Next Article in Journal
Neuroprotective Effect of Morin Hydrate against Attention-Deficit/Hyperactivity Disorder (ADHD) Induced by MSG and/or Protein Malnutrition in Rat Pups: Effect on Oxidative/Monoamines/Inflammatory Balance and Apoptosis
Next Article in Special Issue
Healthcare Facilities as Potential Reservoirs of Antimicrobial Resistant Klebsiella pneumoniae: An Emerging Concern to Public Health in Bangladesh
Previous Article in Journal
A Study on Repositioning Nalidixic Acid via Lanthanide Complexation: Synthesis, Characterization, Cytotoxicity and DNA/Protein Binding Studies
Previous Article in Special Issue
Synthesis of Functionalized N-(4-Bromophenyl)furan-2-carboxamides via Suzuki-Miyaura Cross-Coupling: Anti-Bacterial Activities against Clinically Isolated Drug Resistant A. baumannii, K. pneumoniae, E. cloacae and MRSA and Its Validation via a Computational Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Co-Occurrence of β-Lactam and Aminoglycoside Resistance Determinants among Clinical and Environmental Isolates of Klebsiella pneumoniae and Escherichia coli: A Genomic Approach

1
Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
2
Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
3
Microbiology and Parasitology Department, Soba University Hospital, University of Khartoum, Khartoum 11115, Sudan
4
Embryonic Stem Cells Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
5
Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environmental and Products, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(8), 1011; https://0-doi-org.brum.beds.ac.uk/10.3390/ph15081011
Submission received: 20 June 2022 / Revised: 11 August 2022 / Accepted: 15 August 2022 / Published: 17 August 2022

Abstract

:
The presence of antimicrobial-resistance genes (ARGs) in mobile genetic elements (MGEs) facilitates the rapid development and dissemination of multidrug-resistant bacteria, which represents a serious problem for human health. This is a One Health study which aims to investigate the co-occurrence of antimicrobial resistance determinants among clinical and environmental isolates of K. pneumoniae and E. coli. Various bioinformatics tools were used to elucidate the bacterial strains’ ID, resistome, virulome, MGEs, and phylogeny for 42 isolates obtained from hospitalized patients (n = 20) and environmental sites (including fresh vegetables, fruits, and drinking water) (n = 22). The multilocus sequence typing (MLST) showed that K. pneumoniae belonged to ten sequence types (STs) while the E. coli belonged to seventeen STs. Multidrug-resistant isolates harbored β-lactam, aminoglycoside resistance determinants, and MGE were detected circulating in the environment (drinking water, fresh vegetables, and fruits) and in patients hospitalized with postoperative infections, neonatal sepsis, and urinary tract infection. Four K. pneumoniae environmental isolates (7E, 16EE, 1KE, and 19KE) were multidrug-resistant and were positive for different beta-lactam and aminoglycoside resistance determinants. blaCTX-M-15 in brackets of ISEc 9 and Tn 3 transposases was detected in isolates circulating in the pediatrics unit of Soba hospital and the environment. This study documented the presence of bacterial isolates harboring a similar pattern of antimicrobial resistance determinants circulating in hospitals and environments. A rapid response is needed from stakeholders to initiate a program for infection prevention and control measures to detect such clones disseminated in the communities and hospitals.

1. Introduction

Antimicrobial resistance in Enterobacterales, especially K. pneumoniae and E. coli, is a critical threat to public health [1,2]. K. pneumoniae and E. coli contribute to the rapid evolution of antibiotic-resistance genes due to genomic plasticity [3]. They have the ability to acquire, accumulate, and disseminate the MGEs associated with antimicrobial resistance such as plasmids and transposons [4,5]. Studying MDR mechanisms and the sequencing of ARGs in these organisms is critical to understanding both the molecular mechanisms of resistance and the basis of their dissemination [6,7].
There is a growing concern regarding controlling the global development and spreading of antibiotic-resistant pathogens, especially for MDR bacteria that produce Extended Spectrum β-Lactamases (ESBLs) and carbapenemases [8]. β-lactam antibiotics represent the most common drug class of antimicrobial drugs with broad clinical implications [9]. The β-lactamases produced by the Enterobacteriaceae family, particularly K. pneumoniae and E. coli, are hydrolytic enzymes that confer bacterial resistance to β-lactam antibiotics such as penicillin, cephalosporin, and carbapenem families that are common antimicrobial drugs used all around the world [2]. Nosocomial MDR K. pneumoniae and E. coli have been considered the most frequent agents of infectious diseases and significant threats to patients in hospital settings in Sudan [10,11,12]. The presence of antimicrobial-resistance genes in MGEs of the environment and clinical strains facilitate the rapid development and dissemination of MDR bacteria and thus represent a serious problem for human health [13]. Different transposable elements are associated with the spread of antimicrobial-resistance genes between bacteria, including plasmids, transposons, and integrons [14]. There are many plasmids associated with ARGs in the Enterobacterales group (e.g., IncF, IncA/C, IncH, IncP, IncL/M, or IncX, etc.). The IncF plasmid is more frequently described as carrying genes encodes for resistance to ESBLs, carbapenems, aminoglycosides, or fluoroquinolones, while IncI2, IncX4, and IncP plasmids encode for gene resistance to colistin. The IncHI1 and IncHI2 plasmids are more frequently reported in MDR isolates [15]. Drug-resistant bacteria can be transferred to humans from the surrounding contaminated environment, including soil, animals, water, vegetables, and fruits [16,17]. The contamination of drinking water and daily consumable products with antimicrobial-resistant bacteria represent a serious problem due to their ease of transmission to human settlements, hospitals, and crowded areas [18,19]. The spread of MDR bacteria in the environment and health settings has led to increased mortality and morbidity rates and is now considered one of the most serious public health threats [20]. Khartoum is one of the most crowded, polluted cities in Africa [21,22], which represents a suitable medium for the dissemination of antimicrobial-resistant bacteria. Sudan suffers from the inappropriate use of antibiotics; most of the antibiotics are frequently sold over the counter, and even without a medical prescription [23,24]. Recently, different studies conducted in Khartoum state have documented the dissemination of antibiotic-resistant bacteria in the environment such as banknotes [25] and drinking water [26,27]. Whole-genome sequencing (WGS) and plasmid analysis are essential for accurate epidemiologic tracking of hospital outbreaks and routine surveillance. Recently, WGS has played an important role in speeding up microbial diagnosis and predicting antimicrobial resistance [28]. There is no information regarding the phenotypic and genetic characteristics of the environmental MDR Klebsiella spp. and E. coli isolates and their relation to nosocomial isolates. The present study aims to characterize and to identify the MGEs, antimicrobial-resistance genes, and STs of K. pneumoniae and E. coli spreading in clinical and environmental isolates using WGS.

2. Results

2.1. Isolates

A total of 42 isolates were identified and confirmed as K. pneumoniae and E. coli by phenotypic tests and the PubMLST database. The isolates were as follows: clinical isolates (K. pneumoniae = 7, E. coli = 13) and environmental isolates (K. pneumoniae =13, E. coli = 9) (Table 1).

2.2. Phenotypic Antimicrobial Susceptibility Testing

Antimicrobial susceptibility testing revealed variation in the resistance patterns (Table 2); all studied isolates were susceptible to carbapenems, while some isolates were resistant to beta-lactam, aminoglycoside, and ciprofloxacin (Table 2). Sequencing results confirmed the presence of different types of ARGs. All (100%) clinical and environmental K. pneumoniae isolates (n = 20) harbored beta-lactam, aminoglycoside, and fluoroquinolones resistance genes while 13 (59%) of E. coli clinical and environmental isolates (n = 22) harbored beta-lactam, aminoglycoside, and fluoroquinolones, and about 72% harbored other resistance genes (Table 2).

2.3. Genomic Sequence Features and Strains Typing

All assembled sequences had greater than 88× coverage, the E. coli had an average GC content of 50.65%, N50 of 106319, 395 contigs, and coding sequences (CDS) of 4914. K. pneumoniae had an average GC content of 57.16%, N50 of 158435, 249 contigs, and 5414.3 CDS (Supplementary File, Table S1).
The MLST showed that K. pneumoniae isolates belonged to ten STs, while the E. coli isolates belonged to seventeen STs. ST45 was detected in eight K. pneumoniae isolates (six were clinical and two environmental isolates) and ST405 was detected in five clinical E. coli isolates. The most common strain of E. coli in urinary tract infection (UTI) patients was ST405, while K. pneumoniae ST45 was the most common in patients with septicemia. The list of all different STs was presented in Table 1. One K. pneumoniae isolate (23KE) showed one novel allele (phoE_4) in the phoE gene and was assigned a novel sequence type with ID:1504, while isolates 16KE and 17KE were identified with novel alleles and assigned a novel sequence type with IDs, 22233 and 22234. Isolate 14EE was submitted to the Pasteur MLST database and assigned with the ID: 1508.

2.4. Detection of Antibiotic-Resistance Genes (ARGs)

Analysis of resistome revealed that 40% (17/42) of the isolates harbored one or more of the aminoglycoside-resistant genes (aph(6)-Id and aph(3″)-Ib, aac(6′)-Ib-cr, aadA5, and rmtB). These genes were detected in ten E. coli isolates (one environmental and nine clinical) and seven K. pneumoniae isolates (one environmental and six clinical).
The β-lactam-resistant determinants were detected in 79% (33/42) of the isolates including 13 E. coli (2 environmental and 11 clinical) and 20 K. pneumoniae (13 environmental and 7 clinical).
K. pneumoniae isolates possessed ompK37 19 (95%), ompK36 17(85%), and blaSHV group 18 (90%), and other β-lactamases including blaCTX-M-15 (n = 7), blaOXA-1 (n = 6), and blaTEM-1D (n = 2). Aminoglycoside-resistant genes (aac(6)-Ib-cr and aac(3)-IIa) were present in seven K. pneumoniae isolates. The OqxB, OqxA, and acrR genes contributing to fluoroquinolones resistance were detected together in 18 K. pneumoniae isolates. Analysis of resistome associated with E. coli isolates revealed that blaCTX-M-15 was the most dominant gene (n = 8). Other β-lactamases including blaNDM-5 (n = 4), blaOXA-1 (n = 4), blaTEM-1D (n = 5), blaTEM-35 (n = 2), blaCMY (n = 2), blaDHA-1 (n = 1), and blaSHV-12 (n = 1) were also detected. Aminoglycoside- and fluoroquinolone-resistant genes were detected in E. coli isolates: aac(6)-Ib-cr, aph(3)-Ib, mdf(A), qnrB4, rmtB, and qepA4. Genes resistant to fosfomycin, tetracycline, macrolides, and trimethoprim were identified in both K. pneumoniae and E. coli, and they include: fosA, tet(A), tet(B), catB3, dfrA12, dfrA14, dfrA17, floR sul1, sul2, and mph(A), in addition to efflux genes such as qacE and sitABCD (Table 3).
Regarding the relationship of isolate sequence types (STs) with the presence of resistant genes, six clinical K. pneumoniae ST45 were reported with different resistant genes (blaOXA-1, blaCTX-M-15, blaSHV-1, ompK37, and ompK36). Four environmental strains of K. pneumoniae (1KE, 12KE, 17KE, and 21KE) were harboring blaSHV-11. Other blaSHV variants including blaSHV-1, blaSHV-26, blaSHV-38, and blaSHV-71 were also reported in environmental strains (Table 3). blaNDM-5 and blaCTX-M-15 genes were most common in E. coli ST405 strains. Four environmental isolates (7E, 16EE, 1KE, and 19KE) were MDR and were positive for different beta-lactamase genes (Table 3). The K. pneumoniae strain (ST1504) was isolated from drinking water at the Khartoum locality, and it was positive for blaSHV-1, ompK37, ompK36, acrR, OqxB, OqxA, and fosA genes. Mutations associated with fluoroquinolone and fosfomycin resistance were investigated, and amino acid substitutions were reported in genes involved in resistance to fosfomycin (cyaA, UhpT, and GlpT) and fluoroquinolone (parC, gyrA, and marR). The cyaA gene mutation (S352T) was observed in eight clinical isolates of E. coli; UhpT (E350Q) was observed in two isolates of E. coli (clinical and environment) and seventeen of K. pneumoniae. The GlpT (E448K) was present only in 20 E. coli isolates (90%). The parC (S80I) and gyrA (D87N, S83L) were reported only in eight E. coli isolates. Mutations (Y137H, G103S) in the marR gene were common in E. coli, reported in 15 isolates (Table S2). Genes associated with antibiotic efflux, antibiotic target alteration, and protection were also investigated; nine efflux pump genes (acrB, emrB, mdtG, AcrE, cpxA, evgA, mdtE, TolC, and mdtH) were dominantly and exclusively reported in E. coli, while in K. pneumoniae LptD, oqxA, K. pneumoniae KpnF, K. pneumoniae KpnH, K. pneumoniae KpnG, adeF, and CRP, efflux pump genes were dominant and exclusive. Most isolates of K. pneumoniae harbored ArnT (19 isolates) and eptB (16 isolates) genes, which are associated with antibiotic target protection (Table S3).

2.5. Analysis of Mobile Genetic Elements (MGEs)

Regarding the analysis of MGEs (plasmid, transposases, and virulence factors), they were present in most clinical isolates with few in environmental strains. The most prevalent plasmid in E. coli isolates were IncFIA (n = 12), IncFIB (AP001918) (n = 11), Col (BS512) (n = 9), and IncY (n = 6), while the prevalent K. pneumonia-encoding plasmids were IncFIB(K) (n = 12), IncFII(K) (n = 9), and Col440I (n = 10). Table 4 presents the distributions of plasmids on the study isolates; more details about transposases and virulence genes are in the additional file, Table S4.

2.6. Co-Occurrence of ARGs with Transposases

The mobile element finder revealed the co-occurrence of ARGs, insertion sequences, transposons, and plasmids, at both environmental and clinical isolates. Some genes clustered together at the same contig (Table S5). blaCTX-M-15 was located on contigs bracketed by ISEc9 and Tn3 transposases, and detected in seven isolates; one clinical E. coli (8EP) isolate and six K. pneumoniae, one environmental (1KE) (Figure 1) and five clinical isolates (5KP, 6KP, 7KP, 13KP, and 14KP) (Figures S1–S5), were detected from patients with septicemia (four of them were neonates). They were characterized by the presence of sulfonamide-resistant dihydropteroate synthase genes (Sul2) and aminoglycoside-resistant genes (aph(6)-Id and aph(3)-Ib) bracketed by IS5075 and IS91 insertion sequences (Table S5 and Figure 2).
As shown in Figure 3, IS6100 clustered with ARGs and was observed similarly in five MDR isolates including E. coli (1EP, 8EP, 10EP, 11EP, and 27EP); three of them were isolated from a wound of a postoperative infection and two from urinary tract infection (UTI) patients. Four isolates of E. coli harbored a set of MDR genes cassettes (mph(A), qacE, dfrA17, sul1, aadA5), in addition to IS6100, located closely at the same contigs.
Six MGEs were detected in the same cassette adjacent to each other in one MDR water isolate (16EE), containing po111 plasmid flanked by ISKpn19 and IS102 insertion sequences, and two ARGs (floR and qnrS1) which fell in brackets of ISKpn19 and ISVsa3.
A K. pneumoniae fruit isolate (12KE) showed the presence of aminoglycoside-resistant genes (aph(3)-Ib, aph(6)-Id) and the chromosomally mediated nhaA gene flanked by three transposases genes (two Tn3 and one 1S110) (Table S5 and Figure 4).
Two MDR isolates of E. coli (20EP and 25EP) were isolated from the urine of patients with chronic kidney disease, characterized by the presence of rmtB and blaTEM-1B, and were harbored in Tn2 transposon. These two isolates were located in the same clade as shown in the phylogenetic tree (Figure 5). Three environmental isolates of K. pneumoniae (15KE, 16KE, and 20KE) showed the co-existence of insertion sequences (ISKpn14, ISKpn41, and ISEhe3) and plasmids (IncHI1B, IncFIB(Mar), and repA) in the same contigs (Table S5).

2.7. Phylogenetic Analysis

The phylogenetic tree and metadata revealed that most isolates were clustered according to the source of the isolate (environmental or clinical), while few were mixed. Isolate 13EE from the environment and isolate 28EP from the clinical source clustered together. Water isolates (1EE, 4EE, 6EE, 7EE, 14EE, 13EE, and 16EE) and three clinical isolates (8EP, 12EP, and 28EP) were clustered together. Isolates 1KE and 11EE clustered with isolates 10EP and 27EP from patients with a wound infection and UTI, respectively. Isolates 12KE from fruits and 15KE from the table surface were closely related to isolate (13KP) from patients with septicemia; isolates 12KE and 13KE belong to ST45. The four clinical isolates (5KP, 6KP, 7KP, 12KP, 13KP, and 14KP) were closely related to isolate 19KE from fruits, and all belong to ST45 (Figure 5 and Figure 6).

3. Discussion

K. pneumoniae and E. coli have been associated with epidemic and endemic nosocomial infections caused by multidrug infections, mainly ESBL-producing bacteria in Sudan and worldwide [2,10,29]. β-lactamases-producing K. pneumoniae and E. coli, especially TEM, CTX-M, and SHV type, are the most prevalent species that have spread globally within the hospital and environment [30,31,32,33,34]. ESBL-producing bacteria spread through consumption or cross-contamination. Environmental exposure produces considerably high ESBL-positive E. coli levels in vegetables and foods due to insufficient hygiene in irrigation water systems [8,34,35]. In this study, all K. pneumoniae and a few numbers of E. coli isolated from water and vegetables were ESBL producers, which gives evidence of the spreading of ESBL genes in our environment. This could be as a result of the strong selection pressure exerted by the indiscriminate use of beta-lactam antibiotics in our community [36].
blaCTX-M-15 was documented recently as the most prevalent ESBL gene in Sudan, which was reported in environmental and clinical samples in previous studies [26,27]. This study is in line with these studies, in which we detected the blaCTX-M-15 gene bracketed by ISEc9 and Tn3 transposases in six clinical and one environmental isolate. These transposases play a crucial role in gene transfer and could be one of the reasons behind horizontal gene transfers [37]. Similar to our findings, Madni et al. [14] recently reported K. pneumoniae with blaCTX-M-15 bracketed by ISEc9 and Tn3 from South African patients. Four of blaCTX-M-15, ISEc9-, and Tn3-positive isolates were from neonatal sepsis in the pediatric ward at Soba University Hospital caused by K. pneumoniae ST45, suggesting a hospital-acquired infection which is more common in Sudan hospitals [10,38]. Similarly, the mph(A), qacE, dfrA17, sul1, and aadA5 ARGs have co-existed closely with IS6100 transposase in five MDR E. coli isolates, three being from a wound of a postoperative infection. A similar set of genes cluster with transposons in MDR E. coli was reported by Roy Chowdhury et al. [39] from Australian patients with UTIs.
blaCTX-M-15-positive isolates were phenotypically resistant to cephalosporin; this finding is expected because this gene was documented with a high affinity to hydrolyze cephalosporin [13]. Here, the blaCTX-M-15 was detected among 15 isolates from clinical and environmental isolates, which is in agreement with a study conducted in Khartoum, which revealed the blaCTX-M-15 gene in isolates recovered from diverse non-clinical niches and belonging to different Enterobacteriaceae species [40,41]. CTX-M-15 β-lactamases are mainly encoded in IncFII plasmids, the host plasmid of the high-risk clone K. pneumoniae, and play an important role in its international dissemination [42]. Moreover, different studies from Tanzania, Nigeria, and Tunisia reported the presence of blaCTX-M-15 in IncF-type plasmids from clinical and environmental isolates [33,43].
E. coli ST38 is an international high-risk clone responsible for the spreading of the OXA-48 gene [44]. In this study, ST38 was detected in one MDR E. coli isolate from cerebrospinal fluid (CSF), and this isolate was positive for blaCTX-M-15 and blaTEM-35.
In this study, we documented one E. coli strain (ST120) containing blaDHA-1 and qnrB4 associates with four plasmids: IncFIA, IncFIB (AP001918), IncFIC(FII), and IncI1-I(Alpha). The co-occurrence of blaDHA-1 and qnrB4 with IncL/M and IncR plasmids has been reported in Europe and Asia, in Serratia marcescens, E. coli, K. pneumoniae (ST11 and ST37), and Enterobacter cloacae ST528 [45,46,47]. Additionally, blaNDM-5 was found on three E. coli ST405 isolates in association with IncFII-I ncFIB[pB171] plasmid replicons, similar to a study from Japan [48].
E. coli and K. pneumoniae have the potential to increase the widespread transmission of ARGs via MGEs through the processes of natural competence, transformation, and plasmid trans conjugation that can occur in any environment [8]. There are many plasmids associated with antimicrobial resistance genes in the Enterobacterial group (e.g., IncF, IncA/C, IncH, IncP, IncL/M, or IncX, etc.) [15]. Some of these plasmids, such as IncF, are encoded with specific resistance genes such as ESBLs, carbapenems, aminoglycosides, or fluoroquinolones, while IncI2, IncX4, and IncP plasmids are associated with resistance to colistin encoded by the mcr-1 gene; IncHI1 and IncHI2 plasmids are reported to be associated with MDR [15]. IncI1-Ic and IncFIA-FIB plasmid types were reported in co-existence with multiple types of ESBL-encoding genes (blaCTX-M-3, blaTEM-1, and blaSHV-12) in K. pneumoniae from North India [49]. This is similar to our finding in which multiple ESBL-encoding genes (blaOXA-1, blaCTX-M-15, blaSHV-26, blaSHV-11, and blaSHV-38), (blaNDM-5, blaCTX-M-15, and blaTEM-1B) and (blaNDM-5, blaCTX-M-15, and blaOXA-1) have been documented with IncI1-Ic and IncFIA-FIB.
The fosfomycin-resistant genes (fosA and fosA5) were detected as co-resistance in ESBL-producing K pneumoniae, this finding is similar to many studies reporting co-resistance of fosfomycin and ESBL in plasmid-mediated resistance [50,51,52].
The genes encoding for sulfonamide-resistant dihydropteroate synthase (Sul2), which is consistently co-carried with aminoglycoside-resistant genes (aph(6)-Id and aph(3″)-Ib) [13], were detected in this study, bracketed by two transposase (IS5075 and IS91) in 5 K. pneumoniae isolates. These transposases could be the vehicle for the horizontal gene transfer and dissemination of these genes in our isolates [53]. Interestingly, we reported six MGEs clustered together in one MDR E. coli (16EE) from water, which contained a cluster of sul2, dfrA14, and tet(A) genes, and a po111 plasmid flanked by ISKpn19 and IS102 insertion sequences, and two ARGs: the quinolone resistance determinant (qnrS1) and florfenicol resistance gene (floR); these fall in the brackets of ISKpn19 and ISVsa3. The presence of ARGs between transposable elements will help in the easy transposition of genes and can mediate their mobility between drug-sensitive organisms [54]. In later studies from the Khartoum locality, they noted an increase in the presence of ARGs in E. coli isolated from drinking water [40,41].
Future studies from different geographical regions in Sudan with large sample sizes should be considered to better understand the possible role of the environment in the dissemination of ARGs.

4. Materials and Methods

4.1. Bacterial Strains and Antimicrobial Susceptibility Testing

A total of 42 consecutive isolates were collected randomly between March and July 2021 from hospitalized patients in Soba University Hospital (n = 20) and the environment (n = 22). The clinical isolates were collected at the clinical microbiology laboratory of Soba University Hospital as part of their routine clinical procedure; the isolates were from various samples, including blood, urine, pus, wounds, cerebrospinal fluid, and catheter tips. The environmental isolates were from swabs obtained from vegetables, markets, and water stations (Table 1). Environmental samples were collected and processed according to published protocols [40,55]. Isolates were primarily identified using Gram staining and standard biochemical tests, which include citrate utilization test, glucose and lactose fermentation in Kligler iron agar tubes, urease and indole [56], and according to their colors and growth characteristics on chromogenic media. Phenotypic antimicrobial susceptibility testing (AST) was performed using the disk diffusion method against an antibiotic panel including amoxicillin-clavulanate (30 µg), cefuroxime (30 µg), ceftriaxone (30 µg), ceftazidime (30 µg), cephalexin (30 µg), meropenem (10 µg), imipenem (10 µg), amikacin (30 µg), gentamicin (10 µg), ciprofloxacin (5 µg), trimethoprim-sulfamethoxazole (25 µg), and nitrofurantoin (300 μg). American Type Culture Collection (ATCC) strains including P. aeruginosa ATCC 27853 and E. coli ATCC 25922 were used as quality controls. Results were interpreted according to the Clinical and Laboratory Standards Institute (CLSI) guidelines [57].

4.2. Whole-Genome Sequencing and Molecular Analysis

Genomic DNA was extracted from overnight bacterial growth using the Guanidine hydrochloride method as described by Sabeel et al. [58]. The integrity and quantity of extracted DNA was estimated by gel electrophoresis and Nanodrop, Qubit (Thermo Scientific, Waltham, MA, USA).
WGS was performed by Novogene Company (China) using an Illumina HiSeq 2500 platform (Illumina, San Diego, CA, USA), 2 × 150 bp paired-end reads were generated with 100 X coverage. Trimmomatic 0.36 [59] was used to remove low-quality reads, adapters, and reads containing n > 10%. ContEst16S was used to screen assembled genomes for contamination by either cells or DNAs from other organisms. De novo assembly was conducted by Velvet v1.2.10 [60] and PATRIC (Pathosystems Resource Integration Center) server. The assembled bacterial genomes were identified in species, strain levels, and STs using MLST 2.0 and PubMLST [61] databases. The novel sequence types (ST) were assigned by the Pasteur MLST database. Genome annotation was achieved by the RAST server [62] and NCBI Prokaryotic Genome Annotation Pipeline (PGAP) [63]. Antimicrobial resistomes were predicted using Resistance Gene Identifier (RGI) and ResFinder [64]. Virulence genes were also investigated using VirulenceFinder 2.0 (Center for Genomic Epidemiology, DTU, Lyngby, Denmark) and Mobile Element Finder [65]. Plasmids, insertion sequences and transposons, and Human pathogen probability were predicted by Plasmid Finder 2.1, IS Finder, and Pathogen Finder [66], respectively. Resistant genes and MGEs maps were visualized by Geneious Prime 2021 trial version. The assembled contigs were submitted to GenBank under the Bioproject PRJNA767482.

4.3. Phylogenetic Analysis

The phylogenetic tree for E. coli and K. pneumoniae was constructed via tools available through the Galaxy platform [67], the assembled contigs were annotated using Prokka [68], and the generated GFF3 format was used as input for Roary [69] with a default minimum of 95% identity for BLASTp to estimate the pan-genome. The aligned core genes generated from Roary were used as input for RaXML [70] for the reconstruction of the phylogenetic tree. The phylogenetic graph was visualized by Phandango using files generated from RAxML (raxml tree) and Roary (gene presence absence cvs).

5. Conclusions

This study sheds light on the spread of ARGs and/or MGEs in clinical and environmental isolates of E. coli and K. pneumoniae. blaCTX-M-15 bracketed between ISEc9 and Tn3 transposases is disseminated in environmental and clinical isolates. Four K. pneumoniae strains possessed blaCTX-M-15 bracketed by ISEc9 and Tn3, Sul2, and contained aph(6)-Id and aph(3″)-Ib) bracketed by IS5075, and IS91 belonging to ST45, which were potentially endemic in the delivery room or pediatrics unit of Soba University Hospital. A similar pattern of the clustering of IS6100 with MDR genes cassettes (mph(A), qacE, dfrA17, sul1, and aadA5) was observed in E. coli isolated from patients with postoperative wound infections at Soba University Hospital, indicating possible hospital-acquired infections. This finding necessitates a rapid response from stakeholders to initiate a program for infection prevention and control measures to detect such clones disseminated in communities and hospitals.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/ph15081011/s1. Table S1: Genomic features of E. coli and K. pneumonia isolates; Table S2: Point mutations associated with antibiotic resistance; Table S3: Genes associated with antibiotic efflux, antibiotic target alteration and protection; Table S4: Virulence and transposases genes detected in K. pneumonia and E. coli isolates; Table S5: Co-occurrence of antimicrobial-resistance genes and transposases; Figure S1: Map of different antibiotic-resistant genes in clinical K. pneumonia (5KP); Map of different antibiotic resistant genes, transposases, and plasmid, clustered in clinical K. pneumonia (7KP); Figure S3: Map of different antibiotic resistant genes, transposases, and plasmid, clustered in clinical K. pneumonia (6KP); Figure S4: Map of different antibiotic resistant genes, transposases, and plasmid, clustered in clinical K. pneumonia (13KP) isolate; Figure S5. Map of different antibiotic resistant genes, transposases, and plasmid, clustered in contig 156 of clinical K. pneumonia (14KP) isolate.

Author Contributions

H.N.A., Conceptualization, supervision, bioinformatics analysis, Writing—review and editing, funding acquisition; H.S.E.: Data acquisition, carried out the microbiological analysis, Writing—review and editing; O.B. and I.K.: Methodology, software, data curation, formal analysis, Writing—review and editing; F.A.A.: Methodology, software, data curation, writing—review and editing. M.S.N.: Methodology, analysis, writing—review and editing. S.H.: Investigations, resources, writing—review and editing. K.C.: Validation, supervision, visualization, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

The authors extend their appreciation to the Deputyship for Research and Innovation, Ministry of Education in Saudi Arabia, for funding this research work through the project number IFPRC-072-130-2020 and King Abdulaziz University DSR, Jeddah, Saudi Arabia.

Institutional Review Board Statement

This study was approved by the Ethics Committee of the SUST (No: DSR-IEC3-01-21).

Informed Consent Statement

Not applicable because we are collecting sample remnants without patient’s identifiable information.

Data Availability Statement

The data of this project were submitted to GenBank under the Bioproject PRJNA767482, and in the additional files.

Acknowledgments

The authors extend their appreciation to the Deputyship for Research and Innovation, Ministry of Education in Saudi Arabia, for funding this research work through the project number IFPRC-072-130-2020 and King Abdulaziz University DSR, Jeddah, Saudi Arabia. We thank the Institute Pasteur teams for the curation and maintenance of the BIGSdb-Pasteur databases at http://bigsdb.pasteur.fr, for their role in assigning numbers for the novel strains.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Antimicrobial Resistance: Global Report on Surveillance; World Health Organization: Geneva, Switzerland, 2014. [Google Scholar]
  2. Tacconelli, E.; Magrini, N.; Kahlmeter, G.; Singh, N. Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics. World Health Organ. 2017, 27, 318–327. [Google Scholar]
  3. Holt, K.E.; Wertheim, H.; Zadoks, R.N.; Baker, S.; Whitehouse, C.A.; Dance, D.; Jenney, A.; Connor, T.R.; Hsu, L.Y.; Severin, J. Genomic analysis of diversity, population structure, virulence, and antimicrobial resistance in Klebsiella pneumoniae, an urgent threat to public health. Proc. Natl. Acad. Sci. USA 2015, 112, E3574–E3581. [Google Scholar] [CrossRef] [PubMed]
  4. Evans, D.R.; Griffith, M.P.; Sundermann, A.J.; Shutt, K.A.; Saul, M.I.; Mustapha, M.M.; Marsh, J.W.; Cooper, V.S.; Harrison, L.H.; Van Tyne, D. Systematic detection of horizontal gene transfer across genera among multidrug-resistant bacteria in a single hospital. Elife 2020, 9, e53886. [Google Scholar] [CrossRef] [PubMed]
  5. Xanthopoulou, K.; Carattoli, A.; Wille, J.; Biehl, L.M.; Rohde, H.; Farowski, F.; Krut, O.; Villa, L.; Feudi, C.; Seifert, H. Antibiotic Resistance and Mobile Genetic Elements in Extensively Drug-Resistant Klebsiella pneumoniae Sequence Type 147 Recovered from Germany. Antibiotics 2020, 9, 675. [Google Scholar] [CrossRef]
  6. Blair, J.M.; Webber, M.A.; Baylay, A.J.; Ogbolu, D.O.; Piddock, L.J. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 2015, 13, 42–51. [Google Scholar] [CrossRef]
  7. Ahmed, A.; Sirag, B.; Raees, F.; Kidir, E.-S.; Ali, T.; Rahman, M.A.; Ashgar, S.; Barhameen, A.; Elsawy, A.; Mostafa, A. Bacterial whole genome sequencing as powerful tool for hospital molecular epidemiology: Acinetobacter baumannii as a model. Clin. Microbiol. Infect. Dis. 2016, 1, 10–16. [Google Scholar] [CrossRef]
  8. Stadler, T.; Meinel, D.; Aguilar-Bultet, L.; Huisman, J.S.; Schindler, R.; Egli, A.; Seth-Smith, H.M.; Eichenberger, L.; Brodmann, P.; Hübner, P. Transmission of ESBL-producing Enterobacteriaceae and their mobile genetic elements—Identification of sources by whole genome sequencing: Study protocol for an observational study in Switzerland. BMJ Open 2018, 8, e021823. [Google Scholar] [CrossRef]
  9. Palzkill, T. Structural and mechanistic basis for extended-spectrum drug-resistance mutations in altering the specificity of TEM, CTX-M, and KPC β-lactamases. Front. Mol. Biosci. 2018, 5, 16. [Google Scholar] [CrossRef]
  10. Elbadawi, H.S.; Elhag, K.M.; Mahgoub, E.; Altayb, H.N.; Ntoumi, F.; Elton, L.; McHugh, T.D.; Tembo, J.; Ippolito, G.; Osman, A.Y. Detection and characterization of carbapenem resistant Gram-negative bacilli isolates recovered from hospitalized patients at Soba University Hospital, Sudan. BMC Microbiol. 2021, 21, 1–9. [Google Scholar] [CrossRef]
  11. Osman, E.A.; El-Amin, N.I.; Al-Hassan, L.L.; Mukhtar, M. Multiclonal spread of Klebsiella pneumoniae across hospitals in Khartoum, Sudan. J. Glob. Antimicrob. Resist. 2021, 24, 241–245. [Google Scholar] [CrossRef]
  12. Mohamed, S.B.; Kambal, S.; Munir, A.; Abdalla, N.; Hassan, M.; Hamad, A.; Mohammed, S.; Ahmed, F.; Hamid, O.; Ismail, A. First Whole-Genome Sequence of a Highly Resistant Klebsiella pneumoniae Sequence Type 14 Strain Isolated from Sudan. Microbiol. Resour. Announc. 2019, 8, e00552-19. [Google Scholar] [CrossRef]
  13. Mbanga, J.; Amoako, D.G.; Abia, A.L.; Allam, M.; Ismail, A.; Essack, S.Y. Genomic Insights of Multidrug-Resistant Escherichia coli From Wastewater Sources and Their Association With Clinical Pathogens in South Africa. Front. Vet. Sci. 2021, 8, 137. [Google Scholar] [CrossRef]
  14. Madni, O.; Amoako, D.G.; Abia, A.L.K.; Rout, J.; Essack, S.Y. Genomic Investigation of Carbapenem-Resistant Klebsiella pneumonia Colonization in an Intensive Care Unit in South Africa. Genes 2021, 12, 951. [Google Scholar] [CrossRef]
  15. Rozwandowicz, M.; Brouwer, M.; Fischer, J.; Wagenaar, J.; Gonzalez-Zorn, B.; Guerra, B.; Mevius, D.; Hordijk, J. Plasmids carrying antimicrobial resistance genes in Enterobacteriaceae. J. Antimicrob. Chemother. 2018, 73, 1121–1137. [Google Scholar] [CrossRef]
  16. Mayor, S. Doctors to get real time data to support antibiotic prescribing and reduce resistance. BMJ 2019, 25, l406. [Google Scholar] [CrossRef]
  17. Balali, G.I.; Yar, D.D.; Dela, V.G.A.; Adjei-Kusi, P. Microbial contamination, an increasing threat to the consumption of fresh fruits and vegetables in today’s world. Int. J. Microbiol. 2020, 2020, 3029295. [Google Scholar] [CrossRef]
  18. Kraemer, S.A.; Ramachandran, A.; Perron, G.G. Antibiotic pollution in the environment: From microbial ecology to public policy. Microorganisms 2019, 7, 180. [Google Scholar] [CrossRef]
  19. Laxminarayan, R.; Duse, A.; Wattal, C.; Zaidi, A.K.; Wertheim, H.F.; Sumpradit, N.; Vlieghe, E.; Hara, G.L.; Gould, I.M.; Goossens, H. Antibiotic resistance—The need for global solutions. Lancet Infect. Dis. 2013, 13, 1057–1098. [Google Scholar] [CrossRef]
  20. Van Duin, D.; Paterson, D.L. Multidrug-resistant bacteria in the community: Trends and lessons learned. Infect. Dis. Clin. 2016, 30, 377–390. [Google Scholar] [CrossRef]
  21. Elhadary, Y.; Ali, S. A new trend in urban housing: Gated communities in Khartoum, Sudan. Am. J. Sociol. Res. 2017, 7, 45–55. [Google Scholar]
  22. Gatari, M.J. Air pollution over East Africa. In Proceedings of the First International Workshop on Climate Variability over Africa, Alexandria, Egypt, 15–26 May 2005. [Google Scholar]
  23. Kheder, S.I. Physcians knowledge and perception of antimicrobial resistance: A survey in Khartoum Stata Hospital settings. Br. J. Pharmaceut. Res. 2013, 3, 347–362. [Google Scholar] [CrossRef]
  24. Musa, M.M.Y.A. Medicine Prices, Availability and Affordability in Sudan. In Partial Fulfillment of Master of Sciences of Health Economics and Health Care Management; Chulalongkorn University: Bangkok, Thailand, 2013. [Google Scholar]
  25. Alfadil, A.; Ahmed, N.; Mohamed, M.S.; Ali, M.M.; El Nima, E.A.I. Characterization of pathogenic bacteria isolated from Sudanese banknotes and determination of their resistance profile. Int. J. Microbiol. 2018, 2018, 4375164. [Google Scholar] [CrossRef]
  26. Mohammed, S.A.S.; Musa, A.; Mohammed, A.A.; Mohammed, H. CTX-M B-lactamase–producing Escherichia coli in Sudan tertiary hospitals: Detection genotypes variants and bioinformatics analysis. Int. J. Med. Biomed. Stud. 2019, 3, 146–157. [Google Scholar]
  27. Altayb, H.N.; Salih, E.K.; Moglad, E.H. Molecular detection of beta-lactamase blaCTX-M group 1 in Escherichia coli isolated from drinking water in Khartoum State. J. Water Health 2020, 18, 1091–1097. [Google Scholar] [CrossRef]
  28. Runcharoen, C.; Moradigaravand, D.; Blane, B.; Paksanont, S.; Thammachote, J.; Anun, S.; Parkhill, J.; Chantratita, N.; Peacock, S.J. Whole genome sequencing reveals high-resolution epidemiological links between clinical and environmental Klebsiella pneumoniae. Genome Med. 2017, 9, 1–10. [Google Scholar] [CrossRef]
  29. Malik, I.; Elhag, K.; Gustaw, K. Characterisation of extended-spectrum β-lactamases among multidrug resistant Enterobacteriaceae from Sudan. J. Pure Appl. Microbiol. 2019, 13, 8. [Google Scholar]
  30. Ahmed, O.B.; Omar, A.O.; Asghar, A.H.; Elhassan, M.M.; Al-Munawwarah, A.-M.; Arabia, S. Prevalence of TEM, SHV and CTX-M genes in Escherichia coli and Klebsiella spp Urinary Isolates from Sudan with confirmed ESBL phenotype. Life Sci. J. 2013, 10, 191–195. [Google Scholar]
  31. Bougnom, B.; Thiele-Bruhn, S.; Ricci, V.; Zongo, C.; Piddock, L. Raw wastewater irrigation for urban agriculture in three African cities increases the abundance of transferable antibiotic resistance genes in soil, including those encoding extended spectrum β-lactamases (ESBLs). Sci. Total Environ. 2020, 698, 134201. [Google Scholar] [CrossRef]
  32. Yaici, L.; Haenni, M.; Métayer, V.; Saras, E.; Zekar, F.M.; Ayad, M.; Touati, A.; Madec, J.-Y. Spread of ESBL/AmpC-producing Escherichia coli and Klebsiella pneumoniae in the community through ready-to-eat sandwiches in Algeria. Int. J. Food Microbiol. 2017, 245, 66–72. [Google Scholar] [CrossRef]
  33. Moremi, N.; Manda, E.V.; Falgenhauer, L.; Ghosh, H.; Imirzalioglu, C.; Matee, M.; Chakraborty, T.; Mshana, S.E. Predominance of CTX-M-15 among ESBL producers from environment and fish gut from the shores of Lake Victoria in Mwanza, Tanzania. Front. Microbiol. 2016, 7, 1862. [Google Scholar] [CrossRef]
  34. Ojer-Usoz, E.; González, D.; Vitas, A.I. Clonal diversity of ESBL-producing Escherichia coli isolated from environmental, human and food samples. Int. J. Environ. Res. Public Health 2017, 14, 676. [Google Scholar] [CrossRef] [PubMed]
  35. van Hoek, A.H.; Veenman, C.; van Overbeek, W.M.; Lynch, G.; de Roda Husman, A.M.; Blaak, H. Prevalence and characterization of ESBL-and AmpC-producing Enterobacteriaceae on retail vegetables. Int. J. Food Microbiol. 2015, 204, 1–8. [Google Scholar] [CrossRef] [PubMed]
  36. Falagas, M.E.; Bliziotis, I.A. Pandrug-resistant Gram-negative bacteria: The dawn of the post-antibiotic era? Int. J. Antimicrob. Agents 2007, 29, 630–636. [Google Scholar] [CrossRef] [PubMed]
  37. Darphorn, T.S.; Bel, K.; Koenders-van Sint Anneland, B.B.; Brul, S.; Ter Kuile, B.H. Antibiotic resistance plasmid composition and architecture in Escherichia coli isolates from meat. Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
  38. Babiker, W.; Ahmed, A.; Babiker, T.; Ibrahim, E.; Almugadam, B. Prevalence and Causes of Neonatal Sepsis in Soba University Hospital, Sudan. Med. Microbiol. Rep. 2018, 3, 11–13. [Google Scholar]
  39. Chowdhury, P.R.; McKinnon, J.; Liu, M.; Djordjevic, S.P. Multidrug resistant uropathogenic Escherichia coli ST405 with a novel, composite IS26 transposon in a unique chromosomal location. Front. Microbiol. 2019, 9, 3212. [Google Scholar] [CrossRef] [PubMed]
  40. Mahmoud, N.E.; Altayb, H.N.; Gurashi, R.M. Detection of Carbapenem-Resistant Genes in Escherichia coli Isolated from Drinking Water in Khartoum, Sudan. J. Environ. Public Health 2020, 2020, 2571293. [Google Scholar] [CrossRef]
  41. Moglad, E.; Adam, O.J.; Alnosh, M.; Altayb, H. Detection of virulence genes of diarrheagenic Escherichia coli strains from drinking water in Khartoum State. J. Water Health 2020, jwh2020097. [Google Scholar] [CrossRef]
  42. Stercz, B.; Farkas, F.B.; Tóth, Á.; Gajdács, M.; Domokos, J.; Horváth, V.; Ostorházi, E.; Makra, N.; Kocsis, B.; Juhász, J. The influence of antibiotics on transitory resistome during gut colonization with CTX-M-15 and OXA-162 producing Klebsiella pneumoniae ST15. Sci. Rep. 2021, 11, 1–10. [Google Scholar] [CrossRef]
  43. Alonso, C.; Zarazaga, M.; Ben Sallem, R.; Jouini, A.; Ben Slama, K.; Torres, C. Antibiotic resistance in Escherichia coli in husbandry animals: The African perspective. Lett. Appl. Microbiol. 2017, 64, 318–334. [Google Scholar] [CrossRef]
  44. Pitout, J.D.; Peirano, G.; Kock, M.M.; Strydom, K.-A.; Matsumura, Y. The global ascendency of OXA-48-type carbapenemases. Clin. Microbiol. Rev. 2019, 33, e00102–e00119. [Google Scholar] [CrossRef]
  45. Mata, C.; Miró, E.; Mirelis, B.; Garcillán-Barcia, M.P.; De La Cruz, F.; Coll, P.; Navarro, F. In vivo transmission of a plasmid coharbouring bla DHA-1 and qnr B genes between Escherichia coli and Serratia marcescens. FEMS Microbiol. Lett. 2010, 308, 24–28. [Google Scholar] [CrossRef]
  46. Guo, Q.; Spychala, C.N.; McElheny, C.L.; Doi, Y. Comparative analysis of an IncR plasmid carrying armA, bla DHA-1 and qnrB4 from Klebsiella pneumoniae ST37 isolates. J. Antimicrob. Chemother. 2016, 71, 882–886. [Google Scholar] [CrossRef]
  47. Wang, S.; Zhou, K.; Xiao, S.; Xie, L.; Gu, F.; Li, X.; Ni, Y.; Sun, J.; Han, L. A Multidrug Resistance Plasmid pIMP26, Carrying bla IMP-26, fosA5, bla DHA-1, and qnrB 4 in Enterobacter cloacae. Sci. Rep. 2019, 9, 1–7. [Google Scholar]
  48. Takayama, Y.; Sekizuka, T.; Matsui, H.; Adachi, Y.; Eda, R.; Nihonyanagi, S.; Wada, T.; Matsui, M.; Suzuki, S.; Takaso, M. Characterization of the IncFII-IncFIB (pB171) plasmid carrying blaNDM-5 in Escherichia coli ST405 clinical isolate in Japan. Infect. Drug Resist. 2020, 13, 561. [Google Scholar] [CrossRef]
  49. Maurya, A.P.; Dhar, D.; Basumatary, M.K.; Paul, D.; Ingti, B.; Choudhury, D.; Talukdar, A.D.; Chakravarty, A.; Mishra, S.; Bhattacharjee, A. Expansion of highly stable bla OXA-10 β-lactamase family within diverse host range among nosocomial isolates of Gram-negative bacilli within a tertiary referral hospital of Northeast India. BMC Res. Notes 2017, 10, 1–6. [Google Scholar] [CrossRef]
  50. Ma, Y.; Xu, X.; Guo, Q.; Wang, P.; Wang, W.; Wang, M. Characterization of fosA5, a new plasmid-mediated fosfomycin resistance gene in E scherichia coli. Lett. Appl. Microbiol. 2015, 60, 259–264. [Google Scholar] [CrossRef]
  51. Wang, H.; Min, C.; Li, J.; Yu, T.; Hu, Y.; Dou, Q.; Zou, M. Characterization of fosfomycin resistance and molecular epidemiology among carbapenem-resistant Klebsiella pneumoniae strains from two tertiary hospitals in China. BMC Microbiol. 2021, 21, 1–8. [Google Scholar] [CrossRef]
  52. Sorlozano-Puerto, A.; Lopez-Machado, I.; Albertuz-Crespo, M.; Martinez-Gonzalez, L.J.; Gutierrez-Fernandez, J. Characterization of fosfomycin and nitrofurantoin resistance mechanisms in Escherichia coli isolated in clinical urine samples. Antibiotics 2020, 9, 534. [Google Scholar] [CrossRef]
  53. Botelho, J.; Roberts, A.P.; León-Sampedro, R.; Grosso, F.; Peixe, L. Carbapenemases on the move: It’s good to be on ICEs. Mob. DNA 2018, 9, 1–11. [Google Scholar] [CrossRef]
  54. He, J.; Li, C.; Cui, P.; Wang, H. Detection of Tn7-Like Transposons and Antibiotic Resistance in Enterobacterales from Animals Used for Food Production With Identification of Three Novel Transposons Tn6813, Tn6814, and Tn6765. Front. Microbiol. 2020, 11, 2049. [Google Scholar] [CrossRef]
  55. Biswas, B.; Azad, M.A.K.; Absar, N.; Islam, S.; Amin, S. Isolation and Identification of Pathogenic Bacteria from Fresh Fruits and Vegetables in Chittagong, Bangladesh. J. Microbiol. Res. 2020, 10, 55–58. [Google Scholar]
  56. Wauters, G.; Vaneechoutte, M. Approaches to the identification of aerobic Gram-negative bacteria. In Manual of Clinical Microbiology; Wiley: Hoboken, NJ, USA, 2015; pp. 613–634. [Google Scholar]
  57. Wayne, P. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing. Inform. Suppl. 2020, 31, 100–121. [Google Scholar]
  58. Sabeel, S.; Salih, M.A.; Ali, M.; El-Zaki, S.-E.; Abuzeid, N.; Elgadi, Z.A.M.; Altayb, H.N.; Elegail, A.; Ibrahim, N.Y.; Elamin, B.K. Phenotypic and genotypic analysis of multidrug-resistant Mycobacterium tuberculosis isolates from Sudanese patients. Tuberc. Res. Treat. 2017, 2017, 8340746. [Google Scholar]
  59. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef]
  60. Zerbino, D.R.; Birney, E. Velvet: Algorithms for de novo short read assembly using de Bruijn graphs. Genome Res. 2008, 18, 821–829. [Google Scholar] [CrossRef]
  61. Jolley, K.A.; Bray, J.E.; Maiden, M.C. Open-access bacterial population genomics: BIGSdb software, the PubMLST.org website and their applications. Wellcome Open Res. 2018, 3, 124. [Google Scholar] [CrossRef]
  62. Aziz, R.K.; Bartels, D.; Best, A.A.; DeJongh, M.; Disz, T.; Edwards, R.A.; Formsma, K.; Gerdes, S.; Glass, E.M.; Kubal, M. The RAST Server: Rapid annotations using subsystems technology. BMC Genom. 2008, 9, 1–15. [Google Scholar] [CrossRef]
  63. Tatusova, T.; DiCuccio, M.; Badretdin, A.; Chetvernin, V.; Nawrocki, E.P.; Zaslavsky, L.; Lomsadze, A.; Pruitt, K.D.; Borodovsky, M.; Ostell, J. NCBI prokaryotic genome annotation pipeline. Nucleic Acids Res. 2016, 44, 6614–6624. [Google Scholar] [CrossRef]
  64. Zankari, E.; Hasman, H.; Cosentino, S.; Vestergaard, M.; Rasmussen, S.; Lund, O.; Aarestrup, F.M.; Larsen, M.V. Identification of acquired antimicrobial resistance genes. J. Antimicrob. Chemother. 2012, 67, 2640–2644. [Google Scholar] [CrossRef]
  65. Johansson, M.H.; Bortolaia, V.; Tansirichaiya, S.; Aarestrup, F.M.; Roberts, A.P.; Petersen, T.N. Detection of mobile genetic elements associated with antibiotic resistance in Salmonella enterica using a newly developed web tool: MobileElementFinder. J. Antimicrob. Chemother. 2021, 76, 101–109. [Google Scholar] [CrossRef] [PubMed]
  66. Cosentino, S.; Larsen, M.V.; Aarestrup, F.M.; Lund, O. PathogenFinder-distinguishing friend from foe using bacterial whole genome sequence data. PLoS ONE 2013, 8, e77302. [Google Scholar] [CrossRef]
  67. Afgan, E.; Baker, D.; Batut, B.; Van Den Beek, M.; Bouvier, D.; Čech, M.; Chilton, J.; Clements, D.; Coraor, N.; Grüning, B.A. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update. Nucleic Acids Res. 2018, 46, W537–W544. [Google Scholar] [CrossRef] [PubMed]
  68. Seemann, T. Prokka: Rapid prokaryotic genome annotation. Bioinformatics 2014, 30, 2068–2069. [Google Scholar] [CrossRef]
  69. Page, A.J.; Cummins, C.A.; Hunt, M.; Wong, V.K.; Reuter, S.; Holden, M.T.; Fookes, M.; Falush, D.; Keane, J.A.; Parkhill, J. Roary: Rapid large-scale prokaryote pan genome analysis. Bioinformatics 2015, 31, 3691–3693. [Google Scholar] [CrossRef]
  70. Stamatakis, A. RAxML version 8: A tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics 2014, 30, 1312–1313. [Google Scholar] [CrossRef]
Figure 1. Map of different antibiotic-resistance genes, transposases, and plasmid, clustered in contig 33 of the K. pneumoniae (1KE) environmental strain. Showing an example of the presence of the CTX-M-15 gene which is located between two transposases. The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Figure 1. Map of different antibiotic-resistance genes, transposases, and plasmid, clustered in contig 33 of the K. pneumoniae (1KE) environmental strain. Showing an example of the presence of the CTX-M-15 gene which is located between two transposases. The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Pharmaceuticals 15 01011 g001
Figure 2. Map of antibiotic resistance and transposases cassette, identified in contig 188 of clinical K. pneumonia (14KP), showing the aminoglycoside-resistant genes flanked by three transposase genes. The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Figure 2. Map of antibiotic resistance and transposases cassette, identified in contig 188 of clinical K. pneumonia (14KP), showing the aminoglycoside-resistant genes flanked by three transposase genes. The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Pharmaceuticals 15 01011 g002
Figure 3. Map of ARGs and IS6 transposase cassette, identified in contig 66 of E. coli (1EP). The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Figure 3. Map of ARGs and IS6 transposase cassette, identified in contig 66 of E. coli (1EP). The outer black circle indicates the contig length, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Pharmaceuticals 15 01011 g003
Figure 4. Map of antibiotic resistance and transposases cassette, identified in contig 303 of environmental K. pneumoniae (12KE), showing the aminoglycoside-resistant genes flanked by three transposases genes. The outer black circle indicates the length of the contig, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Figure 4. Map of antibiotic resistance and transposases cassette, identified in contig 303 of environmental K. pneumoniae (12KE), showing the aminoglycoside-resistant genes flanked by three transposases genes. The outer black circle indicates the length of the contig, black arrows indicate coding sequences (CDS), green arrows indicate genes, and the inner blue zigzag circle indicates GC content.
Pharmaceuticals 15 01011 g004
Figure 5. Phylogenomic tree for the clinical and environmental isolates of E. coli from different sources in Khartoum and reference strains (Escherichia coli J53, K-12, and ATCC_43887), Sudan. Environmental isolates were EE while clinical isolates were EP. The blue blocks indicate gene presence and absence. The Klebsiella pneumoniae ATCC_BAA-2146 was used as an outgroup for rooting the tree.
Figure 5. Phylogenomic tree for the clinical and environmental isolates of E. coli from different sources in Khartoum and reference strains (Escherichia coli J53, K-12, and ATCC_43887), Sudan. Environmental isolates were EE while clinical isolates were EP. The blue blocks indicate gene presence and absence. The Klebsiella pneumoniae ATCC_BAA-2146 was used as an outgroup for rooting the tree.
Pharmaceuticals 15 01011 g005
Figure 6. Phylogenomic tree for the clinical and environmental isolates of K. pneumoniae from different sources in Khartoum and reference strains (K. pneumoniae NUHL24835, PittNDM01, and ATCC_BAA-2146), Sudan. Environmental isolates were KE, while clinical isolates were KP. The blue blocks indicate gene presence and absence, Escherichia coli strain ATCC_43887 was used as an outgroup for rooting the tree.
Figure 6. Phylogenomic tree for the clinical and environmental isolates of K. pneumoniae from different sources in Khartoum and reference strains (K. pneumoniae NUHL24835, PittNDM01, and ATCC_BAA-2146), Sudan. Environmental isolates were KE, while clinical isolates were KP. The blue blocks indicate gene presence and absence, Escherichia coli strain ATCC_43887 was used as an outgroup for rooting the tree.
Pharmaceuticals 15 01011 g006
Table 1. Information about the study isolates, source, Specimen type, pubMLST, ST, Human pathogen probability, Accession numbers and AST.
Table 1. Information about the study isolates, source, Specimen type, pubMLST, ST, Human pathogen probability, Accession numbers and AST.
IDSourceSpecimen TypePubMLSTSTAccession No.
1EPClinical sampleWound swabE. coli120JAJGXP000000000
5EPClinical sampleCSFE. coli38JAJGXO000000000
8EPClinical sampleUrineE. coli405JAJHQC000000000
10EPClinical sampleWound swabE. coli773JAJGXN000000000
11EPClinical sampleWound swabE. coli648JAJGXM000000000
15EPClinical sampleUrineE. coli73JAJGXK000000000
20EPClinical sampleUrineE. coli405JAJGXJ000000000
24EPClinical sampleAcetic fluidE. coli405JAJGXI000000000
25EPClinical sampleUrineE. coli405JAJGXH000000000
26EPClinical sampleWound SwabE. coli340JAJHGQ000000000
27EPClinical sampleUrineE. coli656JAJGXG000000000
28EPClinical sampleUrineE. coli410JAJGXF000000000
30EPClinical sampleUrineE. coli405JAJGXE000000000
1EEHandsSwabE. coli2522JAJGXX000000000
4EEHandsSwabE. coli58JAJGXW000000000
6EEWaterWater sampleE. coli4038JAJGXV000000000
7EEWaterWater sampleE. coli448JAJGXU000000000
11EEVegetablesSwab E. coli522JAJGXU000000000
12EPSurfaceSwabE. coli2280JAJGXL000000000
13EESurfaceSwabE. coli1308JAJGXS000000000
14EEWaterWater sampleE. coli1508JAJGXR000000000
16EEWaterWater sampleE. coli1146JAJGXQ000000000
1KEVegetablesSwabK. pneumoniae2365JAJHHI000000000
2KESurfaceSwabK. pneumoniae2177JAJHHH000000000
3KPClinical sampleUrineK. pneumoniae76JAJHHG000000000
5KPClinical sampleBloodK. pneumoniae45JAJHHF000000000
6KPClinical sampleBloodK. pneumoniae45JAJHHE000000000
7KESurfaceSwabK. pneumoniae700JAJHHD000000000
7KPClinical sampleBloodK. pneumoniae45JAJHHC000000000
8KEWaterWater sampleK. pneumoniae1584JAJHNR000000000
11KEVegetablesSwabK. pneumoniae1507JAJHHB000000000
12KEFruitsSwabK. pneumoniae45JAJHHA000000000
12KPClinical sampleBloodK. pneumoniae45JAJHGZ000000000
13KPClinical sampleBloodK. pneumoniae45JAJHGY000000000
14KPClinical sampleBloodK. pneumoniae45JAJHGX000000000
16KESurfaceSwabK. pneumoniae22,233 *JAJHNQ000000000
17KEVegetables SwabK. pneumoniae22,234 *JAJHGW000000000
19KEVegetables SwabK. pneumoniae45JAJHGV000000000
20KEVegetables SwabK. pneumoniae5624JAJHGU000000000
15KESurfaceSwabK. pneumoniae5808JAJHGT000000000
21KESurfaceSwabK. pneumoniae3335JAJHGS000000000
23KEWaterWater samplesK. pneumoniae1504 *JAJHGR000000000
* Assigned novel ST.
Table 2. Presentation of antimicrobial susceptibility profiles.
Table 2. Presentation of antimicrobial susceptibility profiles.
IDPhenotypic AST
Beta-LactamAminoglycosides and Fluoroquinolones
MRIMPCAZCTRAMCAKGENCIP
1EPSSRRRSSS
5EPSSRRRSRS
8EPSSRRRSSR
10EPSSRRRSSS
11EPSSRRRRRS
12EPSSSSRIIS
15EPSSSSRSSS
20EPSSRRRRRR
24EPSSSSRSSS
25EPSSRRRSSR
26EPSSRRRSSR
27EPSSRRRSSS
28EPSSRRRSSS
30EPSSRRRRRR
1EESSSSSSSS
4EESSSSSSSS
6EESSSSSSSS
7EESSSRRSSS
11EESSSSRSRS
13EESSSSRSSS
14EESSSSRIIS
16EESSSSRSRR
3KPSSSSRSSS
5KPSSRRRSRS
6KPSSRRRSRR
7KPSSRRRSRR
12KPSSRRRSSR
13KPSSRRRSSS
14KPSSRRRSSS
1KESSSSRSSS
2KESSSSRSSS
7KESSSSRSSS
8KESSSSRSSS
11KESSSSRSSS
12KESSSSRSSS
16KESSSSRSSS
15KESSSSRSSS
17KESSSSRSSS
19KESSRRRSSS
20KESSSSRSSS
21KESSSSRSSS
23KESSSSRSSS
Abbreviations: ST, strain; S, sensitive; R, resistant; I, Intermediate; -, negative; MR, meropenem; IMP, imipenem; CAZ, cefatzidime; CTX, cefotaxime; GEN, gentamicin; CTR, Co-trimoxazole; CIP, ciprofloxacin; AK, amikacin; AMC, amoxicillin-clavulanic acid.
Table 3. Antimicrobial-resistance genes identified in the isolates, red color means detected.
Table 3. Antimicrobial-resistance genes identified in the isolates, red color means detected.
IDBeta LactamAminoglycosides and FluoroquinolonesOthers
blaCTX-M-15blaCTX-M-216blaOXA-1blaDHA-1blaTEM-35blaTEM-1blaCMY-141blaCMY-42blaNDM-5blaSHV-1blaSHV-12blaSHV-11blaSHV-26blaSHV-38blaSHV-71blaLEN16ompK37ompK36blaLEN16blaECqnrB4aac(6′)-Ib-craac(3)-IIaaadA5aph(3″)-Ibaph(6)-Idmdf(A)MdfrmtBqepA4qacEqnrS1OqxBOqxAacrRsul2Sul1dfrAmph(A)catB3catA1tet(A)tet(B)tet(39)sitABCDfosAerm(C)
1EP
5EP
8EP
10EP
11EP
12EP
15EP
20EP
24EP
25EP
26EP
27EP
28EP
30EP
1EE
4EE
6EE
7EE
11EE
13EE
14EE
16EE
1KE
2KE
3KP
5KP
6KP
7KE
7KP
8KE
11KE
12KE
12KP
13KP
14KP
15KE
16KE
17KE
19KE
20KE
21KE
23KE
Table 4. Types and distributions of plasmids between studied isolates. Blue color means detected.
Table 4. Types and distributions of plasmids between studied isolates. Blue color means detected.
IDIncFIAIncFIBIncFIIIncI1-I(Alpha)IncYIncRIncFIB(H89-PhagePlasmid)IncFIB(pB171)IncFIB(pKPHS1)IncFII(pHN7A8)IncFIC(FII)ncFII(pRSB107)IncFIB(AP001918)Col156Col(BS512)Col440IIncI(Gamma)IncFII(pAMA1167-NDM-5)IncFIB(pNDM-Mar)IncHI1B(pNDM-MAR)IncFII(pCoo)IncFIA(HI1)p0111IncFII(29)IncFIB(K)(pCAV1099-114)FIA(pBK30683)IncFIB(K)IncFII(K)IncFII(pKP91)ColpVCpKP1433
1EP
5EP
8EP
10EP
11EP
12EP
15EP
20EP
24EP
25EP
26EP
27EP
28EP
30EP
1EE
4EE
6EE
7EE
11EE
13EE
14EE
16EE
1KE
2KE
3KP
5KP
6KP
7KE
7KP
8KE
11KE
12KE
12KP
13KP
14KP
16KE
17KE
19KE
20KE
15KE
21KE
23KE
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Altayb, H.N.; Elbadawi, H.S.; Alzahrani, F.A.; Baothman, O.; Kazmi, I.; Nadeem, M.S.; Hosawi, S.; Chaieb, K. Co-Occurrence of β-Lactam and Aminoglycoside Resistance Determinants among Clinical and Environmental Isolates of Klebsiella pneumoniae and Escherichia coli: A Genomic Approach. Pharmaceuticals 2022, 15, 1011. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15081011

AMA Style

Altayb HN, Elbadawi HS, Alzahrani FA, Baothman O, Kazmi I, Nadeem MS, Hosawi S, Chaieb K. Co-Occurrence of β-Lactam and Aminoglycoside Resistance Determinants among Clinical and Environmental Isolates of Klebsiella pneumoniae and Escherichia coli: A Genomic Approach. Pharmaceuticals. 2022; 15(8):1011. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15081011

Chicago/Turabian Style

Altayb, Hisham N., Hana S. Elbadawi, Faisal A. Alzahrani, Othman Baothman, Imran Kazmi, Muhammad Shahid Nadeem, Salman Hosawi, and Kamel Chaieb. 2022. "Co-Occurrence of β-Lactam and Aminoglycoside Resistance Determinants among Clinical and Environmental Isolates of Klebsiella pneumoniae and Escherichia coli: A Genomic Approach" Pharmaceuticals 15, no. 8: 1011. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15081011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop