pmc logo imageJournal ListSearchpmc logo image
Logo of jcmJ Clin Microbiol SubscriptionsJ Clin Microbiol Web Site
J Clin Microbiol. 1999 May; 37(5): 1591–1594.
PMCID: PMC84841
Note
Evaluation of MRSA-Screen, a Simple Anti-PBP 2a Slide Latex Agglutination Kit, for Rapid Detection of Methicillin Resistance in Staphylococcus aureus
Matthias Cavassini,1 Aline Wenger,1 Katia Jaton,1 Dominique S. Blanc,2 and Jacques Bille1*
Institut de Microbiologie1 and Division Autonome de Médecine Préventive Hospitalière,2 Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
*Corresponding author. Mailing address: Institut de Microbiologie, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland. Phone: 0041 21 314 40 57. Fax: 0041 21 314 40 60. E-mail: jacques.bille/at/chuv.hospvd.ch.
Received September 8, 1998; Revisions requested November 2, 1998; Accepted February 7, 1999.
Abstract
The MRSA-Screen test (Denka Seiken Co., Ltd., Tokyo, Japan), consisting of a slide latex agglutination kit that detects PBP 2a with a monoclonal antibody, was blindly compared to the oxacillin disk diffusion test, the oxacillin-salt agar screen, and PCR of the mecA gene for the detection of methicillin resistance in Staphylococcus aureus. A total of 120 methicillin-susceptible S. aureus (MSSA) and 80 methicillin-resistant S. aureus (MRSA) isolates, defined by the absence or presence of the mecA gene, respectively, were tested. The MRSA-Screen test, the oxacillin disk diffusion test, and the oxacillin-salt agar screening test showed sensitivities of 100, 61.3, and 82.5% and specificities of 99.2, 96.7, and 98.3%, respectively. We conclude that the MRSA-Screen is a very accurate, reliable, and fast test (15 min) for differentiation of MRSA from MSSA colonies on agar plates.
 
Resistance to methicillin (and to all β-lactam antibiotics) in Staphylococcus aureus is primarily associated with the acquisition of the mecA gene coding for the penicillin-binding protein 2a (PBP 2a), involved in bacterial cell wall synthesis (1, 13, 31). The detection of methicillin resistance, however, is complicated by the fact that its phenotypic expression in many strains is heterogeneous (9, 14). This has resulted in the development of various laboratory techniques to enhance the expression of this resistance in vitro (2, 6, 17). mecA gene detection tests based on PCR or DNA hybridization performed only by specialized laboratories has proved to be more specific and sensitive than conventional tests, particularly in very heterogeneous strains (16, 22). So far, no simple and rapid method aimed at the direct detection of PBP 2a has been commercialized (11). The purpose of the present study is to test such a candidate.

Bacterial isolates. A total of 200 S. aureus clinical isolates collected between 1987 and 1998 from our hospital and 15 neighboring hospitals were used in this study. All isolates were identified by conventional tests. Isolates from the neighboring hospitals (n = 33) were sent to our laboratory because of the difficulty of assessing susceptibility to oxacillin by phenotypic methods. The definitions of methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) were based on the presence or absence, respectively, of the mecA gene by PCR. Among 120 MSSA isolates (19 penicillin susceptible and 101 penicillin resistant), 40 were susceptible to all non-β-lactam antibiotics tested, 51 showed resistance to one, and 29 showed resistance to more than one non-β-lactam antibiotic. Eighty MRSA isolates were carefully selected on the basis of molecular typing (60 different pulsed-field gel electrophoresis [PFGE] patterns) and on the basis of various levels of heterogeneous resistance to oxacillin (35 heterogeneously and 45 homogeneously resistant isolates).

Before being tested, the isolates were removed from storage (−80°C), streaked onto Columbia blood agar plates, and incubated under aerobic conditions at 35°C for 24 h. An isolated colony was picked from each plate, streaked onto two new Columbia blood agar plates, and incubated for 24 h. All inocula were prepared from these subcultures. All isolates were blindly tested. Two control strains, one MRSA (ATCC 33591) and one MSSA (ATCC 29213) strain, were included in each batch.

