Acute exacerbation of chronic obstructive pulmonary disease (COPD) is often induced by infection and often has a poor prognosis. Bacterial LPS activates innate immune receptor TLR4 followed by activation of a transcriptional factor IFN regulatory factor-3 (IRF3) as well as NF-κB, resulting in upregulation of various inflammatory mediators. To clarify the role of IRF3 in the pathogenesis of LPS-triggered COPD exacerbation, porcine pancreatic elastase (PPE) followed by LPS was administered intranasally to wild-type (WT) or IRF3−/− male mice. Sequential quantitative changes in emphysema were evaluated by microcomputed tomography, and lung histology was evaluated at the sixth week. WT mice treated with PPE and LPS exhibited enlarged alveolar spaces, whereas this feature was attenuated in similarly treated IRF3−/− mice. Moreover, LPS-induced emphysema aggravation was detected only in WT mice. Analysis of acute inflammation induced by PPE plus LPS revealed that the lungs of treated IRF3−/− mice had decreased mRNA transcripts for MCP-1, MIP-1α, TNF-α, and IFN-γ–inducible protein-10 but had increased neutrophils. IRF3 was involved in the production of mediators from macrophages, alveolar epithelial cells, and neutrophils. Furthermore, compared with isolated WT neutrophils from inflamed lung, those of IRF3−/− neutrophils exhibited impaired autophagic activation, phagocytosis, and apoptosis. These results suggest that IRF3 accelerated emphysema formation based on distinct profiles of mediators involved in LPS-induced COPD exacerbation. Regulation of the IRF3 pathway can affect multiple cell types and contribute to ameliorate pathogenesis of infection-triggered exacerbation of COPD.

Chronic obstructive pulmonary disease (COPD) is a progressive disease that affects ∼10% of people worldwide (1). By 2030, COPD is predicted to be the third leading cause of mortality and fifth leading cause of being disabled (2, 3). Acute exacerbation of COPD is often induced by infection with Gram-negative bacteria, which occurs frequently during the clinical course of COPD and is a major cause of mortality and decreased quality of life (4). Despite intensive treatment with antibiotics, bronchodilators, and steroids during episodes of exacerbation, respiratory function often remains worse than before the exacerbation (5, 6).

Gram-negative bacteria contain LPS, also called endotoxin, which is recognized by host innate immune receptor TLR4 and subsequently triggers inflammation (7). Downstream signaling of TLR4 consists of two major pathways: the MyD88-dependent pathway and the Toll/IL-1R domain–containing adaptor inducing IFN-β (TRIF)-dependent pathway. The MyD88 pathway activates NF-κB, resulting in the production of inflammatory mediators, such as IL-6 and TNF-α, which are involved in COPD pathogenesis (8, 9). The TRIF pathway activates IFN regulatory factor-3 (IRF3) and produces type 1 IFN and IFN-γ–inducible protein-10 (IP-10) (10, 11). Although each pathway potentiates a distinct profile of cytokines and chemokines, NF-κB subunit p35 and IRF3 cooperate as an enhanceosome (1214), suggesting that the inflammatory mediators mainly regulated by NF-κB may also be regulated by IRF3. Although the TLR4–MyD88–NF-κB pathway is thought to have a role in COPD exacerbation (15), the contribution of the TLR4–TRIF–IRF3 pathway remains to be elucidated.

In the present study, we hypothesized that IRF3 has an essential role in the pathogenesis of LPS-induced acute exacerbation of COPD. We used a mouse model of COPD acute exacerbation in which LPS was administered after emphysema was induced by using porcine pancreatic elastase (PPE). In PPE plus LPS–treated IRF3−/− mice, emphysema formation was attenuated, and the expression of multiple cytokines and chemokines associated with COPD in the acute inflammatory phase was suppressed. Alveolar epithelial cells, lung macrophages, and neutrophils were partially involved in these differences of expressions. Although the number of neutrophils infiltrating into the lungs of IRF3−/− mice was increased compared with similarly treated wild-type (WT) mice, the neutrophils isolated from the inflamed lungs exhibited decreased apoptosis, phagocytosis, and autophagy. Consequently, IRF3 contributes to the pathogenesis of LPS-induced COPD exacerbation based on different production of inflammatory mediators from multiple cell types that may lead to altered neutrophil function.

Specific pathogen-free male C57BL6/N mice (8–12 wk old) were purchased from Sankyo Laboratory (Tokyo, Japan). IRF3−/− mice (C57BL/6J background) (16) were purchased from RIKEN BioResource Center (Tsukuba, Japan), with the approval of Dr. T. Taniguchi, who originally generated this strain. The mice were housed in a specific pathogen-free environment with free access to food and water, under approval of the Institutional Animal Experiments Committee of Musashino University. Care and use of the animals followed the Principles of Laboratory Animal Care formulated by the National Society for Medical Research of Japan.

In our COPD exacerbation model, WT and IRF3−/− mice were anesthetized with ketamine (90 mg/kg i.p.) and medetomidine (1 mg/kg i.p.); 0.5 U of PPE (Sigma-Aldrich, St. Louis, MO) was administered intranasally on day 0; and then 25 μg of LPS from Escherichia coli 0111:B4 (Sigma-Aldrich) or vehicle (PBS) was administered intranasally on days 7, 10, and 14 (Fig. 1A). In our acute inflammation model, mice intranasally received 0.5 U of PPE or 25 μg of LPS or PBS (PPE group, LPS group, and PBS group, respectively); mice in the PPE plus LPS group intranasally received 25 μg of LPS 1 wk after PPE treatment (Fig. 2A).

FIGURE 1.

IRF3−/− mice exhibit suppressed emphysema formation in COPD exacerbation model. (A) Study design for COPD exacerbation model. Mice were intranasally administered PPE on day 0 and then LPS (PPE plus LPS group) or PBS (PPE group) on days 7, 10, and 14. (B) Representative chest micro-CT images of normal (left) and emphysematous (right) lung. White arrow indicates emphysematous area. (C) Sequential change of percentage of low attenuation area of WT or IRF3−/− mice in the PPE or PPE plus LPS group measured by micro-CT. *p < 0.05, corresponds to WT PPE plus LPS group compared with IRF3−/− PPE plus LPS group. (D) Change in percentage of low attenuation area of WT (upper panel) and IRF3−/− (lower panel) mice since first administration of LPS, described as difference from percentage of low attenuation area at week 1. *p < 0.05. (E) Representative lung section of untreated WT (upper), PPE plus LPS-treated WT (left lower), and PPE plus LPS-treated IRF3−/− group (right lower). H&E staining. Scale bar, 100 μm; original magnification, ×100. (F) The mean linear intercept calculated from pathological images of WT or IRF3−/− PPE plus LPS group. Data represent mean ± SEM. *p < 0.05; PPE plus LPS group, n = 12–14 in each group; PPE group, n = 5–6 in each group.

FIGURE 1.

IRF3−/− mice exhibit suppressed emphysema formation in COPD exacerbation model. (A) Study design for COPD exacerbation model. Mice were intranasally administered PPE on day 0 and then LPS (PPE plus LPS group) or PBS (PPE group) on days 7, 10, and 14. (B) Representative chest micro-CT images of normal (left) and emphysematous (right) lung. White arrow indicates emphysematous area. (C) Sequential change of percentage of low attenuation area of WT or IRF3−/− mice in the PPE or PPE plus LPS group measured by micro-CT. *p < 0.05, corresponds to WT PPE plus LPS group compared with IRF3−/− PPE plus LPS group. (D) Change in percentage of low attenuation area of WT (upper panel) and IRF3−/− (lower panel) mice since first administration of LPS, described as difference from percentage of low attenuation area at week 1. *p < 0.05. (E) Representative lung section of untreated WT (upper), PPE plus LPS-treated WT (left lower), and PPE plus LPS-treated IRF3−/− group (right lower). H&E staining. Scale bar, 100 μm; original magnification, ×100. (F) The mean linear intercept calculated from pathological images of WT or IRF3−/− PPE plus LPS group. Data represent mean ± SEM. *p < 0.05; PPE plus LPS group, n = 12–14 in each group; PPE group, n = 5–6 in each group.

