table

1

).

MICs of b-lactams for OXA-48-producing bacteria

The OXA-48-producing isolates were homogeneously resistant to

penicillins and their combinations with b-lactamase inhibitors.

The MIC of temocillin, which has been proposed as a screening

test for OXA-48 detection, was .32 mg/L for all isolates except

E. aerogenes EA2. The isolates exhibited various levels of

resist-ance to carbapenems (Table

1

). The MICs of oxyimino

cephalos-porins were in the resistance range for isolates producing

both OXA-48 and an ESBL. The MIC values of cefotaxime and

ceftazidime were in the susceptible range (0.06 –2 mg/L) for the

isolates devoid of an ESBL, except for three K. pneumoniae isolates

(KP9, KP26 and KP112) and E. cloacae ECL8.

Plasmidic genetic support of the bla

OXA-48

gene

Except for eight E. coli strains, transconjugants (n ¼ 15) and

transformants (n¼ 2) were obtained from the OXA-48-encoding

strains by a mating-out assay and electroporation. All of the

trans-conjugants and transformants exhibited a b-lactam resistance

phenotype compatible with OXA-48 production, and the presence

of the corresponding gene was confirmed by PCR experiments.

Plasmids from natural isolates and their transformants or

trans-conjugants were extracted and hybridized with a probe specific

for the bla

OXA-48

gene. The results showed that the bla

OXA-48

gene was predominantly located on

62 kb conjugative plasmids

(Table

1

). Further analysis of these plasmids from the

transconju-gants revealed similar restriction profiles (data not shown).

However, the bla

OXA-48

gene was also harboured by plasmids of

50 and 85 kb in Citrobacter freundii CF29 and K. pneumoniae

KP112 (Figure

1

and Table

1

). Resistance to non-b-lactam

antibio-tics was not observed in the transconjugants and transformants,

except for that containing the

85 kb plasmid, which exhibited

resistance to sulphonamides and spectinomycin. No plasmids

could be assigned to an incompatibility group by the PBRT method.

We detected the repA, traU and parA genes of the pOXA-48a

plas-mid in all of the transformants or transconjugants (Table

1

). These

results suggested that the plasmids had an IncL/M-pOXA-48a-like

backbone, including the

50 and 85 kb plasmids.

Chromosomal genetic support of the bla

OXA-48

gene

Despite many attempts, no transconjugants or transformants

were obtained from six clinical and two faecal E. coli isolates.

The insertion of the bla

OXA-48

gene into the bacterial chromosome

was therefore investigated by PFGE after I-CeuI nuclease

treat-ment and hybridization. Different DNA fragtreat-ments (size range

75 to 0.88 Mb) hybridized with the bla

OXA-48

probe (Figure

2

).

These results show that the insertion of the bla

OXA-48

gene into

the E. coli chromosome is not a rare event (33%, n ¼ 8/24) and

may occur at different sites. The parA gene of pOXA-48a (but

not repA and traU) was detected by PCR and sequenced in the

bla

OXA-48

-harbouring DNA fragments, which were eluted from

PFGE. These results suggest that the strains contain a large

frag-ment inserted into the chromosome and that the DNA fragfrag-ments

harbouring the bla

OXA-48

gene originated in a pOXA-48a-type

plasmid.

Genetic environment of the bla

OXA-48

gene

To characterize the genetic environment of the bla

OXA-48

gene in

our strains, we mapped Tn1999-type transposons harbouring

the bla

OXA-48

gene by PCR and sequencing. The bla

OXA-48

gene

was predominantly associated with Tn1999.2 (63%, n ¼ 15/24).

In addition, seven bla

OXA-48

genes (29%, n ¼ 7/24) were located

in incomplete Tn1999.2 elements. These incomplete Tn1999.2

85kb 62kb 50kb A B B 1 2 1 2 3 4 3 4 5 6 5 6 A A B

Figure 1. Plasmid DNA content of representative strains and their transconjugants and transformants. A, DNA on a 0.7% agarose gel; B, hybridization using a probe specific for blaOXA-48. 1 and 2, plasmid content of KP112 and the corresponding transformant, respectively; 3 and 4, plasmid content of

RA35 and the corresponding transconjugant, respectively; 5 and 6, plasmid content of CF29 and its transformant, respectively.

Beyrouthy et al.

(5)

elements were observed only in the isolates containing a

chromosome-mediated bla

OXA-48

gene.

Genetic background and virulence factors

of K. pneumoniae and E. coli

The genetic background of K. pneumoniae and E. coli isolates

pro-ducing OXA-48 was investigated by PFGE and MLST analyses

(Table

2

). Most of the OXA-48-producing K. pneumoniae and

E. coli isolates were different according to Tenover’s

14

criteria for

interpreting PFGE patterns (data not shown). Only K. pneumoniae

isolates KP9, KP26 and KP112 were closely related. Two E. coli

isolates presented identical pulsotypes (EC254 and EC265) and

one was closely related (EC49).

Four STs were represented among the six K. pneumoniae

isolates (Table

1

). No K. pneumoniae isolate exhibited the virulent

capsular serotype K1 or K2. Among the genes encoding adhesins,

only mrkD was detected. However, all isolates harboured the

genes wabG and uge, which favour host colonization and

viru-lence (Table

2

). The ferric iron uptake system encoded by kfu

was detected in four isolates. All strains produced biofilms; four

isolates in particular showed biofilm production that was not

sig-nificantly different from that observed for the reference strain TG1

(Table

2

; ANOVA, P. 0.05).

Seven STs were represented among the E. coli isolates

(Table

1

), belonging predominantly to the commensal

phy-logroups A (n ¼ 6) and B1 (n ¼ 1). Five isolates were assigned to

phylogroup D or B2. These latter isolates harboured virulence

genes and pathogenicity islands previously observed in ExPEC

strains (Table

3

). Strains EC8 and EC15, which belonged to

phy-logroup B2, contained a higher number of pathogenicity islands

(five or more) and virulence genes (six or more) than the E. coli

isolates of phylogroup D.

Discussion

In this study, we showed that the rate of Enterobacteriaceae

exhibiting a decrease in susceptibility to ertapenem in Nini

Hospital, North Lebanon, increased from 0.4% in 2008 – 10 to

1.6% in 2012. This rise was associated with the emergence of

carbapenemase OXA-48, which had been previously reported

in Lebanon in 2008 – 10.

28–30

E. coli was the predominant

species producing OXA-48, in contrast to previous studies, which

generally cite K. pneumoniae as the main OXA-48-producing

Enterobacteriaceae.

4,6,7,31–35

E. coli isolates represented 10% of

the OXA-48 clinical producers in 2008 – 11 and 73% in 2012. In

addition, intestinal carriage of OXA-48-producing E. coli was

observed in the community and was marked by a diversity of

A B C A B C A B C A B C A B C

600kb_

840kb_ 870kb_

740kb_

880kb_

Figure 2. Chromosomal location of blaOXA-48in five representative E. coli isolates. A, I-CeuI fragments of chromosomes analysed by PFGE; B, hybridization

after transfer to a nylon membrane using probes specific for the 16S RNA gene; C, hybridization after transfer to a nylon membrane using probes specific for blaOXA-48.

Table 2. STs, virulence factor-encoding genes and biofilm formation in OXA-48-producing K. pneumoniae isolates

Isolate Source ST

Virulence factor-encoding genes

Biofilm formation relative to E. coli strain TG1

ureA wabG uge cf29a kfu mrkD rmpA allS magA pks

KP9 urine 866 + + + 2 + + 2 2 2 2 0.99+0.28 KP26 urine 866 + + + 2 + + 2 2 2 2 1.11+0.17 KP34 urine 45 + + + 2 + + 2 2 2 2 0.15+0.01 KP78 blood 1157 + + + 2 2 + 2 2 2 2 0.71+0.18 KP87 pus 1156 + + + 2 2 + 2 + 2 2 0.23+0.07 KP112 pus 866 + + + 2 + + 2 2 2 2 0.84+0.20

OXA-48 carbapenemase in North Lebanon

JAC

(6)

strains, suggesting that OXA-48 has become endemic in North

Lebanon.

OXA-48 was observed in all major phylogroups of E. coli. Most

of the isolates belonged to phylogroup A, which predominantly

comprises commensal strains. However, D and B2

OXA-48-producing E. coli strains harboured virulence genes of

extra-intestinal pathogenic E. coli. No highly virulent OXA-48-producing

K. pneumoniae isolate was found. However, OXA-48-producing

K. pneumoniae isolates produced a significant amount of biofilm

and harboured genes previously associated with virulence and

favouring host colonization.

A recent study showed that pOXA-48a is highly transferable

in vitro.

36

Our findings showed that the emergence of OXA-48

car-bapenemase was facilitated by the horizontal transfer of plasmids

related to pOXA-48a-like plasmids.

32

A recent study reported the

presence of the bla

OXA-48

gene in the bacterial chromosome of a

sporadic E. coli isolate.

13

In this study, the bla

OXA-48

gene was

located in the chromosome in 33% of the OXA-48-producing

Enterobacteriaceae and in 67% of the OXA-48-producing E. coli.

A chromosomal location of the bla

OXA-48

gene is therefore not

rare and is observed in unrelated E. coli isolates. The Tn1999.2

elem-ent was predominant in the plasmids and the E. coli chromosomes

harbouring the bla

OXA-48

gene. However, chromosome-mediated

Tn1999.2 was truncated, most likely as a result of its insertion

into the E. coli chromosome. Furthermore, the parA gene, which

is located

13 kb from the bla

OXA-48

gene on pOXA-48a, was

observed in a chromosomal DNA fragment harbouring the

bla

OXA-48

gene, suggesting that Tn1999.2 is most likely not the

only DNA fragment inserted. These results suggest that multiple

acquisitions of the bla

OXA-48

gene in E. coli chromosomes involve

a Tn1999.2-containing fragment of a pOXA-48a-like plasmid.

In conclusion, the distribution and diversity of Enterobacteriaceae

producing OXA-48 carbapenemase demonstrate that this

resist-ance mechanism is becoming widespread in hospitals and

in the community in North Lebanon. The occurrence of the

bla

OXA-48

gene is worrying due to the spread of both highly

diffusing plasmids and their presence in E. coli producers. This

situation resembles the ecological success story of CTX-M-type

ESBLs, the worldwide emergence of which is most likely due to

their association with E. coli.

Acknowledgements

We thank Laurent Guillouard and Alexis Pontvianne for their technical assistance. We also thank the two schools for their helpful collaboration.

Funding

This research project was supported by the National Council for Scientific Research, Lebanon, the AZM Research Centre of Biotechnology, Lebanese University, Lebanon, the Ministe`re de la Recherche et de la Technologie, l’Institut National de la Recherche Agronomique (USC-2018) and the Centre Hospitalier Re´gional Universitaire de Clermont-Ferrand, France.

Transparency declarations

None to declare. Table 3. ST s, pa thog enicity isla nds and other virulence fa ctor-encoding gene s in O XA-4 8-pr oducing E. co li st ra in s belo ngin g to th e D and B2 ph ylogr ou ps Isola te Sour ce ST (p h ylogr oup) Pa thogen icity islands Virulence fa ctor-encod ing genes I536 II536 III 536 IV 536 V536 VI536 ICF073 IICFT073 IJ96 IIJ96 hr a fuyA ear ir oN mal X pks sfa hly cnf pap C pap GI papGII papGIII EC8 urine 131 (B2) + 22 + 2 ++ + 22 ++ + 2 + 22 2 2 + 2 + 2 EC15 sputu m 127 (B2) ++ + + + + + + 2 ++ + + + + + 2 ++ + 22 + EC49 stool 38 (D) 22 ++ 2 + 22 2 2 ++ 22 2 2 2 2 2 2 2 2 2 EC254 urine 38 (D) 22 ++ 2 + 22 2 2 ++ 22 2 2 2 2 2 2 2 2 2 EC265 pus 38 (D) 22 ++ 2 + 22 2 2 ++ 22 2 2 2 2 2 2 2 2 2

Beyrouthy et al.

(7)

Supplementary data

Table S1 is available as Supplementary data at JAC Online (http://jac. oxfordjournals.org/).

References

1 Paterson DL, Bonomo RA. Extended-spectrum b-lactamases: a clinical update. Clin Microbiol Rev 2005; 18: 657– 86.

2 Nordmann P, Dortet L, Poirel L. Carbapenem resistance in Enterobacteriaceae: here is the storm! Trends Mol Med 2012; 18: 263–72. 3 Nordmann P, Naas T, Poirel L. Global spread of carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis 2011; 17: 1791–8. 4 Poirel L, He´ritier C, Tolu¨n V et al. Emergence of oxacillinase-mediated resistance to imipenem in Klebsiella pneumoniae. Antimicrob Agents Chemother 2004; 48: 15–22.

5 Canto´n R, Ako´va M, Carmeli Y et al. Rapid evolution and spread of carbapenemases among Enterobacteriaceae in Europe. Clin Microbiol Infect 2012; 18: 413– 31.

6 Poirel L, Potron A, Nordmann P. OXA-48-like carbapenemases: the phantom menace. J Antimicrob Chemother 2012; 67: 1597–606. 7 Poirel L, Bonnin RA, Nordmann P. Genetic features of the widespread plasmid coding for the carbapenemase OXA-48. Antimicrob Agents Chemother 2012; 56: 559–62.

8 Berger S, Alauzet C, Aissa N et al. Characterization of a new blaOXA-48

-carrying plasmid in Enterobacteriaceae. Antimicrob Agents Chemother 2013; 57: 4064– 7.

9 Espedido BA, Steen JA, Ziochos H et al. Whole genome sequence analysis of the first Australian OXA-48-producing outbreak-associated Klebsiella pneumoniae isolates: the resistome and in vivo evolution. PLoS One 2013; 8: e59920.

10 Brisse S, Fevre C, Passet V et al. Virulent clones of Klebsiella pneumoniae: identification and evolutionary scenario based on genomic and phenotypic characterization. PLoS One 2009; 4: e4982.

11 Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nat Rev Microbiol 2004; 2: 123–40.

12 Dobrindt U, Hentschel U, Kaper JB et al. Genome plasticity in pathogenic and nonpathogenic enterobacteria. Curr Top Microbiol Immunol 2002; 264: 157–75.

13 Beyrouthy R, Robin F, Cougnoux A et al. Chromosome-mediated OXA-48 carbapenemase in highly virulent Escherichia coli. J Antimicrob Chemother 2013; 68: 1558– 61.

14 Tenover FC, Arbeit RD, Goering RV et al. Interpreting chromosomal DNA restriction patterns produced by pulsed-field gel electrophoresis: criteria for bacterial strain typing. J Clin Microbiol 1995; 33: 2233– 9.

15 Diancourt L, Passet V, Verhoef J et al. Multilocus sequence typing of Klebsiella pneumoniae nosocomial isolates. J Clin Microbiol 2005; 43: 4178– 82.

16 Bidet P, Metais A, Mahjoub-Messai F et al. Detection and identification by PCR of a highly virulent phylogenetic subgroup among extraintestinal pathogenic Escherichia coli B2 strains. Appl Env Microbiol 2007; 73: 2373– 7.

17 Giske CG, Gezelius L, Samuelsen Ø et al. A sensitive and specific phenotypic assay for detection of metallo-b-lactamases and KPC in Klebsiella pneumoniae with the use of meropenem disks supplemented with aminophenylboronic acid, dipicolinic acid and cloxacillin. Clin Microbiol Infect 2011; 17: 552– 6.

18 Doyle D, Peirano G, Lascols C et al. Laboratory detection of Enterobacteriaceae that produce carbapenemases. J Clin Microbiol 2012; 50: 3877–80.

19 Kado CI, Liu ST. Rapid procedure for detection and isolation of large and small plasmids. J Bacteriol 1981; 145: 1365– 73.

20 Carattoli A, Bertini A, Villa L et al. Identification of plasmids by PCR-based replicon typing. J Microbiol Methods 2005; 63: 219– 28. 21 Bonnet R, Marchandin H, Chanal C et al. Chromosome-encoded class D b-lactamase OXA-23 in Proteus mirabilis. Antimicrob Agents Chemother 2002; 46: 2004– 6.

22 Fang C-T, Lai S-Y, Yi W-C et al. Klebsiella pneumoniae genotype K1: an emerging pathogen that causes septic ocular or central nervous system complications from pyogenic liver abscess. Clin Infect Dis 2007; 45: 284– 93.

23 Nassif X, Fournier JM, Arondel J et al. Mucoid phenotype of Klebsiella pneumoniae is a plasmid-encoded virulence factor. Infect Immun 1989; 57: 546– 52.

24 Sabate´ M, Moreno E, Pe´rez T et al. Pathogenicity island markers in commensal and uropathogenic Escherichia coli isolates. Clin Microbiol Infect 2006; 12: 880– 6.

25 Bronowski C, Smith SL, Yokota K et al. A subset of mucosa-associated Escherichia coli isolates from patients with colon cancer, but not Crohn’s disease, share pathogenicity islands with urinary pathogenic E. coli. Microbiology 2008; 154: 571–83.

26 O’Toole GA, Kolter R. Flagellar and twitching motility are necessary for Pseudomonas aeruginosa biofilm development. Mol Microbiol 1998; 30: 295–304.

27 Ghigo JM. Natural conjugative plasmids induce bacterial biofilm development. Nature 2001; 412: 442–5.

28 Matar GM, Cuzon G, Araj GF et al. Oxacillinase-mediated resistance to carbapenems in Klebsiella pneumoniae from Lebanon. Clin Microbiol Infect 2008; 14: 887–8.

29 Carre¨r A, Poirel L, Yilmaz M et al. Spread of OXA-48-encoding plasmid in Turkey and beyond. Antimicrob Agents Chemother 2010; 54: 1369–73. 30 Matar GM, Dandache I, Carre¨r A et al. Spread of OXA-48-mediated resistance to carbapenems in Lebanese Klebsiella pneumoniae and Escherichia coli that produce extended spectrum b-lactamase. Ann Trop Med Parasitol 2010; 104: 271– 4.

31 Oteo J, Herna´ndez JM, Espasa M et al. Emergence of OXA-48-producing Klebsiella pneumoniae and the novel carbapenemases OXA-244 and OXA-245 in Spain. J Antimicrob Chemother 2013; 68: 317– 21.

32 Potron A, Kalpoe J, Poirel L et al. European dissemination of a single OXA-48-producing Klebsiella pneumoniae clone. Clin Microbiol Infect 2011; 17: E24–26.

33 Pitart C, Sole´ M, Roca I et al. First outbreak of a plasmid-mediated carbapenem-hydrolyzing OXA-48 b-lactamase in Klebsiella pneumoniae in Spain. Antimicrob Agents Chemother 2011; 55: 4398– 401.

34 Lascols C, Peirano G, Hackel M et al. Surveillance and molecular epidemiology of Klebsiella pneumoniae isolates that produce carbapenemases: first report of OXA-48-like enzymes in North America. Antimicrob Agents Chemother 2013; 57: 130–6.

35 Poirel L, Bernabeu S, Fortineau N et al. Emergence of OXA-48-producing Escherichia coli clone ST38 in France. Antimicrob Agents Chemother 2011; 55: 4937–8.

36 Potron A, Poirel L, Nordmann P. Derepressed transfer properties leading to the efficient spread of the plasmid encoding carbapenemase OXA-48. Antimicrob Agents Chemother 2014; 58: 467–71.

OXA-48 carbapenemase in North Lebanon

JAC

Figure

Figure 1. Plasmid DNA content of representative strains and their transconjugants and transformants
Table 2. STs, virulence factor-encoding genes and biofilm formation in OXA-48-producing K

Références

Documents relatifs

These segments are precisely the Ptol- emy segments which can be isometrically embedded in E 2 (this follows essentially from the classical characterization of circles by the

This editorial refers to ‘Relationship between circulating levels of RANTES (regulated on activation, normal T-cell expressed, and secreted) and carotid plaque characteristics:

Previous experiments have shown that in B cells there are two pathways of gene activation through octamer sites ( 23 , 40 ): (i) activation from a promoter octamer site requires

Conversely, parliamentary systems with a proportional representation electoral rule are likely to provide fewer environmental public goods, which allows for a larger marginal impact

Taken together, our genetic analysis, RNA blot hybrid- ization and cRT–PCR data suggest the following model for the biogenesis of dsRNA precursor of TAS3a tasiRNAs (Figure 1A):

The beam current determinations were estimated to be within 10.8% of the values obtained and were determined from the following errors: target density (1 %), proton ränge

in a non-Western context and, in particular, the legitimacy of such an attempt in the case of Early China, where the Warring States period is traditionally con- sidered as the

A. ATTIN: OCCUPATIONAL DENTAL EROSION 171.. Cross-sectional and non-randomized case–control studies investigating the prevalence of erosion in battery, galvanizing and