Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cells Tissues Organs ; 210(1): 10-23, 2021.
Article in English | MEDLINE | ID: mdl-33910192

ABSTRACT

The basement membrane interacts directly with the vocal fold epithelium. Signaling between the basement membrane and the epithelium modulates gene regulation, differentiation, and proliferation. The purpose of this study was to identify an appropriate simple single-protein substrate for growth of rabbit vocal fold epithelial cells. Vocal folds from 3 New Zealand white rabbits (Oryctolagus cuniculus) were treated to isolate epithelial cells, and cells were seeded onto cell culture inserts coated with collagen I, collagen IV, laminin, or fibronectin. Transepithelial electrical resistance (TEER) was measured, and phase contrast microscopy, PanCK, CK14, and E-cadherin immunofluorescence were utilized to assess for epithelial cell-type characteristics. Further investigation via immunofluorescence labeling was conducted to assess proliferation (Ki67) and differentiation (Vimentin). There was a significant main effect of substrate on TEER, with collagen IV eliciting the highest, and laminin the lowest resistance. Assessment of relative TEER across cell lines identified a larger range of TEER in collagen I and laminin. Phase contrast imaging identified altered morphology in the laminin condition, but cell layer depth did not appear to be related to TEER, differentiation, or morphology. Ki67 staining additionally showed no significant difference in proliferation. All conditions had confluent epithelial cells and dispersed mesenchymal cells, with increased mesenchymal cell numbers over time; however, a higher proportion of mesenchymal cells was observed in the laminin condition. The results suggest collagen IV is a preferable basement membrane substrate for in vitro vocal fold epithelial primary cell culture, providing consistent TEER and characteristic cell morphology, and that laminin is an unsuitable substrate for vocal fold epithelial cells and may promote mesenchymal cell proliferation.


Subject(s)
Epithelial Cells , Vocal Cords , Animals , Basement Membrane , Cell Adhesion , Cell Count , Cells, Cultured , Collagen Type IV , Laminin , Rabbits
2.
Am J Respir Crit Care Med ; 187(6): 630-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23306543

ABSTRACT

RATIONALE: Alveolar epithelial cells (AECs) play central roles in the response to lung injury and the pathogenesis of pulmonary fibrosis. OBJECTIVES: We aimed to determine the role of ß-catenin in alveolar epithelium during bleomycin-induced lung fibrosis. METHODS: Genetically modified mice were developed to selectively delete ß-catenin in AECs and were crossed to cell fate reporter mice that express ß-galactosidase (ßgal) in cells of AEC lineage. Mice were given intratracheal bleomycin (0.04 units) and assessed for AEC death, inflammation, lung injury, and fibrotic remodeling. Mouse lung epithelial cells (MLE12) with small interfering RNA knockdown of ß-catenin underwent evaluation for wound closure, proliferation, and bleomycin-induced cytotoxicity. MEASUREMENTS AND MAIN RESULTS: Increased ß-catenin expression was noted in lung parenchyma after bleomycin. Mice with selective deletion of ß-catenin in AECs had greater AEC death at 1 week after bleomycin, followed by increased numbers of fibroblasts and enhanced lung fibrosis as determined by semiquantitative histological scoring and total collagen content. However, no differences in lung inflammation or protein levels in bronchoalveolar lavage were noted. In vitro, ß-catenin-deficient AECs showed increased bleomycin-induced cytotoxicity as well as reduced proliferation and impaired wound closure. Consistent with these findings, mice with AEC ß-catenin deficiency showed delayed recovery after bleomycin. CONCLUSIONS: ß-Catenin in the alveolar epithelium protects against bleomycin-induced fibrosis. Our studies suggest that AEC survival and wound healing are enhanced through ß-catenin-dependent mechanisms. Activation of the developmentally important ß-catenin pathway in AECs appears to contribute to epithelial repair after epithelial injury.


Subject(s)
Lung Injury/pathology , Pulmonary Alveoli/physiology , Pulmonary Fibrosis/pathology , beta Catenin/physiology , Animals , Bleomycin/adverse effects , Disease Models, Animal , Epithelium , In Situ Nick-End Labeling , Lung Injury/chemically induced , Mice , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Wound Healing/physiology
3.
Exp Lung Res ; 38(3): 124-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22394286

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by interstitial lung infiltrates, dyspnea, and progressive respiratory failure. Reports linking telomerase mutations to familial interstitial pneumonia (FIP) suggest that telomerase activity and telomere length maintenance are important in disease pathogenesis. To investigate the role of telomerase in lung fibrotic remodeling, intratracheal bleomycin was administered to mice deficient in telomerase reverse transcriptase (TERT) or telomerase RNA component (TERC) and to wild-type controls. TERT-deficient and TERC-deficient mice were interbred to the F6 and F4 generation, respectively, when they developed skin manifestations and infertility. Fibrosis was scored using a semiquantitative scale and total lung collagen was measured using a hydroxyprolinemicroplate assay. Telomere lengths were measured in peripheral blood leukocytes and isolated type II alveolar epithelial cells (AECs). Telomerase activity in type II AECs was measured using a real-time polymerase chain reaction (PCR)-based system. Following bleomycin, TERT-deficient and TERC-deficient mice developed an equivalent inflammatory response and similar lung fibrosis (by scoring of lung sections and total lung collagen content) compared to controls, a pattern seen in both early (F1) and later (F6 TERT and F4 TERC) generations. Telomere lengths were reduced in peripheral blood leukocytes and isolated type II AECs from F6 TERT-deficient and F4 TERC-deficient mice compared to controls. Telomerase deficiency in a murine model leads to telomere shortening, but does not predispose to enhanced bleomycin-induced lung fibrosis. Additional genetic or environmental factors may be necessary for development of fibrosis in the presence of telomerase deficiency.


Subject(s)
Bleomycin/toxicity , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/enzymology , Telomerase/deficiency , Telomere Homeostasis/drug effects , Airway Remodeling/drug effects , Airway Remodeling/genetics , Animals , Antibiotics, Antineoplastic/toxicity , Collagen/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Idiopathic Pulmonary Fibrosis/genetics , Leukocytes/drug effects , Leukocytes/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , RNA/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere Homeostasis/genetics , Telomere Shortening/drug effects , Telomere Shortening/genetics
4.
Proc Natl Acad Sci U S A ; 108(26): 10562-7, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670280

ABSTRACT

Evidence of endoplasmic reticulum (ER) stress has been found in lungs of patients with familial and sporadic idiopathic pulmonary fibrosis. We tested whether ER stress causes or exacerbates lung fibrosis by (i) conditional expression of a mutant form of surfactant protein C (L188Q SFTPC) found in familial interstitial pneumonia and (ii) intratracheal treatment with the protein misfolding agent tunicamycin. We developed transgenic mice expressing L188Q SFTPC exclusively in type II alveolar epithelium by using the Tet-On system. Expression of L188Q SFTPC induced ER stress, as determined by increased expression of heavy-chain Ig binding protein (BiP) and splicing of X-box binding protein 1 (XBP1) mRNA, but no lung fibrosis was identified in the absence of a second profibrotic stimulus. After intratracheal bleomycin, L188Q SFTPC-expressing mice developed exaggerated lung fibrosis and reduced static lung compliance compared with controls. Bleomycin-treated L188Q SFTPC mice also demonstrated increased apoptosis of alveolar epithelial cells and greater numbers of fibroblasts in the lungs. With a complementary model, intratracheal tunicamycin treatment failed to induce lung remodeling yet resulted in augmentation of bleomycin-induced fibrosis. These data support the concept that ER stress produces a dysfunctional epithelial cell phenotype that facilitates fibrotic remodeling. ER stress pathways may serve as important therapeutic targets in idiopathic pulmonary fibrosis.


Subject(s)
Endoplasmic Reticulum/metabolism , Lung/pathology , Pulmonary Fibrosis/pathology , Animals , Apoptosis/genetics , Bleomycin/toxicity , Intercellular Signaling Peptides and Proteins , Lung/drug effects , Lung/metabolism , Mice , Mice, Transgenic , Mutation , Peptides/genetics , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Surfactant-Associated Protein C , Reverse Transcriptase Polymerase Chain Reaction , Tunicamycin/toxicity
5.
Am J Physiol Lung Cell Mol Physiol ; 300(6): L887-97, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441353

ABSTRACT

The response of alveolar epithelial cells (AECs) to lung injury plays a central role in the pathogenesis of pulmonary fibrosis, but the mechanisms by which AECs regulate fibrotic processes are not well defined. We aimed to elucidate how transforming growth factor-ß (TGFß) signaling in lung epithelium impacts lung fibrosis in the intratracheal bleomycin model. Mice with selective deficiency of TGFß receptor 2 (TGFßR2) in lung epithelium were generated and crossed to cell fate reporter mice that express ß-galactosidase (ß-gal) in cells of lung epithelial lineage. Mice were given intratracheal bleomycin (0.08 U), and the following parameters were assessed: AEC death by terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling assay, inflammation by total and differential cell counts from bronchoalveolar lavage, fibrosis by scoring of trichrome-stained lung sections, and total lung collagen content. Mice with lung epithelial deficiency of TGFßR2 had improved AEC survival, despite greater lung inflammation, after bleomycin administration. At 3 wk after bleomycin administration, mice with epithelial TGFßR2 deficiency showed a significantly attenuated fibrotic response in the lungs, as determined by semiquantitatve scoring and total collagen content. The reduction in lung fibrosis in these mice was associated with a marked decrease in the lung fibroblast population, both total lung fibroblasts and epithelial-to-mesenchymal transition-derived (S100A4(+)/ß-gal(+)) fibroblasts. Attenuation of TGFß signaling in lung epithelium provides protection from bleomycin-induced fibrosis, indicating a critical role for the epithelium in transducing the profibrotic effects of this cytokine.


Subject(s)
Bleomycin/adverse effects , Epithelium/metabolism , Fibroblasts/metabolism , Lung Injury/chemically induced , Protein Serine-Threonine Kinases/physiology , Pulmonary Alveoli/drug effects , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Blotting, Western , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelium/drug effects , Epithelium/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Fluorescent Antibody Technique , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Transgenic , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , beta-Galactosidase/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 299(4): L442-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20562227

ABSTRACT

Single-dose intratracheal bleomycin has been instrumental for understanding fibrotic lung remodeling, but fails to recapitulate several features of idiopathic pulmonary fibrosis (IPF). Since IPF is thought to result from recurrent alveolar injury, we aimed to develop a repetitive bleomycin model that results in lung fibrosis with key characteristics of human disease, including alveolar epithelial cell (AEC) hyperplasia. Wild-type and cell fate reporter mice expressing ß-galactosidase in cells of lung epithelial lineage were given intratracheal bleomycin after intubation, and lungs were harvested 2 wk after a single or eighth biweekly dose. Lungs were evaluated for fibrosis and collagen content. Bronchoalveolar lavage (BAL) was performed for cell counts. TUNEL staining and immunohistochemistry were performed for pro-surfactant protein C (pro-SP-C), Clara cell 10 (CC-10), ß-galactosidase, S100A4, and α-smooth muscle actin. Lungs from repetitive bleomycin mice had marked fibrosis with prominent AEC hyperplasia, similar to usual interstitial pneumonia (UIP). Compared with single dosing, repetitive bleomycin mice had greater fibrosis by scoring, morphometry, and collagen content; increased TUNEL+ AECs; and reduced inflammatory cells in BAL. Sixty-four percent of pro-SP-C+ cells in areas of fibrosis expressed CC-10 in the repetitive model, suggesting expansion of a bronchoalveolar stem cell-like population. In reporter mice, 50% of S100A4+ lung fibroblasts were derived from epithelial mesenchymal transition compared with 33% in the single-dose model. With repetitive bleomycin, fibrotic remodeling persisted 10 wk after the eighth dose. Repetitive intratracheal bleomycin results in marked lung fibrosis with prominent AEC hyperplasia, features reminiscent of UIP.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Animals , Apoptosis , Bronchoalveolar Lavage Fluid , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Immunoenzyme Techniques , In Situ Nick-End Labeling , Integrases , Intubation, Intratracheal , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , beta-Galactosidase/metabolism
7.
Am J Respir Crit Care Med ; 180(7): 657-65, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19556518

ABSTRACT

RATIONALE: Lung fibroblasts are key mediators of fibrosis resulting in accumulation of excessive interstitial collagen and extracellular matrix, but their origins are not well defined. OBJECTIVES: We aimed to elucidate the contribution of lung epithelium-derived fibroblasts via epithelial-mesenchymal transition (EMT) in the intratracheal bleomycin model. METHODS: Primary type II alveolar epithelial cells were cultured from Immortomice and exposed to transforming growth factor-beta(1) and epidermal growth factor. Cell fate reporter mice that permanently mark cells of lung epithelial lineage with beta-galactosidase were developed to study EMT, and bone marrow chimeras expressing green fluorescent protein under the control of the fibroblast-associated S100A4 promoter were generated to examine bone marrow-derived fibroblasts. Mice were given intratracheal bleomycin (0.08 unit). Immunostaining was performed for S100A4, beta-galactosidase, green fluorescent protein, and alpha-smooth muscle actin. MEASUREMENTS AND MAIN RESULTS: In vitro, primary type II alveolar epithelial cells undergo phenotypic changes of EMT when exposed to transforming growth factor-beta(1) and epidermal growth factor with loss of prosurfactant protein C and E-cadherin and gain of S100A4 and type I procollagen. In vivo, using cell fate reporter mice, approximately one-third of S100A4-positive fibroblasts were derived from lung epithelium 2 weeks after bleomycin administration. From bone marrow chimera studies, one-fifth of S100A4-positive fibroblasts were derived from bone marrow at this same time point. Myofibroblasts rarely derived from EMT or bone marrow progenitors. CONCLUSIONS: Both EMT and bone marrow progenitors contribute to S100A4-positive fibroblasts in bleomycin-induced lung fibrosis. However, neither origin is a principal contributor to lung myofibroblasts.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Epithelial Cells/metabolism , Fibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Animals , Bleomycin , Blotting, Western/methods , Cell Culture Techniques , Disease Models, Animal , Epithelium/metabolism , Fibroblasts/cytology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...