Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2016 Jun 23;84(7):1957–1965. doi: 10.1128/IAI.00117-16

Infectious Dose Dictates the Host Response during Staphylococcus aureus Orthopedic-Implant Biofilm Infection

Debbie Vidlak 1, Tammy Kielian 1,
Editor: A Camilli2
PMCID: PMC4936354  PMID: 27091926

Abstract

Staphylococcus aureus is a leading cause of prosthetic joint infections (PJIs) that are typified by biofilm formation. Given the diversity of S. aureus strains and their propensity to cause community- or hospital-acquired infections, we investigated whether the immune response and biofilm growth during PJI were conserved among distinct S. aureus clinical isolates. Three S. aureus strains representing USA200 (UAMS-1), USA300 (LAC), and USA400 (MW2) lineages were equally effective at biofilm formation in a mouse model of PJI and elicited similar leukocyte infiltrates and cytokine/chemokine profiles. Another factor that may influence the course of PJI is infectious dose. In particular, higher bacterial inocula could accelerate biofilm formation and alter the immune response, making it difficult to discern underlying pathophysiological mechanisms. To address this issue, we compared the effects of two bacterial doses (103 or 105 CFU) on inflammatory responses in interleukin-12p40 (IL-12p40) knockout mice that were previously shown to have reduced myeloid-derived suppressor cell recruitment concomitant with bacterial clearance after low-dose challenge (103 CFU). Increasing the infectious dose of LAC to 105 CFU negated these differences in IL-12p40 knockout animals, demonstrating the importance of bacterial inoculum on infection outcome. Collectively, these observations highlight the importance of considering infectious dose when assessing immune responsiveness, whereas biofilm formation during PJI is conserved among clinical isolates commonly used in mouse S. aureus infection models.

INTRODUCTION

Staphylococcal species (Staphylococcus aureus and coagulase-negative staphylococcus) are a primary cause of prosthetic joint infections (PJIs) that are typified by biofilm formation (13). Many PJIs are thought to originate from bacterial colonization at the time of surgery, although clinical signs of infection may not appear until months to years following joint replacement (4, 5). The often protracted period between prosthetic joint replacement and infectious complications suggests that prostheses may also be colonized following systemic bacterial infection. Since S. aureus biofilms are adept at circumventing immune-mediated clearance (69) and are recalcitrant to antibiotic therapy, S. aureus PJIs have become increasingly difficult to treat (1013). Understanding the series of inflammatory events that occur during S. aureus PJI is a critical step toward identifying potential therapeutic targets.

An important factor to consider when investigating mechanisms pertinent to human infection is whether animal models accurately reflect clinical features of human disease. Given its frequency in clinical infections (13), S. aureus is a logical choice of pathogen to demonstrate clinical relevance with regard to PJIs. However, it has been well documented that S. aureus strains can behave differently in terms of clinical presentation and pathogenicity (1, 1418). The fact that ca. 30% of individuals are carriers for S. aureus and the recent emergence of community-acquired, methicillin-resistant S. aureus (CA-MRSA) clones as causative pathogens in hospital-associated infections (19, 20) highlight the importance of examining the immune response to different S. aureus strains and doses in the context of PJI. Therefore, we compared the immune responses to three distinct S. aureus clinical isolates from various epidemiological backgrounds during PJI. S. aureus LAC is a CA-MRSA USA300 strain that was isolated from a patient with a skin and soft tissue infection (16, 21). S. aureus MW2 is a CA-MRSA USA400 strain that was recovered from a pediatric patient with fatal septicemia and septic arthritis (19, 22, 23). S. aureus UAMS-1 is a community-acquired, methicillin-sensitive S. aureus (CA-MSSA) USA200 strain that was isolated from a patient with osteomyelitis (24). These strains were selected since they were originally responsible for human infections and are commonly used in mouse infection models and, as such, are well characterized. Since the purpose of this study was to demonstrate the influence of infectious dose on immune responses, we considered it important to focus on clinical isolates that are widely used in mouse models and that will best contribute to the current body of knowledge. Recent studies from our laboratory have focused on characterizing the immune response during S. aureus PJI using a mouse model of orthopedic-implant biofilm infection, which has provided a better understanding of the key leukocyte populations that contribute to chronic biofilm establishment. In particular, myeloid-derived suppressor cells (MDSCs) and anti-inflammatory polarized macrophages have been implicated in promoting S. aureus biofilm persistence (2527). In this report, we determined whether biofilm formation or the resultant immune response was strain-dependent using our orthopedic-implant biofilm infection model with LAC, MW2, and UAMS-1. Differences in bacterial burdens between the various strains were negligible. Additionally, the immune response, as measured by immune cell influx and inflammatory mediator production in the soft tissue surrounding the orthopedic implant, revealed little variability between the three strains tested, an important finding considering the diverse origin of the bacterial strains examined.

