ArticlePDF Available

Phenotypic and WGS-derived antimicrobial resistance profiles of clinical and non-clinical Acinetobacter baumannii isolates from Germany and Vietnam

Authors:
  • Friedrich-Löffler-Institut, Jena

Abstract

Objectives: This study aimed to combine in vitro phenotyping analysis and whole-genome-sequencing (WGS) to characterise the phenotype and genetic determinants associated with intrinsic resistance in 100 clinical and non-clinical Acinetobacter baumannii strains originating from Germany and Vietnam. Moreover, it aimed to assess whether powdered milk as a food source functions as a potential reservoir of antibiotic resistance and possesses similar antimicrobial resistance (AMR) genes as in clinical strains isolated from Germany. Methods: Antimicrobial susceptibility testing was performed using the broth microdilution method and the minimum inhibitory concentration (MIC) was determined for 18 antibiotics. The WGS data from all isolates were mapped to intrinsic genes known to be associated with phenotypic AMR. Results: The highest resistance frequency was observed for chloramphenicol (100%), followed by fosfomycin (96%) and cefotaxime (95%). The lowest resistant rates were observed for colistin (3%), trimethoprim/sulfamethoxazole (17%), tigecycline (19%), and amikacin (19%). Thirty-five percent of tested strains displayed resistance to at least one of the carbapenems. Resistance to fluoroquinolones, aminoglycosides, tigecycline, penicillins, trimethoprim/sulfamethoxazole, and fourth-generation cephalosporins was determined only in human strains. About one-quarter of isolates (24%) was multidrug-resistant (MDR) and all were of human origin. Among them, 16 isolates were extensively drug resistant (XDR) and 10 from those 16 isolates showed resistance to all tested antibiotics except colistin. In silico detection of intrinsic AMR genes revealed the presence of 36 β-lactamases and 24 non-β-lactamase resistance genes. Two colistin-resistant and 10 ertapenem-resistant strains were isolated from powdered milk produced in Germany. Thirty-eight AMR genes associated with resistance to antibiotics were found in isolates recovered from milk powder. Several resistance mechanisms towards many classes of antibiotics existed in A. baumannii including β-lactamases, multidrug efflux pumps and aminoglycoside-modifying enzymes. Conclusion: The use of WGS for routine public health surveillance is a reliable method for the rapid detection of emerging AMR in A. baumannii isolates. Milk powder poses a risk to contain MDR Acinetobacter strains or resistance genes in Germany.
International Journal of Antimicrobial Agents 56 (2020) 106 127
Contents lists available at ScienceDirect
International Journal of Antimicrobial Agents
journal homepage: www.elsevier.com/locate/ijantimicag
Phenotypic and WGS-derived antimicrobial resistance profiles of
clinical and non-clinical Acinetobacter baumannii isolates from
Germany and Vietnam
Gamal Wareth
a , b ,
, Jörg Linde
a
, Philipp Hammer
c
, Ngoc H. Nguyen
d , g
, Tuan N.M. Nguyen
d
,
Wolf D. Splettstoesser
e
, Oliwia Makarewicz
b , f
, Heinrich Neubauer
a
, Lisa D. Sprague
a
,
Mathias W. Pletz
b , f
a
Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses (IBIZ), Jena, Germany
b
Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
c
Department of Safety and Quality of Milk and Fish Products, Max Rubner-Institut, Kiel, Germany
d
The Center of Training and Direction of Healthcare Activities, General Hospital of Phutho, Vietnam
e
Department of Microbiology & Hygiene, LADR GmbH, Medical Laboratory Braunschweig, Germany
f
Research Campus Infectognostics, Jena, Germany
g
Department of Health, General Hospital of Phutho, Phutho, Vietnam
a r t i c l e i n f o
Article history:
Received 20 March 2020
Accepted 29 July 2020
Keywo rds:
Acinetobacter baumannii
Intrinsic resistance
Multidrug-resistant
Whole genome sequencing
Germany
Vietnam
a b s t r a c t
Objectives: This study aimed to combine in vitro phenotyping analysis and whole-genome-sequencing
(WGS) to characterise the phenotype and genetic determinants associated with intrinsic resistance in 100
clinical and non-clinical Acinetobacter baumannii strains originating from Germany and Vietnam. More-
over, it aimed to assess whether powdered milk as a food source functions as a potential reservoir of
antibiotic resistance and possesses similar antimicrobial resistance (AMR) genes as in clinical strains iso-
lated from Germany.
Methods: Antimicrobial susceptibility testing was performed using the broth microdilution method and
the minimum inhibitory concentration (MIC) was determined for 18 antibiotics. The WGS data from all
isolates were mapped to intrinsic genes known to be associated with phenotypic AMR.
Results: The highest resistance frequency was observed for chloramphenicol (100%), followed by fos-
fomycin (96%) and cefotaxime (95%). The lowest resistant rates were observed for colistin (3%), trimetho-
prim/sulfamethoxazole (17%), tigecycline (19%), and amikacin (19%). Thirty-five percent of tested strains
displayed resistance to at least one of the carbapenems. Resistance to fluoroquinolones, aminoglycosides,
tigecycline, penicillins, trimethoprim/sulfamethoxazole, and fourth-generation cephalosporins was deter-
mined only in human strains. About one-quarter of isolates (24%) was multidrug-resistant (MDR) and all
were of human origin. Among them, 16 isolates were extensively drug resistant (XDR) and 10 from those
16 isolates showed resistance to all tested antibiotics except colistin. In silico detection of intrinsic AMR
genes revealed the presence of 36 β-lactamases and 24 non- β-lactamase resistance genes. Two colistin-
resistant and 10 ertapenem-resistant strains were isolated from powdered milk produced in Germany.
Thirty-eight AMR genes associated with resistance to antibiotics were found in isolates recovered from
milk powder. Several resistance mechanisms towards many classes of antibiotics existed in A. baumannii
including β-lactamases, multidrug efflux pumps and aminoglycoside-modifying enzymes.
Conclusion: The use of WGS for routine public health surveillance is a reliable method for the rapid de-
tection of emerging AMR in A. baumannii isolates. Milk powder poses a risk to contain MDR Acinetobacter
strains or resistance genes in Germany.
©2020 Elsevier Ltd and International Society of Antimicrobial Chemotherapy. All rights reserved.
Corresponding Author at: Friedrich-Loeffler-Institut, Institute of Bacterial Infec-
tions and Zoonoses (IBIZ), Jena, Germany. E-mail address: gamal.wareth@fli.de (G. Wareth).
https://doi.org/10.1016/j.ijantimicag.2020.106127
0924-8579/© 2020 Elsevier Ltd and International Society of Antimicrobial Chemotherapy. All rights reserved.
2 G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127
1. Introduction
Acinetobacter baumannii ( A. baumannii ) is a Gram-negative op-
portunistic nosocomial pathogen. It is responsible for approxi-
mately 7.7% of all infections in intensive care units [1] , and the ma-
jority ( > 90 %) of A. baumannii isolates in intensive care units are
multidrug-resistant (MDR) and challenging to treat. A. baumannii
can cause a multitude of severe nosocomial infections of the skin
and soft tissue, urinary and respiratory tracts, and bloodstream.
It is also associated with secondary meningitis and ventilator-
associated pneumonia in humans [2-5] . In animals it causes masti-
tis, pneumonia, sepsis, bloodstream, wound and urinary tract in-
fections, septicaemia, bronchopneumonia, neonatal encephalopa-
thy, and eye infections [6-9] . It has been found in humans, an-
imals, foods and the environment. The bacterium is always as-
sociated with severe infection, high mortality rates and massive
economic loss [10] . The World Health Organisation (WHO) has
classified carbapenem-resistant A. baumannii as a priority critical
pathogen [11] regarding the development of novel antibiotics. It is
also a member of the ESKAPE pathogens [12] , a group of bacte-
rial pathogens commonly associated with antibiotic-resistant infec-
tions worldwide: Enterococcus faecium , Staphylococcus aureus , Kleb-
siella pneumoniae , Acinetobacter baumannii , Pseudomonas aerugi-
nosa , and Enterobacter spp. [13] . Spread and dissemination of MDR
A. baumannii has become a public health concern in both, devel-
oping and developed countries, and the prevalence of resistance
towards the last-resort antibiotics such as carbapenems and even
colistin is increasing globally [14] .
Since the late 20
th century, A. baumannii has emerged as a sig-
nificant nosocomial pathogen in Germany [15-17] . The rapid spread
of A. baumannii resistance to carbapenems [18] and to the last-
resort antimicrobial compounds such as colistin and tigecycline
[19] has been reported nationwide and become a major cause of
concern in the modern healthcare system [20] . The prevalence of
MDR A. baumannii has also dramatically increased in other Euro-
pean countries from 15.4% in 2004 to 48.5% in 2014 [21] . Vietnam
has the highest prevalence of resistance amongst Gram-negative
pathogens in the Asia-Pacific region [22] . It is considered one of
the hottest spots of MDR A. baumannii prevalence in Asia. The
prevalence of carbapenem non-susceptible and MDR A. baumannii
has been found to be 80–90% among inpatients [ 23 , 24 ]. A. bauman-
nii possesses the ability to develop intrinsic resistance via reduc-
ing membrane permeability, efflux pump activity and production
of wide varieties of β-lactamases [25] . It is also able to acquire re-
sistance via mutational changes in chromosomal structure as well
as through horizontal gene transfer [25] . Moreover, it presents high
genetic plasticity, which contributes to the accumulation of these
resistance determinants and leads to the MDR pattern [26] .
Dissemination and infection rates of A. baumannii are on the
rise. However, several aspects of resistance development and
pathogenesis are not fully understood. The reservoir for A. bauman-
nii is increasing, not only in the hospitals but also in the commu-
nity and the environment. Its prevalence has gradually increased in
dairy farms and food-producing animals [ 27 , 28 ]. However, it seems
to be neglected in veterinary medicine and food chains, and the
possible risk of human infection concerning animal contact, food
consumption and environmental contamination is unknown.
The current study aimed to combine in vitro phenotyping anal-
ysis and whole-genome-sequencing (WGS) to characterise the phe-
notype and genetic determinants associated with intrinsic resis-
tance in 100 clinical and non-clinical A. baumannii strains origi-
nating from Germany and Vietnam. Moreover, it aimed to assess
whether powdered milk as a food source functions as a potential
reservoir of antibiotic resistance and possesses similar antimicro-
bial resistance (AMR) genes as in clinical strains isolated from Ger-
many.
2. Materials and Methods
2.1. Bacterial isolates and identification
This study used 10 0 non-repetitive A. baumannii strains: 89
isolated in Germany (14 from humans, 71 from milk powder,
two from animals, and two reference strains of A. baumannii
DSM30 0 07 and DSM105126 of German preferential origin), and 11
clinical strains isolated from humans in Vietnam. Human and an-
imal strains were isolated between 2015–2018, while strains ob-
tained from powdered milk were isolated between 2005–2012. The
strains were submitted to the Institute of Bacterial Infections and
Zoonoses (IBIZ, Jena) for confirmation and typing. All strains had
been identified at species level using a combination of Matrix-
Assisted Laser Desorption/Ionization Mass Spectrometry (MALDI-
TOF MS) and the bla OXA-51-like-PCR [29] . WGS was used to con-
firm the identity of the strains by Kraken [30] and Average Nu-
cleotide Identity (ANI) approach [31] .
