Various states of inflammation, including sepsis, are associated with hypoferremia, which limits iron availability to pathogens and reduces iron-mediated oxidative stress. Lipocalin 2 (Lcn2; siderocalin, 24p3) plays a central role in iron transport. Accordingly, Lcn2-deficient (Lcn2KO) mice exhibit elevated intracellular labile iron. In this study, we report that LPS induced systemic Lcn2 by 150-fold in wild-type mice at 24 h. Relative to wild-type littermates, Lcn2KO mice were markedly more sensitive to endotoxemia, exhibiting elevated indices of organ damage (transaminasemia, lactate dehydrogenase) and increased mortality. Such exacerbated endotoxemia was associated with substantially increased caspase-3 cleavage and concomitantly elevated immune cell apoptosis. Furthermore, cells from Lcn2KO mice were hyperresponsive to LPS ex vivo, exhibiting elevated cytokine secretion. Additionally, Lcn2KO mice exhibited delayed LPS-induced hypoferremia despite normal hepatic hepcidin expression and displayed decreased levels of the tissue redox state indicators cysteine and glutathione in liver and plasma. Desferroxamine, an iron chelator, significantly protects Lcn2KO mice from LPS-induced toxicity, including mortality, suggesting that Lcn2 may act as an antioxidant in vivo by regulating iron homeostasis. Thus, Lcn2-mediated regulation of labile iron protects the host against sepsis. Its small size and simple structure may make Lcn2 a deployable treatment for sepsis.

Sepsis, a catastrophically amplified and dysregulated induction of proinflammatory cytokines often eventuating in multiorgan failure and death (1), is a major public health problem resulting in >200,000 deaths in the United States (2) and a significant burden on the global healthcare system (3). Sepsis is triggered by the host response to bacterial components such as LPS, a ligand for TLR4. The death of immune cells, particularly leukocytes, via extensive apoptosis is a major contributor to significant immune suppression and mortality in septic patients (4, 5). Although sepsis is typically characterized by robust production of proinflammatory cytokines, it has been recognized that some genes induced during sepsis serve as dampening agents that seek to restore immunological equilibrium (1). Identifying such agents and defining their mechanisms of action may result in new treatment regimens for sepsis.

Sepsis is not only associated with an extreme proinflammatory response but is also accompanied by hypoferremia of inflammation, a primitive defensive mechanism to drastically reduce the concentration of circulating iron, limiting its availability to pathogens (6, 7) and also ameliorating iron-dependent oxidative stress generated via a Fenton reaction (8). Lipocalin 2 (Lcn2; human ortholog neutrophil gelatinase-associated lipocalin [NGAL]) has been shown to be dramatically upregulated in various inflammatory conditions and is considered as an acute phase protein in mice (912). Several in vitro studies have demonstrated that Lcn2 protects against cellular stress and exposure to H2O2 and that overexpression of Lcn2 allows cells to tolerate superphysiological iron concentrations (1315). Under physiologic conditions Lcn2 can transport iron, perhaps with the aid of mammalian siderophores (16, 17). It has also been suggested that Lcn2 stabilizes the labile iron/siderophore complex (18, 19). Additionally, by chelating bacterial siderophores, Lcn2 exerts direct antibacterial activity and, accordingly, Lcn2-deficient (Lcn2KO) mice are sensitive to bacterial sepsis (11, 20, 21). Although such correlation could be consistent with a role for Lcn2 in driving or dampening an inflammatory response, the known bioactivities of Lcn2 led us to hypothesize that it might play a role in hypoferremia of inflammation and, thereby, the resolution of severe inflammation.

In this study, we tested the hypothesis that Lcn2 protects against severe inflammation using a murine model of LPS-induced sepsis. We observed that LPS dramatically induced Lcn2 and that Lcn2KO mice were highly sensitive to LPS-induced mortality, correlating with greater immune cell apoptosis and upregulation of proinflammatory gene expression. Such increased pathology to LPS in Lcn2KO mice is associated with both delayed hypoferremia and increased oxidative stress. Desferroxamine (DFO), an iron chelator, offers significant protection against LPS-induced toxicity and mortality in Lcn2KO mice. Thus, our study demonstrates that Lcn2 not only protects against bacterial sepsis but also regulates host proinflammatory cytokine expression by limiting iron-mediated oxidative stress.

Abs used were cyclooxygenase (COX)-2 (Cayman Chemical, Ann Arbor, MI), inducible NO synthase (iNOS; Upstate Biotechnology, Bedford, MA), and cleaved caspase-3 (Cell Signaling Technology, Danvers, MA), as well as anti–β-actin, LPS, and desferoxamine mesylate (Sigma-Aldrich, St. Louis, MO). Anti-mouse Lcn2 and all cytokine ELISA kits were from R&D Systems (Minneapolis, MN).

Lcn2KO mice (backcrossed to BL6 mice for >10 generations) generated by Dr. Shizuo Akira (Japan) were obtained via Dr. Alan Aderem (University of Washington) and crossed with BL6 wild-type (WT) mice. The offspring were crossed to obtain Lcn2KO mice and their WT littermates, whose direct descendants were used for all experiments in this study. Mice studies were approved by Georgia State University Animal Ethics Committee.

Eight-week-old mice (both male and female) were challenged i.p. with indicated dose of LPS from Escherichia coli 0128:B12 and monitored for body weight, symptoms of sepsis, mortality, and inflammatory parameters. Blood was collected retro-orbitally at indicated time points, and hemolysis-free serum was collected by centrifugation using serum separator tubes from BD Biosciences. Mice that lost 25% of their initial body weight or became moribund were considered dead. In some experiments, mice were pretreated with 100 μM DFO i.p. 1 h before the administration of LPS and monitored for cleaved caspase-3, systemic proinflammatory cytokines, organ damage markers, and mortality.

Peritoneal exudate cells (PECs) were collected by injecting 5 ml cold PBS i.p. and plated in six-well plates with DMEM supplemented with 10% FBS and 1% penicillin and streptomycin (22). Cells were cultured overnight at 37°C, washed three times with PBS to remove nonadherent cells, and stimulated with E. coli LPS (100 ng/ml) in serum-free DMEM. At various time points supernatants and lysates were collected for cytokine analysis and immunoblotting and stored at −80°C until analysis.

Bone marrow-derived macrophages (BMDMs) were cultured as described by Weischenfedlt and Porse (23). Briefly, bone marrow cells were isolated and cultured in six-well plates in DMEM supplemented with 10% FBS, 1% penicillin and streptomycin, and 10% conditioned L929 media as a source of M-CSF. On day 7, BMDM cultures with nearly 100% confluence were stimulated with the indicated dose of E. coli LPS in serum-free DMEM for 24 h. Cytokines were analyzed in the supernatants.

Cleaved caspase-3 in spleen and bone marrow was analyzed by immunoblotting. TUNEL-positive cells in 10% formalin-fixed tissue were identified using TUNEL assay (Roche, Indianapolis, IN). The images were captured using a Zeiss Axioskop2 Plus (Carl Zeiss Microimaging).

The apoptotic cell population in tissue was measured as in Zhang et al. (24) with all Abs procured from eBioscience (San Diego, CA) except for Ly-6G, which was from BD Biosciences (San Jose, CA). Annexin V staining was performed according to the manufacturer’s protocol (Molecular Probes, San Diego, CA). Data were acquired on a BD LSR II multiparameter flow cytometer and analyzed using FlowJo software (Tree Star, Ashland, OR).

Immunoblotting was performed as described in Vijay-Kumar et al. (25). Briefly, serum was diluted (1:10) in 2× Laemmli sample buffer (Bio-Rad) with 2-ME and 100 mM DTT, boiled for 5 min, immediately cooled on ice for 10 min, and subjected to SDS-PAGE on a 4–20% gel (Bio-Rad, Hercules, CA). Proteins were transferred to nitrocellulose, blocked with 5% milk for 1 h, probed with primary Ab biotinylated anti-mouse Lcn2 (0.2 μg/ml) overnight at 4°C, washed three times, and probed with streptavidin HRP (Invitrogen, Grand Island, NY) for 1 h at room temperature. Immunoblots were developed by ECL (GE Healthcare, Buckinghamshire, U.K.). For splenic cleaved-3 caspase, tissue was homogenized in RIPA buffer (Cell Signaling Technology) with protease inhibitor mixture (Roche). Total protein levels were quantitated by Bio-Rad method and 25 μg protein was loaded under denaturing conditions. Immunoblots were developed using Abs from Cell Signaling Technology as above. β-actin was probed as a loading control.

Cytokines in serum and cell supernatants were measured by DuoSet ELISA cytokine kits according to the manufacturer’s instructions. Serum IL-18 levels were measured by ELISA using an MBL kit.

Serum alanine transaminase (ALT) and aspartate transaminase (AST) were measured using the Cholestech LDX system (Alere, San Diego, CA) according to the manufacturer’s instructions.

Hemolysis-free serum samples obtained by retro-orbital plexus were analyzed using automated iron and lactate dehydrogenase (LDH) biochemical assays on an Olympus AU680 chemistry analyzer (Beckman-Coulter, Brea, CA). Analysis was performed at the Comparative Clinical Pathology Services (Columbia, MO) under the supervision of a board-certified veterinary clinical pathologist (Charles E. Wiedmeyer). Serum triglycerides were measured using a kit from BioVision (Milpitas, CA).

Tissue RNA was isolated using a Qiagen kit. Quantitative RT-PCR was employed to measure hepatic hepcidin, superoxide dismutase (SOD)2, and glutathione peroxidase (Gpx) using the following primers: hepcidin, forward, 5′-AGAGCTGCAGCCTTTGCAC-3′, reverse, 5′-GAAGATGCAGATGGGGAAGT-3′; SOD2, forward, 5′-CAGCCTCCCAGACCTGCCTT-3′, reverse, 5′-GTGCAGGCTGAAGAGCGACC-3′; Gpx, forward, 5′-GGCACCACGATCCGGGACTA-3′, reverse, 5′-AAATTGGGCTCGAACCCGCC-3′; and housekeeping gene 36B4, forward, 5′-TCCAGGCTTTGGGCATCA-3′, reverse, 5′-CTTTATCAGCTGCACATCACTCAGA-3′. Quantitative RT-PCR was carried out using a QuantiFast RT kit with conditions 20 min incubation at 50°C and 5 min at 95°C and 35–40 cycles of 10 s at 95°C and 30 s at 60°C using a Bio-Rad C1000 thermal cycler.

Liver and plasma (collected in EDTA tubes as anticoagulant) levels of cysteine, cystine (CySS), reduced glutathione (GSH), and oxidized glutathione (GSSG) were quantitated by HPLC and redox potential was calculated as described in Jones et al. (26) and Iyer et al (27).

Statistical analysis for significance (p < 0.05) was determined using a Student t test (GraphPad Prism).

To begin to investigate the role of Lcn2 in sepsis, we first analyzed the kinetics of Lcn2 secretion in response to an i.p. administered nonlethal dose of E. coli LPS (5 mg/kg body weight), a TLR4 ligand, in C57BL/6 mice. This resulted in a dramatic increase (150-fold) in serum Lcn2 that peaked at 24 h and returned to basal levels by 48 h (Fig. 1A). LPS induced Lcn2 with kinetics similar to the highly studied acute phase protein C-reactive protein in murine inflammatory models (28). Immunoblotting sera confirmed our ELISA results (Fig. 1B). Similar results were obtained by challenging WT mice with the TLR5 ligand, flagellin (data not shown).

FIGURE 1.

LPS-induced Lcn2 in vivo. Eight-week-old male WT BL6 mice were given E. coli LPS (5 mg/kg body weight) i.p., bled at indicated time points, and analyzed for (A) serum Lcn2 by ELISA. (B) Immunoblot of serum Lcn2 (2 and 24 h after LPS) under reducing (100 mM DTT and 2-ME) conditions.

FIGURE 1.

LPS-induced Lcn2 in vivo. Eight-week-old male WT BL6 mice were given E. coli LPS (5 mg/kg body weight) i.p., bled at indicated time points, and analyzed for (A) serum Lcn2 by ELISA. (B) Immunoblot of serum Lcn2 (2 and 24 h after LPS) under reducing (100 mM DTT and 2-ME) conditions.

Close modal

Lcn2 is a substantially upregulated host protein during the acute phase response not only during infection but also in sterile inflammation and aseptic diseases (29). To study the role of Lcn2 in sepsis, we subjected Lcn2KO mice and WT littermates to systemic treatment with a relatively high dose of LPS (20 mg/kg body weight) and monitored them for classical sepsis symptoms every day. LPS induced significant loss of body weight in Lcn2KO mice when compared with WT mice (Fig. 2A). Additionally, most Lcn2KO mice exhibited periocular discharge, piloerection, and lethargy, whereas in WT mice, only one of six mice exhibited the above symptoms. Accordingly, this treatment resulted in the death of 20% of WT mice within 8 d (Fig. 2B), whereas remaining WT mice appeared healthy at this time and remained so through 4 wk posttreatment. In contrast, 100% of Lcn2KO mice died within 8 d LPS injection.

FIGURE 2.

Lcn2KO mice are highly susceptible to LPS-induced mortality. Eight-week-old Lcn2KO mice and their WT littermates were given E. coli LPS (20 mg/kg body weight) and monitored for (A) mean body weight and (B) percentage survival. Data are results of an individual experiment (n = 6) and representative of three separate experiments that all yielded a very similar pattern of results.

FIGURE 2.

Lcn2KO mice are highly susceptible to LPS-induced mortality. Eight-week-old Lcn2KO mice and their WT littermates were given E. coli LPS (20 mg/kg body weight) and monitored for (A) mean body weight and (B) percentage survival. Data are results of an individual experiment (n = 6) and representative of three separate experiments that all yielded a very similar pattern of results.

Close modal

Sepsis results in multiple organ damage and dysfunction associated with elevated levels of circulating transaminases and LDH levels. Therefore, we measured serum levels of AST, ALT, and LDH at different time points in response to a nonlethal dose of LPS (5 mg/kg body weight) in WT and Lcn2KO mice. In accordance with greater mortality, Lcn2KO mice exhibited greater increases in serum AST, ALT, and LDH (Fig. 3A–C) when compared with WT mice. Furthermore, Lcn2KO mice also showed significantly elevated levels of serum triglycerides (Fig. 3D). Such elevated triglycerides, clinically termed lipemia of sepsis, correlates with TNF-α levels and is considered a host immune response intended to bind and neutralize LPS (30, 31). These results indicate that increases in Lcn2 expression in response to LPS serve to protect against sepsis.

FIGURE 3.

Lcn2KO mice display elevated systemic inflammatory markers. Mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight), bled at different time points, and analyzed for the organ damage markers in the serum at 6 and 24 h after LPS treatment. (A) AST, (B) ALT, (C) LDH, and (D) 24 h after LPS-treated serum triglycerides. *p < 0.05.

FIGURE 3.

Lcn2KO mice display elevated systemic inflammatory markers. Mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight), bled at different time points, and analyzed for the organ damage markers in the serum at 6 and 24 h after LPS treatment. (A) AST, (B) ALT, (C) LDH, and (D) 24 h after LPS-treated serum triglycerides. *p < 0.05.

Close modal

LPS provokes caspase-mediated apoptosis of immune cells, resulting in immunosuppression and mortality (32). Thus, we next investigated the extent to which Lcn2 might affect LPS-induced immune cell apoptosis. Mice were administered a modest dose of LPS (5 mg/kg body weight), which does not induce overt illness in either mouse strain. Spleen and bone marrow cells were isolated 24 h after LPS and analyzed for levels of cleaved caspase-3, a hallmark of apoptosis. This treatment did not elicit significant caspase-3 cleavage in WT mice. Strikingly, LPS treatment resulted in increased cleavage of executioner caspase-3 in Lcn2KO spleen and bone marrow cells (Fig. 4A, 4B). To rule out the possibility that WT mice may induce cleavage of caspase-3 at earlier time points, we monitored caspase-3 cleavage at 2, 4, 8, and 12 h in WT mice by immunoblotting, but at no time did we detect caspase-3 cleavage in the spleen (data not shown). In accordance, TUNEL assays performed on spleens showed increased numbers of apoptotic cell aggregates in LPS-treated Lcn2KO mice that were not apparent in similarly treated WT mice (Fig. 4C). Similarly, LPS induced more TUNEL-positive cells in the colon, specifically at the base of the crypts (data not shown).

FIGURE 4.

Lcn2 deficiency sensitizes LPS-induced apoptosis. WT or Lcn2KO mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and after 24 h apoptosis markers were measured in spleen and bone marrow cells. (A) Immunoblot showing cleaved caspase-3 in spleen lysates. (B) Immunoblot showing cleaved caspase-3 in bone marrow cells. (C) TUNEL staining of spleen. Original magnification ×20. White arrows show aggregates of TUNEL-positive cells in Lcn2KO mice.

FIGURE 4.

Lcn2 deficiency sensitizes LPS-induced apoptosis. WT or Lcn2KO mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and after 24 h apoptosis markers were measured in spleen and bone marrow cells. (A) Immunoblot showing cleaved caspase-3 in spleen lysates. (B) Immunoblot showing cleaved caspase-3 in bone marrow cells. (C) TUNEL staining of spleen. Original magnification ×20. White arrows show aggregates of TUNEL-positive cells in Lcn2KO mice.

Close modal

Next, to identify the immune cell populations most prone to apoptosis, we analyzed cells for annexin V staining by flow cytometry. We observed significant increases in LPS-induced apoptosis of CD4+ T cells (CD3+CD4+), neutrophils (CD11b+Ly-6G+), and dendritic cells (CD11b+CD11c+) isolated from spleens of Lcn2KO mice when compared with those derived from WT mice (Fig. 5). Analysis of bone marrow showed a similar trend of increased apoptosis in Lcn2KO neutrophils (not shown), while B cell (CD3CD19+B220+) and macrophage (CD45+CD11b+MHC class II+F4/80+) fractions were not different between the two genotypes in both tissues assayed (data not shown).

FIGURE 5.

Increased LPS-induced immune cell apoptosis in Lcn2KO mice. WT or Lcn2KO mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and after 24 h (%) apoptotic cells were measured in the spleen by flow cytometry. (A) CD3+CD4+ T cells. (B) CD11b+Ly-6G+ neutrophils. (C) CD11b+CD11c+ dendritic cells. *p < 0.05.

FIGURE 5.

Increased LPS-induced immune cell apoptosis in Lcn2KO mice. WT or Lcn2KO mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and after 24 h (%) apoptotic cells were measured in the spleen by flow cytometry. (A) CD3+CD4+ T cells. (B) CD11b+Ly-6G+ neutrophils. (C) CD11b+CD11c+ dendritic cells. *p < 0.05.

Close modal

LPS is known to induce extensive production of cytokines, especially TNF-α, a main determinant of multiple organ damage and death. Furthermore, LPS induced Lcn2 robustly in WT peritoneal exudate cells (data not shown). Therefore, we studied proinflammatory cytokine response in WT and Lcn2KO mice in vivo. Lcn2KO mice exhibited significantly elevated levels of serum TNF-α and IL-18, master proinflammatory mediators that are highly linked to sepsis (Fig. 6A, 6B). Such increased cytokine production was also seen in peritoneal immune cells stimulated with LPS ex vivo. Specifically, relative to WT mice, Lcn2KO PECs exhibited greater production of KC, IL-6, and TNF-α (Fig. 6C–E). Analogous results were seen for proinflammatory enzymes. Specifically, Western blots revealed that COX-2 levels were markedly upregulated in Lcn2KO cells and only moderately upregulated in WT cells, especially at the 12 h time point. Similar results were seen for iNOS, which was also modestly induced by LPS in WT cells but markedly induced in cells of LPS-treated Lcn2KO PECs (Fig. 6H). Such elevated proinflammatory mediators were accompanied by concomitant increases of the anti-inflammatory mediators IL-10 and secretory (s)IL-1Ra (Fig. 6F, 6G).

FIGURE 6.

Lcn2 deficiency potentiates LPS-induced proinflammatory gene expression. Mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and, after 6 h, serum was analyzed for (A) TNF-α and (B) IL-18 (pg/ml). (CH) PECs stimulated in triplicate with either PBS or LPS (100 ng/ml) in serum-free media. Supernatants were collected at 12 and 24 h and analyzed for cytokines by ELISA or lysates subjected to immunoblotting for COX2 and iNOS. (C) KC, (D) IL-6, (E) TNF-α, (F) IL-10, (G) sIL-1Ra (expressed in ng/mg protein). (H) Immunoblot showing iNOS and COX2 expression in PEC lysates. (A) and (B) and (C)–(H) are representatives of two and three independent experiments, respectively. *p < 0.05.

FIGURE 6.

Lcn2 deficiency potentiates LPS-induced proinflammatory gene expression. Mice (n = 3–6) were given either PBS or LPS (5 mg/kg body weight) and, after 6 h, serum was analyzed for (A) TNF-α and (B) IL-18 (pg/ml). (CH) PECs stimulated in triplicate with either PBS or LPS (100 ng/ml) in serum-free media. Supernatants were collected at 12 and 24 h and analyzed for cytokines by ELISA or lysates subjected to immunoblotting for COX2 and iNOS. (C) KC, (D) IL-6, (E) TNF-α, (F) IL-10, (G) sIL-1Ra (expressed in ng/mg protein). (H) Immunoblot showing iNOS and COX2 expression in PEC lysates. (A) and (B) and (C)–(H) are representatives of two and three independent experiments, respectively. *p < 0.05.

Close modal

Next, to examine the extent to which hyperresponsiveness of Lcn2KO PECs resulted from loss of Lcn2 in these cells per se or was an indirect consequence of a Lcn2-deficient environment, we compared responses of naive BMDMs that had been cultured from WT and Lcn2KO mice. First, we verified that WT BMDMs induced Lcn2 in response to LPS (data not shown). Similar to PECs, BMDMs from Lcn2KO mice exhibited greater LPS-induced cytokine secretion relative to BMDMs from WT mice. Such elevated cytokine secretion was seen for both proinflammatory cytokines (IL-1β, IL-6, and KC) and the anti-inflammatory cytokine (sIL-1Ra) at both 100 and 500 ng/ml concentrations of LPS (Supplemental Fig. 1), suggesting the increased septic response observed in Lcn2KO mice was not due to a broad defect in counterregulatory mediators but rather reflects an important direct role for Lcn2 in containing sepsis.

Inflammatory responses are accompanied by hypoferremia of inflammation, a primitive host protective mechanism that limits the iron available to invading pathogens and that can participate in a Fenton reaction, exacerbating oxidative stress (7, 33). Lcn2 is known to chelate iron and play a role in its transport (16, 17). Thus, we reasoned that loss of Lcn2 may disrupt iron transport while rendering intracellular siderophore-bound iron more labile (18, 19), thus mediating the high toxicity to LPS seen in Lcn2KO mice. Measurement of serum iron revealed a modest increase in basal iron levels in Lcn2KO mice relative to WT littermates. Moreover, Lcn2KO mice exhibited only modest hypoferremia in response to LPS, ∼25% by 4 h, whereas iron levels dropped by >50% in WT mice. At later times, serum iron levels fell in Lcn2KO mice such that they were similar to those of WT mice from 6 to 24 h after LPS treatment (Fig. 7). Interestingly, there was no significant difference in basal or LPS-induced hepatic hepcidin, a major hypoferremic hormone, in WT and Lcn2KO mice (Supplemental Fig. 2A).

FIGURE 7.

Lcn2KO mice exhibit delayed LPS-induced hypoferremia. Mice (n = 4–7) were given either PBS or LPS (5 mg/kg body weight) and, at indicated time points, serum iron levels were measured as described in 2Materials and Methods.

FIGURE 7.

Lcn2KO mice exhibit delayed LPS-induced hypoferremia. Mice (n = 4–7) were given either PBS or LPS (5 mg/kg body weight) and, at indicated time points, serum iron levels were measured as described in 2Materials and Methods.

Close modal

Lcn2 not only transports and stabilizes iron but also regulates the intracellular iron concentration. Nairz et al. (34) demonstrated that Lcn2KO mice have increased cellular iron levels. We hypothesized that such defective iron homeostasis in Lcn2KO mice may render them more susceptible to oxidative stress via participation of iron in the Fenton reaction. To study this, we analyzed GSH and cysteine, as well as GSSG and CySS, in plasma and liver as indicators of redox state, itself an important determinant of cytokine expression as changes in cysteine and GSH metabolism modulate inflammation (35). As shown in Fig. 8A and 8B, Lcn2KO mice exhibited significantly elevated levels of CySS and GSSG in the liver 24 h after LPS administration when compared with similarly treated WT mice. Similar, but more modest, elevations in CySS and GSSG were observed in plasma (data not shown). Next, we analyzed expression of the antioxidant enzymes SOD2 and Gpx1 in livers of WT and Lcn2KO mice. Whereas WT mice livers upregulated SOD2 expression by 10-fold 6 h after LPS with expression returning to basal levels by 24 h, Lcn2KO mice exhibited significantly less SOD2 expression when compared with WT mice despite decreased levels of tissue redox state indicators (Fig. 8C). No significant differences were observed in Gpx levels between the two strains (Supplemental Fig. 2B).

FIGURE 8.

Lcn2KO mice exhibit greater LPS-induced oxidative stress than did WT mice. Mice (n = 3–5) were given either PBS or LPS (5 mg/kg body weight) and at indicated time points for liver cysteine (Cys), CySS, GSH, and GSSG levels were measured and redox potential was calculated. A portion of liver was used to isolate RNA for quantitative RT-PCR of SOD2. (A) Liver (CySS/Cys), (B) liver GSSG/GSH, (C) liver SOD2. Data are from representative of two independent experiments. *p < 0.05.

FIGURE 8.

Lcn2KO mice exhibit greater LPS-induced oxidative stress than did WT mice. Mice (n = 3–5) were given either PBS or LPS (5 mg/kg body weight) and at indicated time points for liver cysteine (Cys), CySS, GSH, and GSSG levels were measured and redox potential was calculated. A portion of liver was used to isolate RNA for quantitative RT-PCR of SOD2. (A) Liver (CySS/Cys), (B) liver GSSG/GSH, (C) liver SOD2. Data are from representative of two independent experiments. *p < 0.05.

Close modal

DFO is a siderophore-based iron chelator routinely used clinically to treat iron overload. DFO-bound iron is metabolically inactive and unable to participate in a Fenton reaction, thus limiting free radical generation (3638). To study our hypothesis that delayed LPS-induced hypoferremia, as well as increased intracellular labile iron, in Lcn2KO mice may render them more susceptible to LPS-induced septic shock, we pretreated Lcn2KO mice with DFO (100 μM) i.p. 1 h before LPS (5 mg/kg body weight) administration. Control mice were given PBS. Such pretreatment of Lcn2KO mice with DFO resulted in significant reduction of cleaved caspase-3 in the spleen (Fig. 9A) and systemic proinflammatory gene expression. Specifically, in DFO-pretreated Lcn2KO mice, LPS-induced IL-6 and KC were reduced by >50% at 4 h relative to Lcn2KO mice pretreated with PBS. Additionally, whereas at 24 h cytokine levels remained elevated in PBS-pretreated LPS-challenged Lcn2KO mice, they were undetectable in DFO-pretreated Lcn2KO mice (Fig. 9B, 9C). Furthermore, DFO pretreatment substantially reduced LPS-induced AST and ALT, markers of liver and multiple organ damage, respectively (Fig. 9D, 9E). Finally, to test whether DFO-mediated reduction in cleaved caspase-3 and serum inflammatory markers would protect against LPS-induced mortality, we challenged Lcn2KO mice with a high dose of LPS (20 mg/kg body weight) 1 h after DFO administration. Lcn2KO mice that were pretreated with DFO still exhibited symptoms of sepsis initially but recovered and survived (70%), whereas Lcn2KO mice given PBS succumbed to death within 7 d (Fig. 9F). Collectively, our studies demonstrate that Lcn2 is a host protective factor against LPS-induced toxicity (Fig. 10).

FIGURE 9.

DFO, an iron chelator, neutralized LPS-induced toxicity in Lcn2KO mice. Mice (n = 3–5) were given either PBS or 100 μM DFO in PBS 1 h before administration of LPS (5 mg/kg body weight). At indicated time points, mice were bled, euthanized, and analyzed for apoptosis marker caspase-3 cleavage in spleen, systemic proinflammatory cytokines, and organ damage markers. (A) Caspase-3 cleavage in spleen. (B) Serum IL-6, (C) serum KC, (D) serum AST, and (E) serum ALT. For DFO-induced protection against mortality, Lcn2KO mice (n = 6) were given 100 μM DFO 1 h before lethal dose of LPS (20 mg/kg body weight) i.p. (F) DFO-induced protection against LPS-induced mortality in Lcn2KO mice. *p < 0.05.

FIGURE 9.

DFO, an iron chelator, neutralized LPS-induced toxicity in Lcn2KO mice. Mice (n = 3–5) were given either PBS or 100 μM DFO in PBS 1 h before administration of LPS (5 mg/kg body weight). At indicated time points, mice were bled, euthanized, and analyzed for apoptosis marker caspase-3 cleavage in spleen, systemic proinflammatory cytokines, and organ damage markers. (A) Caspase-3 cleavage in spleen. (B) Serum IL-6, (C) serum KC, (D) serum AST, and (E) serum ALT. For DFO-induced protection against mortality, Lcn2KO mice (n = 6) were given 100 μM DFO 1 h before lethal dose of LPS (20 mg/kg body weight) i.p. (F) DFO-induced protection against LPS-induced mortality in Lcn2KO mice. *p < 0.05.

Close modal
FIGURE 10.

Potential mechanism for increased susceptibility of Lcn2KO mice to LPS-induced mortality.

FIGURE 10.

Potential mechanism for increased susceptibility of Lcn2KO mice to LPS-induced mortality.

Close modal

Lcn2 is an acute phase protein known to be highly upregulated during inflammatory conditions such as sepsis. Induced acute phase proteins may amplify or dampen the inflammatory response. For instance, HMGB1 and SAA are known to amplify the inflammatory response by activating TLR4. However, the role of Lcn2 in sepsis is not known. In this study we demonstrate that Lcn2 is highly upregulated in WT mice in response to the TLR4 ligand, LPS. In Lcn2KO mice, LPS-induced sepsis resulted in increased toxicity as accessed by immune cell apoptosis, proinflammatory cytokine gene expression, systemic organ damage markers, and mortality. Furthermore, we demonstrated that Lcn2KO mice exhibited delayed hypoferremia of inflammation at the onset of inflammation, which eventuated in increased oxidative stress. Whereas delayed hypoferremia was most prominent at 4 h, elevated oxidative stress was only observed at later time points (24 h). This temporal difference likely reflects that the major nonprotein thiol and cellular reductant, GSH, is generally present in high concentrations (39), likely high enough to protect from oxidative damage for a limited period, thus manifesting in depletion of GSH stores only at 24 h (39). Importantly, reducing levels of free iron with the iron chelator DFO substantially ameliorated LPS-induced toxicity and mortality.

Sepsis is characterized by an exacerbated host proinflammatory response and elevated immune cell apoptosis eventuating in immunosuppression. The increased sensitivity of Lcn2-deficient cells to LPS suggests that Lcn2 may negatively regulate proinflammatory gene expression. Our results also support the previous study that used siderophore/iron-free recombinant Lcn2 to substantially reduce LPS-induced proinflammatory cytokines in macrophages (40). However, it is not clear at present whether the highly upregulated Lcn2 observed during inflammatory conditions is apo-Lcn2 (iron- and siderophore-free) or whether it is bound to a siderophore, another aspect of Lcn2 biology that warrants further investigation. It would be reasonable to predict that mammalian-derived Lcn2 would be in a siderophore-containing form, as Lcn2 can only bind iron through the action of a mammalian siderophore such as catechol or 2,5-dihydroxybenzoic acid (19). Exploring the role of apo- and holo-Lcn2 (siderophore- or siderophore- and iron-bound) during inflammatory conditions, their targets and molecular fates could reveal key insights regarding Lcn2 signaling and iron homeostasis. However, current technology does not easily permit in-depth study of the iron binding states of Lcn in vivo.

Immune cell apoptosis is a major factor underlying the increased mortality seen in septic patients (5). The increased immune cell apoptosis observed in Lcn2KO mice may contribute to the high mortality of the mice. Furthermore, Lcn2-deficient cells not only exhibited elevated proinflammatory cytokines but also substantially upregulated anti-inflammatory molecules (IL-10 and sIL-1Ra), which may also contribute to immunosuppression, a phenomenon known as compensatory anti-inflammatory response syndrome (41). Additionally, apoptotic immune cells appeared as aggregates in the spleens of Lcn2KO mice, suggesting the possibility that impaired autophagy results in a delay in the clearance of apoptotic cells contributing to the leakage of cellular debris, including many activators of innate immunity (danger-associated molecular patterns such as Hsp70 and HMGB1), which would further amplify the proinflammatory response (42).

Furthermore, our study clearly demonstrates that Lcn2 facilitates LPS-induced hypoferremia at early time points, allowing the host to control iron equilibrium based on inflammatory conditions. It is likely that Lcn2KO mice with significantly elevated proinflammatory cytokines accompanied with modestly elevated basal systemic iron levels may further aggravate LPS-induced toxicity in these mice. It is also possible that elevated concentrations of intracellular “labile” iron (34) in Lcn2KO mice may readily participate in a Fenton reaction, potentiating LPS-induced toxicity (18, 19). The substantial reduction of LPS-induced caspase-3 cleavage, organ damage markers, and proinflammatory cytokines by DFO suggests that dysregulated iron homeostasis plays a role in LPS-induced toxicity in Lcn2KO mice.

Macrophages play a central role in sepsis via secretion of proinflammatory cytokines, particularly TNF-α, and also by regulating iron homeostasis (43). The secretion of proinflammatory cytokines in response to LPS is NF-κB dependent (44, 45). It has been shown that treatment of cultured hepatic macrophages with 1,2-dimethyl-3-hydroxypyrid-4-one, a lipophilic iron chelator, blocked LPS-induced NF-κB activation and proinflammatory TNF-α and IL-6 expression (46). Furthermore, Darshan et al. (47) demonstrated that 6 h pretreatment of RAW264.7 macrophages with DFO inhibited LPS-induced IL-6 in vitro and LPS-induced TNF-α and IL-6 were significantly lower in rats on an iron-deficient diet. Similarly, DFO treatment reduced LPS-induced mortality in mice (48). Iron supplementation increases proinflammatory cytokines in IL-10KO mice, which are prone to develop spontaneous colitis (49). Furthermore, Sindrilaru et al. (50) have shown that in humans with chronic venous leg ulcers, iron-loaded macrophages exhibit increased proinflammatory cytokines. This study also showed that coadministering iron/dextran delays wound healing whereas DFO improves wound healing. Taken together, these studies demonstrate that iron homeostasis plays a crucial role not only in ROS generation but also in host proinflammatory gene expression. Accordingly, neutralization of LPS-induced toxicity by DFO in Lcn2KO mice strongly supports previous work, which demonstrated elevated intracellular labile iron levels in Lcn2KO mice (34). Thus, elevated Lcn2 during sepsis serves to induce hypoferremia and stabilize iron, which can limit oxidative stress, cellular apoptosis, and proinflammatory gene expression as shown schematically in Fig. 10.

That dramatic induction of Lcn2 protects mice against catastrophic inflammation suggests the possibility that Lcn2, and its human ortholog NGAL, may have therapeutic potential in the treatment of septic shock. Indeed, our results suggest that Lcn2 not only exerts antiseptic activity via bacterial siderophore sequestration (51), but it also acts as an anti-inflammatory agent by regulating hypoferremia of inflammation, which rescues host immune cell apoptosis/proinflammatory gene expression by maintaining thiol/disulfide redox homeostasis. The fact that NGAL is not an acute phase protein in humans (52) and thus does not exhibit the dramatic induction during infection seen in mice may bolster the potential for use of NGAL in humans as a dampener of the systemic inflammatory response to severe infections. That Lcn2/NGAL is a small, structurally simple yet functionally diverse molecule further contributes to its therapeutic potential against sepsis (53, 54).

We thank Daniel Moore, III for technical assistance.

This work was supported by National Institutes of Health Grants K01 DK083275 (to M.V.-K.) and R01 DK061417 and R01 DK083890 (to A.T.G.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ALT

alanine transaminase

AST

aspartate transaminase

BMDM

bone marrow-derived macrophage

COX

cyclooxygenase

CySS

cystine

DFO

desferroxamine

Gpx

glutathione peroxidase

GSH

reduced glutathione

GSSG

oxidized glutathione

iNOS

inducible NO synthase

Lcn2

lipocalin 2

Lcn2KO

lipocalin 2-deficient

LDH

lactate dehydrogenase

NGAL

neutrophil gelatinase-associated lipocalin

PEC

peritoneal exudate cell

s

secretory

SOD

superoxide dismutase

WT

wild-type.

1
Cohen
J.
2002
.
The immunopathogenesis of sepsis.
Nature
420
:
885
891
.
2
Martin
G. S.
,
Mannino
D. M.
,
Eaton
S.
,
Moss
M.
.
2003
.
The epidemiology of sepsis in the United States from 1979 through 2000.
N. Engl. J. Med.
348
:
1546
1554
.
3
Edbrooke
D. L.
,
Hibbert
C. L.
,
Kingsley
J. M.
,
Smith
S.
,
Bright
N. M.
,
Quinn
J. M.
.
1999
.
The patient-related costs of care for sepsis patients in a United Kingdom adult general intensive care unit.
Crit. Care Med.
27
:
1760
1767
.
4
Felmet
K. A.
,
Hall
M. W.
,
Clark
R. S.
,
Jaffe
R.
,
Carcillo
J. A.
.
2005
.
Prolonged lymphopenia, lymphoid depletion, and hypoprolactinemia in children with nosocomial sepsis and multiple organ failure.
J. Immunol.
174
:
3765
3772
.
5
Boomer
J. S.
,
To
K.
,
Chang
K. C.
,
Takasu
O.
,
Osborne
D. F.
,
Walton
A. H.
,
Bricker
T. L.
,
Jarman
S. D.
 II
,
Kreisel
D.
,
Krupnick
A. S.
, et al
.
2011
.
Immunosuppression in patients who die of sepsis and multiple organ failure.
JAMA
306
:
2594
2605
.
6
Ganz
T.
,
Nemeth
E.
.
2009
.
Iron sequestration and anemia of inflammation.
Semin. Hematol.
46
:
387
393
.
7
Collins
H. L.
2008
.
Withholding iron as a cellular defence mechanism: friend or foe?
Eur. J. Immunol.
38
:
1803
1806
.
8
Hentze
M. W.
,
Muckenthaler
M. U.
,
Andrews
N. C.
.
2004
.
Balancing acts: molecular control of mammalian iron metabolism.
Cell
117
:
285
297
.
9
Mori
K.
,
Lee
H. T.
,
Rapoport
D.
,
Drexler
I. R.
,
Foster
K.
,
Yang
J.
,
Schmidt-Ott
K. M.
,
Chen
X.
,
Li
J. Y.
,
Weiss
S.
, et al
.
2005
.
Endocytic delivery of lipocalin-siderophore-iron complex rescues the kidney from ischemia-reperfusion injury.
J. Clin. Invest.
115
:
610
621
.
10
Mishra
J.
,
Dent
C.
,
Tarabishi
R.
,
Mitsnefes
M. M.
,
Ma
Q.
,
Kelly
C.
,
Ruff
S. M.
,
Zahedi
K.
,
Shao
M.
,
Bean
J.
, et al
.
2005
.
Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery.
Lancet
365
:
1231
1238
.
11
Flo
T. H.
,
Smith
K. D.
,
Sato
S.
,
Rodriguez
D. J.
,
Holmes
M. A.
,
Strong
R. K.
,
Akira
S.
,
Aderem
A.
.
2004
.
Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron.
Nature
432
:
917
921
.
12
Vijay-Kumar
M.
,
Sanders
C. J.
,
Taylor
R. T.
,
Kumar
A.
,
Aitken
J. D.
,
Sitaraman
S. V.
,
Neish
A. S.
,
Uematsu
S.
,
Akira
S.
,
Williams
I. R.
,
Gewirtz
A. T.
.
2007
.
Deletion of TLR5 results in spontaneous colitis in mice.
J. Clin. Invest.
117
:
3909
3921
.
13
Roudkenar
M. H.
,
Halabian
R.
,
Bahmani
P.
,
Roushandeh
A. M.
,
Kuwahara
Y.
,
Fukumoto
M.
.
2011
.
Neutrophil gelatinase-associated lipocalin: a new antioxidant that exerts its cytoprotective effect independent on heme oxygenase-1.
Free Radic. Res.
45
:
810
819
.
14
Roudkenar
M. H.
,
Halabian
R.
,
Ghasemipour
Z.
,
Roushandeh
A. M.
,
Rouhbakhsh
M.
,
Nekogoftar
M.
,
Kuwahara
Y.
,
Fukumoto
M.
,
Shokrgozar
M. A.
.
2008
.
Neutrophil gelatinase-associated lipocalin acts as a protective factor against H2O2 toxicity.
Arch. Med. Res.
39
:
560
566
.
15
Hu
L.
,
Hittelman
W.
,
Lu
T.
,
Ji
P.
,
Arlinghaus
R.
,
Shmulevich
I.
,
Hamilton
S. R.
,
Zhang
W.
.
2009
.
NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells.
Lab. Invest.
89
:
531
548
.
16
Devireddy
L. R.
,
Hart
D. O.
,
Goetz
D. H.
,
Green
M. R.
.
2010
.
A mammalian siderophore synthesized by an enzyme with a bacterial homolog involved in enterobactin production.
Cell
141
:
1006
1017
.
17
Bao
G.
,
Clifton
M.
,
Hoette
T. M.
,
Mori
K.
,
Deng
S. X.
,
Qiu
A.
,
Viltard
M.
,
Williams
D.
,
Paragas
N.
,
Leete
T.
, et al
.
2010
.
Iron traffics in circulation bound to a siderocalin (Ngal)-catechol complex.
Nat. Chem. Biol.
6
:
602
609
.
18
Cherayil
B. J.
2011
.
The role of iron in the immune response to bacterial infection.
Immunol. Res.
50
:
1
9
.
19
Correnti
C.
,
Strong
R. K.
.
2012
.
Mammalian siderophores, siderophore-binding lipocalins, and the labile iron pool.
J. Biol. Chem.
287
:
13524
13531
.
20
Goetz
D. H.
,
Holmes
M. A.
,
Borregaard
N.
,
Bluhm
M. E.
,
Raymond
K. N.
,
Strong
R. K.
.
2002
.
The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron acquisition.
Mol. Cell
10
:
1033
1043
.
21
Berger
T.
,
Togawa
A.
,
Duncan
G. S.
,
Elia
A. J.
,
You-Ten
A.
,
Wakeham
A.
,
Fong
H. E.
,
Cheung
C. C.
,
Mak
T. W.
.
2006
.
Lipocalin 2-deficient mice exhibit increased sensitivity to Escherichia coli infection but not to ischemia-reperfusion injury.
Proc. Natl. Acad. Sci. USA
103
:
1834
1839
.
22
Vijay-Kumar
M.
,
Carvalho
F. A.
,
Aitken
J. D.
,
Fifadara
N. H.
,
Gewirtz
A. T.
.
2010
.
TLR5 or NLRC4 is necessary and sufficient for promotion of humoral immunity by flagellin.
Eur. J. Immunol.
40
:
3528
3534
.
23
Weischenfeldt
J.
,
Porse
B.
.
2008
.
Bone marrow-derived macrophages (BMM): isolation and applications.
CSH Protoc.
2008
:
pdb.prot5080
.
24
Zhang
B.
,
Zhang
Y.
,
Niu
L.
,
Vella
A. T.
,
Mittler
R. S.
.
2010
.
Dendritic cells and Stat3 are essential for CD137-induced CD8 T cell activation-induced cell death.
J. Immunol.
184
:
4770
4778
.
25
Vijay-Kumar
M.
,
Wu
H.
,
Aitken
J.
,
Kolachala
V. L.
,
Neish
A. S.
,
Sitaraman
S. V.
,
Gewirtz
A. T.
.
2007
.
Activation of toll-like receptor 3 protects against DSS-induced acute colitis.
Inflamm. Bowel Dis.
13
:
856
864
.
26
Jones
D. P.
,
Carlson
J. L.
,
Samiec
P. S.
,
Sternberg
P.
 Jr.
,
Mody
V. C.
 Jr.
,
Reed
R. L.
,
Brown
L. A.
.
1998
.
Glutathione measurement in human plasma. Evaluation of sample collection, storage and derivatization conditions for analysis of dansyl derivatives by HPLC.
Clin. Chim. Acta
275
:
175
184
.
27
Iyer
S. S.
,
Torres-Gonzalez
E.
,
Neujahr
D. C.
,
Kwon
M.
,
Brigham
K. L.
,
Jones
D. P.
,
Mora
A. L.
,
Rojas
M.
.
2010
.
Effect of bone marrow-derived mesenchymal stem cells on endotoxin-induced oxidation of plasma cysteine and glutathione in mice.
Stem Cells Int.
2010
:
868076
.
28
Paul
A.
,
Ko
K. W.
,
Li
L.
,
Yechoor
V.
,
McCrory
M. A.
,
Szalai
A. J.
,
Chan
L.
.
2004
.
C-reactive protein accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice.
Circulation
109
:
647
655
.
29
Sultan
S.
,
Pascucci
M.
,
Ahmad
S.
,
Malik
I. A.
,
Bianchi
A.
,
Ramadori
P.
,
Ahmad
G.
,
Ramadori
G.
.
2012
.
LIPOCALIN-2 is a major acute-phase protein in a rat and mouse model of sterile abscess.
Shock
37
:
191
196
.
30
Memon
R. A.
,
Grunfeld
C.
,
Moser
A. H.
,
Feingold
K. R.
.
1993
.
Tumor necrosis factor mediates the effects of endotoxin on cholesterol and triglyceride metabolism in mice.
Endocrinology
132
:
2246
2253
.
31
Barcia
A. M.
,
Harris
H. W.
.
2005
.
Triglyceride-rich lipoproteins as agents of innate immunity.
Clin. Infect. Dis.
41
(
Suppl. 7
):
S498
S503
.
32
Gautier
E. L.
,
Huby
T.
,
Saint-Charles
F.
,
Ouzilleau
B.
,
Chapman
M. J.
,
Lesnik
P.
.
2008
.
Enhanced dendritic cell survival attenuates lipopolysaccharide-induced immunosuppression and increases resistance to lethal endotoxic shock.
J. Immunol.
180
:
6941
6946
.
33
Malyszko
J.
,
Tesar
V.
,
Macdougall
I. C.
.
2010
.
Neutrophil gelatinase-associated lipocalin and hepcidin: what do they have in common and is there a potential interaction?
Kidney Blood Press. Res.
33
:
157
165
.
34
Nairz
M.
,
Theurl
I.
,
Schroll
A.
,
Theurl
M.
,
Fritsche
G.
,
Lindner
E.
,
Seifert
M.
,
Crouch
M. L.
,
Hantke
K.
,
Akira
S.
, et al
.
2009
.
Absence of functional Hfe protects mice from invasive Salmonella enterica serovar Typhimurium infection via induction of lipocalin-2.
Blood
114
:
3642
3651
.
35
Biswas
S. K.
,
Rahman
I.
.
2009
.
Environmental toxicity, redox signaling and lung inflammation: the role of glutathione.
Mol. Aspects Med.
30
:
60
76
.
36
Halliwell
B.
,
Gutteridge
J. M.
.
1984
.
Role of iron in oxygen radical reactions.
Methods Enzymol.
105
:
47
56
.
37
Simeonova
P. P.
,
Luster
M. I.
.
1995
.
Iron and reactive oxygen species in the asbestos-induced tumor necrosis factor-alpha response from alveolar macrophages.
Am. J. Respir. Cell Mol. Biol.
12
:
676
683
.
38
Kamp
D. W.
,
Graceffa
P.
,
Pryor
W. A.
,
Weitzman
S. A.
.
1992
.
The role of free radicals in asbestos-induced diseases.
Free Radic. Biol. Med.
12
:
293
315
.
39
Jones
D. P.
2008
.
Radical-free biology of oxidative stress.
Am. J. Physiol. Cell Physiol.
295
:
C849
C868
.
40
Zhang
J.
,
Wu
Y.
,
Zhang
Y.
,
Leroith
D.
,
Bernlohr
D. A.
,
Chen
X.
.
2008
.
The role of lipocalin 2 in the regulation of inflammation in adipocytes and macrophages.
Mol. Endocrinol.
22
:
1416
1426
.
41
Kumpf
O.
,
Schumann
R. R.
.
2010
.
Genetic variation in innate immunity pathways and their potential contribution to the SIRS/CARS debate: evidence from human studies and animal models.
J. Innate Immun.
2
:
381
394
.
42
Kono
H.
,
Rock
K. L.
.
2008
.
How dying cells alert the immune system to danger.
Nat. Rev. Immunol.
8
:
279
289
.
43
Ganz
T.
2012
.
Macrophages and systemic iron homeostasis.
J. Innate Immun
.
44
Libermann
T. A.
,
Baltimore
D.
.
1990
.
Activation of interleukin-6 gene expression through the NF-κB transcription factor.
Mol. Cell. Biol.
10
:
2327
2334
.
45
Ghosh
S.
,
Karin
M.
.
2002
.
Missing pieces in the NF-κB puzzle.
Cell
109
(
Suppl.
):
S81
S96
.
46
Lin
M.
,
Rippe
R. A.
,
Niemelä
O.
,
Brittenham
G.
,
Tsukamoto
H.
.
1997
.
Role of iron in NF-κB activation and cytokine gene expression by rat hepatic macrophages.
Am. J. Physiol.
272
:
G1355
G1364
.
47
Darshan
D.
,
Frazer
D. M.
,
Wilkins
S. J.
,
Anderson
G. J.
.
2010
.
Severe iron deficiency blunts the response of the iron regulatory gene Hamp and pro-inflammatory cytokines to lipopolysaccharide.
Haematologica
95
:
1660
1667
.
48
Vulcano
M.
,
Meiss
R. P.
,
Isturiz
M. A.
.
2000
.
Deferoxamine reduces tissue injury and lethality in LPS-treated mice.
Int. J. Immunopharmacol.
22
:
635
644
.
49
Oldenburg
B.
,
van Berge Henegouwen
G. P.
,
Rennick
D.
,
Van Asbeck
B. S.
,
Koningsberger
J. C.
.
2000
.
Iron supplementation affects the production of pro-inflammatory cytokines in IL-10 deficient mice.
Eur. J. Clin. Invest.
30
:
505
510
.
50
Sindrilaru
A.
,
Peters
T.
,
Wieschalka
S.
,
Baican
C.
,
Baican
A.
,
Peter
H.
,
Hainzl
A.
,
Schatz
S.
,
Qi
Y.
,
Schlecht
A.
, et al
.
2011
.
An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice.
J. Clin. Invest.
121
:
985
997
.
51
Borregaard
N.
,
Cowland
J. B.
.
2006
.
Neutrophil gelatinase-associated lipocalin, a siderophore-binding eukaryotic protein.
Biometals
19
:
211
215
.
52
Klausen
P.
,
Niemann
C. U.
,
Cowland
J. B.
,
Krabbe
K.
,
Borregaard
N.
.
2005
.
On mouse and man: neutrophil gelatinase associated lipocalin is not involved in apoptosis or acute response.
Eur. J. Haematol.
75
:
332
340
.
53
Gebauer
M.
,
Skerra
A.
.
2012
.
Anticalins small engineered binding proteins based on the lipocalin scaffold.
Methods Enzymol.
503
:
157
188
.
54
Skerra
A.
2008
.
Alternative binding proteins: anticalins - harnessing the structural plasticity of the lipocalin ligand pocket to engineer novel binding activities.
FEBS J.
275
:
2677
2683
.

The authors have no financial conflicts of interest.