Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1205, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864068

ABSTRACT

Pulmonary alveolar microlithiasis is an autosomal recessive lung disease caused by a deficiency in the pulmonary epithelial Npt2b sodium-phosphate co-transporter that results in accumulation of phosphate and formation of hydroxyapatite microliths in the alveolar space. The single cell transcriptomic analysis of a pulmonary alveolar microlithiasis lung explant showing a robust osteoclast gene signature in alveolar monocytes and the finding that calcium phosphate microliths contain a rich protein and lipid matrix that includes bone resorbing osteoclast enzymes and other proteins suggested a role for osteoclast-like cells in the host response to microliths. While investigating the mechanisms of microlith clearance, we found that Npt2b modulates pulmonary phosphate homeostasis through effects on alternative phosphate transporter activity and alveolar osteoprotegerin, and that microliths induce osteoclast formation and activation in a receptor activator of nuclear factor-κB ligand and dietary phosphate dependent manner. This work reveals that Npt2b and pulmonary osteoclast-like cells play key roles in pulmonary homeostasis and suggest potential new therapeutic targets for the treatment of lung disease.


Subject(s)
Lung Diseases , Osteogenesis , Humans , Homeostasis , Lung
2.
bioRxiv ; 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36824953

ABSTRACT

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored the mechanisms of silica-induced pulmonary fibrosis in a mouse model using multiple modalities including whole-lung single-nucleus RNA sequencing. These analyses revealed that in addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor-κB ligand (RANKL) in pulmonary lymphocytes and alveolar type II cells. Furthermore, anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated silica-induced pulmonary fibrosis. We conclude that silica induces osteoclast-like differentiation of distinct recruited and tissue resident monocyte populations, leading to progressive lung injury, likely due to sustained elaboration of bone resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.

3.
Front Microbiol ; 12: 774711, 2021.
Article in English | MEDLINE | ID: mdl-34956139

ABSTRACT

Mannose-binding lectins effectively inhibit most seasonal strains of influenza A virus and contribute to the innate host defense vs. these viruses. In contrast, pandemic IAV strains are largely resistant to these lectins, likely contributing to increased spread and worse outcomes. In this paper, we evaluated the inhibition of IAV by mannose-binding lectins of human, bacterial, and fungal origin to understand and possibly increase activity vs. the pandemic IAV. A modified version of the human surfactant protein D (SP-D) neck and carbohydrate recognition domain (NCRD) with combinatorial substitutions at the 325 and 343 positions, previously shown to inhibit pandemic H3N2 IAV in vitro and in vivo, and to inhibit pandemic H1N1 in vitro, failed to protect mice from pandemic H1N1 in vivo in the current study. We attempted a variety of maneuvers to improve the activity of the mutant NCRDs vs. the 2009 pandemic H1N1, including the formation of full-length SP-D molecules containing the mutant NCRD, cross-linking of NCRDs through the use of antibodies, combining SP-D or NCRDs with alpha-2-macroglobulin, and introducing an additional mutation to the double mutant NCRD. None of these substantially increased the antiviral activity for the pandemic H1N1. We also tested the activity of bacterial and algal mannose-binding lectins, cyanovirin, and griffithsin, against IAV. These had strong activity against seasonal IAV, which was largely retained against pandemic H1N1. We propose mechanisms to account for differences in activity of SP-D constructs against pandemic H3N2 and H1N1, and for differences in activity of cyanovirin vs. SP-D constructs.

4.
Proc Natl Acad Sci U S A ; 114(32): E6613-E6622, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739896

ABSTRACT

Development of pneumonia is the most lethal consequence of influenza, increasing mortality more than 50-fold compared with uncomplicated infection. The spread of viral infection from conducting airways to the alveolar epithelium is therefore a pivotal event in influenza pathogenesis. We found that mitogenic stimulation with keratinocyte growth factor (KGF) markedly accelerated mortality after infectious challenge with influenza A virus (IAV). Coadministration of KGF with IAV markedly accelerated the spread of viral infection from the airways to alveoli compared with challenge with IAV alone, based on spatial and temporal analyses of viral nucleoprotein staining of lung tissue sections and dissociated lung cells. To better define the temporal relationship between KGF administration and susceptibility to IAV infection in vivo, we administered KGF 120, 48, 24, and 0 h before intrapulmonary IAV challenge and assessed the percentages of proliferating and IAV-infected, alveolar type II (AECII) cells in dispersed lung cell populations. Peak AECII infectivity coincided with the timing of KGF administration that also induced peak AECII proliferation. AECII from mice that were given intrapulmonary KGF before isolation and then infected with IAV ex vivo exhibited the same temporal pattern of proliferation and infectious susceptibility. KGF-induced increases in mortality, AECII proliferation, and enhanced IAV susceptibility were all reversed by pretreatment of the animals with the mTOR inhibitor rapamycin before mitogenic stimulation. Taken together, these data suggest mTOR signaling-dependent, mitogenic conditioning of AECII is a determinant of host susceptibility to infection with IAV.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cell Proliferation/drug effects , Fibroblast Growth Factor 7/pharmacology , Influenza A virus/metabolism , Mitogens/pharmacology , Orthomyxoviridae Infections/metabolism , Alveolar Epithelial Cells/pathology , Animals , Disease Susceptibility/chemically induced , Female , Mice , Mice, Inbred DBA , Orthomyxoviridae Infections/pathology
5.
Biochemistry ; 56(31): 4095-4105, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28719181

ABSTRACT

Lung surfactant proteins (SPs) play critical roles in surfactant function and innate immunity. SP-A and SP-D, members of the collectin family of C-type lectins, exhibit distinct ligand specificities, effects on surfactant structure, and host defense functions despite extensive structural homology. SP-A binds to dipalmitoylphosphatidylcholine (DPPC), the major surfactant lipid component, but not phosphatidylinositol (PI), whereas SP-D shows the opposite preference. Additionally, SP-A and SP-D recognize widely divergent pathogen-associated molecular patterns. Previous studies suggested that a ligand-induced surface loop conformational change unique to SP-A contributes to lipid binding affinity. To test this hypothesis and define the structural features of SP-A and SP-D that determine their ligand binding specificities, a structure-guided approach was used to introduce key features of SP-D into SP-A. A quadruple mutant (E171D/P175E/R197N/K203D) that introduced an SP-D-like loop-stabilizing calcium binding site into the carbohydrate recognition domain was found to interconvert SP-A ligand binding preferences to an SP-D phenotype, exchanging DPPC for PI specificity, and resulting in the loss of lipid A binding and the acquisition of more avid mannan binding properties. Mutants with constituent single or triple mutations showed alterations in their lipid and sugar binding properties that were intermediate between those of SP-A and SP-D. Structures of mutant complexes with inositol or methyl-mannose revealed an attenuation of the ligand-induced conformational change relative to wild-type SP-A. These studies suggest that flexibility in a key surface loop supports the distinctive lipid binding functions of SP-A, thus contributing to its multiple functions in surfactant structure and regulation, and host defense.


Subject(s)
Models, Molecular , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , 1,2-Dipalmitoylphosphatidylcholine/chemistry , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Amino Acid Substitution , Animals , Binding Sites , Crystallography, X-Ray , Kinetics , Lectins, C-Type/chemistry , Lectins, C-Type/metabolism , Ligands , Lipid A/chemistry , Lipid A/metabolism , Liposomes , Mutagenesis, Site-Directed , Mutation , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Protein Stability , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L868-79, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26919897

ABSTRACT

Keratinocyte growth factor (KGF) is an epithelial mitogen that has been reported to protect the lungs from a variety of toxic and infectious insults. In prior studies we found that recombinant human KGF accelerates clearance of bacteria from the murine lung by augmenting the function of alveolar macrophages (AM). In this study we tested the hypothesis that endogenous KGF plays a role in the maintenance of innate pulmonary defense against gram-negative bacterial infections. KGF-deficient mice exhibited delayed clearance of Escherichia coli from the lungs, attenuated phagocytosis by AM, and decreased antimicrobial activity in bronchoalveolar lavage (BAL) fluid, due in part to reductions in levels of surfactant protein A, surfactant protein D, and lysozyme. These immune deficits were accompanied by lower alveolar type II epithelial cell counts and reduced alveolar type II epithelial cell expression of collectin and lysozyme genes on a per cell basis. No significant between-group differences were detected in selected inflammatory cytokines or BAL inflammatory cell populations at baseline or after bacterial challenge in the wild-type and KGF-deficient mice. A single intranasal dose of recombinant human KGF reversed defects in bacterial clearance, AM function, and BAL fluid antimicrobial activity. We conclude that KGF supports alveolar innate immune defense through maintenance of alveolar antimicrobial protein levels and functions of AM. Together these data demonstrate a role for endogenous KGF in maintenance of normal pulmonary innate immune function.


Subject(s)
Escherichia coli Infections/immunology , Fibroblast Growth Factor 7/physiology , Immunity, Innate , Macrophages, Alveolar/immunology , Pneumonia, Bacterial/immunology , Animals , Cells, Cultured , Collectins/genetics , Collectins/metabolism , Escherichia coli Infections/metabolism , Female , Gene Expression , Humans , Macrophages, Alveolar/microbiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muramidase/genetics , Muramidase/metabolism , Pneumonia, Bacterial/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology
7.
Sci Transl Med ; 7(313): 313ra181, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560359

ABSTRACT

Pulmonary alveolar microlithiasis (PAM) is a rare, autosomal recessive lung disorder associated with progressive accumulation of calcium phosphate microliths. Inactivating mutations in SLC34A2, which encodes the NPT2b sodium-dependent phosphate cotransporter, has been proposed as a cause of PAM. We show that epithelial deletion of Npt2b in mice results in a progressive pulmonary process characterized by diffuse alveolar microlith accumulation, radiographic opacification, restrictive physiology, inflammation, fibrosis, and an unexpected alveolar phospholipidosis. Cytokine and surfactant protein elevations in the alveolar lavage and serum of PAM mice and confirmed in serum from PAM patients identify serum MCP-1 (monocyte chemotactic protein 1) and SP-D (surfactant protein D) as potential biomarkers. Microliths introduced by adoptive transfer into the lungs of wild-type mice produce marked macrophage-rich inflammation and elevation of serum MCP-1 that peaks at 1 week and resolves at 1 month, concomitant with clearance of stones. Microliths isolated by bronchoalveolar lavage readily dissolve in EDTA, and therapeutic whole-lung EDTA lavage reduces the burden of stones in the lungs. A low-phosphate diet prevents microlith formation in young animals and reduces lung injury on the basis of reduction in serum SP-D. The burden of pulmonary calcium deposits in established PAM is also diminished within 4 weeks by a low-phosphate diet challenge. These data support a causative role for Npt2b in the pathogenesis of PAM and the use of the PAM mouse model as a preclinical platform for the development of biomarkers and therapeutic strategies.


Subject(s)
Biomarkers/blood , Calcinosis/etiology , Calcinosis/physiopathology , Calcinosis/therapy , Genetic Diseases, Inborn/etiology , Genetic Diseases, Inborn/physiopathology , Genetic Diseases, Inborn/therapy , Lung Diseases/etiology , Lung Diseases/physiopathology , Lung Diseases/therapy , Sodium-Phosphate Cotransporter Proteins, Type IIb/deficiency , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Animals , Diet , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Lung/metabolism , Lung/pathology , Mice , Mutation , Phosphates/metabolism , Pulmonary Alveoli/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 306(11): L1036-44, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24705721

ABSTRACT

We recently reported that a trimeric neck and carbohydrate recognition domain (NCRD) fragment of human surfactant protein D (SP-D), a host defense lectin, with combinatorial substitutions at the 325 and 343 positions (D325A+R343V) exhibits markedly increased antiviral activity for seasonal strains of influenza A virus (IAV). The NCRD binds to glycan-rich viral envelope proteins including hemagglutinin (HA). We now show that replacement of D325 with serine to create D325S+R343V provided equal or increased neutralizing activity compared with D325A+R343V. The activity of the double mutants was significantly greater than that of either single mutant (D325A/S or R343V). D325A+R343V and D325S+R343V also strongly inhibited HA activity, and markedly aggregated, the 1968 pandemic H3N2 strain, Aichi68. D325S+R343V significantly reduced viral loads and mortality of mice infected with Aichi68, whereas wild-type SP-D NCRD did not. The pandemic H1N1 strains of 1918 and 2009 have only one N-linked glycan side on the head region of the HA and are fully resistant to inhibition by native SP-D. Importantly, we now show that D325A+R343V and D325S+R343V inhibited Cal09 H1N1 and related strains, and reduced uptake of Cal09 by epithelial cells. Inhibition of Cal09 was mediated by the lectin activity of the NCRDs. All known human pandemic strains have at least one glycan attachment on the top or side of the HA head, and our results indicate that they may be susceptible to inhibition by modified host defense lectins.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/virology , Pulmonary Surfactant-Associated Protein D/genetics , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Disease Resistance , Dogs , Female , Host-Pathogen Interactions , Humans , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred DBA , Mutation, Missense , Pandemics , Protein Binding , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Viral Load
9.
J Immunol ; 192(5): 2405-17, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24470495

ABSTRACT

Traumatic injury is generally considered to have a suppressive effect on the immune system, resulting in increased susceptibility to infection. Paradoxically, we found that thermal injury to the skin induced a robust time-dependent protection of mice from a lethal Klebsiella pneumoniae pulmonary challenge. The protective response was neutrophil dependent and temporally associated with a systemic increase in neutrophils resulting from a reprioritization of hematopoiesis toward myeloid lineages. A prominent and specific activation of STAT3 in the bone marrow preceded the myeloid shift in that compartment, in association with durable increases in STAT3 activating serum cytokines G-CSF and IL-6. Neutralization of the postburn increase in serum G-CSF largely blocked STAT3 activation in marrow cells, reversing the hematopoietic changes and systemic neutrophilia. Daily administration of rG-CSF was sufficient to recapitulate the changes induced by injury including hematopoietic reprioritization and protection from pulmonary challenge with K. pneumoniae. Analysis of posttraumatic gene expression patterns in humans reveals that they are also consistent with a role for G-CSF as a switch that activates innate immune responses and suppresses adaptive immune responses. Our findings suggest that the G-CSF STAT3 axis constitutes a key protective mechanism induced by injury to reduce the risk for posttraumatic infection.


Subject(s)
Burns/immunology , Granulocyte Colony-Stimulating Factor/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Pneumonia, Bacterial/immunology , Adaptive Immunity , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Burns/blood , Burns/complications , Burns/pathology , Granulocyte Colony-Stimulating Factor/blood , Immunity, Innate , Interleukin-6/blood , Interleukin-6/immunology , Klebsiella Infections/blood , Klebsiella Infections/etiology , Klebsiella Infections/pathology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia, Bacterial/blood , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism
10.
Innate Immun ; 17(6): 499-507, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21088048

ABSTRACT

Previous studies have shown that the Ron receptor tyrosine kinase is an important regulator of the acute lung inflammatory response induced by intranasal administration of bacterial LPS. Compared to wild-type mice, complete loss of the Ron receptor in all cell types in vivo was associated with increased lung damage as determined by histological analyses and several markers of lung injury including increases in pro-inflammatory cytokines such as TNF-α. Tumor-necrosis factor-α is a multifunctional cytokine secreted by macrophages, which plays a major role in inflammation and is a central mediator of several disease states including rheumatoid arthritis and sepsis. Based on increased TNF-α production observed in the Ron-deficient mice, we hypothesized that Ron receptor function in the inflammatory cell compartment is essential for the regulating lung injury in vivo. To test this hypothesis, we generated myeloid lineage-specific Ron-deficient mice. In this study, we report that loss of Ron signaling selectively in myeloid cells results in increased lung injury following intranasal administration of LPS as measured by increases in TNF-α production, ensuing neutrophil accumulation and increased lung histopathology. These findings corroborate the role of Ron receptor tyrosine kinase as a negative regulator of inflammation and further demonstrate the in vivo significance of Ron signaling selectively in myeloid cells as a major regulator of this response in vivo. These data authenticate Ron as a potential target in innate immunity and TNF-α-mediated pathologies.


Subject(s)
Acute Lung Injury/pathology , Myeloid Cells/pathology , Pulmonary Alveoli/pathology , Receptor Protein-Tyrosine Kinases/deficiency , Tumor Necrosis Factor-alpha/biosynthesis , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Gene Expression , Immunity, Innate/drug effects , Immunity, Innate/physiology , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
11.
Shock ; 33(2): 197-204, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19487969

ABSTRACT

The Ron receptor tyrosine kinase (TK) plays a regulatory role in the inflammatory response to acute lung injury induced by intranasal administration of bacterial LPS. Previously, we have shown that mice with a targeted deletion of the TK signaling domain of the Ron receptor exhibited more severe lung injury in response to intranasal LPS administration as evidenced by an increased leakage of albumin in the lungs and a greater thickening of the alveolar septa compared with wild-type mice. In addition, lung injury in the Ron TK-deficient (TK(-/-)) mice was associated with increased activation of the transcription factor, nuclear factor-kappaB (NF-kappaB), and significantly increased intrapulmonary expression of TNFalpha. TNFalpha, a multifunctional proinflammatory cytokine, is a central mediator in several disease states, including rheumatoid arthritis and sepsis. On the basis of the observation that TNFalpha production is increased in the Ron TK-/- mice and that macrophages are a major source of this cytokine, we hypothesized that the alterations observed in the Ron TK(-/-) mice may be due, in part, to Ron signaling, specifically in alveolar macrophages. To test this hypothesis, we used the wild-type and Ron TK(-/-) primary alveolar macrophages and the murine alveolar macrophage cell line, MH-S, to examine the effects of Ron activation on LPS-induced TNFalpha production and NF-kappaB activity. Here, we reported that Ron is expressed on alveolar macrophages and MH-S cells. Activation of Ron by its ligand, hepatocyte growth factor-like protein, decreases TNFalpha production in alveolar macrophages after LPS challenge. Decreased TNFalpha is associated with hepatocyte growth factor-like protein-induced decreases in NF-kappaB activation and increases in the NF-kappaB inhibitory protein, IkappaB. We also provided the first evidence for Ron as a negative regulator of Adam17, the metalloprotease involved in TNFalpha processing. These results indicate that Ron plays a critical role in regulation of alveolar macrophage signaling and validates this receptor as a target in TNFalpha-mediated pulmonary pathologies.


Subject(s)
ADAM Proteins/metabolism , Macrophages, Alveolar/metabolism , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein , Animals , Cell Line , Cells, Cultured , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Mutant Strains , Mutation , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics
12.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L305-13, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17545488

ABSTRACT

Resistin-like molecule (RELM)-beta is a cysteine-rich cytokine implicated in insulin resistance and asthmatic responses, but its function remains an enigma. We now report that RELM-beta has a role in promoting airway inflammation and lung remodeling in the mouse lung. RELM-beta is strongly induced by diverse allergens and T helper type 2 (Th2) cytokines by an IL-13- and STAT6-dependent mechanism. To understand the in vivo role of RELM-beta, we delivered recombinant murine RELM-beta intratracheally to naïve mice. RELM-beta induced dose-dependent leukocyte accumulation (most prominently involving macrophages) and goblet cell hyperplasia. The most prominent effect induced by RELM-beta was increased perivascular and peribronchial collagen deposition. Mice genetically deficient in RELM-beta had reduced accumulation of collagen and goblet cell hyperplasia in an experimental model of allergic airway inflammation. In vitro experiments demonstrated that RELM-beta had fibroblast motogenic activity. These results identify RELM-beta as a Th2-associated cytokine with potent inflammatory and remodeling activity.


Subject(s)
Allergens/immunology , Hormones, Ectopic/immunology , Lung/immunology , Pneumonia/immunology , Allergens/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Collagen/metabolism , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Goblet Cells/immunology , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Intercellular Signaling Peptides and Proteins , Interleukin-13/metabolism , Interleukin-4/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , NIH 3T3 Cells , Pneumonia/metabolism , Pneumonia/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , STAT6 Transcription Factor/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
13.
Exp Lung Res ; 32(10): 483-97, 2006.
Article in English | MEDLINE | ID: mdl-17169855

ABSTRACT

Asthma is a complex pulmonary disorder characterized by reversible airflow obstruction, airway hyperresponsiveness, mucus cell metaplasia, and inflammation. Employing animal models of pulmonary inflammation induced by different allergens and Th2 cytokines, the authors have previously described the up-regulation of trefoil factor 2 (TFF2) in the lung. Given the known biological role of trefoil factors in epithelial restitution, it has been postulated that allergen-induced TFF2 might have an important role in asthmatic responses. Here the authors show that TFF2 is induced early and maintained for 2 weeks following allergen challenge in the mouse lung. In situ mRNA hybridization demonstrated expression of TFF2 primarily in a subset of bronchial epithelial cells and TFF2 immunohistochemistry identified expression in alcian blue-positive bronchial epithelial cells. TFF2 gene-deleted mice inoculated with allergen displayed a 10-fold increase in total cellularity compared with saline controls. Although this response was modestly attenuated compared to wild type controls, the loss of TFF2 did not affect gross levels of tissue inflammation. Furthermore, the loss of TFF2 did not affect induction or resolution of mucus cell metaplasia as measured by periodic acid-Schiff (PAS) or alcian blue staining. Thus, TFF2 is an allergen-induced gene, which is expressed in mucus-positive airways, but is not a major contributor to allergen-induced goblet cell metaplasia, mucus production, or inflammatory responses in the lung.


Subject(s)
Allergens/pharmacology , Asthma/immunology , Bronchi/drug effects , Mucins/biosynthesis , Mucus/metabolism , Muscle Proteins/biosynthesis , Pneumonia/immunology , Animals , Asthma/genetics , Asthma/metabolism , Bronchi/immunology , Bronchi/metabolism , Disease Models, Animal , Gene Expression/drug effects , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucins/genetics , Mucus/immunology , Muscle Proteins/genetics , Peptides/genetics , Pneumonia/genetics , Pneumonia/metabolism , RNA, Messenger/metabolism , Trefoil Factor-2
14.
Am J Respir Cell Mol Biol ; 35(3): 337-46, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16645178

ABSTRACT

IL-13 overexpression in the lung induces inflammatory and remodeling responses that are prominent features of asthma. Whereas most studies have concentrated on the development of IL-13-induced disease, far fewer studies have focused on the reversibility of IL-13-induced pathologies. This is particularly important because current asthma therapy appears to be poor at reversing lung remodeling. In this manuscript, we used an externally regulatable transgenic system that targets expression of IL-13 to the lung with the aim of characterizing the reversibility process. After 4 wk of doxycycline (dox) exposure, IL-13 expression resulted in mixed inflammatory cell infiltration, mucus cell metaplasia, lung fibrosis, and airspace enlargement (emphysema). After withdrawal of dox, IL-13 protein levels were profoundly reduced by 7 d and below baseline by 14 d. During this time frame, the level of lung eosinophils returned to near normal, whereas macrophages, lymphocytes, and neutrophils remained markedly elevated. IL-13-induced mucus cell metaplasia significantly decreased (91%) 3 wk after withdrawal of dox, showing strong correlation with reduced eosinophil levels. In contrast, IL-13-induced lung fibrosis did not significantly decline 4 wk after dox withdrawal. Importantly, IL-13-induced emphysema persisted, but modestly declined 4 wk after dox. Examination of transcript expression profiles identified a subset of genes that remained increased weeks after transgene expression was no longer detected. Notably, numerous IL-13-induced cytokines and enzymes were reversible (IL-6 and cathepsins), whereas others were sustained (CCL6 and chitinases) after IL-13 withdrawal, respectively. Thus, several hallmark features of IL-13-induced lung pathology persist and are dissociated from eosinophilia after IL-13 overexpression ceases.


Subject(s)
Interleukin-13/metabolism , Lung Diseases/genetics , Lung Diseases/pathology , Lung/pathology , Transcription, Genetic , Animals , Cathepsins/genetics , Cytokines/genetics , Doxycycline/toxicity , Eosinophilia/genetics , Eosinophilia/metabolism , Interleukin-13/genetics , Lung/drug effects , Lung/metabolism , Lung Diseases/chemically induced , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Receptors, Cytokine/genetics
15.
Am J Respir Cell Mol Biol ; 32(5): 428-35, 2005 May.
Article in English | MEDLINE | ID: mdl-15731505

ABSTRACT

Asthma is a complex inflammatory pulmonary disorder that is on the rise despite intense ongoing research. We aimed to elucidate novel pathways involved in the pathogenesis of asthma. Employing asthma models induced by different allergens (ovalbumin and Aspergillus fumigatus), we uncovered the involvement of two members of the small proline-rich protein (SPRR) family, SPRR2a and SPRR2b, known to be involved in epithelial differentiation but not allergic disease. In situ hybridization revealed induction of SPRR2 signal in a subset of bronchial epithelial cells and mononuclear cells associated with inflammation after allergen challenge. Allergen-induced SPRR2 mRNA accumulation in the lung occurred in a time-dependent manner, with peak expression 10-96 h after a second ovalbumin challenge. Transgenic overexpression of interleukin (IL)-13 in the lungs resulted in a marked increase of SPRR2 expression, and allergen-induced SPRR2 expression was significantly decreased in IL-13-deficient mice. Studies in gene-targeted mice revealed that allergen-induced SPRR2 was dependent upon STAT6. Finally, we aimed to determine if the induction of SPRR2 by allergen was tissue specific. Notably, SPRR2 was markedly increased in the small intestine after induction of allergic gastrointestinal inflammation. Thus, SPRR2 is an allergen- and IL-13-induced gene in experimental allergic responses that may be involved in disease pathophysiology.