2.2. Antimicrobial susceptibility testing
The minimum inhibitory concentration (MIC) was determined
via the broth microdilution method in Mueller Hinton II broth us-
ing an automated MICRONAUT-S system (Micronaut, MERLIN Diag-
nostics GmbH, Bornheim-Hersel Germany) according to the man-
ufacturer’s instructions. Briefly, susceptibility testing via this sys-
tem is based on rehydration of antibiotic compounds by adding
a standardized bacterial suspension, and the growth is anal-
ysed using the plate reader after incubation at 37 °C for 18
24 hours. The results were automatically evaluated into sus-
ceptible, intermediate and resistant, with the built-in MICRO-
NAUTS software. Two different plates were used: the Micronaut-
S β-Lactamases and the Micronaut-S-MDR-MRGN-Screening 3
plates. The MIC values were interpreted according to the Clin-
ical and Laboratory Standards Institute (CLSI) breakpoint guide-
lines available for A. baumannii. For A. baumannii, 18 of 26 an-
tibiotics were interpretable by the software. The sensitivity of
all 100 isolates was determined for a panel of the 18 an-
tibiotics: ciprofloxacin (CIP), levofloxacin (LEV), amikacin (AMK),
colistin (COL), chloramphenicol (CMP), fosfomycin (FOS), tigecy-
cline (TGC), trimethoprim/sulfamethoxazole (T/S), piperacillin (PIP),
piperacillin/tazobactam (PIT), cefotaxime (CTX), ceftazidime (CAZ),
ceftazidime/avibactam (CAA), ceftolozane/tazobactam (CTA), ce-
fepime (CEP), imipenem (IMP), meropenem (MER), and ertapenem
(ERT).
2.3. WGS and in silico detection of genes coding for antimicrobial
resistance factors
The genomic DNA was extracted using the High Pure PCR Tem-
plate Preparation Kit (Roche Diagnostics GmbH, Mannheim, Ger-
many) according to the manufacturer’s instructions. The sequenc-
ing library was prepared with the Nextera XT DNA Library Prep
Kit (Illumina, Inc., San Diego, CA, USA) followed by paired-end
sequencing on an Illumina MiSeq sequencer (Illumina, USA). The
Linux-based pipeline WGSBAC that had been developed at the
FLI was used to analyse raw sequencing data ( https://gitlab.com/
FLI _ Bioinfo/WGSBAC ). The pipeline performs quality control of raw
sequencing data by calculating raw sequencing coverage and by us-
ing FastQC (v. 0.11.5) [32] . Results of FastQC were summarised with
MutltiQC [33] . For assembly, the pipeline used SPAdes (v. 3.12.0)
[34] together with the Bayse hammer read correction mode (op-
tion - carefull) and checks the quality of assembled genomes us-
ing QUAST (v. 4.3) [35] . To characterise the assembled genomes,
WGSBAC used ABRIcate (v 0.8.10) ( https://github.com/tseemann/
abricate ). In silico detection of intrinsic AMR genes was performed
G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127 3
by using the Resistance Gene Identifier (RGI) based on the Compre-
hensive Antibiotic Resistance Database (CARD) [36] and the NCBI
databases (Bio Project PRJNA313047). The β-lactamase variants and
non- β-lactamase genes with 100% identity using the A. baumannii
reference genome (Accession ASM74664v1) as input were selected
for further analysis. A few genes with identity < 100% were re-
moved from the analysis.
3. Results
3.1. The phenotypic characteristics and antibiotic susceptibility testing
The phenotypic analysis and antibiotic susceptibility testing
(AST) of the 100 isolates to the panel of 18 antibiotics are shown
in Table 1 . The non-susceptible pattern (resistance and interme-
diate) was identified in isolates of human and animal origins,
and in isolates obtained from milk powder. The highest resis-
tance frequency was found for chloramphenicol (100% of the tested
isolates), followed by fosfomycin (96%) and the third-generation
cephalosporins cefotaxime (95%). Thirty-five isolates (35%) dis-
played resistance to at least one of the carbapenems. Among them,
24 isolates (24%) of human origin from Germany and Vietnam
were resistant to imipenem, meropenem and ertapenem. Addi-
tionally, one reference strain (DSM30 0 07) and ten isolates (10%)
obtained from milk powder produced in Germany were resis-
tant only to ertapenem. About one quarter (25%) of the isolates
displayed resistance to the broad-spectrum β-lactams piperacillin
either alone or with the β-lactamase inhibitor tazobactam. A
quarter of the isolates (24%, 25%) displayed resistance to fluoro-
quinolones, levofloxacin and ciprofloxacin, respectively. The lowest
resistance among the isolates was observed towards colistin (3%),
trimethoprim/sulfamethoxazole (17%), the tetracycline derivative
tigecycline (19%), and the aminoglycoside amikacin (19%). Three
colistin-resistant strains originating from Germany were found.
One of them was a human strain isolated in 2016 that showed
resistance to all tested antibiotics, including colistin and tigecy-
cline, while still sensitive only to trimethoprim/sulfamethoxazole.
The other two strains were isolated from milk powder in 2006
and 2016, and showed resistance only to colistin, fosfomycin,
chloramphenicol, and cefotaxime. Resistance to fluoroquinolones,
aminoglycosides, tigecycline, trimethoprim/sulfamethoxazole, and
to broad-spectrum β-lactam piperacillin and the fourth-generation
cephalosporins was found only in human strains isolated from both
Germany and Vietnam. The 71 strains isolated from milk powder
produced in Germany showed resistance against chloramphenicol
(n = 71), fosfomycin (n = 67), the third-generation cephalosporin
cefotaxime (n = 66), the carbapenem ertapenem (n = 10) , and col-
istin (n = 2). The metadata of the 100 isolates and the antibiotic
profiles are shown in Supplementary Table 1.
3.2. The phenotypic characteristics of MDR and XDR strains
According to the standardised international terminology created
by the European Centre for Disease Control (ECDC) and the Centre
for Disease Control (CDC) and Prevention [37] , A. baumannii is con-
sidered MDR when it is resistant to at least one compound in three
or more antimicrobial categories: aminoglycosides (amikacin), flu-
oroquinolones (ciprofloxacin or levofloxacin), cephalosporins (ce-
fotaxime, ceftazidime, ceftazidime or cefepime), and carbapenems
(imipenem, meropenem or ertapenem). Moreover, the isolate is
considered extensively drug-resistant (XDR) when it is resistant
to at least one agent in all but two or fewer antimicrobial cat-
egories. As shown in Table 2 , 24 isolates (24%) were MDR. All
were of human origin and isolated from Germany (n = 14) and
Vietnam (n = 10). All MDR strains were resistant to carbapen-
ems, fluoroquinolones, chloramphenicol, fosfomycin, piperacillin,
Tabl e 1
Phenotypic analysis of all tested Acinetobacter baumannii isolates (n = 10 0) based on detection of MIC using microdilution methods.
No. Antibiotic Antibiotic class
Tota l number and % of susceptible (S), intermediate
(I) and resistant (R) strains Number of non-susceptible strains based on country and source
Vietnam Germany
S I R
Human
(n = 11 )
Human
(n = 14)
Milk powder
(n = 71)
Animal
(n = 2)
Two DSMZ
reference
1 CIP Fluoroquinolones 75 (75%) 1 (1%) 24 (24%) 10 14 - - 1
2 LEV Fluoroquinolones 76 (76%) 0 24 (24%) 10 14 - - -
3 AMK Aminoglycosides 81 (81%) 0 19 (19%) 9 10 - - -
4 COL Polymyxin B 97 (97%) 0 3 (3%) - 1 2 - -
5 CMP Chloramphenicol 0 5 (5%) 95 (95%) 11 14 71 2 2
6 FOS Produced by Streptomyces 4 (4%) 0 96 (96%) 11 14 67 2 2
7 TGC Tetrac yclines 81 (81%) 7 (7%) 12 (12%) 9 10 - - -
8 T/S Sulfonamides 83 (85%) 0 17 (17%) 9 6 - - 2
9 PIP β-lactams 75 (75%) 0 25 (25%) 10 14 - - 1
10 PIT β-lactams 76 (76%) 0 24 (24%) 10 14 - - -
11 CTX 3
rd
-generation cephalosporins 5 (5%) 14 (14%) 81 (81%) 11 14 66 2 2
12 CAZ 3
rd
-generation cephalosporins 76 (76%) 0 24 (24%) 10 14 - - -
13 CAA 3
rd
-generation cephalosporins 79 (79%) 0 21 (21%) 10 11 - - -
14 CTA 3
rd
-generation cephalosporins 76 (76%) 0 24 (24%) 10 14 - - -
15 CEP 4
th
-generation cephalosporins 76 (76%) 2 (2%) 22 (22%) 10 13 - 1 -
16 IMP Carbapenems 76 (76%) 0 24 (24%) 10 14 - - -
17 MER Carbapenems 76 (76%) 0 24 (24%) 10 14 - - -
18 ERT Carbapenems 65 (65%) 0 35 (35%) 10 14 10 - 1
Abbreviations: CIP, ciprofloxacin; LEV, levofloxacin; AMK, amikacin; COL, colistin; CMP, chloramphenicol; FOS, fosfomycin; TGC, tigecycline; T/S, trimethoprim/sulfamethoxazole; PIP, piperacillin; PIT, piperacillin/tazobactam; CTX,
cefotaxime; CAZ, ceftazidime; CAA, ceftazidime/avibactam; CTA, ceftolozane/tazobactam; CEP, cefepime; IMP, imipenem; MER, meropenem; ERT, ertapenem; DSMZ, Deutsche Sammlung von Mikroorganismen und Zellkulturen
(German Collection of Microorganisms and Cell Cultures).
4 G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127
Tabl e 2
Antibiotic resistance profile of MDR and XDR Acinetobacter baumannii strains toward a panel of 18 antibiotics based on detection of the MIC using microdilution methods.
No. ID Year of isolation Country Origin CIP LEV AMK COL CMP FOS TGC T/S PIP PIT CTX CAZ CAA CTA CEP IMP MER ERT
1 17Y0437 Ab MVZ 2015 Germany Human R R R S R R S R R R R R S R R R R R
2 17Y0438 Ab MVZ 2015 Germany Human R R S S R R R R R R R R R R R R R R
3 17Y0439 Ab MVZ 2015 Germany Human R R R S R R R S R R R R R R R R R R
4 17Y0440 Ab MVZ 2016 Germany Human R R R R R R R S R R R R R R R R R R
5 17Y0441 Ab MVZ 2016 Germany Human R R R S R R I R R R R R S R R R R R
6 17Y0442 Ab MVZ 2016 Germany Human R R R S R R S R R R R R R R I R R R
7 17Y0444 Ab MVZ 2016 Germany Human R R S S R R S S R R R R R R R R R R
8 17Y0446 Ab MVZ 2016 Germany Human R R R S R R I S R R R R S R S R R R
9 17Y0447
Ab MVZ 2016 Germany Human R R S S R R S S R R R R R R R R R R
10 17Y0448 Ab MVZ 2017 Germany Human R R R S R R R R R R R R R R R R R R
11 18Y0258_Ab_TLV 2018 Germany Human R R R S R R I R R R R R R R R R R R
12 18Y0259_Ab_TLV 2018 Germany Human R R R S R R I S R R R R R R R R R R
13 18Y0260_Ab_TLV 2018 Germany Human R R S S R R I S R R R R R R R R R R
14 18Y0261_Ab_TLV 2018 Germany Human R R R S R R R S R R R R R R R R R R
15 18Y0059 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
16 18Y0060 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
17 18Y0061 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
18 18Y0064 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
19 18Y0065 Ab HVU 2017 Vietnam Human R R R S R R R S R R R R R R R R R R
20 18Y0066 Ab HVU 2017 Vietnam Human R R R S R R I R R R R R R R R R R R
21 18Y0067 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
22 18Y0072 Ab HVU 2017 Vietnam Human R R S S R R S S R R R R R R R R R R
23 18Y0074 Ab HVU 2017 Vietnam Human R R R S R R I R R R R R R R R R R R
24 18Y0075 Ab HVU 2017 Vietnam Human R R R S R R R R R R R R R R R R R R
Abbreviations: R, resistant; I, intermediate; S, susceptible; CIP, ciprofloxacin; LEV, levofloxacin; AMK, amikacin; COL, colistin; CMP, chloramphenicol; FOS, fosfomycin; TGC, tigecycline; T/S, trimetho-
prim/Sulfamethoxazole; PIP, piperacillin; PIT, piperacillin/tazobactam; CTX, cefotaxime; CAZ, ceftazidime; CAA, ceftazidime/avibactam; CTA, ceftolozane/tazobactam; CEP, cefepime; IMP, imipenem; MER, meropenem;
ERT, ertapenem.
piperacillin/tazobactam, cefotaxime, ceftazidime, and the novel an-
tibiotics ceftolozane/tazobactam. One of them was sensitive to the
fourth-generation cephalosporin cefepime and five were still sus-
ceptible to amikacin. Sixteen isolates (16%) were classified as XDR
A. baumannii. Among them, ten isolates (10/16) were resistant to
all tested antibiotics except colistin. One human isolate (1/16) from
Germany was resistant to all tested antibiotics, including colistin,
while it was still sensitive only to trimethoprim/sulfamethoxazole.
Five strains (5/16) were resistant to all tested antibiotics and
still sensitive to colistin (n = 5), trimethoprim/sulfamethoxazole
(n = 3), ceftazidime/avibactam (n = 3), tigecycline (n = 2), and
cefepime (n = 1). Metadata of the MDR strains and resistance pro-
files are shown in Table 2 .
3.3. Analysis of intrinsic AMR determinants
The WGS approach identified different AMR-related genes,
among which at least 60 represented intrinsic resistance genes and
51 acquired resistance genes (data not shown). In silico detection
of intrinsic AMR genes using CARD and NCBI databases succeeded
in identifying 36 β-lactamases and ESBLs intrinsic AMR genes
( Table 3 ), as well as 24 non- β-lactamase AMR genes ( Table 4 ).
3.4. Resistance to β-lactams
As shown in Table 3 , three Ambler classes of β-lactamases
(i.e. classes A, C and D) were identified in the current study. At
least three, four and 29 different variants of class A, D and C β-
lactamases were identified, respectively. The members of Ambler
class C β-lactamases were the most predominant genes and en-
compassed 28 Acinetobacter -derived cephalosporinase bla ADC vari-
ants and one A. baumannii AmpC β-lactamase gene. The gene
bla ADC-6 was present in 33 isolates (33%), followed by bla ADC-2
in 24 isolates (24%), bla ADC-52 in 21 isolates (21%), bla ADC-16 in
13 isolates (13%), bla ADC-155 in 12 isolates (12%), bla ADC-156 in
11 isolates (11%), bla ADC-80 in 10 isolates (10%), and bla ADC-73
in nine isolates (9%). The nucleotide sequence of the chromosomal
cephalosporinase gene, which encodes an AmpC β-lactamase, was
found in one strain obtained from milk powder in Germany. Four
AMR genes belonging to the Ambler class D β-lactamases were
identified, the bla OXA-65 gene was present in four isolates (4%),
followed by bla OXA-104, bla OXA.180, and bla OXA.200 ( Table 3 ).
Three AMR genes belonging to Ambler class A β-lactamases were
identified including those of bla TEM-12, which was present in nine
strains (9%), followed by carbenicillin hydrolysing β-lactamase,
bla CARB-16, and bla CARB-14 which were present in three (3%)
and one strain (1%), respectively. Nineteen isolates (19%) had both,
bla ADC-6 and bla ADC-52 of the same Ambler class C, and five iso-
lates (5%) had both, bla ADC-73 and bla TEM-12 of different Ambler
classes. A. baumannii isolates collected from powdered milk sam-
ples from Germany showed wide varieties of β-lactamase AMR
genes from Ambler classes C and D. No AMR genes belonging to
Ambler class B β-lactamase were found. All information regarding
the number of AMR genes, source and origin of strains, as well as
the mechanism of resistance are shown in Table 3 .
3.5. Resistance to aminoglycosides
Among the 100 tested strains, 19 showed resistance to
amikacin. Five aminoglycoside-modifying enzymes (AMEs) genes
were detected. The new subclass of intrinsic aminoglycoside nu-
cleotidyltransferase, ANT(3")-IIa, was widely distributed in human,
animal and milk powder samples. It was found in all tested strains
(100%) followed by the intrinsic aminoglycoside nucleotidyltrans-
ferases aad A and aad A1, which were present in nine strains (9%),
G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127 5
Tabl e 3
β-lactamase and ESBL families intrinsic resistance genes identified in A. baumannii isolates (n = 100) based on WGS data using CARD and NCBI databases.
AMR genes No. (%) Mechanism Source Origin Database
bla ADC.2 23 (23%)/
1 (1%)
Ambler class C
β-lactamases
Milk powder/ human Germany/
Vietnam
CARD
bla ADC.3 1 (1%) Milk powder Germany CARD
bla ADC.4 1 (1%) Animal Germany CARD
bla ADC.5 1 (1%) Animal Germany CARD
bla ADC. 6 33 (33%) Human, milk powder Germany CARD
bla ADC.26 6 (6%) Milk powder Germany NCBI
bla ADC.30 4 (4%) Human Germany NCBI
bla ADC.32 4 (4%) Milk powder Germany NCBI
bla ADC.33 1 (1%) Human Germany NCBI
bla ADC.52 21 (21%) Human, milk powder Germany NCBI
bla ADC.73 9 (9%) Human Germany, Vietnam NCBI
bla ADC.76 4 (4%) Milk powder Germany CARD
bla ADC.78 1 (1%) Human Germany CARD
bla ADC.79 3 (3%) Milk powder Germany CARD
bla ADC.80 10 (10%) Milk powder Germany CARD
bla ADC.81 2 (2%) Human Germany CARD
bla ADC.82 4 (4%) Human Vietnam CARD
bla ADC.90 1 (1%) Milk powder Germany NCBI
bla ADC.107 1 (1%) Milk powder Germany NCBI
bla ADC.154 1 (1%) Animal Germany NCBI
bla ADC.155 12 (12%) Milk powder Germany NCBI
bla ADC.156 10 (10%)/
1 (1%)
Milk powder/ human Germany/
Vietnam
NCBI
bla ADC.157 1 (1%) Milk powder Germany NCBI
bla ADC.158 3 (3%) Animal, milk powder Germany NCBI
bla ADC.165 1 (1%) Milk powder Germany NCBI
bla ADC.16 13
(13%) Milk powder Germany NCBI
bla ADC.169 1 (1%) Milk powder Germany NCBI
blaADC.176 2 (2%) Human Germany NCBI
Ab AmpC_ 1 (1%) Milk powder Germany CARD
bla OXA. 104 2 (2%) Ambler
class D
β-lactamases
Animal, milk powder Germany NCBI
bla OXA.180 2 (2%) Milk powder Germany CARD
bla OXA. 200 1 (1%) Human Germany CARD
bla OXA. 65 4 (4%) Milk powder Germany CARD
bla TEM.12 9 (9%) Ambler
class A
β-lactamases
Human Germany, Vietnam CARD
bla CARB.14 1 (1%) Human Germany CARD
bla CARB.16 3 (3%) Human Germany, Vietnam CARD
Abbreviations: CARD, The Comprehensive Antibiotic Resistance Database; NCBI, National Center for Biotechnology Information.
and aadA2 that was present in one strain (1%). Moreover, the in-
trinsic aminoglycoside acetyltransferase aac A34 was found in one
human isolate from Vietnam ( Table 4 ).
3.6. Antibiotic efflux pumps
Four categories of efflux pumps were found in the current
study, including the multidrug and toxic compound extrusion
(MATE) family, the small multidrug-resistance (SMR) family trans-
porters, the resistance-nodulation-division (RND) superfamily, and
the major facilitator superfamily (MFS). Within these different
pumps the RND ( ade FGH, ade IJK and ade L), MATE ( abe M) and SMR
( abe S) were most frequent and found in 99–100% of isolates. The
RND ( ade N, ade R, ade S, and ade AB) was found in 97%, 76%, 71%, and
66% of strains, respectively. The tetC gene of the MFS transporter
family and the adeC gene of the RND were the genes with the low-
est abundance and were found in one and 22 isolates, respectively.
A. baumannii collected from milk powder showed to have wide va-
rieties of multidrug efflux pump genes ( Table 4 ).
3.7. Resistance to phenicols, glycopeptides and macrolides
Among the 100 tested isolates, one human isolate from Ger-
many harboured the catI gene encoding resistance to pheni-
cols, and one human isolate from Vietnam harboured the gene
brp (MBL) encoding resistance to glycopeptides (e.g. bleomycin).
The mphD gene encoding resistance to macrolide-lincosamide-
streptogramin B (MLS) was found in nine strains (9%) of human
origin from both countries, Germany and Vietnam ( Table 4 ).
4. Discussion
Acinetobacter baumannii has become a notorious pathogen in
human medicine and is implicated in nosocomial and community-
acquired infections worldwide [38] . The general distribution and
reservoirs of A. baumannii are increasing in hospitals, communities,
food chains and the environment [39] . Data on the widespread dis-
tribution of A. baumannii in the farm animal population and foods
is becoming alarming in Germany [40] . However, the risk of human
infection regarding animal contact, food consumption and environ-
mental contamination is unknown. To assess whether milk pow-
der is an essential route for dissemination of resistance in A. bau-
mannii , 71 strains isolated from milk powder obtained from two
powdered milk producers in Germany were included in the study.
Among them, two strains were resistant to colistin and ten strains
were resistant to ertapenem. Both antibiotics are considered last-
resort antibiotics. None of the isolates recovered from milk pow-
der showed an MDR pattern. In total, 38 AMR genes associated
with resistance to antibiotics (22 β-lactamases, 15 multidrug ef-
flux pumps and one AMEs) were found in strains obtained from
powdered milk ( Tables 3 and 4 ). It could not be defined whether
the strains originated from animals, humans or the environment
during milk powder manufacturing. Most of the strains were iso-
lated from end products. As milk is pasteurised before spray dry-
ing and subsequent heat treatment is applied during concentration
6 G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127
Tabl e 4
Non-
β-lactamases intrinsic resistance genes identified in Acinetobacter baumannii isolates (n = 100 ) based on WGS data using CARD and NCBI databases.
