Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2011 Sep;55(9):4012–4018. doi: 10.1128/AAC.00223-11

In Vitro Cross-Resistance to Daptomycin and Host Defense Cationic Antimicrobial Peptides in Clinical Methicillin-Resistant Staphylococcus aureus Isolates

Nagendra N Mishra 1,*, James McKinnell 1, Michael R Yeaman 1,2, Aileen Rubio 3, Cynthia C Nast 2,4, Liang Chen 5, Barry N Kreiswirth 5, Arnold S Bayer 1,2
PMCID: PMC3165344  PMID: 21709105

Abstract

We investigated the hypothesis that methicillin-resistant Staphylococcus aureus (MRSA) isolates developing reduced susceptibilities to daptomycin (DAP; a calcium-dependent molecule acting as a cationic antimicrobial peptide [CAP]) may also coevolve reduced in vitro susceptibilities to host defense cationic antimicrobial peptides (HDPs). Ten isogenic pairs of clinical MRSA DAP-susceptible/DAP-resistant (DAPs/DAPr) strains were tested against two distinct HDPs differing in structure, mechanism of action, and origin (thrombin-induced platelet microbicidal proteins [tPMPs] and human neutrophil peptide-1 [hNP-1]) and one bacterium-derived CAP, polymyxin B (PMB). Seven of 10 DAPr strains had point mutations in the mprF locus (with or without yyc operon mutations), while three DAPr strains had neither mutation. Several phenotypic parameters previously associated with DAPr were also examined: cell membrane order (fluidity), surface charge, and cell wall thickness profiles. Compared to the 10 DAPs parental strains, their respective DAPr strains exhibited (i) significantly reduced susceptibility to killing by all three peptides (P < 0.05), (ii) increased cell membrane fluidity, and (iii) significantly thicker cell walls (P < 0.0001). There was no consistent pattern of surface charge profiles distinguishing DAPs and DAPr strain pairs. Reduced in vitro susceptibility to two HDPs and one bacterium-derived CAP tracked closely with DAPr in these 10 recent MRSA clinical isolates. These results suggest that adaptive mechanisms involved in the evolution of DAPr also provide MRSA with enhanced survivability against HDPs. Such adaptations appear to correlate with MRSA variations in cell membrane order and cell wall structure. DAPr strains with or without mutations in the mprF locus demonstrated significant cross-resistance profiles to these unrelated CAPs.

INTRODUCTION

Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are often a challenge for clinicians due to limited treatment options and resistance to multiple antibiotics (15, 34). In this regard, daptomycin (DAP; a calcium-dependent antimicrobial lipopeptide) has become a relevant therapeutic option (28, 29).

Cationic antimicrobial host defense peptides (HDPs), including those of hematogenous origin, such as platelets and neutrophils, kill many important blood-borne pathogens, especially S. aureus (35). Although most HDPs initially interact with the bacterial cell membrane, their overall mechanism(s) of action can be quite distinct and multifactorial, involving the cell membrane, the cell wall, and intracellular targets (37). Since both DAP and HDPs target the bacterial cell membrane to initiate their lethal mechanism(s), we hypothesized that common resistance pathways between these molecules might exist (21, 22). Of interest, previous studies from our laboratories indicated that selected DAP-resistant (DAPr) S. aureus strains isolated from clinical infections, as well as following in vitro passage in DAP, exhibited in vitro cross-resistance to several HDPs (13, 21). (Although the official terminology is “daptomycin nonsusceptible,” the term “daptomycin resistant” was employed in this study for a more facile presentation.) However, the overall frequency of the co-occurrence of such reduced in vitro susceptibilities to DAP and HDPs, especially among clinically derived MRSA strains, remains undefined.

The most prevalent and well-known genetic perturbations associated with DAPr in S. aureus are single point mutations in various regions of the mprF open reading frame (ORF), with or without concomitant point mutations in the yyc operon (12, 14, 27, 42). The mprF locus is principally involved in the maintenance of a relative positive surface charge in S. aureus via lysinylation of cell membrane phosphatidylglycerol (PG) (10). Phenotypic mechanisms which have been linked to DAPr in S. aureus include increases in net positive surface charge ostensibly imparting a charge-repulsive milieu against cationic peptides (related to gain-in-function mutations in mprF or dlt [39, 42]), thickened cell walls (5), and/or altered cell membrane order (2, 13, 21, 22). Of interest, most of the same phenotypic perturbations have also been linked to reduced in vitro susceptibilities to cationic HDPs (13, 41). The purposes of the current investigation were to (i) determine the frequency and extent of in vitro concurrence of reduced susceptibility to DAP and selected HDPs in MRSA clinical isolates; (ii) compare the antistaphylococcal efficacies of HDPs that are involved in endovascular defense but that differ in structure, charge, mechanism(s), and origin; (iii) correlate potential cross-resistance with cell surface charge, cell membrane fluidity, and cell wall thickness; and (iv) define the relationship of such concurrently reduced in vitro susceptibilities to the presence of mprF mutations.

(This work was presented in part at the 50th Interscience Conference on Antimicrobial Agents and Chemotherapy [abstr. C1-1459], Boston, MA, 12 to 15 September 2010.)

MATERIALS AND METHODS

Bacterial strains.

The 10 DAPs/DAPr MRSA study pairs employed in this investigation were clinical bloodstream isolates selected from the Cubist Pharmaceuticals isolate collection by one of the investigators (A.R.), including an initial pre-DAP therapy strain and a matched strain which developed in vitro DAPr during DAP treatment. The selection of this strain set was prioritized to encompass DAPr strains with and without mprF and/or yyc operon mutations. Isolates studied were identical on pulse-field gel electrophoresis (PFGE). To confirm the putative isogenicity of each strain pair, these isolates were evaluated by the following genotypic assays: agr typing (20), spa typing and clonal complexing (30), and staphylococcal cassette chromosome mec (SCCmec) typing (6). These studies confirmed the genetic relatedness of each of the 10 strain pairs (Table 1). The in vitro growth rates of all strain pairs were virtually identical over a 24-h period (data not shown).

Table 1.

Genotypes of study strain pairsa

Strain spa type spa motif Clonal complex SCCmec type agr type
CB1483 2 TJMBMDMGMK 5 II II
CB185 2 TJMBMDMGMK 5 II II
CB5079 1 YHGFMBQBLO 8 IV I
CB5080 YHGFMBQBLO 8 IV I
CB5083 1 TMDMGMK 5 II II
CB5082 1 TMDMGMK 5 II II
CB5088 47 YHGFMBQBLO 8 II I
CB5089 47 YHGFMBQBLO 8 II I
CB1631 1 TJMBMDMGMK 5 II II
CB1634 1 TJMBMDMGMK 5 II II
CB1663 2 TJMBMDMGMK 5 II II
CB1664 2 TJMBMDMGMK 5 II II
CB5057 1 YHGFMBQBLO 8 IV I
CB5059 1 YHGFMBQBLO 8 IV I
CB5062 16 WGKAKAOMQQQ 30 II III
CB5063 16 WGKAKAOMQQQ 30 II III
CB5015 2 TJMBMDMGMK 5 II II
CB5016 2 TJMBMDMGMK 5 II II
CB5021 2 TJMBMDMGMK 5 II II
CB5020 2 TJMBMDMGMK 5 II II
a

Pairs of isolates are represented by alternative shading and no shading, with the first strain in each pair being the DAPs parental strain and the second one in each pair being the DAPr strain.

The MICs of the strain set to DAP, vancomycin, oxacillin, and gentamicin were determined by standard Etest (AB Biodisc, Dalvagen, Sweden) on Mueller-Hinton agar (MHA) plates (Difco Laboratories, Detroit, MI). To assess the DAP Etest, plates were calcium supplemented according to the manufacturer's recommendations (50 μg/ml CaCl2). DAP MICs are shown in Table 2; the breakpoint distinguishing the DAPs and DAPr phenotypes was considered ≥2 μg/ml. Vancomycin MICs were 1 μg/ml for 8/10 parental strains and 2 and 4 μg/ml for one strain each. Vancomycin MICs commonly increased 2- to 4-fold among DAPr isolates (9/10 isolates), with 4 isolates exhibiting a vancomycin-intermediate susceptible S. aureus (VISA) phenotype (MIC = 4 μg/ml). For gentamicin, 9/10 DAPs/DAPr pairs exhibited MICs of 2 μg/ml (gentamicin susceptible); one parental strain was intrinsically gentamicin resistant. As expected, oxacillin MICs were >8 μg/ml for all DAPs parental strains, with no change in oxacillin MICs observed in 8/10 DAPr strains. Of interest, a significant reduction of oxacillin MIC was seen in one DAPr strain (the so-called seesaw effect [31, 32, 40]). Seven of the 10 DAPr strains exhibited single nucleotide polymorphisms (SNPs) within the mprF gene locus, with or without concomitant SNPs within the yyc operon in yycG. DAPr strains isolated either clinically or following serial in vitro passage in DAP have often been shown to temporally accumulate mutations in these loci (12). Table 2 lists the specific SNPs identified in these 7 DAPr strains. It should be noted that SNPs in mprF occurred within both its putative lysyl-PG (L-PG) synthase and translocase domains (10) and represented the most common SNP hot spots for mutations in DAPr strains, as previously reported (4, 12, 41).

Table 2.

MICs, PFGE types, and mprF/yycG SNPs among 10 study strain pairsa

Strain MIC (μg/ml)b
USA group SNP
DAP VAN OX GM mprF yycG
CB1483 0.25 1 >8 2 USA100
CB185 4 2 >8 2 L826Fc None
CB5079 0.5 1 >8 2 USA300
CB5080 2 2 >8 2 L826Fc None
CB5083 0.25 1 >8 2 USA100
CB5082 4 2 >8 2 L341Sd None
CB5088 0.5 1 8 2 USA300
CB5089 2–4 2 >8 2 S295Ld None
CB1631 0.5 2 >8 2 USA100
CB1634 4 4 >8 2 L826Fc Frameshift
CB1663 0.5 1 >8 >256 ND
CB1664 4 4 4 >256 L826Fc R86H
CB5057 0.5 1 >8 2 USA300
CB5059 4 4 0.5 2 I420Nc T474I
CB5062 0.5 1 >8 2 NDe
CB5063 8 2 >8 2 None None
CB5015 1 4 >8 2 ND
CB5016 4 4 >8 2 None None
CB5021 0.25 1 >8 2 ND
CB5020 1 4 >8 2 None None
a

Pairs of isolates are represented by alternative shading and no shading, with the first strain in each pair being the DAPs parental strain and the second one in each pair being the DAPr strain.

b

VAN, vancomycin; OX, oxacillin; GM, gentamicin.

c

Mutation in putative mprF synthase domain.

d

Mutation in putative mprF translocase domain.

e

ND, not determined.

HDP and CAP susceptibilities.

HDP and cationic antimicrobial peptide (CAP) bactericidal assays were performed in minimal liquid nutrient medium (Eagle's minimal essential medium [MEM]) in appropriate buffers (37, 43) by a 2-h timed-kill method as previously detailed (22). The following peptides were studied: (i) two prototypical mammal-derived HDPs, thrombin-induced platelet microbicidal proteins (tPMPs) from rabbits and human neutrophil-derived defensin-1 (hNP-1) (both of these HDPs have previously been shown to play a role in innate host defenses against endovascular infections [1, 9, 18, 38, 45]), and (ii) a bacterium-derived cyclic CAP, polymyxin B (PMB). The concentrations of peptides used in the killing assays were 0.25 to 0.50 μg/ml for tPMPs, 5 to 10 μg/ml for hNP-1, and 20 to 40 μg/ml for PMB. These sublethal concentrations were selected on the basis of (i) their ability to reduce survival of the parental DAPs strains by >50% in preliminary studies and (ii) peptide concentrations used in prior investigations of HDP-S. aureus interactions (23, 37). The hNP-1 was purchased from Peptide International (Louisville, KY); PMB was purchased from Sigma-Aldrich (St. Louis, MO). DAP was obtained from Cubist Pharmaceuticals (Lexington, MA). The tPMP was prepared from thrombin-stimulated rabbit platelets as previously described (44). The bioactive tPMP concentration equivalency was determined as detailed elsewhere (22). Stationary-phase cells (overnight cultures) were utilized in all assays.

All HDPs described above were reconstituted in appropriate diluents as described elsewhere (37, 44). S. aureus cells were diluted into the peptide solutions to achieve the desired final inoculum (103 CFU/ml) (38, 39) and peptide concentrations and were then incubated at 37°C. After 2 h exposure, samples were obtained and processed for quantitative culture to evaluate the extent of killing by each CAP. Final data were expressed as mean percent surviving CFU/ml ± standard deviation (SD). Since there is no bona fide resistance breakpoint for HDPs, we compared only the mean percent survivability ± SD in the DAPs versus DAPr groups for statistical assessments. A minimum of three experimental runs were performed on separate days.

Cell membrane fluidity.

Relative cell membrane order was determined by polarization spectrofluorometry as described previously by Mishra et al. (22) using the fluorescent probe 1,6-diphenyl-1,3,5-hexatriene (DPH). An inverse relationship exists between polarization indices and the degree of cell membrane order (i.e., a lower polarization index [PI] value equates to a greater cell membrane fluidity). These assays were performed a minimum of three times for each strain set on separate days.

Cell wall thickness.

All strain pairs were prepared for assessment of cell wall thickness profiles by transmission electron microscopy as described previously (41). The mean thickness ± SD of 100 cells was determined for our strain set at a magnification of ×190,000 (model 100CX; Jeol, Tokyo, Japan) using digital image capture and morphometric measurement (version 54; Advanced Microscopy Techniques, Danvers, MA). Cells were prepared for microscopy by previously published techniques (17). The electron microscopy measurements were performed by one of the authors (C.C.N.), who was blinded to the identity of the strains as DAPs or DAPr.

Surface charge.

To measure relative net surface charge, a fluorescein isothiocyanate (FITC)-labeled poly-l-lysine (PLL) binding assay was performed using flow cytometry (FACS Calibur apparatus; Beckman Instruments, Alameda, CA) as described previously (21, 23). Data were expressed as mean fluorescent units ± SDs. The lower that the residual cell-associated label was, the more positively charged that the S. aureus cell envelope is (21, 23). At least three independent runs were performed on separate days.

Statistical analysis.

Means and SDs were calculated for all variables. Differences between strains for killing and cell membrane/cell wall profile assays were analyzed with the Wilcoxon rank-sum test or two-tailed Student t test, as appropriate. For analysis of the relationships between CAP susceptibilities, DAP MIC, or cell membrane/cell wall profiles, comparisons for individual variables were performed using simple linear regression and multiple linear regression, and these techniques were then used to assess the joint relationship of the predictors with the outcome. All variables with P values of <0.2 were included in a stepwise regression analysis, with the criterion for remaining in the model being significance at α equal to 0.05. P values of ≤0.05 were considered significant.

RESULTS

HDP and CAP susceptibilities.

In general, among the individual isogenic pairs, DAPr strains exhibited higher survival profiles than their respective parental DAPs strains when they were exposed in vitro to the cadre of test CAPs (Table 3). For example, 7/10 DAPr strains were significantly more resistant to both peptide exposure concentrations of tPMPs and hNP-1 than their respective DAPs parental strain. In five of these seven comparisons, reduced killing profiles for these two HDPs tracked together for individual strain pairs. For PMB, the concomitant cross-resistance phenomenon was less common and did not consistently track with the two HDPs.

Table 3.

In vitro susceptibilities to three distinct CAPs, cell membrane fluidity, and cell wall thickness of 10 study strain pairsa

Strain % survival (mean ± SD) after 2 h of exposure to:
Cell membrane fluidity (PI value) Cell wall thickness (nm)
tPMPs
hNP-1
PMB
0.5 μg/ml 0.25 μg/ml 10 μg/ml 5 μg/ml 40 μg/ml 20 μg/ml
CB1483 18 ± 12 32 ± 18 45 ± 11 59 ± 11 7 ± 8 31 ± 18 0.333 ± 0.012 35.9 ± 3.8
CB185 67 ± 22* 95 ± 10* 56 ± 8 69 ± 14 9 ± 11 38 ± 16 0.331 ± 0.004 38.8 ± 3.4**
CB5079 3 ± 4 22 ± 8 3 ± 6 21 ± 7 0 ± 0 40 ± 18 0.453 ± 0.042 30.7 ± 3.2
CB5080 48 ± 7* 74 ± 11* 40 ± 5* 60 ± 13* 37 ± 7* 65 ± 19 0.369 ± 0.047 39.3 ± 4.2***
CB5083 14 ± 7 32 ± 15 0 ± 0 0 ± 0 0 ± 0 33 ± 18 0.394 ± 0.064 30.6 ± 3.1
CB5082 78 ± 10* 83 ± 12* 33 ± 13* 48 ± 8* 52 ± 7* 77 ± 14* 0.325 ± 0.046 37.0 ± 4.1***
CB5088 3 ± 2 17 ± 8 9 ± 9 20 ± 13 5 ± 8 52 ± 10 0.388 ± 0.033 33.7 ± 3.3
CB5089 41 ± 3* 76 ± 17* 19 ± 9 40 ± 9 26 ± 7* 59 ± 3 0.343 ± 0.008 34.6 ± 2.6*
CB1631 22 ± 12 31 ± 24 15 ± 14 30 ± 24 6 ± 7 46 ± 25 0.375 ± 0.021 39.7 ± 3.5
CB1634 87 ± 8* 100 ± 12* 72 ± 16* 79 ± 10* 12 ± 9 56 ± 28 0.280 ± 0.045* 39.8 ± 3.5
CB1663 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 20 ± 5 0.422 ± 0.081 29.4 ± 3.0
CB1664 84 ± 36* 82 ± 39* 67 ± 17* 64 ± 14* 57 ± 21* 69 ± 5* 0.334 ± 0.075 44.7 ± 5.3*
CB5057 80 ± 10 80 ± 6 24 ± 6 34 ± 5 40 ± 24 85 ± 1 0.321 ± 0.022 34.2 ± 4.4
CB5059 94 ± 13 95 ± 19 70 ± 21* 76 ± 20* 69 ± 20 83 ± 13 0.265 ± 0.054 36.0 ± 3.6*
CB5062 69 ± 27 74 ± 35 23 ± 9 44 ± 8 0 ± 0 7 ± 12 0.332 ± 0.098 32.3 ± 3.0
CB5063 85 ± 2 96 ± 7 64 ± 33 69 ± 28 20 ± 21 56 ± 23* 0.331 ± 0.014 36.2 ± 4.0***
CB5015 80 ± 36 82 ± 28 65 ± 7 66 ± 3 44 ± 12 58 ± 1 0.375 ± 0.051 41.4 ± 6.7
CB5016 85 ± 32 92 ± 19 87 ± 8* 82 ± 3* 50 ± 10 48 ± 8 0.342 ± 0.035 43.1 ± 6.1b
CB5021 24 ± 11 57 ± 14 16 ± 12 28 ± 24 22 ± 13 66 ± 13 0.295 ± 0.020 30.4 ± 3.8
CB5020 101 ± 16* 111 ± 5* 83 ± 5* 73 ± 6* 82 ± 30* 95 ± 41 0.083 ± 0.04* 47.7 ± 7.5***
a

Pairs of isolates are represented by alternative shading and no shading, with the first strain in each pair being the DAPs parental strain and the second one in each pair being the DAPr strain. *, P < 0.05 versus parental strain; **, P = 0.05 versus parental strain; ***, P < 0.0001 versus parental strain.

b

P = 0.057 versus parental strain value.

When they were analyzed as collective groups of DAPs and DAPr strains, DAPr strains demonstrated significant reductions in killing by all three CAPs assessed at all peptide exposure concentrations tested; this was especially striking for sublethal concentrations of tPMPs and hNP-1 in vitro (Table 4).

Table 4.

Group comparison of CAP susceptibility, cell membrane fluidity, and cell wall thickness of all DAPs and DAPr strainsa

Parameter DAPs strains (n = 10) DAPr strains (n = 10) P value
% survival after 2 h of exposure to CAP (concnb)
    tPMP (0.50) 31 ± 32 77 ± 19 0.0012
    tPMP (0.25) 43 ± 29 90 ± 11 <0.001
    hNP-1 (10) 20 ± 21 59 ± 22 <0.001
    hNP-1 (5) 30 ± 22 66 ± 13 <0.001
    PMB (40) 12 ± 17 41 ± 24 0.0067
    PMB (20) 44 ± 23 65 ± 16 0.0328
Cell membrane fluidity (PI value) 0.369 ± 0.05 0.300 ± 0.08 0.0358
Cell wall thickness (nm) 33.84 ± 4.1 39.70 ± 4.10 0.0051
a

Values are means ± SDs.

b

Concentrations are in micrograms per milliliter.

We next examined the potential correlation of HDP-resistant or CAP-resistant profiles with acquisition of an mprF mutation(s), with or without concomitant yyc operon mutations, in DAPr strains (Table 5). Collectively, acquisition of mprF mutations in DAPr strains was associated with significantly reduced killing by both HDPs (tPMPs and hNP-1). A similar but less robust statistical trend was noted for PMB. To further dissect the specific impact of mprF mutations alone (n = 4 strains) or in combination with yycG mutations (n = 3 strains), we analyzed CAP susceptibility phenotypes in relation to one or more mutations. As seen in Table 6, there was a notable trend of increasing CAP-resistant phenotypes with accumulation of both the mprF and yycG mutations combined compared to mprF mutations alone. However, the small sample sizes precluded definitive statistical evaluation. It should be underscored that the three DAPr strains lacking SNPs in either locus still demonstrated substantially reduced killing by all study peptides compared to their parental DAPs strains.

Table 5.

Comparative analysis of CAP susceptibility, cell membrane fluidity, and cell wall thickness of the DAPs parental strains and the 7 DAPr strains with mutations in mprF (with or without yycG mutations)a

Parameter DAPs strains (n = 10) DAPr strains (n = 7) P value
% survival after 2 h of exposure to CAP (concnb)
    tPMP (0.50) 31 ± 32 71 ± 20 0.0112
    tPMP (0.25) 43 ± 29 86 ± 10 0.016
    hNP-1 (10) 20 ± 21 51 ± 21 0.0085
    hNP-1 (5) 30 ± 22 62 ± 14 0.0041
    PMB (40) 12 ± 17 37 ± 23 0.0207
    PMB (20) 44 ± 23 64 ± 15 0.0599
Cell membrane fluidity (PI value) 0.369 ± 0.05 0.321 ± 0.04 0.0441
Cell wall thickness (nm) 33.84 ± 4.1 38.60 ± 3.3 0.0218
a

Values are means ± SDs.

b

Concentrations are in micrograms per milliliter.

Table 6.

Comparative analysis of CAP susceptibility, cell membrane fluidity, cell wall thickness, and MIC profiles of all DAPs strains and DAPr strains with or without specific point mutation profilesa

Parameter DAPs (n = 10) DAPr mutations in mprF alone (n = 4) DAPr mutations in mprF plus yycG (n = 3) DAPr neither mutation (n = 3)
% survival after 2 h of exposure to CAP (concnb)
    tPMP (0.50) 31 ± 32 59 ± 17 88 ± 5 90 ± 9
    tPMP (0.25) 43 ± 29 82 ± 9 92 ± 9 100 ± 10
    hNP-1 (10) 20 ± 21 37 ± 15 70 ± 3 78 ± 12
    hNP-1 (5) 30 ± 22 54 ± 13 73 ± 8 75 ± 7
    PMB (40) 12 ± 17 31 ± 68 46 ± 30 51 ± 31
    PMB (20) 44 ± 23 60 ± 16 69 ± 13 66 ± 25
Cell membrane fluidity (PI value) 0.369 ± 0.05 0.343 ± 0.19 0.293 ± 0.04 0.252 ± 0.15
Cell wall thickness (nm) 33.84 ± 4.1 37.43 ± 2.1 40.16 ± 4.4 42.13 ± 5.5
MIC (μg/ml) 0.475 ± 0.22 3.25 ± 0.96 4.00 ± 0.00 4.33 ± 3.5
a

Values are means ± SDs. See text for comparative assessments of these data sets.

b

Concentrations are in micrograms per milliliter.

Cell membrane fluidity.

When they were compared individually, each DAPr strain exhibited a clear trend toward more fluid cell membranes than the respective DAPs parental strain (Table 3). This comparison reached statistical significance for two individual strain pair comparisons. When they were compared as collective DAPs versus DAPr isolate groups, the cell membranes of the DAPr strains were significantly more fluid than those of the isolates in the DAPs group (Table 4).

Cell wall thickness.

All DAPr strains had significantly thicker cell walls than their respective DAPs parental strains (Table 3). Similarly, when they were analyzed as collective groups of DAPs and DAPr strains, DAPr strains exhibited significantly thicker cell walls than the DAPs isolates (Table 4). This same relationship held when strains with mprF mutations were compared to the DAPs strains (Table 5).

To refine the phenotypic associations between cell wall/cell membrane profiles with CAP-DAP susceptibility profiles, we performed simple linear regression analysis comparing cell wall thickness and cell membrane fluidity with the outcome variables of CAP and DAP efficacies (reductions in numbers of CFU/ml versus MICs, respectively). Cell wall thickness was directly and significantly associated with reduced killing by hNP-1 (P < 0.001), tPMPs (P < 0.05), and PMB (P < 0.01) among the DAPr strains. Cell wall thickness was related to higher DAP MICs, although this did not reach statistical significance (P = 0.12). Increased cell membrane fluidity correlated well with reduced killing of DAPr strains by all peptides (hNP-1, P < 0.01; tPMPs, P < 0.01; and PMB, P < 0.01). Similarly, enhanced cell membrane fluidity tracked somewhat with higher DAP MICs in the DAPr strains, although not significantly (P = 0.4). Although multiple linear regression analyses with both predictor variables of cell wall thickness and cell membrane fluidity were attempted, they proved to not be feasible due to limited sample sizes.

Genotypic and phenotypic associations.

To further explore phenotypic-genotypic correlates, DAPr strains were subcategorized as to the presence of mutations in mprF alone, mutations in mprF plus yycG, or no mutations in either locus. Of interest, DAPr strains with mutations in mprF alone or in combination with yycG mutations exhibited obvious trends in thicker cell walls than DAPs strains and increased cell membrane fluidity compared to DAPs strains (Table 6). However, small sample sizes precluded adequate statistical comparisons.

Surface charge.

No consistent pattern of surface charge differences was observed between the DAPr and DARs strain pairs, either individually or as overall groups (data not shown).

DISCUSSION

There have been a number of recent reports concerning development of DAPr among clinical strains of S. aureus during the therapy of invasive infections with this agent (13, 33). Recent studies of a limited number of DAPs/DAPr MRSA and methicillin-susceptible S. aureus (MSSA) strain pairs from our laboratories have shown a trend toward coevolution of relative resistance to several HDPs and DAPr (13, 22, 39). In the present investigation, we used a well-characterized set of DAPs and DAPr MRSA strains to examine (i) the frequency and extent of this phenomenon and (ii) potential genotypic and phenotypic associations that may serve as biomarkers of such a co-occurrence in reduced in vitro susceptibilities (relative cross-resistance) of these agents.

A number of interesting findings emerged from this study. First, we employed two HDPs which have been well chronicled to defend against endovascular infections: tPMPs from platelets and hNP-1 from neutrophils (44, 46). DAPr MRSA strains in this study demonstrated a clear trend of reduced in vitro susceptibility to these HDPs, whether they were assessed in individual (head-to-head) or group strain comparisons. In addition, this in vitro cross-resistance phenotype extended to PMB, a cyclic bacterium-derived CAP with no overt structural or mechanistic similarities to either DAP or the two HDPs tested (22). In contrast, no such cross-resistance was found for the cationic ribosome-targeting molecule gentamicin. Since the principle mechanistic feature shared by DAP and the three peptides above is cell membrane targeting (22), this suggested that a general adaptive paradigm for such cross-resistance was operative. This hypothesis prompted our comparison of several prototypical cell membrane and cell surface parameters between the DAPs and DAPr strain pairs. Of interest, 9/10 DAPr isolates exhibited 2- to 4-fold increases in MICs to vancomycin, a minimally cationic cell wall-targeting antibiotic. Previous studies have documented a temporal linkage between vancomycin usage, increased vancomycin MICs, and subsequent resistance in vitro to DAP (8, 14, 24).

Second, recently published studies by our group and others (10, 13, 21, 22, 41) have suggested an important role in DAPr for at least two genes involved in maintenance of staphylococcal positive surface charge, mprF and dlt (13, 21, 39). For mprF, several investigations have identified a series of gain-in-function point mutations within its ORF. These mutations have been associated with either excess production or increased outer cell membrane translocation of the positively charged phospholipid species L-PG. The net result of these effects is believed to be enhancement of relative positive surface charge (10, 13). Our current data were somewhat in line with these prior observations, showing that DAPr strains commonly exhibited SNPs within the mprF ORF (in either its putative synthase or translocase domain) (10). Surprisingly, the presence of such SNPs in mprF among DAPr strains was not consistently associated with significant changes in the relative surface charge profiles compared with the profiles of their respective DAPs parental strains. This suggested several possible explanations: (i) these SNPs may not have altered the mprF gene expression profile in these strains. This phenomenon was recently seen in a DAPr MSSA strain in which a well-defined SNP was identified within the mprF ORF without enhancement of mprF gene expression (39). In that instance, an increased positive surface charge was identified in association with enhanced expression of the dlt operon. (ii) mprF expression was enhanced in our DAPr strains of interest, but compensatory adaptations in other genes involved in surface charge maintenance occurred (e.g., dlt or genes regulating cell wall amidation [7, 25, 26, 36]). (iii) mprF expression and L-PG synthesis/translocation were indeed enhanced, but DAPr in these strains was unrelated to a charge-mediated effect on peptide interactions. In this regard, recent investigations from our laboratory and others provide evidence of an increased association of certain CAPs with the cell membrane and potentially with PG-cardiolipin-enriched regions. In this case, we hypothesize that any effect of charge repulsion would be a secondary mechanism of DAPr (16, 19). Gene expression and phospholipid compositional profiling are currently in progress to examine these possible scenarios. It should be underscored that although acquisition of mprF SNPs (with or without concomitant yycG SNPs) was frequently found in our DAPr strains, isolates without either mutation demonstrated similar frequencies and extents of DAP-CAP cross-resistance. Thus, other genotypic mechanisms of DAPr are likely at play for such strains.

As pointed out above, SNPs within the yyc operon have previously been noted in both in vitro-derived (by serial DAP passage) and clinically derived DAPr strains of S. aureus (13, 21). Among in vitro-generated DAPr strains, accumulation of yyc operon SNPs appears to temporally follow mutations in the mprF operon (12). The precise mechanism(s) by which SNPs within the yyc operon (e.g., yycG, as in three of our strains) cause the DAPr phenotype is not known. This multifunctional S. aureus regulatory operon has been described to be a factor potentially important in (i) virulence (through impacts on ssaA and lytM expression), (ii) influence upon cell wall biosynthesis through regulation of the tag operon expression (involved in cell wall teichoic acid synthesis), (iii) cell membrane fatty acid homeostasis, and (iv) biofilm formation (11).

Third, in addition to the potential issue of surface charge impacts on DAP-CAP cross-resistance, we investigated two other phenotypic characteristics of the DAPs/DAPr strain pairs that might influence peptide-S. aureus interactions: (i) cell membrane order and (ii) cell wall thickness. Our laboratories have reported a number of instances in which S. aureus strains with highly disordered cell membranes demonstrated significantly reduced abilities to be killed in vitro by prototypical HDPs, including tPMPs and hNP-1 (2, 13, 21, 37). The mechanism(s) by which increased cell membrane fluidity leads to reduced HDP-induced killing of S. aureus is not clear but may include perturbations of the HDP-cell membrane association, a reduced capacity for HDP insertion into the target cell membrane, and/or CAP partitioning within such disordered cell membranes (13, 37). Similarly, S. aureus strains which have excessively ordered (rigid) cell membranes by virtue of robust pigment production also show DAP-CAP cross-resistance in vitro (22). In the present study, there was a notable trend among all 10 DAPr strains to have substantially more fluid cell membranes than their respective DAPs parental strains. Studies are in progress to determine whether enhanced cell membrane fluidity is causal in DAPr or whether it is a secondary consequence and surrogate biomarker of this phenotype.

Lastly, the cell walls of these DAPr strains were substantially thicker by electron microscopy than those of their respective DAPs parental isolates. The association of thickened cell walls with DAPr has been previously documented as a common, but not universal, accompaniment of this phenotype in S. aureus, similar to VISA strains (5, 8, 21, 41). It has been postulated that, as with VISA strains, thickened cell walls may represent a physical barrier or an affinity trap to DAP and other HDPs or CAPs, preventing their accessibility to their principle cell membrane target(s) (8, 21). As noted before, 9/10 DAPr strains studied showed 2- to 4-fold increases in vancomycin MICs correlating with such thickened cell walls. The metabolic pathways leading to the thickened cell wall phenotype and associated DAP-CAP relative cross-resistance in S. aureus are likely to be multifactorial and complex (21). Recent data from our laboratories implicate excess synthesis of cell wall teichoic acids as an important contributor to the thickened cell wall and DAPr phenotypes in selected strains (3).

It should be emphasized that the current investigation had several important limitations: (i) the small sample size of DAPr strains with and without mutations within the mprF locus precluded adequate statistical analysis of the impacts of this operon on DAP-CAP cross-resistance phenotypes; (ii) only a limited breadth of cell membrane and cell wall profiling was queried in comparing the strain pairs; (iii) a relatively narrow range of host defense CAPs was investigated, leaving open the question of how specific or nonspecific the DAP-CAP cross-resistance phenotype really is; and (iv) CAPs were assessed for their inhibitory activities against study strains individually, at low-inoculum challenges, using peptide concentrations that are likely well below their physiological concentrations and in assays conducted within austere artificial media in vitro. Such conditions are unlikely to effectively represent those under which S. aureus strains encounter DAP and HDPs within the endovascular compartment in vivo. These limitations are being addressed in current investigations in our laboratories. Finally, the precise sequence of events by which in vitro DAPr coevolves with CAPr in S. aureus is not clear. Thus, do organisms first develop DAPr upon prolonged exposure to this agent in vivo, with the co-occurrence of relative resistance to HDPs being a secondary phenomenon, or are bloodstream organisms first selected in vivo for reduced susceptibility to HDPs by exposure to endovascular host defenses (i.e., those from platelets and neutrophils), setting the stage for DAPr? These and other potential paradigms, while not mutually exclusive, are also under active investigation in our laboratories.

ACKNOWLEDGMENTS

This research was supported by grants AI-39108 (to A.S.B.) and grants AI-39001 and AI-48031 (to M.R.Y.) from the National Institutes of Health and a grant from Cubist Pharmaceuticals, Lexington, MA (to A.S.B.).

Footnotes

Published ahead of print on 27 June 2011.

REFERENCES

  • 1. Bayer A. S., et al. 1997. Hyperproduction of alpha-toxin by Staphylococcus aureus results in paradoxically reduced virulence in experimental endocarditis: a host defense role for platelet microbicidal proteins. Infect. Immun. 65:4652–4660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Bayer A. S., et al. 2000. In vitro resistance of Staphylococcus aureus to thrombin-induced microbicidal protein is associated with alterations in membrane fluidity. Infect. Immun. 68:3548–3553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Bertsche U., et al. 23 May 2011. Correlation of daptomycin-resistance in a clinical Staphylococcus aureus strain with increased cell wall teichoic acid production and d-alanylation. Antimicrob. Agents Chemother. [Epub ahead of print.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Boyle-Vavra S., et al. 2011. Comparative genome sequencing of an isogenic pair of USA800 clinical methicillin-resistant Staphylococcus aureus isolates obtained before and after daptomycin treatment failure. Antimicrob. Agents Chemother. 55:2018–2025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Camargo I. L., Neoh H.-M., Cui L., Hiramatsu K. 2008. Serial daptomycin selection generates daptomycin-nonsusceptible Staphylococcus aureus strains with a heterogeneous vancomycin-intermediate phenotype. Antimicrob. Agents Chemother. 52:4289–4299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Chen L., et al. 2009. Multiplex real-time PCR for rapid staphylococcal cassette chromosome mec typing. J. Clin. Microbiol. 47:3692–3706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Collins L. V., et al. 2002. Staphylococcus aureus strains lacking d-alanine modifications of teichoic acids are highly susceptible to human neutrophil killing and are virulence attenuated in mice. J. Infect. Dis. 186:214–219 [DOI] [PubMed] [Google Scholar]
  • 8. Cui L., Tominaga E., Neoh H. M., Hiramatsu K. 2006. Correlation between reduced daptomycin susceptibility and vancomycin resistance in vancomycin-intermediate Staphylococcus aureus. Antimicrob. Agents Chemother. 50:1079–1082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Dhawan V. K., Bayer A. S., Yeaman M. R. 1998. In vitro resistance to thrombin-induced platelet microbicidal protein is associated with enhanced progression and hematogenous dissemination in experimental Staphylococcus aureus infective endocarditis. Infect. Immun. 66:3476–3479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ernst C. M., et al. 2009. The bacterial defensin resistance protein MprF consists of separable domains for lipid lysinylation and antimicrobial peptide repulsion. PLoS Pathog. 5:e1000660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Fischer A., et al. Daptomycin resistance mechanisms in clinically derived Staphylococcus aureus strains assessed by a combined transcriptomics and proteomic approach. J. Antimicrob. Chemother., in press [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Friedman L., Adler J. D., Silverman J. A. 2006. Genetic changes that correlate with reduced susceptibility to daptomycin in Staphylococcus aureus. Antimicrob. Agents Chemother. 50:2137–2145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Jones T., et al. 2008. Failures in clinical treatment of Staphylococcus aureus infection with daptomycin are associated with alterations in surface charge, membrane phospholipid asymmetry, and drug binding. Antimicrob. Agents Chemother. 52:269–278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Julian K., et al. 2007. Characterization of a daptomycin-nonsusceptible vancomycin-intermediate Staphylococcus aureus strain in a patient with endocarditis. Antimicrob. Agents Chemother. 51:3445–3448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kaatz G. W., Lundstrom T. S., Seo S. M. 2006. Mechanisms of daptomycin resistance in Staphylococcus aureus. Int. J. Antimicrob. Agents 28:280–287 [DOI] [PubMed] [Google Scholar]
  • 16. Kilelee E., Pokorny A., Yeaman M. R., Bayer A. S. 2010. Lysyl-phosphatidylglycerol attenuates membrane perturbation rather than surface association of the cationic antimicrobial peptide 6W-RP-1 in a model membrane system: implications for daptomycin resistance. Antimicrob. Agents Chemother. 54:4476–4479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Koo S.-P., Yeaman M. R., Nast C. C., Bayer A. S. 1997. The cytoplasmic membrane is a primary target for the staphylocidal action of thrombin-induced platelet microbicidal protein. Infect. Immun. 65:4795–4800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Kupferwasser L. I., Yeaman M. R., Shapiro S. M., Nast C. C., Bayer A. S. 2002. In vitro susceptibility to thrombin-induced platelet microbicidal protein is associated with reduced disease progression and complication rates in experimental Staphylococcus aureus endocarditis: microbiological, histopathologic, and echocardiographic analyses. Circulation 105:746–752 [DOI] [PubMed] [Google Scholar]
  • 19. Li M., et al. 2009. Staphylococcus aureus mutant screen reveals interaction of the human antimicrobial peptide dermcidin with membrane phospholipids. Antimicrob. Agents Chemother. 53:4200–4210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Lina G., et al. 2003. Bacterial competition for human nasal cavity colonization: role of staphylococcal agr alleles. Appl. Environ. Microbiol. 69:18–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mishra N. N., et al. 2009. Analysis of cell membrane characteristics of in vitro-selected daptomycin-resistant strains of methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 53:2312–2318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Mishra N. N., et al. 2011. Carotenoid related alteration of cell membrane fluidity impacts Staphylococcus aureus susceptibility to host defense peptides. Antimicrob. Agents Chemother. 55:526–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Mukhopadhyay K., et al. 2007. Reduced in vitro susceptibility of Staphylococcus aureus to thrombin-induced platelet microbicidal protein-1 (tPMP-1) is associated with alterations in cell membrane phospholipid composition and asymmetry. Microbiology 153:1187–1197 [DOI] [PubMed] [Google Scholar]
  • 24. Mwangi M. M., et al. 2007. Tracking the in vivo evolution of multidrug resistance in Staphylococcus aureus by whole-genome sequencing. Proc. Natl. Acad. Sci. U. S. A. 104:9451–9456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Peschel A., et al. 1999. Inactivation of the dlt operon in Staphylococcus aureus confers sensitivity to defensins, protegrins and other antimicrobial peptides. J. Biol. Chem. 274:8405–8410 [DOI] [PubMed] [Google Scholar]
  • 26. Peschel A., Vuong C., Otto M., Götz F. 2000. The d-alanine residues of Staphylococcus aureus teichoic acids alter the susceptibility to vancomycin and the activity of autolysins. Antimicrob. Agents Chemother. 44:2845–2847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Pillai S. K., et al. 2007. Daptomycin nonsusceptibility in Staphylococcus aureus with reduced vancomycin susceptibility is independent of alterations in MprF. Antimicrob. Agents Chemother. 51:2223–2225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Sakoulas G., Eliopoulos G. M., Alder J., Eliopoulos C. T. 2003. Efficacy of daptomycin in experimental endocarditis due to methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 47:1714–1718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Schriever C. A., Fernandez C., Rodvold K. A., Danziger L. H. 2005. Daptomycin: a novel cyclic lipopeptide antimicrobial. Am. J. Health Syst. Pharm. 62:1145–1158 [DOI] [PubMed] [Google Scholar]
  • 30. Shopsin B., et al. 1999. Evaluation of protein A gene polymorphic region DNA sequencing for typing of Staphylococcus aureus strains. J. Clin. Microbiol. 37:3556–3563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Sieradzki K., Leski T., Dick J., Borio L., Tomasz A. 2003. Evolution of a vancomycin-intermediate Staphylococcus aureus strain in vivo: multiple changes in the antibiotic resistance phenotypes of a single lineage of methicillin-resistant S. aureus under the impact of antibiotics administered for chemotherapy. J. Clin. Microbiol. 41:1687–1693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Sieradzki K., Tomasz A. 1997. Inhibition of cell wall turnover and autolysis by vancomycin in a highly vancomycin-resistant mutant of Staphylococcus aureus. J. Bacteriol. 179:2557–2566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Skiest D. J. 2006. Treatment failure resulting from resistance of Staphylococcus aureus to daptomycin. J. Clin. Microbiol. 44:655–656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Steenbergen J. N., Alder J., Thorne G. M., Tally F. P. 2005. Daptomycin: a lipopeptide antibiotic for the treatment of serious Gram-positive infections. J. Antimicrob. Chemother. 55:283–288 [DOI] [PubMed] [Google Scholar]
  • 35. Trier D. A., et al. 2008. Platelet antistaphylococcal responses occur through P2X1 and P2Y12 receptor-induced activation and kinocidin release. Infect. Immun. 76:5706–5713 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Weidenmaier C., et al. 2005. Lack of wall teichoic acids in Staphylococcus aureus leads to reduced interactions with endothelial cells and to attenuated virulence in a rabbit model of endocarditis. J. Infect. Dis. 191:1771–1777 [DOI] [PubMed] [Google Scholar]
  • 37. Xiong Y. Q., Mukhopadhyay K., Yeaman M. R., Adler-Moore J., Bayer A. S. 2005. Functional interrelationships between cell membrane and cell wall in antimicrobial peptide-mediated killing of Staphylococcus aureus. Antimicrob. Agents Chemother. 49:3114–3121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Xiong Y. Q., Yeaman M. R., Bayer A. S. 1999. In vitro antibacterial activities of platelet microbicidal protein and neutrophil defensin against Staphylococcus aureus are influenced by antibiotics differing in mechanism of action. Antimicrob. Agents Chemother. 43:1111–1117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Yang S. J., et al. 2009. Enhanced expression of dltABCD is associated with the development of daptomycin nonsusceptibility in a clinical endocarditis isolate of Staphylococcus aureus. J. Infect. Dis. 200:1916–1920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Yang S. J., et al. 2010. Daptomycin-oxacillin combinations in treatment of experimental endocarditis caused by daptomycin-nonsusceptible strains of methicillin-resistant Staphylococcus aureus with evolving oxacillin susceptibility (the “seesaw effect”). Antimicrob. Agents Chemother. 54:3161–3169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Yang S. J., et al. 2010. Cell wall thickening is not a universal accompaniment of the daptomycin nonsusceptibility phenotype in Staphylococcus aureus: evidence for multiple resistance mechanisms. Antimicrob. Agents Chemother. 54:3079–3085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Yang S. J., et al. 2009. Regulation of mprF in daptomycin-nonsusceptible Staphylococcus aureus strains. Antimicrob. Agents Chemother. 53:2636–2637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Yeaman M. R., Gank K. D., Bayer A. S., Brass E. P. 2002. Synthetic peptides that exert antimicrobial activities in whole blood and blood-derived matrices. Antimicrob. Agents Chemother. 46:3883–3891 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Yeaman M. R., Tang Y. Q., Shen A. J., Bayer A. S., Selsted M. E. 1997. Purification and in vitro activities of rabbit platelet microbicidal proteins. Infect. Immun. 65:1023–1031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Yeaman M. R. 2010. Bacterial-platelet interactions: virulence meets host defense. Future Microbiol. 3:471–506 [DOI] [PubMed] [Google Scholar]
  • 46. Yeaman M. R., Bayer A. S. 2000. Staphylococcus aureus, platelets, and the heart. Curr. Infect. Dis. Rep. 2:281–298 [DOI] [PubMed] [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES