Skip to main content

Molecular analysis and distribution of multidrug-resistant Enterococcus faeciumisolates belonging to clonal complex 17 in a tertiary care center in Mexico City

Abstract

Background

Enterococcus faecium has recently emerged as a multidrug-resistant nosocomial pathogen involved in outbreaks worldwide. A high rate of resistance to different antibiotics has been associated with virulent clonal complex 17 isolates carrying the esp and hyl genes and the purK1 allele.

Results

Twelve clinical vancomycin-resistant Enterococcus faecium (VREF) isolates were obtained from pediatric patients at the Hospital Infantil de México Federico Gómez (HIMFG). Among these VREF isolates, 58.3% (7/12) were recovered from urine, while 41.7% (5/12) were recovered from the bloodstream. The VREF isolates showed a 100% rate of resistance to ampicillin, amoxicillin-clavulanate, ciprofloxacin, clindamycin, chloramphenicol, streptomycin, gentamicin, rifampicin, erythromycin and teicoplanin. In addition, 16.7% (2/12) of the isolates were resistant to linezolid, and 66.7% (8/12) were resistant to tetracycline and doxycycline. PCR analysis revealed the presence of the vanA gene in all 12 VREF isolates, esp in 83.3% (10/12) of the isolates and hyl in 50% (6/12) of the isolates. Phylogenetic analysis via molecular typing was performed using pulsed-field gel electrophoresis (PFGE) and demonstrated 44% similarity among the VREF isolates. MLST analysis identified four different sequence types (ST412, ST757, ST203 and ST612).

Conclusion

This study provides the first report of multidrug-resistant VREF isolates belonging to clonal complex 17 from a tertiary care center in Mexico City. Multidrug resistance and genetic determinants of virulence confer advantages among VREF in the colonization of their host. Therefore, the prevention and control of the spread of nosocomial infections caused by VREF is crucial for identifying new emergent subclones that could be challenging to treat in subsequent years.

Background

Enterococci are opportunistic pathogens of the normal intestinal microbiota of humans and animals [1, 2]. The most common species of Enterococcus involved in nosocomial infections is Enterococcus faecium (E. faecium) [1, 2]. This pathogen is associated with hospital-acquired infections such as UTIs (urinary tract infections), wounds, bacteremia, endocarditis and meningitis [1, 2].

In recent years, the emergence of multidrug-resistant E. faecium has increased [35]. The recommended treatment for Enterococcus infections has been penicillin alone or combined with aminoglycosides. However, due to increased resistance to aminoglycosides, vancomycin is currently the antibiotic employed to treat these infections. In the last several decades, the number of vancomycin-resistant enterococci (VRE) has increased. The first VRE isolates were reported in the United Kingdom in the late 1980s [6]. In the United States, more than 80% of E. faecium isolates from hospitals are now resistant to vancomycin, and virtually all of them (>90%) exhibit ampicillin resistance [7]. Vancomycin-resistant Enterococcus faecium (VREF) has been associated with outbreaks in hospitals worldwide [2]. The rates of VREF colonization and infection have risen steadily, with most cases being caused by strains displaying glycopeptide resistance to VanA and VanB [811].

In addition to multidrug resistance, E. faecium produces diverse factors that contribute to its pathogenesis, including virulence molecules such as secreted antigen SagA [12], cell wall-anchored collagen adhesin (Acm) [13], hyaluronidase (Hyl) [14] and enterococcal surface protein (Esp) [15]. However, the traits that contribute to the transition of E. faecium from a commensal to a nosocomial pathogen have not been identified [16].

Molecular typing methods are essential for identifying hospital-associated outbreaks of E. faecium. Multilocus sequence typing (MLST) has revealed the existence of host-specific genogroups, including a specific genetic lineage designated clonal complex 17, associated with hospital-related isolates [1, 17]. MLST of E. faecium is based on identifying alleles from DNA sequences in internal fragments of housekeeping genes (atpA, ddl, gdh, purK, gyd, pstS and adk), resulting in a numeric allelic profile, with each profile then being assigned a sequence type (ST) [17].

Complex 17 most likely evolved from the primary E. faecium ancestor ST-22, while ST-17 represents an important secondary founder with additional linages designated to complex 17 [18]. Clonal complex 17 is characterized by ampicillin and quinolone resistance and the presence of a putative pathogenicity island that includes the esp and/or hyl genes in the majority of isolates [1, 1820]. Various STs belonging to clonal complex 17, such as ST16, ST17, ST18, ST203 and ST412, are currently being disseminated worldwide [21, 22]. Interestingly, half of the STs within the clonal complex 17 polyclonal subpopulation have also been identified in samples obtained from healthy humans, swine, poultry and pets [16].

In Mexico, there is little available information about the prevalence of VREF isolates, and no study related to clonal complex 17 has been performed in pediatric patients. The aim of this study was to genotypically and phenotypically characterize VREF clinical isolates from 12 immunocompromised pediatric patients at the Hospital Infantil de México Federico Gómez (HIMFG). This study involved amplification of the resistance genes vanA and vanB and two virulence genes (esp and hyl) and molecular typing via pulsed-field gel electrophoresis (PFGE) and MLST.

Methods

Bacterial isolates

Twelve E. faecium isolates of clinical importance were obtained from 12 patients with nosocomial infections in the PICU (Pediatric Intensive Care Unit), oncology, gastroenterology and transplant wards of HIMFG during the period from July 2009 to April 2011. The isolates were maintained at −70°C in skim milk (Becton Dickinson, New Jersey, USA) and cultured on 5% sheep blood agar plates (Becton Dickinson, New Jersey, USA) at 37°C under 5% CO2 for 24 h. The E. faecalis ATCC® 29212, E. faecalis ATCC® 51299 and E. faecium ATCC® 51559 strains (American Type Culture Collection Manassas, VA, USA) were used as controls.

Biochemical tests

Bacteria were grown on blood agar, and identification was performed using manual methods. All colonies were grown in brain heart infusion broth (BHI) (Becton Dickinson, New Jersey, USA) with 6.5% NaCl and on bile esculin agar (Oxoid Sunnyvale, California, USA) to determine their hydrolysis grade. Disks impregnated with the substrate L-pyrrolidonyl-beta-naphthylamide were used to perform pyrrolidonase tests (Oxoid Biochemical Identification System, Oxoid LTD., Basingstoke, Hampshire, England). Reduction of tellurite (Merck, Darmstadt, Germany) was evaluated via growing the bacteria on 0.04% potassium tellurite.

Antibiotic susceptibility

The antibiotic susceptibility profiles of the 12 VREF isolates were determined via the minimum inhibitory concentration (MIC) technique by means of the microdilution method using Mueller-Hinton broth (MHB), as recommended by the Clinical and Laboratory Standards Institute. MIC tests were performed for vancomycin (MP Biomedicals, Solon, Ohio, USA), teicoplanin (Sigma-Aldrich, St. oLouis, Missouri, USA), chloramphenicol (MP Biomedicals, Solon, Ohio, USA), ciprofloxacin (MP Biomedicals, Solon, Ohio, USA), streptomycin (Alexis Biochemical, San Diego California, USA), linezolid (Sigma-Aldrich, St. Louis, Missouri, USA), rifampicin (MP, Biomedicals, Ohio, USA), nitrofurantoin (MP Biomedicals, Solon, Ohio, USA), tetracycline (MP Biomedicals, Solon, Ohio, USA), doxycycline (Sigma-Aldrich, St. Louis, Missouri, USA), erythromycin (MP Biomedicals, Solon, Ohio, USA), tigecycline (Sigma-Aldrich, St. Louis, Missouri, USA), gentamicin (MP Biomedicals, Solon, Ohio, USA) and amoxicillin-clavulanate (Glaxo-Smith-Kline, Philadelphia, Pennsylvania, USA). Several concentrations (256–0.625 μg/ml) of the antibiotics were tested in Mueller Hinton broth, with 100 μl of those dilutions being loaded into each well of a microplate. For each dilution, 100 μl of a bacterial suspension (1.5x108 CFU/ml) was inoculated and grown overnight at 37°C under a CO2 atmosphere. After bacterial growth was detected, the MIC for each isolate of E. faecium was reported as the highest concentration (μg/ml) of antibiotics in which no growth was observed. The E. faecalis ATCC® 29212 strain (American Type Culture Collection Manassas, VA, USA) was used as a control. These isolates were also evaluated for high-level aminoglycoside resistance (HLAR) to streptomycin (1,000 μg/ml) and gentamicin (500 μg/ml).

Detection of the glycopeptide resistance genes vanA and vanB

PCR was performed to detect the glycopeptide resistance genes vanA and vanB in the 12 E. faecium clinical isolates using specific primers (Table 1) [23]. Briefly, genomic DNA was purified using the Wizard Genomic DNA Purification Kit (Promega Madison, Wisconsin, USA) from a bacterial culture grown in BHI broth incubated at 37°C for 24 h. The amplification reactions were prepared in a final volume of 50 μl, as follows: 25 μl of amplification mix (22 mM Tris/HCl, pH 8.4; 55 mM KCl; 1.65 mM MgCl2; 25 μM each dNTP; 0.6 U recombinant Taq DNA polymerase/ml), 100 ng/μl of bacterial DNA, 10 μl of H2O and 5 μl of primer solution (10 pg/μl). A Perkin Elmer 9600 thermocycler was programmed to run for 30 cycles with the following parameters: denaturing at 94°C for 3 m, annealing at 55°C for 45 s and extension at 72°C for 1 m, with a final extension at 72°C for 2 m. The samples were analyzed via electrophoresis in 1% agarose gels (Agarose LE, Promega) using a 100 bp DNA ladder (Gibco/BRL Life Technologies, Breda, The Netherlands). E. faecium strain ATCC 51559 (vanA+) and E. faecalis strain ATCC® 51299 (vanB+) were used as controls in the PCR experiments [24].

Table 1 Primers sequences used in this study

PCR screening for the esp and hylgenes

DNA from bacterial cultures was extracted and amplified via PCR using primers for the esp Efm and hyl Efm genes (Table 1), generating bands of 954 bp and 661 bp, respectively [14, 25].

Molecular typing of VREF

PFGE of the 12 VREF clinical isolates was carried out following the protocols of Morrison et al. [26, 27]. Briefly, the samples were digested with 50 U of SmaI (New England Biolab, Ipswich, MA, USA) for 4 h at 25°C. The digested plugs were separated via electrophoresis in 1% agarose gels (BioRad, Hercules, California, USA) using ultra-pure DNA agarose (BioRad, Hercules, California, USA), with 0.5X TBE as the running buffer in the CHEF MAPPER system (BioRad Laboratories, Hercules, California, USA), run at 6 V/cm at 14°C under two different linear ramped pulse times: 1 to 10 s for 16 h and 10 to 40 s for 22 h. A PFGE lambda ladder (New England Biolabs, Hertfordshire, England, UK) was used as a molecular weight marker, and the gels were stained for 40 m with 0.5 mg/ml of ethidium bromide for visualization under UV light. The obtained banding patterns were initially interpreted via visual inspection according to the criteria specified by Tenover et al. [28]. Cluster analysis was performed with BioNumerics (Applied Maths, Inc., Austin, TX, USA) using the DICE correlation coefficient and the unweighted pair group mathematical average algorithm (UPGMA) as the grouping method [29].

The PFGE pulsotypes of the 12 VREF clinical isolates were also genotyped through multilocus sequence typing (MLST) according to a standard protocol described by Homan et al. [17]. Fragments of seven housekeeping genes (atpA, ddl, gdh, purK, gyd, pstS and adk) were sequenced using a 3730xl DNA Analyzer (Applied Biosystems, Foster City, California, USA), thus obtaining their allelic profiles, and the STs for each unique allelic profile were designated on the basis of information from the MLST website (http://efaecium.mlst.net).

Results

Origin of the strains

A total of 12 VREF clinical isolates obtained during the period from July 2009 to April 2011 were included in this study. The risk factors of the 12 patients were characterized by a minimum hospital stay of 4 days, assistance in the PICU and treatment with vancomycin. During their stay, the 12 patients were subjected to surgical procedures and received a central venous catheter, steroids and immunosuppressive treatment. Among the VREF isolates, 58.3% (7/12) were obtained from urine, while 41.6% (5/12) were obtained from the bloodstream. The VREF isolates were obtained from patients with different pathologies (Table 2).

Table 2 Characteristics of the 12 VREF isolates related to the patients’ clinical diagnosis, source of clinical samples, ward, PFGE, sequence type and clonal complex

Detection of susceptibility patterns and glycopeptide resistance in the VREF isolates

The results obtained for the 12 VREF clinical isolates showed a 100% rate of resistance to ampicillin, amoxicillin-clavulanate, ciprofloxacin, clindamycin, chloramphenicol, streptomycin, gentamicin, rifampicin, erythromycin and teicoplanin. The MIC values for each VREF isolate are presented in Table 3. In addition, 16.7% (2/12) of the VREF clinical isolates were resistant to linezolid, and 67% (8/12) were resistant to tetracycline and doxycycline (Table 3). However, all of the VREF isolates were susceptible to nitrofurantoin and tigecycline (Table 3). The HLAR values for gentamicin (500 μg/ml), streptomycin (1,000 μg/ml) and gentamicin/streptomycin (500/1,000 μg/ml) were determined with to 50% (6/12), 25% (3/12) and 25% (3/12), respectively.

Table 3 Minimum Inhibitory Concentration (MIC) to 12 clinical isolates of multidrug-resistant Enterococcus faecium

The vanA and vanB genes of the 12 VREF clinical isolates were amplified via PCR. Interestingly, only the vanA gene was detected in all the VREF clinical isolates, as a 1,030 bp amplicon (data not shown), whereas the vanB gene, with a length of 433 bp, was not identified in the isolates (data not shown). The E. faecium ATCC® 51559 (vanA+) and E. faecalis ATCC® 51299 (vanB+) strains were used as positive controls in the PCR assays [24].

Prevalence of the esp and hylvirulence genes in the VREF isolates

The esp and hyl virulence genes, which are associated with a clonal subcluster known as clonal complex 17 in VREF clinical isolates, were detected via PCR. The esp and hyl genes were highly prevalent in the isolates. The esp virulence gene was detected in 83.3% (10/12) of the isolates, and the hyl virulence gene was present in 50% (6/12) of them. Therefore, three genotypes were determined for the VREF clinical isolates: esp+/hyl-, esp+/hyl+ and esp-/hyl+, at prevalence rates of 50% (6/12), 33.3% (4/12) and 16.7% (2/12), respectively.

Molecular typing analysis of the E. faeciumisolates via PFGE and MLST

The VREF isolates were analyzed via PFGE following SmaI digestion of genomic DNA. Data obtained through PFGE were analyzed using a dendrogram profile, which included the PFGE pulsotypes obtained from VREF (Figure 1). A total of four clusters (I-IV) with five DNA pulsotypes were identified, showing patterns consisting of 12 to 20 DNA fragments ranging in size from 48.5 to 339.5 Kb (Figure 1). Interestingly, 25% (3/12) of the VREF clinical isolates observed via PFGE were categorized as pulsotype B and 16.7% (2/12) as pulsotype B1, with 92% genetic similarity being observed among these isolates (Figure 1). Meanwhile, 25% (3/12) of the VREF isolates were classified as pulsotype A, showing a different pattern from pulsotypes B, C and D (Figure 1). However, 16.7% (2/12) of the VREF isolates were classified as pulsotypes C and D, which displayed 50% genetic similarity. In addition, a maximum of 44% similarity was observed among all clusters of VREF isolates.

Figure 1
figure 1

PFGE analysis of 12 VREF isolates recovered at HIMFG and detection of the virulence factors esp and hyl , sequence type, isolation ward and type of sample. Phylogenetic analysis was performed using the DICE coefficient in association with the UPGMA algorithm as the grouping method. The dendrogram was evaluated by obtaining the cophenetic correlation coefficient using the Mantel test, which yielded an r value of 0.97769.

In this study, 12 VREF clinical isolates were subjected to MLST genotyping. Six of the 12 VREF isolates (50%) belonged to ST412, three to ST757, two to ST203 and one to ST612 (Table 2). eBURST analysis of the VREF isolates revealed four different STs (ST412, ST612, ST757 and ST203), three of which belonged to clonal complex 17; ST757 was not related to this clonal complex (Figure 2).

Figure 2
figure 2

Clustering of MLST profiles using the eBURST database algorithm. Our profiles showed that ST412, ST612 and ST203, but not ST757, belong to clonal complex 17.

Discussion

E. faecium is a highly resistant nosocomial pathogen and has recently emerged as an important threat in hospitals worldwide [2]. In this study, the 12 examined VREF isolates exhibited multidrug resistance to ampicillin, amoxicillin-clavulanate, ciprofloxacin, clindamycin, chloramphenicol, streptomycin, gentamicin, rifampicin, erythromycin and teicoplanin. At HIMFG, several types of enterococcal infections in pediatric patients are commonly treated with a combination of drugs (aminoglycoside-β-lactams, such as gentamicin/ampicillin) as the first choice, while vancomycin is the second choice; vancomycin-aminoglycoside or linezolid is the third choice; and tigecycline is the fourth choice. Interestingly, 16.7% (2/12) of the VREF clinical isolates were also resistant to linezolid, and 67% (8/12) were resistant to both tetracycline and doxycycline. The emergence of high levels of resistance to the most common anti-enterococcal antibiotics (vancomycin) might constitute a real challenge in the treatment of these infections. In the present study, 100% (12/12) of the examined VREF isolates were susceptible to tigecycline and nitrofurantoin. The VREF resistance patterns observed in this study are in agreement with the findings of other authors [30, 31]. However, these authors observed VREF isolates that were susceptible to linezolid and nitrofurantoin, in contrast to our data, which showed that two of the VREF isolates were resistant to linezolid. Nevertheless, the low resistance to linezolid observed in the VREF clinical isolates is in accord with data reported in other countries [11, 32].

Few instances of the isolation of HLAR E. faecium have been documented worldwide [22, 33, 34]. In this study, the examined VREF clinical isolates showed HLAR to gentamicin (500 μg/ml), streptomycin (1,000 μg/ml) and gentamicin/streptomycin (500/1,000 μg/ml), displaying resistance values of 50%, 25% and 25%, respectively. Treatment of severe enterococcal infection requires combined therapy to achieve a synergistic bactericidal effect [35]. However, the results obtained in cases of severe infections associated with enterococci have shown that HLAR should not be treated with combined therapy (gentamicin/ampicillin) [35]. Therefore, the treatment of HLAR E. faecium is restricted [36].

The enterococcal surface protein Esp, which is encoded by genes that appear to have been acquired and localized within a pathogenicity island, is commonly found in clinical isolates and anchors to the cell wall. This protein also affects biofilm formation and plays a role in experimental UTI and/or endocarditis models [2]. The presence of the esp gene has been associated with hospital outbreaks, although this gene is not exclusively found in epidemic strains [19, 30, 37, 38]. The esp gene was detected in 83.3% of our VREF clinical isolates. In addition, the majority of esp+ strains of E. faecium isolates were multidrug-resistant to more than three antibiotics, in accord with data reported by other researchers [3941].

On the other hand, the hyl gene was found in 50% of the VREF clinical isolates and displayed a higher prevalence compared to the prevalences of 29.8% (29/131) reported in isolates of E. faecium in the Picardy Region of France, 38% (83/220) in isolates from the US and 3% in European clinical isolates. However, in the United Kingdom, a hyl gene prevalence of 71% (20/28) was observed in E. faecium isolates [14, 42, 43]. We believe that the differences observed in the detection rates of the hyl gene are due to the region in which the samples were isolated. The rates of the occurrence of esp+/hyl-, esp+/hyl+ and esp-/hyl+ isolates were found to be 50% (6/12), 33.3% (4/12) and 16.7% (2/12), respectively, which is in accord with the findings of Vankerckhoven et al. and Rice et al. [14, 42, 44]. The VREF clinical isolates of Mexican origin in which the esp and/or hyl gene was amplified (alone or together), were resistant to more than three antibiotics; in contrast, other studies have shown a significant correlation between the presence of the esp gene and resistance to ampicillin, imipenem and ciprofloxacin [40, 41].

PFGE and MLST analyses have been proposed as alternative methods for the molecular characterization of clinical isolates of E. faecium[45]. According to our PFGE analysis, the 12 VREF isolates showed a heterogeneous pattern associated with a profile of multidrug resistance to different antibiotics and the presence of the vanA gene. The data obtained through PFGE revealed four clusters (I-IV), with a low similarity of 44% being detected among the VREF isolates and therefore high diversity. Furthermore, the VREF isolates within clusters I, II-B and III showed an identical banding profile in the PFGE analysis. However, the MLST data indicated different STs due to changes in the nucleotide sequences of the analyzed housekeeping genes; these data are consistent with the findings of Poh et al. [46]. In addition, the VREF isolates within clusters II-B1 and IV displayed identical PFGE and MLST profiles, in agreement with other authors [22, 33]. Nevertheless, pulsotypes from different wards showed similar multidrug resistance profiles, possibly due to horizontal genetic transference between these isolates.

MLST is an important tool for studying the molecular epidemiology of outbreaks of E. faecium and microbial population biology [44]. MLST analysis of VREF clinical isolates revealed four STs: ST203, ST412, ST612 and ST757. As previously reported, clonal complex 17 harbors various STs that have been involved in hospital outbreaks. Our results revealed two allelic profiles, ST203 and ST412, belonging to clonal complex 17 STs involved in hospital outbreaks. However, clonal complex 17 has been resolved into two different subgroups, one of which harbors ST17 and ST18, while the second harbors ST78 [47]. ST17, ST18 and ST203 are the major groups in the genetic lineage of E. faecium; they are distributed worldwide and have been associated with outbreaks [18, 48]. ST412 was the most frequent sequence type found in the VREF isolates from HIMFG and was genetically linked to the ST78 lineage. Interestingly, ST412 has been identified worldwide and associated with outbreaks [49]. According to the eBURST analysis, ST612 showed characteristics of the STs belonging to the 18 lineage. ST757 has not been characterized within clonal complex 17. In addition, ST757 displayed resistance markers (ampicillin and quinolones), virulence genes (esp+ and/or hyl+) and the purK1 allele; however, it has not been associated with outbreaks. Nevertheless, this community of multidrug-resistant strains is able to infect humans and might contribute to the spreading of these bacteria in the hospital, highlighting the importance of molecular typing via MLST to identify STs involved in nosocomial outbreaks.

Recently, it was shown that MLST analysis of typified E. faecium based on selected alleles may generate misleading results due to the recombination of five alleles (atpA, ddl, gdh, gyd and pstS). As only the purk and adk alleles are located in regions where there is no predicted recombination, the results must be interpreted with care [50]. The genome of E. faecium is highly plastic due to the few existing barriers to the acquisition of foreign genetic elements [51, 52]. Recent studies have provided evidence of high levels of recombination through comparative genomics analyses [5154]. Whole-genome sequencing platforms are superior to conventional typing methods, providing an excellent tool for determining phylogenies and regions of recombination and for accurately discriminating between outbreak- and non-outbreak-causing VREF isolates [50, 55]. Thus, whole-genome sequence information, rather than data on just one or a few genes, could be used to distinguish between closely related strains.

In this study, MLST and PFGE analysis were applied for the molecular characterization of clinical VREF isolates to identify different clonal complexes with different pulsotypes that were not related to outbreaks. However, according to the results obtained through PFGE, four multidrug-resistant clones of VREF were identified at HIMFG; in addition, these VREF isolates were identified at different periods. Therefore, these data suggest that these clones have circulated endemically at HIMFG.

In the case of cluster II, the clones have evolved from cluster II-B to cluster II-B1 due to the high similarity (> 90%) observed via PFGE analysis and based on the acquisition of three bands for B1, suggesting a mechanism of horizontal gene transfer. The results obtained in this study highlight the importance of monitoring circulating VREF isolates in different wards of this institution to efficiently control multidrug resistance and prevent outbreaks of these clones.

Conclusion

Little is known about VREF isolates in Mexican hospitals. In this study, the detected virulence genes (esp and hyl), multidrug profiles and allelic patterns were associated with clonal complex 17 VREF clinical isolates obtained from pediatric patients at HIMFG. To our knowledge, this is the first report describing clonal complex 17 VREF isolates in a tertiary care center in Mexico City.

Multidrug resistance and genetic determinants of virulence confer advantages in VREF in the colonization of their hosts. The genome of E. faecium is highly plastic, showing an ability to readily acquire genes involved in environmental persistence, colonization and virulence, favoring the selection of specific clonal complexes in a hospital environment. Therefore, the prevention and control of the propagation of nosocomial infections caused by VREF is crucial for identifying new emergent subclones that could be challenging to treat in subsequent years.

Ethics statement

The study was reviewed and approved by the Research (Dr. Onofre Muñoz Hernández), Ethics (Dr. Amparo Faure Fontenla) and Biosecurity (Dr. Herlinda Vera Hermosillo) Committee of HIMFG, under permit numbers HIM/2011/019. After looking at the medical history of each patient, E. faecium isolates were recovered from clinical samples, and the patients were asked by the physicians in the Infectology Department of HIMFG for their permission for their samples be used in this study. Analyses of E. faecium isolates obtained from clinical samples are not considered routine studies. Informed consent was obtained from the patient for the publication of this report and any accompanying images.

Abbreviations

Am:

Ampicillin

Amc:

Amoxacillin/clavulanate

CIP:

Ciprofloxacin

CC:

Clindamycin

C:

Chloramphenicol

GM:

Gentamicin

S:

Streptomycin

RA:

Rifampin

E:

Erythromycin

Va:

Vancomycin

TEI:

Teicoplanin

Te:

Tetracycline

D:

Doxycycline

LZN:

Linezolid

F/M:

Nitrofurantoin

TGC:

Tigecycline

Hyl:

E. faecium hyaluronidase

Esp:

Enterococcal surface protein.

References

  1. Top J, Willems R, Bonten M: Emergence of CC17 Enterococcus faecium: from commensal to hospital-adapted pathogen. FEMS Immunol Med Microbiol. 2008, 52 (3): 297-308. 10.1111/j.1574-695X.2008.00383.x.

    Article  PubMed  CAS  Google Scholar 

  2. Arias CA, Murray BE: The rise of the Enterococcus: beyond vancomycin resistance. Nat Rev Microbiol. 2012, 10 (4): 266-278. 10.1038/nrmicro2761.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  3. Grayson ML, Eliopoulos GM, Wennersten CB, Ruoff KL, De Girolami PC, Ferraro MJ, Moellering RC: Increasing resistance to beta-lactam antibiotics among clinical isolates of Enterococcus faecium: a 22-year review at one institution. Antimicrob Agents Chemother. 1991, 35 (11): 2180-2184. 10.1128/AAC.35.11.2180.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  4. Jones RN, Sader HS, Erwin ME, Anderson SC: Emerging multiply resistant enterococci among clinical isolates. I. Prevalence data from 97 medical center surveillance study in the United States. Enterococcus Study Group. Diagn Microbiol Infect Dis. 1995, 21 (2): 85-93. 10.1016/0732-8893(94)00147-O.

    Article  PubMed  CAS  Google Scholar 

  5. Rice LB: Emergence of vancomycin-resistant enterococci. Emerg Infect Dis. 2001, 7 (2): 183-187. 10.3201/eid0702.010205.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Uttley AH, Collins CH, Naidoo J, George RC: Vancomycin-resistant enterococci. Lancet. 1988, 1 (8575–6): 57-58.

    Article  PubMed  CAS  Google Scholar 

  7. Hidron AI, Edwards JR, Patel J, Horan TC, Sievert DM, Pollock DA, Fridkin SK: NHSN annual update: antimicrobial-resistant pathogens associated with healthcare-associated infections: annual summary of data reported to the national healthcare safety network at the centers for disease control and prevention, 2006–2007. Infect Control Hosp Epidemiol. 2008, 29 (11): 996-1011. 10.1086/591861.

    Article  PubMed  Google Scholar 

  8. Coque TM, Tomayko JF, Ricke SC, Okhyusen PC, Murray BE: Vancomycin-resistant enterococci from nosocomial, community, and animal sources in the United States. Antimicrob Agents Chemother. 1996, 40 (11): 2605-2609.

    PubMed  PubMed Central  Google Scholar 

  9. Rice LB: Antimicrobial resistance in gram-positive bacteria. Am J Med. 2006, 119 (6): S11-19-S62-70.

    Article  Google Scholar 

  10. Johnson AP, Uttley AH, Woodford N, George RC: Resistance to vancomycin and teicoplanin: an emerging clinical problem. Clin Microbiol Rev. 1990, 3 (3): 280-291.

    PubMed  CAS  PubMed Central  Google Scholar 

  11. Deshpande LM, Fritsche TR, Moet GJ, Biedenbach DJ, Jones RN: Antimicrobial resistance and molecular epidemiology of vancomycin-resistant enterococci from North America and Europe: a report from the SENTRY antimicrobial surveillance program. Diagn Microbiol Infect Dis. 2007, 58 (2): 163-170. 10.1016/j.diagmicrobio.2006.12.022.

    Article  PubMed  CAS  Google Scholar 

  12. Teng F, Kawalec M, Weinstock GM, Hryniewicz W, Murray BE: An Enterococcus faecium secreted antigen, SagA, exhibits broad-spectrum binding to extracellular matrix proteins and appears essential for E. faecium growth. Infect Immun. 2003, 71 (9): 5033-5041. 10.1128/IAI.71.9.5033-5041.2003.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Nallapareddy SR, Singh KV, Murray BE: Contribution of the collagen adhesin Acm to pathogenesis of Enterococcus faecium in experimental endocarditis. Infect Immun. 2008, 76 (9): 4120-4128. 10.1128/IAI.00376-08.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  14. Rice LB, Carias L, Rudin S, Vael C, Goossens H, Konstabel C, Klare I, Nallapareddy SR, Huang W, Murray BE: A potential virulence gene, hylEfm, predominates in Enterococcus faecium of clinical origin. J Infect Dis. 2003, 187 (3): 508-512. 10.1086/367711.

    Article  PubMed  CAS  Google Scholar 

  15. Heikens E, Bonten MJ, Willems RJ: Enterococcal surface protein Esp is important for biofilm formation of Enterococcus faecium E1162. J Bacteriol. 2007, 189 (22): 8233-8240. 10.1128/JB.01205-07.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  16. Willems RJ, Van Schaik W: Transition of Enterococcus faecium from commensal organism to nosocomial pathogen. Future Microbiol. 2009, 4 (9): 1125-1135. 10.2217/fmb.09.82.

    Article  PubMed  Google Scholar 

  17. Homan WL, Tribe D, Poznanski S, Li M, Hogg G, Spalburg E, Van Embden JD, Willems RJ: Multilocus sequence typing scheme for Enterococcus faecium. J Clin Microbiol. 2002, 40 (6): 1963-1971. 10.1128/JCM.40.6.1963-1971.2002.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  18. Willems RJ, Top J, Van Santen M, Robinson DA, Coque TM, Baquero F, Grundmann H, Bonten MJ: Global spread of vancomycin-resistant Enterococcus faecium from distinct nosocomial genetic complex. Emerg Infect Dis. 2005, 11 (6): 821-828. 10.3201/1106.041204.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  19. Leavis H, Top J, Shankar N, Borgen K, Bonten M, Van Embden J, Willems RJ: A novel putative enterococcal pathogenicity island linked to the esp virulence gene of Enterococcus faecium and associated with epidemicity. J Bacteriol. 2004, 186 (3): 672-682. 10.1128/JB.186.3.672-682.2004.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  20. Bonten MJ, Willems R, Weinstein RA: Vancomycin-resistant enterococci: why are they here, and where do they come from?. Lancet Infect Dis. 2001, 1 (5): 314-325. 10.1016/S1473-3099(01)00145-1.

    Article  PubMed  CAS  Google Scholar 

  21. Damani A, Klapsa D, Panopoulou M, Spiliopoulou I, Pantelidi K, Malli E, Kolonitsiou F, Grapsa S, Alepopoulou E, Frantzidou F, et al: A newly described vancomycin-resistant ST412 Enterococcus faecium predominant in Greek hospitals. Eur J Clin Microbiol Infect Dis. 2010, 29 (3): 329-331. 10.1007/s10096-009-0847-9.

    Article  PubMed  CAS  Google Scholar 

  22. Panesso D, Reyes J, Rincon S, Diaz L, Galloway-Pena J, Zurita J, Carrillo C, Merentes A, Guzman M, Adachi JA, et al: Molecular epidemiology of vancomycin-resistant Enterococcus faecium: a prospective, multicenter study in South American hospitals. J Clin Microbiol. 2010, 48 (5): 1562-1569. 10.1128/JCM.02526-09.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  23. Clark NC, Cooksey RC, Hill BC, Swenson JM, Tenover FC: Characterization of glycopeptide-resistant enterococci from U.S. hospitals. Antimicrob Agents Chemother. 1993, 37 (11): 2311-2317. 10.1128/AAC.37.11.2311.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  24. Kariyama R, Mitsuhata R, Chow JW, Clewell DB, Kumon H: Simple and reliable multiplex PCR assay for surveillance isolates of vancomycin-resistant enterococci. J Clin Microbiol. 2000, 38 (8): 3092-3095.

    PubMed  CAS  PubMed Central  Google Scholar 

  25. Shankar V, Baghdayan AS, Huycke MM, Lindahl G, Gilmore MS: Infection-derived Enterococcus faecalis strains are enriched in esp, a gene encoding a novel surface protein. Infect Immun. 1999, 67 (1): 193-200.

    PubMed  CAS  PubMed Central  Google Scholar 

  26. Morrison D, Woodford N, Barrett SP, Sisson P, Cookson BD: DNA banding pattern polymorphism in vancomycin-resistant Enterococcus faecium and criteria for defining strains. J Clin Microbiol. 1999, 37 (4): 1084-1091.

    PubMed  CAS  PubMed Central  Google Scholar 

  27. Turabelidze D, Kotetishvili M, Kreger A, Morris JG, Sulakvelidze A: Improved pulsed-field gel electrophoresis for typing vancomycin-resistant enterococci. J Clin Microbiol. 2000, 38 (11): 4242-4245.

    PubMed  CAS  PubMed Central  Google Scholar 

  28. Tenover FC, Arbeit RD, Goering RV, Mickelsen PA, Murray BE, Persing DH, Swaminathan B: Interpreting chromosomal DNA restriction patterns produced by pulsed-field gel electrophoresis: criteria for bacterial strain typing. J Clin Microbiol. 1995, 33 (9): 2233-2239.

    PubMed  CAS  PubMed Central  Google Scholar 

  29. Mullane NR, Whyte P, Wall PG, Quinn T, Fanning S: Application of pulsed-field gel electrophoresis to characterise and trace the prevalence of Enterobacter sakazakii in an infant formula processing facility. Int J Food Microbiol. 2007, 116 (1): 73-81. 10.1016/j.ijfoodmicro.2006.12.036.

    Article  PubMed  CAS  Google Scholar 

  30. Torres E, Perez S, Vindel A, Rodriguez-Bano J, Camba V, Villanueva R, Coque TM, Bou G: Glycopeptide-resistant Enterococcus faecium in a hospital in northern Spain. Molecular characterization and clinical epidemiology. Enferm Infecc Microbiol Clin. 2009, 27 (9): 511-517. 10.1016/j.eimc.2008.09.014.

    Article  PubMed  Google Scholar 

  31. Pourakbari B, Aghdam MK, Mahmoudi S, Ashtiani MT, Sabouni F, Movahedi Z, Alyari AE, Sadeghi RH, Mamishi S: High frequency of vancomycin-resistant Enterococcus faecalis in an Iranian referral children medical hospital. Maedica. 2012, 7 (3): 201-204.

    PubMed  PubMed Central  Google Scholar 

  32. Werner G, Klare I, Fleige C, Witte W: Increasing rates of vancomycin resistance among Enterococcus faecium isolated from German hospitals between 2004 and 2006 are due to wide clonal dissemination of vancomycin-resistant enterococci and horizontal spread of vanA clusters. Int J Med Microbiol. 2008, 298 (5–6): 515-527.

    Article  PubMed  CAS  Google Scholar 

  33. Weng PL, Ramli R, Shamsudin MN, Cheah YK, Hamat RA: High Genetic Diversity of Enterococcus faecium and Enterococcus faecalis Clinical Isolates by Pulsed-Field Gel Electrophoresis and Multilocus Sequence Typing from a Hospital in Malaysia. Biomed Res Int. 2013, 2013: 938937-

    Article  PubMed  PubMed Central  Google Scholar 

  34. Araoka H, Kimura M, Yoneyama A: A surveillance of high-level gentamicin-resistant enterococcal bacteremia. J Infect Chemother. 2011, 17 (3): 433-434. 10.1007/s10156-010-0175-0.

    Article  PubMed  Google Scholar 

  35. Murray BE: Vancomycin-resistant enterococcal infections. N Engl J Med. 2000, 342 (10): 710-721. 10.1056/NEJM200003093421007.

    Article  PubMed  CAS  Google Scholar 

  36. Watanabe S, Kobayashi N, Quinones D, Nagashima S, Uehara N, Watanabe N: Genetic diversity of enterococci harboring the high-level gentamicin resistance gene aac(6′)-Ie-aph(2″)-Ia or aph(2″)-Ie in a Japanese hospital. Microb Drug Resist. 2009, 15 (3): 185-194. 10.1089/mdr.2009.0917.

    Article  PubMed  CAS  Google Scholar 

  37. Leavis HL, Willems RJ, Top J, Spalburg E, Mascini EM, Fluit AC, Hoepelman A, De Neeling AJ, Bonten MJ: Epidemic and nonepidemic multidrug-resistant Enterococcus faecium. Emerg Infect Dis. 2003, 9 (9): 1108-1115. 10.3201/eid0909.020383.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Coque TM, Willems R, Canton R, Del Campo R, Baquero F: High occurrence of esp among ampicillin-resistant and vancomycin-susceptible Enterococcus faecium clones from hospitalized patients. J Antimicrob Chemother. 2002, 50 (6): 1035-1038. 10.1093/jac/dkf229.

    Article  PubMed  CAS  Google Scholar 

  39. Eaton TJ, Gasson MJ: A variant enterococcal surface protein Esp(fm) in Enterococcus faecium; distribution among food, commensal, medical, and environmental isolates. FEMS Microbiol Lett. 2002, 216 (2): 269-275. 10.1111/j.1574-6968.2002.tb11446.x.

    Article  PubMed  CAS  Google Scholar 

  40. Dupre I, Zanetti S, Schito AM, Fadda G, Sechi LA: Incidence of virulence determinants in clinical Enterococcus faecium and Enterococcus faecalis isolates collected in Sardinia (Italy). J Med Microbiol. 2003, 52 (Pt 6): 491-498.

    Article  PubMed  CAS  Google Scholar 

  41. Billstrom H, Lund B, Sullivan A, Nord CE: Virulence and antimicrobial resistance in clinical Enterococcus faecium. Int J Antimicrob Agents. 2008, 32 (5): 374-377. 10.1016/j.ijantimicag.2008.04.026.

    Article  PubMed  Google Scholar 

  42. Vankerckhoven V, Van Autgaerden T, Vael C, Lammens C, Chapelle S, Rossi R, Jabes D, Goossens H: Development of a multiplex PCR for the detection of asa1, gelE, cylA, esp, and hyl genes in enterococci and survey for virulence determinants among European hospital isolates of Enterococcus faecium. J Clin Microbiol. 2004, 42 (10): 4473-4479. 10.1128/JCM.42.10.4473-4479.2004.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  43. Biendo M, Adjide C, Castelain S, Belmekki M, Rousseau F, Slama M, Ganry O, Schmit JL, Eb F: Molecular characterization of glycopeptide-resistant enterococci from hospitals of the picardy region (france). Int J Microbiol. 2010, 2010: 150464-

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  44. Cha JO, Jung YH, Lee HR, Yoo JI, Lee YS: Comparison of genetic epidemiology of vancomycin-resistant Enterococcus faecium isolates from humans and poultry. J Med Microbiol. 2012, 61 (Pt 8): 1121-1128.

    Article  PubMed  CAS  Google Scholar 

  45. Kuriyama T, Williams DW, Patel M, Lewis MA, Jenkins LE, Hill DW, Hosein IK: Molecular characterization of clinical and environmental isolates of vancomycin-resistant Enterococcus faecium and Enterococcus faecalis from a teaching hospital in Wales. J Med Microbiol. 2003, 52 (Pt 9): 821-827.

    Article  PubMed  CAS  Google Scholar 

  46. Poh LW, Rukman AW, Cheah YK, Norital Z, Nazri AM, Mariana NS: Vancomycin-resistant Enterococcus faecium of multi locus sequence type 18 in Malaysia. Med J Malaysia. 2012, 67 (6): 639-640.

    PubMed  CAS  Google Scholar 

  47. Willems RJ, Top J, Van Schaik W, Leavis H, Bonten M, Siren J, Hanage WP, Corander J: Restricted gene flow among hospital subpopulations of Enterococcus faecium. MBio. 2012, 3 (4): e00151-00112.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  48. Werner G, Coque TM, Hammerum AM, Hope R, Hryniewicz W, Johnson A, Klare I, Kristinsson KG, Leclercq R, Lester CH, et al: Emergence and spread of vancomycin resistance among enterococci in Europe. Euro Surveill. 2008, 13: 47-

    Google Scholar 

  49. Freitas AR, Novais C, Ruiz-Garbajosa P, Coque TM, Peixe L: Dispersion of multidrug-resistant Enterococcus faecium isolates belonging to major clonal complexes in different Portuguese settings. Appl Environ Microbiol. 2009, 75 (14): 4904-4908. 10.1128/AEM.02945-08.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  50. Howden BP, Holt KE, Lam MM, Seemann T, Ballard S, Coombs GW, Tong SY, Grayson ML, Johnson PD, Stinear TP: Genomic insights to control the emergence of vancomycin-resistant enterococci. MBio. 2013, 4: 4-

    Article  Google Scholar 

  51. Galloway-Pena J, Roh JH, Latorre M, Qin X, Murray BE: Genomic and SNP analyses demonstrate a distant separation of the hospital and community-associated clades of Enterococcus faecium. PLoS One. 2012, 7 (1): e30187-10.1371/journal.pone.0030187.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  52. Palmer KL, Godfrey P, Griggs A, Kos VN, Zucker J, Desjardins C, Cerqueira G, Gevers D, Walker S, Wortman J, et al: Comparative genomics of enterococci: variation in Enterococcus faecalis, clade structure in E. faecium, and defining characteristics of E. gallinarum and E. casseliflavus. MBio. 2012, 3 (1): e00318-00311.

    Article  PubMed  PubMed Central  Google Scholar 

  53. De Been M, Van Schaik W, Cheng L, Corander J, Willems RJ: Recent recombination events in the core genome are associated with adaptive evolution in Enterococcus faecium. Genome Biol Evol. 2013, 5 (8): 1524-1535. 10.1093/gbe/evt111.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  54. Van Schaik W, Top J, Riley DR, Boekhorst J, Vrijenhoek JE, Schapendonk CM, Hendrickx AP, Nijman IJ, Bonten MJ, Tettelin H, et al: Pyrosequencing-based comparative genome analysis of the nosocomial pathogen Enterococcus faecium and identification of a large transferable pathogenicity island. BMC Genomics. 2010, 11: 239-10.1186/1471-2164-11-239.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Reuter S, Ellington MJ, Cartwright EJ, Koser CU, Torok ME, Gouliouris T, Harris SR, Brown NM, Holden MT, Quail M, et al: Rapid bacterial whole-genome sequencing to enhance diagnostic and public health microbiology. JAMA Intern Med. 2013, 173 (15): 1397-1404. 10.1001/jamainternmed.2013.7734.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Ma. del Carmen Castellanos Cruz and Ana Karina Espinosa Mazariego for their technical assistance in this study. This work was partially supported by grants HIM/2011/019 from Federal Funds from the Hospital Infantil de México Federico Gómez, CONACyT 133451 and the Fondo Sectorial de Investigación en Salud y Seguridad Social, FOSSIS-CONACYT 2012-01-182651.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juan Xicohtencatl-Cortes.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SAO, LBD and GE performed the susceptibility pattern analysis, molecular genetics experiments and PFGE and MLST assays. SAO, ZS and ACC participated in editing the manuscript and the data analysis. VCD, CAE, BLM, RHC and GAJ conducted the diagnoses of the patients, interpreted data, collaborated in the collection of samples and revised the manuscript. JXC is the principal investigator and conceived the study, designed the experiments, performed data analysis and wrote the manuscript. All authors read and approved the final version.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Cite this article

Ochoa, S.A., Escalona, G., Cruz-Córdova, A. et al. Molecular analysis and distribution of multidrug-resistant Enterococcus faeciumisolates belonging to clonal complex 17 in a tertiary care center in Mexico City. BMC Microbiol 13, 291 (2013). https://doi.org/10.1186/1471-2180-13-291

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2180-13-291

Keywords