The range of diseases caused by S. aureus is represented by the diversity of animal model systems currently being studied (28, 29). In terms of S. aureus biofilm infection, some models utilize a large infectious inoculum or introduce implants that are precoated with bacteria (3036). A high-challenge dose, particularly in a confined space such as the joint/bone, would be predicted to elicit a vigorous proinflammatory cascade that could likely alter the course of the resultant immune response and bacterial survival. In contrast, human PJIs typically result from colonization with low numbers of bacteria, which may provide a survival advantage during acute infection by their inability to trigger a robust proinflammatory response. To examine the effect of infectious dose on the host immune response and biofilm formation, we compared disease progression following the administration of a standard low-dose S. aureus inoculum (103 CFU) that we and others have used to establish chronic biofilm infection (2527, 3739) versus a 2-log-higher dose (105 CFU). These studies utilized IL-12p40 knockout (KO) mice that were previously shown to have reduced myeloid-derived suppressor cell (MDSC) recruitment concomitant with improved bacterial clearance after low-dose challenge (103 CFU) (26) to investigate whether a higher dose (105 CFU) would negate this phenotype. Indeed, bacterial burdens and MDSC infiltrates were similar between IL-12p40 KO and wild-type (WT) mice after high-dose (105 CFU) LAC challenge, whereas both readouts were significantly reduced in IL-12p40 KO animals in response to 103 CFU of bacteria. Taken together, these observations highlight the importance of considering infectious dose when assessing immune responsiveness, whereas biofilm formation during PJI is conserved among clinical isolates commonly used in mouse S. aureus infection models.

MATERIALS AND METHODS

Mice.

For S. aureus strain comparison studies, male C57BL/6 mice (8 to 12 weeks old) were purchased from Charles River Laboratories (Frederick, MD). For S. aureus dose comparison studies, IL-12p40 KO mice (C57BL/6 background) were purchased from The Jackson Laboratory (Bar Harbor, ME) with breeding colonies maintained in an approved facility at the University of Nebraska Medical Center. Age- and sex-matched IL-12p40 KO and C57BL/6 WT mice were utilized at 8 to 12 weeks of age. These studies were conducted in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The animal use protocol was approved by the Institutional Animal Care and Use Committee of the University of Nebraska Medical Center (UNMC).

Mouse model of S. aureus prosthetic joint infection.

A mouse model of S. aureus PJI was utilized as previously described (2527, 39, 40). Briefly, mice were anesthetized with a ketamine/xylazine cocktail (Hospira, Lake Forest, IL, and Akorn, Decatur, IL; 100 and 5 mg/kg of body weight, respectively), and the surgical site was shaved and disinfected with povidone-iodine. A medial parapatellar arthrotomy with lateral displacement of the quadriceps-patella was performed to access the distal femur. Next, a burr hole was created in the femoral intercondylar notch extending into the intramedullary canal using a 26-gauge needle, whereupon a precut 0.8-cm-length, orthopedic-grade Kirschner (K) wire (0.6 mm in diameter; Nitinol [nickel-titanium]; Custom Wire Technologies, Port Washington, WI) was inserted into the intramedullary canal, leaving ∼1 mm protruding into the joint space. A total of 103 or 105 CFU of S. aureus was inoculated at the implant tip depending on the experimental design. Analgesia (Buprenex, 0.1 mg/kg [administered subcutaneously]; Reckitt Benckiser, Hull, United Kingdom) was administered immediately after infection and again 24 h later for pain relief. After this interval, all mice exhibited normal ambulation and no discernible pain behaviors.

Bacterial strains and preparation of the bacterial inoculum.

Three bacterial strains, representing different epidemiological backgrounds, were utilized for these studies. S. aureus USA300 LAC, which was originally isolated from a patient with a skin and soft tissue infection (16, 21), was cured of two antibiotic resistance plasmids and designated USA300 LAC-13C (referred to as LAC in the present study) as previously described (41). S. aureus MW2 is a USA400 CA-MRSA clinical isolate recovered from a fatal pediatric infection and was kindly provided by Keer Sun at UNMC (22, 42). S. aureus UAMS-1, a USA200 MSSA strain, was isolated from a patient with osteomyelitis and was kindly provided by Mark Smeltzer at the University of Arkansas for Medical Sciences (Little Rock, AR) (24). All three isolates were utilized for strain comparison studies, whereas dose-response assessments were only performed with LAC and UAMS-1.

For all experiments, bacterial strains were prepared by inoculating 25 ml of brain heart infusion medium (BD Biosciences, Sparks, MD) with a single colony from a fresh streak plate made 24 h earlier. Cultures were grown at 37°C with constant aeration for 12 h, with a 1:10 volume-flask ratio, whereupon an aliquot was removed and diluted 1:10 with PBS, for optical density measurements. Absorbance readings (an optical density at 620 nm) were used to estimate culture densities, extrapolated from previous studies and growth curves. A total of 1 ml from the 12-h culture was removed and pelleted by centrifugation (20,000 × g, 5 min, 4°C), and the bacteria were washed three times with 1 ml of phosphate-buffered saline (PBS). The final bacterial pellet was resuspended in 1 ml of PBS, and infectious inocula were prepared by diluting organisms to 5 × 105 or 5 × 107 CFU/ml in PBS to achieve the desired infectious doses of 103 or 105 CFU/mouse. Actual bacterial inocula were calculated retrospectively by plating serial dilutions of each working stock onto blood agar plates, which were consistently found to accurately reflect the inoculum estimates (103 or 105 CFU/mouse).

Flow cytometry.

To characterize leukocyte infiltrates associated with S. aureus PJI, soft tissues surrounding the knee joint were dissociated in Hanks balanced salt solution (HBSS) containing 10% fetal bovine serum (FBS; HyClone, Logan, UT) using the flat end of a plunger from a 3-ml syringe and subsequently passed through a 70-μm-pore-size filter (BD Falcon, Bedford, MA). The resulting filtrate was washed with HBSS–10% FBS, and cells were collected by centrifugation (300 × g, 10 min), whereupon red blood cells were lysed using BD Pharm Lyse (BD Biosciences, San Diego, CA) according to the manufacturer's instructions. After lysis, the cells were washed and resuspended in PBS, followed by incubation in mouse Fc block (BD Biosciences) to minimize nonspecific antibody binding. The cells were then stained for 30 min with directly conjugated antibodies for multicolor flow cytometry analysis, which included two separate panels to identify innate immune populations (CD45-APC, Ly6G-PE, Ly6CPerCP-Cy5.5, and F4/80-PE-Cy7) or T cells (CD3ε-APC, CD4-Pacific Blue, CD8a-FITC, and TCR γδ-PE) with a viability dye to exclude dead cells (Live/Dead Fixable Blue dead cell stain kit; Life Technologies, Eugene, OR). All fluorochrome-conjugated antibodies were purchased from either BD Biosciences or eBioscience. Controls included cells stained with isotype control antibodies to assess the degree of nonspecific staining, as well as fluorescence minus one to identify gating thresholds (43). Analysis was performed using BD FACSDiva software with results presented as the percentage of CD45+ or CD3+ gated cells.

Multianalyte microbead arrays.

To compare inflammatory mediator profiles elicited by the various S. aureus strains during PJI, a custom-designed mouse microbead array was used (Milliplex; Millipore, Billerica, MA), which detects the following mediators: granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-1α (IL-1α), IL-1β, IL-6, IL-9, IL-10, IL-12p40, IL-13, IL-17, CCL2, CCL3, CCL5, CXCL2, CXCL9, CXCL10, tumor necrosis factor alpha (TNF-α), and vascular endothelial growth factor (VEGF). A Bio-Plex workstation (Bio-Rad, Hercules, CA) was used to analyze results with values normalized to the total amount of protein recovered from each sample to correct for differences in tissue sampling size.

Statistical analysis.

Significant differences between experimental groups were determined by using an unpaired two-tailed Student t test using GraphPad Prism version 4 (GraphPad Software, La Jolla, CA). For all analyses, P < 0.05 was considered statistically significant.

RESULTS

Distinct S. aureus clinical isolates elicit similar immunological responses and biofilm formation during PJI.

When examining immunological responses to S. aureus, determining the most appropriate isolate to best model clinical disease is an important consideration. To evaluate the importance of strain on infection outcome and inflammation, we compared three commonly used S. aureus isolates, namely, LAC, MW2, and UAMS-1, in a mouse model of orthopedic-implant biofilm infection that models PJI in humans. As previously reported in our laboratory with USA300 LAC (2527), this is a well-established low dose infection model associated with rapid bacterial expansion, with titers most prominent in the soft tissue surrounding the infected knee joint followed by the tendons/ligaments comprising the joint, femur, and implant (Fig. 1). All three S. aureus strains displayed equivalent burdens in the soft tissue surrounding the knee (Fig. 1A) and, in general, the titers were similar in the other compartments, with the exception of MW2, which was significantly greater in the joint and femur at day 7 (Fig. 1B and C). However, these increases were only apparent during the first week after infection and dissipated thereafter, with all three strains displaying roughly equivalent infectious burdens at day 14 (Fig. 1).

FIG 1.

FIG 1

Establishment of S. aureus orthopedic biofilm infection is not strain dependent. C57BL/6 mice were infected with 103 CFU of S. aureus LAC, MW2, or UAMS-1, whereupon the soft tissue surrounding the joint (A), knee joint (B), femur (C), and titanium implant (D) was collected 7 and 14 days after infection for quantitation of bacterial burdens. The results are expressed as CFU per milliliter for the implant and CFU per gram of tissue (for tissue, knee joint, and femur) to normalize for differences in sampling size and are presented from individual animals combined from two independent experiments (n = 8 to 12 mice per group). Significant differences between experimental groups were determined by an unpaired, two-tailed Student t test (*, P < 0.05; **, P < 0.01).

As previously described in our S. aureus PJI model (25, 26), the innate immune response is predominantly composed of MDSCs (50 to 80% of the CD45+ population), which persist throughout infection (Fig. 2A). Monocytes, macrophages, and T cells are also recruited to the site of infection but are much less abundant than the MDSC infiltrate (Fig. 2B to D). Overall, LAC, MW2, and UAMS-1 elicited similar inflammatory cell infiltrates, with the exception of UAMS-1 at day 7, where MDSCs were significantly, although modestly reduced (Fig. 2A). However, the biological significance of this finding is questionable, since it did not translate into enhanced monocyte or macrophage recruitment, which we have previously shown occurs when MDSC infiltrates are decreased (2527). Likewise, levels of inflammatory cytokine/chemokine production in the soft tissue surrounding infected joints of mice challenged with LAC or UAMS-1 were similar (G-CSF, IL-10, CCL2, CCL5, CXCL2, and VEGF; Fig. 3 and data not shown). Although the production of some mediators was increased in UAMS-1-infected tissues compared to LAC at day 7 or 14 (IL-1α, IL-1β, IL-9, CCL3, CXCL9, and CXCL10; Fig. 3 and data not shown), this did not influence bacterial persistence or leukocyte infiltrates at the same time points (Fig. 1 and 2, respectively) and thus was not considered biologically significant. UAMS-1 and LAC were utilized for all subsequent dose-response studies, since UAMS-1 originated from an orthopedic infection and we have a great deal of knowledge about the behavior of LAC in the orthopedic-implant model (2527, 40). This approach was further justified by the finding that MW2 behaved similarly to both UAMS-1 and LAC in terms of immune profiles and biofilm burdens.

FIG 2.

FIG 2

S. aureus clinical isolates recruit similar leukocyte infiltrates after PJI. C57BL/6 mice were infected with 103 CFU of S. aureus LAC, MW2, or UAMS-1, whereupon MDSC (A), monocyte (B), macrophage (C), and T cell (D) infiltrates from the soft tissue surrounding the infected knee joint were analyzed by flow cytometry at days 7 and 14 postinfection. The results are presented as means ± the standard deviations (SD) from two independent experiments (n = 8 to 12 mice per group). Significant differences between experimental groups were determined by an unpaired, two-tailed Student t test (*, P < 0.05).

FIG 3.

FIG 3

S. aureus LAC and UAMS-1 elicit similar cytokine and chemokine profiles. C57BL/6 mice were infected with 103 CFU of S. aureus LAC or UAMS-1, whereupon IL-1β, G-CSF, CXCL2, and CCL5 production was measured in the tissue surrounding the knee joint at days 7 and 14 postinfection by multianalyte bead arrays. The results were normalized to the amount of total protein to correct for differences in tissue sampling size and are presented as means ± the SD from one representative experiment (n = 5 mice per group). Significant differences between experimental groups were determined by an unpaired, two-tailed Student t test (*, P < 0.05).

Infectious dose influences inflammatory responses and bacterial persistence during S. aureus PJI.

Another important factor when translating mouse infection models to human disease is infectious dose. This is particularly relevant when considering the initial bacterial challenge to the immune system, especially since the number of organisms seeding a given site during human infection is usually lower than the high S. aureus inocula used in many mouse models (i.e., 105 to 107 CFU) (30, 4446). For example, a high infectious dose is likely to elicit an immediate and robust proinflammatory response attributed to a greater bacterial biomass (mediated by lipoteichoic acids, peptidoglycan, etc.). In contrast, a low inoculum may afford S. aureus an early advantage to colonize and propagate by not eliciting an initial vigorous proinflammatory response, as suggested by our prior studies (2527).

IL-12p40 KO mice were used to investigate how the immune response to S. aureus was influenced by infectious dose, since our prior study showed that these animals had significantly reduced MDSC infiltrates and improved bacterial clearance in response to low-dose LAC infection (103 CFU), which was confirmed in the present study (Fig. 4 and 5, respectively) (26). However, when the bacterial inoculum was increased to 105 these differences between WT and IL-12p40 KO mice were no longer apparent. Specifically, MDSCs were significantly reduced concomitant with increased monocytes and macrophages in IL-12p40 KO mice in response to 103 CFU (Fig. 4, indicated by asterisks), which was negated with the 105 CFU dose, where all three populations were equivalent between IL-12p40 KO and WT animals at both 7 and 14 days postinfection (Fig. 4).

FIG 4.

FIG 4

Leukocyte recruitment in response to S. aureus LAC is influenced by infectious dose. Wild-type (WT) or IL-12p40 knockout (KO) mice were infected with 103 or 105 CFU of S. aureus LAC, whereupon MDSC (A), monocyte (B), macrophage (C), and T cell (D) infiltrates from the soft tissue surrounding the infected knee joint were analyzed by flow cytometry at days 7 and 14 postinfection. The results are presented as means ± the SD from three independent experiments (n = 11 to 18 mice per group). Significance was determined using an unpaired, two-tailed Student t test, with differences between WT and IL-12p40 KO mice denoted by asterisks (*, P < 0.05; **, P < 0.01; ***, P < 0.005; ****, P < 0.001), whereas differences between WT or IL-12p40 KO animals receiving 103 CFU versus 105 CFU are indicated by hash signs (#, P < 0.05; ##, P < 0.01; ####, P < 0.001).

FIG 5.

FIG 5

Biofilm establishment by S. aureus LAC is dependent on infectious dose. WT or IL-12p40 KO mice were infected with 103 or 105 CFU of S. aureus LAC, whereupon the soft tissue surrounding the joint (A), knee joint (B), femur (C), and titanium implant (D) was collected 7 and 14 days after infection for quantitation of bacterial burdens. The results are expressed as CFU per milliliter for the implant and CFU per gram of tissue (for tissue, knee joint, and femur) to normalize for differences in sampling size and are presented from individual animals combined from three independent experiments (n = 15 to 18 mice per group). Significant differences between experimental groups were determined by an unpaired, two-tailed Student t test (*, P < 0.05; **, P < 0.01; ***, P < 0.005; ****, P < 0.001).

A dose-response relationship was also detected with bacterial burdens. As we previously reported, biofilm titers were significantly reduced in IL-12p40 KO animals at both 7 and 14 days postinfection with an inoculum of 103 CFU. However, when the dose was increased to 105 CFU, bacterial clearance was no longer evident, since the biofilm burdens in IL-12p40 KO animals were now nearly identical or increased compared to WT mice (Fig. 5).

Similar dose-dependent effects were observed with regard to inflammatory mediator production, where numerous cytokines/chemokines (G-CSF, IL-1α, IL-1β, CXCL2, CCL2, CCL3, CCL5; Fig. 6 and data not shown) were significantly reduced in IL-12p40 KO mice after a 103 CFU challenge in agreement with our earlier report (26). In contrast, these same inflammatory mediators were similar between IL-12p40 KO and WT animals that received an inoculum of 105 CFU (Fig. 6 and data not shown). The production of IL-9, CCL2, CXCL9, CXCL10, and VEGF in IL-12p40 KO and WT mice was very low and did not differ between the two infectious inocula. We previously reported that MDSCs support biofilm persistence, in part, through IL-10 induction (25). IL-10 levels were slightly reduced in IL-12p40 KO mice at days 7 and 14 postinfection with the 103 CFU dose; however, these differences did not reach statistical significance. Nevertheless, reduced IL-10 production in IL-12p40 KO animals correlated with improved bacterial clearance (Fig. 5), which is in agreement with our prior report demonstrating a role for IL-10 in promoting biofilm persistence (25). Similar dosage-dependent changes in IL-12p40 KO mice were observed with UAMS-1 (data not shown).

FIG 6.

FIG 6

A high infectious dose of S. aureus LAC alters cytokine and chemokine responses. WT or IL-12p40 KO mice were infected with 103 or 105 CFU of S. aureus LAC, whereupon IL-1β, G-CSF, CXCL2, and CCL5 production was measured in the tissue surrounding the knee joint at days 7 and 14 postinfection by multianalyte bead arrays. The results are normalized to the amount of total protein to correct for differences in tissue sampling size and are presented as means ± the SD from a representative experiment (n = 5 mice per group). Significance was determined using an unpaired, two-tailed Student t test, with differences between WT and IL-12p40 KO mice denoted by asterisks (*, P < 0.05; **, P < 0.01; ***, P < 0.005; ****, P < 0.001), whereas differences between WT or IL-12p40 KO animals receiving 103 CFU versus 105 CFU are indicated by hash signs (#, P < 0.05; ##, P < 0.01; ###, P < 0.005).

Dose-response effects of S. aureus challenge were also observed when comparing differences within WT or IL-12p40 KO animals. For example, MDSC and macrophage recruitment was significantly increased in WT mice receiving 105 CFU of S. aureus compared to 103 CFU at day 7 postinfection (Fig. 4, indicated by hash signs). However, dose-dependent differences in MDSC infiltrates were more pronounced in IL-12p40 KO animals, where 105 CFU resulted in major increases in MDSC recruitment at both days 7 and 14 postinfection (Fig. 4, indicated by hash signs). Differences in biofilm burdens were also observed in WT animals following low- versus high-dose bacterial challenge. Specifically, infection with 103 CFU resulted in bacterial expansion from day 7 to 14 in WT mice; however, this increase did not reach statistical significance. In contrast, when the higher inoculum was used (105 CFU), bacterial burdens were similar at both time points in WT animals. Our prior studies have demonstrated that biofilm burdens reach a threshold titer (normally by day 14 following a 103 CFU challenge) that can no longer expand in vivo, even with increasing time postinfection. Our interpretation for the failure to observe bacterial expansion over time with the 105 CFU dose in WT animals was that biofilm development was accelerated and reached its maximal titer by day 7, since less time is required to reach maximal device colonization, effectively masking bacterial expansion. This could have dramatic consequences on the stages of biofilm development, particularly when considering the prominent role of quorum sensing during biofilm formation. Finally, increasing the dose to 105 CFU resulted in heightened inflammatory mediator production compared to 103 CFU, although this was most evident in IL-12p40 KO mice at both 7 and 14 days after infection (Fig. 6, indicated by hash signs). Collectively, these findings have established that the infectious inoculum can dictate leukocyte recruitment, inflammatory mediator production, and biofilm growth/clearance and highlight the need to carefully consider infectious doses when examining inflammatory attributes of biofilm infection.

DISCUSSION

The number of patients undergoing primary total hip and knee arthroplasties has steadily increased over the past decade, with nearly 800,000 procedures being performed in the United States each year (2). Prosthetic joint infection (PJI) is a serious complication after arthroplasty, with S. aureus being a common inciting pathogen (2). A recent study using National Inpatient Sample data from 1990 to 2003 projected the infection incidence following total hip revision to increase from 3,400 in 2005 to 46,000 in 2030, and for total knee replacements to increase from 6,400 in 2003 to 175,500 in 2030 based on the increased volume of prosthetic joint replacement procedures with an aging population (47). In addition, S. aureus PJIs are challenging because of their treatment complexity and increased risk of recurrent infection (2, 5). It is currently unclear whether certain S. aureus isolates have a propensity to cause PJIs versus colonization of other tissue sites.

The three S. aureus clinical isolates examined in this study (LAC, MW2, and UAMS-1) displayed roughly equivalent abilities to establish infection and elicited comparable immune responses when WT animals were challenged with a low infectious inoculum (103 CFU). Whereas strain-dependent preferences have been suggested for the colonization of native tissues (i.e., USA300 strains preferentially causing skin and soft tissue infections) (48, 49), it appears that the presence of an implant likely negates these differences and facilitates bacterial attachment/persistence regardless of the strain during PJI. In general, the immune responses elicited by all three S. aureus isolates were similar, which from a therapeutic standpoint is advantageous, since a conserved immune response would be more amenable to a singular treatment regimen targeted toward enhancing antimicrobial immunity.

S. aureus is a highly opportunistic pathogen that expresses numerous virulence determinants designed to circumvent host immune responses and promote bacterial persistence. As we previously demonstrated (26), IL-12 is a critical cytokine for shaping the immune response after PJI and determining infection outcome, since IL-12p40 KO mice were more resistant to S. aureus colonization and biofilm formation during PJI than WT animals following the administration of a low inoculum (103 CFU). Importantly, this phenotype was negated when IL-12p40 KO animals were challenged with a 2-log-higher dose of LAC (105 CFU). Since bacterial division is very rapid, we propose that the initial introduction of large numbers of organisms quickly transitions into a large bacterial biomass, which elicits a vigorous proinflammatory response that is not reminiscent of what occurs during native infection in humans where tissues are often colonized with few organisms. This is supported by our findings with IL-12p40 KO animals, showing that the large infectious dose elicits a different series of events that changes the inflammatory infiltrate and prevents S. aureus clearance. This emphasizes the critical importance of the infectious dose, particularly when evaluating the efficacy of potential therapeutics for PJI in preclinical studies.

ACKNOWLEDGMENTS

We thank Casey Gries and Tyler Scherr for detailed discussions, Jessica Snowden and Keer Sun for critical review of the manuscript, and Rachel Fallet for maintenance of IL-12p40 KO mice. We also thank Phillip Hexley, Victoria Smith, and Samantha Wall of the University of Nebraska Medical Center Flow Cytometry Research Facility for their assistance with fluorescence-activated cell sorting analysis.

This study was supported by the National Institutes of Health/National Institute of Allergy and Infectious Disease grant P01 AI083211 (project 4 to T.K.). The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

REFERENCES

  • 1.Boucher H, Miller LG, Razonable RR. 2010. Serious infections caused by methicillin-resistant Staphylococcus aureus. Clin Infect Dis 51(Suppl 2):S183–S197. doi: 10.1086/653519. [DOI] [PubMed] [Google Scholar]
  • 2.Del Pozo JL, Patel R. 2009. Clinical practice: infection associated with prosthetic joints. N Engl J Med 361:787–794. doi: 10.1056/NEJMcp0905029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Grundmann H, Hellriegel B. 2006. Mathematical modeling: a tool for hospital infection control. Lancet Infect Dis 6:39–45. doi: 10.1016/S1473-3099(05)70325-X. [DOI] [PubMed] [Google Scholar]
  • 4.Pulido L, Ghanem E, Joshi A, Purtill JJ, Parvizi J. 2008. Periprosthetic joint infection: the incidence, timing, and predisposing factors. Clin Orthop Relat Res 466:1710–1715. doi: 10.1007/s11999-008-0209-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zimmerli W, Trampuz A, Ochsner PE. 2004. Prosthetic-joint infections. N Engl J Med 351:1645–1654. doi: 10.1056/NEJMra040181. [DOI] [PubMed] [Google Scholar]
  • 6.Vuong C, Kocianova S, Yao Y, Carmody AB, Otto M. 2004. Increased colonization of indwelling medical devices by quorum-sensing mutants of Staphylococcus epidermidis in vivo. J Infect Dis 190:1498–1505. doi: 10.1086/424487. [DOI] [PubMed] [Google Scholar]
  • 7.Hanke ML, Kielian T. 2012. Deciphering mechanisms of staphylococcal biofilm evasion of host immunity. Front Cell Infect Microbiol 2:62. doi: 10.3389/fcimb.2012.00062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kocianova S, Vuong C, Yao Y, Voyich JM, Fischer ER, DeLeo FR, Otto M. 2005. Key role of poly-γ-dl-glutamic acid in immune evasion and virulence of Staphylococcus epidermidis. J Clin Invest 115:688–694. doi: 10.1172/JCI200523523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Scherr TD, Heim CE, Morrison JM, Kielian T. 2014. Hiding in plain sight: interplay between staphylococcal biofilms and host immunity. Front Immunol 5:37. doi: 10.3389/fimmu.2014.00037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.del Pozo JL, Patel R. 2007. The challenge of treating biofilm-associated bacterial infections. Clin Pharmacol Ther 82:204–209. doi: 10.1038/sj.clpt.6100247. [DOI] [PubMed] [Google Scholar]
  • 11.Kilgus DJ, Howe DJ, Strang A. 2002. Results of periprosthetic hip and knee infections caused by resistant bacteria. Clin Orthop Relat Res 116–124. [DOI] [PubMed] [Google Scholar]
  • 12.Salgado CD, Dash S, Cantey JR, Marculescu CE. 2007. Higher risk of failure of methicillin-resistant Staphylococcus aureus prosthetic joint infections. Clin Orthop Relat Res 461:48–53. [DOI] [PubMed] [Google Scholar]
  • 13.Teterycz D, Ferry T, Lew D, Stern R, Assal M, Hoffmeyer P, Bernard L, Uckay I. 2010. Outcome of orthopedic implant infections due to different staphylococci. Int J Infect Dis 14:e913–e918. doi: 10.1016/j.ijid.2010.05.014. [DOI] [PubMed] [Google Scholar]
  • 14.Adem PV, Montgomery CP, Husain AN, Koogler TK, Arangelovich V, Humilier M, Boyle-Vavra S, Daum RS. 2005. Staphylococcus aureus sepsis and the Waterhouse-Friderichsen syndrome in children. N Engl J Med 353:1245–1251. doi: 10.1056/NEJMoa044194. [DOI] [PubMed] [Google Scholar]
  • 15.Francis JS, Doherty MC, Lopatin U, Johnston CP, Sinha G, Ross T, Cai M, Hansel NN, Perl T, Ticehurst JR, Carroll K, Thomas DL, Nuermberger E, Bartlett JG. 2005. Severe community-onset pneumonia in healthy adults caused by methicillin-resistant Staphylococcus aureus carrying the Panton-Valentine leukocidin genes. Clin Infect Dis 40:100–107. doi: 10.1086/427148. [DOI] [PubMed] [Google Scholar]
  • 16.Miller LG, Perdreau-Remington F, Rieg G, Mehdi S, Perlroth J, Bayer AS, Tang AW, Phung TO, Spellberg B. 2005. Necrotizing fasciitis caused by community-associated methicillin-resistant Staphylococcus aureus in Los Angeles. N Engl J Med 352:1445–1453. doi: 10.1056/NEJMoa042683. [DOI] [PubMed] [Google Scholar]
  • 17.Mongkolrattanothai K, Boyle S, Kahana MD, Daum RS. 2003. Severe Staphylococcus aureus infections caused by clonally related community-acquired methicillin-susceptible and methicillin-resistant isolates. Clin Infect Dis 37:1050–1058. doi: 10.1086/378277. [DOI] [PubMed] [Google Scholar]
  • 18.Gonzalez BE, Martinez-Aguilar G, Hulten KG, Hammerman WA, Coss-Bu J, Avalos-Mishaan A, Mason EO Jr, Kaplan SL. 2005. Severe staphylococcal sepsis in adolescents in the era of community-acquired methicillin-resistant Staphylococcus aureus. Pediatrics 115:642–648. doi: 10.1542/peds.2004-2300. [DOI] [PubMed] [Google Scholar]
  • 19.Guimaraes MA, Ramundo MS, Americo MA, de Mattos MC, Souza RR, Ramos-Junior ES, Coelho LR, Morrot A, Melo PA, Fracalanzza SE, Ferreira FA, Figueiredo AM. 2015. A comparison of virulence patterns and in vivo fitness between hospital- and community-acquired methicillin-resistant Staphylococcus aureus related to the USA400 clone. Eur J Clin Microbiol Infect Dis 34:497–509. doi: 10.1007/s10096-014-2253-1. [DOI] [PubMed] [Google Scholar]
  • 20.Kourbatova EV, Halvosa JS, King MD, Ray SM, White N, Blumberg HM. 2005. Emergence of community-associated methicillin-resistant Staphylococcus aureus USA300 clone as a cause of health care-associated infections among patients with prosthetic joint infections. Am J Infect Control 33:385–391. doi: 10.1016/j.ajic.2005.06.006. [DOI] [PubMed] [Google Scholar]
  • 21.Kennedy AD, Otto M, Braughton KR, Whitney AR, Chen L, Mathema B, Mediavilla JR, Byrne KA, Parkins LD, Tenover FC, Kreiswirth BN, Musser JM, DeLeo FR. 2008. Epidemic community-associated methicillin-resistant Staphylococcus aureus: recent clonal expansion and diversification. Proc Natl Acad Sci U S A 105:1327–1332. doi: 10.1073/pnas.0710217105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Centers for Disease Control and Prevention. 1999. Four pediatric deaths from community-acquired methicillin-resistant Staphylococcus aureus–Minnesota and North Dakota, 1997-1999. JAMA 282:1123–1125. [PubMed] [Google Scholar]
  • 23.Baba T, Takeuchi F, Kuroda M, Yuzawa H, Aoki K, Oguchi A, Nagai Y, Iwama N, Asano K, Naimi T, Kuroda H, Cui L, Yamamoto K, Hiramatsu K. 2002. Genome and virulence determinants of high virulence community-acquired MRSA. Lancet 359:1819–1827. doi: 10.1016/S0140-6736(02)08713-5. [DOI] [PubMed] [Google Scholar]
  • 24.Gillaspy AF, Hickmon SG, Skinner RA, Thomas JR, Nelson CL, Smeltzer MS. 1995. Role of the accessory gene regulator (agr) in pathogenesis of staphylococcal osteomyelitis. Infect Immun 63:3373–3380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Heim CE, Vidlak D, Kielian T. 2015. Interleukin-10 production by myeloid-derived suppressor cells contributes to bacterial persistence during Staphylococcus aureus orthopedic biofilm infection. J Leukoc Biol 98:1003–1013. doi: 10.1189/jlb.4VMA0315-125RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Heim CE, Vidlak D, Scherr TD, Hartman CW, Garvin KL, Kielian T. 2015. IL-12 promotes myeloid-derived suppressor cell recruitment and bacterial persistence during Staphylococcus aureus orthopedic implant infection. J Immunol 194:3861–3872. doi: 10.4049/jimmunol.1402689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Heim CE, Vidlak D, Scherr TD, Kozel JA, Holzapfel M, Muirhead DE, Kielian T. 2014. Myeloid-derived suppressor cells contribute to Staphylococcus aureus orthopedic biofilm infection. J Immunol 192:3778–3792. doi: 10.4049/jimmunol.1303408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kim HK, Missiakas D, Schneewind O. 2014. Mouse models for infectious diseases caused by Staphylococcus aureus. J Immunol Methods 410:88–99. doi: 10.1016/j.jim.2014.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Reizner W, Hunter JG, O'Malley NT, Southgate RD, Schwarz EM, Kates SL. 2014. A systematic review of animal models for Staphylococcus aureus osteomyelitis. Eur Cell Mater 27:196–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Prabhakara R, Harro JM, Leid JG, Keegan AD, Prior ML, Shirtliff ME. 2011. Suppression of the inflammatory immune response prevents the development of chronic biofilm infection due to methicillin-resistant Staphylococcus aureus. Infect Immun 79:5010–5018. doi: 10.1128/IAI.05571-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ghosh C, Manjunath GB, Konai MM, Uppu DS, Hoque J, Paramanandham K, Shome BR, Haldar J. 2015. Aryl-alkyl-lysines: agents that kill planktonic cells, persister cells, biofilms of MRSA, and protect mice from skin infection. PLoS One 10:e0144094. doi: 10.1371/journal.pone.0144094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jorgensen NP, Meyer RL, Dagnaes-Hansen F, Fuursted K, Petersen E. 2014. A modified chronic infection model for testing treatment of Staphylococcus aureus biofilms on implants. PLoS One 9:e103688. doi: 10.1371/journal.pone.0103688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kwiecinski J, Na M, Jarneborn A, Jacobsson G, Peetermans M, Verhamme P, Jin T. 2015. Tissue plasminogen activator coating on implant surfaces reduces Staphylococcus aureus biofilm formation. Appl Environ Microbiol 82:394–401. doi: 10.1128/AEM.02803-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nishitani K, Sutipornpalangkul W, de Mesy Bentley KL, Varrone JJ, Bello-Irizarry SN, Ito H, Matsuda S, Kates SL, Daiss JL, Schwarz EM. 2015. Quantifying the natural history of biofilm formation in vivo during the establishment of chronic implant-associated Staphylococcus aureus osteomyelitis in mice to identify critical pathogen and host factors. J Orthop Res 33:1311–1319. doi: 10.1002/jor.22907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rochford ET, Sabate Bresco M, Zeiter S, Kluge K, Poulsson A, Ziegler M, Richards RG, O'Mahony L, Moriarty TF. 2015. Monitoring immune responses in a mouse model of fracture fixation with and without Staphylococcus aureus osteomyelitis. Bone 83:82–92. doi: 10.1016/j.bone.2015.10.014. [DOI] [PubMed] [Google Scholar]
  • 36.Thangamani S, Nepal M, Chmielewski J, Seleem MN. 2015. Antibacterial activity and therapeutic efficacy of Fl-P(R)P(R)P(L)-5, a cationic amphiphilic polyproline helix, in a mouse model of staphylococcal skin infection. Drug Des Dev Ther 9:5749–5754. doi: 10.2147/DDDT.S94505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pribaz JR, Bernthal NM, Billi F, Cho JS, Ramos RI, Guo Y, Cheung AL, Francis KP, Miller LS. 2012. Mouse model of chronic post-arthroplasty infection: noninvasive in vivo bioluminescence imaging to monitor bacterial burden for long-term study. J Orthop Res 30:335–340. doi: 10.1002/jor.21519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bernthal NM, Pribaz JR, Stavrakis AI, Billi F, Cho JS, Ramos RI, Francis KP, Iwakura Y, Miller LS. 2011. Protective role of IL-1β against post-arthroplasty Staphylococcus aureus infection. J Orthop Res 29:1621–1626. doi: 10.1002/jor.21414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Scherr TD, Lindgren KE, Schaeffer CR, Hanke ML, Hartman CW, Kielian T. 2014. Mouse model of post-arthroplasty Staphylococcus epidermidis joint infection. Methods Mol Biol 1106:173–181. doi: 10.1007/978-1-62703-736-5_16. [DOI] [PubMed] [Google Scholar]
  • 40.Scherr TD, Hanke ML, Huang O, James DB, Horswill AR, Bayles KW, Fey PD, Torres VJ, Kielian T. 2015. Staphylococcus aureus biofilms induce macrophage dysfunction through leukocidin AB and alpha-toxin. mBio 6:e01021-15. doi: 10.1128/mBio.01021-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fey PD, Endres JL, Yajjala VK, Widhelm TJ, Boissy RJ, Bose JL, Bayles KW. 2013. A genetic resource for rapid and comprehensive phenotype screening of nonessential Staphylococcus aureus genes. mBio 4:e00537-12. doi: 10.1128/mBio.00537-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.McDougal LK, Steward CD, Killgore GE, Chaitram JM, McAllister SK, Tenover FC. 2003. Pulsed-field gel electrophoresis typing of oxacillin-resistant Staphylococcus aureus isolates from the United States: establishing a national database. J Clin Microbiol 41:5113–5120. doi: 10.1128/JCM.41.11.5113-5120.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Herzenberg LA, Tung J, Moore WA, Herzenberg LA, Parks DR. 2006. Interpreting flow cytometry data: a guide for the perplexed. Nat Immunol 7:681–685. doi: 10.1038/ni0706-681. [DOI] [PubMed] [Google Scholar]
  • 44.Cassat JE, Hammer ND, Campbell JP, Benson MA, Perrien DS, Mrak LN, Smeltzer MS, Torres VJ, Skaar EP. 2013. A secreted bacterial protease tailors the Staphylococcus aureus virulence repertoire to modulate bone remodeling during osteomyelitis. Cell Host Microbe 13:759–772. doi: 10.1016/j.chom.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Prabhakara R, Harro JM, Leid JG, Harris M, Shirtliff ME. 2011. Murine immune response to a chronic Staphylococcus aureus biofilm infection. Infect Immun 79:1789–1796. doi: 10.1128/IAI.01386-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tebartz C, Horst SA, Sparwasser T, Huehn J, Beineke A, Peters G, Medina E. 2015. A major role for myeloid-derived suppressor cells and a minor role for regulatory T cells in immunosuppression during Staphylococcus aureus infection. J Immunol 194:1100–1111. doi: 10.4049/jimmunol.1400196. [DOI] [PubMed] [Google Scholar]
  • 47.Kurtz SM, Lau E, Schmier J, Ong KL, Zhao K, Parvizi J. 2008. Infection burden for hip and knee arthroplasty in the United States. J Arthroplasty 23:984–991. doi: 10.1016/j.arth.2007.10.017. [DOI] [PubMed] [Google Scholar]
  • 48.David MZ, Daum RS. 2010. Community-associated methicillin-resistant Staphylococcus aureus: epidemiology and clinical consequences of an emerging epidemic. Clin Microbiol Rev 23:616–687. doi: 10.1128/CMR.00081-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gordon RJ, Lowy FD. 2008. Pathogenesis of methicillin-resistant Staphylococcus aureus infection. Clin Infect Dis 46(Suppl 5):S350–S359. doi: 10.1086/533591. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES