Rationale: Exudate macrophages are key players in host defense toward invading pathogens. Their antiinflammatory and epithelial-protective potential in gram-negative pneumonia, however, remains elusive.
Objectives: We investigated whether exudate macrophages contributed to preservation of alveolar epithelial barrier integrity and analyzed the molecular pathways involved.
Methods: We evaluated the antiinflammatory and epithelial-protective effects of exudate macrophages in a model of LPS- and Klebsiella pneumoniae–induced lung injury comparing wild-type and CC-chemokine receptor 2 (CCR2)-deficient mice with defective lung macrophage recruitment and in in vitro studies using primary alveolar epithelial cells.
Measurements and Main Results: CCR2−/− mice exhibited enhanced alveolar epithelial cell apoptosis and lung leakage on intratracheal LPS treatment, which could be attributed to lack of exudate macrophage recruitment from the circulating pool as demonstrated in a model of wild-type/CCR2−/− bone-marrow chimeric mice. Among various antiinflammatory and proliferative mediators analyzed, the endogenous counterpart of resident macrophage–expressed IL-1β, IL-1 receptor antagonist (IL-1ra), was highly up-regulated in flow-sorted exudate macrophages in LPS-treated wild-type mice. LPS/IL-1β–induced impairment of alveolar epithelial cell integrity was antagonized by IL-1ra in vitro. Finally, intratracheal substitution of IL-1ra or intravenous adoptive transfer of IL-1ra+/+ but not IL-1ra−/− blood mononuclear cells attenuated alveolar inflammation, epithelial apoptosis, and loss of barrier function in LPS-challenged or K. pneumoniae-infected CCR2−/− mice and enhanced survival after K. pneumoniae infection.
Conclusions: We conclude that recruited lung macrophages attenuate IL-1β–mediated acute lung injury in gram-negative pneumonia by release of IL-1ra.
Little is known about the antiinflammatory and barrier-protective role of alveolar recruited macrophages in pathogen-induced epithelial lung injury.
This study demonstrates that lung-recruited exudate macrophages attenuate alveolar inflammation and epithelial damage in LPS-induced acute lung injury by releasing IL-1 receptor antagonist. Intratracheal substitution with recombinant IL-1 receptor antagonist counteracts epithelial injury and loss of barrier function and enhances survival in exudate macrophage recruitment–deficient mice after intratracheal LPS challenge or Klebsiella pneumoniae infection.
IL-1β activates the IL-1R1 receptor expressed on a variety of tissues. IL1-β actions are counteracted by an endogenous receptor antagonist, IL-1ra, binding to IL-1R1 without receptor activation. The local balance between IL-1β and IL-1ra thereby determines the net effects on IL-1R1 (6). It was shown that IL-1ra ameliorates a variety of lung diseases, such as interstitial pulmonary fibrosis, asthma, or ventilator-induced lung injury (4, 6), and low levels of IL-1ra in bronchoalveolar lavage fluid (BALF) of patients with ARDS were associated with poor prognosis (7). Although systemic administration of IL-1ra reduced mortality in an animal model of septic shock (8), reports on the role of IL-1ra in local and systemic bacterial infections are conflicting (9). Several reports suggest resident alveolar macrophages to be the primary source of IL-1β in lung inflammation (6, 10, 11); however, the role of resident and recruited mononuclear phagocytes in equilibrating the balance between IL-1β and IL-1ra actions in lung infection remains elusive.
Mononuclear phagocytes are long-living cells with broad differentiation potential, residing in the alveoli as resident macrophages and entering lung tissue from the bloodstream in a CC-chemokine receptor 2 (CCR2)-dependent way (12, 13). A role of alveolar resident and exudate mononuclear phagocytes in initiating and maintaining pulmonary inflammation and host defense in lung infection has been convincingly demonstrated (14–16). On the other hand, recent reports suggest that resident and circulating GR-1highCCR2highCX3CR1low phagocyte populations, due to their broad functional plasticity, additionally contribute to resolution and repair processes in acute lung inflammation that are essential for return to tissue homeostasis (17–19).
The aim of this study was to test the hypothesis that CCR2+ exudate macrophages acquire an antiinflammatory and barrier-protective phenotype in pathogen-induced lung injury. Using bone marrow chimeric and adoptive cell transfer mouse models and in vitro alveolar epithelial cell culture systems, we demonstrate that CCR2+ exudate macrophages attenuate alveolar inflammation and preserve epithelial barrier function by the release of IL-1ra in LPS- and K. pneumoniae–induced lung injury. Furthermore, we show that exogenous substitution of IL-1ra in CCR2−/− mice is associated with increased survival after K. pneumonia infection.
Some of the results of this study have been previously reported as abstract (20).
Materials and reagents are listed in the online supplement.
Wild-type (WT) C57BL/6J, CD45.1 alloantigen (Ly5.1), and IL-1ra−/− mice (weight 18–21 g, C57BL/6J background) were purchased from Charles River (Sulzfeld, Germany). CCR2−/− mice (C57BL/6J background) were originally obtained from W. A. Kuziel (21). CX3CR1GFP/GFP mice (C57BL/6J background) were generated as described previously (22). Parent CX3CR1GFP/GFP and CX3CR1+/+ mice were bred to yield heterozygous CX3CR1+/GFP offspring or crossbred with CCR2−/−CX3CR1+/+ mice to obtain CCR2+/+CX3CR1+/GFP and CCR2−/−CX3CR1+/GFP mice. Animals were bred in our facility under special pathogen-free conditions for three to four generations and used at 8 to 11 weeks of age. All animal experiments were approved by the local government committee.
Mice were intratracheally treated with ultrapure LPS (50 μg/mouse in sterile phosphate-buffered saline [PBS] in a total volume of 70 μl) or with K. pneumoniae serotype 2 (1 × 105 or 2 × 105 in sterile PBS in a total volume of 70 μl) as described (18, 23) and killed at the indicated time points. In selected experiments, hIL-1ra (Anakinra; Amgen, Seattle, WA; 10 μg/70 μl PBS) or PBS alone were applied intratracheally. Depletion of circulating monocytes was achieved by intravenous injection of 200 μl clodronate-containing liposomes 12 hours before LPS administration as described (24). Processing of BALF and blood cells and lung homogenates and analysis of alveolar leakage are outlined in the online supplement.
Bone marrow chimeric mice were created by intravenous transplantation of CD45.2+ WT or CCR2−/− bone marrow cells into lethally irradiated CD45.1 recipients as outlined in the online supplement. WT, IL-1ra−/−, or CCR2−/− peripheral blood mononuclear cells were isolated as described in the online supplement (13) and intravenously injected into CCR2−/− mice directly before LPS instillation.
For fluorescence-activated cell sorter (FACS) analysis and cell sorting, cells were processed as described in the online supplement using a FACSCanto or FACSVantage flow cytometer equipped with a FACSDiva software package (BD Biosciences, Heidelberg, Germany).
IL-1β and IL-1ra levels in BALF were measured using commercially available ELISA kits (R&D Systems, Wiesbaden, Germany) according to the manufacturer's instructions.
RNA isolation, cDNA synthesis, and real-time reverse transcriptase–polymerase chain reaction analysis were performed as previously described (18). The ΔCt values for each target gene were calculated using the reference gene Actin or PBGD with the formula ΔCt = Ctreference – Cttarget. Values are given as ΔΔCt or fold expression of control (2ΔΔCt), depending on the experiment.
Murine and human alveolar epithelial cells type II (AEC) were isolated as described (18, 25) and grown to confluency on 24-well plates, transwells, or chamber slides (BD Biosciences). Human lung tissue was obtained from lobectomy specimens distal from tumors after written patient consent (Departments of Pathology and Surgery, Justus-Liebig-University, Giessen; approved by the local ethics committee). Cells were treated with recombinant mouse or human IL-1β (10 ng/ml), LPS (1 μg/ml), or mouse or human IL-1ra (1 μg/ml) and subjected to further analyses as outlined in the supplement.
AEC were methanol/acetone fixed and stained overnight with anti–ZO-1 (1:50) or isotype antibodies. Lung cryosections were prepared from perfused and lavaged lungs and stained with hematoxylin-eosin (online supplement).
Data are presented as means ± SD. Statistical significance was estimated using one-way analysis of variance (SPSS for Windows); paired samples were analyzed using the two-tailed paired Student t test. A value of P ≤ 0.05 was considered as significant.
To investigate whether exudate macrophages (ExMa) might contribute to preservation of alveolar epithelial cell integrity in LPS-induced ALI, we compared AEC apoptosis and alveolar albumin leakage in WT and CCR2-deficient mice, well known to exhibit defective ExMa recruitment (12, 13). Indeed, ExMa recruitment was drastically reduced in CCR2-deficient mice after intratracheal LPS challenge, whereas sustained alveolar ExMa presence was observed in WT mice peaking at 48 hours after treatment as analyzed by differential counts from BALF cytospins (Figure 1A, upper panel). Flow cytometric immunophenotyping of BALF cells at 48 hours after LPS treatment confirmed the absence of CD11c+GR-1+F4/80+CD11bhigh ExMa (Figure 1A, bottom panel, P1, red) in CCR2−/− mice despite equal numbers of CD11c+GR-1lowF4/80+CD11bint resident alveolar macrophages (rAM) compared with WT mice (Figure 1A, bottom panel, P2, red). Likewise, analysis of rAM numbers in the time course after LPS challenge did not reveal significant differences between both mouse strains (Figure 1B), nor did analysis of alveolar lymphocyte subset numbers (CD4 T cells, CD8 T cells, natural killer cells; data not shown). Quantification of ExMa in the interstitial lung compartment (i.e., in lavaged and perfused lung digests) demonstrated strongly reduced numbers in CCR2-deficient mice, most prominent at 12 hours after LPS challenge (Figure 1C). Lack of interstitial ExMa recruitment in CCR2-deficient mice at this time point was associated with significantly elevated alveolar albumin leakage, a feature of lung injury (Figure 1D). Correspondingly, CCR2 deficiency was associated with significantly increased AEC apoptosis as demonstrated by Annexin V binding at 12 hours after LPS treatment (15.1 ± 1.59% vs. 33.2 ± 6.19%; Figure 1E). AEC were defined as CD45−CD31−cytokeratin+ cells from lung digests and consisted of approximately 87% AEC type II (pro-SP-C+) and approximately 13% AEC type I (T1α+; see Figure E1 in the online supplement).
To demonstrate that indeed CCR2 expression on circulating ExMa precursors (i.e., Gr-1+ blood monocytes) rather than CCR2 expression on resident lung myeloid or parenchymal cells accounted for attenuated epithelial injury, we made use of a bone marrow chimeric mouse model whereby we transplanted either WT or CCR2-deficient CD45.2+ bone marrow cells into lethally irradiated CD45.1+ WT recipients. At 14 days after transplantation bone marrow engraftment was always greater than 90%, whereas resident lung cells including rAM were of recipient phenotype as demonstrated by FACS analysis of CD45.1 and CD45.2 alloantigen expression on peripheral blood leukocytes and BALF rAM, respectively (Figure E2). As shown in Figure 2A, chimeric WT mice with CCR2-deficient circulating leukocytes revealed abrogated ExMa recruitment together with sustained increase in alveolar barrier dysfunction in the time course after LPS treatment (Figure 2B). These results clearly demonstrate that CCR2 expressed on circulating blood monocytes is critically involved in both ExMa extravasation into the interstitial and alveolar compartment and attenuation of LPS-induced lung injury.
To confirm that indeed recruitment of circulating blood monocytes attenuated lung injury in our model, we depleted monocytes in WT mice by intravenous injection of liposomal clodronate 12 hours before LPS challenge. As shown in Figure E3, side scatter (SSC)lowCD115+ monocytes were largely depleted in peripheral blood (top panel), resulting in abrogated interstitial accumulation of CD11c+GR-1+F4/80+CD11bhigh ExMa (bottom panel) after 12 hours of LPS treatment. Accordingly, AEC apoptosis (Figure 2C) and albumin leakage (Figure 2D) were enhanced in mice injected with clodronate liposomes as compared with empty liposome controls, demonstrating that depletion of ExMa precursors aggravated LPS-induced epithelial injury.
Given that ExMa recruitment was associated with attenuated epithelial injury on LPS treatment, we hypothesized that transendothelial/epithelial migration of peripheral blood monocytes (PB-Mo), giving rise to ExMa, might induce antiinflammatory, antiapoptotic, or epithelial-mitogenic mediators in these cells. We therefore analyzed the relative expression of potential candidates, such as IL-10, hepatocyte growth factor, keratinocyte growth factor/FGF7, transforming growth factor-β1, and IL-1ra (26–31) in ExMa versus PB-Mo flow-sorted from LPS-challenged CX3CR1+/GFP mice, expressing green fluorescent protein (GFP) in both PB-Mo and freshly recruited ExMa, at 48 hours as previously described (32). Representative FACS plots with gating of GFP-positive cells from BALF and peripheral blood are given in Figure 3A (upper panel). We found IL-10, hepatocyte growth factor, and keratinocyte growth factor gene expression slightly down-regulated in alveolar ExMa compared with PB-Mo, whereas transforming growth factor-β1 expression was moderately up-regulated. Strikingly, IL-1ra gene expression was highly up-regulated in alveolar transmigrated ExMa compared with expression in their circulating precursors, with a mean ddCT of 14.8 ± 1.59 (Figure 3A, bottom panel), whereas IL-1ra gene induction was only moderate in recruited neutrophils (Figure E4). Correspondingly, IL-1ra protein was alveolarly released in significantly higher amounts in WT mice at 48 hours after LPS challenge than in CCR2−/− mice (Figure 3B, upper panel). Given that ExMa-mediated antiapoptotic and barrier-protective effects occurred early, at 12 to 24 hours after LPS instillation when the majority of ExMa was present in the interstitial lung compartment, we speculated that ExMa might exert these effects already during the transmigration process, on close contact with AEC. We therefore measured IL-1ra levels in perfused and lavaged lungs containing mainly interstitial ExMa. Indeed, we found IL-1ra protein accumulating to high levels in WT lung parenchyma starting at 6 hours after LPS treatment, whereas IL-1ra levels remained significantly lower in CCR2-deficient mice at 24 to 72 hours (Figure 3B, bottom panel). Correspondingly, we found the IL-1β/IL-1ra receptor IL-1R1 surface expressed on AEC from lung digests as demonstrated by flow cytometry, and further up-regulated on intratracheal LPS treatment, likely enhancing AEC responsiveness to IL-1β effects (Figure 3C).
IL-β is known to be primarily expressed in mononuclear phagocytes, and it was previously suggested that rAM are a primary source of IL-β in patients with ARDS (10), contributing to lung injury progression. We therefore analyzed IL-1β gene expression in flow-sorted rAM from BALF of 24-hour sham- or LPS-treated CCR2+/+CX3CR1+/GFP and CCR2−/−CX3CR1+/GFP mice. rAM were identified as CD11c+CX3CR1−GR-1lowF4/80+ and could be clearly distinguished from CD11c−CX3CR1−GR-1highF4/80− alveolar neutrophils and from CD11c+CX3CR1+GR-1highF4/80+ ExMa (Figure 3D, left). As shown in Figure 3D (right panel), IL-1β mRNA was highly up-regulated in rAM after LPS challenge in both WT and CCR2-deficient mice (ddCT: WT, 6 ± 0.74; CCR2−/−, 5.27 ± 1.09, representing a mean 68- and 45-fold induction, respectively). Correspondingly, IL-1β protein was present in the alveoli (Figure 3E, top panel) and interstitial lung compartment (Figure 3E, bottom panel) in comparable amounts in both WT and CCR2−/− mice in response to LPS. These data suggest that resident rather than recruited mononuclear phagocytes represent the major source of alveolar IL-1β in our model of LPS-induced ALI.
Lung injury is at least in part initiated and maintained by recruited neutrophils, releasing proteases and reactive metabolites (33). IL-1β is known to promote polymorphonuclear neutrophil (PMN) recruitment, likely by inducing chemokine release and adhesion molecule expression involved in PMN extravasation (11). We therefore hypothesized that LPS-driven PMN recruitment would be enhanced in CCR2-deficient mice lacking IL-1ra expressing ExMa. Indeed, alveolar PMN numbers in CCR2-deficient mice widely exceeded those found in WT mice at 48 hours after LPS treatment (89.3 ± 21.1 × 105 vs. 43.4 ± 7.24 × 105), which was associated with CCR2 expression on circulating ExMa precursors as shown in WT mice transplanted either WT or CCR2-deficient bone marrow as outlined above. Interestingly, substitution of alveolar IL-1ra by a single intratracheal application (10 μg) at 0 hours in CCR2−/− mice reduced LPS-induced PMN recruitment to levels observed in WT mice, whereas intratracheal IL-1ra application into WT mice did not further reduce PMN numbers (Figure 4A). Respective lung sections demonstrate enhanced neutrophilic alveolitis in CCR2−/− mice, which was attenuated by IL-1ra treatment (Figure 4B). Furthermore, IL-1ra but not PBS treatment reduced alveolar release of the PMN-recruiting chemokine macrophage inflammatory protein-2 in CCR2-deficient mice at 12 hours after LPS challenge, but had no effect in WT mice (Figure 4C). Similarly, surface expression of AEC intracellular adhesion molecule-1, known to promote PMN recruitment into the airspaces by interaction with PMN β1- and β2-integrins, was reduced in IL-1ra–treated CCR2-deficient mice 24 hours after LPS treatment (Figure 4D). Flow cytometric analyses of adhesion molecule expression on alveolar PMN (β1- or β2-integrins) revealed no changes between WT and CCR2-deficient mice (data not shown). Together, our data suggest that ExMa-expressed IL-1ra attenuates neutrophilic alveolitis, involving inhibition of chemokine release from and adhesion molecule expression on AEC.
IL-1ra was previously shown to exert antiapoptotic effects on cardiomyocytes (31). We therefore speculated that the increase in AEC apoptosis in CCR2−/− mice might be related to lacking ExMa IL1-ra. Intratracheal substitution of IL-1ra reduced AEC apoptosis rates in LPS-treated CCR2-deficient mice at 12 and 48 hours compared with sham-treated controls but revealed no further antiapoptotic effect in LPS-treated WT mice (Figure 5A). Accordingly, mRNA expression of the proapoptotic cytokine Fas ligand (FasL), known to mediate AEC apoptosis during pathogen-induced ALI (34), was induced on LPS instillation in lung homogenates approximately 20-fold in WT mice and approximately 110-fold in CCR2−/− mice, which could be antagonized by intratracheal IL-1ra application in CCR2−/− mice but not WT mice at 6 hours of LPS treatment (Figure 5B). These data indicate that IL-1ra acts in an antiapoptotic way on AEC after LPS treatment, possibly through inhibition of a FasL-mediated apoptosis pathway.
Given that IL-1ra attenuated alveolar inflammation and AEC apoptosis, we hypothesized that IL-1ra treatment would stabilize alveolar barrier function. Indeed, alveolar IL-1ra deposition attenuated alveolar albumin leakage in CCR2-deficient mice to levels observed in WT mice at 12 hours after LPS treatment (Figure 6A). As it was previously shown that, apart from its proinflammatory actions, IL-1β might directly increase lung epithelial permeability (3), we studied surface expression of the tight junction protein ZO-1 in confluent murine AEC on LPS- or IL-1β treatment in vitro by immunofluorescence. As shown in Figure 6B, ZO-1 was surface expressed on confluent, untreated AEC. LPS treatment alone (1 μg/ml) induced a localized tight junction disruption after 8 hours as indicated by arrowheads. Treatment with IL-1β (10 ng/ml) alone or with LPS plus IL-1β resulted in tight junction disassembly at 4 hours followed by complete disruption of the AEC monolayer after 8 hours, which could be antagonized by the addition of 1 μg/ml IL-1ra. Furthermore, transepithelial electrical resistance in primary murine (top panel) or human (bottom panel) AEC was reduced on a 2-hour treatment with IL-1β alone or with LPS + IL-1β, which was inhibited in the presence of IL-1ra.
As shown in Figure 1C, LPS-induced alveolar albumin leakage in CCR2-deficient mice was not only increased at 12 hours after challenge compared with WT mice, but tended to persist over 72 hours, when values in WT mice had already declined to baseline levels. We therefore speculated that alveolar fluid clearance, maintained mainly by the epithelial sodium channel (ENaC) (35), might be compromised in ExMa-deficient mice. Indeed, CCR2-deficient mice displayed reduced lung gene expression of ENaC subunits α and β on 6 hours of LPS treatment. Intratracheal treatment with IL-1ra counteracted these effects, whereby ENaCα subunit expression even exceeded levels of untreated control mice (Figure 6C). Finally, stimulation of murine or human AEC with IL-1β in the presence of LPS reduced expression of ENaCα subunit compared with unstimulated controls, and this decrease was not only abrogated in the presence of IL-1ra, but, on addition of 1 μg/ml IL-1ra, ENaCα levels even exceeded those of unstimulated AEC (Figure 6D). These data indicate that, apart from indirect proinflammatory or proapoptotic effects on alveolar epithelium, IL-1β directly induces structural and functional alveolar epithelial barrier damage, which can be widely antagonized by IL-1ra.
To further confirm that ExMa-released IL-1ra accounted for the beneficial effects on AEC barrier function in our model, we established an adoptive transfer approach using CCR2−/− recipient mice that were either intravenously transferred IL-1ra+/+CCR2−/−, IL-1ra+/+CCR2+/+, or IL-1ra−/−CCR2+/+ mononuclear cells isolated from peripheral blood of CCR2−/−, WT, or IL-1ra−/− mice, respectively. Of note, in this model, CCR2-deficient recipient mice will largely recruit adoptively transferred CCR2+/+ monocytes into their lungs on LPS challenge. As shown in Figure 7, AEC apoptosis at 12 hours after LPS treatment (Figure 7A) was significantly higher in mice recruiting IL-1ra−/− compared with mice recruiting IL-1ra+/+ ExMa. Recipient mice that were transferred CCR2−/− mononuclear cells correspondingly demonstrated high AEC apoptosis. Similarly, albumin leakage was higher in CCR2-deficient mice recruiting IL-1ra−/− ExMa compared with mice recruiting IL-1ra+/+ ExMa (Figure 7B). These data clearly demonstrate that attenuation of LPS-induced lung injury is mediated by alveolar recruitment of IL-1ra+/+ but not IL-1ra−/− ExMa.
We finally sought to confirm our findings on the beneficial role of ExMa IL-1ra in LPS-induced lung injury in a mouse model of gram-negative pneumonia. We therefore infected WT and CCR2-deficient mice with 105 K. pneumoniae and comparatively addressed the effects of defective alveolar ExMa recruitment and of compensatory alveolar deposition of IL-1ra into CCR2-deficient mice on lung injury parameters. As outlined in Figure 8A, IL-1ra was alveolarly released to significantly higher amounts at 48 hours post-infection (hpi) in WT mice compared with CCR2−/− mice. Of note, intratracheal deposition of 10 μg IL-1ra into K. pneumoniae–challenged CCR2-deficient mice compensated reduced IL-1ra levels to those observed in WT mice. K. pneumoniae infection induced pronounced AEC apoptosis in CCR2−/− mice peaking at 24 to 48 hpi, which was widely reduced in WT mice. A single intratracheal application of IL-1ra at the time point of infection attenuated AEC apoptosis in CCR2-deficient mice at 48 hpi (Figure 8B). Correspondingly, alveolar albumin leakage was increased in CCR2−/− mice at 48 hours after K. pneumoniae infection compared with WT mice, and leakage induction was inhibited in CCR2-deficient mice after IL-1ra treatment (Figure 8C). To determine whether IL-1ra treatment affected host defense functions in terms of bacterial clearance from the lungs, bacteria in BALF from the different treatment groups were counted by plating. We found that CCR2 deficiency significantly impaired bacterial clearance at 24 and 48 hpi; however, IL-1ra treatment of CCR2−/− mice did not further impact on bacterial loads (Figure 8D). Finally, CCR2-deficiency was associated with a 100% mortality rate after 10 days post-infection (dpi), whereas all WT mice survived a dose of 1 × 105 K. pneumoniae. Early intratracheal treatment with IL-1ra significantly increased survival rates of CCR2−/− mice to 67% (Figure 8E). Together, these data suggest that ExMa-derived IL-1ra is protective in terms of epithelial injury, barrier function, and survival in gram-negative pneumonia.
ALI on gram-negative infection is associated with severe damage of the alveolar epithelial barrier, followed by edema formation and impaired gas exchange (2). On infectious challenge, lung resident and recruited mononuclear phagocytes acquire distinct M1 or M2 phenotypes and differentially regulate both induction of proinflammatory host defense mechanisms and, on the other side, termination of inflammation and initiation of repair processes in a tightly controlled fashion, respectively (17–19). However, the antiinflammatory, reparative potential of lung mononuclear phagocyte populations antagonizing early proinflammatory processes in favor of preservation or regeneration of alveolar barrier function in gram-negative pneumonia remains elusive. We hypothesized that CCR2+ macrophages recruited from the circulation and therefore endowed with a broad differentiation potential might acquire such a protective or reparative phenotype on alveolar recruitment after LPS- or K. pneumonia–induced lung inflammation. Using CCR2−/− mice and monocyte depletion strategies, we demonstrate that lack of ExMa accumulation in the lung parenchyma and airspaces of challenged mice resulted in a more severe course of lung injury with reduced release of the endogenous IL-1β antagonist IL-1ra in the airspaces and lung parenchyma. IL-1ra was highly up-regulated in ExMa in challenged WT mice and exerted antiinflammatory, antiapoptotic, and barrier-stabilizing functions toward alveolar epithelium. Hence, we conclude that CCR2+ circulating macrophage precursors, on recruitment to the lung, acquire the potential to counteract detrimental proinflammatory IL-1β effects and are therefore crucial players in balancing inflammatory processes in gram-negative pneumonia, attenuating otherwise fatal lung injury.
Of note, CCR2+ ExMa predominantly exerted these antiapoptotic and barrier-stabilizing effects as early as 12 hours after LPS challenge, when ExMa were mainly present in the interstitial lung compartment rather than in the airspaces in WT mice. These findings suggest that ExMa-derived IL-1ra was induced and released already during the migration process across the endothelial/epithelial barrier, fulfilling its function likely on direct contact of ExMa with the alveolar epithelium, before entering the alveolar space. Indeed, IL-1ra protein release in the interstitial lung tissue was already observed at 6 and 24 hours after LPS treatment in injured WT mice.
Our data provide evidence that antagonizing alveolar IL1-β in gram-negative lung infection attenuates epithelial injury in several ways. First, we observe that AEC apoptosis, increased in CCR2−/− mice defective in ExMa recruitment after LPS or Klebsiella challenge (105 colony-forming units), is attenuated after intratracheal substitution of 10 μg IL-1ra to levels observed in WT mice. IL-1ra has previously been shown to exert antiapoptotic effects toward myocardial cells after ischemic damage in vivo and direct inhibition of caspase-1 and -9 is discussed as an underlying mechanism (31). Furthermore, there is evidence that the IL-1R1 downstream adaptor TRIF activates caspase-8 in primary murine cells on IL-1β binding in bacterial infection (36). AEC apoptosis, however, might also be induced indirectly by proapoptotic factors released from invading neutrophils such as FasL, known to mediate AEC injury in bacterial pneumonia (34, 37). Indeed, FasL mRNA was strongly up-regulated in lung homogenates in challenged CCR2−/− mice compared with WT mice and widely reduced on IL-1ra administration. Correspondingly, IL-1ra attenuated neutrophilic alveolitis increased in CCR2-deficient mice, likely by reduction of epithelial intracellular adhesion molecule-1 and macrophage inflammatory protein-2, two crucial mediators of alveolar neutrophil recruitment that are known to be up-regulated by IL-1β (38, 39). In addition, reduced phagocytosis of apoptotic neutrophils in CCR2-deficient mice due to lack of exudate phagocytes might contribute to elevated neutrophil counts in CCR2−/− mice. Furthermore, we provide data that IL-1ra antagonizes IL-1β–mediated loss of epithelial integrity in vitro using primary murine and human AEC and confirm these findings in our in vivo models by analysis of albumin permeability. We show that transepithelial resistance across AEC monolayers is reduced after 2 hours of LPS/IL-1β stimulation, most likely due to IL-1β–induced tight junctional disassembly as shown by ZO-1 immunostaining. Both ZO-1 disaggregation and enhanced AEC permeability were abolished in the presence of IL-1ra. Similar effects were observed by Ganter and colleagues in an IL-1β overexpressing mouse model, which reveals IL-1β–mediated endothelial and epithelial permeability increase to be dependent on RhoA- and αvβ5/6-induced adherens junction disruption and highlights modulation of these pathways as potential ALI treatment strategy (3). Our data add to the aforementioned study and provide additional evidence that lung recruited CCR2+ macrophages effectively stabilize alveolar epithelial integrity by antagonizing these detrimental IL-1β effects.
As a consequence of alveolar barrier disruption, edema fluid accumulates within the airspaces, severely compromising gas exchange. Therefore, effective alveolar fluid clearance, maintained by the epithelial sodium channel ENaC on the apical epithelial surface, is of utmost importance (35). ENaC expression was previously shown to be down-regulated by IL-1β in a MAPK-dependent way (40); hence, we addressed whether ExMa IL-1ra would counteract these effects. Indeed, ENaC α subunit mRNA was decreased in murine and human AEC on LPS/IL-1β treatment, and this was completely abolished in the presence of IL-1ra. Likewise, down-regulation of ENaC α and β subunits in lung homogenates of CCR2−/− mice after LPS treatment was antagonized by intratracheal IL-1ra delivery, suggesting that CCR2+ ExMa may support ENaC-mediated fluid clearance after gram-negative pneumonia.
The concept that circulating precursor cells exert antiinflammatory effects through IL-1 antagonism has been previously raised by Ortiz and colleagues, who demonstrated that bone marrow–derived mesenchymal stem cells attenuate bleomycin-induced lung inflammation in an IL-1ra–dependent way. Interestingly, stem cell–mediated pulmonary delivery of IL-1ra revealed to be even more effective than continuous systemic application of recombinant IL-1ra (30). Similarly, Lamacchia and colleagues demonstrated circulating myeloid cell–derived IL-1ra to be crucial for survival after lethal endotoxemia (41), highlighting the importance of mapping cell-specific antiinflammatory effectors during lung injury. Different studies revealed that IL1-ra protected from experimental ventilator-induced lung injury and IL-1ra gene polymorphisms correlated with severity of lung injury after community-acquired pneumonia in children (4, 42). Yet, data on therapeutic IL-1ra application in lung injury in general or particularly in bacterial pneumonia are missing. A previous phase III trial testing intravenous IL-1ra treatment of sepsis did not reduce overall 28-day mortality (although in this study IL-1ra treatment was beneficial in a subgroup of patients with multiorgan failure and high predicted mortality), whereas a second sepsis study did not observe any benefit from IL-1ra treatment (43, 44). Parsons and colleagues even showed that elevated serum IL-1ra levels in critically ill patients were associated with death from ARDS (45). In this line, in our study, we noticed only very limited beneficial effects on LPS-induced epithelial injury after systemic (intraperitoneal) IL-1ra application (data not shown) as opposed to the clear-cut effects after intratracheal delivery, suggesting that blocking IL-1β actions locally might be more effective through direct protection of the alveolar epithelium and potentially associated with fewer systemic side effects. These data emphasize the importance of cell-based and lung compartment–specific intervention strategies to dampen lung inflammation and injury during severe pneumonia.
Of note, no substantial effects of a single intratracheal IL-1ra treatment on neutrophil recruitment, AEC apoptosis, or lung permeability could be detected in LPS-challenged WT mice with intact CCR2+ ExMa recruitment. This could be because certain levels of IL-1β might still be needed for recovery, even after sterile inflammation. To this end, previous reports documented a role of IL-1β in AEC proliferation and repair after injury (46, 47), suggesting that general inhibition of IL-1R1 signaling in lung injury might not necessarily be beneficial. Although we could not detect any differences in AEC proliferation in WT versus CCR2−/− mice after LPS challenge in the LPS model (18), this issue should be thoroughly taken into account when considering therapeutic modulation of the IL-1β/IL-1ra balance in patients with pneumonia. In bacterial infection, blockade of IL-1R1 signaling might even be detrimental, as we observe increased mortality in IL-1ra–treated WT mice challenged with a very high Klebsiella dose of 107/mouse (data not shown). We speculate that IL-1β is in some way needed for bacterial clearance, at least on high-dose challenge, although data on this issue are conflicting (9, 48), and we did not notice significant differences in lung bacterial loads in CCR2−/− mice infected with a moderate dose of 105. Tanabe and colleagues could not detect changes in bacterial clearance or in mortality in Klebsiella-infected IL-1 knockout compared with WT mice on low-dose infection (3.6 × 103) and concluded that TNF-α compensates for lacking IL-1β functions (48). In fact, taking these different issues into account, we speculate that IL-1ra treatment should be thoroughly timed during the infection course and likely exerts beneficial effects in later stages of infection, when infection is cleared from the lungs but progression to ARDS is ongoing. Antagonizing IL-1β might be of particular interest against the background of its profibrotic actions (49) in late ARDS characterized by progression to lung fibrosis. Moreover, it should be considered that therapeutic strategies based on cell recruitment blockade to dampen lung inflammation may abrogate important antiinflammatory and repair signals derived from those cells.
On the other hand, our group recently showed that ExMa massively recruited into the airspaces during influenza virus pneumonia in fact acquire a proinflammatory phenotype and induce alveolar epithelial apoptosis via TNF-related apoptosis-inducing ligand, thereby contributing to loss of barrier function (13). Such a divergent role of lung macrophages emerges most likely from the different injury models applied, as the lung mononuclear phagocyte pool represents a functionally diverse cell population whose fate is, at least in part, dependent on signals from surrounding lung cells during inflammatory conditions. These various conditions may impact on the functional differentiation of macrophages acquiring either an inflammatory, tissue-destructive or an antiinflammatory, reparative phenotype. Increasing evidence suggests that macrophages do not remain committed to a single activation state during inflammatory injury and recovery (50). In this line, we previously demonstrated that certain proinflammatory mediators, such as resident macrophage-released TNF-α, may even induce reparative signaling pathways in AEC type II in gram-negative pneumonia (18), demonstrating that the role of mononuclear phagocytes in pulmonary host defense fundamentally differs depending on the causative pathogen.
In conclusion, our study demonstrates a crucial role of recruited CCR2+ macrophages in attenuation of IL-1β–induced lung injury in gram-negative pneumonia and elucidated cell-specific functional profiles relevant for further time- and compartment-specific intervention strategies to equilibrate the imbalance of the alveolar IL-1β/IL1ra system without compromising host defense functions in severe pneumonia.
The authors thank D. Hensel, R. Winkler, and S. Fiedel for excellent technical assistance.
1. | Rubenfeld GD, Herridge MS. Epidemiology and outcomes of acute lung injury. Chest 2007;131:554–562. |
2. | Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 2000;342:1334–1349. |
3. | Ganter MT, Roux J, Miyazawa B, Howard M, Frank JA, Su G, Sheppard D, Violette SM, Weinreb PH, Horan GS, et al. Interleukin-1beta causes acute lung injury via alphavbeta5 and alphavbeta6 integrin-dependent mechanisms. Circ Res 2008;102:804–812. |
4. | Frank JA, Pittet JF, Wray C, Matthay MA. Protection from experimental ventilator-induced acute lung injury by IL-1 receptor blockade. Thorax 2008;63:147–153. |
5. | Pugin J, Ricou B, Steinberg KP, Suter PM, Martin TR. Proinflammatory activity in bronchoalveolar lavage fluids from patients with ARDS, a prominent role for interleukin-1. Am J Respir Crit Care Med 1996;153:1850–1856. |
6. | Arend WP. The balance between IL-1 and IL-1ra in disease. Cytokine Growth Factor Rev 2002;13:323–340. |
7. | Donnelly SC, Strieter RM, Reid PT, Kunkel SL, Burdick MD, Armstrong I, Mackenzie A, Haslett C. The association between mortality rates and decreased concentrations of interleukin-10 and interleukin-1 receptor antagonist in the lung fluids of patients with the adult respiratory distress syndrome. Ann Intern Med 1996;125:191–196. |
8. | Alexander HR, Doherty GM, Buresh CM, Venzon DJ, Norton JA. A recombinant human receptor antagonist to interleukin 1 improves survival after lethal endotoxemia in mice. J Exp Med 1991;173:1029–1032. |
9. | Hirsch E, Irikura VM, Paul SM, Hirsh D. Functions of interleukin 1 receptor antagonist in gene knockout and overproducing mice. Proc Natl Acad Sci USA 1996;93:11008–11013. |
10. | Jacobs RF, Tabor DR, Burks AW, Campbell GD. Elevated interleukin-1 release by human alveolar macrophages during the adult respiratory distress syndrome. Am Rev Respir Dis 1989;140:1686–1692. |
11. | Toews GB. Cytokines and the lung. Eur Respir J Suppl 2001;34:3s–17s. |
12. | Tsou CL, Peters W, Si Y, Slaymaker S, Aslanian AM, Weisberg SP, Mack M, Charo IF. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J Clin Invest 2007;117:902–909. |
13. | Herold S, Steinmueller M, von Wulffen W, Cakarova L, Pinto R, Pleschka S, Mack M, Kuziel WA, Corazza N, Brunner T, et al. Lung epithelial apoptosis in influenza virus pneumonia: the role of macrophage-expressed TNF-related apoptosis-inducing ligand. J Exp Med 2008;205:3065–3077. |
14. | Rosseau S, Hammerl P, Maus U, Walmrath HD, Schutte H, Grimminger F, Seeger W, Lohmeyer J. Phenotypic characterization of alveolar monocyte recruitment in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2000;279:L25–L35. |
15. | Winter C, Taut K, Srivastava M, Langer F, Mack M, Briles DE, Paton JC, Maus R, Welte T, Gunn MD, et al. Lung-specific overexpression of CC chemokine ligand (CCL) 2 enhances the host defense to streptococcus pneumoniae infection in mice: role of the CCL2–CCR2 axis. J Immunol 2007;178:5828–5838. |
16. | Xu F, Droemann D, Rupp J, Shen H, Wu X, Goldmann T, Hippenstiel S, Zabel P, Dalhoff K. Modulation of the inflammatory response to streptococcus pneumoniae in a model of acute lung tissue infection. Am J Respir Cell Mol Biol 2008;39:522–529. |
17. | Amano H, Morimoto K, Senba M, Wang H, Ishida Y, Kumatori A, Yoshimine H, Oishi K, Mukaida N, Nagatake T. Essential contribution of monocyte chemoattractant protein-1/C-C chemokine ligand-2 to resolution and repair processes in acute bacterial pneumonia. J Immunol 2004;172:398–409. |
18. | Cakarova L, Marsh LM, Wilhelm J, Mayer K, Grimminger F, Seeger W, Lohmeyer J, Herold S. Macrophage tumor necrosis factor-alpha induces epithelial expression of granulocyte-macrophage colony-stimulating factor: impact on alveolar epithelial repair. Am J Respir Crit Care Med 2009;180:521–532. |
19. | Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008;8:958–969. |
20. | Herold S, Shafiei Tabar T, Cabanski M, Kuziel WA, Seeger W, Lohmeyer J, Steinmueller M. IL-1 receptor antagonist exerts anti-apoptotic and barrier-protective effects towards alveolar epithelium in a murine model of LPS-induced acute lung injury [abstract]. Am J Respir Crit Care Med 2010;181:A2325. |
21. | Herold S, von Wulffen W, Steinmueller M, Pleschka S, Kuziel WA, Mack M, Srivastava M, Seeger W, Maus UA, Lohmeyer J. Alveolar epithelial cells direct monocyte transepithelial migration upon influenza virus infection: impact of chemokines and adhesion molecules. J Immunol 2006;177:1817–1824. |
22. | Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 2000;20:4106–4114. |
23. | von Wulffen W, Steinmueller M, Herold S, Marsh LM, Bulau P, Seeger W, Welte T, Lohmeyer J, Maus UA. Lung dendritic cells elicited by Fms-like tyrosine 3-kinase ligand amplify the lung inflammatory response to lipopolysaccharide. Am J Respir Crit Care Med 2007;176:892–901. |
24. | Sunderkotter C, Nikolic T, Dillon MJ, Van Rooijen N, Stehling M, Drevets DA, Leenen PJ. Subpopulations of mouse blood monocytes differ in maturation stage and inflammatory response. J Immunol 2004;172:4410–4417. |
25. | Rosseau S, Wiechmann K, Moderer S, Selhorst J, Mayer K, Krull M, Hocke A, Slevogt H, Seeger W, Suttorp N, et al. Moraxella catarrhalis–infected alveolar epithelium induced monocyte recruitment and oxidative burst. Am J Respir Cell Mol Biol 2005;32:157–166. |
26. | Panos RJ, Rubin JS, Csaky KG, Aaronson SA, Mason RJ. Keratinocyte growth factor and hepatocyte growth factor/scatter factor are heparin-binding growth factors for alveolar type II cells in fibroblast-conditioned medium. J Clin Invest 1993;92:969–977. |
27. | Li CM, Khosla J, Hoyle P, Sannes PL. Transforming growth factor-beta(1) modifies fibroblast growth factor-2 production in type ii cells. Chest 2001;120:60S–61S. |
28. | Buff SM, Yu H, McCall JN, Caldwell SM, Ferkol TW, Flotte TR, Virella-Lowell IL. IL-10 delivery by AAV5 vector attenuates inflammation in mice with Pseudomonas pneumonia. Gene Ther 2010;17:567–576. |
29. | Dewberry RM, King AR, Crossman DC, Francis SE. Interleukin-1 receptor antagonist (IL-1ra) modulates endothelial cell proliferation. FEBS Lett 2008;582:886–890. |
30. | Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K, Phinney DG. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci USA 2007;104:11002–11007. |
31. | Abbate A, Salloum FN, Vecile E, Das A, Hoke NN, Straino S, Biondi-Zoccai GG, Houser JE, Qureshi IZ, Ownby ED, et al. Anakinra, a recombinant human interleukin-1 receptor antagonist, inhibits apoptosis in experimental acute myocardial infarction. Circulation 2008;117:2670–2683. |
32. | Cabanski M, Wilhelm J, Zaslona Z, Steinmuller M, Fink L, Seeger W, Lohmeyer J. Genome-wide transcriptional profiling of mononuclear phagocytes recruited to mouse lungs in response to alveolar challenge with the TLR2 agonist Pam3CSK4. Am J Physiol Lung Cell Mol Physiol 2009;297:L608–L618. |
33. | Lee WL, Downey GP. Neutrophil activation and acute lung injury. Curr Opin Crit Care 2001;7:1–7. |
34. | Matute-Bello G, Frevert CW, Liles WC, Nakamura M, Ruzinski JT, Ballman K, Wong VA, Vathanaprida C, Martin TR. Fas/Fas ligand system mediates epithelial injury, but not pulmonary host defenses, in response to inhaled bacteria. Infect Immun 2001;69:5768–5776. |
35. | Morty RE, Eickelberg O, Seeger W. Alveolar fluid clearance in acute lung injury: what have we learned from animal models and clinical studies? Intensive Care Med 2007;33:1229–1240. |
36. | Ruckdeschel K, Pfaffinger G, Haase R, Sing A, Weighardt H, Hacker G, Holzmann B, Heesemann J. Signaling of apoptosis through TLRS critically involves toll/IL-1 receptor domain-containing adapter inducing IFN-beta, but not MyD88, in bacteria-infected murine macrophages. J Immunol 2004;173:3320–3328. |
37. | Glavan BJ, Holden TD, Goss CH, Black RA, Neff MJ, Nathens AB, Martin TR, Wurfel MM. Genetic variation in the Fas gene and associations with acute lung injury. Am J Respir Crit Care Med 2011;183:356–363. |
38. | Ying B, Yang T, Song X, Hu X, Fan H, Lu X, Chen L, Cheng D, Wang T, Liu D, et al. Quercetin inhibits IL-1 beta-induced ICAM-1 expression in pulmonary epithelial cell line A549 through the MAPK pathways. Mol Biol Rep 2009;36:1825–1832. |
39. | Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol 2005;32:311–318. |
40. | Roux J, Kawakatsu H, Gartland B, Pespeni M, Sheppard D, Matthay MA, Canessa CM, Pittet JF. Interleukin-1beta decreases expression of the epithelial sodium channel alpha-subunit in alveolar epithelial cells via a p38 MAPK-dependent signaling pathway. J Biol Chem 2005;280:18579–18589. |
41. | Lamacchia C, Palmer G, Bischoff L, Rodriguez E, Talabot-Ayer D, Gabay C. Distinct roles of hepatocyte- and myeloid cell-derived IL-1 receptor antagonist during endotoxemia and sterile inflammation in mice. J Immunol 2010;185:2516–2524. |
42. | Patwari PP, O'Cain P, Goodman DM, Smith M, Krushkal J, Liu C, Somes G, Quasney MW, Dahmer MK. Interleukin-1 receptor antagonist intron 2 variable number of tandem repeats polymorphism and respiratory failure in children with community-acquired pneumonia. Pediatr Crit Care Med 2008;9:553–559. |
43. | Fisher CJ Jr, Dhainaut JF, Opal SM, Pribble JP, Balk RA, Slotman GJ, Iberti TJ, Rackow EC, Shapiro MJ, Greenman RL, et al. Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group. JAMA 1994;271:1836–1843. |
44. | Opal SM, Fisher CJ Jr, Dhainaut JF, Vincent JL, Brase R, Lowry SF, Sadoff JC, Slotman GJ, Levy H, Balk RA, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. The interleukin-1 receptor antagonist sepsis investigator group. Crit Care Med 1997;25:1115–1124. |
45. | Parsons PE, Moss M, Vannice JL, Moore EE, Moore FA, Repine JE. Circulating IL-1ra and IL-10 levels are increased but do not predict the development of acute respiratory distress syndrome in at-risk patients. Am J Respir Crit Care Med 1997;155:1469–1473. |
46. | Geiser T, Jarreau PH, Atabai K, Matthay MA. Interleukin-1beta augments in vitro alveolar epithelial repair. Am J Physiol Lung Cell Mol Physiol 2000;279:L1184–L1190. |
47. | Geiser T, Atabai K, Jarreau PH, Ware LB, Pugin J, Matthay MA. Pulmonary edema fluid from patients with acute lung injury augments in vitro alveolar epithelial repair by an IL-1beta-dependent mechanism. Am J Respir Crit Care Med 2001;163:1384–1388. |
48. | Tanabe M, Matsumoto T, Shibuya K, Tateda K, Miyazaki S, Nakane A, Iwakura Y, Yamaguchi K. Compensatory response of IL-1 gene knockout mice after pulmonary infection with Klebsiella pneumoniae. J Med Microbiol 2005;54:7–13. |
49. | Gasse P, Mary C, Guenon I, Noulin N, Charron S, Schnyder-Candrian S, Schnyder B, Akira S, Quesniaux VF, Lagente V, et al. IL-1r1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J Clin Invest 2007;117:3786–3799. |
50. | Gordon S. Alternative activation of macrophages. Nat Rev Immunol 2003;3:23–35. |