Next Article in Journal
Construction of a Yeast Cell-Based Assay System to Analyze SNAP25-Targeting Botulinum Neurotoxins
Next Article in Special Issue
Antibiotics Resistance and Adhesive Properties of Clinical Staphylococcus aureus Isolated from Wound Infections
Previous Article in Journal
Impact of Chlorine Dioxide on Pathogenic Waterborne Microorganisms Occurring in Dental Chair Units
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antibiotic Susceptibility Profiling of Human Pathogenic Staphylococcus aureus Strains Using Whole Genome Sequencing and Genome-Scale Annotation Approaches

1
Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
2
Medical and Diagnostic Research Centre, University of Ha’il, Ha’il 55473, Saudi Arabia
3
Department of Medical, Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Ha’il 55476, Saudi Arabia
4
Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
5
Center of Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
6
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.
Microorganisms 2023, 11(5), 1124; https://doi.org/10.3390/microorganisms11051124
Submission received: 1 April 2023 / Revised: 22 April 2023 / Accepted: 24 April 2023 / Published: 26 April 2023
(This article belongs to the Special Issue Biology and Pathogenesis of Staphylococcus Infection 2.0)

Abstract

:
Staphylococcus species are major pathogens with increasing importance due to the rise in antibiotic resistance. Whole genome sequencing and genome-scale annotation are promising approaches to study the pathogenicity and dissemination of virulence factors in nosocomial methicillin-resistant and multidrug-resistant bacteria in intensive care units. Draft genome sequences of eight clinical S. aureus strains were assembled and annotated for the prediction of antimicrobial resistance genes, virulence factors, and phylogenetic analysis. Most of the studied S. aureus strains displayed multi-resistance toward the tested drugs, reaching more than seven drugs up to 12 in isolate S22. The mecA gene was detected in three isolates (S14, S21, and S23), mecC was identified in S8 and S9, and blaZ was commonly identified in all isolates except strain S23. Additionally, two complete mobile genomic islands coding for methicillin resistance SCCmec Iva (2B) were identified in strains S21 and S23. Numerous antimicrobial resistance genes (norA, norC, MgrA, tet(45), APH(3′)-IIIa, and AAC(6′)-APH(2″)) were identified in chromosomes of different strains. Plasmid analysis revealed the presence of blaZ, tetK, and ermC in different plasmid types, located in gene cassettes containing plasmid replicons (rep) and insertion sequences (IS). Additionally, the aminoglycoside-resistant determinants were identified in S1 (APH(3′)-IIIa), while AAC(6)-APH(2″) was detected in strains S8 and S14. The trimethoprim (dfrC) resistance gene was detected in S. aureus S21, and the fosfomycin (fosB) resistance gene was detected only in S. aureus S14. We also noted that S. aureus S1 belongs to ST1-t127, which has been reported as one of the most frequent human pathogen types. Additionally, we noted the presence of rare plasmid-mediated mecC-MRSA in some of our isolates.

1. Introduction

Staphylococcus aureus is a significant human pathogen that causes severe hospital infections. Methicillin-resistant S. aureus (MRSA) is one of the most important causal organisms of nosocomial infections, posing a significant risk to persons with weakened immune systems [1]. Alghaithy et al. [2] reported the prevalence of nasal carriage of S. aureus to be about 26.1% in the community and 25.4% in healthy hospital and non-hospital staff in hospitals in Abha (Saudi Arabia). Furthermore, during a 5-year period, from January 2015 to December 2019, Bazaid and colleagues [3] reported a 12% prevalence of S. aureus related to urinary tract infections in patients from two main hospitals in Ha’il, Saudi Arabia. In addition, S. aureus was reported to cause several infections such as osteoarticular infections, endocarditis, deep soft-tissue infections, and food poisoning [4,5,6,7]. Multidrug-resistant staphylococci may persist in the hospital environment and counteract drugs and biocides by generating a biofilm or by conversion to an atypical form [8]. The pathogenicity of Staphylococcus isolates is associated with the production of various virulence factors expressed by chromosomal genes or mobile genetic elements [9], as well as the combined action of different components of the bacterial surface. These factors code for toxins, enzymes, and cell adhesion and invasion factors, thus allowing the bacterium to fight the immune system, adhere to cells, disseminate in the body, form protective biofilms, develop resistance to various antibiotics, and use the available nutrients and energy [10]. Due to the genetic flexibility of S. aureus, various drug-resistant strains have emerged, causing a serious therapeutic problem [11]. In fact, S. aureus has developed resistance to methicillin because it harbors a genetic element, staphylococcal cassette chromosome mec (SCCmec), which carries the methicillin resistance gene (mecA) responsible for methicillin and penicillin resistance [12]. According to Moussa and Shibl [13], the mecA gene was detected in all strains phenotypically resistant to methicillin recovered from outpatient clinics in Riyadh, Saudi Arabia. Fusidic acid is a steroid antibiotic that has been used to treat S. aureus since the 1960s. Unfortunately, high fusidic acid use may result in the rapid development of resistance [14]. Fusidic acid resistance is frequent in Middle Eastern/Arabic Gulf countries and is mainly caused by plasmid-borne fusB/far1 [15,16] or SCC-associated fusC [17]. According to Albarrag et al. [18], 47% of analyzed MRSA isolated from a nursing home in Riyadh developed multiple-drug resistance (MDR) and were mecA-positive, and the SCCmec types were as follows: SCCmec IVc (41.18%), SCCmec V (29.41%), and SCCmec IVa (11.76%).
The pathogenicity of Staphylococcus spp. is amplified by its ability to form a biofilm that proliferates on various inert or biological surfaces. In fact, the formation of a biofilm reinforces adhesion to materials and protects the bacteria from immune defenses and the action of antimicrobial agents [19]. Several studies have shown that staphylococcal biofilm can also settle on biotic surfaces and even abiotic surfaces. Several genes implicated in adhesion and biofilm formation have been reported in S. aureus [20,21,22,23].
The comparative genomics technique was used to determine the evolutionary processes of clinically relevant S. aureus genomes and to identify areas influencing the acquisition of drug resistance and virulence factors [11]. Several staphylococci clones have been discovered internationally and regionally, and their epidemiological, clinical, and genetic characteristics have been evaluated [24]. Whole genome sequencing (WGS) enables the comparison of entire genomic DNA sequences and detects genetic variation across species [25], and it is considered a high-resolution approach for confirming outbreaks, studying pathogenesis, predicting resistance, assessing virulence, and typing microbial species [26]. This approach has been used to investigate the epidemiology of MRSA and its spread within hospitals as well as from hospitals to the general population [27].
Hence, the main aims of this study were to investigate whole genome sequencing, typing, and prediction of resistome and virulome in some clinical multidrug-resistant S. aureus strains associated with human infections.

2. Materials and Methods

2.1. Isolation of Multidrug-Resistant S. aureus

Various clinical multidrug-resistant S. aureus strains (Table 1) were collected from the microbiology laboratory at King Khalid hospital, Ha’il, Saudi Arabia, in March 2021. Samples from wounds, throat, sputum, and pleural fluid were plated in blood and MacConkey agar plates (Oxoid, Basingstoke, UK) and then incubated at 37 °C for 24 h. The purity of the suspected S. aureus isolates was then confirmed by sub-culturing on mannitol salt agar. Patient information including gender and location were collected from hospital records. The study was approved by the Ethics Committee at Ha’il Affairs (reference H-08-L-074). A consent form was not required because isolates were collected from the laboratory with no interaction with patients. Patient privacy and confidentiality of data were maintained in accordance with The Declaration of Helsinki.

2.2. Antimicrobial Susceptibility Testing

Antimicrobial susceptibility of the suspected S. aureus strains was achieved using a BD Phoenix™ M50 instrument (Becton, Dickinson and Co., Franklin Lakes, NJ, USA) as previously reported [28]. Susceptibility of the identified S. aureus was tested toward several antibiotics including ampicillin, amoxicillin–clavulanate, cefuroxime, cefoxitin, clindamycin, ciprofloxacin, daptomycin, erythromycin, fusidic acid, gentamicin, imipenem, mupirocin, linezolid, penicillin G, trimethoprim-sulfamethoxazole, nitrofurantoin, tetracycline, teicoplanin, vancomycin, rifampicin, moxifloxacin, and oxacillin. The results of the drug-susceptibility tests were interpreted according to Clinical and Laboratory Standards Institute (CLSI) guidelines document M100S-26 [29]. Two indices were used to interpret the obtained results, including the multiple antibiotic resistance index and the antibiotic resistance index as previously described by Abdulhakeem and colleagues [30].

