Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Macrophage PPAR-γ suppresses long-term lung fibrotic sequelae following acute influenza infection

  • Su Huang,

    Roles Investigation, Writing – original draft

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Nick P. Goplen,

    Roles Data curation, Writing – original draft

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Bibo Zhu,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • In Su Cheon,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Youngmin Son,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Zheng Wang,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Chaofan Li,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Qigang Dai,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Li Jiang,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Min Xiang,

    Roles Investigation

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Eva M. Carmona,

    Roles Conceptualization, Writing – review & editing

    Affiliations Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America, Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Robert Vassallo,

    Roles Conceptualization, Writing – review & editing

    Affiliations Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America, Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Andrew H. Limper,

    Roles Funding acquisition, Writing – review & editing

    Affiliation Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

  • Jie Sun

    Roles Conceptualization, Funding acquisition, Writing – original draft, Writing – review & editing

    sun.jie@mayo.edu

    Affiliations Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America, Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Rochester, Minnesota, United States of America

Abstract

Influenza virus causes a heterogeneous respiratory infectious disease ranging from self-limiting symptoms to non-resolving pathology in the lungs. Worldwide, seasonal influenza infections claim ~500,000 lives annually. Recent reports describe pathologic pulmonary sequelae that result in remodeling the architecture of lung parenchyma following respiratory infections. These dysfunctional recovery processes that disproportionately impact the elderly have been understudied. Macrophages are involved in tissue remodeling and are critical for survival of severe influenza infection. Here, we found intrinsic deficiency of the nuclear receptor PPAR-γ in myeloid cells delayed the resolution of pulmonary inflammation following influenza infection. Mice with myeloid cell-specific PPAR-γ deficiency subsequently presented with increased influenza-induced deposition of pulmonary collagen compared to control mice. This dysfunctional lung remodeling was progressive and sustained for at least 3 months following infection of mice with myeloid PPAR-γ deficiency. These progressive changes were accompanied by a pro-fibrotic gene signature from lung macrophages and preceded by deficiencies in activation of genes involved with damage repair. Importantly similar aberrant gene expression patterns were also found in a secondary analysis of a study where macrophages were isolated from patients with fibrotic interstitial lung disease. Quite unexpectedly, mice with PPAR-γ deficient macrophages were more resistant to bleomycin-induced weight loss whereas extracellular matrix deposition was unaffected compared to controls. Therefore PPAR-γ expression in macrophages may be a pathogen-specific limiter of organ recovery rather than a ubiquitous effector pathway in response to generic damage.

Introduction

Influenza virus infects 5–10% of adults and 20–30% of children worldwide [1]. Though seasonal influenza is often thought of as a rapidly cleared self-limiting infection, evidence is mounting that long-term sequelae can accumulate despite clearance of infectious viral particles [26]. A long-awaited plausible explanation for this prolonged immune stimulus may be the recent discovery of transcriptionally active viral RNA remnants. Depots of active RNA remnants are associated with pulmonary regions of chronic inflammation and lesions dispersed throughout the parenchyma and are observable months after clearance of influenza virus [2]. Prolonged activation of innate and/or adaptive immune system modulates severity and longevity of influenza-associated diseases [712]. Accordingly, activation of naïve and memory influenza-specific CD8 T cells can occur in lung draining lymph-nodes months after the lung is cleared of infectious virus particles [1315]. It is therefore plausible that chronic immune stimuli may be driving abnormal remodeling and pathologies in the lung following severe influenza infection.

Respiratory viruses, including influenza, have indeed been shown to cause abnormal remodeling of lung parenchyma dependent on the severity of infection [2, 1618]. The lungs are responsible for gas exchange; coupled with circulation, this results in supplying the body with oxygen for cellular respiration. Replacing pulmonary parenchyma (alveoli) with scar tissue therefore has functional consequences. Similar to physiological lung changes in chronic asthma models, matrix deposition, mucus secretion, and airway hypersensitivity to chronic stimuli have been demonstrated weeks after influenza clearance [2, 16, 17, 19]. Consequences of the pathologic sequelae include airway dysfunction, non-resolving inflammatory foci including tertiary lymphoid-like structures, and chronic lesions in the face of prolonged activation of the innate and adaptive immune systems. While these are now established chronic hallmarks of severe influenza infection in animal models with primate correlates [2, 1618], how the immune system tunes infection response from benign to pathologic remodeling of the lung parenchyma, is not clear.

The innate immune system responds to damage- and pathogen-associated molecular patterns (DAMPs and PAMPs) to simultaneously initiate anti-microbial defense, immune-response resolution, and repair mechanisms [4, 2022]. Repair of immune response induced damage occurs early after injury and is predisposed to dysfunction with age [2326]. Macrophages are a versatile and plastic cell type capable of homeostatic, pro-inflammatory, pro-repair, and fibrotic promoting functions all in the same immune response [2730]. Following influenza infection, the lung macrophage compartment is a heterogeneous population of monocyte-derived and embryonic derived cells with similar surface phenotypes, but distinct transcriptomes [31, 32].

PPAR-γ is a member of the nuclear hormone receptor superfamily. It is a lipid-sensing nuclear transcription factor with many intracellular ligands, including essential fatty acids and eicosanoids. PPAR-γ is known to regulate downstream gene expression mediating lipid storage and glucose homeostasis, cell growth, proliferation, and differentiation [33, 34]. Outside of adipocytes, under homoeostatic conditions, protein levels of PPAR-γ in the lung primarily derive from macrophages where it is required for their differentiation and normal recovery of influenza-infected animals [33, 35, 36]. Following influenza infection, PPAR-γ expression in alveolar macrophages is down-regulated in a manner dependent on intrinsic Type I interferon receptor signaling [37, 38]. PPAR-γ expression in AM was critical for the suppression of exaggerated antiviral and inflammatory responses of AM following influenza virus infection [37, 38]. As such, myeloid PPAR-γ deficiency resulted in enhanced acute host morbidity and delayed tissue recovery following influenza virus infection [37, 38].

Here, we demonstrate that myeloid cell PPAR-γ is important for recovery of influenza- induced injury. Whereas inflammatory and repair responses were largely resolved in wild type (WT) mice 30 days post-infection, they are sustained and delayed in myeloid PPAR-γ deficient mice. Subsequently, 60 and 90 days post-infection lungs from animals with PPAR-γ deficient macrophages exhibited a pro-fibrotic gene signature coupled with increased collagen deposition compared to wild type lungs. These findings of pro-fibrosis and pro-inflammatory signatures aligned well with macrophages from pulmonary fibrosis patients, who experience non-resolving progressive lung remodeling. However, mice with the same genetic defect exhibited enhanced recovery from bleomycin induced weight loss, with no effects on collagen deposition during the resolution phase of bleomycin-induced fibrosis. Thus, expression of PPAR-γ in macrophages limits pathogenic influenza-induced lung remodeling. Our data indicate existing agonists of PPAR-γ may alter the severity of influenza disease in the elderly that disproportionately experience long-term complications from seasonal flu infections.

Methods

Mouse and infection

WT C57/BL6, Lyz2-cre, Ppargfl/fl, were purchased from the Jackson Laboratory and bred in house. PpargΔLyz2 (PPAR-γ cKO) mice were generated by crossing Ppargfl/fl (WT) mice with Lyz2-cre mice. All mice housed in a specific pathogen-free environment. For influenza virus infection, influenza A/PR8/34 strain (~200 pfu/mouse) was diluted in FBS-free DMEM media (Corning) on ice and inoculated in ketamine-anesthetized mice through intranasal route as described before [39]. All animals are sex and age matched. Animal experiments and procedures were approved by Mayo Clinic Institutional Animal Care and Use Committee (IACUC study #: A00002027 and A00002246). Animals were sacrificed with overdose of ketamine followed with cervical dislocation as approved in the IACUC protocols. Animal numbers used in each experiment are indicated in the figure legends and total animal numbers used in the experiments were 124 mice. Infected animals are monitored daily. Experimental mice were unrestricted with food or water. Moribund mice, mice under severe respiratory distress or mice lost more than 30% of weight loss were humanely sacrificed according to the IACUC protocols listed.

Bleomycin treatment

Ppargfl/fl or PpargΔLyz2 mice were used at 12 weeks old. All mice were anesthetized with ketamine before bleomycin administration. Bleomycin was dissolved in saline and was given at the dosage of ~2.4 units per kilogram body weight (0.072 unit/mouse in 50 μl saline). Bleomycin was given by intratracheal instillation through an aerosolizer under visualization for each mouse [40]. After bleomycin administration, antisedan (atipamezole hydrochloride) was immediately given by IP (100μl diluted in 5 ml saline, 200μl for each mouse) and mice were put on heat pad with oxygen supply. Body weight was monitored in following days. At day 42 post bleomycin treatment, lungs were removed for determination of hydroxyproline levels in the whole lung.

Lung histopathology

Following euthanasia, mice were perfused with PBS (10 mL) via the right ventricle. 10% paraformaldehyde (PF) was then gently instilled into the lung and left inflated for 1 minute before excising and moving lobe to 10% PF for 48 hours followed by transfer to ethanol (70%). Samples were shipped to Mayo Clinic Histology Core Lab (Scottsdale, AZ) where they were embedded in paraffin and 5 um sections were cut for Hematoxylin and eosin (H&E) stain.

Quantitative RT-PCR

mRNA from cultured AM (pooled from multiple mice), in vivo sorted AM (pooled from multiple mice) or homogenates from individual lungs as indicated in the text was isolated with Total RNA purification kit (Sigma) and treated with DNase I (Invitrogen) as described [41]. Random primers (Invitrogen) and MMLV reverse transcriptase (Invitrogen) were used to synthesize first-strand cDNAs from equivalent amounts of RNA from each sample. RT-PCR was performed with Fast SYBR Green PCR Master Mix (Applied Biosystems). qPCR was conducted in duplicates in QuantStudio3 (Applied Bioscience). Data were generated with the comparative threshold cycle (Delta CT) method by normalizing to hypoxanthine phosphoribosyltransferase (HPRT). Sequences of primers will be provided if requested.

RT2 profiler PCR array

Total RNA from lung tissue or AM was extracted as described above. Equal amount of total RNA was used for the synthesis of first strand cDNA with kit from Qiagen. First strand cDNA was mixed with 2xFast SYBR Green Master Mix (Applied Bioscience) and water in a formula directed in the manual. 25 μl of the mixture was added into each well of the 96 well plate provided by manufacture. The wells in the plate include different primers in each well to detect 84 target genes, housekeeping genes, negative and positive control genes. qPCR was conducted in QuantStudio3 (Applied Bioscience). Obtained raw data was analyzed in software provided by Qiagen (accessible online on the website of Qiagen). Following the instruction step by step, upload Excel file, designating control group, select housekeeping gene to normalize result and calculate the relative expression quantity.

Hydroxyproline assay

Hydroxyproline assay for determining total lung collagen content was performed as previously reported [6]. Briefly, isolated whole lungs were cut into small pieces. Lung tissue was hydrolyzed in 1 ml 6 M HCL. The hydro lysate was cooled down to room temperature. Hydroxyproline standard solution was purchased from Sigma. Diluted standard solution were added into different wells in the assay plate for generation of standard curve. Freshly-made Chloramine-T Solution (2.0 ml N-Propanol, 0.282g Chloramine-T, 2.0ml H2O in 20ml citrate acetate buffer) was added into each well and the mixture was incubated at room temperature for 20 minutes. Then, 100 μl of fresh Ehrlich solution (4.5g 4-dimethylaminobenzaldehyde in 18.6 ml N-Propanol and 7.8 ml perchloric acid) was added into each well. Following that, the assay plate was placed at a 60°C oven for 60 minutes before reading at 560 nm wavelength in Thermax plate reader.

Flow cytometry and flow sorting

Fluorescence-conjugated FACS Abs were purchased from Biolegend or Tonbo Bioscience. Lung single cell suspensions were prepared according to previous publications [42, 43]. Lung cells were then washed with FACS buffer (PBS, 2 mM EDTA, 2% FBS, 0.09% Sodium Azide) and stained with CD64, Ly6G, CD11b, Siglec F, CD11c and MertK. Samples were collected on FACS Attune NXT flow cytometer (Life technologies) and analyzed using Flow Jo software (Tree Star). Total lung macrophages were defined as CD64+ Ly6G-. For cell sorting, total lung cells were pooled from infected WT or PPAR-γ cKO mice and stained with CD45, Ly6G, CD64 and MertK. CD45+ Ly6G- CD64+ MertK+ lung total macrophages (including resident macrophages and recruited macrophages) were sorted by BD FACSAria II. mRNA was extracted from sorted macrophages and realtime RT-PCR was performed as above.

Statistical analysis

Data are mean ± SEM of values from individual mice (in vivo experiments). Unpaired two-tailed Student’s t-test (two group comparison), Mann-Whitney test, One-way ANOVA with Tukey multiple comparison test (multi-group comparison) and Multiple t-tests (weight loss) were used to determine statistical significance by GraphPad Prism software. We consider P values < 0.05 as significant.

Data availability

The dataset we used has been publicly deposited in the GEO database: GSE49072 by Shi et al [44].

