Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Physiol ; 596(8): 1397-1417, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29380370

ABSTRACT

KEY POINTS: The mechanisms by which bacteria alter endothelial cell phenotypes and programme inflammatory angiogenesis remain unclear. In lung endothelial cells, we demonstrate that toll-like receptor 4 (TLR4) signalling induces activation of forkhead box protein C2 (FOXC2), a transcriptional factor implicated in lymphangiogenesis and endothelial specification, in an extracellular signal-regulated kinase (ERK)-dependent manner. TLR4-ERK-FOXC2 signalling regulates expression of the Notch ligand DLL4 and signals inflammatory angiogenesis in vivo and in vitro. Our work reveals a novel link between endothelial immune signalling (TLR pathway) and a vascular transcription factor, FOXC2, that regulates embryonic vascular development. This mechanism is likely to be relevant to pathological angiogenesis complicating inflammatory diseases in humans. ABSTRACT: Endothelial cells (ECs) mediate a specific and robust immune response to bacteria in sepsis through the activation of toll-like receptor (TLR) signalling. The mechanisms by which bacterial ligands released during sepsis programme EC specification and altered angiogenesis remain unclear. We postulated that the forkhead box protein C2 (FOXC2) transcriptional factor directs EC cell-fate decisions and angiogenesis during TLR signalling. In human lung ECs, lipopolysaccharide (LPS) induced ERK phosphorylation, FOXC2, and delta-like 4 (DLL4, the master regulator of sprouting angiogenesis expression) in a TLR4-dependent manner. LPS-mediated ERK phosphorylation resulted in FOXC2-ERK protein ligation, ERK-dependent FOXC2 serine and threonine phosphorylation, and subsequent activation of DLL4 gene expression. Chemical inhibition of ERK or ERK-2 dominant negative transfection disrupted LPS-mediated FOXC2 phosphorylation and transcriptional activation of FOXC2. FOXC2-siRNA or ERK-inhibition attenuated LPS-induced DLL4 expression and angiogenic sprouting in vitro. In vivo, intraperitoneal LPS induced ERK and FOXC2 phosphorylation, FOXC2 binding to DLL4 promoter, and FOXC2/DLL4 expression in the lung. ERK-inhibition suppressed LPS-induced FOXC2 phosphorylation, FOXC2-DLL4 promoter binding, and induction of FOXC2 and DLL4 in mouse lung ECs. LPS induced aberrant retinal angiogenesis and DLL4 expression in neonatal mice, which was attenuated with ERK inhibition. FOXC2+/- mice treated with LPS showed a mitigated increase in FOXC2 and DLL4 compared to FOXC2+/+ mice. These data reveal a new mechanism (TLR4-ERK-FOXC2-DLL4) by which sepsis-induced EC TLR signalling programmes EC specification and altered angiogenesis.


Subject(s)
Endothelial Cells/immunology , Forkhead Transcription Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Signal Transduction , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Differentiation , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/toxicity , Lung/blood supply , Lung/embryology , Lung/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
2.
Am J Respir Crit Care Med ; 189(3): 301-13, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24392884

ABSTRACT

RATIONALE: Goblet cell metaplasia accompanies common pulmonary disorders that are prone to recurrent viral infections. Mechanisms regulating both goblet cell metaplasia and susceptibility to viral infection associated with chronic lung diseases are incompletely understood. OBJECTIVES: We sought to identify the role of the transcription factor FOXA3 in regulation of goblet cell metaplasia and pulmonary innate immunity. METHODS: FOXA3 was identified in airways from patients with asthma and chronic obstructive pulmonary disease. We produced transgenic mice conditionally expressing Foxa3 in airway epithelial cells and developed human bronchial epithelial cells expressing Foxa3. Foxa3-regulated genes were identified by immunostaining, Western blotting, and RNA analysis. Direct binding of FOXA3 to target genes was identified by chromatin immunoprecipitation sequencing correlated with RNA sequencing. MEASUREMENTS AND MAIN RESULTS: FOXA3 was highly expressed in airway goblet cells from patients with asthma and chronic obstructive pulmonary disease. FOXA3 was induced by either IL-13 or rhinovirus. Foxa3 induced goblet cell metaplasia and enhanced expression of a network of genes mediating mucus production. Paradoxically, FOXA3 inhibited rhinovirus-induced IFN production, IRF-3 phosphorylation, and IKKε expression and inhibited viral clearance and expression of genes required for antiviral defenses, including MDA5, RIG-I, TLR3, IRF7/9, and nuclear factor-κB. CONCLUSIONS: FOXA3 induces goblet cell metaplasia in response to infection or Th2 stimulation. Suppression of IFN signaling by FOXA3 provides a plausible mechanism that may serve to limit ongoing Th1 inflammation during the resolution of acute viral infection; however, inhibition of innate immunity by FOXA3 may contribute to susceptibility to viral infections associated with chronic lung disorders accompanied by chronic goblet cell metaplasia.