2.3. Molecular Typing of Multi-Drug-Resistant S. aureus

2.3.1. S. aureus DNA Extraction

Total DNA was extracted from overnight 24 h bacterial culture growth. The guanidine chloride procedure was used for DNA extraction as detailed previously [31]. DNA quality and quantity were determined by gel-electrophoresis and Qubit (Thermo Scientific, Waltham, MA, USA).

2.3.2. Whole Genome Sequencing, Typing, and Prediction of Resistome and Virulome

The sequencing of the genomes of the selected strains was done by Novogene Company (Hong Kong, China) using Illumina HiSeq 2500 (Illumina, San Diego, CA, USA), and paired-end 150 bp reads with 100X coverage were achieved. Genome sequences were reassembled using SPAdes version 3.7 software [31,32]. Plasmids were assembled using plasmidSPAdes tool v3.15.4, applying different k-mer sizes (21, 33, and 55) [33]. The assembled contigs were submitted to the MLST 2.0 and PubMLST database [34] for species identification. The assembled genomes were run through the annotation pipeline PATRIC server and the NCBI Prokaryotic Genome Annotation Pipeline (PGAP) [35]. The determination of isolate sequence types (ST) and S. aureus-specific staphylococcal protein A (spa) genes were achieved using the global platform for genomic surveillance (Pathogenwatch) and spaTyper 1.0, respectively [36]. MLST clonal complexes (CCs) were determined according to the Ridom StaphType database’s previous publications [37,38]. Plasmids generated from plasmidSPAdes were identified using BLASTn and Mobile Element Finder [39]. Genomic and plasmid-mediated antimicrobial resistance were identified using Resistance Gene Identifier (RGI) and ResFinder [40]. Point mutations associated with drug resistance were screened using the RGI database. Staphylococcal chromosomal cassettes mec (SCCmec) were identified using SCCmec Finder 1.2. Virulence genes were identified from the assembled contigs using the Virulent Factor Database (VFDB, Version: 2016.03) applying MRSA252 as a reference genome. Phages were screened by PHASTER (PHAge Search Tool Enhanced Release) [41].

2.3.3. Pan-Genome Analysis and Phylogenomic Tree

Pan-genome analysis of all isolates’ genomes was conducted using Roary V. 3.11.2 software, and the general feature format (GFF) files generated from Prokka [42] annotation were used as input for Roary with default settings [43]. The alignment FASTA file generated from Roary was used as input for RaXML [44] for building the phylogenetic tree, which was then visualized by the interactive visualizer of genome phylogenies (Phandango) [45]. The Gene presence–absence file generated from Roary was uploaded to Scoary [46] to calculate the association between gene presence and absence with different traits such as methicillin resistance and source of samples.

3. Results and Discussion

3.1. Antimicrobial Susceptibility of the Isolated S. aureus

In this study, eight clinical S. aureus strains were tested for their antibiotic resistance profile using a BD Phoenix™ M50 instrument. Based on the minimum inhibitory concentration (MIC) values obtained, our results showed that almost all tested S. aureus strains were resistant to 3 to 12 antibiotics used (out of 21 tested). Interestingly, all tested S. aureus strains were highly resistant to cefoxitin (87.5%), Penicillin G (87.5%), and erythromycin (62.5%). In addition, the tested strains were completely resistant to ampicillin and cefotaxime, and sensitive to tigecycline, nitrofurantoin, and rifampicin. All these data are summarized in Table 2 in below.
Using the multiple antibiotic resistance index (MARI) and the antibiotic resistance index (ARI), results obtained showed that the ARI ranged from 0 to 1, while the MARI varied from 0.190 for S. aureus (S14) isolated from eye infection to 0.571 for S. aureus (S22) isolated from pleural fluid (Table 3).

3.2. Genome Composition and Genomic Variation

Genomes were assembled with an average of 2.75 Mb (2.74–2.85 Mb), at least 2601 predicted coding sequences (CDS), and a GC content of 32.7% for all isolates (Table 4); these results are consistent with S. aureus genome characteristics [47]. Isolates belonged to ST1 (S1 and S20), ST97 (S8 and S9), ST121 (S14), ST22 (S21), ST291 (S22), and ST6 (S23). Isolate S1 belonged to ST1-t127, which has been reported as one of the most frequent human pathogen types [48].
Data obtained from Saudi Arabia showed that ST239 is the most common in Riyadh and Dammam, located in the eastern region of Saudi Arabia [49], while in the western region this clone is not common [50], which is consistent with our findings. All clones identified here have been identified sporadically in the western region by Al-Zahrani et al. [51].

3.3. Prediction of Antimicrobial Resistance Mechanism in the Studied S. aureus

The isolates were identified with resistance genes to several antimicrobial classes as shown in Table 2 and Table 3; these genes are associated with phenotypic findings in which the isolates were multidrug-resistant to different classes of antibiotics as shown in Table 1. Genes responsible for beta-lactam resistance (blaZ, mecA, and mecC) were identified in the studied isolates: the mecA gene was detected in three MRSA isolates (S14, S21, and S23), mecC was identified in S8 and S9 isolates, and blaZ was commonly identified in all isolates except S23 (Table 5). Interestingly, we identified the presence of mecC-MRSA for the first time in our region; mecC is a new divergent from mecA usually associated with animal transmission, and these patients are usually from rural areas [51].
Eight antimicrobial-resistance-associated genes were identified in all of the isolates: the quinolone resistance genes (norA and norC) and their regulators (MgrA for norC and arlS for norA), the major facilitator superfamily MDR efflux pump in S. aureus (LmrS), the multidrug efflux pump (sdrM), the mepA gene which acts as a repressor for multidrug export protein, and the SepA gene that confers resistance to disinfecting agents and dyes [52]. SepA was also demonstrated to have induction activity on biofilm accumulation [53].
Although isolate S1 lacked the mecA and mecC genes, it exclusively harbored genes associated with resistance to different drug classes, including the tetracycline resistance gene (tet(45)), aminoglycoside resistance gene (APH(3′)-IIIa), nucleoside antibiotic (SAT-4), and erythromycin resistance genes (ermC) (Table 3). A total of 4 out of 8 (50%) isolates were positive for a gene (fusC) conferring resistance to fusidane antibiotic; a high prevalence of fusC in S. aureus has been documented recently in Saudi Arabia [54]. Aminoglycosides are among the important antibiotics that are used to treat a range of bacterial infections, especially those caused by Staphylococcus species [54]. Here, the aminoglycoside-resistant determinants were identified in S1 (APH(3′)-IIIa), and the AAC(6′)-APH(2″) was detected in strains S8 and S14. Additionally, strains S8 and S14 are MRSA and harbor multiple genes associated with resistance to different classes of antibiotics (Table 5). The presence of additional aminoglycoside-resistance genes will limit the therapeutic options, and this makes treatment for such isolates more challenging and eventually leads to the continuing spread of MRSA strains.
Point mutations (L27F, A100V, E291D, and T396N) associated with phosphonic acid resistance have been identified in isolate S14, while isolate S23 was identified with a fluoroquinolone-resistant parC gene mutation (S80F), GlpT gene mutations (F3I, A100V) conferring resistance to fosfomycin, and a gyrA gene mutation (S84L) conferring resistance to fluoroquinolones (Supplementary Table S1).

3.4. Plasmid-Mediated Antimicrobial Resistance Genes

Plasmid analysis revealed the presence of blaZ, tetK, and ermC in different plasmid types, located in gene cassettes containing plasmid replicons (rep) and insertion sequences (IS) (Table 6). The presence of these genes in a plasmid in association with plasmid replicons and insertion sequences will facilitate their distribution and transmission between resistant and susceptible isolates, which is reflected in their high distribution in our isolates. A gene cassette consisting of rep20, blaZ, and ISSau6 was identified in plasmid pUSA300HOUMS circulating in both S. aureus S8 and S9 (Table 6). The blaZ gene is a plasmid-mediated β-lactamase causing penicillin resistance and could also resist all β-lactam antibiotics [55,56]. The co-existence of the blaZ gene with the chromosomally mediated mec genes could be the reason for the high resistance of β-lactam antibiotics observed in our isolates. As presented in Table 4, we found isolate S22 with two rep genes (rep5 and rep16) and blaZ in one plasmid (pLDNT_611); the co-occurrence of two rep genes in one plasmid could indicate the occurrence of recombination between different S. aureus plasmids [57]. A gene encoding macrolides–lincosamides–streptogramin resistance (ermC) and rep10 were identified in plasmid pE5 circulating in three isolates: S1, S14, and S23. These results indicate that these isolates have acquired these genes from others via plasmids, which is in accordance with previous studies [58,59]. Horizontal transfer of the ermC gene in a small plasmid was commonly documented in animal and human staphylococcal species [59]. In accordance with a previous study conducted in Saudi Arabia, the prevalence of ermC in S. aureus isolates was 28.8% in this study [60]. The MRSA-S14 strain harbored an additional plasmid (pUSA02) that carried tetK; the presence of tetK in S. aureus pUSA02 plasmid has been documented in a previous study [61].

3.5. Analysis of Staphylococcal Chromosomal Cassettes mec (SCCmec)

The SCCmec is a mobile genomic island coding for methicillin resistance, classified into different subtypes according to the SCCmec gene cassette arrangement [62]. In this study, two complete SCCmec IVa(2B) were identified in isolates S21 and S23, while isolates S1, S8, S9, and S14 were documented with incomplete cassettes (Table 7, Figure 1). Isolate S21 belongs to ST22 and harbors the toxic shock syndrome gene (TSST-1), and CC22-MRSA-IV is a pandemic MRSA strain mainly found in Western Europe; recently, CC22-MRSA-IV with or without the TSST gene has been commonly identified in different Middle Eastern countries including Saudi Arabia [60].
Additionally, CC22-MRSA-IV strains positive for TSST are epidemically reported in Gaza, Palestine [63], which suggests an epidemiological link to Saudi Arabia. Isolate S23 belonged to ST6 and has spa type 304 (CC6-ST6-IV/t304), which is called the Western Australia (WA) MRSA-51 clone; this clone is reported dominantly in different Gulf countries, including Saudi Arabia, UAE, Kuwait, and Oman, with a prevalence of more than 36% in Oman and UAE [64]. The presence of incomplete SCCmec cassettes suggests a probable sequence deletion or insertion due to an unknown process [65].

3.6. Pan-Genome Analysis

Analysis of the staphylococcal pan-, core-, and accessory genomes revealed the presence of a total of 3837 genes, from which 2027 genes represent the core genes that are shared in >99% of all isolates, indicating their high similarity, ribosomes, and proteins associated with biogenesis [66,67]. A total of 992 genes were identified as shell genes present in >15% of the isolates; cloud genes represented 0–15% (818 genes), and soft-core genes were not identified in our studied isolates (Table 8). More details about core genes are represented in Supplementary File Table S2 and Figure 2.

3.7. Molecular Analysis of Virulence Factors in the Studied S. aureus

3.7.1. Detection of Panton–Valentine Leukocidin Gene

Panton–Valentine leukocidin (PVL) toxin is a virulence factor in S. aureus associated with deep skin and soft-tissue infections (furunculosis, cutaneous abscesses, and severe necrotizing pneumonia) [68,69,70]. In 1932, Panton and Valentine defined the PVL as a virulence factor in the family of synergohymenotropic toxins in S. aureus [71]. PVL is a toxin with two components that are encoded by the prophage-associated genes lukF-PV and lukS-PV [72].
We detected two PVL (lukF-PV and lukS-PV) in the S. aureus isolate S14 on the orf02119 and orf02120, respectively (Supplementary File Tables S1 and S2).
Recently, El-Deeb et al. [73] analyzed the whole genome of nine methicillin-resistant staphylococci (MRS) collected in Eastern Province, Saudi Arabia, and they found that only isolate SA1 recovered from goat’s milk had the PVL toxin gene and the Staphylococcus enterotoxin B gene seb. Ullah et al. [70] conducted a genomic investigation of an S. aureus strain isolated from Pakistan and discovered that the strain has many prophage-associated virulence factors, including PVL and toxic shock syndrome toxin (TSST). Additionally, all 52 isolates of S. aureus isolated from the vascular accesses in hemodialysis patients at Assiout University Hospitals were negative for the PVL gene [74]. Moreover, Alghizzi and Shami [75] analyzed 112 S. aureus strains isolated from raw milk and cheese and were unable to detect the PVL gene in any of the analyzed strains [75].

3.7.2. Detection of Genes Related to Iron Uptake System in S. aureus

S. aureus needs iron to survive, multiply, and infect cells, hence it has evolved specific proteins to steal heme from its host. The iron surface determinant (Isd) system is a protein family that obtains nutritional iron from the host body, allowing the bacteria to multiply during infection [76]. Staphylococcal cell-surface proteins (IsdA, IsdB, and IsdH) are assumed to channel their molecular payload to IsdC, which subsequently facilitates the transfer of the iron-containing nutrition to the membrane translocation system IsdDEF [77]. According to Valenciano-Bellido et al. [76], IsdH, a surface protein, binds to hemoglobin (Hb) and absorbs the heme moiety carrying the iron atom. A cluster of iron surface determinants (Isd) have been identified similarly in all isolates, which includes LPXTG-anchored heme-scavenging protein (IsdA), heme uptake protein (IsdB), heme uptake protein (IsdC), iron-regulated surface determinant protein (IsdD), heme ABC transporter substrate-binding protein (IsdE), and hemin ABC transporter permease protein (IsdF) (Figure 3). The staphylobilin-forming heme oxygenase (IsdI) was detected separately in another contig. The Isd locus, which is required for the acquisition of iron from hemoglobin, was identified in this study in all of the isolates; the common presence of this locus in different staphylococcus species has been documented previously [2].

3.7.3. Detection of Autolysin Genes in the Studied S. aureus Strains

Autolysin (Atl) is considered a peptidoglycan hydrolase (PGH) that is primarily responsible for the breakdown of the bacterial cell wall as well as daughter cell separation during cell division [78,79]. Atl of S. aureus is a cell-surface-associated peptidoglycan hydrolase containing amidase and glucosaminidase domains. Atl enzymes have been linked to biofilm development as well as staphylococcal adhesion to host extracellular and plasma proteins [80]. More recently, Zheng and colleagues [81] reported that Atl controls the sorting of LukAB from the cell envelope to the extracellular milieu. In our study, autolysin was detected in seven clinical S. aureus strains as well as the control strain (Supplementary Tables S1 and S2).

3.7.4. Detection of Enterotoxin and Exotoxin in the Studied S. aureus

Supplementary Table S1 lists genes related to staphylococcal enterotoxin A (sea) (37.5%), B (seb) (25%), G (seg) (25%), H (seh) (25%), I (sei) (25%), and enterotoxin-like K (selk) (25%), M (selm) (25%), N (seln) (25%), O (selo) (25%), Q (selq) (25%), and U (selu) (25%), while the following genes were absent; C (sec), D (sed), E (see), J (sej), L (sell), P (selp), and R (selr). According to Alghizzi and Shami [75], seh enterotoxin gene (51%) showed the highest ratio among the analyzed ones followed by see (27.5%), sem (21.6%), seo (19.6%), sea and sen (17.6%), seb (13.7%), seg (11.8%), sed and sei (3.9%), and lastly sec and sek (1.96%).
We noted the absence of sec, selk, see, sej, sell, selp, and selr (Supplementary Table S1). Similarly, neither sej nor sel existed in any of the MRSA isolates [75]. I have been demonstrated the most prevalent enterotoxin gene from S. aureus strains isolated from cell phones were sea (30%) followed by the sec gene (2.5%), while sed and seb genes not detected in any of the isolates [82]. Moreover, the analysis of 88 MRSA collected from 5 hospitals in Makkah demonstrated the presence of 2.3% S. aureus positive for the etb toxin gene, and none of the tested strains harbored the eta toxin gene [83]. According to Hamdan-Partida et al. [84], the seb gene (63.8%), which was more prevalent in strains isolated from the pharynx, and the tsst gene (57.8%), which was more prevalent in nose strains, were the most common toxin genes discovered. Additionally, we did not detect yent1 (Enterotoxin Yent1) and yent2 (Enterotoxin Yent2) genes. Moreover, the exfoliative toxin type A (eta), B (etb), C (etc), and D (etd) were also not detected in the analyzed samples (Supplementary Table S1).

3.7.5. Detection of Toxic Shock Syndrome Toxin in S. aureus

The toxic shock syndrome toxin 1 was considered among the main virulence factors elaborated by S. aureus associated with scalded skin syndrome [85]. Tsst-1 is frequently seen in conjunction with septic shock and toxic shock syndromes, and exfoliative toxins are common in isolates producing staphylococcal scalded skin syndrome [86]. In our investigation, the toxic shock syndrome toxin (tsst-1) was detected only in one S. aureus isolate (S21) located in the orf01006 (Supplementary Tables S1 and S2, Figure 4). In 2016, Ahmed [83] used multiplex PCR to analyze 88 MRSA isolates collected from 5 hospitals in Makkah, and they reported the presence of 3.4% positive strains for toxic shock syndrome toxin. Additionally, Udo et al. [87] demonstrated the presence of tsst in 23 out of 37 MRSA collected from patients in Kuwait hospitals.