Antibiotic family AMR genes No. (%) Mechanism Source Origin Database
AMEs aac A34 1 (1%) ACT Human Vietnam NCBI
aad A 9 (9%) NUT Human Germany, Vietnam NCBI
aad A1 9 (9%) NUT Human Germany, Vietnam CARD
aad A2 1 (1%) NUT Human Vietnam CARD
ANT .3...IIa 100 (100%) NUT Human / animal, milk powder Germany, Vietnam / Germany CARD
MATE Family abe M 99 (99%) Antibiotic efflux Human / animal, milk powder Germany, Vietnam / Germany CARD
SMR family abe S 100 (100%) Human / animal, milk powder Germany, Vietnam / Germany CARD
RND family ade AB 66 (66%) Human / animal, milk powder Germany, Vietnam / Germany CARD
ade
C 22 (22%) Human / milk powder Germany, Vietnam / Germany CARD
ade FGH 99-100 (99-100%) Human / animal, milk powder Germany, Vietnam / Germany CARD
ade IJK 99-100 (99-100%) Human / animal, milk powder Germany, Vietnam / Germany CARD
ade NL 97, 100 (97, 100%) Human / animal, milk powder Germany, Vietnam / Germany CARD
ade R 76 (76%) Human / animal, milk powder Germany, Vietnam / Germany CARD
ade S 71 (71%) Human / animal, milk powder Germany, Vietnam / Germany CARD
MFS family Tetra cyc li ne s tet. C 1 (1%) Human Germany CARD
Phenicols cat I 1 (1%) Enzymes inactivation Human Germany CARD
MLS family mph D 9 (9%) Human Germany, Vietnam CARD
Glycopeptides Determinant of bleomycin resistance 1 (1%) Human Vietnam CARD
Abbreviations: AMEs, aminoglycoside-modifying enzymes; ACT, acetyltransferase; NUT, nucleotidyltransferase; MATE, multidrug and toxic compound extrusi on transporter; SMR, smallest multidrug trans-
porters; RND, resistance-nodulation-cell division antibiotic efflux pump; MFS, major facilitator superfamily; MLS, macrolide-lincosamide-streptogramin B; CARD, The Comprehensive Antibiotic Resistance
Database; NCBI, National Center for Biotechnology Information.
and spray drying, the survival of Acinetobacter is unlikely. However,
processing of contaminated concentrate and rework of filter pow-
der may result in a contaminated end product [41] . In addition, the
powder might be re-contaminated from the environment of the
factory. Acinetobacter exposure can occur via many sources such
as humans, pests, air, packing material, forklift traffic, etc. Conse-
quently, milk powder probably poses a risk for the dissemination
of resistant A. baumannii to humans and further investigations are
required. Powdered infant milk has been identified as a source of
bacterial infection in neonates and infants, especially in hospital
nurseries [41] . The results in the present study are consistent with
previously published observations [ 27 , 42 ].
Clinically, the presence of MDR A. baumannii is of great con-
cern in modern healthcare systems. Detection and characterisation
of AMR genes are gradually moving from PCR to high through-
put identification via sequencing and in silico detection using free-
ware programs and public databases. Although several PCR proto-
cols have been designed to identify resistance genes, these tools
are very limited because they identify only the expected resistance
genes according to the chosen primers. In the current study, the
implementation of high-throughput WGS for the 100 A. baumannii
isolates facilitated rapid and comprehensive AMR detection. In sil-
ico detection of AMR genes based on CARD and NCBI databases re-
vealed the presence of 60 intrinsic resistance genes. Depending on
one database to discriminate the presence of all AMR genes was
insufficient. Moreover, sequencing-based approaches identify only
the known resistance-associated genes but do not provide any in-
formation if these are active. Thus, this study highlights the neces-
sity of combining in vivo phenotyping with in silico detection of
resistance genes from different databases to determine the resis-
tance profiles of A. baumannii isolates.
Different resistance mechanisms towards many classes of an-
tibiotics are known to exist in A. baumannii [43] . Among them, β-
lactamases, multidrug efflux pumps and aminoglycoside-modifying
enzymes were detected in the current study. A. baumannii ex-
hibits intrinsic resistance due to the presence of two chromo-
somally encoded β-lactamases, the bla ADC cephalosporinase and
bla OXA oxacillinase variants, in addition to the natural membrane
impermeability and basal efflux activity [44] . The chromosomally
encoded cephalosporinase gene ( Amp C type) [45] and oxacillinase
bla OXA-69 [46] play an important role in β-lactam resistance in A.
baumannii. Besides, A. baumannii encode 72 further sequence vari-
ants of the intrinsic, non-transferable OXA-51-like enzymes, repre-
senting one of the largest groups of ß-lactamases [47] . In the cur-
rent study, resistance conferred by ß-lactamases was detected by
36 predicted genes; about two-thirds of the genes (n = 22) were
present in strains obtained from milk powder. Inactivation of β-
lactams constitutes an important part of MDR in A. baumannii , es-
pecially for β-lactam antibiotic resistance. All four Ambler classes
of β-lactamases (i.e . classes A, B, C, and D) can be identified in this
pathogen [48] . However, no resistance gene belonging to Ambler
classes B was found. Thirty-six encoding genes initiating resistance
due to β-lactamases were found, and the chromosomally encoded
Acinetobacter -derived cephalosporinase bla ADC gene was predomi-
nant with 28 allelic variants detected. The variants bla ADC-6 and
bla ADC-2 were the most predominant being present in 33% and
24% of the strains, respectively.
Resistance to aminoglycosides in A. baumannii is mainly me-
diated by enzymes, which chemically modify aminoglycosides
(AMEs) [49] . Four-aminoglycoside nucleotidyltransferase (NUT) and
one acetyltransferase (ACT) genes were found. Resistance due to
antibiotic efflux was mediated by 13 RND antibiotic efflux pump-
encoding genes ( adeABC, adeFGH, adeIJK, adeLN and adeRS ). At least
five RND genes were present in 100 % of strains and another four
were present in 99% of strains. The role of RND efflux pump genes
and antimicrobial resistance in A. baumannii is well documented
G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127 7
[50] . The RND genes conferring resistance to clinically essential an-
tibiotics were among the majority of MDR strains in A. bauman-
nii [51] , and were prevalent in the current study. RND proteins
are found in both, prokaryotic and eukaryotic cells. They are sec-
ondary transporters and mediate resistance by pumping the an-
tibiotic from the cell [52] . The present study found one MATE
transporter coded by abe M. The protein encoded by this gene uses
the cationic gradient across the membrane as an energy source
to pump antimicrobial agents from cells [53] . The SMR gene abeS ,
which is restricted to prokaryotic cells [54] , and the MFS gene tetC
were found in 10 0% and 1% of isolates, respectively. Efflux pumps
help bacteria to survive in the presence of high concentrations of
antibiotics but may also confer resistance to antiseptics [55] , and
play an important role in development of AMR A. baumannii [56] .
Although no gene associated with fosfomycin resistance was
found, 96% of strains were resistant to fosfomycin in culture . The
mechanism behind the intrinsic resistance to fosfomycin is not
fully understood in A. baumannii. Despite the fact that fosfomycin
is not the first-line treatment of A. baumannii infections, it has
recently been suggested as a possible treatment for XDR Gram-
negative bacteria [57] . Fosfomycin has been used to treat a wide
variety of bacterial infections in humans. It is not widely used
in animals and its use is limited to treat infectious diseases of
broiler chickens and pigs [58] . Fosfomycin resistance is typically
plasmid-mediated in most members of Enterobacteriaceae . How-
ever, mutants in the ampD and anmK genes of the peptidoglycan-
recycling pathway also contributed to the intrinsic resistance to-
wards fosfomycin in A . baumannii [59] . In general, most A . bauman-
nii strains are intrinsically resistant to chloramphenicol and the re-
sistance is mediated by the major facilitator superfamily (MFS) ef-
flux pump [60] . All isolates (100%) in the current study were chlo-
ramphenicol resistant, while the MFS gene was present only in one
strain. The ade IJK is the second RND efflux system described for
A. baumannii and it contributes to intrinsic resistance to chloram-
phenicol [61] . Besides, inactivation of and trans complementation
with the abe S gene confers low-level resistance to chlorampheni-
col. Among sequenced A. baumannii strains, abe S and ade IJK were
found in 100% of isolates. In Germany, strains resistant to chlo-
ramphenicol have been reported in milk powder obtained from
the same source [27] . A. baumannii strains resistant to fosfomycin
have been described in pets [62] , adult moth flies captured in the
vicinity of hospitals [63] and in patients [ 64 , 65 ]. All A. bauman-
nii strains isolated from humans in Germany (n = 14) and Viet-
nam (n = 10/11) were resistant to imipenem, meropenem and
ertapenem. These results support previous data from Vietnamese
hospitals [ 23 , 24 ], where a substantial increase in the rate of isola-
tion of MDR-carbapenem-resistant A. baumannii was demonstrated .
Until 2005, resistance to carbapenems remained low in Germany
[66] , and the first outbreak of carbapenem-resistant A. bauman-
nii (CRAb) was in 2006 [67] . Since then, carbapenem-resistant
strains have been recovered from several outbreaks every year [68-
72] , and in 2011, CRAb was found in 96.3% of tested strains at
the National Reference Laboratory for Gram-negative nosocomial
pathogens [73] .
5. Conclusion
In conclusion, this study exposed the role of milk powder as
a potential reservoir for antibiotic-resistant A. baumannii strains .
Strains obtained from powdered milk possessed similar AMR genes
as those in clinical isolates from Germany and Vietnam. However, a
direct relationship between milk powder isolates and human iso-
lates has not been established until now. Taken together, the re-
sults of this work emphasise the necessity of combining in vivo
phenotyping and implementing high-throughput WGS for AMR de-
tection and profiling. Almost all isolates of human origin investi-
gated in the current study were MDR and carbapenem-resistant.
The high resistance rates found among clinical isolates are of great
concern and pose an essential threat to healthcare systems. The
current study was more concerned about intrinsic resistance genes
rather than acquired ones. Identification of both intrinsic and hor-
izontally transferable resistance genes could enrich the knowledge
about the dynamics of resistance evolution in A. baumannii , par-
ticularly in cases of foodborne transmission. Multilocus sequence
types (MLST) of isolates, presence of acquired resistance genes and
genes associated with motility and biofilm formation can also ex-
tracted from WGS data. Retrieving these kinds of data is essential
to explore the genetic relatedness of isolates, and to understand
survival and resistance development. Comparing the results of clin-
ical isolates with more strains from animals and environmental
sources is advantageous.
Acknowledgements:
We thank Dr Gernot Schmoock, Johannes Solle, Claudia Grosser
and Birgit Schikowski for excellent technical assistance.
Declarations
Funding: This work was supported by internal funding of the
Friedrich-Loeffler-Institut (FLI). OM and MP were co-funded by
grants from the Federal Ministry of Education and Research (Ger-
many), Grant Numbers: ‘13GW0096D and 01KI1501’.
Competing Interests: None to declare.
Ethical Approval: Not required.
Supplementary materials
Supplementary material associated with this article can be
found in the online version at doi:10.1016/j.ijantimicag.2020.
106127 .
