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










Publication year range
1.
Am J Respir Crit Care Med ; 200(4): 431-443, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30950644

ABSTRACT

Rationale: Histologic stains have been used as the gold standard to visualize extracellular matrix (ECM) changes associated with airway remodeling in asthma, yet they provide no information on the biochemical and structural characteristics of the ECM, which are vital to understanding alterations in tissue function.Objectives: To demonstrate the use of nonlinear optical microscopy (NLOM) and texture analysis algorithms to image fibrillar collagen (second harmonic generation) and elastin (two-photon excited autofluorescence), to obtain biochemical and structural information on the remodeled ECM environment in asthma.Methods: Nontransplantable donor lungs from donors with asthma (n = 13) and control (n = 12) donors were used for the assessment of airway collagen and elastin fibers by NLOM, and extraction of lung fibroblasts for in vitro experiments.Measurements and Main Results: Fibrillar collagen is not only increased but also highly disorganized and fragmented within large and small asthmatic airways compared with control subjects, using NLOM imaging. Furthermore, such structural alterations are present in pediatric and adult donors with asthma, irrespective of fatal disease. In vitro studies demonstrated that asthmatic airway fibroblasts are deficient in their packaging of fibrillar collagen-I and express less decorin, important for collagen fibril packaging. Packaging of collagen fibrils was found to be more disorganized in asthmatic airways compared with control subjects, using transmission electron microscopy.Conclusions: NLOM imaging enabled the structural assessment of the ECM, and the data suggest that airway remodeling in asthma involves the progressive accumulation of disorganized fibrillar collagen by airway fibroblasts. This study highlights the future potential clinical application of NLOM to assess airway remodeling in vivo.


Subject(s)
Airway Remodeling/physiology , Asthma/metabolism , Elastin/metabolism , Fibrillar Collagens/metabolism , Fibroblasts/metabolism , Lung/metabolism , Adolescent , Adult , Asthma/pathology , Child , Collagen Type I/metabolism , Decorin/metabolism , Elastin/ultrastructure , Extracellular Matrix , Female , Fibrillar Collagens/ultrastructure , Humans , In Vitro Techniques , Lung/cytology , Lung/ultrastructure , Male , Microscopy, Electron, Transmission , Nonlinear Optical Microscopy , Young Adult
2.
BMC Pulm Med ; 17(1): 24, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28137284

ABSTRACT

BACKGROUND: Recognition of the airway epithelium as a central mediator in the pathogenesis of asthma has necessitated greater understanding of the aberrant cellular mechanisms of the epithelium in asthma. The architecture of chromatin is integral to the regulation of gene expression and is determined by modifications to the surrounding histones and DNA. The acetylation, methylation, phosphorylation, and ubiquitination of histone tail residues has the potential to greatly alter the accessibility of DNA to the cells transcriptional machinery. DNA methylation can also interrupt binding of transcription factors and recruit chromatin remodelers resulting in general gene silencing. Although previous studies have found numerous irregularities in the expression of genes involved in asthma, the contribution of epigenetic regulation of these genes is less well known. We propose that the gene expression of epigenetic modifying enzymes is cell-specific and influenced by asthma status in tissues derived from the airways. METHODS: Airway epithelial cells (AECs) isolated by pronase digestion or endobronchial brushings and airway fibroblasts obtained by outgrowth technique from healthy and asthmatic donors were maintained in monolayer culture. RNA was analyzed for the expression of 82 epigenetic enzymes across 5 families of epigenetic modifying enzymes. Western blot and immunohistochemistry were also used to examine expression of 3 genes. RESULTS: Between AECs and airway fibroblasts, we identified cell-specific gene expression in each of the families of epigenetic modifying enzymes; specifically 24 of the 82 genes analyzed showed differential expression. We found that 6 histone modifiers in AECs and one in fibroblasts were differentially expressed in cells from asthmatic compared to healthy donors however, not all passed correction. In addition, we identified a corresponding increase in Aurora Kinase A (AURKA) protein expression in epithelial cells from asthmatics compared to those from non-asthmatics. CONCLUSIONS: In summary, we have identified cell-specific variation in gene expression in each of the families of epigenetic modifying enzymes in airway epithelial cells and airway fibroblasts. These data provide insight into the cell-specific variation in epigenetic regulation which may be relevant to cell fate and function, and disease susceptibility.


Subject(s)
Asthma/genetics , Epigenesis, Genetic , Epithelial Cells/enzymology , Fibroblasts/enzymology , Histones/metabolism , Asthma/enzymology , Cell Differentiation , Cells, Cultured , DNA Methylation , Gene Expression , Gene Silencing , Humans , Linear Models , Protein Processing, Post-Translational , Respiratory System/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Am J Respir Cell Mol Biol ; 56(3): 291-299, 2017 03.
Article in English | MEDLINE | ID: mdl-27788019