Close modal
FIGURE 2.

IRF3 involvement in production of multiple cytokines and chemokines in PPE plus LPS acute inflammation model. (A) In vivo study design. Lung and BALF are harvested 24 h after last stimulation. (B and C) Lung mRNA expression levels in each group investigated by quantitative PCR. Factors namely associated with (B) COPD and (C) neutrophil recruitment are shown. Data represent mean ± SEM. *p < 0.05; for (A)–(C) n = 3 in the PBS group, n = 8–12 in the other groups.

FIGURE 2.

IRF3 involvement in production of multiple cytokines and chemokines in PPE plus LPS acute inflammation model. (A) In vivo study design. Lung and BALF are harvested 24 h after last stimulation. (B and C) Lung mRNA expression levels in each group investigated by quantitative PCR. Factors namely associated with (B) COPD and (C) neutrophil recruitment are shown. Data represent mean ± SEM. *p < 0.05; for (A)–(C) n = 3 in the PBS group, n = 8–12 in the other groups.

Close modal

At weeks 0, 1, 2, 4, and 6, mice were anesthetized with ketamine (90 mg/kg i.p.) and medetomidine (1 mg/kg i.p.) and fixed in a dorsal position in a chamber for microcomputed tomographic scanning of small animals (LaTheta LCT-200; Aloka, Tokyo, Japan) (17).

After the scanning range was determined, computed tomography (CT) of the chest was performed in respiratory-gated mode. Acquired data were analyzed according to CT number in Hounsfield units (HU). First, low-attenuation areas, the volume of which reflects emphysema, were quantified as those having a CT number of −871 to −610 HU, as previously described (18). Second, the total lung volume without vasculature volume was quantified as regions having a value in the range of −871 to −250 HU and was confirmed by calculating the lung area from actual images. Third, the total intratracheal air volume was determined with the same CT number range of −871 to −250 HU. The percentage of low-attenuation areas was calculated as the ratio of the volume of low-attenuation areas to total lung volume. Because it was included in both original values, intratracheal air volume was subtracted from the low-attenuation volume and the total lung volume.

At week 6, mice were euthanized, a tracheal cannula was inserted, and the lungs were inflated with 4% paraformaldehyde for fixation, paraffin embedded, and cut in 5-μm sections. To histopathologically assess emphysema formation, we stained the lung sections with H&E and calculated the mean linear intercept, as previously described (19, 20).

In our acute inflammation model, mice were euthanized on the day after their last treatment. To obtain bronchoalveolar lavage fluid (BALF), the trachea was catheterized, and the lungs were washed with 1 ml of sterile saline three times. The isolated BALF was centrifuged at 450 × g for 10 min at 4°C, pellets were dissolved in 1 ml of PBS, and the cells were counted. Differential cell counts were performed on cytospin preparations stained with Diff-Quik (Sysmex, Hyogo, Japan) according to the manufacturer’s instructions. The methods for total RNA extraction from harvested lungs, cDNA synthesis, and quantitative PCR have been previously described (20). The expression of genes of interest was calculated according to the comparative threshold cycle method, using β-actin as an internal control.

Naive macrophages were isolated from BALF as previously described (21), and alveolar epithelial cells were isolated as previously described (22, 23), with some modifications. Briefly, after lungs were perfused with PBS, 0.1% Dispase II (Wako Pure Chemical Industries, Osaka, Japan) in HBSS was instilled via a catheter inserted in the trachea. Then lungs were incubated in the Dispase solution for 45 min at room temperature and then cut into small pieces in DMEM containing 0.01% DNase (Sigma-Aldrich). The cell suspensions were filtered through cell strainers (Corning, Corning, NY), centrifuged at 150 × g for 10 min at 4°C, and washed with 0.01% DNase in DMEM. Cells were then purified using centrifugation on a discontinuous Percoll gradient, followed by incubation in Primaria (Corning) tissue culture plates at 37°C for 12 h. Nonattached cells were used for culture. The purity of alveolar epithelial cells was confirmed by immunostaining for pro-surfactant protein C.

Macrophages were cultured in DMEM with 5% FBS and 1% penicillin/streptomycin. The next day, cells were simultaneously treated with filtered 8 mU/ml PPE in PBS and 20 ng/ml LPS. After 24 h, RNA was harvested from these cells and used for quantitative PCR analysis. Alveolar epithelial cells were cultured in bronchial epithelial cell growth medium (Lonza, Walkersville, MD) supplemented with 5% FBS and 1% penicillin/streptomycin for 4–6 d to ∼90% confluence. After cells were treated for 24 h with 8 mU/ml PPE plus 20 ng/ml LPS (PPE plus LPS), RNA was harvested from these cells.

For the experiments using lung neutrophils and macrophages, lungs were perfused with 3 ml of PBS via the right ventricle, and both lobes were pooled for cell sorting. The lobes were cut into small pieces and digested in HBSS supplemented with 0.2% collagenase (Wako), 0.1% Dispase II, and 0.025% DNase for 60 min at 37°C. Cells were passed through a 40-μm cell strainer and washed with PBS. RBCs were lysed with lysis buffer (BD Biosciences, San Diego, CA), and the remaining cells were suspended in sorting buffer (PBS with 2% FBS and 1% penicillin/streptomycin). Then cells were incubated with biotinylated anti–Gr-1 Ab and allophycocyanin-conjugated anti-CD11c Ab (eBioscience, San Diego, CA) before incubation with streptavidin-PE or streptavidin-PerCP-Cy5.5 (eBioscience) for an apoptosis assay; Gr-1highCD11clow cells were sorted (SH800 cell sorter; Sony, Tokyo, Japan) as lung neutrophils, and Gr-1intCD11chigh cells were sorted as lung macrophages (24). Dead cells were identified by propidium iodide (PI; Sigma-Aldrich) staining and excluded from our analyses except for an apoptosis assay. Cell purity was confirmed by cytospin preparations stained with Diff-Quik (Sysmex). For RNA extraction, ∼4 × 105 neutrophils for each sample were used immediately after sorting.

Lung neutrophils (2.5 × 105 cells per tube) that had not been affected by PI were incubated with FITC-labeled annexin V and PI in an apoptosis detection kit (Takara, Tokyo, Japan) or with MitoPT in a MitoPT apoptosis detection kit (ImmunoChemistry Technologies, Bloomington, MN) and analyzed by flow cytometry (SH800 cell sorter; Sony). Data were analyzed according to the manufacturer’s instructions. Data were expressed as the difference between test samples and controls without annexin V.

Sorted lung neutrophils or macrophages (2 × 105 cells per tube) were washed with PBS, suspended in sorting buffer, and incubated with carboxylate-modified, fluorescent yellow-green latex beads (Sigma-Aldrich) at 37°C, under 5% CO2 for 2 h. After cells were washed with PBS and suspended in sorting buffer, phagocytosing cells were detected as FITC+ cells by flow cytometry. Data were calculated as the difference in fluorescence intensity of the cells with or without beads. In some experiments, cytochalasin D (Sigma-Aldrich) at final concentration of 20 μg/ml was used as an inhibitor for phagocytosis. Additionally, cytospin preparations underwent nuclear staining by using mounting medium containing DAPI (Vectashield, Burlingame, CA) according to the manufacturer’s instructions, followed by microscopy (FSX100 Bio Imaging Navigator; Olympus, Tokyo, Japan) for analysis of phagocytosing cells.

Sorted lung neutrophils or macrophages (2 × 106 cells/ml) were washed and suspended in HBSS. For 2 h at 37°C, under 5% CO2, half of the cells were incubated in starving condition (in HBSS without supplement) alone, and the remaining cells were incubated in starving condition with chloroquine, an inhibitor of lysosomal acidification. To prepare whole-cell protein extracts for immunoblotting, cells were washed and lysed in cell lysis buffer (Medical and Biological Laboratories, Aichi, Japan). After incubation, partial cells were prepared for immunocytochemistry via cytospin. Autophagic flux was assessed by the difference in immunoblotting of autophagosome protein LC3-II expression in the presence and absence of chloroquine (25).

A DC protein assay kit (Bio-Rad Laboratories, Hercules, CA) was used to quantify neutrophil and macrophage proteins in LDS sample buffer (Life Technologies, Carlsbad, CA). A Multigel II mini 10/20 (Cosmo Bio, Tokyo, Japan) was used for electrophoresis. For immunoblotting, 10 μg of neutrophil or macrophage protein was used. After electrophoresis, proteins were transferred to a polyvinylidene difluoride membrane; membranes were blocked with with ECL prime blocking reagent (GE Healthcare, Buckinghamshire, U.K.) and incubated with anti-LC3b Ab (Abcam, Cambridge, U.K.) or anti–β-actin Ab (control; Cell Signaling Technology, Beverly, MA) as the primary Ab and HRP-conjugated anti-rabbit IgG Ab (GE Healthcare) as the secondary Ab. Bands were detected by chemiluminescence, and band intensities were measured by using ImageQuant TL software (GE Healthcare).

For immunocytochemistry, cytospin preparations of lung neutrophils treated with or without chloroquine were fixed with 4% paraformaldehyde. Cells were treated with permeabilization buffer (BD Biosciences), blocked with 3% FBS in PBS for 1 h, and incubated with anti-LC3 mAb (Medical and Biological Laboratories) as the primary Ab for 1 h and FITC-conjugated anti-mouse IgG1 (eBioscience) as the secondary Ab for 1 h. Nuclei were stained and cells were evaluated as for the phagocytosis assay.

Cigarette smoke extract (CSE) was prepared based on the method previously described (26). Briefly, smoke from four Marlboro cigarettes (Philip Morris, Victoria, Australia) was bubbled into 10 ml of PBS for 2 min per cigarette. Each cigarette contained 1.0 mg of nicotine, 12 mg of tar. Crude extract was filter sterilized using a 0.22-μm filter and frozen in aliquots until use. Alveolar macrophages and alveolar epithelial cells were incubated in medium containing 2% CSE for 24 h and then supernatants and mRNA were harvested. For induction of apoptosis on neutrophils, cells were incubated in medium containing 8 or 16% CSE for 1 h and then used for the measurement. For measurement of TNF-α in culture supernatants, a Quantikine mouse TNF-α ELISA kit (R&D Systems, Minneapolis, MN) was used according to the manufacturer’s instructions.

A Student t test was used for comparison of two groups, whereas ANOVA and a Tukey–Kramer honestly significant difference test were used for comparison among multiple groups (JMP Pro 11.2.0; SAS Institute, Cary, NC). The level of statistical significance was set at p < 0.05. Data were expressed as the mean ± SEM.

Using chest microcomputed tomography (micro-CT), we examined the morphologic changes of the lung. To quantify the volume of emphysematous changes, the ratio of the volume of low-attenuation regions to total lung volume was calculated as the percentage of low-attenuation regions according to acquired images (Fig. 1B). Compared with those in WT PPE plus LPS mice, emphysematous changes in the IRF3−/− PPE plus LPS group were significantly (p < 0.05) suppressed at 4 wk (Fig. 1C). Next, we analyzed the change in percentage of low attenuation area after LPS administration. In the WT group, LPS administration induced a significant increase in PPE-induced emphysema formation only at 4 and 6 wk (Fig. 1D). Histopathological examination at 6 wk showed enlarged alveolar spaces with destruction of alveolar septae in both the WT and IRF3−/− PPE plus LPS groups; however, according to our quantitative examination, the mean linear intercept of the IRF3−/− PPE plus LPS group was significantly lower than that of the WT group (Fig. 1E, 1F). These data suggest that LPS administration exacerbates the PPE-induced emphysematous change in the lung in an IRF3-dependent manner.

To examine the mechanisms underlying the distinct morphologic changes in the lung, we assessed the cell, cytokine, and chemokine profiles during acute inflammation induced by PPE plus LPS. The stimulation protocol is shown in Fig. 2A. Compared with WT mice, the PPE plus LPS group of IRF3−/− mice exhibited suppressed mRNA expression of MCP-1, TNF-α, MIP-1α, IP-10, and keratinocyte chemoattractant. Among these mediators, IP-10 expression in the PPE plus LPS group of WT mice was also higher than that in the PPE or LPS only WT group. In contrast, a significant difference of MIP-2 expression was not detected, and the concentration of IL-17A transcripts was higher in IRF3−/− mice than in WT mice (Fig. 2B, 2C).

Alternatively, compared with those in the PPE plus LPS group of WT mice, total cell and neutrophil counts were significantly increased in the BALF of the PPE plus LPS group of IRF3−/− mice (Fig. 3A). The numbers of total cells and neutrophils were greater in the PPE, LPS, and PPE plus LPS groups than in the PBS group. It is notable that LPS-induced augmentation of the number of total cells and neutrophils in BALF after PPE treatment was detected only in IRF3−/− mice (Fig. 3B). However, the augmentation was observed in macrophages both in WT and IRF3−/− mice. These data suggest that various mediators related to COPD were produced by PPE plus LPS treatment, and augmentation by LPS after PPE treatment was observed in IP-10 expression and cell number of macrophages. In IRF3−/− mice, multiple mediators were suppressed, resulting in the attenuated morphologic changes in these mice. Controversially, neutrophil recruitment to lung was somewhat increased during acute inflammation.

FIGURE 3.

IRF3 has a role in increased neutrophil recruitment in BALF in PPE plus LPS acute inflammation model. See Fig. 2 for the study design. (A) Differential counts of cell number from BALF in the PBS or PPE plus LPS group, and (B) the number of total cells and each cell type in each group are shown. Data represent mean ± SEM. *p < 0.05; n = 3 in the PBS group, n = 8–12 in the other groups.

FIGURE 3.

IRF3 has a role in increased neutrophil recruitment in BALF in PPE plus LPS acute inflammation model. See Fig. 2 for the study design. (A) Differential counts of cell number from BALF in the PBS or PPE plus LPS group, and (B) the number of total cells and each cell type in each group are shown. Data represent mean ± SEM. *p < 0.05; n = 3 in the PBS group, n = 8–12 in the other groups.

Close modal

To further investigate pathogenic roles of IRF3 in this exacerbation model, we isolated alveolar epithelial cells and alveolar or lung macrophages to examine their function. As for alveolar epithelial cells treated by PPE plus LPS in IRF3−/− mice, the expression of MCP-1 was significantly suppressed, although IP-10 expression showed no significant difference (Fig. 4A). Naive alveolar macrophages expressed IP-10 and MIP-1α in response to PPE plus LPS in WT mice, but their expression was both suppressed in IRF3−/− mice (Fig. 4B).

FIGURE 4.

IRF3−/− alveolar epithelial cells and naive macrophages show suppressed expression of mediators. (A) Alveolar epithelial cells from WT or IRF3−/− mice were harvested and cultured as described in 2Materials and Methods. Harvested mRNA after the stimulation of PPE plus LPS or PBS was used for quantitative PCR. The data were normalized by the expression in the PBS control. (B) Primary alveolar macrophages were harvested and cultured as described in 2Materials and Methods. Data represent mean ± SEM. *p < 0.05; each sample is derived from three to four mice.

FIGURE 4.

IRF3−/− alveolar epithelial cells and naive macrophages show suppressed expression of mediators. (A) Alveolar epithelial cells from WT or IRF3−/− mice were harvested and cultured as described in 2Materials and Methods. Harvested mRNA after the stimulation of PPE plus LPS or PBS was used for quantitative PCR. The data were normalized by the expression in the PBS control. (B) Primary alveolar macrophages were harvested and cultured as described in 2Materials and Methods. Data represent mean ± SEM. *p < 0.05; each sample is derived from three to four mice.

Close modal

We next isolated lung macrophages (considered to include inflammatory monocytes) from lungs treated by PPE plus LPS and examined their function. IP-10 expression was diminished in IRF3−/− lung macrophages; in contrast, MIP-1α expression showed no difference (Fig. 5A). Then, we assessed the phagocytic activity of harvested macrophages using fluorescent latex beads. FITC mean fluorescence intensity (MFI) and the proportion of FITC+ macrophages showed no significant change between the WT and the IRF3−/− group (Fig. 5B). The extent of its activity inhibited by the phagocytic inhibitor cytochalasin D in WT or IRF3−/− macrophages was 11 ± 1 and 9 ± 1%, respectively, which was also almost comparable. These data suggest the partial involvement of these cells in the distinct inflammatory mediator profiles induced by PPE plus LPS, and this effect was dependent on IRF3, although phagocytic activity appeared to be almost the same between WT and IRF3−/− lung macrophages.

FIGURE 5.

IRF3−/−-activated macrophages show suppressed expression of mediators but comparable phagocytic activity compared with WT counterparts. (A) Lung macrophages (including inflammatory monocytes) were sorted from the inflamed lung 1 d after PPE plus LPS treatment using CD11c and Gr-1 expression. mRNA expression level of each mediator was investigated by quantitative PCR. (B) Lung macrophages (2 × 105) were incubated with green fluorescent latex beads, and cellular fluorescence was measured by flow cytometry (B, upper panel) and fluorescence microscopy (B, lower panel). FITC MFI and FITC+ proportion of macrophages are shown. For fluorescence microscopy, macrophages preincubated with latex beads were stained with DAPI (blue) for nuclei. Original magnification, ×400. Data represent mean ± SEM and are shown as an average of three independent experiments. *p < 0.05.

FIGURE 5.

IRF3−/−-activated macrophages show suppressed expression of mediators but comparable phagocytic activity compared with WT counterparts. (A) Lung macrophages (including inflammatory monocytes) were sorted from the inflamed lung 1 d after PPE plus LPS treatment using CD11c and Gr-1 expression. mRNA expression level of each mediator was investigated by quantitative PCR. (B) Lung macrophages (2 × 105) were incubated with green fluorescent latex beads, and cellular fluorescence was measured by flow cytometry (B, upper panel) and fluorescence microscopy (B, lower panel). FITC MFI and FITC+ proportion of macrophages are shown. For fluorescence microscopy, macrophages preincubated with latex beads were stained with DAPI (blue) for nuclei. Original magnification, ×400. Data represent mean ± SEM and are shown as an average of three independent experiments. *p < 0.05.

Close modal

Given the apparently contradictory data of increased neutrophil counts but deceased morphologic changes in IRF3−/− mice, we hypothesized that the function of the neutrophils recruited to the lung due to PPE plus LPS treatment was altered in these mice. First, we investigated the profiles of mediators produced by neutrophils that recruited to the lung after PPE plus LPS treatment. IP-10 mRNA expression was significantly increased in lung neutrophils derived from PPE plus LPS–treated WT mice compared with those from LPS only–treated WT mice (Fig. 6A). In contrast, this augmentation was not induced in IRF3−/− mice (Fig. 6A). Regarding MIP-1α mRNA, the expression was significantly impaired in IRF3−/− mice compared with WT mice (Fig. 6A).

FIGURE 6.

IRF3−/− lung neutrophils show suppressed expression of mediators and exhibit reduced apoptotic or phagocytic activity. (A) Lung neutrophils were sorted from the inflamed lung 1 d after PPE plus LPS treatment as described in 2Materials and Methods, and mRNA expression level of each mediator was investigated by quantitative PCR. For comparison, neutrophils derived from LPS-treated lung were used. (B) Neutrophils (2.5 × 105) from PPE plus LPS–treated lung were stained with annexin-FITC (left panel) or incubated with MitoPT-JC1 (right panel), then measured by flow cytometry. Data were obtained by subtracting a baseline fluorescerent population without annexin-FITC. Proportion of annexin-FITC+ cells (left panel) or cells of disrupted mitochondrial membrane potential are shown. (C) Phagocytic activity of lung neutrophils (2 × 105) in the same manner as measured in Fig. 5C. Original magnification, ×400. Data represent mean ± SEM and are shown as an average of three independent experiments. *p < 0.05.

FIGURE 6.

IRF3−/− lung neutrophils show suppressed expression of mediators and exhibit reduced apoptotic or phagocytic activity. (A) Lung neutrophils were sorted from the inflamed lung 1 d after PPE plus LPS treatment as described in 2Materials and Methods, and mRNA expression level of each mediator was investigated by quantitative PCR. For comparison, neutrophils derived from LPS-treated lung were used. (B) Neutrophils (2.5 × 105) from PPE plus LPS–treated lung were stained with annexin-FITC (left panel) or incubated with MitoPT-JC1 (right panel), then measured by flow cytometry. Data were obtained by subtracting a baseline fluorescerent population without annexin-FITC. Proportion of annexin-FITC+ cells (left panel) or cells of disrupted mitochondrial membrane potential are shown. (C) Phagocytic activity of lung neutrophils (2 × 105) in the same manner as measured in Fig. 5C. Original magnification, ×400. Data represent mean ± SEM and are shown as an average of three independent experiments. *p < 0.05.

Close modal

Next, to investigate the survival ability of lung neutrophils, we examined apoptotic activity, as measured by using annexin V and the detection of the change in mitochondrial membrane potential. The apoptotic activity of neutrophils from mice treated with PPE plus LPS was significantly reduced in the IRF3−/− group compared with the WT group in both assays (Fig. 6B).

Then we assessed the phagocytic activity of harvested neutrophils in the same way used for lung macrophages. FITC MFI and the proportion of FITC+ neutrophils indicated that neutrophils of the WT group exhibited elevated phagocytic activity of latex beads than did those of the IRF3−/− group (Fig. 6C). The extent of their phagocytic activity inhibited by cytochalasin D was 17 ± 3% in WT neutrophils, whereas almost none of this effect was observed in IRF3−/− neutrophils. Therefore, in the IRF3−/− mice treated with PPE plus LPS, lung neutrophils exhibited suppressed phagocytic activity.

Finally, using the difference in the production of autophagy-related protein LC3-II in the presence or absence of chloroquine (ΔLC3-II), we investigated the autophagic activity of lung neutrophils and macrophages. Lung neutrophils from both WT and IRF3−/− mice treated with PPE plus LPS showed increased LC3-II in the presence of chloroquine, whereas this difference was indistinct in the lung macrophages isolated from these two groups (Fig. 7A). As detected by Western blotting, ΔLC3-II of lung neutrophils was significantly greater in WT mice than in IRF3−/− mice (Fig. 7B). Additionally, incubation with chloroquine increased LC3B expression in lung neutrophils from the WT group but not from the IRF3−/− group (Fig. 7C). Therefore, treatment with PPE plus LPS activated autophagy in the lung neutrophils from WT mice, but that activity was suppressed in IRF3−/− mice. These data, together with the production of chemokines, suggest that various functions of neutrophils were altered in neutrophils derived from PPE plus LPS–treated IRF3−/− mice.

FIGURE 7.

Autophagic activities of lung neutrophils are suppressed in IRF3−/− PPE plus LPS mice. Lung neutrophils or macrophages obtained from WT or IRF3−/− mice sensitized by PPE plus LPS were incubated under starving conditions with or without chloroquine (CQ). (A) Western blotting analysis of LC3B protein from lysates of these cells. Autophagic flux is measured by LC3-II turnover as the difference between with and without chloroquine. Left, lung neutrophils; right, lung macrophages. (B) Quantification by densitometry of ΔLC3-II obtained from (A). (C) Immunocytochemistry staining was performed with neutrophils. Cytoplasmic LC3B (green, arrows) and nuclei (blue) are indicated. Original magnification, ×400. Data represent mean ± SEM and are shown from three to five independent experiments. *p < 0.05.

FIGURE 7.

Autophagic activities of lung neutrophils are suppressed in IRF3−/− PPE plus LPS mice. Lung neutrophils or macrophages obtained from WT or IRF3−/− mice sensitized by PPE plus LPS were incubated under starving conditions with or without chloroquine (CQ). (A) Western blotting analysis of LC3B protein from lysates of these cells. Autophagic flux is measured by LC3-II turnover as the difference between with and without chloroquine. Left, lung neutrophils; right, lung macrophages. (B) Quantification by densitometry of ΔLC3-II obtained from (A). (C) Immunocytochemistry staining was performed with neutrophils. Cytoplasmic LC3B (green, arrows) and nuclei (blue) are indicated. Original magnification, ×400. Data represent mean ± SEM and are shown from three to five independent experiments. *p < 0.05.

Close modal

As cigarette smoke is profoundly involved in the pathogenesis of COPD, we examined the effect of CSE on alveolar macrophages, alveolar epithetilal cells, and lung neutrophils derived from PPE plus LPS–treated mice. Alveolar macrophages showed suppressed TNF-α production by 2% CSE administration in both WT and IRF3−/− cell types (Fig. 8A). A similar result was detected in the expression of MCP-1 mRNA in alveolar epithetilal cells (Fig. 8B). As for neutrophils, we measured apoptotic activity using annexin, the change in mitochondrial membrane potential, and necrotic activity using PI staining. At 4% CSE, no significant change was detected in all assays compared with PBS-treated neutrophils (data not shown). In contrast, at 8% CSE, the apoptotic activity was significantly increased both in WT and IRF3−/− group, and 16% CSE showed further increased apoptotic activity (Fig. 8C). In all conditions, WT neutrophils exhibited greater apoptotic activity compared with IRF3−/− neutrophils. Additionally, necrotic activity detected by PI showed only in WT neutrophils at 16% CSE (Fig. 8C). These results indicate that the CSE effect on the apoptotic and necrotic change may be different between WT and IRF3−/− neutrophils, although its effect on the production of chemokines showed similar change between both strains.

FIGURE 8.

CSE induces suppression of some mediators, and IRF3−/− neutrophils are more tolerable in cell death induced by dense CSE. (A) Naive alveolar macrophages from WT or IRF3−/− mice were incubated with 2% CSE or PBS for 24 h. Supernatants were collected and used for measurement of TNF-α by ELISA. (B) Alveolar epithelial cells from WT or IRF3−/− mice were incubated with 2% CSE or PBS for 24 h. Harvested mRNA after stimulation was used for quantitative PCR. (C) Neutrophils (2.5 × 105) from PPE plus LPS–treated lung stained without PI were incubated with 8 or 16% CSE or PBS for 1 h and thereafter used for apoptosis assay in the same manner described in Fig. 6B. PI staining was also performed with annexin V simultaneously and then measured by flow cytometry. Proportion of annexin-FITC+ cells (left panel), cells of disrupted mitochondrial membrane potential (middle panel), and PI+ cells (right panel) are shown. (A and B) Each sample is derived from three to four mice. (C) Data are shown as an average of three independent experiments. Data represent mean ± SEM. *p < 0.05.

FIGURE 8.

CSE induces suppression of some mediators, and IRF3−/− neutrophils are more tolerable in cell death induced by dense CSE. (A) Naive alveolar macrophages from WT or IRF3−/− mice were incubated with 2% CSE or PBS for 24 h. Supernatants were collected and used for measurement of TNF-α by ELISA. (B) Alveolar epithelial cells from WT or IRF3−/− mice were incubated with 2% CSE or PBS for 24 h. Harvested mRNA after stimulation was used for quantitative PCR. (C) Neutrophils (2.5 × 105) from PPE plus LPS–treated lung stained without PI were incubated with 8 or 16% CSE or PBS for 1 h and thereafter used for apoptosis assay in the same manner described in Fig. 6B. PI staining was also performed with annexin V simultaneously and then measured by flow cytometry. Proportion of annexin-FITC+ cells (left panel), cells of disrupted mitochondrial membrane potential (middle panel), and PI+ cells (right panel) are shown. (A and B) Each sample is derived from three to four mice. (C) Data are shown as an average of three independent experiments. Data represent mean ± SEM. *p < 0.05.

Close modal

In this study, we demonstrated a novel role of IRF3 in a COPD exacerbation model. According to our morphologic studies using micro-CT and histopathology, the IRF3-associated pathway contributed to the additional emphysematous formation induced after LPS stimulation.

Micro-CT revealed that a single dose of LPS after PPE administration causes more severe emphysema at 12 wk after treatment than does PPE administration alone, suggesting that the sequential use of PPE followed by LPS in mice generates a useful model of COPD exacerbation (18) In our model, we observed additional changes of emphysema formation earlier after LPS administration than found in the previous study (Fig. 1).

The pathogenesis of COPD exacerbation is complicated and many types of the cells are involved, depending on the trigger of exacerbation. In a viral exacerbation model using dsRNA polyinosinic-polycytidylic acid or respiratory viruses, numbers of macrophages, neutrophils, and lymphocytes in BALF are elevated, and many mediators such as type 1 IFNs, IL-18, IP-10, and TNF-α are induced, leading to progression of emphysema or airway hyperresponsiveness (27, 28). This effect is partially dependent on TLR3 (27). Meanwhile, LPS or bacteria are used for bacterial exacerbation (29). LPS is recognized by TLR4, and subsequently many inflammatory mediators that are augmented in COPD patients are released by LPS stimulation (3036).

In this LPS-triggered exacerbation model, a previous study demonstrated that CD8+ T cells infiltrated into the alveolar space and might be involved in pathogenesis (18). In the present study, neutrophils and macrophages in BALF are increased by PPE plus LPS treatment in WT and IRF3−/− mice. Some inflammatory mediators in the mice treated with PPE plus LPS were dependent on IRF3. Among these mediators, IP-10 reportedly is IRF3-dependent (37). The chemokine IP-10 is known to be involved in a murine model of acute lung injury (38). Although the expression of IP-10 was increased in WT mice treated with PPE plus LPS compared with PPE or LPS only treated group, the neutralization of IP-10 by using anti–IP-10 Ab did not significantly modulate emphysematous formation after PPE plus LPS administration (T. Ishii and N. Yamashita, unpublished observations). In IRF3−/− mice, other mediators were also suppressed; these include TNF-α, which is dependent on the MyD88–NF-κB pathway (39). This finding might be attributed to cooperation between NF-κB subunit p35 and IRF3 as an enhanceosome (1214). In contrast, in IRF3−/− mice, MIP-2 and IL-17A, which are associated with neutrophil recruitment, did not decrease compared with similarly treated WT mice. In a previous study, IRF3 interacted with retinoic acid receptor–related orphan receptor γt, a transcriptional regulator of differentiation for Th17 cells, and suppressed IL-17A production (40). This interaction may reflect overexpression of IL-17A and increased neutrophils in BALF.

As the origin of these mediators, many types of cells, including alveolar epithelial cells, macrophages, and neutrophils, were involved in our study. IRF3 is constitutively expressed both in immune cells and structural cells (11). These cells are known to be involved in COPD pathogenesis (41). In our observation, we presumed that alveolar epithelial cells and alveolar macrophages are involved in initial inflammation, and then activated macrophages, including inflammatory monocytes, and neutrophils augment the mediators, although other cell types, including lymphocytes and endothelial cells, may also be involved, and an analysis using bone marrow chimeric mice would provide a deeper insight. Elastolytic protease such as matrix metalloproteinases and cathepsins may also be involved in COPD pathogenesis, but in some models no apparent relationship is observed (42). We did not detect any difference between two strains (data not shown).

Neutrophils also have a pivotal role in COPD, and neutrophilic airway inflammation correlates with the decline in lung function (43). In the present study, despite less prominent morphologic changes of emphysema, neutrophil recruitment increased in PPE plus LPS–treated IRF3−/− mice. Lung neutrophils isolated from IRF3−/− mice exhibited significantly low levels of mediator production, apoptosis, phagocytosis, and autophagy. Apoptotic neutrophils are associated with COPD in two aspects (44): 1) reduced apoptotic activity, which potentiates prolonged neutrophil inflammation, and 2) a high proportion of apoptotic neutrophils, which results in secondary necrosis, releasing proinflammatory components into the air spaces (45). In the present study, we speculated that suppression of both the apoptotic and perhaps necrotic activities of IRF3−/− neutrophils contribute to the increased neutrophil number in BALF, resulting in the decrease in the COPD morphologic changes despite the augmented neutrophil number in IRF3−/− mice.

The influence of the phagocytic activity of neutrophils on COPD remains unclear; however, in a study using Haemophilus influenzae, the phagocytosis of bacteria by neutrophils led to cellular necrosis due to the release of toxic granules from cytotoxic neutrophils, thus augmenting COPD (46). We similarly used phagocytic activity as a marker of neutrophil function. The attenuated phagocytosis of bacteria by inhibition of IRF3 has also been reported in mouse peritoneal macrophages (47) and human monocytes (48). Our findings suggest that nonspecific phagocytosis by neutrophils differed between WT and IRF3−/− mice and might be associated with necrosis and tissue damage via the release of the cytoplasmic contents of neutrophils.

Autophagy and apoptosis are related to diseases in both inductive and suppressive mechanisms (49). Autophagy may be associated with COPD pathogenesis in many aspects, including bronchial and alveolar epithelial cells and macrophages (50, 51). For example, elevated autophagy in lung epithelial cells promotes apoptosis through LC3b activation (51). Suppression of autophagy in neutrophils is suggested to be involved in autoimmune encephalomyelitis (52) and LPS-induced breakdown of the blood–brain barrier (52). To our knowledge, we are the first to describe alteration of autophagy in neutrophils in a COPD mouse model, suggesting that progressive autophagy and apoptosis in lung neutrophils may influence COPD exacerbation. The induced autophagy may be relevant to apoptosis and phagocytosis mutually, although it has been demonstrated that autophagy is also involved in differentiation and proliferation of neutrophils in lymphoid organs (53). Because autophagy is also activated by infection with dsDNA virus such as HSV1 through the cytoplasmic adaptor protein stimulator of IFN genes protein that is associated with IRF3 activation (54, 55), the observed difference in autophagic activity may be applicable in some virus-associated exacerbation model.

As cigarette smoking has an essential role in COPD pathogenesis, we examined the effect of CSE on various cell types. CSE suppressed expression of chemokines in alveolar epithelial cells and macrophages as previously described (56), and these suppressions were similar in IRF3−/− counterparts. Cigarette smoke exposure can increase cellular stress, leading to apoptosis in many types of lung cells (57). In our experiments, CSE induced suppressed apoptosis and necrosis in IRF3−/− neutrophils compared with WT neutrophils, indicating that IRF3 affects neutrophil function induced by CSE.

In summary, in this study we clarified the role of IRF3 in the pathogenesis of LPS-triggered COPD exacerbation. IRF3 knockout in mice decreases several inflammatory mediators that are involved in COPD in multiple types of lung cells, although impaired neutrophil function is observed in IRF3−/− mice. Abs that target specific cytokines are expensive therapeutics and therefore have limited utility. In contrast, targeting a single transcriptional factor that influences multiple mediators important for the disease pathogenesis, such as IRF3, may be a promising, practical solution.

We thank Prof. T. Taniguchi (Department of Molecular Immunology, Institute of Industrial Science, University of Tokyo, Tokyo, Japan) and the RIKEN BioResource Center for providing IRF3−/− mice. We thank Emi Kishimoto for excellent secretarial assistance.

This study was supported in part by Ministry of Education, Culture, Sports, Science and Technology, Japan Grant-in-Aid for Scientific Research 25460660 (to Naomi Yamashita).

Abbreviations used in this article:

BALF

bronchoalveolar lavage fluid

COPD

chronic obstructive pulmonary disease

CSE

cigarette smoke extract

CT

computed tomography

HU

Hounsfield unit

IP-10

IFN-γ–inducible protein-10

IRF3

IFN regulatory factor-3

MFI

mean fluorescence intensity

micro-CT

microcomputed tomography

PI

propidium iodide

PPE

porcine pancreatic elastase

TRIF

Toll/IL-1R domain–containing adaptor inducing IFN-β

WT

wild-type.

1
Buist
A. S.
,
McBurnie
M. A.
,
Vollmer
W. M.
,
Gillespie
S.
,
Burney
P.
,
Mannino
D. M.
,
Menezes
A. M.
,
Sullivan
S. D.
,
Lee
T. A.
,
Weiss
K. B.
, et al
BOLD Collaborative Research Group
.
2007
.
International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study.
Lancet
370
:
741
750
.
2
Lopez
A. D.
,
Mathers
C. D.
,
Ezzati
M.
,
Jamison
D. T.
,
Murray
C. J.
.
2006
.
Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data.
Lancet
367
:
1747
1757
.
3
Vestbo
J.
,
Hurd
S. S.
,
Agustí
A. G.
,
Jones
P. W.
,
Vogelmeier
C.
,
Anzueto
A.
,
Barnes
P. J.
,
Fabbri
L. M.
,
Martinez
F. J.
,
Nishimura
M.
, et al
.
2013
.
Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary.
Am. J. Respir. Crit. Care Med.
187
:
347
365
.
4
Spencer
S.
,
Calverley
P. M.
,
Burge
P. S.
,
Jones
P. W.
.
2004
.
Impact of preventing exacerbations on deterioration of health status in COPD.
Eur. Respir. J.
23
:
698
702
.
5
Donaldson
G. C.
,
Seemungal
T. A.
,
Bhowmik
A.
,
Wedzicha
J. A.
.
2002
.
Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease.
Thorax
57
:
847
852
.
6
Connors
Jr.
 A. F.
,
Dawson
N. V.
,
Thomas
C.
,
Harrell
F. E.
 Jr.
,
Desbiens
N.
,
Fulkerson
W. J.
,
Kussin
P.
,
Bellamy
P.
,
Goldman
L.
,
Knaus
W. A.
.
1996
.
Outcomes following acute exacerbation of severe chronic obstructive lung disease. The SUPPORT investigators (study to understand prognoses and preferences for outcomes and risks of treatments)
.
Am. J. Respir. Crit. Care Med.
154
:
959
967
.
7
Akira
S.
,
Uematsu
S.
,
Takeuchi
O.
.
2006
.
Pathogen recognition and innate immunity.
Cell
124
:
783
801
.
8
He
J. Q.
,
Foreman
M. G.
,
Shumansky
K.
,
Zhang
X.
,
Akhabir
L.
,
Sin
D. D.
,
Man
S. F.
,
DeMeo
D. L.
,
Litonjua
A. A.
,
Silverman
E. K.
, et al
.
2009
.
Associations of IL6 polymorphisms with lung function decline and COPD.
Thorax
64
:
698
704
.
9
Sakao
S.
,
Tatsumi
K.
,
Igari
H.
,
Shino
Y.
,
Shirasawa
H.
,
Kuriyama
T.
.
2001
.
Association of tumor necrosis factor α gene promoter polymorphism with the presence of chronic obstructive pulmonary disease.
Am. J. Respir. Crit. Care Med.
163
:
420
422
.
10
Kawai
T.
,
Takeuchi
O.
,
Fujita
T.
,
Inoue
J.
,
Mühlradt
P. F.
,
Sato
S.
,
Hoshino
K.
,
Akira
S.
.
2001
.
Lipopolysaccharide stimulates the MyD88-independent pathway and results in activation of IFN-regulatory factor 3 and the expression of a subset of lipopolysaccharide-inducible genes.
J. Immunol.
167
:
5887
5894
.
11
Honda
K.
,
Taniguchi
T.
.
2006
.
IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors.
Nat. Rev. Immunol.
6
:
644
658
.
12
Leung
T. H.
,
Hoffmann
A.
,
Baltimore
D.
.
2004
.
One nucleotide in a κB site can determine cofactor specificity for NF-κB dimers.
Cell
118
:
453
464
.
13
Ogawa
S.
,
Lozach
J.
,
Benner
C.
,
Pascual
G.
,
Tangirala
R. K.
,
Westin
S.
,
Hoffmann
A.
,
Subramaniam
S.
,
David
M.
,
Rosenfeld
M. G.
,
Glass
C. K.
.
2005
.
Molecular determinants of crosstalk between nuclear receptors and Toll-like receptors.
Cell
122
:
707
721
.
14
Wietek
C.
,
Miggin
S. M.
,
Jefferies
C. A.
,
O’Neill
L. A.
.
2003
.
Interferon regulatory factor-3-mediated activation of the interferon-sensitive response element by Toll-like receptor (TLR) 4 but not TLR3 requires the p65 subunit of NF-κ.
J. Biol. Chem.
278
:
50923
50931
.
15
Edwards
M. R.
,
Bartlett
N. W.
,
Clarke
D.
,
Birrell
M.
,
Belvisi
M.
,
Johnston
S. L.
.
2009
.
Targeting the NF-κB pathway in asthma and chronic obstructive pulmonary disease.
Pharmacol. Ther.
121
:
1
13
.
16
Sato
M.
,
Suemori
H.
,
Hata
N.
,
Asagiri
M.
,
Ogasawara
K.
,
Nakao
K.
,
Nakaya
T.
,
Katsuki
M.
,
Noguchi
S.
,
Tanaka
N.
,
Taniguchi
T.
.
2000
.
Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-α/β gene induction.
Immunity
13
:
539
548
.
17
Kawakami
M.
,
Matsuo
Y.
,
Yoshiura
K.
,
Nagase
T.
,
Yamashita
N.
.
2008
.
Sequential and quantitative analysis of a murine model of elastase-induced emphysema.
Biol. Pharm. Bull.
31
:
1434
1438
.
18
Kobayashi
S.
,
Fujinawa
R.
,
Ota
F.
,
Kobayashi
S.
,
Angata
T.
,
Ueno
M.
,
Maeno
T.
,
Kitazume
S.
,
Yoshida
K.
,
Ishii
T.
, et al
.
2013
.
A single dose of lipopolysaccharide into mice with emphysema mimics human chronic obstructive pulmonary disease exacerbation as assessed by micro-computed tomography.
Am. J. Respir. Cell Mol. Biol.
49
:
971
977
.
19
Robbesom
A. A.
,
Versteeg
E. M.
,
Veerkamp
J. H.
,
van Krieken
J. H.
,
Bulten
H. J.
,
Smits
H. T.
,
Willems
L. N.
,
van Herwaarden
C. L.
,
Dekhuijzen
P. N.
,
van Kuppevelt
T. H.
.
2003
.
Morphological quantification of emphysema in small human lung specimens: comparison of methods and relation with clinical data.
Mod. Pathol.
16
:
1
7
.
20
Niikura
Y.
,
Ishii
T.
,
Hosoki
K.
,
Nagase
T.
,
Yamashita
N.
.
2015
.
Ovary-dependent emphysema augmentation and osteopontin induction in adult female mice.
Biochem. Biophys. Res. Commun.
461
:
642
647
.
21
Zhang, X., R. Goncalves, and D. M. Mosser. 2008. The isolation and characterization of murine macrophages. Curr. Protoc. Immunol. Chapter 14: Unit 14.11. doi:10.1002/0471142735.im1401s83
22
Dobbs
L. G.
1990
.
Isolation and culture of alveolar type II cells.
Am. J. Physiol.
258
:
L134
L147
.
23
Corti
M.
,
Brody
A. R.
,
Harrison
J. H.
.
1996
.
Isolation and primary culture of murine alveolar type II cells.
Am. J. Respir. Cell Mol. Biol.
14
:
309
315
.
24
Zaynagetdinov
R.
,
Sherrill
T. P.
,
Kendall
P. L.
,
Segal
B. H.
,
Weller
K. P.
,
Tighe
R. M.
,
Blackwell
T. S.
.
2013
.
Identification of myeloid cell subsets in murine lungs using flow cytometry.
Am. J. Respir. Cell Mol. Biol.
49
:
180
189
.
25
Mizushima
N.
,
Yoshimori
T.
,
Levine
B.
.
2010
.
Methods in mammalian autophagy research.
Cell
140
:
313
326
.
26
Mio
T.
,
Romberger
D. J.
,
Thompson
A. B.
,
Robbins
R. A.
,
Heires
A.
,
Rennard
S. I.
.
1997
.
Cigarette smoke induces interleukin-8 release from human bronchial epithelial cells.
Am. J. Respir. Crit. Care Med.
155
:
1770
1776
.
27
Kang
M. J.
,
Lee
C. G.
,
Lee
J. Y.
,
Dela Cruz
C. S.
,
Chen
Z. J.
,
Enelow
R.
,
Elias
J. A.
.
2008
.
Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice.
J. Clin. Invest.
118
:
2771
2784
.
28
Singanayagam
A.
,
Glanville
N.
,
Walton
R. P.
,
Aniscenko
J.
,
Pearson
R. M.
,
Pinkerton
J. W.
,
Horvat
J. C.
,
Hansbro
P. M.
,
Bartlett
N. W.
,
Johnston
S. L.
.
2015
.
A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD.
Clin. Sci.
129
:
245
258
.
29
Ganesan
S.
,
Faris
A. N.
,
Comstock
A. T.
,
Sonstein
J.
,
Curtis
J. L.
,
Sajjan
U. S.
.
2012
.
Elastase/LPS-exposed mice exhibit impaired innate immune responses to bacterial challenge: role of scavenger receptor A.
Am. J. Pathol.
180
:
61
72
.
30
Martin
T. R.
2000
.
Recognition of bacterial endotoxin in the lungs.
Am. J. Respir. Cell Mol. Biol.
23
:
128
132
.
31
Bafadhel
M.
,
McKenna
S.
,
Terry
S.
,
Mistry
V.
,
Reid
C.
,
Haldar
P.
,
McCormick
M.
,
Haldar
K.
,
Kebadze
T.
,
Duvoix
A.
, et al
.
2011
.
Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers.
Am. J. Respir. Crit. Care Med.
184
:
662
671
.
32
Capelli
A.
,
Di Stefano
A.
,
Gnemmi
I.
,
Balbo
P.
,
Cerutti
C. G.
,
Balbi
B.
,
Lusuardi
M.
,
Donner
C. F.
.
1999
.
Increased MCP-1 and MIP-1beta in bronchoalveolar lavage fluid of chronic bronchitics.
Eur. Respir. J.
14
:
160
165
.
33
Keatings
V. M.
,
Collins
P. D.
,
Scott
D. M.
,
Barnes
P. J.
.
1996
.
Differences in interleukin-8 and tumor necrosis factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or asthma.
Am. J. Respir. Crit. Care Med.
153
:
530
534
.
34
Ravi
A. K.
,
Khurana
S.
,
Lemon
J.
,
Plumb
J.
,
Booth
G.
,
Healy
L.
,
Catley
M.
,
Vestbo
J.
,
Singh
D.
.
2014
.
Increased levels of soluble interleukin-6 receptor and CCL3 in COPD sputum.
Respir. Res.
15
:
103
.
35
Saetta
M.
,
Mariani
M.
,
Panina-Bordignon
P.
,
Turato
G.
,
Buonsanti
C.
,
Baraldo
S.
,
Bellettato
C. M.
,
Papi
A.
,
Corbetta
L.
,
Zuin
R.
, et al
.
2002
.
Increased expression of the chemokine receptor CXCR3 and its ligand CXCL10 in peripheral airways of smokers with chronic obstructive pulmonary disease.
Am. J. Respir. Crit. Care Med.
165
:
1404
1409
.
36
Shanley
T. P.
,
Schmal
H.
,
Friedl
H. P.
,
Jones
M. L.
,
Ward
P. A.
.
1995
.
Role of macrophage inflammatory protein-1 alpha (MIP-1 alpha) in acute lung injury in rats.
J. Immunol.
154
:
4793
4802
.
37
Sakaguchi
S.
,
Negishi
H.
,
Asagiri
M.
,
Nakajima
C.
,
Mizutani
T.
,
Takaoka
A.
,
Honda
K.
,
Taniguchi
T.
.
2003
.
Essential role of IRF-3 in lipopolysaccharide-induced interferon-β gene expression and endotoxin shock.
Biochem. Biophys. Res. Commun.
306
:
860
866
.
38
Ichikawa
A.
,
Kuba
K.
,
Morita
M.
,
Chida
S.
,
Tezuka
H.
,
Hara
H.
,
Sasaki
T.
,
Ohteki
T.
,
Ranieri
V. M.
,
dos Santos
C. C.
, et al
.
2013
.
CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin.
Am. J. Respir. Crit. Care Med.
187
:
65
77
.
39
Kawai
T.
,
Adachi
O.
,
Ogawa
T.
,
Takeda
K.
,
Akira
S.
.
1999
.
Unresponsiveness of MyD88-deficient mice to endotoxin.
Immunity
11
:
115
122
.
40
Ysebrant de Lendonck
L.
,
Tonon
S.
,
Nguyen
M.
,
Vandevenne
P.
,
Welsby
I.
,
Martinet
V.
,
Molle
C.
,
Charbonnier
L. M.
,
Leo
O.
,
Goriely
S.
.
2013
.
Interferon regulatory factor 3 controls interleukin-17 expression in CD8 T lymphocytes.
Proc. Natl. Acad. Sci. USA
110
:
E3189
E3197
.
41
Barnes
P. J.
2016
.
Inflammatory mechanisms in patients with chronic obstructive pulmonary disease.
J. Allergy Clin. Immunol.
138
:
16
27
.
42
Atkinson
J. J.
,
Lutey
B. A.
,
Suzuki
Y.
,
Toennies
H. M.
,
Kelley
D. G.
,
Kobayashi
D. K.
,
Ijem
W. G.
,
Deslee
G.
,
Moore
C. H.
,
Jacobs
M. E.
, et al
.
2011
.
The role of matrix metalloproteinase-9 in cigarette smoke-induced emphysema.
Am. J. Respir. Crit. Care Med.
183
:
876
884
.
43
O’Donnell
R. A.
,
Peebles
C.
,
Ward
J. A.
,
Daraker
A.
,
Angco
G.
,
Broberg
P.
,
Pierrou
S.
,
Lund
J.
,
Holgate
S. T.
,
Davies
D. E.
, et al
.
2004
.
Relationship between peripheral airway dysfunction, airway obstruction, and neutrophilic inflammation in COPD.
Thorax
59
:
837
842
.
44
Hoenderdos
K.
,
Condliffe
A.
.
2013
.
The neutrophil in chronic obstructive pulmonary disease.
Am. J. Respir. Cell Mol. Biol.
48
:
531
539
.
45
Borges
V. M.
,
Vandivier
R. W.
,
McPhillips
K. A.
,
Kench
J. A.
,
Morimoto
K.
,
Groshong
S. D.
,
Richens
T. R.
,
Graham
B. B.
,
Muldrow
A. M.
,
Van Heule
L.
, et al
.
2009
.
TNFα inhibits apoptotic cell clearance in the lung, exacerbating acute inflammation.
Am. J. Physiol. Lung Cell. Mol. Physiol.
297
:
L586
L595
.
46
Naylor
E. J.
,
Bakstad
D.
,
Biffen
M.
,
Thong
B.
,
Calverley
P.
,
Scott
S.
,
Hart
C. A.
,
Moots
R. J.
,
Edwards
S. W.
.
2007
.
Haemophilus influenzae induces neutrophil necrosis: a role in chronic obstructive pulmonary disease?
Am. J. Respir. Cell Mol. Biol.
37
:
135
143
.
47
Deng
T.
,
Feng
X.
,
Liu
P.
,
Yan
K.
,
Chen
Y.
,
Han
D.
.
2013
.
Toll-like receptor 3 activation differentially regulates phagocytosis of bacteria and apoptotic neutrophils by mouse peritoneal macrophages.
Immunol. Cell Biol.
91
:
52
59
.
48
Husebye
H.
,
Aune
M. H.
,
Stenvik
J.
,
Samstad
E.
,
Skjeldal
F.
,
Halaas
O.
,
Nilsen
N. J.
,
Stenmark
H.
,
Latz
E.
,
Lien
E.
, et al
.
2010
.
The Rab11a GTPase controls Toll-like receptor 4-induced activation of interferon regulatory factor-3 on phagosomes.
Immunity
33
:
583
596
.
49
Mariño
G.
,
Niso-Santano
M.
,
Baehrecke
E. H.
,
Kroemer
G.
.
2014
.
Self-consumption: the interplay of autophagy and apoptosis.
Nat. Rev. Mol. Cell Biol.
15
:
81
94
.
50
Ryter
S. W.
,
Choi
A. M.
.
2015
.
Autophagy in lung disease pathogenesis and therapeutics.
Redox Biol.
4
:
215
225
.
51
Chen
Z. H.
,
Lam
H. C.
,
Jin
Y.
,
Kim
H. P.
,
Cao
J.
,
Lee
S. J.
,
Ifedigbo
E.
,
Parameswaran
H.
,
Ryter
S. W.
,
Choi
A. M.
.
2010
.
Autophagy protein microtubule-associated protein 1 light chain-3B (LC3B) activates extrinsic apoptosis during cigarette smoke-induced emphysema.
Proc. Natl. Acad. Sci. USA
107
:
18880
18885
.
52
Bhattacharya
A.
,
Wei
Q.
,
Shin
J. N.
,
Abdel Fattah
E.
,
Bonilla
D. L.
,
Xiang
Q.
,
Eissa
N. T.
.
2015
.
Autophagy is required for neutrophil-mediated inflammation.
Cell Rep.
12
:
1731
1739
.
53
Rožman
S.
,
Yousefi
S.
,
Oberson
K.
,
Kaufmann
T.
,
Benarafa
C.
,
Simon
H. U.
.
2015
.
The generation of neutrophils in the bone marrow is controlled by autophagy.
Cell Death Differ.
22
:
445
456
.
54
McFarlane
S.
,
Aitken
J.
,
Sutherland
J. S.
,
Nicholl
M. J.
,
Preston
V. G.
,
Preston
C. M.
.
2011
.
Early induction of autophagy in human fibroblasts after infection with human cytomegalovirus or herpes simplex virus 1.
J. Virol.
85
:
4212
4221
.
55
Deretic
V.
,
Saitoh
T.
,
Akira
S.
.
2013
.
Autophagy in infection, inflammation and immunity.
Nat. Rev. Immunol.
13
:
722
737
.
56
Witherden
I. R.
,
Vanden Bon
E. J.
,
Goldstraw
P.
,
Ratcliffe
C.
,
Pastorino
U.
,
Tetley
T. D.
.
2004
.
Primary human alveolar type II epithelial cell chemokine release: effects of cigarette smoke and neutrophil elastase.
Am. J. Respir. Cell Mol. Biol.
30
:
500
509
.
57
Tuder
R. M.
,
Petrache
I.
.
2012
.
Pathogenesis of chronic obstructive pulmonary disease.
J. Clin. Invest.
122
:
2749
2755
.

The authors have no financial conflicts of interest.