MRSA-Screen test. The MRSA-Screen test (Denka Seiken Co., Ltd., Tokyo, Japan) was performed according to the manufacturer’s instructions by batches of 20 isolates, including the control strains. The sample preparation was made as follows. Ten to 20 S. aureus colonies from a fresh blood agar plate were suspended in a 1.5-ml microtube containing 4 drops (200 μl) of extraction reagent no. 1 (0.1 M NaOH). The suspension was boiled for 3 min, and then 1 drop (50 μl) of extraction reagent no. 2 (0.5 M KH2PO4) was added and mixed well. After a centrifugation step (at 1,500 × g for 5 min at room temperature), 50 μl of the supernatant was placed on the slide for testing and mixed with 1 drop (25 μl) of anti-PBP 2a monoclonal antibody-sensitized latex. For the negative control, 50 μl of the supernatant was placed on the slide for testing and mixed with 1 drop (25 μl) of negative-control latex. Mixing for 3 min was performed with a shaker. When agglutination occurred within 3 min, it was visually quantified as a score between 1+ and 3+. All the isolates were tested twice, and the results were interpreted blindly by two different persons.

Phenotypic methods. The oxacillin disk diffusion test and oxacillin-salt agar screening test were carried out on all the isolates according to the recommendations of the National Committee for Clinical Laboratory Standards (NCCLS) (2325). Both tests were read after 24 h of incubation. Susceptibility to 11 other antibiotics (penicillin, cephalothin, ceftriaxone, gentamicin, ciprofloxacin, clindamycin, fusidic acid, erythromycin, trimethoprim-sulfamethoxazole, rifampin, and vancomycin) was also tested by the disk diffusion method according to NCCLS recommendations.

mecA gene. Detection of the mecA gene was performed blindly for all isolates by using the method described by Tokue et al. with some modifications (28). The extraction technique was simplified by directly suspending 2 to 5 colonies in 200 μl of water; the suspension was diluted 1:10 and 1:100 in water, and the DNA was released by boiling the suspensions for 5 min at 95°C. One positive MRSA control strain (ATCC 33591), one negative MSSA control strain (ATCC 25923), and water as an extraction control were included in each run.

Molecular typing. Molecular typing was performed by PFGE on all MRSA isolates (4).

Repeat testing. When S. aureus isolates yielded discrepant results among the MRSA-Screen test, oxacillin-salt agar screening test, and mecA gene detection, the tests were repeated blindly twice from the same Columbia blood agar plate.

β-Lactamase testing. Chromogenic nitrocefin disks (Cefinase; BBL Becton Dickinson, Cockeysville, Md.) were used according to the manufacturer’s instructions to test MSSA isolates that appeared resistant to oxacillin by phenotypic methods.

E-tests. Oxacillin MICs were measured for all isolates showing discordant results between phenotypic methods and the MRSA-Screen. E-tests (AB Biodisk, Solna, Sweden) were performed according to the manufacturer’s advice on Mueller-Hinton agar supplemented with 2% NaCl and read after 24 h of incubation. In addition, penicillin, amoxicillin, and amoxicillin-clavulanate MICs were measured for the four MSSA isolates that appeared resistant to oxacillin by phenotypic methods.

Results. Table 1 summarizes the results obtained with the MRSA-Screen test, oxacillin disk diffusion method, and oxacillin-salt agar screening test. Table 2 lists the isolates with discrepant results. The positive and negative control strains, included blindly in each of 11 batches of MRSA-screen tests, gave the expected results. No agglutination occurred when the 200 S. aureus isolates were tested with the negative-control latex. The MRSA-Screen test by slide latex agglutination showed complete concordance with the detection by PCR of the mecA gene, except for one false-positive result among the 120 mecA gene-negative S. aureus isolates tested. This false-positive agglutination was graded 1+ and gave no agglutination upon retesting. Results of duplicate testing were in agreement for the remaining 199 isolates, which proves the good reproducibility of the test (99.5%). Quantification of the agglutination test reaction (1+ to 3+) for the 80 MRSA isolates did not correlate with their level of heterogeneous resistance to methicillin. Among the 35 heterogeneously resistant MRSA isolates, 31 showed a 3+ agglutination test reaction and 4 showed a 2+ reaction. Of the remaining 45 isolates homogeneously resistant to methicillin, one showed a 1+, 6 showed a 2+, and 38 showed a 3+ agglutination test reaction. Upon retesting, the grading of the agglutination reaction was identical for 69 of 80 MRSA isolates; of the remaining 11 isolates (4 heterogeneously resistant and 7 homogeneously resistant), the grading changed from 3+ to 2+ or from 2+ to 3+ for 10 and from 1+ to 2+ for the last isolate.

TABLE 1TABLE 1
Comparison of the MRSA-Screen test and two phenotypic tests to classify 200 S. aureus clinical isolates as MRSA or MSSAa
TABLE 2TABLE 2
Discrepancies between the MRSA-Screen test, mecA gene detection by PCR, and phenotypic tests for the detection of methicillin resistance among 200 S. aureus clinical isolates

Of 80 true MRSA isolates, the oxacillin disk diffusion method falsely read 20 as MSSA (false negative), with inhibition zone diameters ranging from 14 to 27 mm (Table 2). All of them were heterogeneously resistant, with oxacillin MICs ranging from 0.25 to 3.0 μg/ml by the E-test. These 20 isolates were easily classified as MRSA by the MRSA-Screen test. Among them, only six were also detected as MRSA by the oxacillin-salt agar screening test read after 24 h of incubation.

Among 120 true MSSA isolates, the oxacillin disk diffusion method falsely classified 4 as MRSA (false positive), with inhibition zone diameters between 6 and 8 mm. Two of these isolates were correctly classified as MSSA by the oxacillin-salt agar screening test, whereas the two remaining isolates were also false positive by the latter method. Oxacillin MICs by the E-test ranged from 2.0 to 3.0 μg/ml for these four isolates. Production of β-lactamase, associated with a reduction in the MIC from 24 to 1 μg/ml when clavulanate was added to amoxicillin, was demonstrated in only one isolate. Overproduction of β-lactamase in this isolate may contribute to the oxacillin resistance observed with the oxacillin disk diffusion test (21). The three remaining strains were not β-lactamase producers and could thus be classified as MOD-SA (modified S. aureus) isolates (15, 29).

Discussion. The MRSA-Screen test detected all the 80 MRSA isolates and misclassified as MRSA only 1 of 120 MSSA isolates. Compared to the standard agar disk diffusion test and to the oxacillin-salt agar screening test, the MRSA-Screen test showed higher sensitivity and specificity. Fourteen heterogeneously resistant isolates, while they were mecA gene positive by PCR, did not express any resistance to oxacillin by the conventional phenotypic methods. However, these mecA gene-positive isolates were categorized as MRSA by the MRSA-Screen test, which detects PBP 2a, confirming that expression of the resistance is under the control of other determinants, such as the BlaI, mecR1-mecI, and fem genes, whose mechanisms and interactions are still not fully understood (3, 8, 12, 18).

Technically the test was easy to perform, requiring only microtubes, boiling water, a table centrifuge, and a manual pipetter. The test gave results in 15 min, making it the fastest test to our knowledge to reliably detect oxacillin resistance in S. aureus isolates. Even if most MRSA isolates agglutinated after 30 s of mixing time, some needed almost the 3 min recommended by the manufacturer to produce visible agglutination. The correct inoculum is also important, as at the beginning of the study we experienced two false-positive agglutination reactions when the inoculum used was about 10 times heavier than that recommended. The influences of various solid growth media on the sensitivity and specificity of the MRSA-Screen test were not investigated in the present study.

As already reported, the oxacillin disk diffusion test was the least reliable test for detection of resistance to oxacillin in S. aureus (5, 20, 30). Of the 80 MRSA isolates, 20 had inhibitory-zone diameters in the susceptible range (≥13 mm), 11 in the intermediate range (11 to 12 mm), and 49 in the resistant range (≤10 mm). As recommended by the NCCLS, we looked for associated resistance and cross-resistance to other antibiotics as clues to suspect methicillin resistance. Of the 11 antibiotics tested in addition to oxacillin, ceftriaxone gave the best indication for oxacillin resistance when both intermediate and resistant zone diameters were considered as clues for oxacillin resistance. Among 120 MSSA isolates, the ceftriaxone inhibitory-zone diameters would have suggested resistance to oxacillin in only 4 cases. Of these four, two were also falsely resistant to oxacillin by the oxacillin disk diffusion test. Among 80 MRSA isolates, only 4 were ceftriaxone susceptible, 3 of which were also susceptible to oxacillin by disk diffusion. Although associated resistance to non-β-lactam antibiotics is often found among MRSA isolates, there were 13 MRSA isolates without any associated resistance in this study.

The oxacillin-salt agar screening test is recognized as a sensitive and specific test (7, 10, 27). In our study the sensitivity was low (82.5%). This is due to a bias of selection of the MRSA isolates, as we included strains that were difficult to detect as being resistant to methicillin by the oxacillin-salt agar screening test and oxacillin disk diffusion. However, 13 of the 14 MRSA isolates not detected by the oxacillin-salt agar screen test after 24 h of incubation showed clear growth after reincubation for an additional 24 h. Therefore, in order to further increase the sensitivity of the test, we suggest looking for growth after 24 and 48 h of incubation. In our study, reading all the oxacillin-salt agar screening plates after 48 h of incubation would not have decreased the specificity of the test compared to reading after 24 h of incubation.

Many methods have been evaluated for more accurate detection of methicillin resistance in S. aureus (10, 19, 26). The cost and workload of DNA probes or PCR to detect the mecA gene have prevented their broad use in a clinical microbiology laboratory. Therefore, detection of PBP 2a with this simple agglutination kit offers an interesting new approach to the rapid characterization of S. aureus as MRSA or MSSA. The test has the major advantage over the phenotypic methods of not being influenced by the various levels of expression of the resistance, a parameter which in highly heterogeneously resistant isolates tends to render classical and automated methods less accurate (11). When applied to overnight primary culture agar media, the MRSA-Screen test will shorten the delay for the detection of MRSA to 1 day, versus 2 or 3 days for conventional methods, a potentially significant improvement for both directed antibiotherapy and epidemiological measures.

Acknowledgments

This work was supported in part by Pharma Consulting, Burgdorf, Switzerland.

We thank Marica Galazzo, Patricia Rudaz, and Dorothée Raffalli for their valuable technical help in this study.

REFERENCES
1.
Archer, G L; Pennell, E. Detection of methicillin resistance in staphylococci by using a DNA probe. Antimicrob Agents Chemother. 1990;34:1720–1724. [PubMed]
2.
Baker, C N; Huang, M B; Tenover, F C. Optimizing testing of methicillin-resistant Staphylococcus species. Diagn Microbiol Infect Dis. 1994;19:167–170. [PubMed]
3.
Berger-Bächi, B. Update on methicillin resistance mechanisms in staphylococci. Chemotherapy. 1996;42(Suppl. 2):19–26.
4.
Brosch, R; Buchrieser, C; Rocourt, J. Subtyping of Listeria monocytogenes serovar 4b by use of low-frequency-cleavage restriction endonucleases and pulsed-field gel electrophoresis. Res Microbiol. 1991;142:667–675. [PubMed]
5.
Chambers, H F. Detection of methicillin resistant staphylococci. Infect Dis Clin N Am. 1993;7:425–433. [PubMed]
6.
Chambers, H F; Hackbarth, C J. Effect of NaCl and nafcillin on penicillin-binding protein 2a and heterogeneous expression of methicillin resistance in Staphylococcus aureus. Antimicrob Agents Chemother. 1987;31:1982–1988. [PubMed]
7.
Chambers, H F. Methicillin resistance in staphylococci: molecular and biochemical basis and clinical implications. Clin Microbiol Rev. 1997;10:781–791. [PubMed]
8.
de Lencastre, H; Tomasz, A. Reassessment of the number of auxiliary genes essential for expression of high-level methicillin resistance in Staphylococcus aureus. Antimicrob Agents Chemother. 1994;38:2590–2598. [PubMed]
9.
de Lencastre, H; Figueiredo, A M S; Urban, C; Rahal, J; Tomasz, A. Multiple mechanisms of methicillin resistance and improved methods for detection in clinical isolates of Staphylococcus aureus. Antimicrob Agents Chemother. 1991;35:632–639. [PubMed]
10.
Dillard, S C; Waites, K B; Brookings, E S; Moser, S A. Detection of oxacillin-resistance in Staphylococcus aureus by MicroScan MIC panels in comparison to four other methods. Diagn Microbiol Infect Dis. 1996;24:93–100. [PubMed]
11.
Gerberding, J L; Miick, C; Liu, H H; Chambers, H F. Comparison of conventional susceptibility tests with direct detection of penicillin-binding protein 2a in borderline oxacillin-resistant strains of Staphylococcus aureus. Antimicrob Agents Chemother. 1991;35:2574–2579. [PubMed]
12.
Hackbarth, C J; Chambers, H F. blaI and blaR1 regulate β-lactamase and PBP 2a production in methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 1993;37:1144–1149. [PubMed]
13.
Hartman, B J; Tomasz, A. Low-affinity penicillin-binding protein associated with β-lactam resistance in Staphylococcus aureus. J Bacteriol. 1984;158:513–516. [PubMed]
14.
Hartman, B J; Tomasz, A. Expression of methicillin resistance in heterogeneous strains of Staphylococcus aureus. Antimicrob Agents Chemother. 1986;29:85–92. [PubMed]
15.
Henze, U U; Berger-Bächi, B. Penicillin-binding protein 4 overproduction increases β-lactam resistance in Staphylococcus aureus. Antimicrob Agents Chemother. 1996;40:2121–2125. [PubMed]
16.
Hiramatsu, K; Kihara, H; Yokota, T. Analysis of borderline-resistant strains of methicillin-resistant Staphylococcus aureus using polymerase chain reaction. Microbiol Immunol. 1992;36:445–453. [PubMed]
17.
Huang, M B; Gay, T E; Baker, C N; Banerjee, S N; Tenover, F C. Two percent sodium chloride is required for susceptibility testing of staphylococci with oxacillin when using agar-based dilution methods. J Clin Microbiol. 1993;31:2683–2688. [PubMed]
18.
Hürlimann-Dalel, R L; Ryffel, C; Kayser, F H; Berger-Bächi, B. Survey of the methicillin resistance-associated genes mecA, mecR1-mecI, and femA-femB in clinical isolates of methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 1992;36:2617–2621. [PubMed]
19.
Knapp, C C; Ludwig, M D; Washington, J A. Evaluation of BBL Crystal MRSA ID system. J Clin Microbiol. 1994;32:2588–2589. [PubMed]
20.
Knapp, C C; Ludwig, M D; Washington, J A; Chambers, H F. Evaluation of the Vitek GPS-SA card for testing of oxacillin against borderline-susceptible staphylococci that lack mec. J Clin Microbiol. 1996;34:1603–1605. [PubMed]
21.
McDougal, L K; Thornsberry, C. The role of β-lactamase in staphylococcal resistance to penicillinase-resistant penicillins and cephalosporin. J Clin Microbiol. 1986;23:832–839. [PubMed]
22.
Murakami, K; Minamide, W; Wada, K; Nakamura, E; Teraoka, H; Watanabe, S. Identification of methicillin-resistant strains of staphylococci by polymerase chain reaction. J Clin Microbiol. 1991;29:2240–2244. [PubMed]
23.
National Committee for Clinical Laboratory Standards. Performance standards for antimicrobial disk susceptibility tests, 6th ed., vol. 17, no. 1. Approved standard M2-A6. Wayne, Pa: National Committee for Clinical Laboratory Standards; 1997.
24.
National Committee for Clinical Laboratory Standards. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically, 4th ed., vol. 17, no. 2. Approved standard M7-A4. Wayne, Pa: National Committee for Clinical Laboratory Standards; 1997.
25.
National Committee for Clinical Laboratory Standards. Performance standards for antimicrobial susceptibility testing, vol. 18, no. 1. Eighth informational supplement. M100-S8. Wayne, Pa: National Committee for Clinical Laboratory Standards; 1998.
26.
Petersson, A; Miorner, H; Kamme, C. Identification of mecA-related oxacillin resistance in staphylococci by the E test and the broth microdilution method. J Antimicrob Chemother. 1996;37:445–456. [PubMed]
27.
Skulnick, M; Simor, A E; Gregson, D; Patel, M; Small, G W; Kreiswirth, B; Hathoway, D; Low, D E. Evaluation of commercial and standard methodology for determination of oxacillin susceptibility in Staphylococcus aureus. J Clin Microbiol. 1992;30:1985–1988. [PubMed]
28.
Tokue, Y; Shoji, S; Satoh, K; Watanabe, A; Motomiya, M. Comparison of a polymerase chain reaction assay and a conventional microbiologic method for detection of methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 1992;36:6–9. [PubMed]
29.
Tomasz, A; Drugeon, H B; de Lencastre, H M; Jabes, D; McDougall, L; Bille, J. New mechanism for methicillin resistance in Staphylococcus aureus: clinical isolates that lack the PBP 2a gene and contain normal penicillin-binding proteins with modified penicillin-binding capacity. Antimicrob Agents Chemother. 1989;33:1869–1874. [PubMed]
30.
Ünal, S; Werner, K; DeGirolami, P; Barsanti, F; Eliopoulos, G. Comparison of tests for detection of methicillin-resistant Staphylococcus aureus in a clinical microbiology laboratory. Antimicrob Agents Chemother. 1994;38:345–347. [PubMed]
31.
Utsui, Y; Yokota, T. Role of an altered penicillin-binding protein in methicillin- and cephem-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 1985;28:397–403. [PubMed]