ABSTRACT

Limited in vivo models exist to investigate the lung airway epithelial role in repair, regeneration, and pathology of chronic lung diseases. Herein, we introduce a novel animal model in asthma-a xenograft system integrating a differentiating human asthmatic airway epithelium with an actively remodeling rodent mesenchyme in an immunocompromised murine host. Human asthmatic and nonasthmatic airway epithelial cells were seeded into decellularized rat tracheas. Tracheas were ligated to a sterile cassette and implanted subcutaneously in the flanks of nude mice. Grafts were harvested at 2, 4, or 6 weeks for tissue histology, fibrillar collagen, and transforming growth factor-ß activation analysis. We compared immunostaining in these xenografts to human lungs. Grafted epithelial cells generated a differentiated epithelium containing basal, ciliated, and mucus-expressing cells. By 4 weeks postengraftment, asthmatic epithelia showed decreased numbers of ciliated cells and decreased E-cadherin expression compared with nonasthmatic grafts, similar to human lungs. Grafts seeded with asthmatic epithelial cells had three times more fibrillar collagen and induction of transforming growth factor-ß isoforms at 6 weeks postengraftment compared with nonasthmatic grafts. Asthmatic epithelium alone is sufficient to drive aberrant mesenchymal remodeling with fibrillar collagen deposition in asthmatic xenografts. Moreover, this xenograft system represents an advance over current asthma models in that it permits direct assessment of the epithelial-mesenchymal trophic unit.


Subject(s)
Asthma/pathology , Heterografts/pathology , Lung/pathology , Pulmonary Fibrosis/pathology , Adult , Airway Remodeling , Animals , Asthma/physiopathology , Demography , Disease Models, Animal , Epidermal Growth Factor/metabolism , Extracellular Matrix/metabolism , Female , Heterografts/physiopathology , Humans , Male , Middle Aged , Rats, Inbred F344 , Signal Transduction , Tissue Donors , Transforming Growth Factor beta1/metabolism , Young Adult
4.
Int J Biochem Cell Biol ; 68: 59-69, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26315281

ABSTRACT

The epithelium of asthmatics is characterized by reduced expression of E-cadherin and increased expression of the basal cell markers ck-5 and p63 that is indicative of a relatively undifferentiated repairing epithelium. This phenotype correlates with increased proliferation, compromised wound healing and an enhanced capacity to undergo epithelial-mesenchymal transition (EMT). The transcription factor ß-catenin plays a vital role in epithelial cell differentiation and regeneration, depending on the co-factor recruited. Transcriptional programs driven by the ß-catenin/CBP axis are critical for maintaining an undifferentiated and proliferative state, whereas the ß-catenin/p300 axis is associated with cell differentiation. We hypothesized that disrupting the ß-catenin/CBP signaling axis would promote epithelial differentiation and inhibit EMT. We treated monolayer cultures of human airway epithelial cells with TGFß1 in the presence or absence of the selective small molecule ICG-001 to inhibit ß-catenin/CBP signaling. We used western blots to assess expression of an EMT signature, CBP, p300, ß-catenin, fibronectin and ITGß1 and scratch wound assays to assess epithelial cell migration. Snai-1 and -2 expressions were determined using q-PCR. Exposure to TGFß1 induced EMT, characterized by reduced E-cadherin expression with increased expression of α-smooth muscle actin and EDA-fibronectin. Either co-treatment or therapeutic administration of ICG-001 completely inhibited TGFß1-induced EMT. ICG-001 also reduced the expression of ck-5 and -19 independent of TGFß1. Exposure to ICG-001 significantly inhibited epithelial cell proliferation and migration, coincident with a down regulation of ITGß1 and fibronectin expression. These data support our hypothesis that modulating the ß-catenin/CBP signaling axis plays a key role in epithelial plasticity and function.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Epithelial Cells/metabolism , Peptide Fragments/genetics , Pyrimidinones/pharmacology , Sialoglycoproteins/genetics , Transforming Growth Factor beta1/pharmacology , beta Catenin/genetics , Actins/genetics , Actins/metabolism , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Keratin-19/genetics , Keratin-19/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Primary Cell Culture , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
5.
Respir Res ; 16: 95, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26243279

ABSTRACT

BACKGROUND: Epigenetic adjustments of the chromatin architecture through histone modifications are reactive to the environment and can establish chromatin states which are permissive or repressive to gene expression. Epigenetic regulation of gene expression is cell specific and therefore, it is important to understand its contribution to individual cellular responses in tissues like the airway epithelium which forms the mucosal barrier to the inhaled environment within the lung. The airway epithelium of asthmatics is abnormal with dysregulation of genes such as epidermal growth factor receptor (EGFR), the ΔN isoform of the transcription factor p63 (ΔNp63), and signal transducer and activator of transcription 6 (STAT6), integral to differentiation, proliferation, and inflammation. It is important to establish in diseases like asthma how histone modifications affect tissue responses such as proliferation and differentiation. OBJECTIVES: To characterize the global histone acetylation and methylation status in the epithelium of asthmatic compared to healthy subjects and to identify the impact of these variations on genes involved in epithelial functions. METHODS: Whole lungs were obtained from healthy and asthmatic subjects (n = 6) from which airway epithelial cells (AECs) were isolated and airway sections were taken for analysis of histone lysine acetylation and methylation by immunohistochemistry. AECs were subjected to chromatin immunoprecipitation (ChIP) using anti-H3K18ac and anti-H3K4me2 antibodies followed by RT-PCR targeting ΔNp63, EGFR, and STAT6. AECs were also treated with TSA and changes in ΔNp63, EGFR, and STAT6 expression were determined. RESULTS: We identified an increase in the acetylation of lysine 18 on histone 3 (H3K18ac) and trimethylation of lysine 9 on histone 3 (H3K9me3) in the airway epithelium of asthmatic compared to healthy subjects. We found increased association of H3K18ac around the transcription start site of ΔNp63, EGFR, and STAT6 in AECs of asthmatics. However, we were unable to modify the expression of these genes with the use of the HDAC inhibitor TSA in healthy subjects. DISCUSSION: The airway epithelium from asthmatic subjects displays increased acetylation of H3K18 and association of this mark around the transcription start site of ΔNp63, EGFR, and STAT6. These findings suggest a complex interaction between histone modifications and gene regulation in asthma.


Subject(s)
Asthma/metabolism , Histone Acetyltransferases/metabolism , Lysine/metabolism , Respiratory Mucosa/metabolism , Acetylation , Adolescent , Adult , Asthma/pathology , Cell Differentiation/physiology , Cells, Cultured , Child , Child, Preschool , Female , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Male , Respiratory Mucosa/pathology , Young Adult
6.
Am J Respir Cell Mol Biol ; 51(6): 830-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24922586

ABSTRACT

Cigarette smoke-induced emphysema and small airway remodeling are the anatomic bases of chronic obstructive pulmonary disease (COPD), but the pathogenesis of these changes is unclear, and current treatments for COPD are minimally effective. To evaluate the role of signal transducer and activator of transcription (STAT)-4 in cigarette smoke-induced small airway remodeling, we used C57BL/6J (wild type [WT]) and STAT4-/- mice exposed to air or cigarette smoke for 6 months and isolated airway and parenchymal fibroblasts. We also compared the results with those obtained with human fibroblasts. We found that STAT4-/- mice were protected against smoke-induced small airway remodeling but not emphysema. STAT4 is abundantly expressed in airway compared with parenchymal-derived fibroblasts isolated from normal human and murine lung. WT airway fibroblasts proliferate faster than STAT4-/- airway fibroblasts, whereas there is no difference between strains for parenchymal fibroblasts. IL-12 is up-regulated in the lung after smoke exposure, and IL-12 receptor B2 is expressed on airway and parenchymal fibroblasts in mouse and human lung. Treatment with IL-12 causes phosphorylation of STAT4 in WT airway fibroblasts. Exposure of WT airway, but not parenchymal, fibroblasts to IL-12 causes increased expression of collagen 1α1 and transforming growth factor ß1, factors involved in small airway remodeling, whereas STAT4-/- fibroblasts are unresponsive to IL-12. These results indicate that IL-12 can drive small airway remodeling via STAT4 signaling and suggest that treatment with clinically available anti-IL-12p40 drugs might provide a new approach to preventing small airway remodeling in cigarette smokers.


Subject(s)
Pulmonary Fibrosis/metabolism , STAT4 Transcription Factor/physiology , Smoking/metabolism , Airway Remodeling , Animals , Cell Proliferation , Cell Shape , Cells, Cultured , Humans , Interleukin-12/metabolism , Mice, Inbred BALB C , Phosphorylation , Protein Processing, Post-Translational , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Signal Transduction , Smoking/adverse effects
7.
Am J Respir Cell Mol Biol ; 49(6): 978-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23837456

ABSTRACT

The airway epithelium in asthma displays altered repair and incomplete barrier formation. Basal cells are the progenitor cells of the airway epithelium, and can repopulate other cell types after injury. We previously reported increased numbers of basal cells expressing the transcription factor p63 in the airway epithelium of patients with asthma. Here we sought to determine the molecular consequences of p63 expression in basal human airway epithelial cells during wound repair. Because at least six isoforms of p63 exist (N-terminally truncated [ΔN] versus transcriptional activation promoter variants and α, ß, or γ 3' splice variants), the expression of all isoforms was investigated in primary human airway epithelial cells (pHAECs). We modulated p63 expression, using small interfering RNA (siRNA) and adenoviral constructs to determine the effects of p63 on 21 candidate target genes by RT-PCR, and on repair using a scratch wound assay. We found that basal pHAECs from asthmatic and nonasthmatic donors predominantly expressed the N-terminally truncated p63α variant (ΔNp63α) isoform, with no disease-specific differences in expression. The knockdown of ΔNp63, using specific siRNA, decreased the expression of 11 out of 21 genes associated with epithelial repair and differentiation, including ß-catenin, epidermal growth factor receptor, and Jagged1. The loss of ΔNp63 significantly inhibited wound closure (which was associated with the decreased expression of ß-catenin and Jagged1), reduced epithelial proliferation as measured by Ki-67 staining, and increased E-cadherin expression, potentially preventing cytokinesis. In conclusion, ΔNp63α is the major isoform expressed in basal pHAECs, and is essential for epithelial wound repair. The role of ΔNp63α in epithelial barrier integrity requires further study to understand its role in health and disease.


Subject(s)
Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wound Healing/genetics , Wound Healing/physiology , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
8.
Am J Respir Cell Mol Biol ; 49(4): 662-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23742006

ABSTRACT

The molecular basis for airway epithelial fragility in asthma has remained unclear. We investigated whether the loss of caveolin-1, the major component of caveolae and a known stabilizer of adherens junctions, contributes to epithelial barrier dysfunction in asthma. We studied the expression of caveolin-1 and adhesion molecules E-cadherin and ß-catenin in airway sections, and we cultured bronchial epithelial cells from patients with asthma and from healthy control subjects. To determine the functional role of caveolin-1, we investigated the effects of caveolin-1 up-regulation and down-regulation on E-cadherin expression, barrier function, and proallergic activity in the human bronchial epithelial cell lines 16HBE and BEAS-2B. The membrane expression of caveolin-1 was significantly lower in airway epithelia from patients with asthma than from subjects without asthma, and this lower expression was maintained in vitro upon air-liquid interface and submerged culturing. Importantly, reduced caveolin-1 expression was accompanied by a loss of junctional E-cadherin and ß-catenin expression, disrupted epithelial barrier function, and increased levels of the proallergic cytokine thymic stromal lymphopoietin (TSLP). Furthermore, E-cadherin redistribution upon exposure to epidermal growth factor or house dust mite was paralleled by the internalization of caveolin-1 in 16HBE cells. These effects appear to be causally related, because the short, interfering RNA down-regulation of caveolin-1 resulted in the delocalization of E-cadherin and barrier dysfunction in 16HBE cells. Moreover, caveolin-1 overexpression improved barrier function and reduced TSLP expression in BEAS-2B cells. Together, our data demonstrate a crucial role for caveolin-1 in epithelial cell-cell adhesion, with important consequences for epithelial barrier function and the promotion of Th2 responses in asthma.


Subject(s)
Asthma/metabolism , Bronchi/immunology , Bronchi/metabolism , Caveolin 1/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Adherens Junctions/genetics , Adherens Junctions/immunology , Adherens Junctions/metabolism , Adolescent , Adult , Animals , Asthma/genetics , Asthma/immunology , Cadherins/genetics , Cadherins/immunology , Cadherins/metabolism , Caveolin 1/genetics , Caveolin 1/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Child , Down-Regulation , Epidermal Growth Factor/genetics , Epidermal Growth Factor/immunology , Epidermal Growth Factor/metabolism , Female , Humans , Male , Pyroglyphidae/immunology , Respiratory Mucosa/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Up-Regulation , beta Catenin/genetics , beta Catenin/immunology , beta Catenin/metabolism
9.
PLoS One ; 7(7): e40478, 2012.
Article in English | MEDLINE | ID: mdl-22808170

ABSTRACT

BACKGROUND: Normal airway epithelial barrier function is maintained by cell-cell contacts which require the translocation of adhesion proteins at the cell surface, through membrane vesicle trafficking and fusion events. Myoferlin and dysferlin, members of the multiple-C2-domain Ferlin superfamily, have been implicated in membrane fusion processes through the induction of membrane curvature. The objectives of this study were to examine the expression of dysferlin and myoferlin within the human airway and determine the roles of these proteins in airway epithelial homeostasis. METHODS: The expression of dysferlin and myoferlin were evaluated in normal human airway sections by immunohistochemistry, and primary human airway epithelial cells and fibroblasts by immuno blot. Localization of dysferlin and myoferlin in epithelial cells were determined using confocal microscopy. Functional outcomes analyzed included cell adhesion, protein expression, and cell detachment following dysferlin and myoferlin siRNA knock-down, using the human bronchial epithelial cell line, 16HBE. RESULTS: Primary human airway epithelial cells express both dysferlin and myoferlin whereas fibroblasts isolated from bronchi and the parenchyma only express myoferlin. Expression of dysferlin and myoferlin was further localized within the Golgi, cell cytoplasm and plasma membrane of 16HBE cells using confocal micrscopy. Treatment of 16HBE cells with myoferlin siRNA, but not dysferlin siRNA, resulted in a rounded cell morphology and loss of cell adhesion. This cell shedding following myoferlin knockdown was associated with decreased expression of tight junction molecule, zonula occludens-1 (ZO-1) and increased number of cells positive for apoptotic markers Annexin V and propidium iodide. Cell shedding was not associated with release of the innate inflammatory cytokines IL-6 and IL-8. CONCLUSIONS/SIGNIFICANCE: This study demonstrates the heterogeneous expression of myoferlin within epithelial cells and fibroblasts of the respiratory airway. The effect of myoferlin on the expression of ZO-1 in airway epithelial cells indicates its role in membrane fusion events that regulate cell detachment and apoptosis within the airway epithelium.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Zonula Occludens-1 Protein/metabolism , Animals , Calcium-Binding Proteins/genetics , Cattle , Caveolin 1/metabolism , Cell Adhesion , Cell Death , Cell Line , Cell Shape/genetics , Down-Regulation , Dysferlin , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Knockdown Techniques , Gene Silencing , Humans , Inflammation/genetics , Inflammation/pathology , Membrane Proteins/genetics , Muscle Proteins/genetics , Protein Transport , Tight Junctions/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
J Allergy Clin Immunol ; 129(4): 1116-25.e6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22227418

ABSTRACT

BACKGROUND: The airway epithelium is the first line of defense against inhaled insults and therefore must be capable of coordinating appropriate inflammatory and immune responses. OBJECTIVE: We sought to test the hypothesis that the nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome, an intracellular danger-sensing complex, plays a critical role in airway epithelium-mediated immune responses to urban particulate matter (PM) exposure. METHODS: In this study we (1) identified NLRP3 and caspase-1 expression in human airway epithelium bronchus and primary cells, (2) characterized NLRP3 inflammasome-mediated IL-1ß production from human airway epithelium in response to PM, and (3) performed in vivo PM exposure experiments with wild-type and Nlrp3(-/-) mice. RESULTS: Our results demonstrate that human airway epithelium contains a functional NLRP3 inflammasome that responds to PM exposure with caspase-1 cleavage and production of IL-1ß. Exposure of Nlrp3(-/-) and wild-type mice to PM in vivo demonstrates NLRP3-dependent production of IL-1ß in the lung, airway neutrophilia, and increases in CD11c(+hi)/MHC class II(+hi) cell numbers in intrathoracic lymph nodes. CONCLUSION: Our study is the first to characterize airway epithelial NLRP3 inflammasome-mediated immune responses to PM exposure, which might have implications in patients with asthma and other lung diseases.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Particulate Matter/immunology , Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Immunophenotyping , Interleukin-1beta/metabolism , Leucine-Rich Repeat Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Transport , Proteins/genetics
11.
Am J Respir Cell Mol Biol ; 45(5): 1090-100, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21642587

ABSTRACT

A substantial proportion of healthcare cost associated with asthma is attributable to exacerbations of the disease. Within the airway, the epithelium forms the mucosal immune barrier, the first structural cell defense against common environmental insults such as respiratory syncytial virus (RSV) and particulate matter. We sought to characterize the phenotype of differentiated asthmatic-derived airway epithelial cultures and their intrinsic inflammatory responses to environmental challenges. Air-liquid interface (ALI) cultures were generated from asthmatic (n = 6) and nonasthmatic (n = 6) airway epithelial cells. Airway tissue and ALI cultures were analyzed by immunohistochemistry for cytokeratin-5, E-cadherin, Ki67, Muc5AC, NF-κB, the activation of p38, and apoptosis. ALI cultures were exposed to RSV (4 × 10(6) plaque forming unit/ml), particulate matter collected by Environmental Health Canada (EHC-93, 100 µg/ml), or mechanically wounded for 24, 48, and 96 hours and basolateral supernatants analyzed for inflammatory cytokines, using Luminex and ELISA. The airway epithelium in airway sections of patients with asthma as well as in vitro ALI cultures demonstrated a less differentiated epithelium, characterized by elevated numbers of basal cells marked by the expression of cytokeratin-5, increased phosphorylation of p38 mitogen-activated protein kinase, and less adherens junction protein E-cadherin. Transepithelial resistance was not different between asthmatic and nonasthmatic cultures. In response to infection with RSV, exposure to EHC-93, or mechanical wounding, asthmatic ALI cultures released greater concentrations of IL-6, IL-8, and granulocyte macrophage colony-stimulating factor, compared with nonasthmatic cultures (P < 0.05). This parallel ex vivo and in vitro study of the asthmatic epithelium demonstrates an intrinsically altered phenotype and aberrant inflammatory response to common environmental challenges, compared with nonasthmatic epithelium.


Subject(s)
Air Pollution/adverse effects , Asthma/metabolism , Asthma/virology , Particulate Matter/adverse effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Respiratory Syncytial Viruses/metabolism , Adult , Apoptosis , Asthma/chemically induced , Cadherins/metabolism , Cells, Cultured , Child , Child, Preschool , Cytokines/metabolism , Female , Humans , Keratin-5/metabolism , Ki-67 Antigen/metabolism , Male , Mucin 5AC/metabolism , NF-kappa B/metabolism , Phosphorylation , Young Adult , p38 Mitogen-Activated Protein Kinases
12.
Arterioscler Thromb Vasc Biol ; 30(11): 2196-204, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20724702

ABSTRACT

OBJECTIVE: Ferlins are known to regulate plasma membrane repair in muscle cells and are linked to muscular dystrophy and cardiomyopathy. Recently, using proteomic analysis of caveolae/lipid rafts, we reported that endothelial cells (EC) express myoferlin and that it regulates membrane expression of vascular endothelial growth factor receptor 2 (VEGFR-2). The goal of this study was to document the presence of other ferlins in EC. METHODS AND RESULTS: EC expressed another ferlin, dysferlin, and that in contrast to myoferlin, it did not regulate VEGFR-2 expression levels or downstream signaling (nitric oxide and Erk1/2 phosphorylation). Instead, loss of dysferlin in subconfluent EC resulted in deficient adhesion followed by growth arrest, an effect not observed in confluent EC. In vivo, dysferlin was also detected in intact and diseased blood vessels of rodent and human origin, and angiogenic challenge of dysferlin-null mice resulted in impaired angiogenic response compared with control mice. Mechanistically, loss of dysferlin in cultured EC caused polyubiquitination and proteasomal degradation of platelet endothelial cellular adhesion molecule-1 (PECAM-1/CD31), an adhesion molecule essential for angiogenesis. In addition, adenovirus-mediated gene transfer of PECAM-1 rescued the abnormal adhesion of EC caused by dysferlin gene silencing. CONCLUSIONS: Our data describe a novel pathway for PECAM-1 regulation and broaden the functional scope of ferlins in angiogenesis and specialized ferlin-selective protein cargo trafficking in vascular settings.