Subject(s)
Asthma/metabolism , Goblet Cells/pathology , Hepatocyte Nuclear Factor 3-gamma/metabolism , Immunity, Innate/physiology , Picornaviridae Infections/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Animals , Asthma/complications , Asthma/immunology , Asthma/pathology , Biomarkers/metabolism , Blotting, Western , Chromatin Immunoprecipitation , Disease Susceptibility , Goblet Cells/immunology , Goblet Cells/metabolism , Hepatocyte Nuclear Factor 3-gamma/immunology , Humans , Interferons/metabolism , Metaplasia , Mice , Mice, Transgenic , Picornaviridae Infections/etiology , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Rhinovirus , Sequence Analysis, RNA , Th1-Th2 Balance
3.
Proc Natl Acad Sci U S A ; 109(41): 16630-5, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23012424

ABSTRACT

Airway mucus plays a critical role in clearing inhaled toxins, particles, and pathogens. Diverse toxic, inflammatory, and infectious insults induce airway mucus secretion and goblet cell metaplasia to preserve airway sterility and homeostasis. However, goblet cell metaplasia, mucus hypersecretion, and airway obstruction are integral features of inflammatory lung diseases, including asthma, chronic obstructive lung disease, and cystic fibrosis, which cause an immense burden of morbidity and mortality. These chronic lung diseases are united by susceptibility to microbial colonization and recurrent airway infections. Whether these twinned phenomena (mucous metaplasia, compromised host defenses) are causally related has been unclear. Here, we demonstrate that SAM pointed domain ETS factor (SPDEF) was induced by rhinoviral infection of primary human airway cells and that cytoplasmic activities of SPDEF, a transcriptional regulator of airway goblet cell metaplasia, inhibited Toll-like receptor (TLR) activation of epithelial cells. SPDEF bound to and inhibited activities of TLR signaling adapters, MyD88 and TRIF, inhibiting MyD88-induced cytokine production and TRIF-induced interferon ß production. Conditional expression of SPDEF in airway epithelial cells in vivo inhibited LPS-induced neutrophilic infiltration and bacterial clearance. SPDEF-mediated inhibition of both TLR and type I interferon signaling likely protects the lung against inflammatory damage when inciting stimuli are not eradicated. Present findings provide, at least in part, a molecular explanation for increased susceptibility to infection in lung diseases associated with mucous metaplasia and a mechanism by which patients with florid mucous metaplasia may tolerate microbial burdens that are usually associated with fulminant inflammatory disease in normal hosts.


Subject(s)
Epithelial Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Blotting, Western , Doxycycline/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression/drug effects , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interleukin-13/pharmacology , Lipopolysaccharides/pharmacology , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lung Diseases/pathology , Metaplasia , Mice , Microscopy, Confocal , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Binding , Proto-Oncogene Proteins c-ets/genetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Rhinovirus/physiology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L726-35, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24508732

ABSTRACT

A number of growth factors and signaling pathways regulate matrix deposition and fibroblast proliferation in the lung. The epidermal growth factor receptor (EGFR) family of receptors and the transforming growth factor-ß (TGF-ß) family are active in diverse biological processes and are central mediators in the initiation and maintenance of fibrosis in many diseases. Transforming growth factor-α (TGF-α) is a ligand for the EGFR, and doxycycline (Dox)-inducible transgenic mice conditionally expressing TGF-α specifically in the lung epithelium develop progressive fibrosis accompanied with cachexia, changes in lung mechanics, and marked pleural thickening. Although recent studies demonstrate that EGFR activation modulates the fibroproliferative effects involved in the pathogenesis of TGF-ß induced pulmonary fibrosis, in converse, the direct role of EGFR induction of the TGF-ß pathway in the lung is unknown. The αvß6 integrin is an important in vivo activator of TGF-ß activation in the lung. Immunohistochemical analysis of αvß6 protein expression and bronchoalveolar analysis of TGF-ß pathway signaling indicates activation of the αvß6/TGF-ß pathway only at later time points after lung fibrosis was already established in the TGF-α model. To determine the contribution of the αvß6/TGF-ß pathway on the progression of established fibrotic disease, TGF-α transgenic mice were administered Dox for 4 wk, which leads to extensive fibrosis; these mice were then treated with a function-blocking anti-αvß6 antibody with continued administration of Dox for an additional 4 wk. Compared with TGF-α transgenic mice treated with control antibody, αvß6 inhibition significantly attenuated pleural thickening and altered the decline in lung mechanics. To test the effects of genetic loss of the ß6 integrin, TGF-α transgenic mice were mated with ß6-null mice and the degree of fibrosis was compared in adult mice following 8 wk of Dox administration. Genetic ablation of the ß6 integrin attenuated histological and physiological changes in the lungs of TGF-α transgenic mice although a significant degree of fibrosis still developed. In summary, inhibition of the ß6 integrin led to a modest, albeit significant, effect on pleural thickening and lung function decline observed with TGF-α-induced pulmonary fibrosis. These data support activation of the αvß6/TGF-ß pathway as a secondary effect contributing to TGF-α-induced pleural fibrosis and suggest a complex contribution of multiple mediators to the maintenance of progressive fibrosis in the lung.


Subject(s)
Integrins/antagonists & inhibitors , Pulmonary Fibrosis/pathology , Transforming Growth Factor alpha/pharmacology , Animals , Anti-Bacterial Agents/toxicity , Antibodies, Neutralizing , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Bronchoalveolar Lavage , Collagen , Doxycycline/toxicity , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoenzyme Techniques , Integrins/genetics , Integrins/metabolism , Male , Mice , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology , Uteroglobin/physiology
5.
Am J Respir Cell Mol Biol ; 49(5): 845-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23795648