4. Conclusions

In this study, eight clinical multidrug-resistant S. aureus strains were collected from the microbiology laboratory at King Khalid hospital, Ha’il, Saudi Arabia. The analysis of whole genomes revealed the presence of virulent and multidrug resistance determinants in the studied S. aureus strains. Interestingly, according to our literature survey, no previous study has been conducted in Saudi Arabia that has documented the presence of plasmid-mediated mecC-MRSA, so this report is considered the first one in our region. Additionally, mecA, norA, and norC, MgrA, tet(45), APH(3′)-IIIa, blaZ, tetK, and AAC(6′)-APH(2″) were also identified in the isolates. The pathogenic TSST-1-positive Western European MRSA strain (CC22-MRSA-IV) was also reported in this study.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11051124/s1, Table S1: Details of virulence factors identified in the analyzed S. aureus; Table S2: Predicted virulence factors in the whole genome of the studied S. aureus.

Author Contributions

Conceptualization, K.C., H.N.A., E.N. and M.S.; methodology, N.B., M.M.A., A.S.B. and M.S.; software, K.C., H.N.A. and M.S.; validation, M.S.; writing—original draft preparation, K.C., H.N.A., E.N. and M.S., writing—review and editing, A.S.B., K.C., H.N.A., E.N. and M.M.A.; supervision, M.S.; project administration, N.B., K.C., H.N.A., E.N. and M.S.; funding acquisition, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Scientific Research Deanship at University of Ha’il-Saudi Arabia through project number MDR-22 018.

Data Availability Statement

The Whole Genome Shotgun project PRJNA890445 has been deposited at DDBJ/ENA/GenBank under accession numbers JAOXRK000000000, JAOXRJ000000000, JAOXRI000000000, JAOXRH000000000, JAOXRG000000000, JAOXRF000000000, JAOXRE000000000, JAOXRD000000000 for the studied strains S1, S8, S9, S14, S20, S21, S22, and S23, respectively.

Acknowledgments

This research was funded by Scientific Research Deanship at University of Ha’il-Saudi Arabia through project number MDR-22 018.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Samia, N.I.; Robicsek, A.; Heesterbeek, H.; Peterson, L.R. Methicillin-resistant Staphylococcus aureus nosocomial infection has a distinct epidemiological position and acts as a marker for overall hospital-acquired infection trends. Sci. Rep. 2022, 12, 17007. [Google Scholar] [CrossRef] [PubMed]
  2. Alghaithy, A.A.; Bilal, N.E.; Gedebou, M.; Weily, A.H. Nasal carriage and antibiotic resistance of Staphylococcus aureus isolates from hospital and non-hospital personnel in Abha, Saudi Arabia. Trans. R. Soc. Trop. Med. Hyg. 2000, 94, 504–547. [Google Scholar] [CrossRef]
  3. Bazaid, A.S.; Saeed, A.; Alrashidi, A.; Alrashidi, A.; Alshaghdali, K.; AHammam, S.; Alreshidi, T.; Alshammary, M.; Alarfaj, A.; Thallab, R.; et al. Antimicrobial Surveillance for Bacterial Uropathogens in Ha’il, Saudi Arabia: A Five-Year Multicenter Retrospective Study. Infect Drug Resist. 2021, 14, 1455–1465. [Google Scholar] [CrossRef]
  4. Ash, S.; Kennedy, L.E. Deep soft-tissue infections. In Schlossberg’s Clinical Infectious Disease; Oxford University Press: Oxford, UK, 2022; Volume 154. [Google Scholar]
  5. Bourget, M.; Pasquie, M.; Charbonneau, H.; Bonnet, E. Comparable clinical course between coagulase-negative staphylococcal and Staphylococcus aureus endocarditis. Infection 2022, 50, 483–490. [Google Scholar] [CrossRef]
  6. Moutaouakkil, K.; Abdellaoui, H.; Arhoune, B.; Atarraf, K.; El Fakir, S.; Yahyaoui, G.; Mahmoud, M.; Afifi, M.A.; Oumokhtar, B. Paediatric osteoarticular infections caused by Staphylococcus aureus producing panton–valentine leucocidin in morocco: Risk factors and clinical features. Afr. J. Paediatr. Surg. 2022, 19, 78. [Google Scholar] [PubMed]
  7. Zhang, F.; Wu, S.; Lei, T.; Wu, Q.; Zhang, J.; Huang, J.; Dai, J.; Chen, M.; Ding, Y.; Wang, J. Presence and characterization of methicillin-resistant Staphylococcus aureus co-carrying the multidrug resistance genes cfr and lsa (E) in retail food in China. Int. J. Food Microbiol. 2022, 363, 109512. [Google Scholar] [CrossRef]
  8. Guo, H.; Tong, Y.; Cheng, J.; Abbas, Z.; Li, Z.; Wang, J.; Zhou, Y.; Si, D.; Zhang, R. Biofilm and Small Colony Variants An Update on Staphylococcus aureus Strategies toward Drug Resistance. Int. J. Mol. Sci. 2022, 23, 1241. [Google Scholar] [CrossRef]
  9. Sanchini, A. Recent Developments in Phenotypic and Molecular Diagnostic Methods for Antimicrobial Resistance Detection in Staphylococcus aureus: A Narrative Review. Diagnostics 2022, 12, 208. [Google Scholar] [CrossRef]
  10. Naber, C.K. Staphylococcus aureus bacteremia: Epidemiology, pathophysiology, and management strategies. Clin. Infect. Infect. Dis. 2009, 48, S231–S237. [Google Scholar] [CrossRef]
  11. Holden, M.T.; Feil, E.J.; Lindsay, J.A.; Peacock, S.J.; Day, N.P.; Enright, M.C.; Foster, T.J.; Moore, C.E.; Hurst, L.; Atkin, R. Complete genomes of two clinical Staphylococcus aureus strains: Evidence for the rapid evolution of virulence and drug resistance. Proc. Natl. Acad. Sci. USA 2004, 101, 9786–9791. [Google Scholar] [CrossRef]
  12. Hiramatsu, K.; Katayama, Y.; Yuzawa, H.; Ito, T. Molecular genetics of methicillin-resistant Staphylococcus aureus. Int. J. Med. Microbiol. 2002, 292, 67–74. [Google Scholar] [CrossRef] [PubMed]
  13. Moussa, I.; Shibl, A.M. Molecular characterization of methicillin-resistant Staphylococcus aureus recovered from outpatient clinics in Riyadh, Saudi Arabia. Saudi Med. J. 2009, 30, 611–617. [Google Scholar]
  14. Williamson, D.A.; Monecke, S.; Heffernan, H.; Ritchie, S.R.; Roberts, S.A.; Upton, A.; Thomas, M.G.; Fraser, J.D. High usage of topical fusidic acid and rapid clonal expansion of fusidic Acid–Resistant Staphylococcus aureus: A cautionary tale. Clin. Infect. Dis. 2014, 59, 1451–1454. [Google Scholar] [CrossRef]
  15. Udo, E.; O’brien, F.; Al-Sweih, N.; Noronha, B.; Matthew, B.; Grubb, W. Genetic lineages of community-associated methicillin-resistant Staphylococcus aureus in Kuwait hospitals. J. Clin. Microbiol. 2008, 46, 3514–3516. [Google Scholar] [CrossRef]
  16. Udo, E.E.; Al-Lawati, B.-H.; Al-Muharmi, Z.; Thukral, S. Genotyping of methicillin-resistant Staphylococcus aureus in the Sultan Qaboos University Hospital, Oman reveals the dominance of Panton–Valentine leucocidin-negative ST6-IV/t304 clone. New Microbes New Infect. 2014, 2, 100–105. [Google Scholar] [CrossRef] [PubMed]
  17. Udo, E.E.; Boswihi, S.S.; Mathew, B.; Noronha, B.; Verghese, T.; Al-Jemaz, A.; Al Saqer, F. Emergence of methicillin-resistant Staphylococcus aureus belonging to clonal complex 15 (CC15-MRSA) in Kuwait hospitals. Infect. Drug Resist. 2020, 13, 617. [Google Scholar] [CrossRef]
  18. Albarrag, A.; Shami, A.; Almutairi, A.; Alsudairi, S.; Aldakeel, S.; Al-Amodi, A. Prevalence and molecular genetics of methicillin-resistant Staphylococcus aureus colonization in nursing homes in Saudi Arabia. Can. J. Infect. Dis. Med. Microbiol. 2020, 2020, 2434350. [Google Scholar] [CrossRef]
  19. Donlan, R.M.; Costerton, J.W. Biofilms: Survival mechanisms of clinically relevant microorganisms. Clin. Microbiol. Rev. 2002, 15, 167–193. [Google Scholar] [CrossRef]
  20. Piechota, M.; Kot, B.; Frankowska-Maciejewska, A.; Grużewska, A.; Woźniak-Kosek, A. Biofilm Formation by Methicillin-Resistant and Methicillin-Sensitive Staphylococcus aureus Strains from Hospitalized Patients in Poland. Biomed. Res. Int. 2018, 27, 4657396. [Google Scholar]
  21. Alabdullatif, M.; Alzahrani, A. Expression of biofilm-associated genes in Staphylococcus aureus during storage of platelet concentrates. Transfus. Apher. Sci. 2022, 61, 103456. [Google Scholar] [CrossRef] [PubMed]
  22. Bui, L.M.; Conlon, B.P.; Kidd, S.P. Antibiotic tolerance and the alternative lifestyles of Staphylococcus aureus. Essays Biochem. 2017, 61, 71–79. [Google Scholar]
  23. Cucarella, C.; Solano, C.; Valle, J.; Amorena, B.; Lasa, I.N.I.; Penadés, J.R. Bap, a Staphylococcus aureus surface protein involved in biofilm formation. J. Bacteriol. 2001, 183, 2888–2896. [Google Scholar] [CrossRef]
  24. Yamamoto, T.; Nishiyama, A.; Takano, T.; Yabe, S.; Higuchi, W.; Razvina, O.; Shi, D. Community-acquired methicillin-resistant Staphylococcus aureus: Community transmission, pathogenesis, and drug resistance. J. Infect. Chemother. 2010, 16, 225–254. [Google Scholar] [CrossRef]
  25. Eyre, D.W.; Golubchik, T.; Gordon, N.C.; Bowden, R.; Piazza, P.; Batty, E.M.; Ip, C.L.; Wilson, D.J.; Didelot, X.; O’Connor, L. A pilot study of rapid benchtop sequencing of Staphylococcus aureus and Clostridium difficile for outbreak detection and surveillance. BMJ Open 2012, 2, e001124. [Google Scholar] [CrossRef]
  26. Price, J.; Claire Gordon, N.; Crook, D.; Llewelyn, M.; Paul, J. The usefulness of whole genome sequencing in the management of Staphylococcus aureus infections. Clin. Microbiol. Infect. 2013, 19, 784–789. [Google Scholar] [CrossRef]
  27. Humphreys, H.; Coleman, D.C. Contribution of whole-genome sequencing to understanding of the epidemiology and control of meticillin-resistant Staphylococcus aureus. J. Hosp. Infect. 2019, 102, 189–199. [Google Scholar] [CrossRef]
  28. Bazaid, A.S.; Barnawi, H.; Qanash, H.; Alsaif, G.; Aldarhami, A.; Gattan, H.; Alharbi, B.; Alrashidi, A.; Al-Soud, W.A.; Moussa, S.; et al. Bacterial Coinfection and Antibiotic Resistance Profiles among Hospitalised COVID-19 Patients. Microorganisms 2022, 10, 495. [Google Scholar] [CrossRef]
  29. Patel, J.B. Performance Standards for Antimicrobial Susceptibility Testing; Clinical and Laboratory Standards Institute: Wayne, MI, USA, 2017. [Google Scholar]
  30. 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]
  31. Abdulhakeem, M.A.; Alreshidi, M.; Bardakci, F.; Hamadou, W.S.; De Feo, V.; Noumi, E.; Snoussi, M. Molecular Identification of Bacteria Isolated from Marketed Sparus aurata and Penaeus indicus Sea Products: Antibiotic Resistance Profiling and Evaluation of Biofilm Formation. Life 2023, 13, 548. [Google Scholar] [CrossRef]
  32. Bankevich, A.; Nurk, S.; Antipov, D.; Gurevich, A.A.; Dvorkin, M.; Kulikov, A.S.; Lesin, V.M.; Nikolenko, S.I.; Pham, S.; Prjibelski, A.D. SPAdes: A new genome assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. 2012, 19, 455–477. [Google Scholar] [CrossRef]
  33. Antipov, D.; Hartwick, N.; Shen, M.; Raiko, M.; Lapidus, A.; Pevzner, P.A. plasmidSPAdes: Assembling plasmids from whole genome sequencing data. Bioinformatics 2016, 32, 3380–3387. [Google Scholar] [CrossRef]
  34. 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] [PubMed]
  35. 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]
  36. Bartels, M.D.; Petersen, A.; Worning, P.; Nielsen, J.B.; Larner-Svensson, H.; Johansen, H.K.; Andersen, L.P.; Jarløv, J.O.; Boye, K.; Larsen, A.R. Comparing whole-genome sequencing with Sanger sequencing for spa typing of methicillin-resistant Staphylococcus aureus. J. Clin. Microbiol. 2014, 52, 4305–4308. [Google Scholar] [CrossRef]
  37. Monecke, S.; Coombs, G.; Shore, A.C.; Coleman, D.C.; Akpaka, P.; Borg, M.; Chow, H.; Ip, M.; Jatzwauk, L.; Jonas, D. A field guide to pandemic, epidemic and sporadic clones of methicillin-resistant Staphylococcus aureus. PLoS ONE 2011, 6, e17936. [Google Scholar] [CrossRef] [PubMed]
  38. Strommenger, B.; Kettlitz, C.; Weniger, T.; Harmsen, D.; Friedrich, A.; Witte, W. Assignment of Staphylococcus isolates to groups by spa typing, SmaI macrorestriction analysis, and multilocus sequence typing. J. Clin. Microbiol. 2006, 44, 2533–2540. [Google Scholar] [CrossRef]
  39. 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]
  40. 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]
  41. Arndt, D.; Grant, J.R.; Marcu, A.; Sajed, T.; Pon, A.; Liang, Y.; Wishart, D.S. PHASTER: A better, faster version of the PHAST phage search tool. Nucleic Acids Res. 2016, 44, W16–W21. [Google Scholar] [CrossRef]
  42. Seemann, T. Prokka: Rapid prokaryotic genome annotation. Bioinformatics 2014, 30, 2068–2069. [Google Scholar] [CrossRef]
  43. 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]
  44. Stamatakis, A. RAxML version 8: A tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics 2014, 30, 1312–1313. [Google Scholar] [CrossRef] [PubMed]
  45. Hadfield, J.; Croucher, N.J.; Goater, R.J.; Abudahab, K.; Aanensen, D.M.; Harris, S.R. Phandango: An interactive viewer for bacterial population genomics. Bioinformatics 2018, 34, 292–293. [Google Scholar] [CrossRef]
  46. Brynildsrud, O.; Bohlin, J.; Scheffer, L.; Eldholm, V. Rapid scoring of genes in microbial pan-genome-wide association studies with Scoary. Genome Biol. 2016, 17, 238. [Google Scholar] [CrossRef] [PubMed]
  47. Åvall-Jääskeläinen, S.; Koort, J.; Simojoki, H.; Taponen, S. Genomic analysis of Staphylococcus aureus Isolates associated with peracute non-gangrenous or gangrenous mastitis and comparison with other mastitis-associated Staphylococcus aureus Isolates. Front. Microbiol. 2021, 12, 688819. [Google Scholar] [CrossRef] [PubMed]
  48. Tsergouli, K.; Karampatakis, T.; Kontopoulou, K.; Pappa, S.; Kampouridou, P.; Kallasidou, G.; Tsioka, K.; Zotou, S.; Farmaki, Ε.-E.; Kotzamanidis, C. Spa diversity and genetic characterization of t127 methicillin-resistant Staphylococcus aureus in a tertiary Greek hospital. Acta Microbiol. Et Immunol. Hung. 2022, 69, 185–192. [Google Scholar] [CrossRef]
  49. Al Yousef, S.A.; Taha, E.M. Methicillin-resistant Staphylococcus aureus in Saudi Arabia: Genotypes distribution review. Saudi J. Med. Med. Sci. 2016, 4, 2. [Google Scholar] [CrossRef]
  50. Al-Zahrani, I.A.; Azhar, E.I.; Jiman-Fatani, A.A.; Siddig, L.A.; Yasir, M.; Al-Ghamdi, A.K.; Harwood, C.R. Impact of mass migrations on the clonal variation of clinical Staphylococcus aureus strains isolated from the Western region of Saudi Arabia. J. Infect. Public Health 2019, 12, 317–322. [Google Scholar] [CrossRef]
  51. Moghaddam, P.Z.; Azimian, A.; Sepahy, A.A.; Iranbakhsh, A. Isolation and Genetic Characterization of Vancomycin-resistant and mecC+ Methicillin-resistant Staphylococcus aureus Strains in Clinical Samples of Bojnurd, Northeastern Iran. Jundishapur J. Microbiol. 2021, 14, e118949. [Google Scholar] [CrossRef]
  52. Narui, K.; Noguchi, N.; Wakasugi, K.; Sasatsu, M. Cloning and characterization of a novel chromosomal drug efflux gene in Staphylococcus aureus. Biol. Pharm. Bull. 2002, 25, 1533–1536. [Google Scholar] [CrossRef]
  53. Paharik, A.E.; Kotasinska, M.; Both, A.; Hoang, T.M.N.; Büttner, H.; Roy, P.; Fey, P.D.; Horswill, A.R.; Rohde, H. The metalloprotease S ep A governs processing of accumulation-associated protein and shapes intercellular adhesive surface properties in Staphylococcus epidermidis. Mol. Microbiol. 2017, 103, 860–874. [Google Scholar] [CrossRef]
  54. Senok, A.; Somily, A.M.; Nassar, R.; Garaween, G.; Sing, G.K.; Müller, E.; Reissig, A.; Gawlik, D.; Ehricht, R.; Monecke, S. Emergence of novel methicillin-resistant Staphylococcus aureus strains in a tertiary care facility in Riyadh, Saudi Arabia. Infect. Drug Resist. 2019, 12, 2739. [Google Scholar] [CrossRef]
  55. Mahdiyoun, S.M.; Kazemian, H.; Ahanjan, M.; Houri, H.; Goudarzi, M. Frequency of aminoglycoside-resistance genes in methicillin-resistant Staphylococcus aureus (MRSA) isolates from hospitalized patients. Jundishapur J. Microbiol. 2016, 9, e35052. [Google Scholar] [CrossRef] [PubMed]
  56. Okiki, P.; Eromosele, E.; Ade-Ojo, P.; Sobajo, O.; Idris, O.; Agbana, R. Occurrence of mecA and blaZ genes in methicillin-resistant Staphylococcus aureus associated with vaginitis among pregnant women in Ado-Ekiti, Nigeria. New Microbes New Infect. 2020, 38, 100772. [Google Scholar] [CrossRef] [PubMed]
  57. McCarthy, A.J.; Lindsay, J.A. The distribution of plasmids that carry virulence and resistance genes in Staphylococcus aureus is lineage associated. BMC Microbiol. 2012, 12, 1–8. [Google Scholar] [CrossRef] [PubMed]
  58. Gan, T.; Shu, G.; Fu, H.; Yan, Q.; Zhang, W.; Tang, H.; Yin, L.; Zhao, L.; Lin, J. Antimicrobial resistance and genotyping of Staphylococcus aureus obtained from food animals in Sichuan Province, China. BMC Vet. Res. 2021, 17, 177. [Google Scholar] [CrossRef]
  59. Wang, H.; Zhuang, H.; Ji, S.; Sun, L.; Zhao, F.; Wu, D.; Shen, P.; Jiang, Y.; Yu, Y.; Chen, Y. Distribution of erm genes among MRSA isolates with resistance to clindamycin in a Chinese teaching hospital. Infect. Genet. Evol. 2021, 96, 105127. [Google Scholar] [CrossRef]
  60. Senok, A.; Somily, A.; Raji, A.; Gawlik, D.; Al-Shahrani, F.; Baqi, S.; Boswihi, S.; Skakni, L.; Udo, E.E.; Weber, S. Diversity of methicillin-resistant Staphylococcus aureus CC22-MRSA-IV from Saudi Arabia and the Gulf region. Int. J. Infect. Dis. 2016, 51, 31–35. [Google Scholar] [CrossRef]
  61. Shittu, A.O.; Udo, E.E.; Lin, J. Insights on virulence and antibiotic resistance: A review of the accessory genome of Staphylococcus aureus. Wounds 2007, 19, 237. [Google Scholar]
  62. Ito, T.; Kuwahara-Arai, K.; Katayama, Y.; Uehara, Y.; Han, X.; Kondo, Y.; Hiramatsu, K. Staphylococcal cassette chromosome mec (SCCmec) analysis of MRSA. In Methicillin-Resistant Staphylococcus aureus (MRSA) Protocols; Springer: Berlin/Heidelberg, Germany, 2014; pp. 131–148. [Google Scholar]
  63. Laham, N.A.; Mediavilla, J.R.; Chen, L.; Abdelateef, N.; Elamreen, F.A.; Ginocchio, C.C.; Pierard, D.; Becker, K.; Kreiswirth, B.N. MRSA clonal complex 22 strains harboring toxic shock syndrome toxin (TSST-1) are endemic in the primary hospital in Gaza, Palestine. PLoS ONE 2015, 10, e0120008. [Google Scholar] [CrossRef]
  64. Al-Saleh, A.; Shahid, M.; Farid, E.; Bindayna, K. Trends in methicillin-resistant Staphylococcus aureus in the Gulf Cooperation Council countries: Antibiotic resistance, virulence factors and emerging strains. East. Mediterr. Health J. 2022, 28, 434–443. [Google Scholar] [CrossRef]
  65. Zong, Z.; Lü, X. Characterization of a new SCC mec element in Staphylococcus cohnii. PLoS ONE 2010, 5, e14016. [Google Scholar] [CrossRef] [PubMed]
  66. Jalil, M.; Quddos, F.; Anwer, F.; Nasir, S.; Rahman, A.; Alharbi, M.; Alshammari, A.; Alshammari, H.K.; Ali, A. Comparative Pan-Genomic Analysis Revealed an Improved Multi-Locus Sequence Typing Scheme for Staphylococcus aureus. Genes 2022, 13, 2160. [Google Scholar] [CrossRef]
  67. Naushad, S.; Nobrega, D.B.; Naqvi, S.A.; Barkema, H.W.; De Buck, J. Genomic analysis of bovine Staphylococcus aureus isolates from milk to elucidate diversity and determine the distributions of antimicrobial and virulence genes and their association with mastitis. Msystems 2020, 5, e00063-20. [Google Scholar] [CrossRef] [PubMed]
  68. Hussain, K.; Bandyopadhyay, A.; Roberts, N.; Mughal, N.; Moore, L.S.; Fuller, L.C. Panton-Valentine Leukocidin producing Staphylococcus aureus: A clinical review. Clin. Exp. Dermatol. 2022, 47, 2150–2158. [Google Scholar] [CrossRef]
  69. Mohamadou, M.; Essama, S.R.; Ngonde Essome, M.C.; Akwah, L.; Nadeem, N.; Gonsu Kamga, H.; Sattar, S.; Javed, S. High prevalence of Panton-Valentine leukocidin positive, multidrug resistant, Methicillin-resistant Staphylococcus aureus strains circulating among clinical setups in Adamawa and Far North regions of Cameroon. PLoS ONE 2022, 17, e0265118. [Google Scholar] [CrossRef]
  70. Ullah, N.; Nasir, S.; Ishaq, Z.; Anwer, F.; Raza, T.; Rahman, M.; Alshammari, A.; Alharbi, M.; Bae, T.; Rahman, A. Comparative Genomic Analysis of a Panton–Valentine Leukocidin-Positive ST22 Community-Acquired Methicillin-Resistant Staphylococcus aureus from Pakistan. Antibiotics 2022, 11, 496. [Google Scholar] [CrossRef] [PubMed]
  71. Panton, P.; Came, M.; Valentine, F. Staphylococcal toxin. Lancet 1932, 219, 506–508. [Google Scholar] [CrossRef]
  72. Boakes, E.; Kearns, A.; Ganner, M.; Perry, C.; Hill, R.; Ellington, M. Distinct bacteriophages encoding Panton-Valentine leukocidin (PVL) among international methicillin-resistant Staphylococcus aureus clones harboring PVL. J. Clin. Microbiol. 2011, 49, 684–692. [Google Scholar] [CrossRef]
  73. El-Deeb, W.; Cave, R.; Fayez, M.; Alhumam, N.; Quadri, S.; Mkrtchyan, H.V. Methicillin Resistant Staphylococci Isolated from Goats and Their Farm Environments in Saudi Arabia Genotypically Linked to Known Human Clinical Isolates: A Pilot Study. Microbiol. Spectr. 2022, 10, e00387-322. [Google Scholar] [CrossRef]
  74. Hassan, E.A.; Abdel-Rahim, M.H.; Hassan, T.H.M.; A Azoz, N.M.; Mohamed, M.E. Some virulence genes of Staphylococcus aureus isolated from infected vascular accesses in hemodialysis patients at Assiut University Hospitals. Bull. Pharm. Sci. Assiut 2022, 45, 371–387. [Google Scholar] [CrossRef]
  75. Alghizzi, M.; Shami, A. The prevalence of Staphylococcus aureus and methicillin resistant Staphylococcus aureus in milk and dairy products in Riyadh, Saudi Arabia. Saudi J. Biol. Sci. 2021, 28, 7098–7104. [Google Scholar] [CrossRef] [PubMed]
  76. Valenciano-Bellido, S.; Caaveiro, J.M.M.; Morante, K.; Sushko, T.; Nakakido, M.; Nagatoishi, S.; Tsumoto, K. Structure and role of the linker domain of the iron surface-determinant protein IsdH in heme transportation in Staphylococcus aureus. J. Biol. Chem. 2022, 298, 101995. [Google Scholar] [CrossRef]
  77. Sharp, K.H.; Schneider, S.; Cockayne, A.; Paoli, M. Crystal structure of the heme-IsdC complex, the central conduit of the Isd iron/heme uptake system in Staphylococcus aureus. J. Biol. Chem. 2007, 282, 10625–10631. [Google Scholar] [CrossRef] [PubMed]
  78. Pishchany, G.; Sheldon, J.R.; Dickson, C.F.; Alam, M.T.; Read, T.D.; Gell, D.A.; Heinrichs, D.E.; Skaar, E.P. IsdB-dependent hemoglobin binding is required for acquisition of heme by Staphylococcus aureus. J. Infect. Dis. 2014, 209, 1764–1772. [Google Scholar] [CrossRef]
  79. Hemmadi, V.; Biswas, M. An overview of moonlighting proteins in Staphylococcus aureus infection. Arch. Microbiol. 2021, 203, 481–498. [Google Scholar] [CrossRef]
  80. Porayath, C.; Suresh, M.K.; Biswas, R.; Nair, B.G.; Mishra, N.; Pal, S. Autolysin mediated adherence of Staphylococcus aureus with Fibronectin, Gelatin and Heparin. Int. J. Biol. Macromol. 2018, 110, 179–184. [Google Scholar] [CrossRef] [PubMed]
  81. Zheng, X.; Ma, S.X.; St. John, A.; Torres, V.J. The Major Autolysin Atl Regulates the Virulence of Staphylococcus aureus by Controlling the Sorting of LukAB. Infect. Immun. 2022, 90, e00056-22. [Google Scholar]
  82. de Souza, G.M.; de Jesus, M.F.A.; de Souza Ferreira, M.V.; de Oliveira Vieira, K.C.; Nakagaki, W.R.; Lordelo, E.P.; Cataneli, V.P.; Winkelstroter, L.K. Virulence, biofilm formation ability and antimicrobial resistance of Staphylococcus aureus isolated from cell phones of university students. ABCS Health Sci. 2022, 47, e022203. [Google Scholar] [CrossRef]
  83. Ahmed, O.B. Prevalence of Exfoliative and Toxic Shock Syndrome Toxin Genes in Methicillin-resistant Staphylococcus aureus Strains Isolated from Clinical Specimens in Makkah, Saudi Arabia. Br. J. Med. Med. Res. 2016, 11, 1. [Google Scholar] [CrossRef]
  84. Hamdan-Partida, A.; González-García, S.; Martínez-Ruíz, F.J.; Zavala-Sánchez, M.Á.; Bustos-Hamdan, A.; Bustos-Martínez, J. Molecular Characterization of Staphylococcus aureus Strains Isolated from Mobile Phones. Microorganisms 2022, 10, 669. [Google Scholar] [CrossRef]
  85. Ahmed, O.B. Prevalence of Exfoliative and Toxic Shock Syndrome Genes in Methicillin-Resistant Staphylococcus aureus Strains Isolated from Clinical Specimens. Curr. Overv. Dis. Health Res. 2022, 4, 120–128. [Google Scholar]
  86. Opal, S.; Johnson-Winegar, A.; Cross, A. Staphylococcal scalded skin syndrome in two immunocompetent adults caused by exfoliatin B-producing Staphylococcus aureus. J. Clin. Microbiol. 1988, 26, 1283–1286. [Google Scholar] [CrossRef] [PubMed]
  87. Udo, E.E.; Boswihi, S.S.; Al-Sweih, N. High prevalence of toxic shock syndrome toxin–producing epidemic methicillin-resistant Staphylococcus aureus 15 (EMRSA-15) strains in Kuwait hospitals. New Microbes New Infect. 2016, 12, 24–30. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Staphylococcal chromosomal cassettes mec (SCCmec) (IVa(2B)) identified in isolate S21. The cassette chromosomal recombinase genes are in turquoise, the mecA gene and its regulator are in pink, transposase is in red, Transposase in green, and recombinase in cyan, purple 9ndicate the rest of genes.
Figure 1. Staphylococcal chromosomal cassettes mec (SCCmec) (IVa(2B)) identified in isolate S21. The cassette chromosomal recombinase genes are in turquoise, the mecA gene and its regulator are in pink, transposase is in red, Transposase in green, and recombinase in cyan, purple 9ndicate the rest of genes.
Microorganisms 11 01124 g001
Figure 2. Core-genome phylogenetic analysis showing core and non-core genes in S. aureus isolates. On the left, the phylogenetic tree is built from 3837 core genes of the isolates. The blue heat map shows core genes (dark blue) and the blue boxes and white gaps show non-core genes. Each row represents the aligned sample.
Figure 2. Core-genome phylogenetic analysis showing core and non-core genes in S. aureus isolates. On the left, the phylogenetic tree is built from 3837 core genes of the isolates. The blue heat map shows core genes (dark blue) and the blue boxes and white gaps show non-core genes. Each row represents the aligned sample.
Microorganisms 11 01124 g002
Figure 3. Map of isd locus and surrounding chromosomal genes (purple) in isolate S1; isd genes are colored in green.
Figure 3. Map of isd locus and surrounding chromosomal genes (purple) in isolate S1; isd genes are colored in green.
Microorganisms 11 01124 g003
Figure 4. Map of a group of virulence genes located at the same contig in isolate S21 and surrounded by other staphylococcal proteins (purple). The green color shows the toxic shock syndrome toxin (TSST-1); leukocidins (lukGH) are colored in turquoise, the staphylokinase (sak) gene is colored in yellow, the leukocidins G and H are in cyan color, the rest of genes are colored in purple.
Figure 4. Map of a group of virulence genes located at the same contig in isolate S21 and surrounded by other staphylococcal proteins (purple). The green color shows the toxic shock syndrome toxin (TSST-1); leukocidins (lukGH) are colored in turquoise, the staphylokinase (sak) gene is colored in yellow, the leukocidins G and H are in cyan color, the rest of genes are colored in purple.
Microorganisms 11 01124 g004
Table 1. Type and antibiotic resistance profile of S. aureus collected in this study with patient gender and location.
Table 1. Type and antibiotic resistance profile of S. aureus collected in this study with patient gender and location.
StrainGenderLocationSample
S1MaleWardWound
S8FemaleWardWound
S9MaleICUThroat swab
S14MalePICUEye swab
S20MaleICUSputum
S21FemaleWardWound
S22MaleWardPleural Fluid
S23MaleWardWound
Table 2. Antibiotic susceptibility profiles obtained by using a BD Phoenix™ M50 instrument.
Table 2. Antibiotic susceptibility profiles obtained by using a BD Phoenix™ M50 instrument.
StrainResistance Profile
S1Cefoxitin; Cefotaxime; Ampicillin; Penicillin G; Oxacillin; Trimethoprim; Erythromycin; Ciprofloxacin; Tetracycline
S8Gentamicin; Cefoxitin; Cefotaxime; Ampicillin; Penicillin G
S9Gentamicin; Cefoxitin; Cefotaxime; Ampicillin; Penicillin G; Oxacillin
S14Cefotaxime; Ceftaroline; Ampicillin
S20Cefoxitin; Cefotaxime; Ceftaroline; Ampicillin; Penicillin G; Teicoplanin; Vancomycin; Clindamycin; Erythromycin; Linezolid; Tetracycline
S21Cefoxitin; Cefotaxime; Ceftaroline; Ampicillin; Penicillin G; Oxacillin; Daptomycin; Trimethoprim; Vancomycin
S22Cefoxitin; Cefotaxime; Ampicillin; Penicillin G; Teicoplanin; Vancomycin; Clindamycin; Erythromycin; Ciprofloxacin; Levofloxacin; Moxifloxacin; Tetracycline
S23Cefoxitin; Cefotaxime; Ampicillin; Penicillin G; Oxacillin; Clindamycin; Erythromycin
Table 3. Multiple antibiotic resistance index (MARI) and antibiotic resistance index (ARI) distribution in the six S. aureus strains tested.
Table 3. Multiple antibiotic resistance index (MARI) and antibiotic resistance index (ARI) distribution in the six S. aureus strains tested.
Antibiotics TestedS1S8S9S14S20S21S22S23ARI
GentamicinSRRSSSSS2/8 = 0.25
CefoxitinRRRSRRRR7/8 = 0.875
CefotaximeRRRRRRRR8/8 = 1
CeftarolineSSSRRRIS3/8 = 0.375
AmpicillinRRRRRRRR8/8 = 1
Penicillin GRRRSRRRR7/8 = 0.875
OxacillinRSRSSRSR4/8 = 0.5
DaptomycinSSSSSRSS1/8 = 0.125
TrimethoprimRSSSSRSS2/8 = 0.25
TeicoplaninSSSSRSRS2/8 = 0.25
VancomycinSSSSRRRS3/8 = 0.375
ClindamycinSSSSRSRR3/8 = 0.375
ErythromycinRSSRRSRR5/8 = 0.625
LinezolidSSSSRSSS1/8 = 0.125
NitrofurantoinSSSSSSSS0/8 = 0
CiprofloxacinRSSSSSRS2/8 = 0.25
LevofloxacinSSSSSSRS1/8 = 0.125
MoxifloxacinSSSSSSRS1/8 = 0.125
RifampinSSSSSSSS0/8 = 0
TetracyclineRSSSRSRS3/8 = 0.375
TigecyclineSSSSSSSS0/8 = 0
MARI0.4280.2380.2850.1900.5230.4280.5710.333
Table 4. Genome characteristics of the studied S. aureus and assembly statistics.
Table 4. Genome characteristics of the studied S. aureus and assembly statistics.
IsolateSTSpa TypeMLST CCsGenome LengthNo of Contigs Coverage N50GCCDStRNArRNA
S11t127CC012,806,63421438336,51132.7%2659544
S897t189CC972,758,54132463300,84432.7%2632563
S997t2297CC972,853,55348403286,66832.7%2777553
S14121t314CC1212,778,59561331113,58232.7%2634584
S201t5388CC012,785,40514331547,17432.7%2645553
S2122t845C222,742,17444471173,81332.7%2607535
S22291t3649CC2912,755,47134526211,29832.7%2632533
S236t304CC62,750,77226485298,52432.7%2601574
Biosample/Accession number: S1 (SAMN31278742/JAOXRK000000000); S8 (SAMN31278743/JAOXRJ000000000);
S9 (SAMN31278744/JAOXRI000000000); S14 (SAMN31278745/JAOXRH000000000); S20 (SAMN31278746/JAOXRG000000000); S21 (SAMN31278747/JAOXRF000000000); S22 (SAMN31278748/JAOXRE000000000); S23 (SAMN31278749/JAOXRD000000000).
Table 5. Predicted antimicrobial resistance genes among the studied S. aureus.
Table 5. Predicted antimicrobial resistance genes among the studied S. aureus.
GeneResistance MechanismS1S8S9S14S20S21S22S23
mecAAntibiotic target replacement x x x
mecCAntibiotic target replacement xx
arlRAntibiotic effluxxxxxxxxx
arlSAntibiotic effluxx xx x
S. aureus norAAntibiotic effluxxxxxxxxx
mgrAAntibiotic effluxxxxxxxxx
dfrCAntibiotic target replacement x
fusCAntibiotic target protectionxxxx
S. aureus FosBAntibiotic inactivation x
mepRAntibiotic effluxxxxxxxxx
APH(3′)-IIIaAntibiotic inactivationx
AAC(6′)-APH(2″)Antibiotic inactivation x x
norCAntibiotic effluxxxxxxxxx
S. aureus LmrSAntibiotic effluxxxxxxxxx
sepAAntibiotic effluxxxxxxxxx
sdrMAntibiotic effluxxxxxxxxx
tet(45)Antibiotic effluxx
tetKAntibiotic efflux x
PC1 beta-lactamase (blaZ)Antibiotic inactivationxxxxxxx
SAT-4Antibiotic inactivationx
ErmCAntibiotic target alterationx x
Table 6. Plasmids identified in the studied S. aureus associated with drug resistance genes and mobile elements.
Table 6. Plasmids identified in the studied S. aureus associated with drug resistance genes and mobile elements.
StrainPlasmidContigCoveragePlasmid IdentityReferenceGenes
NamePosition in ContigIdentity
S1pE51157399%M17990.1erm(C)1542–2276100%
rep10251–727100%
S8pUSA300HOUMS169799%CP000732.1rep2019,264–20,253100%
blaZ10,806–9961100%
ISSau610,806–9961100%
S9pUSA300HOUMS342699%CP000732.1rep202896–3885100%
blaZ13,996–14,841100%
ISSau613,996–14,841100%
S14pUSA022147799%CP000257.1tetK1425–46100%
rep7a3379–4323100%
pE535.7399%M17990.1rep10512–988100%
erm(C)1803–2537100%
S20pER07993.3A.1164899%CP049391.1rep5a1669–2529100%
blaZ8989–9834100%
S21--------
S22pLDNT_611189799%CP080252.1rep5a1066–206100%
rep162357–3100100%
blaZ13,545–14,390100%
S23pE52229699%M17990.1rep101746–2222100%
erm(C)197–931100%
Table 7. Staphylococcal chromosomal cassettes mec (SCCmec) identified in the studied S. aureus.
Table 7. Staphylococcal chromosomal cassettes mec (SCCmec) identified in the studied S. aureus.
IsolateSCCmec TypeSCCmec Gene CassettesIdentity (%)Ref. CoverageContig NoContig Position
S1N/AccrB1:1:COL:CP00004692.251626/1625S1_contig_263,208–64,832
ccrA1:1:COL:CP00004694.371350/1350S1_contig_264,854–66,203
S8N/AccrC1-allele-8:1:AB46239399.941677/1677S8_contig_31522–3198
mec-class-C2:3:AB47878099.912290/2398S8_contig_182493–4782
S9N/AccrC1-allele-8:1:AB46239398.451677/1677S9_contig_201462–3138
mec-class-C2:3:AB4787801002198/2398S9_contig_232400–4597
ccrC1-allele-2:1:AB5127671001680/1680S9_contig_17153–8832
S14N/Amec-class-C2:5:AB50562999.864408/4408S14_contig_44295–4702
ccrC1-allele-8:1:AB46239399.941677/1677S14_contig_1455,301–56,977
S20No SCCmec
S21IVa(2B)mecA:5:CP0000461002007/2007S21_contig_181366–3372
IS1272:2:AB03376399.681550/1585S21_contig_341–1550
subtype-IVa(2B):1:CA05:AB0631721001491/1491S21_contig_2027,585–29,075
dmecR1:1:AB033763100987/987S21_contig_18280–1266
ccrA2:7:81108:AB0962171001350/1350S21_contig_2033,027–34,376
ccrB2:9:JCSC4469:AB09767799.941650/1650S21_contig_2034,377–36,026
S22No SCCmec
S23IVa(2B)subtype-a(2B):1:CA05:AB06317299.931491/1491S23_contig_1628,081–29,571
mecA:12:AB5056281002010/2010S23_contig_4285,112–287,121
dmecR1:1:AB033763100987/987S23_contig_4287,218–288,204
IS1272:3:AM2923041001843/1843S23_contig_4288,193–290,035
ccrB2:9:JCSC4469:AB09767799.941650/1650S23_contig_4291,877–293,526
ccrA2:7:81108:AB0962171001350/1350S23_contig_4293,527–294,876
Abbreviations: N/A = The sequencing result was not complete and the SCCmec type was not determined.
Table 8. Frequency of staphylococcal isolates’ pan-genomes, categorized into core, soft-core, shell, and cloud genes.
Table 8. Frequency of staphylococcal isolates’ pan-genomes, categorized into core, soft-core, shell, and cloud genes.
Core Genes(99% ≤ strains ≤ 100%)2027
Soft-core genes(95% ≤ strains < 99%)0
Shell genes(15% ≤ strains < 95%)992
Cloud genes(0% ≤ strains < 15%)818
Total genes(0% ≤ strains ≤ 100%)3837
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Snoussi, M.; Noumi, E.; Bouali, N.; Bazaid, A.S.; Alreshidi, M.M.; Altayb, H.N.; Chaieb, K. Antibiotic Susceptibility Profiling of Human Pathogenic Staphylococcus aureus Strains Using Whole Genome Sequencing and Genome-Scale Annotation Approaches. Microorganisms 2023, 11, 1124. https://doi.org/10.3390/microorganisms11051124

AMA Style

Snoussi M, Noumi E, Bouali N, Bazaid AS, Alreshidi MM, Altayb HN, Chaieb K. Antibiotic Susceptibility Profiling of Human Pathogenic Staphylococcus aureus Strains Using Whole Genome Sequencing and Genome-Scale Annotation Approaches. Microorganisms. 2023; 11(5):1124. https://doi.org/10.3390/microorganisms11051124

Chicago/Turabian Style

Snoussi, Mejdi, Emira Noumi, Nouha Bouali, Abdulrahman S. Bazaid, Mousa M. Alreshidi, Hisham N. Altayb, and Kamel Chaieb. 2023. "Antibiotic Susceptibility Profiling of Human Pathogenic Staphylococcus aureus Strains Using Whole Genome Sequencing and Genome-Scale Annotation Approaches" Microorganisms 11, no. 5: 1124. https://doi.org/10.3390/microorganisms11051124

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