Subject(s)
Cell Adhesion/physiology , Endothelial Cells/physiology , Membrane Proteins/physiology , Muscle Proteins/physiology , Neovascularization, Pathologic/physiopathology , Animals , Cattle , Cell Proliferation , Cells, Cultured , Down-Regulation , Dysferlin , Humans , Membrane Proteins/biosynthesis , Mice , Muscle Proteins/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
13.
Fibrogenesis Tissue Repair ; 3: 11, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20602770

ABSTRACT

PURPOSE: To evaluate the effect of the anti-fibrotic protein serum amyloid P (SAP) on radiation-induced oral mucositis (OM) and fibrosis in a hamster cheek-pouch model. EXPERIMENTAL DESIGN: Hamsters received a single dose of radiation (40 Gy) to the left everted cheek pouch to induce significant OM. The protective therapeutic potential of SAP was evaluated using varying dosing regimens. The extent of OM was measured using a validated six-point scoring scheme ranging from 0 (normal tissue, no mucositis) to 5 (complete ulceration). Fibrotic remodeling was also visualized histologically and quantified at later time points using collagen gene expression. RESULTS: SAP treatment attenuated the profile of radiation-induced oral mucositis by delaying the time of onset, reducing the peak value, and enhancing the resolution of injury. The peak mucositis score was reduced by approximately 0.5 grade in SAP-treated animals. The number of animal days with a score of >/= 3 was reduced by 48% in the SAP-treated group, compared with the saline control group (P < 0.01). SAP also inhibited the extent of tissue remodeling and decreased radiation-induced increases in myofibroblast number. Attenuated collagen deposition and gene expression was also observed in the cheek pouches of hamsters treated with SAP at both 16 and 28 days post-radiation. CONCLUSIONS: SAP treatment significantly attenuated radiation-induced injury. In particular, SAP attenuated the severity of OM and inhibited pathogenic remodeling. This suggests that SAP may be a useful therapy for the palliation of side effects observed during treatment for head and neck cancer.

14.
Am J Respir Cell Mol Biol ; 43(6): 641-51, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20061511

ABSTRACT

Given the contribution various fibroblast subsets make to wound healing and tissue remodeling, the concept of lung fibroblast heterogeneity is of great interest. However, the mechanisms contributing to this heterogeneity are unknown. To this aim, we compared molecular and biophysical characteristics of fibroblasts concurrently isolated from normal human proximal bronchi (B-FBR) and distal lung parenchyma (P-FBR). Using quantitative RT-PCR, spontaneous expression of more than 30 genes related to repair and remodeling was analyzed. All P-FBR lines demonstrated significantly increased basal α-smooth muscle actin (α-SMA) mRNA and protein expression levels when compared with donor-matched B-FBR. These differences were not associated with sex, age, or disease history of lung tissue donors. In contrast to B-FBR, P-FBR displayed enhanced transforming growth factor (TGF)-ß/Smad signaling at baseline, and inhibition of either ALK-5 or neutralization of endogenously produced and activated TGF-ß substantially decreased basal α-SMA protein in P-FBR. Both B-FBR and P-FBR up-regulated α-SMA after stimulation with TGF-ß1, and basal expression levels of TGF-ß1, TGF-ßRI, and TGF-ßRII were not significantly different between fibroblast pairs. Blockade of metalloproteinase-dependent activation of endogenous TGF-ß did not significantly modify α-SMA expression in P-FBR. However, resistance to mechanical tension of these cells was significantly higher in comparison with B-FBR, and added TGF-ß1 significantly increased stiffness of both cell monolayers. Our data suggest that in contrast with human normal bronchial tissue explants, lung parenchyma produces mesenchymal cells with a myofibroblastic phenotype by intrinsic mechanisms of TGF-ß activation in feed-forward manner. These results also offer a new insight into mechanisms of human fibroblast heterogeneity and their function in the airway and lung tissue repair and remodeling.


Subject(s)
Actins/metabolism , Bronchi/cytology , Fibroblasts/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Actins/genetics , Adolescent , Adult , Aged , Child, Preschool , Demography , Dipeptides/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad3 Protein/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta1/pharmacology , Young Adult
15.
Int Arch Allergy Immunol ; 152(1): 12-27, 2010.
Article in English | MEDLINE | ID: mdl-19940501

ABSTRACT

BACKGROUND: Although the accepted paradigm is that the proteins stored in eosinophil crystalloid granules are translated from messenger RNA transcribed in the cell nucleus, recent ultrastructural evidence suggests that protein synthesis may also take place within eosinophilic granules. METHODS: We used 2 different methods to detect the presence of DNA and RNA in eosinophil secretory granules. Using bromodeoxyuridine, a thymidine analogue, and bromouridine, a uracil analogue, we labeled the DNA and RNA in eosinophils in vivo in rabbits. Immunoelectron microscopy to localize these molecules was performed on ultrathin sections of blood and bone marrow eosinophils using monoclonal anti-bromodeoxyuridine antibody with IgG as a control. The immunogold grain density was measured in each subcellular compartment within the eosinophils and analyzed using image analysis software. A combination of DNA/CD63 immunofluorescence staining and a fluorescently labeled molecular probe that stains RNA was used to examine the presence of DNA and RNA in the secretory granules of human blood eosinophils. RESULTS: The mean density of bromodeoxyuridine-labeled DNA and bromouridine-labeled RNA immunogold grains in the secretory granules of blood and bone marrow eosinophils were significantly higher (p < 0.0005) than cytoplasmic or background staining. We also demonstrated the existence of DNA and RNA in the CD63-positive secretory granules of human peripheral blood eosinophils by means of immunofluorescent staining and a fluorescently labeled molecular probe. CONCLUSIONS: These results provide evidence that eosinophil granules are the site of DNA and RNA synthesis and suggest the potential for a new role(s) for eosinophil-secretory granules.


Subject(s)
DNA/metabolism , Eosinophils/metabolism , Eosinophils/ultrastructure , RNA/metabolism , Secretory Vesicles/metabolism , Animals , Bone Marrow Cells/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Bromodeoxyuridine/metabolism , Bromouracil/analogs & derivatives , DNA/analysis , Eosinophils/chemistry , Eosinophils/cytology , Female , Fluorescent Dyes/metabolism , Humans , Immunohistochemistry , Microscopy, Immunoelectron , Organic Chemicals/metabolism , RNA/analysis , Rabbits , Uridine/analogs & derivatives , Uridine/metabolism
16.
Am J Respir Crit Care Med ; 180(2): 122-33, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19406982

ABSTRACT

RATIONALE: Airway remodeling in asthma is associated with the accumulation of fibroblasts, the primary cell responsible for synthesis and secretion of extracellular matrix proteins. The process by which the number of fibroblasts increases in asthma is poorly understood, but epithelial-mesenchymal transition (EMT) may play a significant role. OBJECTIVES: To evaluate whether EMT occurs in primary airway epithelial cells (AECs), the mechanisms involved, and if this process is altered in asthmatic AECs. METHODS: AECs were obtained from subjects with asthma (n = 8) and normal subjects without asthma (n = 10). Monolayer and air-liquid interface-AEC (ALI-AEC) cultures were treated with transforming growth factor (TGF)-beta1 (10 ng/ml) for 72 hours and assayed for mesenchymal and epithelial markers using quantitative polymerase chain reaction, confocal microscopy, and immunoblot. The involvement of BMP-7, Smad3, and MAPK-mediated signaling were also evaluated. MEASUREMENTS AND MAIN RESULTS: TGF-beta1-induced EMT in AEC monolayers derived from subjects with asthma and normal donors. EMT was characterized by changes in cell morphology, increased expression of mesenchymal markers EDA-fibronectin, vimentin, alpha-smooth muscle actin, and collagen-1, and loss of epithelial markers E-cadherin and zonular occludin-1. Inhibition of TGF-beta1-induced signaling with Smad3-inhibiting siRNA or TGF-beta1-neutralizing antibodies prevented and reversed EMT, respectively, whereas BMP-7 had no effect. In ALI-AEC cultures derived from normal subjects, EMT was confined to basally situated cells, whereas in asthmatic ALI-AEC cultures EMT was widespread throughout the epithelium. CONCLUSIONS: TGF-beta1 induces EMT in a Smad3-dependent manner in primary AECs. However, in asthmatic-derived ALI-AEC cultures, the number of cells undergoing EMT is greater. These findings support the hypothesis that epithelial repair in asthmatic airways is dysregulated.


Subject(s)
Asthma/pathology , Cell Dedifferentiation/drug effects , Epithelial Cells/drug effects , Transforming Growth Factor beta1/pharmacology , Adolescent , Asthma/etiology , Asthma/metabolism , Case-Control Studies , Cell Culture Techniques , Child , Child, Preschool , Epithelial Cells/physiology , Extracellular Matrix Proteins/physiology , Female , Humans , Intercellular Signaling Peptides and Proteins/physiology , Male , Mesenchymal Stem Cells , Recombinant Proteins , Signal Transduction , Young Adult
17.
PLoS One ; 3(12): e4039, 2008.
Article in English | MEDLINE | ID: mdl-19112509

ABSTRACT

Bone morphogenic protein (BMP)-7 is a member of the BMP family which are structurally and functionally related, and part of the TGFbeta super family of growth factors. BMP-7 has been reported to inhibit renal fibrosis and TGFbeta1-induced epithelial-mesenchymal transition (EMT), in part through negative interactions with TGFbeta1 induced Smad 2/3 activation. We utilized in vivo bleomycin-induced fibrosis models in the skin and lung to determine the potential therapeutic effect of BMP-7. We then determined the effect of BMP-7 on TGFbeta1-induced EMT in lung epithelial cells and collagen production by human lung fibroblasts. We show that BMP-7 did not affect bleomycin-induced fibrosis in either the lung or skin in vivo; had no effect on expression of pro-fibrotic genes by human lung fibroblasts, either at rest or following exposure to TGFbeta1; and did not modulate TGFbeta1-induced EMT in human lung epithelial cells. Taken together our data indicates that BMP-7 has no anti-fibrotic effect in lung or skin fibrosis either in vivo or in vitro. This suggests that the therapeutic options for BMP-7 may be confined to the renal compartment.


Subject(s)
Bleomycin/pharmacology , Bone Morphogenetic Protein 7/physiology , Fibrosis/chemically induced , Fibrosis/metabolism , Gene Expression Regulation , Lung/pathology , Skin/pathology , Animals , Bone Morphogenetic Protein 7/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Skin/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism
18.
Am J Respir Crit Care Med ; 178(12): 1227-37, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18849495

ABSTRACT

RATIONALE: Acute respiratory distress syndrome (ARDS) manifests clinically as a consequence of septic and/or traumatic injury in the lung. Oxygen therapy remains a major therapeutic intervention in ARDS, but this can contribute further to lung damage. Patients with ARDS are highly susceptible to viral infection and it may be due to altered Toll-like receptor (TLR) expression. OBJECTIVES: To evaluate the role of TLR3 in ARDS. METHODS: TLR3 expression and signaling was determined in airway epithelial cells after in vitro hyperoxia challenge. Using a murine model of hyperoxia-induced lung injury, the role of TLR3 was determined using either TLR3-gene deficient mice or a specific neutralizing antibody directed to TLR3. MEASUREMENTS AND MAIN RESULTS: Increased TLR3 expression was observed in airway epithelial cells from patients with ARDS. Further, hyperoxic conditions alone were a major stimulus for increased TLR3 expression and activation in cultured human epithelial cells. Interestingly, TLR3(-/-) mice exhibited less acute lung injury, activation of apoptotic cascades, and extracellular matrix deposition after 5 days of 80% oxygen compared with wild-type (TLR3(+/+)) mice under the same conditions. Administration of a monoclonal anti-TLR3 antibody to TLR3(+/+) mice exposed to hyperoxic conditions likewise protected these mice from lung injury and inflammation. CONCLUSIONS: The potential for redundancy in function as well as cross-talk between distinct TLRs may indeed contribute to whether the inflammatory cascade can be effectively disrupted once signaling has been initiated. Together, these data show that TLR3 has a major role in the development of ARDS-like pathology in the absence of a viral pathogen.


Subject(s)
Gene Expression , Hyperoxia/complications , RNA/genetics , Respiratory Distress Syndrome/genetics , Toll-Like Receptor 3/genetics , Animals , Apoptosis , Biopsy , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Flow Cytometry , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Immunohistochemistry , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 3/biosynthesis
19.
Stem Cells ; 26(10): 2576-85, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18653771

ABSTRACT

The airway epithelium is the first line of contact with the inhaled external environment and is continuously exposed to and injured by pollutants, allergens, and viruses. However, little is known about epithelial repair and in particular the identity and role of tissue resident stem/progenitor cells that may contribute to epithelial regeneration. The aims of the present study were to identify, isolate, and characterize side population (SP) cells in human tracheobronchial epithelium. Epithelial cells were obtained from seven nontransplantable healthy lungs and four asthmatic lungs by pronase digestion. SP cells were identified by verapamil-sensitive efflux of the DNA-binding dye Hoechst 33342. Using flow cytometry, CD45(-) SP, CD45(+) SP, and non-SP cells were isolated and sorted. CD45(-) SP cells made up 0.12% +/- 0.01% of the total epithelial cell population in normal airway but 4.1% +/- 0.06% of the epithelium in asthmatic airways. All CD45(-) SP cells showed positive staining for epithelial-specific markers cytokeratin-5, E-cadherin, ZO-1, and p63. CD45(-) SP cells exhibited stable telomere length and increased colony-forming and proliferative potential, undergoing population expansion for at least 16 consecutive passages. In contrast with non-SP cells, fewer than 100 CD45(-) SP cells were able to generate a multilayered and differentiated epithelium in air-liquid interface culture. SP cells are present in human tracheobronchial epithelium, exhibit both short- and long-term proliferative potential, and are capable of generation of differentiated epithelium in vitro. The number of SP cells is significantly greater in asthmatic airways, providing evidence of dysregulated resident SP cells in the asthmatic epithelium. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Epithelial Cells/cytology , Respiratory System/cytology , Adolescent , Adult , Asthma/pathology , Bronchi/cytology , Cell Count , Cell Differentiation , Cells, Cultured , Child , Child, Preschool , Colony-Forming Units Assay , Demography , Female , Humans , Leukocyte Common Antigens/metabolism , Male , Phenotype , Telomere/metabolism , Trachea/cytology
20.
J Biol Chem ; 283(19): 12898-908, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18353785

ABSTRACT

In response to transforming growth factor beta1 (TGFbeta) stimulation, fibroblasts modify their integrin repertoire and adhesive capabilities to certain extracellular matrix proteins. Although TGFbeta has been shown to increase the expression of specific alphav integrins, the mechanisms underlying this are unknown. In this study we demonstrate that TGFbeta1 increased both beta3 integrin subunit mRNA and protein levels as well as surface expression of alphavbeta3 in human lung fibroblasts. TGFbeta1-induced alphavbeta3 expression was strongly adhesion-dependent and associated with increased focal adhesion kinase and c-Src kinase phosphorylation. Inhibition of beta3 integrin activation by the Arg-Gly-Asp tripeptide motif-specific disintegrin echistatin or alphavbeta3 blocking antibody prevented the increase in beta3 but not beta5 integrin expression. In addition, echistatin inhibited TGFbeta1-induced p38 MAPK but not Smad3 activation. Furthermore, inhibition of the Src family kinases, but not focal adhesion kinase, completely abrogated TGFbeta1-induced expression of alphavbeta3 and p38 MAPK phosphorylation but not beta5 integrin expression and Smad3 activation. The TGFbeta1-induced alphavbeta3 expression was blocked by pharmacologic and genetic inhibition of p38 MAPK- but not Smad2/3-, Sp1-, ERK-, phosphatidylinositol 3-kinase, and NF-kappaB-dependent pathways. Our results demonstrate that TGFbeta1 induces alphavbeta3 integrin expression via a beta3 integrin-, c-Src-, and p38 MAPK-dependent pathway. These data identify a novel mechanism for TGFbeta1 signaling in human lung fibroblasts by which they may contribute to normal and pathological wound healing.


Subject(s)
Gene Expression Regulation/drug effects , Integrin alphaVbeta3/metabolism , Integrin beta3/metabolism , Lung/cytology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Enzyme Activation/drug effects , Fibroblasts , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Smad Proteins/metabolism , Sp1 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...