References
[1] Taba h A , Ko ulent i D , Laupland K , Misset B , Valles J , Bruzzi de Carvalho F ,
et al. Characteristics and determinants of outcome of hospital-acquired blood-
stream infections in intensive care units: the EUROBACT international cohort
study. Intensive care medicine 2012;38:1930–45 .
[2] Tian H, Chen L, Wu X, Li F, Ma Y, Cai Y, et al. Infectious complications in severe
acute pancreatitis: pathogens, drug resistance, and status of nosocomial infec-
tion in a university-affiliated teaching hospital. Digestive diseases and sciences
2019. doi: 10.1007/s10620- 019- 05924- 9 .
[3] Metan G, Zarakolu P, Otlu B, Tekin
I, Aytac H, Bolek EC, et al. Emergence of col-
istin and carbapenem-resistant Acinetobacter calcoaceticus-Acinetobacter bau-
mannii (CCR-Acb) complex in a neurological intensive care unit followed by
successful control of the outbreak. Journal of infection and public health 2019
pii: S1876-0341(19)30313-2. doi: 10.1016/j.jiph.2019.09.013 .
[4] Sharma R, Goda R, Borkar SA, Katiyar V, Agarwal S, Kumar A, et al. Out-
come following postneurosurgical Acinetobacter meningitis: an institutional
experience of 72 cases. Neurosurgical focus 2019;47:E8. doi: 10.3171/2019.5.
FOCUS19278 .
[5] Raza MS , Das BK , Goyal V , Lodha R , Chaudhry R , Sood S , et al. Emerging mul-
tidrug resistance isolates of hospital-acquired bacterial meningitis in a tertiary
care centre in North India. Journal of medical microbiology 2019;68:1585–90 .
[6] Eroksuz Y , Otlu B , Eroksuz H , Gursoy NC , Yer li ka ya Z , Akdeniz Incili C ,
et al. Brain abscess and bronchopneumonia caused by Acinetobacter baumannii
in a 2-year-old female sheep. The veterinary quarterly 2018;38:67–71 .
[7] Linz B , Mukhtar N , Shabbir MZ , Rivera I , Ivanov YV , Tahi r Z , et al. Virulent
epidemic pneumonia in sheep caused by the human pathogen Acinetobacter
baumannii . Frontie rs in microbiology 2018;9:2616 .
[8] Vaneechoutte M , Devriese LA , Dijkshoorn L , Lamote B , Deprez P , Verschrae-
gen G , et al. Acinetobacter baumannii -infected vascular catheters collected from
horses in an equine clinic. Journal of clinical microbiology 20 0 0;38:4280–1 .
[9] Ware th G , Abdel-Glil MY , Schmoock G , Steinacker U , Kaspar H , Neubauer H ,
et al. Draft genome sequence of an Acinetobacter baumannii isolate recov-
ered from a horse with conjunctivitis in Germany. Microbiol Resour Announc
2019;8 e01128-19 .
[10] Antunes LC , Visca P , Towner KJ . Acinetobacter baumannii : evolution of a
global
pathogen. Pathogens and disease 2014;71:292–301 .
[11] Tacconelli E , Carrara E , Savoldi A , Harbarth S , Mendelson M , Monnet DL ,
et al. Discovery, research, and development of new antibiotics: the WHO pri-
ority list of antibiotic-resistant bacteria and tuberculosis. The Lancet Infectious
diseases 2018;18:318–27 .
[12] Peterson LR . Bad bugs, no drugs: no ESCAPE revisited. Clinical infectious dis-
eases: an official publication of the Infectious Diseases Society of America
2009;49:992–3 .
8 G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127
[13] Pendleton JN , Gorman SP , Gilmore BF . Clinical relevance of the ESKAPE
pathogens. Expert review of anti-infective therapy 2013;11:297–308 .
[14] Pormohammad A, Mehdinejadiani K, Gholizadeh P, Mohtavinejad N,
Dadashi M, Karimaei S, et al. Global prevalence of colistin resistance in clinical
isolates of Acinetobacter baumannii : A systematic review and meta-analysis.
Microbial pathogenesis 2020;139:103887. doi: 10.1016/j.micpath.2019.103887 .
[15] Graser Y , Klare I , Halle E , Gantenberg R , Buchholz P , Jacobi HD , et al. Epidemi-
ological study of an Acinetobacter baumannii outbreak by using polymerase
chain reaction fingerprinting. Journal of clinical microbiology 1993;31:2417–20
.
[16] Seifert H , Baginski R , Schulze A , Pulverer G . The distribution of Acinetobac-
ter species in clinical culture materials. Zentralblatt fur Bakteriologie: inter-
national journal of medical microbiology 1993;279:544–52 .
[17] Thurm V , Ritter E . Genetic diversity and clonal relationships of Acinetobacter
baumannii strains isolated in a neonatal ward: epidemiological investigations
by allozyme, whole-cell protein and antibiotic resistance analysis. Epidemiol-
ogy and infection 1993;111:491–8 .
[18] Wendel AF , Malecki M , Otchwemah R , Tellez-Castillo CJ , Sakka SG , Mattner F .
One-year molecular surveillance of carbapenem-susceptible A. baumannii on a
German
intensive care unit: diversity or clonality. Antimicrobial resistance and
infection control 2018;7:145 .
[19] Gottig S , Gruber TM , Higgins PG , Wachsmuth M , Seifert H , Kempf VA . Detec-
tion of pan drug-resistant Acinetobacter baumannii in Germany. The Journal of
antimicrobial chemotherapy 2014;69:2578–9 .
[20] Ware th G, Brandt C, Sprague LD, Neubauer H, Pletz MW. Spatio-temporal dis-
tribution of Acinetobacter baumannii in Germany: A comprehensive systematic
review of studies on resistance development in humans (20 0 0-2018). Microor-
ganisms 2020;8(3):375. doi: 10.3390/microorganisms8030375 .
[21] Rodloff AC , Dowzicky MJ . Antimicrobial susceptibility among European
Gram-negative and Gram-positive isolates
collected as part of the Tigecycline
Evaluation and Surveillance Trial (2004-2014). Chemotherapy 2017;62:1–11 .
[22] Kiratisin P , Chongthaleong A , Tan TY , Lagamayo E , Rober ts S , Garcia J ,
et al. Comparative in vitro activity of carbapenems against major Gram-nega-
tive pathogens: results of Asia-Pacific surveillance from the COMPACT II study.
International journal of antimicrobial agents 2012;39:311–16 .
[23] Hoang Quoc C, Nguyen Thi Phuong T, Nguyen Duc H, Tran Le T, Tran Thi Thu
H, Nguyen Tuan S, et al. Carbapenemase genes and multidrug resistance of
Acinetobacter baumannii : A cross sectional study of patients with
pneumonia
in Southern Vietnam. Antibiotics (Basel, Switzerland). 2019;8(3). pii: E148. doi:
10.3390/antibiotics8030148.
[24] Tuan Anh N , Nga TV , Tuan HM , Tuan NS , Y DM , Vinh Chau NV , et al. Molecu-
lar epidemiology and antimicrobial resistance phenotypes of Acinetobacter bau-
mannii isolated from patients in three hospitals in southern Vietnam. Journal
of medical microbiology 2017;66:46–53 .
[25] Lupo A, Haenni M, Madec JY. Antimicrobial resistance in Acinetobacter
spp. and Pseudomonas spp. Microbiology spectrum 2018;6(3). doi: 10.1128/
microbiolspec.ARBA-0 0 07-2017 .
[26] Pagano M , Martins AF , Barth AL . Mobile genetic elements related to car-
bapenem resistance in Acinetobacter baumannii. Brazilian journal of microbi-
ology: [publication of the Brazilian Society for Microbiology] 2016;47:785–92 .
[27] Cho GS , Li B , Rostalsky A , Fiedler G , Rosch N , Igbinosa E , et al. Diversity and
antibiotic susceptibility of Acinetobacter strains from milk powder produced in
Germany. Frontiers in microbiology 2018;9:536 .
[28] Klotz P , Higgins PG , Schaubmar AR , Failing K , Leidner U , Seifert H , et al. Sea-
sonal occurrence and carbapenem susceptibility of bovine Acinetobacter bau-
mannii in Germany. Fro ntiers in microbiology 2019;10:272 .
[29] Turton JF ,
Woodford N , Glover J , Yar de S , Kaufmann ME , Pitt TL . Identification
of Acinetobacter baumannii by detection of the blaOXA-51-like carbapenemase
gene intrinsic to this species. Journal of clinical microbiology 2006;44:2974–6 .
[30] Wood DE , Lu J , Langmead B . Improved metagenomic analysis with Kraken 2.
Genome biology 2019;20:257 .
[31] Richter M , Rossello-Mora R . Shifting the genomic gold standard for the
prokaryotic species definition. Proceedings of the National Academy of Sci-
ences of the United States of America 2009;106:19126–31 .
[32] Andrews S. FastQC: a quality control tool for high throughput sequence
data;
2010. Available online at: http://www.bioinformatics.babraham.ac.uk/projects/
fastqc .
[33] Ewels P , Magnusson M , Lundin S , Kaller M . MultiQC: summarize analysis re-
sults for multiple tools and samples in a single report. Bioinformatics (Oxford,
England) 2016;32:3047–8 .
[34] Bankevich A , Nurk S , Antipov D , Gurevich AA , Dvorkin M , Kulikov AS ,
et al. SPAdes: a new genome assembly algorithm and its applications to sin-
gle-cell sequencing. Journal of computational biology: a journal of computa-
tional molecular cell biology 2012;19:455–77 .
[35] Gurevich A , Saveliev V , Vyahhi N , Tesler G .
QUAST: quality assessment tool for
genome assemblies. Bioinformatics (Oxford, England) 2013;29:1072–5 .
[36] Jia B , Raphenya AR , Alcock B , Waglechner N , Guo P , Tsang KK , et al. CARD
2017: expansion and model-centric curation of the comprehensive antibiotic
resistance database. Nucleic acids research 2017;45:D566–Dd73 .
[37] Magiorakos AP , Srinivasan A , Carey RB , Carmeli Y , Fala gas ME , Giske CG ,
et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant
bacteria: an international expert proposal for interim standard definitions for
acquired resistance. Clinical microbiology and infection: the official publica-
tion of the European Society of
Clinical Microbiology and Infectious Diseases
2012;18:268–81 .
[38] Hamidian M, Emergence Nigro SJ. molecular mechanisms and global
spread of carbapenem-resistant Acinetobacter baumannii . Microbial genomics
2019;5(10):e0 0 0306. doi: 10.1099/mgen.0.0 0 0306 .
[39] Hamouda A . Identification of Acinetobacter baumannii of human and animal
origins by a gene-specific PCR. Current microbiology 2017;74:1118–22 .
[40] Ware th G , Neubauer H , Sprague LD . Acinetobacter baumannii - a neglected
pathogen in veterinary and environmental health in Germany. Veterinary re-
search communications 2019;43:1–6 .
[41] Jacobs C , Braun P , Hammer P . Reservoir and routes of transmission of Enter-
obacter sakazakii ( Cronobacter spp.) in a milk
powder-producing plant. Journal
of dairy science 2011;94:3801–10 .
[42] Carvalheira A , Ferreira V , Silva J , Teixeir a P . Enrichment of Acinetobacter spp.
from food samples. Food microbiology 2016;55:123–7 .
[43] Gordon NC , Wa reham DW . Multidrug-resistant Acinetobacter baumannii : mech-
anisms of virulence and resistance. International journal of antimicrobial
agents 2010;35:219–26 .
[44] Bonomo RA , Szabo D . Mechanisms of multidrug resistance in Acinetobac-
ter species and Pseudomonas aeruginosa . Clinical infectious diseases: an offi-
cial publication of the Infectious Diseases Society of America 2006;43(Suppl
2):S49–56 .
[45] Lin MF , Lan CY . Antimicrobial resistance in Acinetobacter baumannii : From
bench to bedside. Worl d journal of clinical cases 2014;2:787–814 .
[46] Héritier C , Poirel L , Fournier PE , Claverie JM , Raoult D , Nordmann P . Character-
ization of the naturally occurring oxacillinase of Acinetobacter baumannii . An-
timicrobial agents and chemotherapy 2005;49:4174–9 .
[47] Evans BA , Hamouda A , Amyes SG . The rise of carbapenem-resistant Acinetobac-
ter baumannii . Current pharmaceutical design 2013;19:223–38 .
[48] Perez F , Hujer AM , Hujer KM , Decker BK , Rather PN , Bonomo RA . Global chal-
lenge of multidrug-resistant Acinetobacter baumannii
. Antimicrobial agents and
chemotherapy 2007;51:3471–84 .
[49] Cho YJ , Moon DC , Jin JS , Choi CH , Lee YC , Lee JC . Genetic basis of resistance
to aminoglycosides in Acinetobacter spp. and spread of arm A in Acinetobacter
baumannii sequence group 1 in Korean hospitals. Diagnostic microbiology and
infectious disease 2009;64:185–90 .
[50] Leus IV, Weeks JW, Bonifay V, Smith L, Richardson S, Zgurskaya HI. Sub-
strate specificities and efflux efficiencies of RND efflux pumps of Acinetobac-
ter baumannii . Journal of bacteriology 2020;202(8) e0 0 088-20. doi: 10.1128/JB.
0 0 088- 20 .
[51] Abdi SN , Ghotaslou R , Asgharzadeh M , Mehramouz B , Hasani A , Baghi HB ,
et al. Ad eB efflux pump gene knockdown by mRNA mediated peptide nucleic
acid in multidrug resistance Acinetobacter baumannii . Microbial pathogenesis
2019;139:103825 .
[52] Blair JM , Structure Piddock LJ . function and inhibition of RND efflux pumps
in Gram-negative bacteria: an update. Current opinion in microbiology
2009;12:512–19 .
[53] Otsuka M , Matsumoto T , Morimoto R , Arioka S , Omote H , Moriyama Y . A hu-
man transporter protein that mediates the final excretion step for toxic organic
cations. In: Proceedings of the National Academy of Sciences of the United
States of America, 102; 2005. p. 17923–8 .
[54] Bay DC , Rommens KL , Turner RJ . Small multidrug resistance proteins: a mul-
tidrug transporter family that continues to grow. Biochimica et biophysica acta
2008;1778:1814–38 .
[55] Stein C , Vincze S , Kipp F , Makarewicz O , Al Dahouk S , Pletz MW . Carbapen-
em-resistant Klebsiella pneumoniae with low chlorhexidine susceptibility. The
Lancet Infectious diseases 2019;19:932–3 .
[56] Coyne S , Courvalin P , Perichon B . Efflux-mediated antibiotic resistance in
Acinetobacter spp. Antimicrobial agents and chemotherapy 2011;55:947–53 .
[57] Fritzenwanker M , Imirzalioglu C , Herold S , Wagenlehner FM , Zimmer KP ,
Chakraborty T . Treatment Options for Carbapenem- Resistant Gram-Negative
Infections. Deutsches Arzteblatt international 2018;115:345–52 .
[58] Perez DS , Tapia MO , Soraci AL . Fosfomycin: Uses and potentialities in veteri-
nary medicine. Open veterinary journal 2014;4:26–43 .
[59] Gil-Marques ML , Moreno-Martinez P , Costas C , Pachon J , Blazquez J , Mc-
Connell MJ . Peptidoglycan recycling contributes to intrinsic resistance to fos-
fomycin in Acinetobacter baumannii . The Journal of antimicrobial che motherapy
2018;73:2960–8 .
[60] Roca I , Marti S , Espinal P , Martinez
P , Gibert I , Vila J . CraA, a major facilitator
superfamily efflux pump associated with chloramphenicol resistance in Acine-
tobacter baumannii . Antimicrobial agents and chemotherapy 2009;53:4013–14 .
[61] Coyne S , Courvalin P , Périchon B . Efflux-mediated antibiotic resistance in
Acinetobacter spp. Antimicrobial agents and chemotherapy 2011;55:947–53 .
[62] Ewers C , Klotz P , Leidner U , Stamm I , Prenger-Berninghoff E , Gottig S ,
et al. OXA-23 and ISAba1-OXA-66 class D beta-lactamases in Acinetobacter bau-
mannii isolates from companion animals. International journal of antimicrobial
agents 2017;49:37–44 .
[63] Fauld e M , Spiesberger M . Role of
the moth fly Clogmia albipunctata (Diptera:
Psychodinae) as a mechanical vector of bacterial pathogens in German hospi-
tals. The Journal of hospital infection 2013;83:51–60 .
[64] Tafels ki S , Wagner L , Angermair S , Deja M . Therapeutic drug monitoring for
colistin therapy in severe multi-resistant Acinetobacter intracerebral abscess:
A single case study with high-dose colistin and review of literature. SAGE open
medical case reports 2017;5 2050313x17711630 .
[65] Wagner JA , Nenoff P , Handrick W , Renner R , Simon J , Treudler R . [Necrotiz-
ing fasciitis caused by Acinetobacter baumannii: A case report], 62. Der Hau-
tarzt:
Zeitschrift fur Dermatologie, Venerologie, und verwandte Gebiete.; 2011.
p. 128–30 .
G. Wareth, J. Linde and P. Hammer et al. / International Journal of Antimicrobial Agents 56 (2020) 10 6127 9
[66] Brauers J , Frank U , Kresken M , Rodloff AC , Seifert H . Activities of vario us
beta-lactams and beta-lactam/beta-lactamase inhibitor combinations against
Acinetobacter baumannii and Acinetobacter DNA group 3 strains. Clinical mi-
crobiology and infection: the official publication of the European Society of
Clinical Microbiology and Infectious Diseases 2005;11:24–30 .
[67] Kohlen berg A , Brummer S , Higgins PG , Sohr D , Piening BC , de Grahl C ,
et al. Outbreak of carbapenem-resistant Acinetobacter baumannii carrying the
carbapenemase OXA-23 in a German university medical centre. Journal of
medical microbiology 2009;58:1499–507 .
[68] Pfeifer Y , Cho S
, Higgins P , Fahr A , Wichelhaus T , Hunfeld K , et al. Molecu-
lar characterisation and outbreak analysis of multidrug- resistant Acinetobacter
baumannii from German hospitals Abstr 20th European Congress of Clinical
Microbiology and Infectious Diseases Abstr P797; 2010 .
[69] Hischebeth GT , Wimmer MD , Molitor E , Seifert H , Gravius S , Bekeredjian-D-
ing I . Multidrug resistant Acinetobacter baumannii reaches a new frontier: pros-
thetic hip joint infection. Infection 2015;43:95–7 .
[70] Molter G , Seifert H , Mandraka F , Kasper G , Weidmann B , Hornei B , et al. Out-
break of carbapenem-resistant Acinetobacter baumannii in the intensive care
unit: a multi-level strategic management approach. The Journal of hospital in-
fection 2016;92:194–8 .
[71] Rieber H , Frontzek A , Pfeifer Y . Molecular investigation of carbapenem-resis-
tant Acinetobacter spp. from hospitals in North Rhine-Westphalia, Germany.
Microbial drug resistance (Larchmont, NY) 2017;23:25–31 .
[72] Reinheimer C, Kempf VA, Gottig S, Hogardt M, Wichelhaus TA, O’Rourke F,
et al. Multidrug-resistant organisms detected in refugee patients admitted
to a University Hospital, Germany June-December 2015. Euro surveillance
2016;21(2). doi: 10.2807/1560-7917 .
[73] Kaase M . Carbapenemasen bei gramnegativen Erregern in Deutschland. Daten
des Nationalen Referenzzentrums fur gramnegative Krankenhauserreger. Bun-
desgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2012;55:1401–4 .
... Several of these important animal and zoonotic bacteria are also ESKAPE pathogens, an acronym devised to refer to six bacteria (Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) which critically impact human health and exhibit high levels of AMR [68]. Increasing AMR, including carbapenem resistance, in these critical ESKAPE pathogens, can be linked to animal agriculture practices, particularly in LMICs [69,70]. ...
Article
Full-text available
The industrialization of animal agriculture has undoubtedly contributed to the improvement of human well-being by increasing the efficiency of food animal production. At the same time, it has also drastically impacted the natural environment and human society. The One Health initiative emphasizes the interdependency of the health of ecosystems, animals, and humans. In this paper, we discuss some of the most profound consequences of animal agriculture practices from a One Health perspective. More specifically, we focus on impacts to host-microbe interactions by elaborating on how modern animal agriculture affects zoonotic infections, specifically those of bacterial origin, and the concomitant emergence of antimicrobial resistance (AMR). A key question underlying these deeply interconnected issues is how to better prevent, monitor, and manage infections in animal agriculture. To address this, we outline approaches to mitigate the impacts of agricultural bacterial zoonoses and AMR, including the development of novel treatments as well as non-drug approaches comprising integrated surveillance programs and policy and education regarding agricultural practices and antimicrobial stewardship. Finally, we touch upon additional major environmental and health factors impacted by animal agriculture within the One Health context, including animal welfare, food security, food safety, and climate change. Charting how these issues are interwoven to comprise the complex web of animal agriculture's broad impacts on One Health will allow for the development of concerted, multidisciplinary interventions which are truly necessary to tackle these issues from a One Health perspective.
... A. baumanni can be found in several foods such as fruit, vegetables and raw milk, as well as dairy products (Elbehiry et al., 2021). For Wareth et al. (2020), this microorganism causes a series of severe infections in the skin and soft tissues. A. baumannii was recently detected as one of the pathogenic bacteria of some fish species such as Ictalurus punctatus and Channa striatus (Malick et al., 2020). ...
Article
Full-text available
Milk is a rich food, source of several indispensable nutrients, much consumed by the population. Its quality and microbiota are influenced by several factors. The objective of the present study was to determine the microbiota of refrigerated raw milk and processed milk in dairy products in Vale do Taquari - RS, through genetic sequencing. Three types of milk were analyzed in two dairies in the region: raw refrigerated milk, which arrives at the dairies by means of tank trucks, pasteurized milk and milk treated by Ultra High Temperature (UHT). The determination of the milk microbiota was performed through sequencing of the 16S rRNA gene. The results showed that refrigerated raw milk has the highest number of microorganisms in the two dairy products, followed by pasteurized milk and milk sterilized by the UHT process, successively. Dairy processing 2 proved to be more efficient, especially for UHT treated milk, considerably reducing the microbiota. Eighty-seven species of the Bacteria Kingdom were identified and the samples showed considerable microbiological diversity. Psychrotrophic microorganisms such as Kurthia gibsonii were expressive in the samples. Lactic acid bacteria such as Streptococcus macedonicus were found in refrigerated raw milk and pasteurized milk and Streptococcus thermophilus in sterilized milk. Harmful species such as Bacillus cereus group, Aeromonas dhakensis and Acinetobacter haemolyticus were found in UHT treated milk from both dairy products, in addition to Aeromonas caviae, Enterobacter mori and Viridibacillus arenosi. Index terms: Microorganisms; metagenomics; genetic sequencing; milk quality; milk production
Article
Full-text available
ARTICLE INFO ABSTRACT Keywords The purpose of this study is to assess the accuracy of the MALDI TOF technique in identifying Klebsiella spp. Plus, application of MICRONAUT susceptibility testing for detection of MDR klebsiella isolates. A total of 293 samples (233 nasal swabs, 45 milk sample, 10 anal swabs and 5 vaginal swabs) were collected from 258 clinically diseased animals. All samples were examined and all typical colonies were analyzed biochemically and confirmed by MALDI TOF method. All confirmed klebislla spp. isolates were tested for antimicrobial susceptibility to 18 antimicrobials. Typical colonies were confirmed biochemically in 30 samples as klebsiella (10.2%). They were identified as klebsiella spp. (n=24), k. pneumoniae (n=3) and enterobacter spp. (n=3) by MALDI TOF. Antibiotic susceptibility testing of 27 k. pneumoniea strains by using MICRONAUT susceptibility testing discovered that all of the isolated strains were sensitive to 7/16 tested antibiotics (43.75%) amoxicillin/clavulanic acid, ceftiofur, cephalothin, enrofloxacin, gentamicin, spectinomycin, trimethoprim/sulfamethoxazol, complete resistance was observed to only 6 antibiotics, i.e., ampicillin erythromycin , tiamulin, tilmicosin, penicillin G, tulathromycin. All strains showed intermediate susceptible to florfenicol except only one strain. Eighty five percent of the isolates recorded sensitivity to colistin and the remaining show intermediate susceptibility. Ten strains were unsusceptible to tetracycline (6 resistance, 4 intermediate) and the other were susceptible to tetracycline. Klebsiella pneumonia Multidrug resistant (MDR) MALDI TOF MS MICRONAUT Susceptibility testing
Article
Full-text available
Acinetobacter baumannii (A. baumannii) is a highly problematic pathogen with an enormous capacity to acquire or upregulate antibiotic drug resistance determinants. The genomic epidemiology and resistome structure of 46 A. baumannii clinical isolates were studied using whole-genome sequenc-ing. The isolates were chosen based on reduced susceptibility to at least three classes of antimicrobial compounds and were initially identified using MALDI-TOF/MS, followed by polymerase chain reaction amplification of bla OXA-51-like genes. The susceptibility profiles were determined using a broth microdilution assay. Multi-, extensive-, and pan-drug resistance was shown by 34.8%, 63.0%, and 2.2% of the isolates, respectively. These were most susceptible to colistin (95.7%), amikacin, and trimethoprim/sulfamethoxazole (32.6% each), while only 26.1% of isolates were susceptible to tigecycline. In silico multi-locus sequence typing revealed 8 Pasteur and 22 Oxford sequence types (STs) including four novel STs (ST Oxf 2805, 2806, 2807, and 2808). The majority of the isolates belonged to Global Clone (GC) 2 (, and GC7 (2.2%) lineages. An extensive resistome potentially conferring resistance to the majority of the tested antimicrobials was identified in silico. Of all known carbapenem resistance genes, bla OXA-23 was carried by most of the isolates (69.6%), followed by ISAba1-amplified bla ADC (56.5%), bla NDM-1 and bla GES-11 (21.7% each), and bla GES-35 (2.2%) genes. A significant correlation was found between carbapenem resistance and carO mutations, which were evident in 35 (76.0%) isolates. A lower proportion of carbapenem resistance was noted for strains possessing both bla OXA-23-and bla GES-11. Amikacin resistance was most probably mediated by armA, aac(6)-Ib9, and aph(3)-VI, most commonly coexisting in GC2 isolates. No mutations were found in pmrABC or lpxACD operons in the colistin-resistant isolates. Tigecycline resistance was associated with adeS (N268Y) and baeS (A436T) mutations. While the lineage-specific distribution of some genes (e.g., bla ADC and bla OXA-51-like alleles) was evident, some resistance genes, such as bla OXA-23 and sul1, were found in all GCs. The data generated here highlight the contribution of five GCs in A. baumannii infections in Egypt and enable the comprehensive analysis of GC-specific resistomes, thus revealing the dissemination of the carbapenem resistance gene bla OXA-23 in isolates encompassing all GCs.
Article
Full-text available
Acinetobacter baumannii is widely distributed in nature and in hospital settings and is a common pathogen causing various infectious diseases. Currently, the drug resistance rate of A. baumannii has been persistently high, showing a worryingly high resistance rate to various antibiotics commonly used in clinical practice, which greatly limits antibiotic treatment options. Tigecycline and polymyxins show rapid and effective bactericidal activity against CRAB, and they are both widely considered to be the last clinical line of defense against multidrug resistant A. baumannii. This review focuses with interest on the mechanisms of tigecycline resistance in A. baumannii. With the explosive increase in the incidence of tigecycline-resistant A. baumannii, controlling and treating such resistance events has been considered a global challenge. Accordingly, there is a need to systematically investigate the mechanisms of tigecycline resistance in A. baumannii. Currently, the resistance mechanism of A. baumannii to tigecycline is complex and not completely clear. This article reviews the proposed resistance mechanisms of A. baumannii to tigecycline, with a view to providing references for the rational clinical application of tigecycline and the development of new candidate antibiotics.
Article
The assessment of antibiotic resistance and related genes of foodborne Acinetobacter spp. and the analysis of whether they are genetically related to clinical infection-agent strains are crucial in terms of sustainability of food safety. The study at hand investigated antibiotic resistance, aminoglycoside-modifying enzyme (AME), and colistin resistance (PmrA) genes, clonal relationships while evaluating a possible correlation between antibiotic resistance and related genes between 27 foodborne and 50 clinical Acinetobacter spp. in Turkey. Antimicrobial susceptibilities, (AME), PmrA genes, and clonal relatedness of the strains were performed by disc diffusion, PCR, and Pulsed Field gel Electrophoresis (PFGE) methods, respectively. The aph-AI, aph-6, anth (3’’)-I, aadA1, aadB and PmrA genes were found as (24, 48%), (11, 22%), (7, 14%), (1, 2%), (2, 4%), and (46, 92%) respectively in clinical strains. This rate was found as (14, 51.9%), (16, 59.3%), (19, 70.4%), (2, 7.4%), (0, 0%), and (27, 100%) respectively in foodborne isolates. A positive correlation existed between the number of aph-AI gene positivity and trimethoprim-sulfamethoxazole and gentamycin resistance; anth (3’’)-I gene positivity, and colistin resistance; PmrA gene positivity and piperacillin-tazobactam, ceftazidime, meropenem, amikacin, and imipenem resistance in clinical strains (P
Article
Full-text available
Background and Aim: Carbapenem-resistant Acinetobacter baumannii (CRAb) has ability to develop and acquire resistance makes it one of the most critical nosocomial pathogens globally. Whole genome sequemcing (WGS) technology was employed to map genes associated with antimicrobial resistance (AMR),virulene factors and to identify multilocus sequence types (MLST). In order to understand the resistance mechanism for A. baumannii species, this study set out to establish the genetic makeup of the species. Materials and Methods: Whole-genome sequencing (WGS) of A.b4, A.b49 and A.b75 was performed using Illumina MiSeq and the genomes were assembled with SPAdes. ARG-ANNOT, CARD-RGI, VFDB, PHAST, PlasmidFinder were used to analyse all genomes. Results: Genome analysis revealed that Ab4 belongs to ST944, represented singletons that could not be attributed to any, A.b49 belongs to ST1104, represented unique ST.While A.b75 belongs to ST195 which represented known international clones of high risk .Molecular characterisation showed the presence 23 antibiotic resistance genes in all straines of A. baumannii. 12 of them are shared by all 3 strains and 11 are common between A. baumannii 4(ST/944), 49 (ST/1104) and 75(ST/195). The common drug-resistance genes shared by all 3 strains include bla OXA-72 (resistance to carbapenems), ade genes, RND (adeFJK, adeLN & adeR), and SMR (abeS) encoding for efflux pumps. Conclusion: We present WGS analysis of three A. baumannii strains belonging to three different STs. The presence of strains harboring acquired AMR genes makes them more dangerous. Acquired resistance genes and chromosomal gene mutation are successful routes for disseminating AMR determinants among A. baumannii. Identification of chromosomal and plasmid-encoded AMR in the genome of A. baumannii may help understand the mechanism behind the genetic mobilization and spread of AMR genes.
Article
The bacteriophage vB8388 can lyse multi-drug resistant Klebsiella oxytoca strain FK-8388 and maintain stability in a wide range of temperatures (from 4 °C to 80 °C) and pHs (3–11). Bioinformatics analysis showed that vB8388 is a linear double-stranded DNA virus that is 39,750 long with 50.65% G + C content and 44 putative open reading frames (ORFs). Phage vB8388 belongs to the family Autographviridae and possesses a non-contractile tail. The latency period of vB8388 was approximately 20 min. The combination of phage vB8388 and gentamicin, amikacin, or tobramycin could effectively inhibit the growth of K. oxytoca strain FK-8388, with a decrease of more than 4 log units within 12 h in vitro. Phage vB8388 showed a strong synergistic effect with gentamicin that could enhance the anti-biofilm effect of vB8388. The phage + gentamicin combination also showed synergy in vivo in the larval infection model of Galleria mellonella. In conclusion, the findings of this study suggest the potential of phage + antibiotic combination therapy to be used as an alternative therapeutic approach for treating infectious diseases caused by multidrug-resistant bacteria.
Article
Full-text available
Acinetobacter (A.) baumannii has gained global notoriety as a significant nosocomial pathogen because it is frequently associated with multi-drug resistance and hospital-based outbreaks. There is a substantial difference in the incidence of A. baumannii infections between different countries and within Germany. However, its continuous spread within Germany is a matter of concern. A systematic literature search and analysis of the literature published between 2000 and 2018 on A. baumannii in humans was performed. Forty-four studies out of 216 articles met the criteria for inclusion, and were selected and reviewed. The number of published articles is increasing over time gradually. Case reports and outbreak investigations are representing the main body of publications. North Rhine-Westphalia, Hesse and Baden-Wuerttemberg were states with frequent reports. Hospitals in Cologne and Frankfurt were often mentioned as specialized institutions. Multiresistant strains carrying diverse resistance genes were isolated in 13 of the 16 German states. The oxacillinase blaOXA-23-like, intrinsic blaOXA-51-like, blaOXA-58 variant, blaNDM-1, blaGES-11, blaCTX-M and blaTEM are the most predominant resistance traits found in German A. baumannii isolates. Five clonal lineages IC-2, IC-7, IC-1, IC-4 and IC-6 and six sequence types ST22, ST53, ST195, ST218, ST944/ST78 and ST348/ST2 have been reported. Due to multidrug resistance, colistin, tigecycline, aminoglycosides, fosfomycin, ceftazidime/avibactam and ceftolozan/tazobactam were often reported to be the only effective antibiotics left to treat quadruple multi-resistant Gram-negative (4MRGN) A. baumannii. Dissemination and infection rates of A. baumannii are on the rise nationwide. Hence, several aspects of resistance development and pathogenesis are not fully understood yet. Increased awareness, extensive study of mechanisms of resistance and development of alternative strategies for treatment are required. One-Health genomic surveillance is needed to understand the dynamics of spread, to identify the main reservoirs and routes of transmission and to develop targeted intervention strategies
Article
Full-text available
Although Kraken's k-mer-based approach provides a fast taxonomic classification of metagenomic sequence data, its large memory requirements can be limiting for some applications. Kraken 2 improves upon Kraken 1 by reducing memory usage by 85%, allowing greater amounts of reference genomic data to be used, while maintaining high accuracy and increasing speed fivefold. Kraken 2 also introduces a translated search mode, providing increased sensitivity in viral metagenomics analysis.
Article
Full-text available
Here, we announce the draft genome sequence of Acinetobacter baumannii strain 161514, which was recovered from a horse with conjunctivitis, and determine the genetic basis of its antimicrobial resistance phenotype. The genome has a size of 3,839,365 bp and a G+C content of 38.93% and is predicted to contain 3,529 coding sequences. The isolate belongs to sequence type 462 (ST462) according to the Oxford scheme (Abaumannii1) and to ST46 according to the Pasteur scheme (Abaumannii2).
Article
Full-text available
Background Secondary infection is an important factor affecting mortality and quality of life in patients with severe acute pancreatitis. The characteristics of secondary infection, which are well known to clinicians, need to be re-examined in detail, and their understanding among clinicians needs to be updated accordingly. Aim This study aims to investigate the characteristics and drug resistance of pathogens causing severe acute pancreatitis (SAP) secondary infection, to objectively present infection situation, and to provide reference for improved clinical management. Methods A retrospective analysis was performed on 55 consecutive patients with SAP who developed secondary infection with an accurate evidence of bacterial/fungal culture from 2016 to 2018. The statistics included the spectrum and distribution of pathogens, the drug resistance of main pathogens, and associations between multiple infectious parameters and mortality. Results A total of 181 strains of pathogens were isolated from (peri)pancreas; bloodstream; and respiratory, urinary, and biliary systems in 55 patients. The strains included 98 g-negative bacteria, 58 g-positive bacteria, and 25 fungi. Bloodstream infection (36.5%) was the most frequent infectious complication, followed by (peri)pancreatic infection (32.0%). Acinetobacter baumannii, Pseudomonas aeruginosa, Klebsiella pneumoniae, Escherichia coli, and Stenotrophomonas maltophilia were predominant among gram-negative bacteria. Gram-positive bacterial infections were mainly caused by Enterococcus faecium and Staphylococcus spp. Fungal infections were predominantly caused by Candida spp. The drug resistance of pathogens causing SAP secondary infection was generally higher than the surveillance level. Patients in the death group were older (55 ± 13 years vs. 46 ± 14 years; p = 0.039) and had longer intensive care unit (ICU) stay (14 vs. 8; p = 0.026) than those in the survival group. A. baumannii infection (68.4% vs. 33%; p = 0.013), number of pathogens ≥ 4 (10 vs. 6; p = 0.005), pancreatic infection (14 vs. 15, p = 0.024), and urinary infection (8 vs. 5; p = 0.019) were significantly associated with mortality. Conclusion Gram-negative bacteria are the main pathogens causing SAP secondary infection, in which nosocomial infections play a major role. The drug resistance profile of gram-negative bacteria is seriously threatening, and the commonly used antibiotics in SAP are gradually losing their effectiveness. Much attention should be paid to the rational use of antibiotics, and strategies should be established for infection prevention in SAP.
Article
Full-text available
Background: Colistin and carbapenem-resistant Acinetobacter calcoaceticus- Acinetobacter baumannii complex (CCR-Acb complex) was isolated from two consecutive patients in the neurological intensive care unit (NICU). An urgent reaction to this desperate situation was required. Patients and methods: Screening cultures were taken from the other patients sharing the NICU with index patients and repeated periodically. NICU was closed for new admissions. Infection control precautions (ICP) such as hand hygiene, cohorting patients colonized with CCR-Acb complex, cohorting the staff caring for these patients, daily bathing with chlorhexidine gluconate impregnated clothes, using gowns when contacting with patients and patient care area, and sodium hypochlorite tablets for environmental cleaning were enforced. Results: Screening cultures revealed carbapenem-resistant Acb complex in 12 out of 32 patients and 8 of them were colonized with CCR-Acb complex. NICU was opened for new admissions one month later. No further new cases with CCR-Acb complex were detected by screening cultures after 6 weeks with enforcement of ICP. Moreover, the rate of nosocomial infections caused by other multi-drug resistant Gram-negative bacilli (MDR-GNB) decreased significantly when rates before and after closing the NICU were compared. Conclusion: ICP were effective not only to limit the spread of CCR-Acb complex but also decreased the incidence of other MDR-GNB infections when applied adequately.
Article
Full-text available
Purpose. Acute bacterial meningitis continues to be a potentially life threatening condition. Hospital-acquired meningitis is rapidly increasing and adding an immense burden to the health system due to the emergence of multidrug resistance isolates. The purpose of this study is to find the antibiotic susceptibility pattern of the bacteria detected from hospital- and community-acquired meningitis.Methodology. A total of 400 Cerebrospinal fluid (CSF) samples from the suspected meningitis cases were collected and processed for cell count, biochemical examination, Gram staining, latex agglutination and culture. Bacteria grown on blood, chocolate and Mac-conkey agar were identified by matrix-assisted laser desorption/ionization-time of flight. Antibiotic susceptibility tests were performed as per Clinical and Laboratory Standard Institute guidelines.Results. Of the isolates, most prevalent Gram negative organisms in hospital-acquired bacterial meningitis were Escherichia coli 13 (27.08 %), Acinetobacter baumannii 12 (25 %), Klebsiella pneumoniae 5 (10.42 %), Pseudomonas aeruginosa 4 (8.33 %) and Gram positive organisms were Staphylococcus aureus 4 (8.33 %), Enterococcus faecium 3 (6.25 %) and CONS 2 (4.16 %). Streptococcus pneumoniae 3 (6.25 %) was the predominant organism in community-acquired bacterial meningitis. All the Gram negative isolates were multidrug resistance. Only colistin and imipenem were effective antibiotics against them. Likewise Gram positive organisms were susceptible to most of the antibiotics tested. However, E. faecium was only susceptible to Vanco+Teicoplanin.Conclusion. In hospital-acquired bacterial meningitis, multidrug resistance Gram negative bacteria are a huge challenge for the treatment of patients. Hence, antimicrobial stewardship should be followed to counteract with the emerging multidrug resistance isolates.
Article
Full-text available
Acinetobacter baumannii is a nosocomial pathogen that has emerged as a global threat because of high levels of resistance to many antibiotics, particularly those considered to be last-resort antibiotics, such as carbapenems. Although alterations in the efflux pump and outer membrane proteins can cause carbapenem resistance, the main mechanism is the acquisition of carbapenem-hydrolyzing oxacillinase-encoding genes. Of these, oxa23 is by far the most widespread in most countries, while oxa24 and oxa58 appear to be dominant in specific regions. Historically, much of the global spread of carbapenem resistance has been due to the dissemination of two major clones, known as global clones 1 and 2, although new lineages are now common in some parts of the world. The analysis of all publicly available genome sequences performed here indicates that ST2, ST1, ST79 and ST25 account for over 71 % of all genomes sequenced to date, with ST2 by far the most dominant type and oxa23 the most widespread carbapenem resistance determinant globally, regardless of clonal type. Whilst this highlights the global spread of ST1 and ST2, and the dominance of oxa23 in both clones, it could also be a result of preferential selection of carbapenem-resistant strains, which mainly belong to the two major clones. Furthermore, ~70 % of the sequenced strains have been isolated from five countries, namely the USA, PR China, Australia, Thailand and Pakistan, with only a limited number from other countries. These genomes are a vital resource, but it is currently difficult to draw an accurate global picture of this important superbug, highlighting the need for more comprehensive genome sequence data and genomic analysis.
Article
Introduction Acinetobacter baumannii antimicrobial resistance is a public health concern in developing and developed countries, especially in the hospital setting. Understanding the antibiotic resistance profile can help to provide better guidelines for the prescription of appropriate antibiotics, reduction of antibiotic resistance, and introducing new and effective treatment options. Method Using the PRISMA guidelines, databases of PubMed, Embase, and Cochrane Library were searched systematically from January 1, 2000, to January 1, 2018. All statistical analyses were carried out via Comprehensive Meta-Analysis Software Version 2.0 (Biostat, Englewood, NJ). Depending on the heterogeneity test, either random or fix effect models were used for determining the pooled prevalence of drug resistance. Result A total of 150 studies were included from 41 countries of six different WHO regional offices worldwide. The highest and the lowest rate of resistance were observed for cefotaxime (99%, 95% CI: 95–99.9) in Africa and colistin (1.1%, 95% CI: 0.3–4.5) in Western Pacific, respectively. Lebanon (17.5%, 95% CI: 16–19) and China (12%, 95% CI: 3.5–32.5) had the highest and Germany (0.2%, 95% CI: 0–2.5) had the lowest rate of resistance for colistin. Conclusion: Our analysis showed that prevalence and rate of increased colistin resistance in South-East Asia and Eastern Mediterranean countries are higher than other regions of the world. Therefore, the establishment of appropriate antibiotic usage guidelines should be essential in these countries.
Article
Multidrug-resistant Acinetobacter baumannii isolates cause critical problems in health-care environments. AdeABC is a resistance-nodulation-cell division (RND)-type multidrug efflux pump conferring resistance to clinically essential antibiotics in A. baumannii, such as ciprofloxacin. This study aimed to target adeB gene with antisense peptide nucleic acid (PNA) and investigate its effect on resistance to antibiotics. NCBI database was used to design appropriate PNA to target adeB gene, by connecting PNA to mRNA, the translation of mRNA can be prevented. Three clinical isolates and A. baumannii ATCC 17978 were treated with the designed PNA by electroporation and competence procedure. Minimum Inhibitory concentration (MIC) of ciprofloxacin, colistin, and tetracycline were determined by microbroth dilution method. In addition, the expression level of adeB gene was measured by quantitative real-time PCR (qRT-PCR). Isolates used in this study had mutations in gyrA and parC genes corresponding to resistance to ciprofloxacin. MIC of resistance to ciprofloxacin after treatment with PNA was reduced from32 μg/ml to16 μg/ml in A. baumannii ATCC 17978 isolate. Susceptibility level of tetracycline, in the 2 clinical isolates was decreased from 64 μg/ml to 32 μg/ml and in the other isolate was reduced from128 μg/ml to 64 μg/ml. The expression level of adeB gene was decreased in A. baumannii ATCC 17978 (P > 0.01) but not in clinical isolate (P = 0.107). Findings of the present study indicate overexpression of adeB efflux pump has extra effect on resistance to antibiotics in isolates with a defined mechanism of resistance. Antisense technology is a feasible technique to suppress the function of these genes, which may be further exploited to control multidrug-resistant isolates.