ABSTRACT

Pulmonary surfactant protein-C (SP-C) gene-targeted mice (Sftpc(-/-)) develop progressive lung inflammation and remodeling. We hypothesized that SP-C deficiency reduces the ability to suppress repetitive inflammatory injury. Sftpc(+/+) and Sftpc(-/-) mice given three doses of bacterial LPS developed airway and airspace inflammation, which was more intense in the Sftpc(-/-) mice at 3 and 5 days after the final dose. Compared with Sftpc(+/+)mice, inflammatory injury persisted in the lungs of Sftpc(-/-) mice 30 days after the final LPS challenge. Sftpc(-/-) mice showed LPS-induced airway goblet cell hyperplasia with increased detection of Sam pointed Ets domain and FoxA3 transcription factors. Sftpc(-/-) type II alveolar epithelial cells had increased cytokine expression after LPS exposure relative to Sftpc(+/+) cells, indicating that type II cell dysfunction contributes to inflammatory sensitivity. Microarray analyses of isolated type II cells identified a pattern of enhanced expression of inflammatory genes consistent with an intrinsic low-level inflammation resulting from SP-C deficiency. SP-C-containing clinical surfactant extract (Survanta) or SP-C/phospholipid vesicles blocked LPS signaling through the LPS receptor (Toll-like receptor [TLR] 4/CD14/MD2) in human embryonic kidney 293T cells, indicating that SP-C blocks LPS-induced cytokine production by a TLR4-dependent mechanism. Phospholipid vesicles alone did not modify the TLR4 response. In vivo deficiency of SP-C leads to inflammation, increased cytokine production by type II cells, and persistent inflammation after repetitive LPS stimulation.


Subject(s)
Endotoxins , Lung/metabolism , Peptides/deficiency , Pneumonia/metabolism , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Biological Products/pharmacology , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/pathology , HEK293 Cells , Hepatocyte Nuclear Factor 3-gamma/metabolism , Humans , Hyperplasia , Immunity, Innate , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins , Lipopolysaccharide Receptors/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Peptides/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Proto-Oncogene Proteins c-ets/metabolism , Pulmonary Surfactant-Associated Protein C , Signal Transduction , Time Factors , Toll-Like Receptor 4/metabolism
6.
Respir Res ; 14: 19, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23399055

ABSTRACT

BACKGROUND: Individuals with deficiencies of pulmonary surfactant protein C (SP-C) develop interstitial lung disease (ILD) that is exacerbated by viral infections including respiratory syncytial virus (RSV). SP-C gene targeted mice (Sftpc -/-) lack SP-C, develop an ILD-like disease and are susceptible to infection with RSV. METHODS: In order to determine requirements for correction of RSV induced injury we have generated compound transgenic mice where SP-C expression can be induced on the Sftpc -/- background (SP-C/Sftpc -/-) by the administration of doxycycline (dox). The pattern of induced SP-C expression was determined by immunohistochemistry and processing by Western blot analysis. Tissue and cellular inflammation was measured following RSV infection and the RSV-induced cytokine response of isolated Sftpc +/+ and -/- type II cells determined. RESULTS: After 5 days of dox administration transgene SP-C mRNA expression was detected by RT-PCR in the lungs of two independent lines of bitransgenic SP-C/Sftpc -/- mice (lines 55.3 and 54.2). ProSP-C was expressed in the lung, and mature SP-C was detected by Western blot analysis of the lavage fluid from both lines of SP-C/Sftpc -/- mice. Induced SP-C expression was localized to alveolar type II cells by immunostaining with an antibody to proSP-C. Line 55.3 SP-C/Sftpc -/- mice were maintained on or off dox for 7 days and infected with 2.6x107 RSV pfu. On day 3 post RSV infection total inflammatory cell counts were reduced in the lavage of dox treated 55.3 SP-C/Sftpc -/- mice (p = 0.004). The percentage of neutrophils was reduced (p = 0.05). The viral titers of lung homogenates from dox treated 55.3 SP-C/Sftpc -/- mice were decreased relative to 55.3 SP-C/Sftpc -/- mice without dox (p = 0.01). The cytokine response of Sftpc -/- type II cells to RSV was increased over that of Sftpc +/+ cells. CONCLUSIONS: Transgenic restoration of SP-C reduced inflammation and improved viral clearance in the lungs of SP-C deficient mice. The loss of SP-C in alveolar type II cells compromises their response to infection. These findings show that the restoration of SP-C in Sftpc -/- mice in response to RSV infection is a useful model to determine parameters for therapeutic intervention.


Subject(s)
Lung Injury/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Viruses , Animals , Cells, Cultured , Down-Regulation/genetics , Lung Injury/genetics , Lung Injury/prevention & control , Mice , Mice, 129 Strain , Mice, Transgenic , Pulmonary Surfactant-Associated Protein C/biosynthesis , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/prevention & control , Viral Load/methods
7.
Respir Res ; 13: 51, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726462

ABSTRACT

BACKGROUND: Resistin-like molecule alpha or found in inflammatory zone protein (Fizz1) is increased in pulmonary epithelial cells and also in limited amounts by other lung cells during various lung injuries and fibrosis. However, the direct role of Fizz1 produced in the pulmonary epithelium has not been determined. METHODS: Fizz1 Transgenic mice (CCSP/Fizz1) were generated that overexpress Fizz1 in the lung epithelium under the control of a doxycycline (Dox) inducible lung epithelial cell specific promoter Scgb1a1 (Clara cell secretory protein, CCSP). Histology and FACS analysis of lung cells were used to identify the direct effects of Fizz1 in the transgenic mice (Dox treated) when compared with control (CCSP/-) mice. Intratracheal bleomycin sulfate or silica in saline and saline alone were used to study the role of Fizz1 during bleomycin- and silica-induced pulmonary fibrosis in CCSP/Fizz1 and CCSP/- mice. Weight change, pulmonary inflammation, and fibrosis were assessed 10 days post bleomycin or 28 days post silica challenge. RESULTS: When CCSP/Fizz1 mice were fed Dox food, elevated Fizz1 protein was detected in lung homogenates by western blot. Lungs of mice in which Fizz1 was induced in the epithelium contained increased lung cells staining for CD11c and F4/80 by FACS analysis consistent with increased dendritic cells however, no changes were observed in the percentage of interstitial macrophages compared to CCSP/- controls. No significant changes were found in the lung histology of CCSP/Fizz1 mice after up to 8 weeks of overexpression compared to CCSP/- controls. Overexpression of Fizz1 prior to challenge or following challenge with bleomycin or silica did not significantly alter airway inflammation or fibrosis compared to control mice. CONCLUSIONS: The current study demonstrates that epithelial cell derived Fizz1 is sufficient to increase the bone-marrow derived dendritic cells in the lungs, but it is not sufficient to cause lung fibrosis or alter chemical or particle-induced fibrosis.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Lung/metabolism , Lung/pathology , Pulmonary Fibrosis , Animals , Dendritic Cells/pathology , Female , Mice , Mice, Transgenic , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology
8.
Am J Respir Cell Mol Biol ; 44(2): 175-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20348208

ABSTRACT

Surfactant protein A (SP-A) mediates innate immune cell responses to LPS, a cell wall component of gram-negative bacteria that is found ubiquitously in the environment and is associated with adverse health effects. Inhaled LPS induces lung inflammation and increases airway responsiveness (AR). However, the role of SP-A in mediating LPS-induced AR is not well-defined. Nitric oxide (NO) is described as a potent bronchodilator, and previous studies showed that SP-A modulates the LPS-induced production of NO. Hence, we tested the hypothesis that increased AR, observed in response to aerosolized LPS exposure, would be significantly reduced in an SP-A-deficient condition. Wild-type (WT) and SP-A null (SP-A(-/-)) mice were challenged with aerosolized LPS. Results indicate that despite similar inflammatory indices, LPS-treated SP-A(-/-) mice had attenuated AR after methacholine challenge, compared with WT mice. The attenuated AR could not be attributed to inherent differences in SP-D concentrations or airway smooth muscle contractile and relaxation properties, because these measures were similar between WT and SP-A(-/-) mice. LPS-treated SP-A(-/-) mice, however, had elevated nitrite concentrations, inducible nitric oxide synthase (iNOS) expression, and NOS activity in their lungs. Moreover, the administration of the iNOS-specific inhibitor 1400W completely abrogated the attenuated AR. Thus, when exposed to aerosolized LPS, SP-A(-/-) mice demonstrate a relative airway hyporesponsiveness that appears to be mediated at least partly via an iNOS-dependent mechanism. These findings may have clinical significance, because recent studies reported associations between surfactant protein polymorphisms and a variety of lung diseases.


Subject(s)
Lipopolysaccharides/pharmacology , Lung/immunology , Lung/physiopathology , Nitric Oxide/physiology , Pulmonary Surfactant-Associated Protein A/deficiency , Animals , Immunity, Innate , Lung/drug effects , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein A/physiology , Pulmonary Surfactant-Associated Protein D/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L414-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21224214

ABSTRACT

Increases in the epidermal growth factor receptor (EGFR) have been associated with the severity of airway thickening in chronic asthmatic subjects, and EGFR signaling is induced by asthma-related cytokines and inflammation. The goal of this study was to determine the role of EGFR signaling in a chronic allergic model of asthma and specifically in epithelial cells, which are increasingly recognized as playing an important role in asthma. EGFR activation was assessed in mice treated with intranasal house dust mite (HDM) for 3 wk. EGFR signaling was inhibited in mice treated with HDM for 6 wk, by using either the drug erlotinib or a genetic approach that utilizes transgenic mice expressing a mutant dominant negative epidermal growth factor receptor in the lung epithelium (EGFR-M mice). Airway hyperreactivity (AHR) was assessed by use of a flexiVent system after increasing doses of nebulized methacholine. Airway smooth muscle (ASM) thickening was measured by morphometric analysis. Sensitization to HDM (IgG and IgE), inflammatory cells, and goblet cell changes were also assessed. Increased EGFR activation was detected in HDM-treated mice, including in bronchiolar epithelial cells. In mice exposed to HDM for 6 wk, AHR and ASM thickening were reduced after erlotinib treatment and in EGFR-M mice. Sensitization to HDM and inflammatory cell counts were similar in all groups, except neutrophil counts, which were lower in the EGFR-M mice. Goblet cell metaplasia with HDM treatment was reduced by erlotinib, but not in EGFR-M transgenic mice. This study demonstrates that EGFR signaling, especially in the airway epithelium, plays an important role in mediating AHR and remodeling in a chronic allergic asthma model.


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Bronchial Hyperreactivity/complications , Epithelial Cells/enzymology , ErbB Receptors/metabolism , Signal Transduction , Animals , Asthma/complications , Asthma/parasitology , Asthma/pathology , Bronchial Hyperreactivity/parasitology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/physiopathology , Chronic Disease , Disease Models, Animal , Enzyme Activation , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , Goblet Cells/pathology , Inflammation/complications , Inflammation/pathology , Lung/parasitology , Lung/pathology , Lung/physiopathology , Metaplasia , Mice , Muscle, Smooth/pathology , Pyroglyphidae/physiology
10.
Am J Pathol ; 176(2): 679-86, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20042669

ABSTRACT

Transforming growth factor-alpha (TGFalpha) is a ligand for the epidermal growth factor receptor (EGFR). EGFR activation is associated with fibroproliferative processes in human lung disease and animal models of pulmonary fibrosis. EGFR signaling activates several intracellular signaling pathways including phosphatidylinositol 3'-kinase (PI3K). We previously showed that induction of lung-specific TGFalpha expression in transgenic mice caused progressive pulmonary fibrosis over a 4-week period. The increase in levels of phosphorylated Akt, detected after 1 day of doxycycline-induced TGFalpha expression, was blocked by treatment with the PI3K inhibitor, PX-866. Daily administration of PX-866 during TGFalpha induction prevented increases in lung collagen and airway resistance as well as decreases in lung compliance. Treatment of mice with oral PX-866 4 weeks after the induction of TGFalpha prevented additional weight loss and further increases in total collagen, and attenuated changes in pulmonary mechanics. These data show that PI3K is activated in TGFalpha/EGFR-mediated pulmonary fibrosis and support further studies to determine the role of PI3K activation in human lung fibrotic disease, which could be amenable to targeted therapy.


Subject(s)
Gonanes/pharmacology , Gonanes/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Transforming Growth Factor alpha , Administration, Oral , Animals , Disease Progression , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gonanes/administration & dosage , Mice , Mice, Transgenic , Oncogene Protein v-akt/metabolism , Phosphorylation/drug effects , Uteroglobin/genetics
11.
J Clin Invest ; 117(4): 978-88, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17347682

ABSTRACT

Goblet cell hyperplasia and mucous hypersecretion contribute to the pathogenesis of chronic pulmonary diseases including cystic fibrosis, asthma, and chronic obstructive pulmonary disease. In the present work, mouse SAM pointed domain-containing ETS transcription factor (SPDEF) mRNA and protein were detected in subsets of epithelial cells lining the trachea, bronchi, and tracheal glands. SPDEF interacted with the C-terminal domain of thyroid transcription factor 1, activating transcription of genes expressed selectively in airway epithelial cells, including Sftpa, Scgb1a1, Foxj1, and Sox17. Expression of Spdef in the respiratory epithelium of adult transgenic mice caused goblet cell hyperplasia, inducing both acidic and neutral mucins in vivo, and stainined for both acidic and neutral mucins in vivo. SPDEF expression was increased at sites of goblet cell hyperplasia caused by IL-13 and dust mite allergen in a process that was dependent upon STAT-6. SPDEF was induced following intratracheal allergen exposure and after Th2 cytokine stimulation and was sufficient to cause goblet cell differentiation of Clara cells in vivo.


Subject(s)
Goblet Cells/physiology , Hyperplasia/physiopathology , Proto-Oncogene Proteins c-ets/genetics , Respiratory Mucosa/physiology , Animals , Binding Sites , Cell Differentiation , Cell Line , Cytokines/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation , Genetic Variation , Goblet Cells/cytology , History, 16th Century , Humans , Lung/growth & development , Lung/physiology , Lung Neoplasms , Mice , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-ets/physiology , RNA, Messenger/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology , Th2 Cells/physiology , Trachea/physiology , Transcription Factors , Transcription, Genetic
12.
Am J Respir Crit Care Med ; 180(9): 834-45, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19661247

ABSTRACT

RATIONALE: Induced mainly by cigarette smoking, chronic obstructive pulmonary disease (COPD) is a global public health problem characterized by progressive difficulty in breathing and increased mucin production. Previously, we reported that acrolein levels found in COPD sputum could activate matrix metalloproteinase-9 (MMP9). OBJECTIVES: To determine whether acrolein increases expression and activity of MMP14, a critical membrane-bound endopeptidase that can initial a MMP-activation cascade. METHODS: MMP14 activity and adduct formation were measured following direct acrolein treatment. MMP14 expression and activity was measured in human airway epithelial cells. MMP14 immunohistochemistry was performed with COPD tissue, and in acrolein- or tobacco-exposed mice. MEASUREMENTS AND MAIN RESULTS: In a cell-free system, acrolein, in concentrations equal to those found in COPD sputum, directly adducted cysteine 319 in the MMP14 hemopexin-like domain and activated MMP14. In cells, acrolein increased MMP14 activity, which was inhibited by a proprotein convertase inhibitor, hexa-d-arginine. In the airway epithelium of COPD subjects, immunoreactive MMP14 protein increased. In mouse lung, acrolein or tobacco smoke increased lung MMP14 activity and protein. In cells, acrolein-induced MMP14 transcripts were inhibited by an epidermal growth factor receptor (EGFR) neutralizing antibody, EGFR kinase inhibitor, metalloproteinase inhibitor, or mitogen-activated protein kinase (MAPK) 3/2 or MAPK8 inhibitors, but not a MAPK14 inhibitor. Decreasing the MMP14 protein and activity in vitro by small interfering (si)RNA to MMP14 diminished the acrolein-induced MUC5AC transcripts. In acrolein-exposed mice or transgenic mice with lung-specific transforming growth factor-alpha (an EGFR ligand) expression, lung MMP14 and MUC5AC levels increased and these effects were inhibited by a EGFR inhibitor, erlotinib. CONCLUSIONS: Taken together, these findings implicate acrolein-induced MMP14 expression and activity in mucin production in COPD.


Subject(s)
Matrix Metalloproteinase 14/metabolism , Mucins/biosynthesis , Respiratory Mucosa/metabolism , Acrolein/metabolism , Animals , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Erlotinib Hydrochloride , Gene Expression Regulation, Enzymologic , Humans , Lung/enzymology , Lung/metabolism , Mice , Mucins/metabolism , Protein Kinase Inhibitors/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Quinazolines/metabolism , Respiratory Mucosa/ultrastructure
13.
Am J Respir Cell Mol Biol ; 41(5): 562-72, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19244201

ABSTRACT

Transforming growth factor (TGF)-alpha is a ligand for the epidermal growth factor receptor (EGFR). EGFR activation is associated with fibroproliferative processes in human lung disease and animal models of pulmonary fibrosis. Overexpression of TGF-alpha in transgenic mice causes progressive and severe pulmonary fibrosis; however, the intracellular signaling pathways downstream of EGFR mediating this response are unknown. Using a doxycycline-regulatable transgenic mouse model of lung-specific TGF-alpha expression, we observed increased PCNA protein and phosphorylation of Akt and p70S6K in whole lung homogenates in association with induction of TGF-alpha. Induction in the lung of TGF-alpha caused progressive pulmonary fibrosis over a 7-week period. Daily administration of rapamycin prevented accumulation of total lung collagen, weight loss, and changes in pulmonary mechanics. Treatment of mice with rapamycin 4 weeks after the induction of TGF-alpha prevented additional weight loss, increases in total collagen, and changes in pulmonary mechanics. Rapamycin prevented further increases in established pulmonary fibrosis induced by EGFR activation. This study demonstrates that mammalian target of rapamycin (mTOR) is a major effector of EGFR-induced pulmonary fibrosis, providing support for further studies to determine the role of mTOR in the pathogenesis and treatment of pulmonary fibrosis.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Lung/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Signal Transduction/drug effects , Sirolimus/pharmacology , Transforming Growth Factor alpha/metabolism , Animals , Carrier Proteins/metabolism , Collagen/metabolism , Disease Models, Animal , Disease Progression , Doxycycline/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gene Expression Regulation , Humans , Lung/enzymology , Lung/physiopathology , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/physiopathology , Quinazolines/pharmacology , Respiratory Mechanics/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transforming Growth Factor alpha/genetics , Uteroglobin/genetics
14.
Am J Respir Cell Mol Biol ; 41(4): 415-25, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19188657

ABSTRACT

Transforming growth factor (TGF)-alpha and its receptor, the epidermal growth factor receptor, are induced after lung injury and are associated with remodeling in chronic pulmonary diseases, such as pulmonary fibrosis and asthma. Expression of TGF-alpha in the lungs of adult mice causes fibrosis, pleural thickening, and pulmonary hypertension, in addition to increased expression of a transcription factor, early growth response-1 (Egr-1). Egr-1 was increased in airway smooth muscle (ASM) and the vascular adventitia in the lungs of mice conditionally expressing TGF-alpha in airway epithelium (Clara cell secretory protein-rtTA(+/-)/[tetO](7)-TGF-alpha(+/-)). The goal of this study was to determine the role of Egr-1 in TGF-alpha-induced lung disease. To accomplish this, TGF-alpha-transgenic mice were crossed to Egr-1 knockout (Egr-1(ko/ko)) mice. The lack of Egr-1 markedly increased the severity of TGF-alpha-induced pulmonary disease, dramatically enhancing airway muscularization, increasing pulmonary fibrosis, and causing greater airway hyperresponsiveness to methacholine. Smooth muscle hyperplasia, not hypertrophy, caused the ASM thickening in the absence of Egr-1. No detectable increases in pulmonary inflammation were found. In addition to the airway remodeling disease, vascular remodeling and pulmonary hypertension were also more severe in Egr-1(ko/ko) mice. Thus, Egr-1 acts to suppress epidermal growth factor receptor-mediated airway and vascular muscularization, fibrosis, and airway hyperresponsiveness in the absence of inflammation. This provides a unique model to study the processes causing pulmonary fibrosis and ASM thickening without the complicating effects of inflammation.


Subject(s)
Bronchial Hyperreactivity/physiopathology , Early Growth Response Protein 1/physiology , Lung/pathology , Pulmonary Fibrosis/pathology , Transforming Growth Factor alpha/physiology , Airway Resistance , Albuterol/pharmacology , Animals , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/genetics , Cells, Cultured/drug effects , Cells, Cultured/pathology , Disease Models, Animal , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , ErbB Receptors/antagonists & inhibitors , Fibroblasts/cytology , Humans , Hyperplasia , Lung Compliance , Methacholine Chloride/toxicity , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Smooth/pathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/cytology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/physiopathology , Recombinant Fusion Proteins/physiology , Transforming Growth Factor alpha/adverse effects , Weight Loss
15.
Am J Respir Cell Mol Biol ; 41(2): 226-36, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19131640

ABSTRACT

The etiology of acute lung injury is complex and associated with numerous, chemically diverse precipitating factors. During acute lung injury in mice, one key event is epithelial cell injury that leads to reduced surfactant biosynthesis. We have previously reported that transgenic mice that express transforming growth factor alpha (TGFA) in the lung were protected during nickel-induced lung injury. Here, we find that the mechanism by which TGFA imparts protection includes maintenance of surfactant-associated protein B (SFTPB) transcript levels and epidermal growth factor receptor-dependent signaling in distal pulmonary epithelial cells. This protection is complex and not accompanied by a diminution in inflammatory mediator transcripts or additional stimulation of antioxidant transcripts. In mouse lung epithelial (MLE-15) cells, microarray analysis demonstrated that nickel increased transcripts of genes enriched in MTF1, E2F-1, and AP-2 transcription factor-binding sites and decreased transcripts of genes enriched in AP-1-binding sites. Nickel also increased Jun transcript and DNA-binding activity, but decreased SFTPB transcript. Expression of SFTPB under the control of a doxycycline-sensitive promoter increased survival during nickel-induced injury as compared with control mice. Together, these findings support the idea that maintenance of SFTPB expression is critical to survival during acute lung injury.


Subject(s)
Acute Lung Injury/chemically induced , Nickel/toxicity , Pulmonary Surfactant-Associated Protein B/metabolism , Administration, Inhalation , Aerosols , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Pulmonary Surfactant-Associated Protein B/genetics , Respiratory Mucosa/cytology , Survival Rate , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L64-72, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19304906

ABSTRACT

Patients with mutations in the pulmonary surfactant protein C (SP-C) gene develop interstitial lung disease and pulmonary exacerbations associated with viral infections including respiratory syncytial virus (RSV). Pulmonary infection with RSV caused more severe interstitial thickening, air space consolidation, and goblet cell hyperplasia in SP-C-deficient (Sftpc(-/-)) mice compared with SP-C replete mice. The RSV-induced pathology resolved more slowly in Sftpc(-/-) mice with lung inflammation persistent up to 30 days postinfection. Polymorphonuclear leukocyte and macrophage counts were increased in the bronchoalveolar lavage (BAL) fluid of Sftpc(-/-) mice. Viral titers and viral F and G protein mRNA were significantly increased in both Sftpc(-/-) and heterozygous Sftpc(+/-) mice compared with controls. Expression of Toll-like receptor 3 (TLR3) mRNA was increased in the lungs of Sftpc(-/-) mice relative to Sftpc(+/+) mice before and after RSV infection. Consistent with the increased TLR3 expression, BAL inflammatory cells were increased in the Sftpc(-/-) mice after exposure to a TLR3-specific ligand, poly(I:C). Preparations of purified SP-C and synthetic phospholipids blocked poly(I:C)-induced TLR3 signaling in vitro. SP-C deficiency increases the severity of RSV-induced pulmonary inflammation through regulation of TLR3 signaling.


Subject(s)
Pulmonary Surfactant-Associated Protein C/deficiency , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/pathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/virology , Cell Count , Cell Line , Collectins/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation, Viral , Goblet Cells/pathology , Goblet Cells/virology , Humans , Hypertrophy , Ligands , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Pneumonia/complications , Pneumonia/pathology , Pneumonia/virology , Pulmonary Surfactant-Associated Protein C/metabolism , RNA, Double-Stranded/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Time Factors , Toll-Like Receptor 3/metabolism
17.
Am J Respir Cell Mol Biol ; 38(4): 446-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18006877

ABSTRACT

Chronic obstructive pulmonary disease (COPD), a global public health problem, is characterized by progressive difficulty in breathing, with increased mucin production, especially in the small airways. Acrolein, a constituent of cigarette smoke and an endogenous mediator of oxidative stress, increases airway mucin 5, subtypes A and C (MUC5AC) production; however, the mechanism remains unclear. In this study, increased mMUC5AC transcripts and protein were associated with increased lung matrix metalloproteinase 9 (mMMP9) transcripts, protein, and activity in acrolein-exposed mice. Increased mMUC5AC transcripts and mucin protein were diminished in gene-targeted Mmp9 mice [Mmp9((-/-))] or in mice treated with an epidermal growth factor receptor (EGFR) inhibitor, erlotinib. Acrolein also decreased mTissue inhibitor of metalloproteinase protein 3 (an MMP9 inhibitor) transcript levels. In a cell-free system, acrolein increased pro-hMMP9 cleavage and activity in concentrations (100-300 nM) found in sputum from subjects with COPD. Acrolein increased hMMP9 transcripts in human airway cells, which was inhibited by an MMP inhibitor, EGFR-neutralizing antibody, or a mitogen-activated protein kinase (MAPK) 3/2 inhibitor. Together these findings indicate that acrolein can initiate cleavage of pro-hMMP9 and EGFR/MAPK signaling that leads to additional MMP9 formation. Augmentation of hMMP9 activity, in turn, could contribute to persistent excessive mucin production.


Subject(s)
Acrolein/pharmacology , Matrix Metalloproteinase 9/metabolism , Mucins/biosynthesis , Animals , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Lung/drug effects , Lung/enzymology , Lung/pathology , MAP Kinase Signaling System/drug effects , Male , Matrix Metalloproteinase 9/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mucin 5AC , Mucins/genetics , Mucins/metabolism , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sputum/drug effects , Sputum/enzymology , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
18.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135315

ABSTRACT

Wilms' tumor 1 (WT1) is a critical transcriptional regulator of mesothelial cells during lung development but is downregulated in postnatal stages and adult lungs. We recently showed that WT1 is upregulated in both mesothelial cells and mesenchymal cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a fatal fibrotic lung disease. Although WT1-positive cell accumulation leading to severe fibrotic lung disease has been studied, the role of WT1 in fibroblast activation and pulmonary fibrosis remains elusive. Here, we show that WT1 functions as a positive regulator of fibroblast activation, including fibroproliferation, myofibroblast transformation, and extracellular matrix (ECM) production. Chromatin immunoprecipitation experiments indicate that WT1 binds directly to the promoter DNA sequence of α-smooth muscle actin (αSMA) to induce myofibroblast transformation. In support, the genetic lineage tracing identifies WT1 as a key driver of mesothelial-to-myofibroblast and fibroblast-to-myofibroblast transformation. Importantly, the partial loss of WT1 was sufficient to attenuate myofibroblast accumulation and pulmonary fibrosis in vivo. Further, our coculture studies show that WT1 upregulation leads to non-cell autonomous effects on neighboring cells. Thus, our data uncovered a pathogenic role of WT1 in IPF by promoting fibroblast activation in the peripheral areas of the lung and as a target for therapeutic intervention.