Subject(s)
Asthma/immunology , Hypersensitivity, Immediate/metabolism , Inflammation/immunology , Intermediate Filament Proteins/immunology , Membrane Proteins/immunology , Protein Precursors/immunology , Allergens/metabolism , Animals , Cornified Envelope Proline-Rich Proteins , Gastrointestinal Tract/immunology , Humans , In Situ Hybridization , Interleukin-13/genetics , Interleukin-13/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , STAT6 Transcription Factor , Trans-Activators/genetics , Trans-Activators/metabolism
16.
J Biol Chem ; 280(14): 13952-61, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15647285

ABSTRACT

Pulmonary eosinophilia, a hallmark pathologic feature of allergic lung disease, is regulated by interleukin-13 (IL-13) as well as the eotaxin chemokines, but the specific role of these cytokines and their cooperative interaction are only partially understood. First, we elucidated the essential role of IL-13 in the induction of the eotaxins by comparing IL-13 gene-targeted mice with wild type control mice by using an ovalbumin-induced model of allergic airway inflammation. Notably, ovalbumin-induced expressions of eotaxin-1 and eotaxin-2 mRNA in the lungs were almost completely dependent upon IL-13. Second, in order to address the specific role of eotaxin-2 in IL-13-induced pulmonary eosinophilia, we generated eotaxin-2 gene-deficient mice by homologous recombination. Notably, in contrast to observations made in eotaxin-1-deficient mice, eotaxin-2-deficient mice had normal base-line eosinophil levels in the hematopoietic tissues and gastrointestinal tract. However, following intratracheal IL-13 administration, eotaxin-2-deficient mice showed a profound reduction in airway eosinophilia compared with wild type mice. Most interestingly, the level of peribronchial lung tissue eosinophils in IL-13-treated eotaxin-2-deficient mice was indistinguishable from wild type mice. Furthermore, IL-13 lung transgenic mice genetically engineered to be deficient in eotaxin-2 had a marked reduction of luminal eosinophils. Mechanistic analysis identified IL13-induced eotaxin-2 expression by macrophages in a distinct lung compartment (luminal inflammatory cells) compared with eotaxin-1, which was expressed solely in the tissue. Taken together, these results demonstrate a cooperative mechanism between IL-13 and eotaxin-2. In particular, IL-13 mediates allergen-induced eotaxin-2 expression, and eotaxin-2 mediates IL-13-induced airway eosinophilia.


Subject(s)
Chemokines, CC/immunology , Eosinophils/immunology , Inflammation/metabolism , Interleukin-13/immunology , Lung/immunology , Animals , Base Sequence , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL11 , Chemokine CCL24 , Chemokines, CC/genetics , Chemokines, CC/metabolism , Eosinophils/cytology , Gene Expression Regulation , Humans , In Situ Hybridization , Interleukin-13/genetics , Lung/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Sequence Alignment , Tissue Distribution
17.
Am J Respir Cell Mol Biol ; 31(3): 257-65, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15087305

ABSTRACT

Asthma, a complex chronic inflammatory pulmonary disorder, is on the rise despite intense ongoing research. To elucidate novel pathways involved in asthma pathogenesis, we used transcript expression profiling in a murine model of asthma. Employing asthma models induced by different allergens (ovalbumin and Aspergillus fumigatus) we uncovered the involvement of ADAM8, a member of a disintegrin and metalloproteinase (ADAM) family. In situ hybridization of mouse lungs revealed strong ADAM8 induction in peribronchial and perivascular inflammatory cells as well as in bronchiolar epithelial cells following allergen challenge. Sequence analysis of lung ADAM8 cDNA identified a novel splice variant of ADAM8 that contained an additional exon in juxtaposition to the transmembrane domain. Allergen-induced ADAM8 mRNA accumulation in the lung was dose- and time-dependent. Transgenic or pharmacologic delivery of interleukin (IL)-4 or IL-13 to the lungs resulted in a marked increase of ADAM8 expression. Gene-targeted mice studies revealed that ovalbumin-induced ADAM8 was largely dependent upon signal transducer and activator of transcription (STAT) 6 and the IL-4 receptor alpha-chain. Thus, ADAM8 is an allergen-, IL-4-, and IL-13-induced gene in the experimental asthmatic lung. Taken together with the role of ADAM33 in asthma, these results suggest that allergic lung responses involve the interplay of diverse members of the ADAM family.


Subject(s)
Antigens, CD/genetics , Asthma/enzymology , Asthma/genetics , Gene Expression Regulation, Enzymologic/genetics , Lung/enzymology , Membrane Proteins/genetics , Metalloendopeptidases/genetics , ADAM Proteins , Allergens , Alternative Splicing/genetics , Animals , Antigens, CD/biosynthesis , Asthma/physiopathology , Base Sequence/genetics , Bronchi/enzymology , Bronchi/pathology , Bronchi/physiopathology , Disease Models, Animal , Interleukin-13/genetics , Interleukin-13/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Lung/pathology , Lung/physiopathology , Membrane Proteins/biosynthesis , Metalloendopeptidases/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Protein Isoforms/genetics , Protein Structure, Tertiary/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Respiratory Mucosa/enzymology , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology , STAT6 Transcription Factor , Trans-Activators/genetics , Trans-Activators/metabolism
18.
J Immunol ; 172(3): 1815-24, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734765

ABSTRACT

The analysis of polygenic diseases such as asthma poses a challenging problem. In an effort to provide unbiased insight into disease pathogenesis, we took an empirical approach involving transcript expression profiling of lung tissue from mice with experimental asthma. Asthmatic responses were found to involve sequential induction of 4.7% of the tested genome; notably, there was ectopic expression of a series of genes not previously implicated in allergic or pulmonary responses. Genes were widely distributed throughout all chromosomes, but preferentially included genes involved in immunity, development, and homeostasis. When asthma was induced by two independent experimental regimens, unique gene transcript profiles were found depending upon the mode of disease induction. However, the majority of genes were common to both models representing an asthma signature genome. Analysis of STAT6-deficient mice revealed that an unexpectedly large segment of the asthma genes were STAT6 independent; this correlated with sustained inflammatory events in these mice. Notably, induction of asthma in STAT6-deficient mice resulted in gene induction not seen in wild-type mice. These results raise concern that therapeutic blockade of STAT6 in the asthmatic setting may reprogram the genetic signature, resulting in alternative lung pathology, which we indeed observed in STAT6-deficient mice. These results provide unprecedented insight into the complex steps involved in the pathogenesis of allergic airway responses; as such, these results have significant therapeutic and clinical implications.


Subject(s)
Asthma/genetics , Asthma/immunology , Disease Models, Animal , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Trans-Activators/physiology , Transcription, Genetic/immunology , Administration, Intranasal , Allergens/administration & dosage , Animals , Antigens, Fungal/administration & dosage , Antigens, Fungal/immunology , Aspergillus fumigatus/immunology , Asthma/physiopathology , Gene Expression Profiling/methods , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligonucleotide Array Sequence Analysis/methods , Ovalbumin/administration & dosage , Ovalbumin/immunology , STAT6 Transcription Factor , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/deficiency , Trans-Activators/genetics
19.
Am J Respir Cell Mol Biol ; 29(4): 458-64, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12702542

ABSTRACT

Asthma, a complex chronic inflammatory pulmonary disorder, is on the rise despite intense ongoing research, underscoring the need for new scientific inquiry. In an effort to provide unbiased insight into the pathogenesis of this disease, we took an empirical approach involving transcript expression profiling of lung tissue from mice with experimental asthma. Employing asthma models induced by different allergens (ovalbumin [OVA] and Aspergillus fumigatus), we found strong induction of trefoil factor-2 (TFF2), a gene involved in epithelial restitution and mucosal secretion in the gastrointestinal tract. Using a combination of pharmacologic delivery and transgenic overexpression, TFF2 was demonstrated to be strongly induced in the lung by interleukin (IL)-4 and IL-13. Notably, TFF2 induction by both OVA and pharmacologic delivery of IL-4 and IL-13 was dependent upon signal transducer and activator of transcription (STAT)6. However, the upregulation of TFF2 by both chronic expression of IL-4 and Aspergillus fumigatus antigen was independent of STAT6. These results establish that TFF2 is an allergen-induced lung gene product differentially regulated by Th2 cytokines and STAT6. Given the important role of trefoil factors in wound healing, epithelial restitution, and maintenance of mucosal integrity in the gastrointestinal tract, these results support a potential role for TFF2, in both the acute and chronic phase of experimental asthma, via separate induction pathways.


Subject(s)
Asthma/metabolism , Cytokines/immunology , Growth Substances/immunology , Lung/physiopathology , Mucins , Muscle Proteins , Neuropeptides , Peptides/immunology , Th2 Cells/immunology , Trans-Activators/immunology , Allergens/pharmacology , Animals , Aspergillus fumigatus/immunology , Asthma/genetics , Asthma/physiopathology , Bronchi/immunology , Bronchi/metabolism , Bronchi/physiopathology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Growth Substances/genetics , Interleukin-13/immunology , Interleukin-13/pharmacology , Interleukin-4/immunology , Interleukin-4/pharmacology , Lung/drug effects , Lung/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Ovalbumin/pharmacology , Peptides/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , STAT6 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/immunology , Trefoil Factor-2 , Trefoil Factor-3
SELECTION OF CITATIONS
SEARCH DETAIL
...