Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1206-17, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27106291

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a critical driver of acute lung injury and fibrosis. Injury leads to activation of TGF-ß, which regulates changes in the cellular and matrix makeup of the lung during the repair and fibrosis phase. TGF-ß can also initiate alveolar epithelial cell (AEC) apoptosis. Injury leads to destruction of the laminin-rich basement membrane, which is replaced by a provisional matrix composed of arginine-glycine-aspartate (RGD) motif-containing plasma matrix proteins, including vitronectin and fibronectin. To determine the role of specific matrix proteins on TGF-ß-induced apoptosis, we studied primary AECs cultured on different matrix conditions and utilized mice with deletion of vitronectin (Vtn(-/-)) or mice in which the vitronectin RGD motif is mutated to nonintegrin-binding arginine-glycine-glutamate (RGE) (Vtn(RGE/RGE)). We found that AECs cultured on fibronectin and vitronectin or in wild-type mouse serum are resistant to TGF-ß-induced apoptosis. In contrast, AECs cultured on laminin or in serum from Vtn(-/-) or Vtn(RGE/RGE) mice undergo robust TGF-ß-induced apoptosis. Plasminogen activator inhibitor-1 (PAI-1) sensitizes AECs to greater apoptosis by disrupting AEC engagement to vitronectin. Inhibition of integrin-associated signaling proteins augments AEC apoptosis. Mice with transgenic deletion of PAI-1 have less apoptosis after bleomycin, but deletion of vitronectin or disruption of the vitronectin RGD motif reverses this protection, suggesting that the proapoptotic function of PAI-1 is mediated through vitronectin inhibition. Collectively, these data suggest that integrin-matrix signaling is an important regulator of TGF-ß-mediated AEC apoptosis and that PAI-1 functions as a natural regulator of this interaction.


Subject(s)
Alveolar Epithelial Cells/physiology , Apoptosis , Transforming Growth Factor beta/physiology , Vitronectin/physiology , Amino Acid Motifs , Animals , Cells, Cultured , Integrins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Interaction Domains and Motifs , Signal Transduction , Vitronectin/chemistry
2.
Am J Pathol ; 185(4): 1001-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25661109

ABSTRACT

Fibrosis is a multicellular process leading to excessive extracellular matrix deposition. Factors that affect lung epithelial cell proliferation and activation may be important regulators of the extent of fibrosis after injury. We and others have shown that activated alveolar epithelial cells (AECs) directly contribute to fibrogenesis by secreting mesenchymal proteins, such as type I collagen. Recent evidence suggests that epithelial cell acquisition of mesenchymal features during carcinogenesis and fibrogenesis is regulated by several mesenchymal transcription factors. Induced expression of direct inhibitors to these mesenchymal transcription factors offers a potentially novel therapeutic strategy. Inhibitor of DNA-binding 2 (Id2) is an inhibitory helix-loop-helix transcription factor that is highly expressed by lung epithelial cells during development and has been shown to coordinate cell proliferation and differentiation of cancer cells. We found that overexpression of Id2 in primary AECs promotes proliferation by inhibiting a retinoblastoma protein/c-Abl interaction leading to greater c-Abl activity. Id2 also blocks transforming growth factor ß1-mediated expression of type I collagen by inhibiting Twist, a prominent mesenchymal basic helix-loop-helix transcription factor. In vivo, Id2 induced AEC proliferation and protected mice from lung fibrosis. By using a high-throughput screen, we found that histone deacetylase inhibitors induce Id2 expression by adult AECs. Collectively, these findings suggest that Id2 expression by AECs can be induced, and overexpression of Id2 affects AEC phenotype, leading to protection from fibrosis.


Subject(s)
Inhibitor of Differentiation Protein 2/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Pulmonary Fibrosis/metabolism , Twist-Related Protein 1/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bleomycin , Cell Proliferation/drug effects , Collagen/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice, Inbred C57BL , Protein Binding/drug effects , Pulmonary Fibrosis/pathology , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism
3.
J Immunol ; 193(10): 5229-39, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25281715

ABSTRACT

Progressive fibrosis involves accumulation of activated collagen-producing mesenchymal cells. Fibrocytes are hematopoietic-derived cells with mesenchymal features that potentially have a unique and critical function during fibrosis. Fibrocytes have been proposed as an important direct contributor of type I collagen deposition during fibrosis based largely on fate-mapping studies. To determine the functional contribution of hematopoietic cell-derived type I collagen to fibrogenesis, we use a double-transgenic system to specifically delete the type I collagen gene across a broad population of hematopoietic cells. These mice develop a robust fibrotic response similar to littermate genotype control mice injured with bleomycin indicating that fibrocytes are not a necessary source of type I collagen. Using collagen-promoter GFP mice, we find that fibrocytes express type I collagen. However, fibrocytes with confirmed deletion of the type I collagen gene have readily detectable intracellular type I collagen indicating that uptake of collagen from neighboring cells account for much of the fibrocyte collagen. Collectively, these results clarify several seemingly conflicting reports regarding the direct contribution of fibrocytes to collagen deposition.


Subject(s)
Collagen Type I/deficiency , Pulmonary Fibrosis/genetics , Animals , Bleomycin , Cell Differentiation , Cell Lineage , Cells, Cultured , Collagen Type I/genetics , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Transport , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
4.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L786-96, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24508728

ABSTRACT

Fibrogenesis involves a pathological accumulation of activated fibroblasts and extensive matrix remodeling. Profibrotic cytokines, such as TGF-ß, stimulate fibroblasts to overexpress fibrotic matrix proteins and induce further expression of profibrotic cytokines, resulting in progressive fibrosis. Connective tissue growth factor (CTGF) is a profibrotic cytokine that is indicative of fibroblast activation. Epithelial cells are abundant in the normal lung, but their contribution to fibrogenesis remains poorly defined. Profibrotic cytokines may activate epithelial cells with protein expression and functions that overlap with the functions of active fibroblasts. We found that alveolar epithelial cells undergoing TGF-ß-mediated mesenchymal transition in vitro were also capable of activating lung fibroblasts through production of CTGF. Alveolar epithelial cell expression of CTGF was dramatically reduced by inhibition of Rho signaling. CTGF reporter mice demonstrated increased CTGF promoter activity by lung epithelial cells acutely after bleomycin in vivo. Furthermore, mice with lung epithelial cell-specific deletion of CTGF had an attenuated fibrotic response to bleomycin. These studies provide direct evidence that epithelial cell activation initiates a cycle of fibrogenic effector cell activation during progressive fibrosis. Therapy targeted at epithelial cell production of CTGF offers a novel pathway for abrogating this progressive cycle and limiting tissue fibrosis.


Subject(s)
Autocrine Communication , Connective Tissue Growth Factor/physiology , Epithelial Cells/pathology , Paracrine Communication , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Blotting, Western , Bronchoalveolar Lavage , Connective Tissue Growth Factor/antagonists & inhibitors , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fluorescent Antibody Technique , Hydroxyproline/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
Am J Pathol ; 183(5): 1559-1570, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012677

ABSTRACT

Fibrosis is characterized by accumulation of activated fibroblasts and pathological deposition of fibrillar collagens. Activated fibroblasts overexpress matrix proteins and release factors that promote further recruitment of activated fibroblasts, leading to progressive fibrosis. The contribution of epithelial cells to this process remains unknown. Epithelium-directed injury may lead to activation of epithelial cells with phenotypes and functions similar to activated fibroblasts. Prior reports that used a reporter gene fate-mapping strategy are limited in their ability to investigate the functional significance of epithelial cell-derived mesenchymal proteins during fibrogenesis. We found that lung epithelial cell-derived collagen I activates fibroblast collagen receptor discoidin domain receptor-2, contributes significantly to fibrogenesis, and promotes resolution of lung inflammation. Alveolar epithelial cells undergoing transforming growth factor-ß-mediated mesenchymal transition express several other secreted profibrotic factors and are capable of activating lung fibroblasts. These studies provide direct evidence that activated epithelial cells produce mesenchymal proteins that initiate a cycle of fibrogenic effector cell activation, leading to progressive fibrosis. Therapy targeted at epithelial cell production of type I collagen offers a novel pathway for abrogating this progressive cycle and for limiting tissue fibrosis but may lead to sustained lung injury/inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Mesoderm/metabolism , Proteins/metabolism , Animals , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Deletion , Humans , Mice , Organ Specificity , Pneumonia/metabolism , Pneumonia/pathology , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...