Results

Myeloid PPAR-γ deficiency leads to enhanced lung inflammation and collagen deposition following the resolution of influenza virus infection

We previously demonstrated that the transcription factor PPAR-γ was downregulated in alveolar macrophages (AM) following influenza virus infection [37, 38]. Within the lung compartment, myeloid cells, particularly AM expressed high levels of PPAR-γ (S1 Fig). Strikingly, myeloid deficiency of PPAR-γ (PpargΔLyz) resulted in increased host disease development, enhanced pulmonary inflammation and impaired tissue recovery at 15 days post influenza virus infection (d.p.i.) [37, 38]. Furthermore, PpargΔLyz mice showed delayed host weight recovery. However, majority of the infected PpargΔLyz survived and recovered most of their weight loss at 15 d.p.i. [37, 38]. Interestingly, recent reports have suggested that acute influenza infection can lead to chronic development of pathology, including fibrotic responses in the lungs [26]. We wondered whether myeloid deficiency of PPAR-γ leads to exacerbated development of chronic lung sequela following acute influenza infection. To this end, we infected WT (Ppargfl/fl) and PpargΔLyz mice with sublethal influenza virus A PR/8 strain. Consistent with our previous observations, PpargΔLyz mice displayed increased morbidity and delayed recovery compared to control mice (Fig 1A) [37, 38]. By 30 d.p.i., however, PpargΔLyz mice exhibited comparable weight gain as WT mice (Fig 1A). To examine whether myeloid PPAR-γ deficiency causes long-term effects on lung tissue following influenza infection, we examined the lung tissue secions at day 30 p.i. H&E staining of lung tissue sections revealed lesions in lungs from PPAR-γ cKO mice were far denser than controls indicating enhanced non-resolving inflammation and tissue injury (Fig 1B). Since PPAR-γ mainly expressed in macrophages but not neutrophils, we next examined macrophage composition in the lungs from WT or PPAR-γ cKO mice at 30 d.p.i. We found that total lung macrophage numbers (CD64+) were comparable between WT and PPAR-γ cKO mice (Fig 1C and 1D). However, WT mice had enhanced percentages of Siglec Fhi CD11blow macrophages, representative of tissue-resident alveolar macrophages, while PPAR-γ cKO mice showed higher percentages of Siglec Flow CD11bhi macrophages, a phenotype that associates with recruited monocyte-derived macrophages [32]. We next examined that cytokine gene expression in the lungs and found that lungs from WT and PPAR-γ cKO mice show comparable Il1b and Tgfb1 expression, but PPAR-γ cKO mice have modestly diminished Il6 expression and enhanced Ccl2 expression, suggesting that myeloid deficiency of PPAR-γ altered inflammatory responses in the lungs following the clearance of influenza virus infection (Fig 1F). Strikingly, at this time-point, late deficits in repair in the lung were observed by a decrease in Sftpb and Abca3; genes associated with type II airway epithelial cells (ATII), which are present in increased numbers in repaired tissue following injury (Fig 1G). Since persistent injury often drives tissue fibrotic responses, we next examined lung collagen content as the surrogate of lung fibrosis. To this end, hydroxyproline content, which was frequently used as a surrogate for collagen deposition, in the whole lungs of WT or PPAR-γ cKO mice was quantified at 30 d.p.i. We found that collagen deposition was quantitatively increased in whole lungs from PPAR-γ cKO mice compared to those of WT mice. (Fig 1H). Together, these data indicate that myeloid PPAR-γ deficiency results in delayed host weight recovery, altered macrophage and inflammatory responses and impaired repair of the lungs, which ultimately result in enhanced collagen deposition weeks following initial influenza exposure.

thumbnail
Fig 1. Myeloid PPAR-γ deficiency causes impaired lung inflammation resolution and enhanced collagen deposition.

WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were infected with influenza virus. Mice were sacrificed for various analysis at 30 d.p.i. (A) Host morbidity (% of initial weight) was monitored every other day till 30 d.p.i. (B) H.E stainning of lung sections collected from WT or PPAR-γ cKO mice. Low magnitude of whole left lung section image and high magnitude of indicated section images are depicted. (C) Lung cells from WT or PPAR-γ cKO mice were stained with CD64, Ly6G, Siglec F and CD11b and analyzed by flow cytometry. CD64+ macrophages were then subdivided into Siglec Fhi CD11blow and Siglec Flow CD11bhi populations. (D) Total numbers of CD64+ macrophages in the lungs from WT or PPAR-γ cKO mice were enumerated by flow cytometry. (E) Percentages of Siglec Fhi CD11blow or Siglec Flow CD11bhi macrophage populations were determined by flow cytometry. (F) Il1b, Tgfb1, Il6 and Ccl2 gene expression levels in the lungs from WT or PPAR-γ cKO mice were determined by realtime RT-PCR. (G) ATII gene, Sftpb and Abca3, expression levels in the lungs from WT or PPAR-γ cKO mice were determined by realtime RT-PCR. (H) Collagen content in the lungs from WT or PPAR-γ cKO mice was measured by Hydroxyproline assay. Data are representative of at least two independent experiments (n = 2–4 mice/group/experiment). Statistical differences are indicated. *, P < 0.05 (two tailed t-test).

https://doi.org/10.1371/journal.pone.0223430.g001

PPAR-γ deficient macrophages exhibited pro-inflammatory and pro-fibrotic gene activation

To assess the role of PPAR-γ deficient macrophages in the aberrant recovery following influenza virus infection, we sorted pulmonary macrophages (CD45+/ly6G-/CD64+/MerTK+) from WT and myeloid PPAR-γ deficient mice at day 30 p.i. (isolated from pooled 3–4 mice/group/experiment) (Fig 2A), and examined the expression of several genes associated with inflammation and fibrosis. Expression of Il1b, Ccl2 and Arg1 but not Tnf was elevated in PPAR-γ cKO macrophages (Fig 2B). Furthermore, we observed PPAR-γ deficient macrophages exhibited moderate increased expression of epithelial and fibroblast growth factors (Igf1, Pdgfa and Ctgf) (Fig 2C) and enhanced lung remodeling factors including Mmp2, Mmp7, Mmp8, Mmp9, Mmp12 and Timp1 (Fig 2D). In accordance, analysis of published microarray dataset (GSE60249 [36]) has revealed that PPAR-γ deficiency leaded to the upregulation of these pro-inflammatory and/or pro-fibrotic genes in AM even in the absence of influenza infection (Fig 2E). Thus, loss of PPAR-γ in macrophages led to sustained macrophage-intrinsic inflammation and lung remodeling programs at a time when they are largely resolved or tempered in wild type control macrophages.

thumbnail
Fig 2. PPAR-γ deficient macrophages exhibited pro-inflammatory and pro-fibrotic gene expression.

WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were infected with influenza. Mice were sacrificed for macrophage isolation at 30 d.p.i. (A) Lung cells were stained with anti-CD45, anti-Ly6G, anti-MerTK and anti-CD64. MerTK+/CD64+ lung macrophages in CD45+ Ly6G- cells were sorted by flow cytometry for realtime RT-PCR analysis of gene expression. (B) Expression of Tnf, Ccl2, Il1b and Arg1 in lung macrophages (isolated from pooled 3–4 mice/group/experiment) by realtime RT-PCR. (C) Expression of growth factors Igf1, Pdgfa, Tgfb and Ctgf in sorted lung macrophages (isolated from pooled 3–4 mice/group/experiment) by realtime RT-PCR. (D) Expression of several tissue remodeling enzymes and fibrotic genes Mmp2, Mmp8, Mmp12, Mmp7, Mmp9 and Timp1 in sorted lung macrophages (isolated from pooled 3–4 mice/group/experiment) by realtime RT-PCR. (E) Heat-map expression of the pro-inflammatory and profibrotic genes shown in B-D in lung alveolar macrophages from adult WT or conditional PPAR-γ-deficient mice (PpargΔCD11c) in published microarray dataset (GSE60249). Data are representative of two independent experiments of pooled lung macrophages from 3–4 mice except (E).

https://doi.org/10.1371/journal.pone.0223430.g002

Macrophages from human pulmonary fibrosis patients have overlapping repair, pro-inflammatory, and fibrotic gene signatures with PPAR-γ deficient macrophages

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic lung disease of unknown etiology. The roles of macrophages in IPF development and/or pathogenesis have been increasingly appreciated [5, 30]. However, the underlying molecular mechanisms regulating macrophage function in IPF remain largely elusive. Since we observed macrophage PPAR-γ deficiency increased lung collagen deposition, we wondered whether IPF patients exhibited altered PPAR-γ expression in macrophages. A secondary analysis from a gene-array expression profile database of macrophages sorted from healthy controls and pulmonary fibrosis patients indicates decreased PPAR-γ expression in patient macrophages (GEO #: GSE49072, 61 healthy volunteers and 23 pulmonary fibrosis patients) (Fig 3A). These data suggest that dysregulated PPAR-γ expression in pulmonary macrophages may play a role in lung fibrosis development and/or pathogenesis. In consistent with the idea, as was indicated in PPAR-γ deficient macrophages from influenza infected mice 30 days after viral infection (20 days after viral clearance), there were differences in pro-inflammatory (Ccl2), growth factor (Pdgfa), and fibrosis-associated gene expression (Mmp7, Mmp12, and Timp1) in macrophages from human IPF patients versus healthy control (Fig 3B). The parallel changes of PPAR-γ and a group of fibrosis related genes observed between murine PPAR-γ deficient macrophages (from influenza virus infection) and macrophages from pulmonary fibrosis patients might reflect important roles of PPAR-γ in macrophages in regulating the development and/or maintenance of progressive fibrosis leading to respiratory failure in IPF patients.

thumbnail
Fig 3. Macrophages from human pulmonary fibrosis patients exhibited diminished PPARG and dysregulated pro-inflammatory and pro-fibrotic gene expression.

PPARG, pro-inflammatory and pro-fibrotic gene expression in lung macrophages isolated from 61 healthy volunteers or 23 pulmonary fibrosis patients in the GSE49072 microarray datasets. (A) PPARG expression in lung alveolar macrophages from healthy or pulmonary fibrosis patients. (B) MMP7, MMP12, TIMP1, CCL2 or PDGFA expression in lung alveolar macrophages from healthy or pulmonary fibrosis patients. Statistical values are indicated (two-tailed Mann-Whitney test).

https://doi.org/10.1371/journal.pone.0223430.g003

Myeloid PPAR-γ deficiency facilitates chronic influenza-induced pulmonary fibrosis

To further assess the effect of myeloid deficiency of PPAR-γ on recovery following influenza infection, we examined lesions and collagen deposition in the lung tissue at 60 days p.i. H&E staining of lung tissue sections revealed sustained inflammation and damage from mice with PPAR-γ deficient macrophages compared to those of WT lungs that were largely resolved in these regards at 60 days p.i. (Fig 4A). At this time, higher collagen deposition was found in lungs of mice with myeloid PPAR-γ deficiency compared to those of WT mice (Fig 4B). Consistent with this observation, RT-qPCR array data exhibited globally enhanced pro-inflammatory and pro-fibrotic gene profiles in lungs of myeloid PPAR-γ deficient mice (Fig 4C), confirming that macrophage PPAR-γ has functional consequences for the lung at the level of gene transcription. To see if this persisted another month, we measured collagen content and fibrotic gene transcription in the lungs of WT or PPAR-γ cKO mice at 90 d.p.i. (Fig 4D and 4E). Notably, influenza-infected control animals had a measurable increase in total lung collagen at 60 d.p.i. relative to non-infected counterparts, but this is largely resolved by day 90 p.i., suggesting that lung fibrotic responses due to influenza infection were largely resolved in WT mice at three months after infection. Remarkably, lungs from PPAR-γ cKO animals exhibited enhanced lung collagen content than the lungs from non-infected or influenza-infected WT mice, suggesting that myeloid PPAR-γ deficiency exacerbated extracellular matrix deposition and/or delayed the clearance of deposited collagen. The data likely indicate myeloid PPAR-γ deficiency leads to persistent progressive pro-fibrotic activity in the lungs following influenza infection.

thumbnail
Fig 4. Myeloid PPAR-γ deficiency leads to unresolved persistent fibrosis.

(A) WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were infected with influenza virus. H&E staining of lung sections from WT or PPAR-γ cKO mice at day 60 p.i. (B) WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were treated with saline (mock) or infected with influenza as indicated. Collagen content in the whole lungs from indicated groups mice was measured by Hydroxyproline assay at day 60 p.i. (C) WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were infected with influenza virus. Lung fibrotic genes expression was measured by QIAGEN Fibrosis RT2 RT-PCR array at day 60 p.i. Red dots, genes upregulated in PPAR-γ cKO lungs, green dots, genes downregulated in PPAR-γ cKO lungs. (D) WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were treated with saline (mock) or infected with influenza virus as indicated. Collagen content in the lungs from indicated groups mice was measured by Hydroxyproline assay at day 90 p.i. (E) WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were infected with influenza virus. Lung fibrotic genes expression was measured by QIAGEN Fibrosis RT2 RT-PCR array at day 90 p.i. Red dots, genes upregulated in PPAR-γ cKO lungs, green dots, genes downregulated in PPAR-γ cKO lungs. Data are representative of two independent experiments (3–5 mice/group/experiment). Statistical differences are indicated. *, P < 0.05 (one-way ANOVA).

https://doi.org/10.1371/journal.pone.0223430.g004

Mice with PPAR-γ deficient macrophages are more resistant to bleomycin-induced weight loss

To examine if the PPAR-γ signaling pathway in macrophages is involved in other damage induced pro-fibrotic responses, we compared the responses induced by intratracheal instillation of bleomycin between wild type and PPAR-γ cKO mice. In contrast to the enhanced weight loss observed in PPAR-γ cKO mice following influenza virus infection, we found that myeloid PPAR-γ deficiency resulted in decreased host morbidity following bleomycin inoculation (Fig 5A). These data suggest that macrophage PPAR-γ may differentially regulate the acute lung inflammation and injury following influenza or bleomycin treatment (Fig 5A). Previous reports have demonstrated that peak of lung fibrosis development following bleomycin treatment is around 2–4 weeks [45, 46]. To study the roles of myeloid PPAR-γ deficiency in regulating the resolution of pulmonary fibrosis, we compared total lung collagen content in the lungs between WT and PPAR-γ cKO mice at 6 weeks post bleomycin inoculation (Fig 5B). We found that myeloid PPAR-γ deficiency did not resulted in increased collagen deposition following bleomycin treatment, which is in sharp contrast to what we have observed in the influenza infection model. Thus, macrophage PPAR-γ may not be an effector pathway for all types of injury and may be limited to microbial responses in the lung.

thumbnail
Fig 5. Mice with myeloid PPAR-γ deficiency were more resistant to bleomycin damage.

WT (Ppargfl/fl) or PPAR-γ cKO (PpargΔLyz2) mice were inoculated with bleomycin (0.072 Unit/mouse). (A) Host morbidity (% of initial weight loss) was monitored following bleomycin treatment. (B) Collagen content in whole lungs from WT or PPAR-γ cKO mice was measured by Hydroxyproline assay at 42 days after bleomycin treatment. Data are representative of two independent experiments (3–5 mice/group/experiment). Statistical differences are indicated. *, P < 0.05. ns, not significant. (A. t test or B. one-way ANOVA).

https://doi.org/10.1371/journal.pone.0223430.g005

Discussion

Emerging evidence has suggested that acute respiratory viral infections can cause chronic disease sequela including fibrotic responses months following the clearance of infectious viruses [26]. Likewise, post influenza fibrotic responses following the resolution of acute virus-induced diseases have been documented in clinical cases [4749]. Furthermore, respiratory viral infections have been associated with the acute exacerbation of pulmonary fibrosis [5052]. Thus, it is of significant clinical relevance to investigate the cellular and molecular mechanisms regulating the development of pulmonary fibrosis following influenza infection. In this report, we have discovered that myeloid deficiency of PPAR-γ resulted in dysregulated macrophage pro-inflammatory and pro-fibrotic responses following influenza virus infection. Strikingly, we observed that persistent fibrotic responses were persistent in myeloid PPAR-γ deficient mice for at least 3 months following infection. Previously, we have shown that macrophage PPAR-γ is a critical inhibitor of acute disease development (before day 15) following influenza virus infection [37, 38]. Together, these data suggest that macrophage PPAR-γ is critical for the suppression of both acute and chronic pathogenesis following primary influenza virus infection.

The roles of macrophage in regulating the development of tissue fibrosis have been well-established. Particularly, alternative-activated macrophages (M2) have been demonstrated to promote fibrosis development in multi-organs including the lungs [5, 30]. PPAR-γ was shown before as a critical regulator for M2 differentiation following IL-4 treatment [33]. However, PPAR-γ deficiency did not alter the expression of M2 genes, Arg1 and Retnla (encodes Relmα) expression following influenza exposure [38]. Likely, PPAR-γ deficiency increased Arg1 expression in lung macrophages at 30 days post influenza infection. Thus, it is likely that macrophage PPAR-γ expression suppresses pulmonary fibrosis independent of M1 or M2 polarization. Indeed, emerging evidence has suggested that a dichotomous stratification of macrophage phenotypes (M1 versus M2) oversimplifies the functional heterogeneity of these highly plastic cells [5, 53, 54].

Besides the roles of macrophage polarization in fibrosis development, recent data have also suggested important roles of recruited monocyte-derived macrophages in fibrosis development [32]. Monocyte recruitment in the lung during influenza infection is largely dependent on the CCL2-CCR2 axis [55, 56]. Indeed, PPAR-γ deficient macrophages exhibited enhanced CCL2 expression and lungs from PPAR-γ cKO mice also showed increased CCL2 expression. Consistently, we observed disproportionate distribution of macrophages with resident or recruited macrophage phenotypes in PPAR-γ cKO mice. Thus, it is possible that enhanced responses of monocyte-derived macrophages may contribute to the development and/or failed resolution of pulmonary fibrosis following influenza infection. Notably, we measured pro-inflammatory and pro-fibrotic genes in total lung macrophages rather than in recruited or resident macrophage populations. We reasoned that macrophages from patient samples would contains both resident and recruited macrophage populations. Therefore, in order to compare the gene expression of the “global” macrophage population between mouse and human, we chose to examine pro-inflammatory and pro-fibrotic gene expression in the total lung macrophage population. Further studies are warranted to determine whether the increased expression of those genes was largely due to the recruitment of monocyte-derived macrophages or not. Of note, we have found that PPAR-γ deficiency in purified resident macrophages leaded to enhanced Il1b and Ccl2 expression [38], suggesting that enhanced inflammatory and fibrotic genes may also occur in resident macrophage population in the absence of PPAR-γ. Nevertheless, comparison of the gene expression of the “total” macrophage compartment revealed that the effects of PPAR-γ in regulating pro-fibrotic macrophage responses may be conserved in mouse and human. Notably, Lyz2-driven Cre activities were also observed in ATII cells and PPAR-γ expression was observed in airway epithelial cells [57, 58]. Thus, it is possible that PPAR-γ expression in ATII cell may also contribute to the fibrotic phenotypes observed in myeloid PPAR-γ deficient mice following influenza virus infection. Future studies using ATII-specific deficiency of PPAR-γ is required to dissect the roles of PPAR-γ expression in ATII cells in modulating pulmonary fibrosis development following influenza virus infection.

Interestingly, the current study is one of multiple reports that has now identified a beneficial role for PPAR-γ in macrophages following influenza virus infection of mice [37,38]. This indicates that endogenous PPAR-γ agonists whose presence in the lung may temper the severity of disease sequelae. This study therefore warrants investigating the potential PPAR-γ ligands in the lung and how these differ regarding severity of non-resolving pathologies. Discovering the nature of these functional agonists in the context of influenza infection could be important for treatment of those who are highly prone to severe infection and often succumb to the resulting sequela, such as the elderly [59]. Notably, although PPAR-γ action appear to be beneficial to the hosts during primary influenza virus infection, a recent report has suggested that stimulation of PPAR-γ action could suppress efficient host anti-bacterial responses in a influenza and bacterial super infection model [60], although the macrophage-specific PPAR-γ action was not determined in this study. Nevertheless, the proper balance of PPAR-γ activities would be a key to promote efficient recovery from primary influenza infection while minimizing the risk for enhanced secondary bacterial infection.

Bleomycin is a small-molecule used to experimentally induce severe, but reversible fibrosis of the lung parenchyma. Respiratory epithelial cells are susceptible to bleomycin resulting in apoptosis following damage of genomic DNA and ensuing inflammation [61, 62]. After bleomycin insult, PPAR-γ cKO mice exhibited decreased acute weight loss. Furthermore, PPAR-γ cKO mice showed less, but no significant, collagen deposition compared with wild type mice. This is surprising as PPAR-γ ligands have been demonstrated to exhibit anti-fibrotic effects in bleomycin treated mice [63]. Currently, the difference for the fibrotic responses between the disparate models used here is not clear, but it is likely that the PPAR-γ signaling axis in pulmonary macrophages may be important for pathogen induced damage and not general damage. Further comparison on the phenotypes of PPAR-γ-deficient lung macrophages from influenza-infected mice versus those from bleomycin-treated mice is needed to confirm this idea. Nonetheless, our data imply that the early and ongoing inflammatory milieus resulting from the type of various danger signals may dictate which repair pathway(s) are used and when. Of note, we focused on our experiments at the resolution phase (i.e. 6 weeks post bleomycin treatment). Previous reports have demonstrated that peak of lung fibrosis development following bleomycin treatment is around 2–4 weeks [45, 46]. It thus remains possible that myeloid PPAR-γ deficiency may increase lung collagen deposition at the peak of pulmonary fibrosis. However, given that myeloid PPAR-γ deficiency diminished rather than increased host weight loss, we think that this possibility is low. Nonetheless, since we found overlapping patterns in dysfunctional macrophages from influenza-infected animals exhibiting fibrosis and those from IPF patients, this indicates the fibrotic sequelae found in the PPAR-γ cKO mice following acute influenza virus exposure may be a relevant model for human pulmonary fibrosis development. Given the current urgent need for better pre-clinical models for human pulmonary fibrotic diseases, the discoveries in this manuscript is of significant importance to pulmonary research.

Supporting information

S1 Fig. Pparg is highly expressed in alveolar macrophages compared to epithelial cells.

https://doi.org/10.1371/journal.pone.0223430.s001

(TIF)

Acknowledgments

We thank Mayo Clinic Flow Cytometry Core and Histology Core for their technical assistance.

References

  1. 1. Ruf BR, Knuf M. The burden of seasonal and pandemic influenza in infants and children. Eur J Pediatr. 2014;173(3):265–76. pmid:23661234; PubMed Central PMCID: PMC3930829.
  2. 2. Keeler SP, Agapov EV, Hinojosa ME, Letvin AN, Wu K, Holtzman MJ. Influenza A Virus Infection Causes Chronic Lung Disease Linked to Sites of Active Viral RNA Remnants. J Immunol. 2018;201(8):2354–68. pmid:30209189; PubMed Central PMCID: PMC6179922.
  3. 3. Zhang Y, Mao D, Keeler SP, Wang X, Wu K, Gerovac BJ, et al. Respiratory Enterovirus (like Parainfluenza Virus) Can Cause Chronic Lung Disease if Protection by Airway Epithelial STAT1 Is Lost. J Immunol. 2019;202(8):2332–47. Epub 2019/02/26. pmid:30804041; PubMed Central PMCID: PMC6456410.
  4. 4. Yoo JK, Kim TS, Hufford MM, Braciale TJ. Viral infection of the lung: host response and sequelae. J Allergy Clin Immunol. 2013;132(6):1263–76; quiz 77. pmid:23915713; PubMed Central PMCID: PMC3844062.
  5. 5. Desai O, Winkler J, Minasyan M, Herzog EL. The Role of Immune and Inflammatory Cells in Idiopathic Pulmonary Fibrosis. Frontiers in medicine. 2018;5:43. Epub 2018/04/05. pmid:29616220; PubMed Central PMCID: PMC5869935.
  6. 6. Wang Z, Wang S, Goplen NP, Li C, Cheon IS, Dai Q, et al. PD-1(hi) CD8(+) resident memory T cells balance immunity and fibrotic sequelae. Sci Immunol. 2019;4(36). Epub 2019/06/16. pmid:31201259.
  7. 7. Sun J, Madan R, Karp CL, Braciale TJ. Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10. Nat Med. 2009;15(3):277–84. Epub 2009/02/24. pmid:19234462; PubMed Central PMCID: PMC2693210.
  8. 8. Wong SS, Oshansky CM, Guo XJ, Ralston J, Wood T, Seeds R, et al. Severe Influenza Is Characterized by Prolonged Immune Activation: Results From the SHIVERS Cohort Study. J Infect Dis. 2018;217(2):245–56. Epub 2017/11/08. pmid:29112724.
  9. 9. Braciale TJ, Sun J, Kim TS. Regulating the adaptive immune response to respiratory virus infection. Nat Rev Immunol. 2012;12(4):295–305. pmid:22402670; PubMed Central PMCID: PMC3364025.
  10. 10. Sun J, Braciale TJ. Role of T cell immunity in recovery from influenza virus infection. Curr Opin Virol. 2013;3(4):425–9. pmid:23721865; PubMed Central PMCID: PMC3804899.
  11. 11. Newton AH, Cardani A, Braciale TJ. The host immune response in respiratory virus infection: balancing virus clearance and immunopathology. Semin Immunopathol. 2016;38(4):471–82. pmid:26965109; PubMed Central PMCID: PMC4896975.
  12. 12. Guo XJ, Thomas PG. New fronts emerge in the influenza cytokine storm. Semin Immunopathol. 2017;39(5):541–50. Epub 2017/05/31. pmid:28555383; PubMed Central PMCID: PMC5580809.
  13. 13. Zammit DJ, Turner DL, Klonowski KD, Lefrancois L, Cauley LS. Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration. Immunity. 2006;24(4):439–49. Epub 2006/04/19. pmid:16618602; PubMed Central PMCID: PMC2861289.
  14. 14. Jelley-Gibbs DM, Brown DM, Dibble JP, Haynes L, Eaton SM, Swain SL. Unexpected prolonged presentation of influenza antigens promotes CD4 T cell memory generation. J Exp Med. 2005;202(5):697–706. Epub 2005/09/09. pmid:16147980; PubMed Central PMCID: PMC2212871.
  15. 15. Kim TS, Hufford MM, Sun J, Fu YX, Braciale TJ. Antigen persistence and the control of local T cell memory by migrant respiratory dendritic cells after acute virus infection. J Exp Med. 2010;207(6):1161–72. Epub 2010/06/02. pmid:20513748; PubMed Central PMCID: PMC2882836.
  16. 16. Pociask DA, Robinson KM, Chen K, McHugh KJ, Clay ME, Huang GT, et al. Epigenetic and Transcriptomic Regulation of Lung Repair during Recovery from Influenza Infection. Am J Pathol. 2017;187(4):851–63. pmid:28193481; PubMed Central PMCID: PMC5397680.
  17. 17. Qiao J, Zhang M, Bi J, Wang X, Deng G, He G, et al. Pulmonary fibrosis induced by H5N1 viral infection in mice. Respir Res. 2009;10:107. pmid:19909524; PubMed Central PMCID: PMC2783028.
  18. 18. Kanegai CM, Xi Y, Donne ML, Gotts JE, Driver IH, Amidzic G, et al. Persistent Pathology in Influenza-Infected Mouse Lungs. Am J Respir Cell Mol Biol. 2016;55(4):613–5. pmid:27689795; PubMed Central PMCID: PMC5070109.
  19. 19. Goplen N, Karim MZ, Liang Q, Gorska MM, Rozario S, Guo L, et al. Combined sensitization of mice to extracts of dust mite, ragweed, and Aspergillus species breaks through tolerance and establishes chronic features of asthma. J Allergy Clin Immunol. 2009;123(4):925–32 e11. Epub 2009/04/08. pmid:19348928; PubMed Central PMCID: PMC2683988.
  20. 20. Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010;327(5963):291–5. Epub 2010/01/16. pmid:20075244; PubMed Central PMCID: PMC3645875.
  21. 21. Iwasaki A, Foxman EF, Molony RD. Early local immune defences in the respiratory tract. Nat Rev Immunol. 2017;17(1):7–20. pmid:27890913; PubMed Central PMCID: PMC5480291.
  22. 22. Medzhitov R, Schneider DS, Soares MP. Disease tolerance as a defense strategy. Science. 2012;335(6071):936–41. Epub 2012/03/01. pmid:22363001; PubMed Central PMCID: PMC3564547.
  23. 23. Rawlins EL. Stem cells: Emergency back-up for lung repair. Nature. 2015;517(7536):556–7. Epub 2015/01/30. pmid:25631438.
  24. 24. Zuo W, Zhang T, Wu DZ, Guan SP, Liew AA, Yamamoto Y, et al. p63(+)Krt5(+) distal airway stem cells are essential for lung regeneration. Nature. 2015;517(7536):616–20. Epub 2014/11/11. pmid:25383540.
  25. 25. Sueblinvong V, Neujahr DC, Mills ST, Roser-Page S, Ritzenthaler JD, Guidot D, et al. Predisposition for disrepair in the aged lung. Am J Med Sci. 2012;344(1):41–51. Epub 2011/12/17. pmid:22173045; PubMed Central PMCID: PMC3395069.
  26. 26. Vaughan AE, Brumwell AN, Xi Y, Gotts JE, Brownfield DG, Treutlein B, et al. Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury. Nature. 2015;517(7536):621–5. pmid:25533958; PubMed Central PMCID: PMC4312207.
  27. 27. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol. 2014;14(2):81–93. Epub 2014/01/22. pmid:24445666.
  28. 28. Italiani P, Boraschi D. New Insights Into Tissue Macrophages: From Their Origin to the Development of Memory. Immune Netw. 2015;15(4):167–76. Epub 2015/09/04. pmid:26330802; PubMed Central PMCID: PMC4553254.
  29. 29. Faz-Lopez B, Morales-Montor J, Terrazas LI. Role of Macrophages in the Repair Process during the Tissue Migrating and Resident Helminth Infections. Biomed Res Int. 2016;2016:8634603. Epub 2016/09/21. pmid:27648452; PubMed Central PMCID: PMC5014929.
  30. 30. Wynn TA, Vannella KM. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity. 2016;44(3):450–62. Epub 2016/03/18. pmid:26982353; PubMed Central PMCID: PMC4794754.
  31. 31. Guilliams M, Lambrecht BN, Hammad H. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal Immunol. 2013;6(3):464–73. Epub 2013/04/04. pmid:23549447.
  32. 32. Misharin AV, Morales-Nebreda L, Reyfman PA, Cuda CM, Walter JM, McQuattie-Pimentel AC, et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med. 2017;214(8):2387–404. Epub 2017/07/12. pmid:28694385; PubMed Central PMCID: PMC5551573.
  33. 33. Zhang L, Chawla A. Role of PPARgamma in macrophage biology and atherosclerosis. Trends Endocrinol Metab. 2004;15(10):500–5. Epub 2004/11/16. pmid:15541649.
  34. 34. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARgamma signaling and metabolism: the good, the bad and the future. Nat Med. 2013;19(5):557–66. Epub 2013/05/09. pmid:23652116; PubMed Central PMCID: PMC3870016.
  35. 35. Schneider C, Nobs SP, Heer AK, Kurrer M, Klinke G, van Rooijen N, et al. Alveolar macrophages are essential for protection from respiratory failure and associated morbidity following influenza virus infection. PLoS Pathog. 2014;10(4):e1004053. pmid:24699679; PubMed Central PMCID: PMC3974877.
  36. 36. Schneider C, Nobs SP, Kurrer M, Rehrauer H, Thiele C, Kopf M. Induction of the nuclear receptor PPAR-gamma by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat Immunol. 2014;15(11):1026–37. pmid:25263125.
  37. 37. Huang S, Jiang L, Cheon IS, Sun J. Targeting Peroxisome Proliferator-Activated Receptor-Gamma Decreases Host Mortality After Influenza Infection in Obese Mice. Viral immunology. 2019;32(4):161–9. Epub 2019/04/23. pmid:31009317; PubMed Central PMCID: PMC6534095.
  38. 38. Huang S, Zhu B, Cheon IS, Goplen NP, Jiang L, Zhang R, et al. PPAR-gamma in macrophages limits pulmonary inflammation and promotes host recovery following respiratory viral infection. J Virol. 2019. Epub 2019/02/23. pmid:30787149.
  39. 39. Hua L, Yao S, Pham D, Jiang L, Wright J, Sawant D, et al. Cytokine-dependent induction of CD4+ T cells with cytotoxic potential during influenza virus infection. J Virol. 2013;87(21):11884–93. pmid:23986597; PubMed Central PMCID: PMC3807312.
  40. 40. Jung MY, Kang JH, Hernandez DM, Yin X, Andrianifahanana M, Wang Y, et al. Fatty acid synthase is required for profibrotic TGF-beta signaling. FASEB J. 2018;32(7):3803–15. Epub 2018/02/25. pmid:29475397; PubMed Central PMCID: PMC5998981.
  41. 41. Cheon IS, Son YM, Jiang L, Goplen NP, Kaplan MH, Limper AH, et al. Neonatal hyperoxia promotes asthma-like features through IL-33-dependent ILC2 responses. J Allergy Clin Immunol. 2018;142(4):1100–12. pmid:29253513; PubMed Central PMCID: PMC6003836.
  42. 42. Yao S, Jiang L, Moser EK, Jewett LB, Wright J, Du J, et al. Control of pathogenic effector T-cell activities in situ by PD-L1 expression on respiratory inflammatory dendritic cells during respiratory syncytial virus infection. Mucosal Immunol. 2015;8(4):746–59. Epub 2014/12/04. pmid:25465101; PubMed Central PMCID: PMC4632244.
  43. 43. Jiang L, Yao S, Huang S, Wright J, Braciale TJ, Sun J. Type I IFN signaling facilitates the development of IL-10-producing effector CD8(+) T cells during murine influenza virus infection. Eur J Immunol. 2016;46(12):2778–88. pmid:27701741; PubMed Central PMCID: PMC5184847.
  44. 44. Shi Y, Gochuico BR, Yu G, Tang X, Osorio JC, Fernandez IE, et al. Syndecan-2 exerts antifibrotic effects by promoting caveolin-1-mediated transforming growth factor-beta receptor I internalization and inhibiting transforming growth factor-beta1 signaling. Am J Respir Crit Care Med. 2013;188(7):831–41. Epub 2013/08/09. pmid:23924348; PubMed Central PMCID: PMC3826270.
  45. 45. Moeller A, Ask K, Warburton D, Gauldie J, Kolb M. The bleomycin animal model: a useful tool to investigate treatment options for idiopathic pulmonary fibrosis? Int J Biochem Cell Biol. 2008;40(3):362–82. Epub 2007/10/16. pmid:17936056; PubMed Central PMCID: PMC2323681.
  46. 46. Chaudhary NI, Schnapp A, Park JE. Pharmacologic differentiation of inflammation and fibrosis in the rat bleomycin model. Am J Respir Crit Care Med. 2006;173(7):769–76. Epub 2006/01/18. pmid:16415276.
  47. 47. Mineo G, Ciccarese F, Modolon C, Landini MP, Valentino M, Zompatori M. Post-ARDS pulmonary fibrosis in patients with H1N1 pneumonia: role of follow-up CT. Radiol Med. 2012;117(2):185–200. Epub 2011/10/25. pmid:22020433.
  48. 48. Singh V, Sharma BB, Patel V. Pulmonary sequelae in a patient recovered from swine flu. Lung India. 2012;29(3):277–9. Epub 2012/08/25. pmid:22919170; PubMed Central PMCID: PMC3424870.
  49. 49. Li P, Zhang JF, Xia XD, Su DJ, Liu BL, Zhao DL, et al. Serial evaluation of high-resolution CT findings in patients with pneumonia in novel swine-origin influenza A (H1N1) virus infection. Br J Radiol. 2012;85(1014):729–35. Epub 2011/12/15. pmid:22167502; PubMed Central PMCID: PMC3474108.
  50. 50. Juarez MM, Chan AL, Norris AG, Morrissey BM, Albertson TE. Acute exacerbation of idiopathic pulmonary fibrosis-a review of current and novel pharmacotherapies. J Thorac Dis. 2015;7(3):499–519. pmid:25922733; PubMed Central PMCID: PMC4387423.
  51. 51. Lederer DJ, Martinez FJ. Idiopathic Pulmonary Fibrosis. N Engl J Med. 2018;378(19):1811–23. pmid:29742380.
  52. 52. Molyneaux PL, Maher TM. The role of infection in the pathogenesis of idiopathic pulmonary fibrosis. Eur Respir Rev. 2013;22(129):376–81. pmid:23997064.
  53. 53. Misharin AV, Morales-Nebreda L, Reyfman PA, Cuda CM, Walter JM, McQuattie-Pimentel AC, et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. Journal of Experimental Medicine. 2017;214(8):2387–404. pmid:28694385 PubMed PMID: WOS:000407076300016.
  54. 54. Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity. 2016;44(3):439–49. Epub 2016/03/18. pmid:26982352.
  55. 55. Aldridge JR Jr., Moseley CE, Boltz DA, Negovetich NJ, Reynolds C, Franks J, et al. TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection. Proc Natl Acad Sci U S A. 2009;106(13):5306–11. pmid:19279209; PubMed Central PMCID: PMC2664048.
  56. 56. Lin KL, Suzuki Y, Nakano H, Ramsburg E, Gunn MD. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J Immunol. 2008;180(4):2562–72. pmid:18250467.
  57. 57. Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature. 2014;507(7491):190–4. pmid:24499815; PubMed Central PMCID: PMC4013278.
  58. 58. Solleti SK, Simon DM, Srisuma S, Arikan MC, Bhattacharya S, Rangasamy T, et al. Airway epithelial cell PPARgamma modulates cigarette smoke-induced chemokine expression and emphysema susceptibility in mice. Am J Physiol Lung Cell Mol Physiol. 2015;309(3):L293–304. Epub 2015/05/31. pmid:26024894; PubMed Central PMCID: PMC4525123.
  59. 59. Pebody RG, McLean E, Zhao H, Cleary P, Bracebridge S, Foster K, et al. Pandemic Influenza A (H1N1) 2009 and mortality in the United Kingdom: risk factors for death, April 2009 to March 2010. Euro Surveill. 2010;15(20). Epub 2010/05/28. pmid:20504388.
  60. 60. Gopal R, Mendy A, Marinelli MA, Richwalls LJ, Seger PJ, Patel S, et al. Peroxisome Proliferator-Activated Receptor Gamma (PPAR) Suppresses Inflammation and Bacterial Clearance during Influenza-Bacterial Super-Infection. Viruses. 2019;11(6). Epub 2019/06/05. pmid:31159430.
  61. 61. Hamilton RF Jr., Li L, Felder TB, Holian A. Bleomycin induces apoptosis in human alveolar macrophages. Am J Physiol. 1995;269(3 Pt 1):L318–25. Epub 1995/09/11. pmid:7573464.
  62. 62. Della Latta V, Cecchettini A, Del Ry S, Morales MA. Bleomycin in the setting of lung fibrosis induction: From biological mechanisms to counteractions. Pharmacol Res. 2015;97:122–30. Epub 2015/05/12. pmid:25959210.
  63. 63. Lakatos HF, Thatcher TH, Kottmann RM, Garcia TM, Phipps RP, Sime PJ. The Role of PPARs in Lung Fibrosis. PPAR Res. 2007;2007:71323. Epub 2007/08/22. pmid:17710235; PubMed Central PMCID: PMC1940051.