Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Eur Respir Rev ; 29(155)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32198218

ABSTRACT

Pulmonary sarcoidosis presents substantial management challenges, with limited evidence on effective therapies and phenotypes. In the absence of definitive evidence, expert consensus can supply clinically useful guidance in medicine. An international panel of 26 experts participated in a Delphi process to identify consensus on pharmacological management in sarcoidosis with the development of preliminary recommendations.The modified Delphi process used three rounds. The first round focused on qualitative data collection with open-ended questions to ensure comprehensive inclusion of expert concepts. Rounds 2 and 3 applied quantitative assessments using an 11-point Likert scale to identify consensus.Key consensus points included glucocorticoids as initial therapy for most patients, with non-biologics (immunomodulators), usually methotrexate, considered in severe or extrapulmonary disease requiring prolonged treatment, or as a steroid-sparing intervention in cases with high risk of steroid toxicity. Biologic therapies might be considered as additive therapy if non-biologics are insufficiently effective or are not tolerated with initial biologic therapy, usually with a tumour necrosis factor-α inhibitor, typically infliximab.The Delphi methodology provided a platform to gain potentially valuable insight and interim guidance while awaiting evidenced-based contributions.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Algorithms , Biological Products/therapeutic use , Clinical Decision-Making , Decision Support Techniques , Immunologic Factors/therapeutic use , Lung/drug effects , Sarcoidosis, Pulmonary/drug therapy , Adrenal Cortex Hormones/adverse effects , Biological Products/adverse effects , Consensus , Delphi Technique , Humans , Immunologic Factors/adverse effects , Lung/physiopathology , Sarcoidosis, Pulmonary/diagnosis , Sarcoidosis, Pulmonary/physiopathology , Severity of Illness Index
2.
Respir Res ; 17(1): 93, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27460362

ABSTRACT

BACKGROUND: Sarcoidosis is a multisystem immuno-inflammatory disorder of unknown etiology that most commonly involves the lungs. We hypothesized that an unbiased approach to identify pathways activated in bronchoalveolar lavage (BAL) cells can shed light on the pathogenesis of this complex disease. METHODS: We recruited 15 patients with various stages of sarcoidosis and 12 healthy controls. All subjects underwent bronchoscopy with lavage. For each subject, total RNA was extracted from BAL cells and hybridized to an Affymetrix U133A microarray. Rigorous statistical methods were applied to identify differential gene expression between subjects with sarcoidosis vs. CONTROLS: To better elucidate pathways differentially activated between these groups, we integrated network and gene set enrichment analyses of BAL cell transcriptional profiles. RESULTS: Sarcoidosis patients were either non-smokers or former smokers, all had lung involvement and only two were on systemic prednisone. Healthy controls were all non-smokers. Comparison of BAL cell gene expression between sarcoidosis and healthy subjects revealed over 1500 differentially expressed genes. Several previously described immune mediators, such as interferon gamma, were upregulated in the sarcoidosis subjects. Using an integrative computational approach we constructed a modular network of over 80 gene sets that were highly enriched in patients with sarcoidosis. Many of these pathways mapped to inflammatory and immune-related processes including adaptive immunity, T-cell signaling, graft vs. host disease, interleukin 12, 23 and 17 signaling. Additionally, we uncovered a close association between the proteasome machinery and adaptive immunity, highlighting a potentially important and targetable relationship in the pathobiology of sarcoidosis. CONCLUSIONS: BAL cells in sarcoidosis are characterized by enrichment of distinct transcriptional programs involved in immunity and proteasomal processes. Our findings add to the growing evidence implicating alveolar resident immune effector cells in the pathogenesis of sarcoidosis and identify specific pathways whose activation may modulate disease progression.


Subject(s)
Bronchoalveolar Lavage Fluid/cytology , Gene Expression Regulation/genetics , Sarcoidosis, Pulmonary/genetics , Sarcoidosis, Pulmonary/metabolism , Adult , Aged , Bronchoscopy , Cell Count , Cytokines/metabolism , Female , Gene Regulatory Networks/genetics , Humans , Immunity/genetics , Male , Microarray Analysis , Middle Aged , Proteasome Endopeptidase Complex/genetics , RNA/biosynthesis , RNA/isolation & purification , Sarcoidosis, Pulmonary/immunology , Smoking/genetics
3.
Eur Respir J ; 44(5): 1296-307, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25034562

ABSTRACT

Sarcoidosis is characterised by non-caseating granulomas that secrete pro-inflammatory cytokines, including interleukin (IL)-12, IL-23, and tumour necrosis factor (TNF)-α. Ustekinumab and golimumab are monoclonal antibodies that specifically inhibit IL-12/IL-23 and TNF-α, respectively. Patients with chronic pulmonary sarcoidosis (lung group) and/or skin sarcoidosis (skin group) received either 180 mg ustekinumab at week 0 followed by 90 mg every 8 weeks, 200 mg golimumab at week 0 followed by 100 mg every 4 weeks, or placebo. Patients underwent corticosteroid tapering between weeks 16 and 28. The primary end-point was week 16 change in percentage predicted forced vital capacity (ΔFVC % pred) in the lung group. Major secondary end-points were: week 28 for ΔFVC % pred, 6-min walking distance, St George's Respiratory Questionnaire (lung group), and Skin Physician Global Assessment response (skin group). At week 16, no significant differences were observed in ΔFVC % pred with ustekinumab (-0.15, p = 0.13) or golimumab (1.15, p = 0.54) compared with placebo (2.02). At week 28, there were no significant improvements in the major secondary end-points, although a nonsignificant numerically greater Skin Physician Global Assessment response was observed following golimumab treatment (53%) when compared with the placebo (30%). Serious adverse events were similar in all treatment groups. Although treatment was well tolerated, neither ustekinumab nor golimumab demonstrated efficacy in pulmonary sarcoidosis. However, trends towards improvement were observed with golimumab in some dermatological end-points.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Sarcoidosis/drug therapy , Sarcoidosis/physiopathology , Adult , Chronic Disease , Double-Blind Method , Female , Humans , Interleukin-12/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Male , Middle Aged , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ustekinumab
4.
J Leukoc Biol ; 95(1): 9-18, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23964118

ABSTRACT

Members of the MMP family function in various processes of innate immunity, particularly in controlling important steps in leukocyte trafficking and activation. MMP28 (epilysin) is a member of this family of proteinases, and we have found that MMP28 is expressed by macrophages and regulates their recruitment to the lung. We hypothesized that MMP28 regulates other key macrophage responses, such as macrophage polarization. Furthermore, we hypothesized that these MMP28-dependent changes in macrophage polarization would alter fibrotic responses in the lung. We examined the gene expression changes in WT and Mmp28-/- BMDMs, stimulated with LPS or IL-4/IL-13 to promote M1 and M2 cells, respectively. We also collected macrophages from the lungs of Pseudomonas aeruginosa-exposed WT and Mmp28-/- mice to evaluate changes in macrophage polarization. Lastly, we evaluated the macrophage polarization phenotypes during bleomycin-induced pulmonary fibrosis in WT and Mmp28-/- mice and assessed mice for differences in weight loss and total collagen levels. We found that MMP28 dampens proinflammatory macrophage function and promots M2 programming. In both in vivo models, we found deficits in M2 polarization in Mmp28-/- mice. In bleomycin-induced lung injury, these changes were associated with reduced fibrosis. MMP28 is an important regulator of macrophage polarization, promoting M2 function. Loss of MMP28 results in reduced M2 polarization and protection from bleomycin-induced fibrosis. These findings highlight a novel role for MMP28 in macrophage biology and pulmonary disease.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Matrix Metalloproteinases, Secreted/genetics , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Animals , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cytokines/pharmacology , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Macrophages/cytology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Matrix Metalloproteinases, Secreted/metabolism , Mice , Mice, Knockout , Monocytes/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Stress, Physiological/genetics , Toll-Like Receptors/antagonists & inhibitors
5.
Int J Mol Sci ; 14(12): 23858-71, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24322444

ABSTRACT

Sarcoidosis, a chronic granulomatous disease of unknown cause, has been linked to several environmental risk factors, among which are some that may favor carbon nanotube formation. Using gene array data, we initially observed that bronchoalveolar lavage (BAL) cells from sarcoidosis patients displayed elevated mRNA of the transcription factor, Twist1, among many M1-associated genes compared to healthy controls. Based on this observation we hypothesized that Twist1 mRNA and protein expression might become elevated in alveolar macrophages from animals bearing granulomas induced by carbon nanotube instillation. To address this hypothesis, wild-type and macrophage-specific peroxisome proliferator-activated receptor gamma (PPARγ) knock out mice were given oropharyngeal instillation of multiwall carbon nanotubes (MWCNT). BAL cells obtained 60 days later exhibited significantly elevated Twist1 mRNA expression in granuloma-bearing wild-type or PPARγ knock out alveolar macrophages compared to sham controls. Overall, Twist1 expression levels in PPARγ knock out mice were higher than those of wild-type. Concurrently, BAL cells obtained from sarcoidosis patients and healthy controls validated gene array data: qPCR and protein analysis showed significantly elevated Twist1 in sarcoidosis compared to healthy controls. In vitro studies of alveolar macrophages from healthy controls indicated that Twist1 was inducible by classical (M1) macrophage activation stimuli (LPS, TNFα) but not by IL-4, an inducer of alternative (M2) macrophage activation. Findings suggest that Twist1 represents a PPARγ-sensitive alveolar macrophage M1 biomarker which is induced by inflammatory granulomatous disease in the MWCNT model and in human sarcoidosis.


Subject(s)
Macrophages, Alveolar/metabolism , Twist-Related Protein 1/metabolism , Adult , Animals , Bronchoalveolar Lavage Fluid/cytology , Female , Humans , Macrophage Activation , Macrophages, Alveolar/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/toxicity , PPAR gamma/deficiency , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Sarcoidosis, Pulmonary/chemically induced , Sarcoidosis, Pulmonary/metabolism , Sarcoidosis, Pulmonary/pathology , Twist-Related Protein 1/genetics , Up-Regulation
6.
Respir Res ; 14: 7, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23343389

ABSTRACT

BACKGROUND: Although granulomatous inflammation is a central feature of many disease processes, cellular mechanisms of granuloma formation and persistence are poorly understood. Carbon nanoparticles, which can be products of manufacture or the environment, have been associated with granulomatous disease. This paper utilizes a previously described carbon nanoparticle granuloma model to address the issue of whether peroxisome proliferator-activated receptor gamma (PPARγ), a nuclear transcription factor and negative regulator of inflammatory cytokines might play a role in granulomatous lung disease. PPARγ is constitutively expressed in alveolar macrophages from healthy individuals but is depressed in alveolar macrophages of patients with sarcoidosis, a prototypical granulomatous disease. Our previous study of macrophage-specific PPARγ KO mice had revealed an intrinsically inflammatory pulmonary environment with an elevated pro-inflammatory cytokines profile as compared to wild-type mice. Based on such observations we hypothesized that PPARγ expression would be repressed in alveolar macrophages from animals bearing granulomas induced by MWCNT instillation. METHODS: Wild-type C57Bl/6 and macrophage-specific PPARγ KO mice received oropharyngeal instillations of multiwall carbon nanotubes (MWCNT) (100 µg). Bronchoalveolar lavage (BAL) cells, BAL fluids, and lung tissues were obtained 60 days post-instillation for analysis of granuloma histology and pro-inflammatory cytokines (osteopontin, CCL2, and interferon gamma [IFN-γ] mRNA and protein expression. RESULTS: In wild-type mice, alveolar macrophage PPARγ expression and activity were significantly reduced in granuloma-bearing animals 60 days after MWCNT instillation. In macrophage-specific PPARγ KO mice, granuloma formation was more extensive than in wild-type at 60 days after MWCNT instillation. PPARγ KO mice also demonstrated elevated pro-inflammatory cytokine expression in lung tissue, laser-microdissected lung granulomas, and BAL cells/fluids, at 60 days post MWCNT exposure. CONCLUSIONS: Overall, data indicate that PPARγ deficiency promotes inflammation and granuloma formation, suggesting that PPARγ functions as a negative regulator of chronic granulomatous inflammation.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , Granuloma, Respiratory Tract/immunology , Lung/immunology , Nanotubes, Carbon , PPAR gamma/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
J Innate Immun ; 4(5-6): 569-78, 2012.
Article in English | MEDLINE | ID: mdl-22759465

ABSTRACT

BACKGROUND: Dysfunctional immune responses characterize sarcoidosis, but the status of cathelicidin, a potent immunoregulatory and antimicrobial molecule, has not been established in clinical disease activity. METHODS: Alveolar macrophage cathelicidin expression was determined in biopsy-proven sarcoidosis patients classified clinically as 'severe' (requiring systemic treatment) or 'non-severe' (never requiring treatment). Bronchoalveolar lavage (BAL) cells from sarcoidosis patients and healthy controls were analyzed for mRNA expression of cathelicidin, vitamin D receptor (VDR) and the VDR coactivator steroid receptor coactivator-3 (SRC3) by quantitative PCR. Cathelicidin-derived peptide LL-37 was determined by immunocytochemistry. Serum calcidiol (25-hydroxyvitamin D2; vitD2) and calcitriol (1,25-dihydroxyvitamin D3; vitD3) were quantified. RESULTS: The results indicated reduced BAL cell expression of cathelicidin and SRC3 in severe but not non-severe sarcoidosis compared to controls. Serum levels of biologically active vitD3 in both severe and non-severe patients were within the control range even though vitD2 levels in both groups were below the recommended level (30 ng/ml). Sarcoidosis and control alveolar macrophages were studied in vitro to determine cathelicidin responses to vitD3 and tumor necrosis factor-α (TNFα), a vitD3 antagonist elevated in active sarcoidosis. Alveolar macrophage cathelicidin was stimulated by vitD3 but repressed by TNFα, which also repressed SRC3. CONCLUSIONS: These findings suggest that TNFα-mediated repression of SRC3 contributes to alveolar macrophage cathelicidin deficiency in severe sarcoidosis despite healthy vitD3 levels. Deficiency of cathelicidin, a multifunctional regulator of immune cells and proinflammatory cytokines, may impede resolution of inflammation in the lungs of patients with severe sarcoidosis.


Subject(s)
Antimicrobial Cationic Peptides/deficiency , Macrophages, Alveolar/metabolism , Nuclear Receptor Coactivator 3/metabolism , Sarcoidosis, Pulmonary/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Vitamin D/analogs & derivatives , Adult , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bronchoalveolar Lavage Fluid/cytology , Female , Humans , Male , Middle Aged , Nuclear Receptor Coactivator 3/genetics , Sarcoidosis, Pulmonary/metabolism , Severity of Illness Index , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vitamin D/metabolism , Young Adult , Cathelicidins
8.
Respir Res ; 13: 46, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22697800

ABSTRACT

RATIONALE: Pulmonary Alveolar Proteinosis (PAP) patients exhibit an acquired deficiency of biologically active granulocyte-macrophage colony stimulating factor (GM-CSF) attributable to GM-CSF specific autoantibodies. PAP alveolar macrophages are foamy, lipid-filled cells with impaired surfactant clearance and markedly reduced expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) and the PPARγ-regulated ATP binding cassette (ABC) lipid transporter, ABCG1. An open label proof of concept Phase II clinical trial was conducted in PAP patients using rituximab, a chimeric murine-human monoclonal antibody directed against B lymphocyte specific antigen CD20. Rituximab treatment decreased anti-GM-CSF antibody levels in bronchoalveolar lavage (BAL) fluid, and 7/9 patients completing the trial demonstrated clinical improvement as measured by arterial blood oxygenation. OBJECTIVES: This study sought to determine whether rituximab therapy would restore lipid metabolism in PAP alveolar macrophages. METHODS: BAL samples were collected from patients pre- and 6-months post-rituximab infusion for evaluation of mRNA and lipid changes. RESULTS: Mean PPARγ and ABCG1 mRNA expression increased 2.8 and 5.3-fold respectively (p ≤ 0.05) after treatment. Lysosomal phospholipase A2 (LPLA2) (a key enzyme in surfactant degradation) mRNA expression was severely deficient in PAP patients pre-treatment but increased 2.8-fold post-treatment. In supplemental animal studies, LPLA2 deficiency was verified in GM-CSF KO mice but was not present in macrophage-specific PPARγ KO mice compared to wild-type controls. Oil Red O intensity of PAP alveolar macrophages decreased after treatment, indicating reduced intracellular lipid while extracellular free cholesterol increased in BAL fluid. Furthermore, total protein and Surfactant protein A were significantly decreased in the BAL fluid post therapy. CONCLUSIONS: Reduction in GM-CSF autoantibodies by rituximab therapy improves alveolar macrophage lipid metabolism by increasing lipid transport and surfactant catabolism. Mechanisms may involve GM-CSF stimulation of alveolar macrophage ABCG1 and LPLA2 activities by distinct pathways.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Homeostasis , Macrophages, Alveolar/drug effects , Membrane Lipids/physiology , Pulmonary Alveolar Proteinosis/drug therapy , Pulmonary Alveoli/drug effects , Adult , Animals , Female , Homeostasis/drug effects , Homeostasis/immunology , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Rituximab
9.
Surgery ; 151(1): 107-12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21982524

ABSTRACT

BACKGROUND: Classically, activated macrophages in adipose tissue, liver, and muscle have been implicated in many conditions associated with obesity, including insulin resistance and the metabolic syndrome. Despite numerous pulmonary comorbidities and the sentinel role alveolar macrophages play in innate immunity and lung homeostasis, their activation status has not been examined in these patients. Peroxisome proliferator-activated receptor-gamma (PPAR-γ) has been shown to be a negative regulator of inflammation in addition to regulating lipid and glucose metabolism. PPAR-γ is expressed constitutively in healthy alveolar macrophages and decreased on activation. We hypothesized that PPAR-γ would be downregulated in alveolar macrophages from obese patients with obstructive sleep apnea (OSA) in the absence of overt lung disease. METHODS: Alveolar macrophages were obtained by bronchoalveolar lavage from obese individuals with and without OSA and healthy controls. RESULTS: Data indicated that PPAR-γ functional activity was decreased by 48% in obese with OSA and 26% without OSA (P < .05). In obese patients with OSA, PPAR-γ mRNA was decreased 2-fold compared with controls (P < .05), whereas obese patients without OSA, it was not different. Regardless of OSA, alveolar macrophages of obese patients demonstrated increased interleukin-6 mRNA. CONCLUSION: These findings are consistent with the presence of classic macrophage activation and an inflammatory lung environment. Data from this study suggest that alveolar macrophage dysfunction becomes aggravated in OSA and may increase pulmonary disease susceptibility.


Subject(s)
Macrophage Activation , Macrophages, Alveolar/metabolism , Obesity/immunology , PPAR gamma/metabolism , Sleep Apnea, Obstructive/immunology , Adult , Bronchoalveolar Lavage , Case-Control Studies , Cell Nucleus/metabolism , Female , Humans , Interleukin-6/metabolism , Male , Middle Aged , Obesity/metabolism , RNA, Messenger/metabolism , Sleep Apnea, Obstructive/metabolism , Transcription, Genetic , Young Adult
10.
Biochem Biophys Res Commun ; 415(2): 288-93, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22033401

ABSTRACT

We have shown decreased expression of the nuclear transcription factor, peroxisome proliferator-activated receptor-gamma (PPARγ) and the PPARγ-regulated ATP-binding cassette transporter G1 (ABCG1) in alveolar macrophages from patients with pulmonary alveolar proteinosis (PAP). PAP patients also exhibit neutralizing antibodies to granulocyte-macrophage colony stimulating factor (GM-CSF), an upregulator of PPARγ. In association with functional GM-CSF deficiency, PAP lung is characterized by surfactant-filled alveolar spaces and lipid-filled alveolar macrophages. Similar pathology characterizes GM-CSF knock-out (KO) mice. We reported previously that intratracheal instillation of a lentivirus (lenti)-PPARγ plasmid into GM-CSF KO animals elevated ABCG1 and reduced alveolar macrophage lipid accumulation. Here, we hypothesized that instillation of lenti-ABCG1 might be sufficient to decrease lipid accumulation and improve pulmonary function in GM-CSF KO mice. Animals received intratracheal instillation of lenti-ABCG1 or control lenti-enhanced Green Fluorescent Protein (eGFP) plasmids and alveolar macrophages were harvested 10 days later. Alveolar macrophage transduction efficiency was 79% as shown by lenti-eGFP fluorescence. Quantitative PCR analyses indicated a threefold (p=0.0005) increase in ABCG1 expression with no change of PPARγ or ABCA1 in alveolar macrophages of lenti-ABCG1 treated mice. ABCG1 was unchanged in control lenti-eGFP and PBS-instilled groups. Oil Red O staining detected reduced intracellular neutral lipid in alveolar macrophages from lenti-ABCG1 treated mice. Extracellular cholesterol and phospholipids were also decreased as shown by analysis of bronchoalveolar lavage fluid. Lung compliance was diminished in untreated GMCSF KO mice but improved significantly after lenti-ABCG1 treatment. Data demonstrate that in vivo instillation of lenti-ABCG1 in GM-CSF KO mice is sufficient to restore pulmonary homeostasis by: (1) upregulating ABCG1; (2) reducing intra and extracellular lipids; and (3) improving lung function. Results suggest that the ABCG1 lipid transporter is the key downstream target of GM-CSF-induced PPARγ necessary for surfactant catabolism.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lung/physiology , Phospholipids/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Animals , Humans , Lentivirus , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation
11.
Am J Respir Cell Mol Biol ; 45(4): 858-66, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21398620

ABSTRACT

Lung granulomas are associated with numerous conditions, including inflammatory disorders, exposure to environmental pollutants, and infection. Osteopontin is a chemotactic cytokine produced by macrophages, and is implicated in extracellular matrix remodeling. Furthermore, osteopontin is up-regulated in granulomatous disease, and osteopontin null mice exhibit reduced granuloma formation. Animal models currently used to investigate chronic lung granulomatous inflammation bear a pathological resemblance, but lack the chronic nature of human granulomatous disease. Carbon nanoparticles are generated as byproducts of combustion. Interestingly, experimental exposures to carbon nanoparticles induce pulmonary granuloma-like lesions. However, the recruited cellular populations and extracellular matrix gene expression profiles within these lesions have not been explored. Because of the rapid resolution of granulomas in current animal models, the mechanisms responsible for persistence have been elusive. To overcome the limitations of previous models, we investigated whether a model using multiwall carbon nanoparticles would resemble chronic human lung granulomatous inflammation. We hypothesized that pulmonary exposure to multiwall carbon nanoparticles would induce granulomas, elicit a macrophage and T-cell response, and mimic other granulomatous disorders with an up-regulation of osteopontin. This model demonstrates: (1) granulomatous inflammation, with macrophage and T-cell infiltration; (2) resemblance to the chronicity of human granulomas, with persistence up to 90 days; and (3) a marked elevation of osteopontin, metalloproteinases, and cell adhesion molecules in granulomatous foci isolated by laser-capture microdissection and in alveolar macrophages from bronchoalveolar lavage. The establishment of such a model provides an important platform for mechanistic studies on the persistence of granuloma.


Subject(s)
Granuloma/chemically induced , Lung/immunology , Nanotubes, Carbon , Pneumonia/chemically induced , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Adhesion Molecules/genetics , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation , Granuloma/genetics , Granuloma/immunology , Granuloma/metabolism , Granuloma/pathology , Inflammation Mediators/metabolism , Integrins/genetics , Lasers , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/immunology , Metalloproteases/genetics , Mice , Mice, Inbred C57BL , Microdissection/instrumentation , Osteopontin/genetics , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , RNA, Messenger/metabolism , T-Lymphocytes/immunology , Time Factors
12.
Am J Pathol ; 176(1): 64-73, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008147

ABSTRACT

Tissue inhibitor of metalloproteinases 3 (TIMP3) inhibits not only matrix metalloproteinases but also a disintegrin and metalloproteinase domain family members and thus contributes to controlling diverse processes mediated by proteolysis. We used Timp3(-/-) mice to assess the role of this inhibitor in acute lung injury. After bleomycin-induced injury, inflammation, as indicated by the influx of neutrophils in bronchoalveolar lavage (BAL), peaked at 7 days post-injury in the wild-type mice and began to wane thereafter; however, in Timp3(-/-) mice, inflammation persisted up to 28 days. Furthermore, although the level of chemokines in BAL and lung homogenate was similar in both genotypes, BAL from Timp3(-/-) mice 7, 14, and 28 days post-injury had increased neutrophil chemotactic activity compared with wild-type BAL. At day 14, a higher percentage of apoptotic neutrophils were present in wild-type mice compared with Timp3(-/-) mice, further suggesting that TIMP3 constrains continued neutrophil influx. In addition, total matrix metalloproteinase activity was increased in lungs from Timp3(-/-) mice, and treatment of mice with a synthetic inhibitor of metalloproteinases rescued the enhanced neutrophilia phenotype. These data demonstrate that TIMP3 regulates neutrophil influx in the lung following injury through its ability to inhibit metalloproteinase activity and indicates that TIMP3 functions to promote the resolution of inflammation in the lung.


Subject(s)
Acute Lung Injury/complications , Acute Lung Injury/pathology , Inflammation/enzymology , Inflammation/etiology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Acute Lung Injury/enzymology , Animals , Biomarkers/metabolism , Bleomycin , Bronchoalveolar Lavage Fluid/cytology , Chemotaxis , Inflammation/pathology , Matrix Metalloproteinases/metabolism , Mice , Neutrophils/cytology , Peroxidase/metabolism , Tissue Extracts , Tissue Inhibitor of Metalloproteinase-3/deficiency
13.
Am J Pathol ; 175(6): 2319-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893044

ABSTRACT

The E-cadherin receptor CD103 (alpha(E)beta(7)-integrin) is expressed on specific populations of pulmonary dendritic cells (DC) and T cells. However, CD103 function in the lung is not well understood. Matrilysin (MMP-7) expression is increased in lung injury and cleaves E-cadherin from injured lung epithelium. Thus, to assess matrilysin effects on CD103-E-cadherin interactions in lung injury, wild-type, CD103(-/-), and Mmp7(-/-) mice, in which E-cadherin isn't cleaved in the lung, were treated with bleomycin or bleomycin with nFMLP to reverse the defect in acute neutrophil influx seen in Mmp7(-/-) mice. Pulmonary CD103(+) DC were significantly increased in injured wild-type compared with Mmp7(-/-) mice, and CD103(+) leukocytes showed significantly enhanced interaction with E-cadherin on injured wild-type epithelium than with Mmp7(-/-) epithelium in vitro and in vivo. Bleomycin-treated CD103(-/-) mice had persistent neutrophilic inflammation, increased fibrosis, and increased mortality compared with wild-type mice, a phenotype that was partially recapitulated in bleomycin/nFMLP-treated Mmp7(-/-) mice. Soluble E-cadherin increased IL-12 and IL-10 and reduced IL-6 mRNA expression in wild-type bone marrow-derived DC but not in CD103(-/-) bone marrow-derived DC. Similar mRNA patterns were seen in lungs of bleomycin-injured wild-type, but not CD103(-/-) or Mmp7(-/-), mice. In conclusion, matrilysin regulates pulmonary localization of DC that express CD103, and E-cadherin cleavage may activate CD103(+) DC to limit inflammation and inhibit fibrosis.


Subject(s)
Dendritic Cells/metabolism , Lung Injury/metabolism , Matrix Metalloproteinase 7/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/metabolism , Animals , Antigens, CD/biosynthesis , Blotting, Western , Cadherins/immunology , Cadherins/metabolism , Dendritic Cells/immunology , Flow Cytometry , Immunohistochemistry , Integrin alpha Chains/biosynthesis , Lung Injury/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Pneumonia/immunology , Pulmonary Fibrosis/immunology , Reverse Transcriptase Polymerase Chain Reaction
14.
Curr Opin Pulm Med ; 15(5): 491-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19561506

ABSTRACT

PURPOSE OF REVIEW: This review discusses the most recent clinical and basic research literature on pulmonary alveolar proteinosis (PAP) as it relates to pathogenesis, diagnosis, and management. RECENT FINDINGS: The discovery of Granulocyte macrophage-colony stimulating factor (GM-CSF) and the alveolar macrophage as critical regulators of surfactant protein and lipid homeostasis has led to significant advances in PAP. Adults affected by PAP have circulating neutralizing anti-GM-CSF antibodies. Reduced localized GM-CSF activity in the lung (from neutralizing anti-GM-CSF antibodies), decreases alveolar macrophage surfactant degradation with surfactant excess and accumulation. Cause, source of antibodies or downstream effects of GM-CSF deficiency is speculative. GM-CSF antibodies above a threshold level have proved to be a useful diagnostic test. Research towards therapy has focused on improving the technique for therapeutic whole lung lavage as well as overcoming effects of neutralizing anti-GM-CSF, which include GM-CSF therapy (systemic and inhaled) and anecdotal reports of anti-B cell therapy. Whereas this approach has been somewhat successful for primary PAP, other causes of PAP (i.e. alveolar macrophage dysfunction, surfactant protein alterations) are still without therapy. SUMMARY: Understanding of the pathogenesis of PAP has greatly increased in the last decade; study has brought better comprehension of lung biology and recognition of the critical role for GM-CSF and alveolar macrophage in surfactant clearance. Balance between resident immune cell population and normal lung function still needs further study. Resident alveolar macrophages have an essential role in surfactant homeostasis. With this knowledge more effective diagnostic tests (e.g. anti-GM-CSF antibody) and therapies for PAP are under investigation.


Subject(s)
B-Lymphocytes/immunology , Bronchoalveolar Lavage/methods , Immunity, Cellular/immunology , Immunosuppressive Agents/therapeutic use , Pulmonary Alveolar Proteinosis , Biopsy , Bronchoalveolar Lavage Fluid/chemistry , Bronchoscopy , Diagnosis, Differential , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Macrophages, Alveolar/metabolism , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Alveolar Proteinosis/therapy , Tomography, X-Ray Computed
15.
Am J Respir Crit Care Med ; 179(3): 212-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19011156

ABSTRACT

RATIONALE: The insulin-like growth factor-I (IGF-I) pathway is an important determinant of survival and proliferation in many cells. However, little is known about the role of the IGF-I pathway in lung injury. We previously showed elevated levels of IGF-I in bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome. Furthermore, immunodepletion of IGF from acute respiratory distress syndrome bronchoalveolar lavage increased fibroblast apoptosis. OBJECTIVES: We examined the effect of blockade of type 1 IGF tyrosine kinase receptor (IGF-IR) in a murine model of bleomycin-induced lung injury and fibrosis. METHODS: Mice were treated with a monoclonal antibody against the IGF-I receptor (A12) or vehicle after intratracheal bleomycin instillation. MEASUREMENTS AND MAIN RESULTS: Mice treated with A12 antibody had significantly improved survival after bleomycin injury compared with control mice. Both groups of mice had a similar degree of fibrosis on days 7 and 14, but by Day 28 the A12-treated group had significantly less fibrosis. Delayed treatment with A12 also resulted in decreased fibrosis. A12-treated mice had significantly decreased apoptotic cells on Day 28 compared with control mice. We confirmed that A12 treatment induced mouse lung fibroblast apoptosis in vitro. In addition, IGF-I increased lung fibroblast migration. The primary pathway activated by IGF-I in lung fibroblasts was the insulin receptor substrate-2/phosphatidylinositol 3-kinase/Akt axis. CONCLUSIONS: IGF-I regulated survival and migration of fibrogenic cells in the lung. Blockade of the IGF pathway increased fibroblast apoptosis and subsequent resolution of pulmonary fibrosis. Thus, IGF-IR may be a potential target for treatment of lung injury and fibrosis.


Subject(s)
Acute Lung Injury/drug therapy , Antibodies, Monoclonal/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Apoptosis , Bleomycin/toxicity , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/pathology , Follow-Up Studies , Gene Expression , Immunohistochemistry , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Treatment Outcome
16.
Gene Expr Patterns ; 8(7-8): 486-93, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18639653

ABSTRACT

Lung remodeling requires active collagen deposition and degradation. Urokinase plasminogen activator receptor-associated protein (uPARAP), or Endo 180, is a cell-surface receptor for collagens, which leads to collagen internalization and degradation. Thus, uPARAP-mediated collagen degradation is an additional pathway for matrix remodeling in addition to matrix remodeling mediated by matrix metalloproteinases and cathepsins. Using immunohistochemistry, we demonstrate extensive uPARAP expression in the mesenchyme throughout murine lung development. By immunofluorescence, we demonstrate significant overlap of uPARAP expression with collagen IV expression, but minimal overlap with collagen I expression in the developing murine lung. Finally, we compared lung development between wild-type and uPARAP(-/-) mice, and found no significant histologic differences, indicating the presence of alternative collagen degradation pathways during murine lung development.


Subject(s)
Lung/embryology , Membrane Glycoproteins/metabolism , Mice/embryology , Receptors, Cell Surface/metabolism , Animals , Collagen Type I/metabolism , Collagen Type IV/metabolism , Lung/metabolism , Mice/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...