Subject(s)
Actins/genetics , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/pathology , Repressor Proteins/metabolism , WT1 Proteins/metabolism , Adult , Animals , Bleomycin/toxicity , Cell Differentiation/genetics , Cells, Cultured , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Lung/cytology , Lung/drug effects , Lung/pathology , Male , Mice, Transgenic , Primary Cell Culture , Promoter Regions, Genetic/genetics
19.
J Clin Invest ; 125(5): 2021-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25866971

ABSTRACT

Epithelial cells that line the conducting airways provide the initial barrier and innate immune responses to the abundant particles, microbes, and allergens that are inhaled throughout life. The transcription factors SPDEF and FOXA3 are both selectively expressed in epithelial cells lining the conducting airways, where they regulate goblet cell differentiation and mucus production. Moreover, these transcription factors are upregulated in chronic lung disorders, including asthma. Here, we show that expression of SPDEF or FOXA3 in airway epithelial cells in neonatal mice caused goblet cell differentiation, spontaneous eosinophilic inflammation, and airway hyperresponsiveness to methacholine. SPDEF expression promoted DC recruitment and activation in association with induction of Il33, Csf2, thymic stromal lymphopoietin (Tslp), and Ccl20 transcripts. Increased Il4, Il13, Ccl17, and Il25 expression was accompanied by recruitment of Th2 lymphocytes, group 2 innate lymphoid cells, and eosinophils to the lung. SPDEF was required for goblet cell differentiation and pulmonary Th2 inflammation in response to house dust mite (HDM) extract, as both were decreased in neonatal and adult Spdef(-/-) mice compared with control animals. Together, our results indicate that SPDEF causes goblet cell differentiation and Th2 inflammation during postnatal development and is required for goblet cell metaplasia and normal Th2 inflammatory responses to HDM aeroallergen.


Subject(s)
Antigens, Dermatophagoides/toxicity , Epithelial Cells/metabolism , Goblet Cells/physiology , Lung/immunology , Proto-Oncogene Proteins c-ets/physiology , Pulmonary Eosinophilia/immunology , Th2 Cells/immunology , Age Factors , Animals , Animals, Newborn , Cell Differentiation , Chemokine CCL20/biosynthesis , Chemokine CCL20/genetics , Chemotaxis, Leukocyte , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/immunology , Eosinophils/physiology , Hepatocyte Nuclear Factor 3-gamma/physiology , Interleukins/biosynthesis , Interleukins/genetics , Metaplasia , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Proto-Oncogene Proteins c-ets/genetics , Pulmonary Eosinophilia/etiology , Pulmonary Eosinophilia/metabolism , Pulmonary Eosinophilia/pathology , Recombinant Fusion Proteins/metabolism , Transgenes , Thymic Stromal Lymphopoietin
20.
Chest ; 121(3 Suppl): 70S-75S, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11893692

ABSTRACT

Initiated by numerous factors, acute lung injury is marked by epithelial and endothelial cell perturbation and inflammatory cell influx that leads to surfactant disruption, pulmonary edema, and atelectasis. This syndrome has been associated with a myriad of mediators including cytokines, oxidants, and growth factors. To better understand gene-environmental interactions controlling this complex process, the sensitivity of inbred mouse strains was investigated following acute lung injury that was induced by fine nickel sulfate aerosol. Measuring survival time, protein and neutrophil concentrations in BAL fluid, lung wet-to-dry weight ratio, and histology, we found that these responses varied between inbred mouse strains and that susceptibility is heritable. To assess the progression of acute lung injury, the temporal expression of genes and expressed sequence tags was assessed by complementary DNA microarray analysis. Enhanced expression was noted in genes that were associated with oxidative stress, antiprotease function, and extracellular matrix repair. In contrast, expression levels of surfactant proteins (SPs) and Clara cell secretory protein (ie, transcripts that are constitutively expressed in the lung) decreased markedly. Genome-wide analysis was performed with offspring derived from a sensitive and resistant strain (C57BL/6xA F(1) backcrossed with susceptible A strain). Significant linkage was identified for a locus on chromosome 6 (proposed as Aliq4), a region that we had identified previously following ozone-induced acute lung injury. Two suggestive linkages were identified on chromosomes 1 and 12. Using haplotype analysis to estimate the combined effect of these regions (along with putative modifying loci on chromosomes 9 and 16), we found that five loci interact to account for the differences in survival time of the parental strains. Candidate genes contained in Aliq4 include SP-B, aquaporin 1, and transforming growth factor-alpha. Thus, the functional genomic approaches of large gene set expression (complementary DNA microarray) and genome-wide analyses continue to provide novel insights into the genetic susceptibility of lung injury.


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease , Respiratory Distress Syndrome/genetics , Aerosols , Animals , Aquaporin 1 , Aquaporins/genetics , Environmental Exposure , Gene Expression , Genetic Linkage , Lung/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Nickel , Oligonucleotide Array Sequence Analysis , Ozone , Polytetrafluoroethylene , Proteolipids/genetics , Pulmonary Surfactants/genetics , Quantitative Trait, Heritable , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Risk Factors , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL