Skip to main content

Clinical manifestations, antimicrobial resistance and genomic feature analysis of multidrug-resistant Elizabethkingia strains

Abstract

Background

Elizabethkingia is emerging as an opportunistic pathogen in humans. The aim of this study was to investigate the clinical epidemiology, antimicrobial susceptibility, virulence factors, and genome features of Elizabethkingia spp.

Methods

Clinical data from 71 patients who were diagnosed with Elizabethkingia-induced pneumonia and bacteremia between August 2019 and September 2021 were analyzed. Whole-genome sequencing was performed on seven isolates, and the results were compared with a dataset of 83 available Elizabethkingia genomes. Genomic features, Kyoto Encyclopedia of Genes and Genomes (KEGG) results and clusters of orthologous groups (COGs) were analyzed.

Results

The mean age of the patients was 56.9 ± 20.7 years, and the in-hospital mortality rate was 29.6% (21/71). Elizabethkingia strains were obtained mainly from intensive care units (36.6%, 26/71) and emergency departments (32.4%, 23/71). The majority of the strains were isolated from respiratory tract specimens (85.9%, 61/71). All patients had a history of broad-spectrum antimicrobial exposure. Hospitalization for invasive mechanical ventilation or catheter insertion was found to be a risk factor for infection. The isolates displayed a high rate of resistance to cephalosporins and carbapenems, but all were susceptible to minocycline and colistin. Genomic analysis identified five β-lactamase genes (blaGOB, blaBlaB, blaCME, blaOXA, and blaTEM) responsible for β-lactam resistance and virulence genes involved in stress adaptation (ureB/G, katA/B, and clpP), adherence (groEL, tufA, and htpB) and immune modulation (gmd, tviB, cps4J, wbtIL, cap8E/D/G, and rfbC). Functional analysis of the COGs revealed that “metabolism” constituted the largest category within the core genome, while “information storage and processing” was predominant in both the accessory and unique genomes. The unique genes in our 7 strains were mostly enriched in KEGG pathways related to microRNAs in cancer, drug resistance (β-lactam and vancomycin), ABC transporters, biological metabolism and biosynthesis, and nucleotide excision repair mechanisms.

Conclusion

The Elizabethkingia genus exhibits multidrug resistance and carries carbapenemase genes. This study presents a comparative genomic analysis of Elizabethkingia, providing knowledge that facilitates a better understanding of this microorganism.

Background

Elizabethkingia spp. are aerobic, Gram-negative bacilli, belonging to the family Flavobacteriaceae [1, 2]. Since their discovery in 1959, these microorganisms have been associated with a variety of human infectious diseases, such as meningitis, keratitis and sepsis, that particularly affect immunocompromised individuals [3]. There are currently seven known species, namely, Elizabethkingia meningoseptica, Elizabethkingia anophelis, Elizabethkingia miricola, Elizabethkingia argenteiflava, Elizabethkingia occulta, Elizabethkingia ursingii, and Elizabethkingia bruuniana, all of which have undergone various taxonomic and nomenclature changes. In 2003, E. miricola was identified for the first time in condensed water samples from the Russian space station Mir [4] and found to be associated with sepsis, bacteremia and pneumonia [5]. In 2011, E. anophelis was discovered in the midgut of Anopheles gambiae mosquitos [6] and found to be associated with neonatal meningitis, catheter-associated infections, and two infection outbreaks in Wisconsin with high mortality [6, 7]. In 2018, three new species were redefined, namely, E. occulta, E. ursingii, and E. bruuniana [8]. The number of whole-genome sequences released for Elizabethkingia is steadily increasing. Due to their innate resistance to several classes of antibiotics, treatment of Elizabethkingia infections is challenging. Consequently, empirical treatment often leads to a relatively high mortality rate. Early diagnosis and prompt initiation of appropriate antibiotics are critical for improving survival rates and outcomes [9]. In addition, several prior studies have reported that patients with a history of persistent antibiotic exposure are more likely to develop nosocomial infections associated with the Elizabethkingia genus [10]. Despite the great clinical significance of Elizabethkingia infections, gaps remain in our understanding of their demographic characteristics, pathogenicity, and effective treatment options.

Elizabethkingia strains have been reported to harbor various types of β-lactamases, which are enzymes responsible for resistance against β-lactams. There are two significant categories of β-lactamases associated with Elizabethkingia: class A extended-spectrum-β-lactamases (ESBLs) and class B metallo-β-lactamases (MBLs). The chromosomal genes blaBlaB, blaGOB and blaCME are unique to Elizabethkingia spp. and contribute to their natural resistance to several commonly used carbapenem antibiotics. The Virulence Factor Database has been employed to predict numerous common virulence factors for Elizabethkingia. Many virulence genes have been found to be involved in the synthesis of various components, such as lipo-oligosaccharides, capsule polysaccharides, catalases, proteases, and peroxidases [11]. Additionally, a two-component regulatory system, superoxide dismutase, heat shock protein, and several other factors contributing to the bacterium’s virulence have been identified [12].

Materials and methods

Clinical data collection and strains in this study

To accurately investigate the epidemiology and clinical characteristics of Elizabethkingia infections, we collected clinical data from 71 patients at West China Hospital of Sichuan University (Chengdu, China) between 2020 and 2021. Seven representative Elizabethkingia isolates were routinely collected for further analysis. These isolates were identified using matrix-assisted laser desorption ionization-time of flight mass spectrometry platforms (Bruker Daltonics) with an updated reference spectrum database, and their identities were confirmed via whole-genome sequencing. At the time of the study, there were 83 whole-genome sequences of Elizabethkingia species (Supplementary Materials Table S1) available in GenBank (https://www.ncbi.nlm.nih.gov/genome/) (until Dec 01, 2021). All these genome sequences were downloaded for comparative genomic analysis. This database included 43 E. anophelis, 16 E. meningoseptica, 18 E. miricola, 4 E. ursingii, and 2 E. occulta sequences, which were all isolated from the environment. Among these genomic sequences, 26 were complete genomes, and 57 were shotgun sequences presented as scaffolds or contigs.

Whole-genome sequencing, assembly and annotation

Seven representative strains were sequenced using an Illumina HiSeq 2500 sequencing platform (Oebiotech, Shanghai, China). Short reads were assembled with SPAdes v.3.6 and optimized based on paired-end and overlap relationships by mapping reads to contigs and scaffolds. The assembled genomes were submitted to the NCBI Prokaryotic Genome Annotation Pipeline and the Rapid Annotations based onProkaryotic Genome Annotation Server (http://rast.nmpdr.org/) for gene function annotation. The annotations were revised using UniProt (http://www.uniprot.org/) and BLAST (https://blast.ncbi.nlm.nih.gov/blast.cgi). Antibiotic resistance genes were identified using the Antibiotic Resistance Genes Database BLAST Server (https://ardb.cbcb.umd.edu/). The Virulence Factor Database (http://www.mgc.ac.cn/VFs/main.htm) was used to identify virulence genes [13].

Bioinformatics analysis

To ensure uniform and consistent annotations for core and pangenome analyses, all genome sequences were annotated using PROKKA v.1.11 [14]. For genome similarity assessment, average nucleotide identity (ANI) was computed using an ANI calculator (https://www.ezbiocloud.net/tools/ani). Bacterial pangenome analysis (BPGA) [15] was used for comprehensive pan/core genome analysis, including functional annotation of core, accessory and unique genes to cluster of orthologous group (COG) categories and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, using the default parameters. A whole-genome sequence-based phylogenetic tree was constructed using the Reference Sequence Alignment Phylogeny builder (REALPHY) [16]. The whole-genome sequences of the 83 Elizabethkingia strains were submitted to the REALPHY online pipeline in FASTA format. Sequences acquired in our study and reference sequences downloaded from GenBank were used to construct a phylogenetic tree using PhyML 3.1 [17].

Antimicrobial susceptibility test

Antimicrobial susceptibility was determined using the VITEK 2 system (bioMérieux, Lyon, France), and breakpoints were interpreted by using the criteria for non-Enterobacterales according to CLSI M100-S30 [18]. The minimal inhibitory concentration (MIC) of minocycline was determined by a Kindy-Bauer KB (Thermo Fisher Scientific, USA) following the manufacturer’s recommendations. The breakpoint (MIC > 2 µg/mL) for colistin was determined by the broth microdilution method adapted from the EUCAST [19].

Results

Clinical characteristics of Elizabethkingia species infections

As shown in Table 1, the specimens were primarily isolated from the respiratory tract (85.9%, 61/71), including sputum (69.1%, 49/71), tracheal secretory fluid (14.1%, 10/71), and bronchoalveolar lavage fluid (2.8%, 2/71). There were only a few samples of sterile body fluids, including cerebrospinal fluid (4.3%, 3/71), ascites (1.4%, 1/71), drainage fluid (2.8%, 2/71), blood (1.4%, 1/71) and pus (2.8%, 2/71). The specimens were predominantly isolated from the intensive care unit (36.6%, 26/71) and emergency (32.4%, 23/71) departments, with fewer specimens from the neurosurgery department (9.9%, 7/71). Most patients had various underlying conditions, such as pulmonary infection, hypertension, and diabetes. A total of 61 patients (85.9%) were hospitalized for more than two weeks. In addition, most patients underwent invasive treatment, including mechanical ventilation (71.8%, 51/71), nasogastric tube placement (63.4%, 45/71) and catheter insertion (59.2%, 42/71). All patients had a history of antibiotic exposure, and 94.4% of patients (67/71) had used broad-spectrum antibiotics for more than one week. The in-hospital mortality rate was 29.6% (21/71).

Table 1 Characteristics of 71 patients with Elizabethkingia infections

Antimicrobial susceptibility

Table 2 displays the antimicrobial susceptibility of Elizabethkingia isolates and the corresponding MICs for the tested antibiotics. All strains exhibited resistance to 13 ~ 16 antimicrobial agents, including aminoglycosides, macrolides, cephalosporins and carbapenems (imipenem and meropenem). Additionally, they exhibited higher rates of resistance to trimethoprim-sulfamethoxazole. The rate of resistance to piperacillin was 100%, but when piperacillin was combined with β-lactamase inhibitors (cefoperazone/sulbactam and piperacillin/tazobactam), increased sensitivity rates of 90.1% (64/71) and 66.2% (47/71), respectively, were observed. Importantly, all the isolates were found to be susceptible to minocycline and colistin.

Table 2 Antimicrobial susceptibilities of 71 Elizabethkingia strains

General features of Elizabethkingia strains

The next-generation sequencing and assembly data for the 7 genomes are presented in Table 3. The genome size ranged from 4.04 Mb to 4.31 Mb, with an average size of 4.10 Mb, which is consistent with the genome size of the 83 Elizabethkingia strains selected from the NCBI database (Supplementary Materials Table S1). The sizes of these 83 genomes ranged from 3.59 Mb to 4.58 Mb, with an average size of 4.26 Mb. The number of contigs per genome ranged from 1 to 378, with a mean of 32.75. The read depth ranged from 23.26 to 80.63, with a mean of 38.79. The average GC content and tRNA content in 83 Elizabethkingia strains were 35.84% and 45.42%, respectively.

Table 3 General features of seven Elizabethkingia strains

Resistance-associated genes of the various Elizabethkingia strains

We analyzed the resistance genes of the 83 strains from NCBI and 7 strains from our study and found that different genes were involved in resistance to 6 antibiotics. Antimicrobial resistance genes in the 83 Elizabethkingia strains are presented in Supplementary Materials Table S2. These resistance genes included the extended-spectrum β-lactamase genes blaCME,blaOXA−347 and blaTEM−116; the carbapenem resistance genes blaBlaB and blaGOB and their various subtypes; the aminoglycoside resistance genes aadS and aph(3’)-IIa; the tetracycline resistance genes tetX and tet36; and the sulfonamide resistance gene sul2. Molecular analysis did not reveal any genes in the mobile colistin resistance (mcr) gene family. There was no difference in the distribution of drug resistance genes among the five species. The 7 strains carried all three previously described β-lactamase genes unique to Elizabethkingia, including the extended-spectrum β-lactamase blaCME and metallo-β-lactams blaBlaB and blaGOB. The specific genes included macrolide, lincosamide and streptogramin (MLS) resistance genes ermF, ereD, mefC and mphG. In addition, certain aminoglycoside resistance genes, such as aac(3)-IVb and aac(3)-IIIc, were only found in our 7 strains (Table S3).

Virulence-associated genes of the various Elizabethkingia strains

The potential virulence factors and the associated genes of the 83 strains and 7 Elizabethkingia strains in our study are shown in Table S2 and Table S4, respectively. We found that some virulence genes, such as catalase/(hydro)peroxidase (katA) and translation elongation factor (tufA), were widely distributed in our seven strains. We identified a total of 753 virulence genes in all strains, and 23 kinds of virulence factors could be classified into three types: stress adaptation, adherence and immune modulation. The virulence genes catalase/(hydro)peroxidase (katA) and translation elongation factor (tufA) were widely distributed in all 90 strains. In addition, the capsular polysaccharide biosynthesis protein (cps4J) gene was detected only in E. miricola. In addition, 60 K heat shock protein (htpB), urease accessory protein (ureE), urease beta subunit (ureB), Vi polysaccharide biosynthesis (tviB) and chaperonin (groEL) were identified in these strains. The immune modulation gene rfbA and stress survival gene fcl were identified only in the 83 strains from the NCBI.

Core and pangenome analysis and phylogenetic relationships between Elizabethkingia species

To clarify the characteristics and differences in the pangenome between the seven Elizabethkingia strains in this study and those from the database, we performed a pangenome analysis on these 90 strains. Core genome analysis revealed that the number of shared genes decreased with the addition of the input genomes. Overall, E. meningoseptica exhibited an open pangenome feature, with new genes appearing when more sequenced genomes were added to the analysis. Pangenome analysis can be used to determine the diversity of genomes and explore core, accessory, and unique genes. In the 90 strains, 2079 core genes were identified. In each strain, the number of accessory genes ranged from 1097 to 1925, and the number of unique genes ranged from 0 to 364. With the addition of new genome sequences, the number of genes in the pangenome increased from 3265 to 11,813, and the number of core genes decreased from 2546 to 1959 (Fig. 1). The distributions of different gene families and the numbers of new genes are illustrated in Fig. 2A and B, respectively. Whole-genome comparisons allow for the distinction between different strains and species with high resolution. Genome sequences were analyzed using a pairwise method, calculating and comparing the ANI for the 90 Elizabethkingia strains (Fig. 2C). The pairwise comparisons revealed a minimum ANI of 80.72% for the most distant strains, whereas the E. anophelis subspecies showed an ANI of > 98.0%. Additionally, we observed that the ANIs of E. meningoseptica and four other species were notably lower than those of the other species. According to the dendrogram, E. ursingii and E. occulta appeared to be relatively close to E. miricola. The delineation of the five species within the Elizabethkingia genus was clearly evident in the heatmap generated from the similarity matrix. The phylogenetic analysis of all the isolates was based on whole-genome sequences (Figure S1). Similar to the dendrogram generated from the ANI analysis, E. ursingii and E. occulta were located close to E. miricola and were distant from E. anophelis and E. meningoseptica. E. anophelis was divided into two major sublineages.

Fig. 1
figure 1

Pan, core and singleton genome evolution according to the number of selected Elizabethkingia strains

Fig. 2
figure 2

Pangenome analysis of 90 Elizabethkingia strains. (A) The distribution of the various gene families in the 90 Elizabethkingia strains. (B) The distribution of new genes in the 90 Elizabethkingia strains. (C) Dendrogram and heatmap generated using the ANIs of 90 different Elizabethkingia strains

Functional analysis of COGs

COG analysis (Fig. 3A and C) revealed 1,497 conserved genes. There were 1888 accessory genes and 557 unique genes. The functional analysis of the COGs in all the Elizabethkingia genomes revealed core, accessory and unique genes related to the regulation of metabolism, cellular processes and signaling, as well as various poorly characterized functions. Core genes were significantly enriched in pathways related to metabolism and biogenesis, including general function, amino acid transport and metabolism, translation, ribosomal structure and biogenesis, and cell wall/membrane/envelope biogenesis. Unique and accessory genes were significantly enriched in transcription; defense mechanisms; and replication, recombination and repair pathways.

Fig. 3
figure 3

Clusters of orthologous groups (COGs) in the core, accessory, and unique genomes and the associated Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of 90 Elizabethkingia strains. (A) Distribution of functional COGs in each core, accessory, and unique genome. (B) The detailed distribution of KEGG pathways with their functions. (C) The majority of core, accessory, and unique genes were associated with metabolism. (D) Functional annotations showing that the gene families associated with carbohydrate metabolism, amino acid metabolism, cofactor and vitamin metabolism, and energy metabolism accounted for the largest proportion of these 90 Elizabethkingia strains

The functions of COGs involved in information storage and processing are associated with intracellular survival. In addition, according to the functional prediction of genomes, general function prediction accounted for the largest proportion of COGs, followed by transcription and replication, recombination and repair. Regarding the constituents of each functional gene family, the core genes accounted for the largest proportion of genes related to transcription (11.7%); replication, recombination and repair (10.4%); cell wall, membrane, envelope and biogenesis (7.1%); and defense mechanisms (10.5%).

KEGG analysis

According to the KEGG analysis (Fig. 3B and D), the largest proportion of genes were enriched in metabolic functions. The other KEGG categories included cellular processes, environmental information processing, genetic information processing, human diseases and organismal systems. The core and accessory genes were most strongly associated with carbohydrate metabolism, followed by amino acid metabolism. Among the genes associated with carbohydrate metabolism, 15.3% were core genes, 12.7% were accessory genes, and 18.5% were unique genes. In addition, the majority of these core genes were also involved in carbohydrate metabolism, and the regulation of amino acid metabolism and lipid metabolism was the focus of the greatest number of genes, followed by signal transduction, replication and repair. In addition to metabolic functions, these genes were also associated with membrane transport, translation, and modulation of cellular growth and death. These functions collectively provide bacteria with the ability to withstand and adapt to the external environment. However, the unique genes in our 7 strains were mostly enriched in KEGG pathways related to microRNAs in cancer, drug resistance (β-lactam and vancomycin), ABC transporters, biological metabolism and biosynthesis, and nucleotide excision repair.

Discussion

Elizabethkingia spp. are responsible for serious nosocomial infections and outbreaks worldwide; however, they have received relatively little attention to date. In this study, we performed whole-genome sequencing and analyzed the characteristics of Elizabethkingia strains obtained from clinical samples. This allowed us to perform a comparative genomic analysis of representative clinical isolates alongside other isolates from the NCBI database around the world.

Risk factors for patients with Elizabethkingia infections primarily include antibiotic use for more than one week and having three or more underlying diseases. Other risk factors include chronic hemodialysis, multiple injuries, and immunosuppression with prolonged hospitalization. Additionally, in terms of patient diagnosis and treatment, most patients have undergone invasive operations. In this study, mechanical ventilation (71.8%, 51/71), urinary catheter placement (59.2%, 42/71) and nasogastric feeding tube placement (63.4%, 45/71) were identified as the main invasive operations. One of the most significant risk factors for Elizabethkingia infection is the use of mechanical ventilation. Elizabethkingia can form biofilms in moist environments or on water-related equipment, which facilitates its transmission in hospital settings [20]. The combination of these risk factors and a lack of effective therapeutic regimens can lead to a high mortality rate. In patients infected with Elizabethkingia, the mortality rate ranges from 20 to 40% [21]. The immune status of patients and virulence factors of microorganisms may be associated with the high fatality rate of patients with Elizabethkingia infections. Moreover, numerous previous studies have demonstrated that patients with Elizabethkingia infections frequently have chronic illnesses [22,23,24].

Elizabethkingia isolates often exhibit resistance to numerous antimicrobial agents, including β-lactams and inhibitors, aminoglycosides, macrolides, tetracycline, vancomycin, and carbapenems [25]. These strains exhibited variable susceptibilities to piperacillin, piperacillin-tazobactam, fluoroquinolones, minocycline, tigecycline, and trimethoprim-sulfamethoxazole. A wide variety of genes associated with drug resistance in the Elizabethkingia genus have been reported. MBLs are a global concern because they can confer resistance against carbapenems and almost all β-lactams. Therefore, evaluating clinical efficacy is crucial since all Elizabethkingia spp. appear to be inherent MBL producers [26]. Elizabethkingia is the only known organism to carry two distinct MBL chromosomes (blaBlaBand blaGOB) as well as the chromosomal blaCME gene that can confer resistance to cephalosporins [27, 28]. In addition, comparative analysis revealed that all 90 isolates carried the same subtype of blaBlaB, and 77.8% (70/71) of the isolates of Elizabethkingia spp. carried blaCME−1. However, various subtypes of the blaGOB gene, such as blaGOB−10, blaGOB−16, blaGOB−11, and blaGOB−1, were identified. Interestingly, genes capable of hydrolyzing cephalosporins, such as blaOXA−347 and blaTEM−116, were also identified, which are relatively rare in two Elizabethkingia spp. and were not identified in our 7 strains. Regardless of allelic combinations, MIC testing demonstrated that all isolates were highly resistant to carbapenems, penicillin and monobactams. In addition to resistance to carbapenems and β-lactams, several other isolates were found to be resistant to aminoglycosides and macrolides. The resistance genes detected in Elizabethkingia spp. included aadS, aph (3’’)-Ia, ermF, ereD, mef(C), and mph(G), and the specific genes aac(3)-IVb, aph(3’’)-Ia, aac(3)-IIIc, which conferred aminoglycoside resistance, were identified only in our 7 strains. The presence of multiple resistance genes contributes to the multidrug resistance of Elizabethkingia and increases the difficulty of clinical treatment [21]. Based on the results of drug sensitivity tests for this study and previously reported studies [29], the antimicrobial therapy options include minocycline, trimethoprim/sulfamethoxazole, cefoperazone/sulbactam, levofloxacin, colistin or tigecycline, either alone or in combination. Our in-hospital mortality rate of 29.6% (21/71) was lower than that reported in other studies, supporting the value of precision drug combinations in reducing mortality [30, 31].

The potential virulence factor homologs and their associated genes of the five Elizabethkingia species were predicted using the VFDB (Table S2). There was no significant difference in the distribution of virulence genes between the two groups of Elizabethkingia. These virulence factors include genes involved in capsular polysaccharide biosynthesis, chaperonin, elongation factor, heat shock protein, phospholipase, capsular polysaccharide, catalase, and peroxidase and are responsible for stress survival, immune modulation, and adherence to the environment [32]. Adherence-associated genes, including tufA, htpB and groEL, were widely distributed in all 90 strains, which indicates that these strains have a strong ability to colonize humans or other hosts [33]. In addition, the fcl gene encoding the GDP-L-fucose synthetase factor is related to immune modulation [34], but it was not detected in these seven strains. Further studies are necessary to investigate the potential function and source of this putative gene. These candidate genes may constitute promising targets for designing novel strategies to prevent and control infections in species belonging to this highly diverse and environmentally adaptable genus.

Elizabethkingia spp. have caused serious nosocomial infections and outbreaks worldwide, and the main aim of this study was to explore the diversity of the clinical characteristics or transmission events of Elizabethkingia species isolates from a global dataset. We constructed a phylogenetic tree of 90 Elizabethkingia strains based on ANI values, which clearly demonstrated the phylogenetic relationships among these strains. ANI analysis revealed that our seven strains belonged to the same or different species. In addition, comparative genome analysis revealed that the average GC content of E. meningoseptica was greater than that of the other four Elizabethkingia species, indicating a unique evolutionary pattern and potential receptiveness to mobile genetic elements from other strains.

Pangenome analysis has been used to evaluate genome diversity, genome dynamics, species evolution, pathogenesis and other features of microorganisms [35]. This approach helps us better understand the functional differences among these genes. The gene families of the pangenome represent metabolic capacity, while those of the core genome are typically related to bacterial replication, translation, and maintenance of cellular biogenesis [36]. The core genes are crucial for maintaining cell wall/envelope structures, which shield bacteria from environmental assaults. In our investigation, each strain’s unique genes were widely distributed and associated with functions such as transcription; DNA replication, recombination, and repair; and cell wall/membrane/envelope biogenesis. The unique genes involved in defense mechanisms are subject to relaxed mutation pressure and often correlate with pathogen virulence and pathogenicity. The core genes likely play essential roles in helping these intracellular pathogens survive, regulating cellular metabolism, and mediating cell membrane signal transduction events. However, the unique genes were enriched in poorly characterized functions, suggesting that further research should be conducted to explore their genomic function characteristics. This information might provide useful insights related to clinical infection control and treatment.

Conclusions

Elizabethkingia infections have become a significant public health concern, making it crucial to understand their clinical, molecular, and genetic characteristics. In this study, the complete genome sequences of 90 strains of Elizabethkingia were compared and analyzed. The findings contributed new knowledge to our understanding of the population, genome characteristics, and genetic roles of this emerging and life-threatening pathogen. These strains displayed diverse genetic compositions, which might contribute to the particular traits of these genera, as evidenced by the variations in their pangenome sizes.

Data availability

The assembled scaffold genome sequences for the seven Elizabethkingia species described in this study have been deposited in the NCBI GenBank database under accession numbers SAMN30678013 to SAMN30678019.

Abbreviations

CLSI:

Clinical & Laboratory Standards Institute

EUCAST:

The European Committee on Antimicrobial Susceptibility Testing

KEGG:

Kyoto Encyclopedia of Genes and Genomes

COGs:

Clusters of Orthologous Groups

ANI:

Average Nucleotide Identity

ESBL:

Extended-Spectrum β-Lactamase

MIC:

Minimal Inhibitory Concentration

BPGA:

Bacterial Pangenome Analysis

REALPHY:

Reference Sequence Alignment Phylogeny Builder

MBLs:

Metallo-β-lactamases

NCBI:

National Center for Biotechnology Information

BLAST:

Basic Local Alignment Search Tool

References

  1. Kim KK, Kim MK, Lim JH, Park HY, Lee ST. Transfer of Chryseobacterium meningosepticum and Chryseobacterium miricola to Elizabethkingia gen. nov. as Elizabethkingia meningoseptica comb. nov. and Elizabethkingia miricola comb. Nov. Int J Syst Evol MicroBiol. 2005;55(Pt 3):1287–93.

    Article  CAS  PubMed  Google Scholar 

  2. Bellais S, Aubert D, Naas T, Nordmann P. Molecular and biochemical heterogeneity of class B carbapenem-hydrolyzing beta-lactamases in Chryseobacterium meningosepticum. Antimicrob Agents Chemother. 2000;44(7):1878–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. King EO. Studies on a group of previously unclassified bacteria associated with meningitis in infants. Am J Clin Pathol. 1959;31(3):241–7.

    Article  CAS  PubMed  Google Scholar 

  4. Li Y, Kawamura Y, Fujiwara N, Naka T, Liu H, Huang X, et al. Chryseobacterium miricola sp. nov., a novel species isolated from condensation water of space station Mir. Syst Appl Microbiol. 2003;26(4):523–8.

    Article  CAS  PubMed  Google Scholar 

  5. Zdziarski P, Paściak M, Rogala K, Korzeniowska-Kowal A, Gamian A. Elizabethkingia miricola as an opportunistic oral pathogen associated with superinfectious complications in humoral immunodeficiency: a case report. BMC Infect Dis. 2017;17(1):763.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Kämpfer P, Matthews H, Glaeser SP, Martin K, Lodders N, Faye I. Elizabethkingia anophelis sp. nov., isolated from the midgut of the mosquito Anopheles gambiae. Int J Syst Evol Microbiol. 2011;61(Pt 11):2670–5.

    Article  PubMed  Google Scholar 

  7. Perrin A, Larsonneur E, Nicholson AC, Edwards DJ, Gundlach KM, Whitney AM, et al. Evolutionary dynamics and genomic features of the Elizabethkingia anophelis 2015 to 2016 Wisconsin outbreak strain. Nat Commun. 2017;8:15483.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nicholson AC, Gulvik CA, Whitney AM, Humrighouse BW, Graziano J, Emery B, et al. Revisiting the taxonomy of the genus Elizabethkingia using whole-genome sequencing, optical mapping, and MALDI-TOF, along with proposal of three novel Elizabethkingia species: Elizabethkingia bruuniana sp. nov., Elizabethkingia ursingii sp. nov., and Elizabethkingia occulta sp. nov. Antonie Van Leeuwenhoek. 2018;111(1):55–72.

    Article  CAS  PubMed  Google Scholar 

  9. Swami M, Mude P, Kar S, Sarathi S, Mohapatra A, Devi U, et al. Elizabethkingia meningoseptica Outbreak in NICU: an observational study on a Debilitating Neuroinfection in neonates. Pediatr Infect Dis J. 2024;43(1):63–8.

    Article  PubMed  Google Scholar 

  10. Chiang MH, Chang FJ, Kesavan DK, Vasudevan A, Xu H, Lan KL et al. Proteomic Network of Antibiotic-Induced Outer Membrane Vesicles released by extensively drug-resistant Elizabethkingia anophelis. Microbiol Spectr. 2022:e0026222.

  11. Andriyanov PA, Zhurilov PA, Kashina DD, Tutrina AI, Liskova EA, Razheva IV et al. Antimicrobial Resistance and Comparative Genomic Analysis of Elizabethkingia anophelis subsp. Endophytica isolated from raw milk. Antibiot (Basel). 2022;11(5).

  12. Tang HJ, Lin YT, Chen CC, Chen CW, Lu YC, Ko WC, et al. Molecular characteristics and in vitro effects of antimicrobial combinations on planktonic and biofilm forms of Elizabethkingia anophelis. J Antimicrob Chemother. 2021;76(5):1205–14.

    Article  CAS  PubMed  Google Scholar 

  13. Chen L, Yang J, Yu J, Yao Z, Sun L, Shen Y, et al. VFDB: a reference database for bacterial virulence factors. Nucleic Acids Res. 2005;33(Database issue):D325–8.

    Article  CAS  PubMed  Google Scholar 

  14. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinf (Oxford England). 2014;30(14):2068–9.

    CAS  Google Scholar 

  15. Chaudhari NM, Gupta VK, Dutta C. BPGA- an ultra-fast pan-genome analysis pipeline. Sci Rep. 2016;6:24373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bertels F, Silander OK, Pachkov M, Rainey PB, van Nimwegen E. Automated reconstruction of whole-genome phylogenies from short-sequence reads. Mol Biol Evol. 2014;31(5):1077–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel O. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol. 2010;59(3):307–21.

    Article  CAS  PubMed  Google Scholar 

  18. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing: 31th informational supplement M100-S31. Wayne, PA, USA: CLSI; 2021.

    Google Scholar 

  19. The European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 10.0. 2020. http://www.eucast.org.2020.

  20. Choi MH, Kim M, Jeong SJ, Choi JY, Lee IY, Yong TS, et al. Risk factors for Elizabethkingia Acquisition and clinical characteristics of patients, South Korea. Emerg Infect Dis. 2019;25(1):42–51.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Burnard D, Gore L, Henderson A, Ranasinghe A, Bergh H, Cottrell K et al. Comparative Genomics and Antimicrobial Resistance profiling of Elizabethkingia isolates reveal nosocomial transmission and in Vitro susceptibility to fluoroquinolones, tetracyclines, and Trimethoprim-Sulfamethoxazole. J Clin Microbiol. 2020;58(9).

  22. Seong H, Kim JH, Kim JH, Lee WJ, Ahn JY. M DN, Risk factors for mortality in patients with Elizabethkingia Infection and the clinical impact of the Antimicrobial susceptibility patterns of Elizabethkingia Species. J Clin Med. 2020;9(5).

  23. Tai IC, Liu TP, Chen YJ, Lien RI, Lee CY, Huang YC. Outbreak of Elizabethkingia meningoseptica sepsis with meningitis in a well-baby nursery. J Hosp Infect. 2017;96(2):168–71.

    Article  PubMed  Google Scholar 

  24. Reed TAN, Watson G, Kheng C, Tan P, Roberts T, Ling CL et al. Elizabethkingia anophelis Infection in Infants, Cambodia, 2012–2018. Emerging infectious diseases. 2020;26(2):320-2.

  25. Lin JN, Lai CH, Yang CH, Huang YH. Elizabethkingia Infections in humans: from Genomics to Clinics. Microorganisms. 2019;7(9).

  26. Chang TY, Chen HY, Chou YC, Cheng YH, Sun JR. In vitro activities of imipenem, Vancomycin, and rifampicin against clinical Elizabethkingia species producing BlaB and GOB metallo-beta-lactamases. Eur J Clin Microbiol Infect Diseases: Official Publication Eur Soc Clin Microbiol. 2019;38(11):2045–52.

    Article  CAS  Google Scholar 

  27. Opota O, Diene SM, Bertelli C, Prod’hom G, Eckert P, Greub G. Genome of the carbapenemase-producing clinical isolate Elizabethkingia miricola EM_CHUV and comparative genomics with Elizabethkingia meningoseptica and Elizabethkingia anophelis: evidence for intrinsic multidrug resistance trait of emerging pathogens. Int J Antimicrob Agents. 2017;49(1):93–7.

    Article  CAS  PubMed  Google Scholar 

  28. Wang L, Zhang X, Li D, Hu F, Wang M, Guo Q, et al. Molecular characteristics and Antimicrobial susceptibility profiles of Elizabethkingia Clinical isolates in Shanghai, China. Infect drug Resist. 2020;13:247–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lin J-N, Lai C-H, Huang Y-H, Yang C-H. Antimicrobial effects of Minocycline, Tigecycline, Ciprofloxacin, and levofloxacin against Elizabethkingia anophelis using in Vitro Time-kill assays and in vivo zebrafish animal models. Antibiotics. 2021;10(3).

  30. Rastogi N, Mathur P, Bindra A, Goyal K, Sokhal N, Kumar S, et al. Infections due to Elizabethkingia meningoseptica in critically injured trauma patients: a seven-year study. J Hosp Infect. 2016;92(1):30–2.

    Article  CAS  PubMed  Google Scholar 

  31. Huang YC, Lin YT, Wang FD. Comparison of the therapeutic efficacy of fluoroquinolone and non-fluoroquinolone treatment in patients with Elizabethkingia meningoseptica bacteraemia. Int J Antimicrob Agents. 2018;51(1):47–51.

    Article  CAS  PubMed  Google Scholar 

  32. Chen S, Soehnlen M, Blom J, Terrapon N, Henrissat B, Walker ED. Comparative genomic analyses reveal diverse virulence factors and antimicrobial resistance mechanisms in clinical Elizabethkingia meningoseptica strains. PLoS ONE. 2019;14(10):e0222648.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang L, Zhang TL, Xiang Q, Fu CX, Qiao M, Ding LJ, et al. Selective enrichment of virulence factor genes in the plastisphere under antibiotic and heavy metal pressures. J Hazard Mater. 2023;465:133319.

    Article  PubMed  Google Scholar 

  34. Fouts DE, Mongodin EF, Mandrell RE, Miller WG, Rasko DA, Ravel J, et al. Major structural differences and novel potential virulence mechanisms from the genomes of multiple campylobacter species. PLoS Biol. 2005;3(1):e15.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Guimarães LC, Florczak-Wyspianska J, de Jesus LB, Viana MV, Silva A, Ramos RT, et al. Inside the pan-genome - methods and Software Overview. Curr Genom. 2015;16(4):245–52.

    Article  Google Scholar 

  36. Kumar R, Bröms JE, Sjöstedt A. Exploring the Diversity within the Genus Francisella - An Integrated Pan-genome and Genome-Mining Approach. Front Microbiol. 2020;11:1928.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

I would like to thank the following people for helping with this research project: LX and QL collected the strains and performed the experiments, and WZ analyzed the experimental results. CW performed the bioinformatics analysis and wrote the manuscript. YX designed the study and reviewed the manuscript.

Corresponding author

Correspondence to Yi Xie.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Institutional Review Board of West China Hospital of Sichuan University (No. 2022 − 948). The hospital institutional review board waived the need for informed patient consent because retrospective analysis of the clinical data posed only a minimal risk to the subject and did not involve procedures that normally require written consent outside of the research context.

Consent for publication

Not applicable.

Conflict of interest

The authors declare that they have no conflicts of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Funding

This project was supported by the Special Foundation for National Science and Technology Basic Research Program of China (2019FY101200).

Electronic supplementary material

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, C., Xiong, L., Liao, Q. et al. Clinical manifestations, antimicrobial resistance and genomic feature analysis of multidrug-resistant Elizabethkingia strains. Ann Clin Microbiol Antimicrob 23, 32 (2024). https://doi.org/10.1186/s12941-024-00691-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12941-024-00691-6

Keywords