Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 15.
Published in final edited form as: J Immunol. 2021 Dec 20;208(2):454–463. doi: 10.4049/jimmunol.2100752

Altered mitochondrial homeostasis during systemic lupus erythematosus impairs NET formation rendering neutrophils ineffective at combating Staphylococcus aureus

Andrew J Monteith 1, Jeanette M Miller 1, Jonathan M Williams 2,3, Kelsey Voss 1, Jeffrey C Rathmell 1,2,3,4, Leslie J Crofford 1,3,4, Eric P Skaar 1,2,3,4,*
PMCID: PMC8761356  NIHMSID: NIHMS1757204  PMID: 34930781

Abstract

Inflammation involves a delicate balance between pathogen clearance and limiting host tissue damage, and perturbations in this equilibrium promote disease. Patients suffering from autoimmune diseases, such as systemic lupus erythematosus (SLE), have higher levels of serum S100A9 protein and increased risk for infection. S100A9 is highly abundant within neutrophils and modulates antimicrobial activity in response to bacterial pathogens. We reasoned that increased serum S100A9 in SLE patients reflects accumulation of S100A9 protein in neutrophils and may indicate altered neutrophil function. Herein, we demonstrate elevated S100A9 protein within neutrophils from SLE patients and MRL/lpr mice associates with lower mitochondrial superoxide, decreased suicidal neutrophil extracellular trap formation, and increased susceptibility to Staphylococcus aureus infection. Furthermore, increasing mitochondrial superoxide production restored the antibacterial activity of MRL/lpr neutrophils in response to S. aureus. These results demonstrate that accumulation of intracellular S100A9 associates with impaired mitochondrial homeostasis thereby rendering SLE neutrophils inherently less bactericidal.

Introduction

Staphylococcus aureus is a Gram-positive bacterium that is the leading cause of soft tissue infections (1, 2), bacterial endocarditis (35), and the second-most frequent agent of hospital-associated pneumonia (6) and bloodstream infections (79). Neutrophil-mediated inflammation is a hallmark of the innate immune response to S. aureus (10, 11). Calprotectin is a heterodimer formed by the S100A8 and S100A9 proteins (12, 13) and comprises approximately 50% of the cytoplasmic protein in neutrophils (14), and significantly influences disease outcome during S. aureus infection. When S100A9-deficient (A9−/−) mice, which lack calprotectin, are systemically infected with S. aureus they exhibit increased bacterial burdens in the liver and kidney (15, 16) but have lower bacterial burdens in the heart with increased overall survival (17, 18), compared to wild-type (WT) mice. While an emphasis has been placed on the ability of calprotectin to sequester nutrient metals from bacterial pathogens (15, 16, 1924), an intracellular function for S100A9 in regulating the antibacterial activity of neutrophils was described recently; whereby loss of S100A9 coincides with increased mitochondrial superoxide (O2) production (18).

In response to pathogens, neutrophils undergo two distinct mechanisms of neutrophil extracellular trap (NET) formation consisting of a meshwork of DNA and antimicrobial peptides and proteins. Suicidal NET formation occurs within 2–4 hr of activation resulting in the terminal release of NETs through rupture of the cell membrane (25), while vital NET formation occurs within 5–60 min of engaging S. aureus whereby NETs are exocytosed and leave the membrane intact (26). The formation of NETs plays an integral role in combating S. aureus infections by restricting bacterial dissemination and eliciting direct bactericidal activity (27, 28) as well as augmenting macrophage bactericidal activity (18). Increased production of mitochondrial O2 in A9−/− neutrophils causes accelerated and more robust suicidal NET formation, and impaired primary degranulation in response to S. aureus (18). Consistent with this, A9−/− mice exhibit increased survival with reduced bacterial burdens specifically in the heart (17, 18).

Chronic inflammation contributes to autoimmune diseases including systemic lupus erythematosus (SLE). SLE is an autoimmune disease with genetic and environmental components leading to tissue-damaging inflammation (29, 30). Neutrophils contribute to SLE pathology by increasing the burden of nuclear antigens through NET formation (31, 32) and heightens the risk for cardiovascular disease in SLE patients (3335). It has been observed by multiple groups that SLE disease activity correlates with increased serum S100A9 protein levels (34, 36, 37); however, the source of S100A9 during autoimmune disease has not been identified. We reasoned that increased serum concentration of S100A9 is indicative of increased S100A9 protein within neutrophils. Since S100A9 levels affect neutrophil bactericidal activity (18), this could contribute to the correlation between autoimmune diseases and S. aureus being a frequent cause of infections in patients with SLE (3841). Herein, we demonstrate that neutrophils from MRL/lpr mice and SLE patients accumulate increased levels of S100A9 protein coinciding with altered mitochondrial homeostasis and reduced O2 production in neutrophils. As a result, suicidal NET formation is decreased in response to S. aureus by murine and human SLE neutrophils, which impairs bactericidal activity and renders the host more susceptible to S. aureus infection.

Materials and Methods

Mice

C57BL/6J were purchased from Jackson Labs (JAX mice stock no. 000664). MRL/MpJ-Tnfrs6lpr/J mice were purchased from Jackson Labs (JAX mice stock no. 000485) or obtained from Dr. B. Vilen. B6.S100a9−/− mice were maintained in an accredited animal facility. Mice (2–5 per cage) were housed in specific pathogen-free conditions and randomly assigned to experimental groups. Food and water were provided ad libitum. All animal experiments were approved and performed in compliance with the Vanderbilt Institutional Animal Care and Use Committee (IACUC).

Reagents

Antibodies specific for Ly6G, CD3, CD19, Ly6C, CD15, CD16, rabbit IgG, and annexin V were from Biolegend; CD11b was from Tonbo; CD63 was from BD Biosciences; myeloperoxidase, histone citrulline, dsDNA, and mouse IgG-alkaline phosphatase were from Abcam; S100A9 (murine and human) was from Cell Signaling. Anti-nucleosome antibody (PL2–3) was a gift from Dr. B. Vilen. Streptavidin Alexa 488 was from Biolegend. Live/dead stain, mitoSOX, mitoTracker, and BacLight® Red Bacterial Stain were from Invitrogen; Helix NP Blue (Sytox) was from Biolegend. Murine Fc-blocking antibody (2.4G2) was from Tonbo; human Fc-blocking antibody was from BD Biosciences. Paraformaldehyde (PFA) was from Electron Microscopy Sciences. Rotenone was from Sigma. Histopaque® 1119 and Histopaque® 1077 were from Sigma. Dulbecco’s Modified Eagle’s Medium (DMEM) and phosphate buffered saline (PBS) was from Gibco and fetal bovine serum (FBS) from Atlanta biologicals were used for all tissue culture.

Bacterial strains

All experiments utilized the S. aureus strain USA300 (LAC) (42). The S. aureus stock was maintained as a −80 °C and 2 days prior to each experiment, was freshly streaked onto tryptic soy agar (TSA; 2% agar) plates each experiment. Single colonies of S. aureus were transferred to tryptic soy broth (TSB), and grown as liquid cultures overnight at 37 °C with 180 rpm shaking. For all in vitro experiments, overnight cultures were diluted into non-heat inactivated FBS at a 1:10 ratio on ice, 2 hr prior to culturing with neutrophils.

Infections

Retroorbital infections were performed as described (18, 43). In brief, S. aureus was streaked onto TSA, two-days prior to infection. Single colonies were transferred from the plate and grown overnight in TSB. On the day of the infection, overnight cultures were sub-cultured 1:100 into 5 mL TSB. S. aureus was grown to mid-exponential phase, washed, and resuspended in 100 μL ice-cold PBS. All infections used 10–13 week-old female mice. Mice were anesthetized by intraperitoneal injection of 2,2,2-tribromoethanol diluted in PBS. Systemic infections were induced by intravenous injection of the retroorbital sinus with 2×107 CFU S. aureus. Mice were monitored prior to humane euthanasia by inhalation of CO2 at 4 dpi.

Organ harvest for CFU and neutrophil characterization

Organs were harvested as previously described (18). Tissue from the hearts, livers, and kidneys were processed through a 35 μm filter into FACS media (PBS, 2% FBS, 0.02% NaAz) to create single cell suspensions. Scalpels were used to cut the heart tissue into smaller pieces to facilitate processing of the tissue through the filter. In addition, special care was taken to flush the heart chambers with PBS to remove any clotted blood and ensure isolated immune cells are from the tissue. Aliquots were taken from each organ homogenate for CFU enumeration by spot plating or to quantify total S100A9 protein levels in the tissue by ELISA. The remaining organ homogenate was pelleted and the supernatant was isolated to quantify NET abundance by ELISA. The remaining pellet was red blood cell lysed per manufacturer’s instructions (Biolegend) and transferred in FACS media into separate aliquots to be stained ex vivo for flow cytometry.

Assessment of autoimmunity in mice

Three days prior to infection murine levels of proteinuria and serum autoantibody were quantified. Urine protein was assessed according to Uristix 4 (Siemens) instruction. Blood was collected by tail nick and serum levels of IgG reactive to nucleosome and dsDNA was quantified by ELISA.

Neutrophil isolation

Neutrophils were isolated as previously described (18). Bone marrow was flushed from the tibias and femurs of mice and single cell suspensions were created. Polymorphonuclear granulocytes were isolated using density centrifugation by isolating the neutrophils at the interface between the layers of Histopaque® 1119 and Histopaque® 1077. Neutrophils were resuspended in D10 media (DMEM, 10% FBS) and rested on ice for at least 1 hr before transferring to ultra-low cluster round bottom 96-well plates (Costar) in D10 media. Neutrophils were incubated (37 °C, 5% CO2) for 1 hr prior to experiments. The isolated cells were 85–95% neutrophils (CD11b+Ly6G+).

Bacterial killing assay

Bacterial killing assays were performed as previously described (18). Neutrophils were cultured with S. aureus at a MOI of 1 (50,000 CFU per 50,000 neutrophils) in D10 media (37 °C, 5% CO2). At each time point (30 min, 2, 6, 12, and 24 hr), samples were serially diluted and spot-plated onto TSA. Plates were cultured overnight (37 °C), and CFU enumerated the next day. Percent growth was quantified by dividing the CFU of the S. aureus-neutrophil co-culture by the S. aureus alone culture.

Confocal imaging

NET imaging was conducted as previously described (18). Neutrophils were stained in with CellMask Deep Red in PBS for 20 min at room temperature and washed in D10 media prior to plating. Glass bottom Petri dishes (MatTek Corporation; Ashland, MA) were charged using a 0.01% poly-L-lysine solution (Sigma Aldrich) and neutrophils were incubated in the dishes for 2 hr (37 °C, 5% CO2) in D10 to allow cells to properly attach to the dish. S. aureus was stained with BacLight® Green for 30 min on ice and added to the dish at an MOI of 1. Helix NP Blue was added 20 min prior to fixation to stain for extracellular DNA (NET structures). Fixation of the neutrophils occurred in 4% paraformaldehyde at room temperature and incubated for 15 min at 4 °C. Dishes were aspirated and samples preserved using Prolong Gold Antifade Mountant. All microscopy was conducted using a Zeiss 880 microscope with AiryScan and Zeiss Zen Software. Images were analyzed using ImageJ.

Flow cytometry

All flow cytometry experiments were conducted as previously described (18). All data were collected using a BD LSRII flow cytometer with FACSDIVA software and analyzed using FlowJo (FlowJo LLC, Ashland, OR). Samples were gated forward scatter height (FSC-H) by forward scatter area (FSC-A) and side scatter height (SSC-H) by side scatter area (SSC-A) to remove doublet populations. The singlet population was gated SSC-A by FSC-A to isolate the granulocyte population. The resulting cell population was then assessed for assay-specific fluorescent markers as described below:

Exogenous Treatments:

Rotenone (0.5 μM) was added 15 min hr prior to stimulation with S. aureus.

Degranulation Flow:

Single cell suspensions isolated from the tissue, bone marrow isolated neutrophils, or peripheral blood PMNs from patients (referred to as cells for the remainder of the flow cytometry section) were stained with a live/dead dye to monitor membrane integrity 20 min prior to fixation. Cells were fixed in 4% room temperature paraformaldehyde and placed on ice for 15 min. Cells were pelleted, aspirated, and resuspended in Fc block in FACS media for 30 min on ice. Cells were washed and stained with antibodies recognizing appropriate neutrophil surface markers (murine: CD11b, Ly6G; human: CD15, CD16), anti-CD63 (primary), and anti-CD35 (secretory) or appropriate isotype controls in FACS media for 30 min on ice. Cells were washed following staining and resuspended in FACS media for analysis. Cells were gated for live cells (Live/Dead-negative) and then neutrophils (CD11b- and Ly6G-positive or CD15- and CD16-positive). The median fluorescence intensity (MFI) was quantified for surface CD63 and CD35 relative to staining with an isotype control antibody.

Phagocytosis and ROS flow:

S. aureus was stained using BacLight® prior to culturing with neutrophils. Cells were live/dead stained as described in ‘Degranulation Flow’ except DHR123 (3 μM) was also introduced to the cultures to quantify total ROS production. Cells were fixed, blocked, and stained for neutrophils (anti-CD11b and -Ly6G) as described in ‘Degranulation Flow’. Following staining, cells were washed, and resuspended in FACs media for analysis. Cells were gated for live cells (Live/Dead-negative) and then neutrophils (CD11b- and Ly6G-positive). The media MFI was quantified for fluorescent S. aureus and DHR123 and normalized to unstimulated neutrophils.

NET Flow:

To quantify the percentage of neutrophils undergoing NET formation, cells were live/dead stained as described above except Helix NP Blue (2.5 nM) was introduced during the live/dead stain. Cells were fixed, blocked, and stained for neutrophils (murine: anti-CD11b and -Ly6G; human: anti-CD15 and -CD16) as described in ‘Degranulation Flow’. During the staining for neutrophils, cells were also stained with anti-H3Cit and -MPO antibodies. Cells were washed and stained with fluorescent streptavidin and anti-rabbit IgG in FACS media for 30 min on ice. Following staining, cells were washed, and resuspended in FACS media for analysis. Cells were gated for neutrophils and then live or dead cells. Neutrophils positive for extracellular dsDNA (Helix NP), MPO, and H3Cit were defined as having undergone NET formation. If the membrane of the cell undergoing NET formation was intact then the cell was defined as having undergone vital NET formation, and if the membrane of the cell undergoing NET formation was permeabilized then the cell was defined as having undergone suicidal NET formation. Representative gating is provided in Supplemental Fig. 2B. The percentage of neutrophils undergoing vital or suicidal NET formation was quantified relative to the total number of neutrophils.

Apoptosis flow:

Cells were live/dead stained, fixed, and blocked, as described in ‘Degranulation Flow’. Cells were washed and stained and stained for neutrophils (anti-CD11b and -Ly6G) as described above except fluorescent staining by annexin V was used to detect apoptotic cells. Following staining, cells were washed, and resuspended in FACs media for analysis. Cells were gated for neutrophils (CD11b- and Ly6G-positive) and the percentage of cells in early (Live/Dead-negative, annexin V-positive) and late (Live/Dead-posative, annexin V-positive) apoptosis was quantified.

Mitochondrial O2 Flow:

To quantify mitochondrial O2 production, cells were live/dead stained as described in ‘Degranulation Flow’ except mitoSOX red (5 μM) and mitoTracker deep red (500 nM) were introduced during the live/dead stain. Cells were fixed, blocked, and stained for neutrophils (murine: anti-CD11b and -Ly6G; human: anti-CD15 and -CD16) as described in ‘Degranulation Flow’. Following staining, cells were washed and resuspended in FACS media for analysis. Cells were gated for live cells and then neutrophils. mitoSOX and mitoTracker MFI were normalized to untreated or mock WT MFI.

A9 Protein Flow:

To quantify the abundance of intracellular S100A9, cells were live/dead stained, fixed, blocked, and stained for neutrophils (murine: anti-CD11b and -Ly6G; human: anti-CD15 and -CD16) as described in ‘Degranulation Flow’. For peripheral blood samples, T cells (CD3+), B cells (CD19+), monocytes (Ly6C+), were also stained. Following staining, cells were washed and stained for S100A9 or an isotype control in permeabilization buffer (PBS, 0.05% saponin, 0.5% BSA) for 30 min on ice. To detect S100A9, cells were washed and stained with fluorescently labeled anti-rabbit IgG for in permeabilization buffer for 30 min on ice. Following staining, cells were washed and resuspended in FACS media for analysis. The MFI was quantified for surface S100A9 relative to staining with an isotype control antibody.

Mitochondrial and Glycolytic Stress Test - Seahorse

Seahorse plates were coated in Cel-Tak (Corning) for 20 min, aspirated, washed using sterile water, and aspirated prior to the addition of neutrophils. Neutrophils were resuspended in either mitochondria (Seahorse Agilent pH 7.4 media, 1 mM pyruvate, 10 mM glucose, 2 mM glutamine) or glycolysis (Seahorse Agilent pH 7.4 media, 2 mM glutamine) assay media and 2×105 neutrophils were added to each well. No neutrophils were added to the four wells in the corners of the plate as a background control. S. aureus was diluted (2×104CFU/μL) in serum and heat-killed for 10 min at 80 °C. For the mitochondrial stress test, the cartridge was sequentially loaded with heat-killed S. aureus (MOI=1), oligomycin (15 μM), FCCP (15 μM), and rotenone/antimycin A (5 μM) (diluted in mitochondria assay media) for injection into the culture. For the glycolysis stress test, the cartridge was sequentially loaded with heat-killed S. aureus (MOI=1), glucose (100 mM), oligomycin (15 μM), and 2-deoxy-d-glucose (500 mM) (diluted in glycolysis assay media) for injection into the culture. To normalize across wells by cell density, the Seahorse plate was imaged using a BioTek Cytation 5. Oxygen consumption and extracellular acidification rates were collected using a Seahorse XFe96 Analyzer and analyzed using Wave Desktop software (Agilent Technologies).

ELISA

Serum autoantibodies:

Vinyl 96-well plates were charged using 0.01% poly-L-lysine solution (Sigma Aldrich), and the plate was coated with dsDNA (Sigma Aldrich) or histones (Immunovision) and dsDNA. Total IgG reactive to nucleosome with histones (4 mg/well; ImmunoVision) and dsDNA (1 mg/well; Sigma) was captured to charged vinyl plates. Captured antibodies were detected with anti-mouse IgG-alkaline phosphatase and p-nitrophenyl phosphate (Sigma). A standard curve was generated using anti-dsDNA (Abcam) and anti-nucleosome (PL2–3) antibodies. Absorbance at 405 nm for each well was quantified using a BioTek Cytation 5. Samples and standards were read in duplicate and DNA abundance was quantified relative to the standard.

NET quantification:

For the NET ELISA a similar procedure was followed as described above. Vinyl 96-well plates were charged and for samples, wells were coated with anti-neutrophil elastase or -MPO antibody. For the DNA standard, dsDNA was titrated using 2-fold dilutions starting at 10 mg/mL. DNA in the wells was detected using an anti-dsDNA antibody (mouse IgG) followed by a secondary anti-mouse antibody conjugated with alkaline phosphatase and 1 mg/mL p-nitrophenyl phosphate. Absorbance at 405 nm for each well was quantified using a BioTek Cytation 5. Samples and standards were read in duplicate and DNA abundance was quantified relative to the standard.

S100A9 quantification:

Organ homogenates were lysed (PBS, 1% Igepal, protease inhibitors) and pelleted to remove cellular debris. The resulting cell lysates or serum from the blood were diluted 1:100 and S100A9 protein levels were quantified per manufacturer’s instructions (Abcam, #ab213887).

Human samples

SLE patients and healthy controls were enrolled following informed consent as approved by the Vanderbilt University Medical Center institutional review board. HC were matched to SLE patients as close as possible by age and gender. HC donors reported no history of autoimmune disease and had not been sick within 2 weeks of donating blood. Disease activity was measured by the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI), and patients with a score ≥ 6 were defined as active and scores < 6 as inactive. Paired healthy control and SLE peripheral blood samples were collected with sodium heparin (BD Biosciences) and PMNs were isolated using density centrifugation at the interface between the layers of Histopaque® 1119 and Histopaque® 1077.

Statistics

Specific statistical details for each experiment are located in the corresponding figure legend. Error bars for experiments represent variation of the mean across mice. A minimum of three experimental replicates were performed for each assay and the specific number of replicates is noted in the corresponding figure legend. All P values were calculated using a one- or two-way ANOVA (Sidak’s multiple comparisons test or Tukey multiple comparisons test), unpaired t-test, or log-rank (Mantel-Cox) test when applicable. Statistical work was performed using Prism 6 software (GraphPad) and significance is indicated on the graphs as follows: *P≤0.05, **P≤0.01, ***P≤0.001, ****P≤0.0001, ns=not significant.

Results

MRL/lpr mice are more susceptible to systemic S. aureus infections

To identify whether altered neutrophil function contributes to the heightened susceptibility of SLE patients to S. aureus infections (3841), MRL/MpJ-Tnfrs6lpr/J (MRL/lpr) mice were employed as they spontaneously generate systemic autoimmunity and are commonly used as a murine model of SLE (44). Therefore, we systemically infected 10–13-week-old female WT and MRL/lpr mice with S. aureus and monitored disease progression over four days. For all infections, MRL/lpr mice with elevated autoantibody titers and mild glomerulonephritis (proteinuria score ≤ 2) were used (Supplemental Fig. 1AB). Over the four-day infection, MRL/lpr mice were more susceptible to systemic S. aureus infection compared to age-matched WT mice (Fig. 1A), despite comparable weight loss during infection (Supplemental Fig. 1C). Coinciding with decreased survival, MRL/lpr mice had increased bacterial burdens in the heart compared to WT (Fig. 1B) four-days post infection (dpi); however, bacterial burdens in the liver and kidney were comparable. These results demonstrate that MRL/lpr mice are more susceptible to systemic S. aureus infections.

FIGURE 1.

FIGURE 1.

MRL/lpr mice are more susceptible to S. aureus infections with increased bacterial burdens in the heart. (A) During the infection, mouse survival was monitored. Each point represents the percentage of living mice (3 infections; mock n=14, WT; inf. n=19, MRL/lpr; inf. n=21). (B) At 4 dpi, organs were homogenized and (B) colony forming units (CFU) enumerated using spot plating. Each point represents a single mouse (3 infections; mock n=6, WT; inf. n=14, MRL/lpr; inf. n=9). (A) Log-rank (Mantel-Cox) test and (B) one-way analysis of variance (ANOVA) with Tukey multiple comparisons test (*P≤0.05, ****P≤0.0001, ns=not significant).

Increased levels of S100A9 in the serum correlate with SLE disease activity (34, 36, 37); therefore, we reasoned that S100A9 protein levels are elevated in SLE neutrophils. Consistent with what is observed in SLE patients, serum levels of S100A9 are elevated in MRL/lpr mice (Fig. 2A). Coinciding with increased serum S100A9 levels, S100A9 protein abundance within MRL/lpr neutrophils in the blood was higher than WT neutrophils (Fig. 2B, Supplemental Fig. 1D). Intracellular S100A9 levels were not detectable by flow cytometry in other immune cell populations (Fig. 2B, Supplemental Fig. 1D). These results demonstrate that increased serum S100A9 corresponds with MRL/lpr neutrophils accumulating and releasing higher levels of S100A9 protein.

FIGURE 2.

FIGURE 2.

Intracellular S100A9 protein is elevated in MRL/lpr neutrophils. Mice were systemically infected (inf.) with S. aureus (CFU=2×107). 3 days prior to infection, (A) S100A9 levels were quantified from the serum of the mice by ELISA and (B) intracellular S100A9 protein levels were quantified in immune cells isolated from the blood by flow cytometry. (B) Median fluorescence intensity (MFI) normalized by isotype control antibody. Each point represents (A) the mean result (technical duplicate) of serum isolated from a single mouse or (B) immune cells isolated from a single mouse (1 infection, n=5). At 4 dpi, organs were homogenized and (C) intracellular abundance of S100A9 in neutrophils (Ly6G+CD11b+) was quantified by flow cytometry and (D) total S100A9 protein in cell lysates from the tissue was quantified by ELISA. (C) MFI normalized by isotype control antibody. Each point represents (C) a single mouse (3 infections; mock n=6, WT; inf. n=14, MRL/lpr; inf. n=9) or (D) the mean result (technical duplicate) of cell lysate isolated from tissue from a single mouse (1 infection; n=5). (A-B, D) Unpaired t-test or (C) one-way ANOVA with Tukey multiple comparisons test (**P≤0.01, ***P≤0.001, ****P≤0.0001, ns=not significant).

The abundance of intracellular S100A9 influences neutrophil function (18). Therefore, mice were systemically infected with S. aureus and intracellular S100A9 levels within neutrophils were quantified by flow cytometry. Neutrophils in the heart of mock and infected MRL/lpr mice had increased intracellular S100A9 protein levels relative to WT (Fig. 2C, Supplemental Fig. 1E), which coincided with a 0.8-log10 increase S. aureus burdens (Fig. 1B). Neutrophils in the liver and kidney, organs that had no change in bacterial burdens, had comparable intracellular S100A9 protein levels in WT and MRL/lpr mice (Fig. 2C). In confirmation, the concentration of S100A9 was quantified in the cell lysate acquired from organ homogenates by enzyme-linked immunosorbent assay (ELISA). S100A9 protein levels were higher in cell lysates obtained from the hearts of MRL/lpr mice compared to WT (Fig. 2D), which is consistent with MRL/lpr neutrophils retaining higher levels of intracellular S100A9 in the heart. Levels of S100A9 protein in cell lysates from the liver and kidney of WT and MRL/lpr mice were comparable (Fig. 2D). Thus, the accumulation of intracellular S100A9 may negatively impact neutrophil function, contributing to the increased bacterial burdens in the heart and heightened susceptibility of MRL/lpr mice to S. aureus infections.

MRL/lpr neutrophils produce lower levels of mitochondrial O2 in response to S. aureus

Dysregulation of mitochondrial homeostasis in immune cells has been linked to SLE pathology (4549), but whether similar defects are observed in SLE neutrophils responding to bacterial pathogens has not been studied in detail. To assess mitochondrial function, neutrophils were isolated from the bone marrow by density centrifugation. Bone marrow neutrophils from MRL/lpr mice showed an accumulated of intracellular S100A9 protein compared to WT by flow cytometry (Fig. 3A, Supplemental Fig. 2A). Since intracellular levels of S100A9 affects mitochondrial O2 production and downstream neutrophil function (18), mitochondrial respiration was monitored in neutrophils by quantifying the oxygen consumption rate (OCR) using extracellular flux analysis (Fig. 3B). Heat-killed S. aureus was used for these experiments to avoid artifacts associated with oxygen consumption by live bacteria. Proton leak has been described as a mechanism to compensate for the electrons that slip from the electron transport chain and contributes to mitochondrial O2 production (5052). Unstimulated MRL/lpr neutrophils exhibited an 8.2-fold higher proton leak when compared to WT, but this phenotype was lost following stimulation with S. aureus (Fig. 3C). Basal respiration of MRL/lpr neutrophils was significantly higher than WT neutrophils in the absence of heat-killed S. aureus, and a similar trend was observed following stimulation with heat-killed S. aureus (Fig. 3C). In addition, maximal respiration and ATP production were elevated in MRL/lpr neutrophils, although they also did not reach statistical significance. Instead, non-mitochondrial oxygen consumption was elevated in MRL/lpr neutrophils when stimulated with heat-killed S. aureus (Fig. 3C), indicative of activity by peroxisomes and/or non-mitochondrial NADPH oxidases (53). Finally, glycolysis and glycolytic capacity were comparable between WT and MRL/lpr neutrophils, as measured by extracellular acidification rates (ECAR; Supplemental Fig. 2BC). These results suggest that while electron transfer is less efficient in MRL/lpr neutrophils, stimulation with heat-killed S. aureus increases the efficiency of electron transfer and simultaneously diverts oxygen consumption to non-mitochondrial processes.

FIGURE 3.

FIGURE 3.

MRL/lpr neutrophils have altered mitochondrial homeostasis with reduced mitochondrial O2 production in response to S. aureus. Neutrophils were isolated from the bone marrow, (A) the intracellular abundance of S100A9 in neutrophils (Ly6G+CD11b+) was quantified by flow cytometry. Median fluorescent intensity (MFI) normalized by isotype. Each point represents neutrophils isolated from a single mouse (2 experiments, n=4). (B-C) Isolated neutrophils were stimulated with heat-killed S. aureus (HK-SA; MOI=1), and the oxygen consumption rate (OCR) was quantified by Seahorse analysis. A mitochondrial stress test was performed using the sequential addition of oligomycin (oligo.; 15 μM), FCCP (15 μM), and rotenone/antimycin A (Rot./Ant.; 5 μM). OCR normalized by cell number. Each point represents (B) the mean of all mean results of neutrophils isolated from a single mouse ±standard error or (C) the mean result of neutrophils isolated from a single mouse (5 experiments; n=5). (D) Neutrophils pre-treated with rotenone (Rot.; 0.5 μM) for 15 min were cultured with S. aureus (MOI=10) and mitochondrial O2 production was quantified by flow cytometry. MitoSOX MFI normalized to MitoTracker MFI. Each point represents neutrophils isolated from a single mouse (2 experiments; n=4). (E) Mice were systemically infected (inf.) with S. aureus (CFU=2×107) and at 4 dpi, organs were homogenized and mitochondrial O2 production by neutrophils was quantified by flow cytometry. MitoSOX MFI normalized to MitoTracker MFI. Each point represents a single mouse (3 infections; mock n=6, WT; inf. n=14, MRL/lpr; inf. n=9). (A, C, E) One-way or (D) two-way ANOVA or with Tukey multiple comparisons test (*P≤0.05, ***P≤0.001, ****P≤0.0001, ns=not significant).

A9−/− neutrophils produce higher levels of mitochondrial O2 in response to S. aureus (18); therefore, we hypothesized that the accumulation of intracellular S100A9 in MRL/lpr neutrophils may impair mitochondrial O2 production. To quantify mitochondrial O2 generation, neutrophils were stained with mitoSOX, a fluorescent dye that localizes to mitochondrial structures and fluoresces upon oxidation by O2. Consistent with increased proton leak (Fig. 3C), unstimulated MRL/lpr neutrophils produced higher levels of mitochondrial O2 compared to WT (Fig. 3D). However, MRL/lpr neutrophils produced lower levels of mitochondrial O2 60 min post inoculation compared to WT neutrophils (Fig. 3D, Supplemental Fig. 2D). This finding is consistent with the reduced proton leak observed in MRL/lpr neutrophils following stimulation with heat-killed S. aureus (Fig. 3C). Treating MRL/lpr neutrophils with rotenone, an inhibitor of complex I of the electron transport chain, was sufficient to increasing mitochondrial O2 production to levels comparable to WT (Fig. 3D, Supplemental Fig. 2D). These results demonstrate that MRL/lpr neutrophils produce lower levels of mitochondrial O2 in response to S. aureus, which coincides with the accumulation of intracellular S100A9 in MRL/lpr neutrophils disrupting mitochondrial homeostasis.

To determine whether the lower production of mitochondrial O2 observed in vitro was present in MRL/lpr mice, mice were infected and the production of mitochondrial O2 by neutrophils was quantified by flow cytometry 4 dpi. Consistent with increased S100A9 protein, MRL/lpr neutrophils in the heart produced 2.4-fold less mitochondrial O2 during infection than WT (Fig. 3E, Supplemental Fig. 2E). Mitochondrial O2 production by neutrophils in the kidney and liver were comparable (Fig. 3E, Supplemental Fig. 2E). These results suggest that accumulation of S100A9 coinciding with lower mitochondrial O2 production, impacts neutrophil antibacterial activity in the heart and rendering MRL/lpr mice more susceptible to infection.

Decreased mitochondrial O2 production by MRL/lpr neutrophils impairs suicidal NET formation

Neutrophils utilize multiple antimicrobial processes when engaging S. aureus. Upon engaging S. aureus, neutrophils couple phagocytosis with the respiratory burst (5456). Phagocytosis of fluorescently labeled S. aureus by WT and MRL/lpr neutrophils was comparable (Supplemental Fig. 2F). In contrast to what was observed with MRL/lpr macrophages (57), MRL/lpr neutrophils produce lower levels of ROS compared to WT 120 min post inoculation (Supplemental Fig. 2G). However, this phenotype was not present in vivo as neutrophils isolated from the heart and kidney of infected WT and MRL/lpr mice produced comparable levels of ROS, while MRL/lpr neutrophils constitutively produced higher levels of ROS in the liver independent of infection (Supplemental Fig. 2H). These results suggest that impaired ROS production does not account for the increased susceptibility of MRL/lpr mice during infection.

Since the production of mitochondrial O2 promotes suicidal NET formation in response to S. aureus (18), neutrophils were stimulated with S. aureus and cultures were imaged for NET structures after 240 min. NET structures were readily apparent in cultures with WT neutrophils, but NET structures were smaller and less frequent in cultures with MRL/lpr neutrophils (Fig. 4A). To quantify NET formation, we used a flow cytometry-based technique (Supplemental Fig. 3A) that has been previously described in detail (18). In response to S. aureus, MRL/lpr neutrophils showed impaired suicidal NET formation (3.8-fold; Fig. 4B, Supplemental Fig. 3B) and sustained vital NET formation (6.2-fold; Fig. 4C, Supplemental Fig. 3C) compared to WT neutrophils 240 min post inoculation. While the abundance of intracellular S100A9 and mitochondrial O2 production influences release of primary granules (18), primary degranulation was comparable between WT and MRL/lpr neutrophils (Supplemental Fig. 3D). In addition, alternative cell death pathways were similar between WT and MRL/lpr neutrophils except at 240 min post inoculation where a higher percentage of WT neutrophils displayed signs of late apoptosis compared to MRL/lpr (Supplemental Fig. 3E). These results demonstrate that MRL/lpr neutrophils have sustained vital NET formation but impaired suicidal NET formation in response to S. aureus.

FIGURE 4.

FIGURE 4.

Decreased mitochondrial O2 production prevents suicidal NET formation in MRL/lpr neutrophils. (A) Neutrophils were cultured with S. aureus (MOI=10) and representative images of neutrophils (red) stimulated for 4 hr with a nuclease-deficient strain of S. aureus (green) are provided. Extracellular DNA was stained using Helix NP Blue. (B-C) Neutrophils pre-treated with rotenone (Rot.; 0.5 μM) for 15 min were cultured with S. aureus (MOI=10) and the percentage of neutrophils undergoing (B) suicidal (dead: extracellular dsDNA+MPO+H3Cit+) and (C) vital (live: extracellular dsDNA+MPO+H3Cit+) NET formation in response to S. aureus was quantified by flow cytometry. Each point represents neutrophils isolated from a single mouse (2 experiments; n=4). (D) Neutrophils were cultured with S. aureus (SA; MOI=1) and restriction of S. aureus growth after 6 hr was quantified by colony forming unit (CFU) spot plating. Percent growth of S. aureus calculated relative to S. aureus growth in the absence of neutrophils. Each point represents the mean result (technical triplicate) of neutrophils isolated from a single mouse (2 experiments; n=3). (E-F) Mice were systemically infected (inf.) with S. aureus (CFU=2×107) and at 4 dpi, organs were homogenized and (E) the percentage of neutrophils undergoing suicidal NET formation was quantified by flow cytometry. Each point represents a single mouse (3 infections; mock n=6, WT; inf. n=14, MRL/lpr; inf. n=9). (F) Supernatant from homogenized organs were used to quantify NET abundance in the tissues by ELISA. Capture antibodies directed towards myeloperoxidase (MPO) or neutrophil elastase (NE). NET DNA abundance was quantified using a DNA standard and normalized to the mass of the tissue. Each point represents the mean result (technical duplicate) from supernatants isolated from a single mouse (1 infection; n=5). Two-way ANOVA with (B-C) Tukey or (D) Sidak’s multiple comparisons test, (E) one-way ANOVA or with Tukey multiple comparison test, or (F) unpaired t-test (*P≤0.05, **P≤0.01, ***P≤0.001, ****P≤0.0001, ns=not significant).

To identify whether impaired production of mitochondrial O2 in MRL/lpr neutrophils prevents suicidal NET formation, neutrophils were treated with rotenone to increase mitochondrial O2 production (Fig. 3D, Supplemental Fig. 2D). In addition to increasing mitochondrial O2 levels, rotenone accelerated and enhanced suicidal NET formation such that rotenone-treated MRL/lpr neutrophils were comparable to WT (Fig. 4B, Supplemental Fig. 3B). Coinciding with increasing suicidal NET formation at 240 min post inoculation, the percentage of MRL/lpr neutrophils undergoing vital NET formation was decreased to levels comparable to WT (Fig. 4C, Supplemental Fig. 3C). In addition, rotenone treatment decreased primary degranulation in both WT and MRL/lpr neutrophils (Supplemental Fig. 3D). These results demonstrate that impaired production of mitochondrial O2 by MRL/lpr neutrophils prevents suicidal NET formation, while sustaining vital NET formation long term.

Impairing suicidal NET formation decreases bactericidal activity of neutrophils in response to S. aureus (18, 27, 28). To determine whether the impaired suicidal NET formation observed in MRL/lpr neutrophils similarly restricts bactericidal activity, neutrophils were cultured with S. aureus and bacterial killing was quantified by dilution spot plating. Coinciding with altered NET formation, MRL/lpr neutrophils failed to restrict S. aureus growth as well as WT neutrophils at 2 and 6 hr post inoculation (Fig. 4D). These results demonstrate that MRL/lpr neutrophils have a significant loss in bactericidal activity.

To determine whether the impaired suicidal NET formation observed in vitro was present in MRL/lpr mice, mice were infected and the percentage of neutrophils undergoing NET formation was quantified by flow cytometry 4 dpi. Coinciding with higher bacterial burdens (Fig. 1B), suicidal NET formation was diminished 2.3-fold in the heart of MRL/lpr mice but not significantly different in the liver and kidney relative to WT (Fig. 4E, Supplemental Fig. S3F). Consistent with a lower percentage of neutrophils undergoing suicidal NET formation in the heart, a decreased abundance of NETs was quantified from the supernatant of heart tissue from MRL/lpr mice compared to WT by ELISA (Fig. 4F). Finally, vital NET formation and primary degranulation were comparable in the heart and liver, but decreased in the kidney of A9−/− mice compared to WT (Supplemental Fig. 3GH). These data demonstrate that the decrease in suicidal NET formation in the heart during SLE may provide a favorable environment for S. aureus and contribute to the increased bacterial burdens and enhanced mortality during S. aureus infections.

Altered neutrophil activity in patients with SLE

MRL/lpr mice are genetically identical and subjected to a controlled environment; however, multiple genetic and environmental factors contribute to SLE in patients. Therefore, we sought to determine how pervasive the impaired mitochondrial O2 and NET formation phenotypes are in patients with SLE. Polymorphonuclear (PMN) cells were isolated from the peripheral blood of healthy control (HC) and SLE patients (Table 1) and co-cultured with S. aureus. Overall, the level of intracellular S100A9 protein was 1.7-fold higher in neutrophils from patients with SLE compared to HC (Fig. 5A, Supplemental Fig. 4A). Similar to MRL/lpr mice, neutrophils from patients with SLE produced less mitochondrial O2 relative to HC following co-culture with S. aureus (Fig. 5B, Supplemental Fig. 4B). Furthermore, after a 240 min culture with S. aureus, samples from patients with SLE showed a 5.5-fold increase in the percentage of neutrophils undergoing vital NET formation (Fig. 5C, Supplemental Fig. 4C) and a 3.3-fold decrease in suicidal NET formation (Fig. 5D, Supplemental Fig. 4D). Primary degranulation was comparable between SLE and HC neutrophils (Supplemental Fig. 4E). Consistent with impaired suicidal NET formation, PMNs from patients with SLE failed to restrict S. aureus growth to the same levels as PMNs from HC (Fig. 5E), suggesting SLE PMNs have impaired bactericidal activity. Disease activity, as measured by SLE disease activity index (SLEDAI; 58), and patient treatments did not account for the altered response of neutrophils from patients with SLE to S. aureus (Table 1, Fig. 5). These results demonstrate that the diminished mitochondrial O2, suicidal NET formation, and antimicrobial activity identified in neutrophils from MRL/lpr mice and patients with SLE, regardless of disease activity. Thus, accumulation of intracellular S100A9 coinciding with impairing suicidal NET formation may contribute to the increased incidence of S. aureus infections in patients with SLE.

TABLE 1.

Distribution of patient samples.

Healthy (n=12) All SLE (n=12) Active SLE (SLEDAI≥6) (n=7) Inactive SLE (SLEDAI<6) (n=5)
Age
41.7 (24–64) 46.8 (34–57) 50.0 (45–57) 44.9 (34–52)
Sex
 Female 12 12 7 5
 Male 0 0 0 0
Race
 White 8 7 4 3
 Black 4 5 3 2
Ethnicity
 hispanic 1 1 1 0
 nonhispanic 11 11 6 5
Medications
 PDS - 4 4 0
 HCQ - 9 7 2
 cDMARD* - 8 5 3
 bDMARD - 1 1 0
 NSAIDs - 3 1 2

cDMARD = methtrexate, leflunamide, azathioprine,mycophenolate mofetil, tacrolimus

bDMARD= adalimumab

NSAIDs = aspirin, diclofenac, nabumetone

FIGURE 5.

FIGURE 5.

Increased S100A9 in neutrophils from patients with SLE correlates with decreased mitochondrial O2 and impaired suicidal NET formation. PMNs were isolated from human peripheral blood of healthy control (HC) and SLE patients, and (A) the intracellular abundance of S100A9 was quantified by flow cytometry. MFI normalized by isotype. Each point represents PMNs isolated from a single donor (12 experiments; n=12). (B-D) PMNs were cultured with S. aureus (MOI=10) and (B) mitochondrial O2 production in PMNs (CD15+CD16+) and the percentage of PMNs undergoing (C) vital (Live: extracellular dsDNA+MPO+H3Cit+) and (D) suicidal (Dead: extracellular dsDNA+MPO+H3Cit+) NET formation was quantified by flow cytometry. (B) mitoSOX MFI normalized by mitoTracker MFI. Each point represents PMNs isolated from a single donor (12 experiments; n=12). (E) PMNs were cultured with S. aureus (SA; MOI=1) and restriction of S. aureus growth after 6 hr was quantified by CFU spot plating. Percent growth of S. aureus calculated relative to S. aureus growth in the absence of PMNs. Each point represents the mean result (technical triplicate) of PMNs isolated from a single donor (12 experiments; n=12). ▲=Active disease (SLEDAI≥6), ▽=Inactive disease (SLEDAI<6). (A) Unpaired t-test or (B-E) two-way ANOVA with Sidak’s multiple comparisons test (*P≤0.05, **P0.01, ****P≤0.0001, ns=not significant).

Discussion

We report that neutrophils from MRL/lpr mice and SLE patients accumulate higher levels of intracellular S100A9 protein and produce lower levels of mitochondrial O2 in response to S. aureus. As a result of decreased production of mitochondrial O2, MRL/lpr neutrophils have impaired suicidal NET formation but heightened vital NET formation in response to S. aureus. In mice, the decrease in suicidal NET formation correlates with a specific increase in bacterial burdens in the heart, and decreased survival during systemic S. aureus infection. Consistent with prior observations (18), these findings demonstrate that elevated intracellular concentrations of S100A9 renders neutrophils less bactericidal towards S. aureus and may contribute to the increased susceptibility of SLE patients to bacterial infection.

Mitochondrial dysfunction has been associated with SLE pathology (4549), and occurs in a mammalian target of rapamycin (mTOR)-dependent manner prior to overt immune dysregulation and tissue damage in MRL/lpr mice (59). We observe a comparable phenotype whereby MRL/lpr neutrophils produce higher levels of mitochondrial O2 (Fig. 2A) with increased proton leak (Fig. 1C) prior to stimulation with S. aureus. However, S. aureus stimulation results in a decrease in proton leak coinciding with increased oxygen consumption by non-mitochondrial processes (Fig. 1C), most likely reflecting increased NADPH oxidase activity. Similarly, MRL/lpr macrophages maintain a prolonged oxidative burst following phagocytosis of immunoglobulin G (IgG)-containing immune complexes thereby disrupting lysosome degradation in an mTOR-dependent manner (57, 60). As a result, MRL/lpr macrophages accumulate high levels of nuclear antigen on the cell surface that precedes and promotes many lupus-associated pathologies (57, 61, 62). While the accumulation of nuclear antigen has been observed on B cells, T cells, monocytes, dendritic cells, and macrophages (62), it is unclear whether accumulation occurs on neutrophils. Taken together, the accumulation of IgG-containing immune complexes that dysregulate macrophages during lupus may play a similar role in driving mitochondrial dysfunction and the elevated NET formation observed during SLE pathology.

The incidence of S. aureus infections in patients with SLE (3841) may be partially attributed to immunosuppressants; however, MRL/lpr mice are susceptible to S. aureus infections independent of immunosuppressants (Fig. 3). Conversely, S. aureus can exacerbate SLE pathologies. Reflecting the importance of immune activation driven by S. aureus, chronic exposure to superantigens from S. aureus in WT mice results in a disease mimicking lupus (63), whereby type-1 interferon production associated with S. aureus infection and SLE antagonizes cutaneous lupus erythematosus lesions and facilitates colonization of the lesion by S. aureus (64). Our data demonstrate that decreased mitochondrial O2 and suicidal NET formation by neutrophils from MRL/lpr mice and patients with SLE could render neutrophils unsuitable to combat S. aureus (Fig. 24). As a result, this may create a feed-forward mechanism whereby autoimmunity renders the host more susceptible to infection, while increased S. aureus colonization promotes heightened SLE disease activity.

The observation that neutrophils from MRL/lpr mice and SLE patients undergo less suicidal NET formation in response to S. aureus was initially surprising. Whether altered NET formation contributes to SLE pathology by providing a source of nuclear antigen has been debated (31, 32, 65, 66); however, it has been consistently observed that murine and human SLE neutrophils undergoing higher levels of NET formation during SLE pathology (4549, 59). Furthermore, increased NET deposition in the kidney contributes to the inflammatory response and type I interferon production associated with kidney nephritis (32, 67). In contrast, neutrophils from MRL/lpr mice and SLE patients exhibit impaired suicidal NET formation in response to S. aureus (Fig. 4AB, 5D) with reduced NET formation in heart (Fig. 4EF). NET formation in the kidney was comparable between WT and MRL/lpr mice, despite the MRL/lpr mice having elevated autoantibody titers and mild glomerulonephritis (Supplemental Fig. 1AB). These results suggest that the signal transduction associated with autoantibodies triggering NET formation during SLE (68) may differ or conflict with pathogen-mediated NET formation. Therefore, while the kidney provides a unique metabolic/immunologic niche that promotes NET formation during SLE, these same autoimmune signals may impair suicidal NET formation in the heart during infection. In addition, we have determined that murine and human SLE neutrophils sustain heightened vital NET formation and fail to undergo suicidal NET formation (Fig 4BC, 5CD). The molecular mechanism regulating vital NET formation is unclear and may account for discrepant reports regarding the role for NET formation in SLE (31, 32, 65, 66). Identifying whether vital and suicidal NETs are immunogenically distinct is necessary to unravel the complexities of NET formation in SLE.

Supplementary Material

1

Key points.

  • Altered mitochondrial homeostasis in SLE neutrophils impairs superoxide production.

  • NET formation by SLE neutrophils is decreased in response to S. aureus.

  • SLE neutrophils are less bactericidal towards S. aureus.

Acknowledgments

We thank Drs. Barbara Vilen and Sunah Kang for the MRL/lpr mice and PL2-3 antibody, and the patients and support staff at Vanderbilt University Medical Center (VUMC) who provided patient samples. We also thank all the members of the Skaar lab who provided feedback on the project and paper.

Funding

Work in E.P.S.’s lab was supported by R01 AI069233, R01 AI073843, R01 AI101171, a grant from the Edward P. Evans Foundation, and funds from Incyte Pharmaceuticals. J.C.R. was supported by R01 DK105550, R01 AI153167, and the William Paul Distinguished Innovator Award of the Lupus Research Alliance. A.J.M. was supported by the Ruth L. Kirschstein National Research Service Award (NRSA) Individual Postdoctoral Fellowship F32HL144081. K.V. was supported by the Integrated Training in Engineering and Diabetes (ITED) grant T32 DK101003. Flow Cytometry experiments were performed in the VUMC Flow Cytometry Shared Resource, which was supported by the Vanderbilt Ingram Cancer Center (P30 CA68485) and the Vanderbilt Digestive Disease Research Center (DK058404).

Declaration of interests

All materials are available and all data are present in the main text or in the supplementary materials. J.C.R. is a founder, scientific advisory board member, and stockholder of Sitryx Therapeutics, a scientific advisory board member and stockholder of Caribou Biosciences, a member of the scientific advisory board of Nirogy Therapeutics, has consulted for Merck, Pfizer, and Mitobridge within the past three years, and has received research support from Incyte Corp., Calithera Biosciences, and Tempest Therapeutics.

References and Notes:

  • 1.Popovich KJ, Weinstein RA, and Hota B. 2008. Are community-associated methicillin-resistant Staphylococcus aureus (MRSA) strains replacing traditional nosocomial MRSA strains? Clin Infect Dis 46: 787–794. [DOI] [PubMed] [Google Scholar]
  • 2.Talan DA, Krishnadasan A, Gorwitz RJ, Fosheim GE, Limbago B, Albrecht V, Moran GJ, and Group EMINS 2011. Comparison of Staphylococcus aureus from skin and soft-tissue infections in US emergency department patients, 2004 and 2008. Clin Infect Dis 53: 144–149. [DOI] [PubMed] [Google Scholar]
  • 3.Fowler RA, and Gupta S. 2005. Subacute and acute infective endocarditis. Lancet 366: 1964. [DOI] [PubMed] [Google Scholar]
  • 4.Klein M, and Wang A. 2016. Infective Endocarditis. J Intensive Care Med 31: 151–163. [DOI] [PubMed] [Google Scholar]
  • 5.Slipczuk L, Codolosa JN, Davila CD, Romero-Corral A, Yun J, Pressman GS, and Figueredo VM 2013. Infective endocarditis epidemiology over five decades: a systematic review. PLoS One 8: e82665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chastre J, and Fagon JY 2002. Ventilator-associated pneumonia. Am J Respir Crit Care Med 165: 867–903. [DOI] [PubMed] [Google Scholar]
  • 7.Anderson DJ, Moehring RW, Sloane R, Schmader KE, Weber DJ, Fowler VG Jr., Smathers E, and Sexton DJ 2014. Bloodstream infections in community hospitals in the 21st century: a multicenter cohort study. PLoS One 9: e91713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Laupland KB 2013. Defining the epidemiology of bloodstream infections: the ‘gold standard’ of population-based assessment. Epidemiol Infect 141: 2149–2157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Laupland KB, and Church DL 2014. Population-based epidemiology and microbiology of community-onset bloodstream infections. Clin Microbiol Rev 27: 647–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Verdrengh M, and Tarkowski A. 1997. Role of neutrophils in experimental septicemia and septic arthritis induced by Staphylococcus aureus. Infect Immun 65: 2517–2521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Spinardi JR, Berea R, Orioli PA, Gabriele MM, Navarini A, Marques MT, Neto MN, and Mimica MJ 2017. Enterococcus spp. and S. aureus colonization in neutropenic febrile children with cancer. Germs 7: 61–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hunter MJ, and Chazin WJ 1998. High level expression and dimer characterization of the S100 EF-hand proteins, migration inhibitory factor-related proteins 8 and 14. J Biol Chem 273: 12427–12435. [DOI] [PubMed] [Google Scholar]
  • 13.Propper C, Huang X, Roth J, Sorg C, and Nacken W. 1999. Analysis of the MRP8-MRP14 protein-protein interaction by the two-hybrid system suggests a prominent role of the C-terminal domain of S100 proteins in dimer formation. J Biol Chem 274: 183–188. [DOI] [PubMed] [Google Scholar]
  • 14.Edgeworth J, Gorman M, Bennett R, Freemont P, and Hogg N. 1991. Identification of p8,14 as a highly abundant heterodimeric calcium binding protein complex of myeloid cells. J Biol Chem 266: 7706–7713. [PubMed] [Google Scholar]
  • 15.Kehl-Fie TE, Zhang Y, Moore JL, Farrand AJ, Hood MI, Rathi S, Chazin WJ, Caprioli RM, and Skaar EP 2013. MntABC and MntH contribute to systemic Staphylococcus aureus infection by competing with calprotectin for nutrient manganese. Infect Immun 81: 3395–3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Corbin BD, Seeley EH, Raab A, Feldmann J, Miller MR, Torres VJ, Anderson KL, Dattilo BM, Dunman PM, Gerads R, Caprioli RM, Nacken W, Chazin WJ, and Skaar EP 2008. Metal chelation and inhibition of bacterial growth in tissue abscesses. Science 319: 962–965. [DOI] [PubMed] [Google Scholar]
  • 17.Juttukonda LJ, Berends ETM, Zackular JP, Moore JL, Stier MT, Zhang Y, Schmitz JE, Beavers WN, Wijers CD, Gilston BA, Kehl-Fie TE, Atkinson J, Washington MK, Peebles RS, Chazin WJ, Torres VJ, Caprioli RM, and Skaar EP 2017. Dietary Manganese Promotes Staphylococcal Infection of the Heart. Cell Host Microbe 22: 531–542 e538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Monteith AJ, Miller JM, Maxwell CN, Chazin WJ, and Skaar EP 2021. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. Sci Adv 7: eabj2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hood MI, Mortensen BL, Moore JL, Zhang Y, Kehl-Fie TE, Sugitani N, Chazin WJ, Caprioli RM, and Skaar EP 2012. Identification of an Acinetobacter baumannii zinc acquisition system that facilitates resistance to calprotectin-mediated zinc sequestration. PLoS Pathog 8: e1003068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zackular JP, Moore JL, Jordan AT, Juttukonda LJ, Noto MJ, Nicholson MR, Crews JD, Semler MW, Zhang Y, Ware LB, Washington MK, Chazin WJ, Caprioli RM, and Skaar EP 2016. Dietary zinc alters the microbiota and decreases resistance to Clostridium difficile infection. Nat Med 22: 1330–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, Brinkmann V, Jungblut PR, and Zychlinsky A. 2009. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog 5: e1000639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Clark HL, Jhingran A, Sun Y, Vareechon C, de Jesus Carrion S, Skaar EP, Chazin WJ, Calera JA, Hohl TM, and Pearlman E. 2016. Zinc and Manganese Chelation by Neutrophil S100A8/A9 (Calprotectin) Limits Extracellular Aspergillus fumigatus Hyphal Growth and Corneal Infection. J Immunol 196: 336–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gaddy JA, Radin JN, Loh JT, Piazuelo MB, Kehl-Fie TE, Delgado AG, Ilca FT, Peek RM, Cover TL, Chazin WJ, Skaar EP, and Scott Algood HM 2014. The host protein calprotectin modulates the Helicobacter pylori cag type IV secretion system via zinc sequestration. PLoS Pathog 10: e1004450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dow A, Sule P, O’Donnell TJ, Burger A, Mattila JT, Antonio B, Vergara K, Marcantonio E, Adams LG, James N, Williams PG, Cirillo JD, and Prisic S. 2021. Zinc limitation triggers anticipatory adaptations in Mycobacterium tuberculosis. PLoS Pathog 17: e1009570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, and Zychlinsky A. 2004. Neutrophil extracellular traps kill bacteria. Science 303: 1532–1535. [DOI] [PubMed] [Google Scholar]
  • 26.Pilsczek FH, Salina D, Poon KK, Fahey C, Yipp BG, Sibley CD, Robbins SM, Green FH, Surette MG, Sugai M, Bowden MG, Hussain M, Zhang K, and Kubes P. 2010. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol 185: 7413–7425. [DOI] [PubMed] [Google Scholar]
  • 27.Berends ET, Horswill AR, Haste NM, Monestier M, Nizet V, and von Kockritz-Blickwede M. 2010. Nuclease expression by Staphylococcus aureus facilitates escape from neutrophil extracellular traps. J Innate Immun 2: 576–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.D’Cruz AA, Speir M, Bliss-Moreau M, Dietrich S, Wang S, Chen AA, Gavillet M, Al-Obeidi A, Lawlor KE, Vince JE, Kelliher MA, Hakem R, Pasparakis M, Williams DA, Ericsson M, and Croker BA 2018. The pseudokinase MLKL activates PAD4-dependent NET formation in necroptotic neutrophils. Sci Signal 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Harley JB, Kelly JA, and Kaufman KM 2006. Unraveling the genetics of systemic lupus erythematosus. Springer Semin Immunopathol 28: 119–130. [DOI] [PubMed] [Google Scholar]
  • 30.Kamen DL 2014. Environmental influences on systemic lupus erythematosus expression. Rheum Dis Clin North Am 40: 401–412, vii. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, McCune WJ, and Kaplan MJ 2010. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 184: 3284–3297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, Denny MF, Plumas J, Chaperot L, Kretzler M, Bruce AT, and Kaplan MJ 2011. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol 187: 538–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Carlucci PM, Purmalek MM, Dey AK, Temesgen-Oyelakin Y, Sakhardande S, Joshi AA, Lerman JB, Fike A, Davis M, Chung JH, Playford MP, Naqi M, Mistry P, Gutierrez-Cruz G, Dell’Orso S, Naz F, Salahuddin T, Natarajan B, Manna Z, Tsai WL, Gupta S, Grayson P, Teague H, Chen MY, Sun HW, Hasni S, Mehta NN, and Kaplan MJ 2018. Neutrophil subsets and their gene signature associate with vascular inflammation and coronary atherosclerosis in lupus. JCI Insight 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tyden H, Lood C, Gullstrand B, Jonsen A, Nived O, Sturfelt G, Truedsson L, Ivars F, Leanderson T, and Bengtsson AA 2013. Increased serum levels of S100A8/A9 and S100A12 are associated with cardiovascular disease in patients with inactive systemic lupus erythematosus. Rheumatology (Oxford) 52: 2048–2055. [DOI] [PubMed] [Google Scholar]
  • 35.Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA, and Ogryzlo MA 1976. The bimodal mortality pattern of systemic lupus erythematosus. Am J Med 60: 221–225. [DOI] [PubMed] [Google Scholar]
  • 36.Soyfoo MS, Roth J, Vogl T, Pochet R, and Decaux G. 2009. Phagocyte-specific S100A8/A9 protein levels during disease exacerbations and infections in systemic lupus erythematosus. J Rheumatol 36: 2190–2194. [DOI] [PubMed] [Google Scholar]
  • 37.Davies JC, Midgley A, Carlsson E, Donohue S, Bruce IN, Beresford MW, and Hedrich CM 2020. Urine and serum S100A8/A9 and S100A12 associate with active lupus nephritis and may predict response to rituximab treatment. RMD Open 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hellmann DB, Petri M, and Whiting-O’Keefe Q. 1987. Fatal infections in systemic lupus erythematosus: the role of opportunistic organisms. Medicine (Baltimore) 66: 341–348. [DOI] [PubMed] [Google Scholar]
  • 39.Ruiz-Irastorza G, Olivares N, Ruiz-Arruza I, Martinez-Berriotxoa A, Egurbide MV, and Aguirre C. 2009. Predictors of major infections in systemic lupus erythematosus. Arthritis Res Ther 11: R109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chen MJ, Tseng HM, Huang YL, Hsu WN, Yeh KW, Wu TL, See LC, and Huang JL 2008. Long-term outcome and short-term survival of patients with systemic lupus erythematosus after bacteraemia episodes: 6-yr follow-up. Rheumatology (Oxford) 47: 1352–1357. [DOI] [PubMed] [Google Scholar]
  • 41.Conti F, Ceccarelli F, Iaiani G, Perricone C, Giordano A, Amori L, Miranda F, Massaro L, Pacucci VA, Truglia S, Girelli G, Fakeri A, Taliani G, Temperoni C, Spinelli FR, Alessandri C, and Valesini G. 2016. Association between Staphylococcus aureus nasal carriage and disease phenotype in patients affected by systemic lupus erythematosus. Arthritis Res Ther 18: 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tenover FC, McDougal LK, Goering RV, Killgore G, Projan SJ, Patel JB, and Dunman PM 2006. Characterization of a strain of community-associated methicillin-resistant Staphylococcus aureus widely disseminated in the United States. J Clin Microbiol 44: 108–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Klopfenstein N, Cassat JE, Monteith A, Miller A, Drury S, Skaar E, and Serezani CH 2021. Murine Models for Staphylococcal Infection. Curr Protoc 1: e52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Andrews BS, Eisenberg RA, Theofilopoulos AN, Izui S, Wilson CB, McConahey PJ, Murphy ED, Roths JB, and Dixon FJ 1978. Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains. J Exp Med 148: 1198–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Blanco LP, Pedersen HL, Wang X, Lightfoot YL, Seto N, Carmona-Rivera C, Yu ZX, Hoffmann V, Yuen PST, and Kaplan MJ 2020. Improved Mitochondrial Metabolism and Reduced Inflammation Following Attenuation of Murine Lupus With Coenzyme Q10 Analog Idebenone. Arthritis Rheumatol 72: 454–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Buskiewicz IA, Montgomery T, Yasewicz EC, Huber SA, Murphy MP, Hartley RC, Kelly R, Crow MK, Perl A, Budd RC, and Koenig A. 2016. Reactive oxygen species induce virus-independent MAVS oligomerization in systemic lupus erythematosus. Sci Signal 9: ra115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Caza TN, Fernandez DR, Talaber G, Oaks Z, Haas M, Madaio MP, Lai ZW, Miklossy G, Singh RR, Chudakov DM, Malorni W, Middleton F, Banki K, and Perl A. 2014. HRES-1/Rab4-mediated depletion of Drp1 impairs mitochondrial homeostasis and represents a target for treatment in SLE. Ann Rheum Dis 73: 1888–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB, and Kaplan MJ 2016. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med 22: 146–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lopez-Lopez L, Nieves-Plaza M, Castro Mdel R, Font YM, Torres-Ramos CA, Vila LM, and Ayala-Pena S. 2014. Mitochondrial DNA damage is associated with damage accrual and disease duration in patients with systemic lupus erythematosus. Lupus 23: 1133–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ganote CE, and Armstrong SC 2003. Effects of CCCP-induced mitochondrial uncoupling and cyclosporin A on cell volume, cell injury and preconditioning protection of isolated rabbit cardiomyocytes. J Mol Cell Cardiol 35: 749–759. [DOI] [PubMed] [Google Scholar]
  • 51.Speakman JR, Talbot DA, Selman C, Snart S, McLaren JS, Redman P, Krol E, Jackson DM, Johnson MS, and Brand MD 2004. Uncoupled and surviving: individual mice with high metabolism have greater mitochondrial uncoupling and live longer. Aging Cell 3: 87–95. [DOI] [PubMed] [Google Scholar]
  • 52.Turrens JF, and Boveris A. 1980. Generation of superoxide anion by the NADH dehydrogenase of bovine heart mitochondria. Biochem J 191: 421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nordgren M, and Fransen M. 2014. Peroxisomal metabolism and oxidative stress. Biochimie 98: 56–62. [DOI] [PubMed] [Google Scholar]
  • 54.Collins LV, Kristian SA, Weidenmaier C, Faigle M, Van Kessel KP, Van Strijp JA, Gotz F, Neumeister B, and Peschel A. 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]
  • 55.Green JN, Kettle AJ, and Winterbourn CC 2014. Protein chlorination in neutrophil phagosomes and correlation with bacterial killing. Free Radic Biol Med 77: 49–56. [DOI] [PubMed] [Google Scholar]
  • 56.Hampton MB, and Winterbourn CC 1995. Modification of neutrophil oxidant production with diphenyleneiodonium and its effect on bacterial killing. Free Radic Biol Med 18: 633–639. [DOI] [PubMed] [Google Scholar]
  • 57.Monteith AJ, Kang S, Scott E, Hillman K, Rajfur Z, Jacobson K, Costello MJ, and Vilen BJ 2016. Defects in lysosomal maturation facilitate the activation of innate sensors in systemic lupus erythematosus. Proc Natl Acad Sci U S A 113: E2142–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bombardier C, Gladman DD, Urowitz MB, Caron D, and Chang CH 1992. Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum 35: 630–640. [DOI] [PubMed] [Google Scholar]
  • 59.Oaks Z, Winans T, Caza T, Fernandez D, Liu Y, Landas SK, Banki K, and Perl A. 2016. Mitochondrial Dysfunction in the Liver and Antiphospholipid Antibody Production Precede Disease Onset and Respond to Rapamycin in Lupus-Prone Mice. Arthritis Rheumatol 68: 2728–2739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Monteith AJ, Vincent HA, Kang S, Li P, Claiborne TM, Rajfur Z, Jacobson K, Moorman NJ, and Vilen BJ 2018. mTORC2 Activity Disrupts Lysosome Acidification in Systemic Lupus Erythematosus by Impairing Caspase-1 Cleavage of Rab39a. J Immunol 201: 371–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kang S, Fedoriw Y, Brenneman EK, Truong YK, Kikly K, and Vilen BJ 2017. BAFF Induces Tertiary Lymphoid Structures and Positions T Cells within the Glomeruli during Lupus Nephritis. J Immunol 198: 2602–2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kang S, Rogers JL, Monteith AJ, Jiang C, Schmitz J, Clarke SH, Tarrant TK, Truong YK, Diaz M, Fedoriw Y, and Vilen BJ 2016. Apoptotic Debris Accumulates on Hematopoietic Cells and Promotes Disease in Murine and Human Systemic Lupus Erythematosus. J Immunol 196: 4030–4039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chowdhary VR, Tilahun AY, Clark CR, Grande JP, and Rajagopalan G. 2012. Chronic exposure to staphylococcal superantigen elicits a systemic inflammatory disease mimicking lupus. J Immunol 189: 2054–2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sirobhushanam S, Parsa N, Reed TJ, Berthier CC, Sarkar MK, Hile GA, Tsoi LC, Banfield J, Dobry C, Horswill AR, Gudjonsson JE, and Kahlenberg JM 2020. Staphylococcus aureus Colonization Is Increased on Lupus Skin Lesions and Is Promoted by IFN-Mediated Barrier Disruption. J Invest Dermatol 140: 1066–1074 e1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Campbell AM, Kashgarian M, and Shlomchik MJ 2012. NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci Transl Med 4: 157ra141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gordon RA, Herter JM, Rosetti F, Campbell AM, Nishi H, Kashgarian M, Bastacky SI, Marinov A, Nickerson KM, Mayadas TN, and Shlomchik MJ 2017. Lupus and proliferative nephritis are PAD4 independent in murine models. JCI Insight 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rother N, Pieterse E, Lubbers J, Hilbrands L, and van der Vlag J. 2017. Acetylated Histones in Apoptotic Microparticles Drive the Formation of Neutrophil Extracellular Traps in Active Lupus Nephritis. Front Immunol 8: 1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, Banchereau J, and Pascual V. 2011. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med 3: 73ra20. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES