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










Publication year range
1.
Dev Cell ; 58(24): 2974-2991.e6, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-37977149

ABSTRACT

The lung contains multiple progenitor cell types, but how their responses are choreographed during injury repair and whether this changes with age is poorly understood. We report that histone H3 lysine 9 di-methylation (H3K9me2), mediated by the methyltransferase G9a, regulates the dynamics of distal lung epithelial progenitor cells and that this regulation deteriorates with age. In aged mouse lungs, H3K9me2 loss coincided with fewer alveolar type 2 (AT2) cell progenitors and reduced alveolar regeneration but increased the frequency and activity of multipotent bronchioalveolar stem cells (BASCs) and bronchiolar progenitor club cells. H3K9me2 depletion in young mice decreased AT2 progenitor activity and impaired alveolar injury repair. Conversely, H3K9me2 depletion increased chromatin accessibility of bronchiolar cell genes, increased BASC frequency, and accelerated bronchiolar cell injury repair. These findings indicate that during aging, the epigenetic regulation that coordinates lung progenitor cells' regenerative responses becomes dysregulated, aiding our understanding of age-related susceptibility to lung disease.


Subject(s)
Epigenesis, Genetic , Lung , Mice , Animals , Lung/metabolism , Chromatin/metabolism , Methylation , Protein Processing, Post-Translational
2.
JCI Insight ; 8(1)2023 01 10.
Article in English | MEDLINE | ID: mdl-36454643

ABSTRACT

Dysfunction of alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, is implicated in pulmonary disease pathogenesis, highlighting the importance of human in vitro models. However, AEC2-like cells in culture have yet to be directly compared to their in vivo counterparts at single-cell resolution. Here, we performed head-to-head comparisons among the transcriptomes of primary (1°) adult human AEC2s, their cultured progeny, and human induced pluripotent stem cell-derived AEC2s (iAEC2s). We found each population occupied a distinct transcriptomic space with cultured AEC2s (1° and iAEC2s) exhibiting similarities to and differences from freshly purified 1° cells. Across each cell type, we found an inverse relationship between proliferative and maturation states, with preculture 1° AEC2s being most quiescent/mature and iAEC2s being most proliferative/least mature. Cultures of either type of human AEC2s did not generate detectable alveolar type 1 cells in these defined conditions; however, a subset of iAEC2s cocultured with fibroblasts acquired a transitional cell state described in mice and humans to arise during fibrosis or following injury. Hence, we provide direct comparisons of the transcriptomic programs of 1° and engineered AEC2s, 2 in vitro models that can be harnessed to study human lung health and disease.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Animals , Mice , Transcriptome , Alveolar Epithelial Cells/metabolism , Lung/pathology , Pulmonary Alveoli/pathology
3.
Cell Rep ; 39(2): 110662, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417699

ABSTRACT

Lung progenitor cells are crucial for regeneration following injury, yet it is unclear whether lung progenitor cells can be functionally engrafted after transplantation. We transplanted organoid cells derived from alveolar type II (AT2) cells enriched by SCA1-negative status (SNO) or multipotent SCA1-positive progenitor cells (SPO) into injured mouse lungs. Transplanted SNO cells are retained in the alveolar regions, whereas SPO cells incorporate into airway and alveolar regions. Single-cell transcriptomics demonstrate that transplanted SNO cells are comparable to native AT2 cells. Transplanted SPO cells exhibit transcriptional hallmarks of alveolar and airway cells, as well as transitional cell states identified in disease. Transplanted cells proliferate after re-injury of recipient mice and retain organoid-forming capacity. Thus, lung epithelial organoid cells exhibit progenitor cell functions after reintroduction to the lung. This study reveals methods to interrogate lung progenitor cell potential and model transitional cell states relevant to pathogenic features of lung disease in vivo.


Subject(s)
Organoids , Spinocerebellar Ataxias , Animals , Cell Differentiation , Epithelial Cells , Lung , Mice , Stem Cells
4.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35315362

ABSTRACT

Type 2 alveolar epithelial cells (AT2s), facultative progenitor cells of the lung alveolus, play a vital role in the biology of the distal lung. In vitro model systems that incorporate human cells, recapitulate the biology of primary AT2s, and interface with the outside environment could serve as useful tools to elucidate functional characteristics of AT2s in homeostasis and disease. We and others recently adapted human induced pluripotent stem cell-derived AT2s (iAT2s) for air-liquid interface (ALI) culture. Here, we comprehensively characterize the effects of ALI culture on iAT2s and benchmark their transcriptional profile relative to both freshly sorted and cultured primary human fetal and adult AT2s. We find that iAT2s cultured at ALI maintain an AT2 phenotype while upregulating expression of transcripts associated with AT2 maturation. We then leverage this platform to assay the effects of exposure to clinically significant, inhaled toxicants including cigarette smoke and electronic cigarette vapor.


Subject(s)
Electronic Nicotine Delivery Systems , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Environmental Exposure , Epithelium , Humans , Induced Pluripotent Stem Cells/metabolism
5.
Cell Rep ; 35(5): 109055, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33905739

ABSTRACT

Coronavirus disease 2019 (COVID-19) is the latest respiratory pandemic caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2). Although infection initiates in the proximal airways, severe and sometimes fatal symptoms of the disease are caused by infection of the alveolar type 2 (AT2) cells of the distal lung and associated inflammation. In this study, we develop primary human lung epithelial infection models to understand initial responses of proximal and distal lung epithelium to SARS-CoV-2 infection. Differentiated air-liquid interface (ALI) cultures of proximal airway epithelium and alveosphere cultures of distal lung AT2 cells are readily infected by SARS-CoV-2, leading to an epithelial cell-autonomous proinflammatory response with increased expression of interferon signaling genes. Studies to validate the efficacy of selected candidate COVID-19 drugs confirm that remdesivir strongly suppresses viral infection/replication. We provide a relevant platform for study of COVID-19 pathobiology and for rapid drug screening against SARS-CoV-2 and emergent respiratory pathogens.


Subject(s)
Alveolar Epithelial Cells/virology , COVID-19 Drug Treatment , COVID-19/pathology , Lung/virology , SARS-CoV-2/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adult , Aged , Alanine/analogs & derivatives , Alanine/pharmacology , Alveolar Epithelial Cells/metabolism , COVID-19/metabolism , COVID-19/virology , Child, Preschool , Drug Discovery/methods , Epithelial Cells/virology , Epithelium/metabolism , Epithelium/virology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lung/pathology , Male , Middle Aged , Models, Biological , Primary Cell Culture , Respiratory Mucosa/virology , SARS-CoV-2/physiology , Virus Replication/drug effects
6.
Cell ; 184(8): 1990-2019, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33811810

ABSTRACT

The population is aging at a rate never seen before in human history. As the number of elderly adults grows, it is imperative we expand our understanding of the underpinnings of aging biology. Human lungs are composed of a unique panoply of cell types that face ongoing chemical, mechanical, biological, immunological, and xenobiotic stress over a lifetime. Yet, we do not fully appreciate the mechanistic drivers of lung aging and why age increases the risk of parenchymal lung disease, fatal respiratory infection, and primary lung cancer. Here, we review the molecular and cellular aspects of lung aging, local stress response pathways, and how the aging process predisposes to the pathogenesis of pulmonary disease. We place these insights into context of the COVID-19 pandemic and discuss how innate and adaptive immunity within the lung is altered with age.


Subject(s)
Aging , Cellular Senescence , Lung Diseases , Lung , Adaptive Immunity , Aged , Aging/immunology , Aging/pathology , COVID-19/immunology , COVID-19/pathology , Humans , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Oxidative Stress
10.
Nat Commun ; 8: 14922, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387316

ABSTRACT

Adenosquamous lung tumours, which are extremely poor prognosis, may result from cellular plasticity. Here, we demonstrate lineage switching of KRAS+ lung adenocarcinomas (ADC) to squamous cell carcinoma (SCC) through deletion of Lkb1 (Stk11) in autochthonous and transplant models. Chromatin analysis reveals loss of H3K27me3 and gain of H3K27ac and H3K4me3 at squamous lineage genes, including Sox2, ΔNp63 and Ngfr. SCC lesions have higher levels of the H3K27 methyltransferase EZH2 than the ADC lesions, but there is a clear lack of the essential Polycomb Repressive Complex 2 (PRC2) subunit EED in the SCC lesions. The pattern of high EZH2, but low H3K27me3 mark, is also prevalent in human lung SCC and SCC regions within ADSCC tumours. Using FACS-isolated populations, we demonstrate that bronchioalveolar stem cells and club cells are the likely cells-of-origin for SCC transitioned tumours. These findings shed light on the epigenetics and cellular origins of lineage-specific lung tumours.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Histones/metabolism , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Methylation , Mice, 129 Strain , Mice, Knockout , Polycomb Repressive Complex 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Cells, Cultured
11.
PLoS One ; 11(3): e0150383, 2016.
Article in English | MEDLINE | ID: mdl-26934369

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal disease of unknown etiology. A growing body of evidence indicates that it may result from an aberrant activation of alveolar epithelium, which induces the expansion of the fibroblast population, their differentiation to myofibroblasts and the excessive accumulation of extracellular matrix. The mechanisms that activate the alveolar epithelium are unknown, but several studies indicate that smoking is the main environmental risk factor for the development of IPF. In this study we explored the effect of cigarette smoke on the gene expression profile and signaling pathways in alveolar epithelial cells. Lung epithelial cell line from human (A549), was exposed to cigarette smoke extract (CSE) for 1, 3, and 5 weeks at 1, 5 and 10% and gene expression was evaluated by complete transcriptome microarrays. Signaling networks were analyzed with the Ingenuity Pathway Analysis software. At 5 weeks of exposure, alveolar epithelial cells acquired a fibroblast-like phenotype. At this time, gene expression profile revealed a significant increase of more than 1000 genes and deregulation of canonical signaling pathways such as TGF-ß and Wnt. Several profibrotic genes involved in EMT were over-expressed, and incomplete EMT was observed in these cells, and corroborated in mouse (MLE-12) and rat (RLE-6TN) epithelial cells. The secretion of activated TGF-ß1 increased in cells exposed to cigarette smoke, which decreased when the integrin alpha v gene was silenced. These findings suggest that the exposure of alveolar epithelial cells to CSE induces the expression and release of a variety of profibrotic genes, and the activation of TGF-ß1, which may explain at least partially, the increased risk of developing IPF in smokers.


Subject(s)
Epithelial Cells/pathology , Fibroblasts/pathology , Nicotiana/adverse effects , Pulmonary Alveoli/pathology , Smoke/adverse effects , Smoking/adverse effects , Transcriptome , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Integrins/genetics , Integrins/metabolism , Male , Mice , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Rats, Wistar , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
12.
J Immunol Res ; 2015: 984973, 2015.
Article in English | MEDLINE | ID: mdl-26347897

ABSTRACT

Lipoarabinomannan (LAM) is a lipid virulence factor secreted by Mycobacterium tuberculosis (Mtb), the etiologic agent of tuberculosis. LAM can be measured in the urine or serum of tuberculosis patients (TB-patients). Circulating monocytes are the precursor cells of alveolar macrophages and might be exposed to LAM in patients with active TB. We speculated that exposing monocytes to LAM could produce phenotypically and functionally immature macrophages. To test our hypothesis, human monocytes were stimulated with LAM (24-120 hours) and various readouts were measured. The study showed that when monocytes were exposed to LAM, the frequency of CD68(+), CD33(+), and CD86(+) macrophages decreased, suggesting that monocyte differentiation into mature macrophages was affected. Regarding functionality markers, TLR2(+) and TLR4(+) macrophages also decreased, but the percentage of MMR(+) expression did not change. LAM-exposed monocytes generated macrophages that were less efficient in producing proinflammatory cytokines such as TNF-α and IFN-γ; however, their phagocytic capacity was not modified. Taken together, these data indicate that LAM exposure influenced monocyte differentiation and produced poorly functional macrophages with a different phenotype. These results may help us understand how mycobacteria can limit the quality of the innate and adaptive immune responses.


Subject(s)
Cell Differentiation/immunology , Lipopolysaccharides/immunology , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/immunology , Antigens, Surface/metabolism , Cytokines/biosynthesis , Humans , Immunophenotyping , Macrophages/immunology , Monocytes/metabolism , Mycobacterium tuberculosis/immunology , Phagocytosis , Phenotype , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology
13.
Lung ; 193(2): 199-202, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25655494

ABSTRACT

OBJECTIVE: The objective of this study is to determine the effect of two angiotensin-converting enzyme inhibitors (ACEi) (Enalapril and Captopril), an angiotensin-II receptor inhibitor (Losartan) and a renin inhibitor (Aliskiren) on renin, TGF-ß1 and collagen expressions in human lung fibroblast cultures through real-time PCR and ELISA. MATERIALS AND METHODS: Normal commercial fibroblasts (CCD25) were exposed to 10(-6) M of enalapril, captopril, losartan, or aliskiren for 6 h. Subsequently, media were recovered and proteins were concentrated; RNA was extracted from the cells. Real time-PCR and ELISA were performed. RESULTS: ACEi and losartan-stimulated fibroblasts showed an increase in the expression of TGF-ß1, Collagen-Iα1 (Col-Iα1), and renin (except losartan) vs PolR2A (p < 0.05), and upregulation of TGF-ß1 protein (p < 0.01), except with aliskiren. CONCLUSION: Results show that ACEis and losartan could play a profibrosing role by inducing the overexpression of molecules such TGF-ß1 and Collagen.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Lung/pathology , Transcription, Genetic/drug effects , Amides/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Captopril/pharmacology , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Enalapril/pharmacology , Fibroblasts/pathology , Fibrosis , Fumarates/pharmacology , Humans , Losartan/pharmacology , Protein Biosynthesis/drug effects , Renin/antagonists & inhibitors , Renin/genetics , Renin/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
14.
Am J Respir Crit Care Med ; 191(4): 427-36, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25531246

ABSTRACT

RATIONALE: Hypersensitivity pneumonitis (HP) represents a lung inflammation provoked by exposure to a variety of antigens. Chronic HP may evolve to lung fibrosis. Bone marrow-derived fibrocytes migrate to injured tissues and contribute to fibrogenesis, but their role in HP is unknown. OBJECTIVES: To assess the possible participation of fibrocytes in chronic HP. METHODS: CD45(+)/CXCR4(+)/Col-I(+) circulating fibrocytes were evaluated by flow cytometry, and the presence of fibrocytes in HP and normal lungs by confocal microscopy. The concentration of CXCL12 in plasma and bronchoalveolar lavage fluids was quantified by ELISA. The effect of fibrocytes on lung fibroblasts and T lymphocytes was examined in co-cultures. MEASUREMENTS AND MAIN RESULTS: The percentage of circulating fibrocytes was significantly increased in patients with HP compared with healthy individuals (5.3 ± 3.4% vs. 0.8 ± 0.7%; P = 0.00004). Numerous fibrocytes were found infiltrating the HP lungs near fibroblasts and lymphocytes. Plasma CXCL12 concentration was significantly increased in patients with HP (2,303.3 ± 813.7 vs. 1,385.6 ± 318.5 pg/ml; P = 0.00003), and similar results were found in bronchoalveolar lavage fluids. The chemokine was primarily expressed by epithelial cells. In co-cultures, fibrocytes induced on lung fibroblasts a significant increase in the expression of α1 type I collagen, matrix metalloprotease-1, and platelet-derived growth factor-ß. Likewise, fibrocytes induced the up-regulation of CCL2 in HP lymphocytes and fibroblasts. CONCLUSIONS: These findings demonstrate that high levels of fibrocytes are present in the peripheral blood of patients with chronic HP and that these cells infiltrate the HP lungs. Fibrocytes may participate in the pathogenesis of HP, amplifying the inflammatory and fibrotic response by paracrine signaling inducing the secretion of a variety of proinflammatory and profibrotic molecules.


Subject(s)
Alveolitis, Extrinsic Allergic/pathology , Fibroblasts/metabolism , Pulmonary Fibrosis/etiology , Adult , Aged , Alveolitis, Extrinsic Allergic/metabolism , Biomarkers/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Chemokine CXCL12/metabolism , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Leukocyte Common Antigens/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Microscopy, Confocal , Middle Aged , Peptide Fragments/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/metabolism
15.
PLoS One ; 6(9): e25392, 2011.
Article in English | MEDLINE | ID: mdl-21980441

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF), a devastating lung disorder of unknown aetiology, and chronic hypersensitivity pneumonitis (HP), a disease provoked by an immunopathologic reaction to inhaled antigens, are two common interstitial lung diseases with uncertain pathogenic mechanisms. Previously, we have shown in other upper and lower airway diseases that immunoglobulin free light chains (FLCs) are increased and may be involved in initiating a local inflammation. In this study we explored if such a mechanism may also apply to HP and IPF. METHODS: In this study we examined the presence of FLC in serum and BAL fluid from 21 IPF and 22 HP patients and controls. IgG, IgE and tryptase concentrations were measured in BAL fluid only. The presence of FLCs, plasma cells, B cells and mast cells in lung tissue of 3 HP and 3 IPF patients and 1 control was analyzed using immunohistochemistry. RESULTS: FLC concentrations in serum and BAL fluid were increased in IPF and HP patients as compared to control subjects. IgG concentrations were only increased in HP patients, whereas IgE concentrations were comparable to controls in both patient groups. FLC-positive cells, B cells, plasma cells, and large numbers of activated mast cells were all detected in the lungs of HP and IPF patients, not in control lung. CONCLUSION: These results show that FLC concentrations are increased in serum and BAL fluid of IPF and HP patients and that FLCs are present within affected lung tissue. This suggests that FLCs may be involved in mediating pathology in both diseases.


Subject(s)
Alveolitis, Extrinsic Allergic/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Immunoglobulin Light Chains/metabolism , Adult , Aged , Alveolitis, Extrinsic Allergic/blood , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/physiopathology , Bronchoalveolar Lavage Fluid , Case-Control Studies , Female , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/physiopathology , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , Immunoglobulin Light Chains/blood , Lung/immunology , Lung/physiopathology , Male , Mast Cells/metabolism , Middle Aged , Plasma Cells/metabolism , Respiratory Function Tests , Tryptases/metabolism
16.
Am J Respir Crit Care Med ; 182(9): 1144-52, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20622038

ABSTRACT

RATIONALE: Fibrocytes are progenitor cells characterized by the simultaneous expression of mesenchymal, monocyte, and hematopoietic stem cell markers. We previously documented their presence in lungs of patients with idiopathic pulmonary fibrosis. However, the mechanisms involved in their migration, subsequent homing, and local role remain unclear. Matrix metalloproteinases (MMPs) facilitate cell migration and have been implicated in the pathogenesis of pulmonary fibrosis. OBJECTIVES: To evaluate the expression and role of matrix metalloproteinases in human fibrocytes. METHODS: Fibrocytes were purified from CD14(+) monocytes and cultured for 8 days; purity of fibrocyte cultures was 95% or greater as determined by flow cytometry. Conditioned media and total RNA were collected and the expression of MMP-1, MMP-2, MMP-7, MMP-8, and MMP-9 was evaluated by real-time polymerase chain reaction. Protein synthesis was examined using a Multiplex assay, Western blot, fluorescent immunocytochemistry, and confocal microscopy. MMP-2 and MMP-9 enzymatic activities were evaluated by gelatin zymography. Migration was assessed using collagen I-coated Boyden chambers. Stromal cell-derived factor-1α and platelet-derived growth factor-B were used as chemoattractant with or without a specific MMP-8 inhibitor. MEASUREMENTS AND MAIN RESULTS: Fibrocytes showed gene and protein expression of MMP-2, MMP-9, MMP-8, and MMP-7. MMP-2 and MMP-9 enzymatic activities were also demonstrated by gelatin zymography. Likewise, we found colocalization of MMP-8 and MMP-7 with type I collagen in fibrocytes. Fibrocyte migration toward platelet-derived growth factor-B or Stromal cell-derived factor-1α in collagen I-coated Boyden chambers was significantly reduced by a specific MMP-8 inhibitor. CONCLUSIONS: Our findings reveal that fibrocytes express a variety of MMPs and that MMP-8 actively participates in the process of fibrocyte migration.


Subject(s)
Cell Movement/physiology , Fibroblasts/enzymology , Matrix Metalloproteinases/physiology , Blotting, Western , Cell Migration Assays , Cells, Cultured , Collagen/biosynthesis , Extracellular Matrix/physiology , Humans , Immunophenotyping , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinases/metabolism , Microscopy, Confocal , Polymerase Chain Reaction , Pulmonary Fibrosis/physiopathology , Transforming Growth Factor beta1/pharmacology
17.
Am J Physiol Lung Cell Mol Physiol ; 299(2): L222-31, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20495078

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease characterized by the expansion of the fibroblast/myofibroblast population and aberrant remodeling. However, the origin of mesenchymal cells in this disorder is still under debate. Recent evidence indicates that epithelial-mesenchymal transition (EMT) induced primarily by TGF-beta1 plays an important role; however, studies regarding the opposite process, mesenchymal-epithelial transition, are scanty. We have previously shown that fibroblast growth factor-1 (FGF-1) inhibits several profibrogenic effects of TGF-beta1. In this study, we examined the effects of FGF-1 on TGF-beta1-induced EMT. A549 and RLE-6TN (human and rat) alveolar epithelial-like cell lines were stimulated with TGF-beta1 for 72 h, and then, in the presence of TGF-beta1, were cultured with FGF-1 plus heparin for an additional 48 h. After TGF-beta1 treatment, epithelial cells acquired a spindle-like mesenchymal phenotype with a substantial reduction of E-cadherin and cytokeratins and concurrent induction of alpha-smooth muscle actin measured by real-time PCR, Western blotting, and immunocytochemistry. FGF-1 plus heparin reversed these morphological changes and returned the epithelial and mesenchymal markers to control levels. Signaling pathways analyzed by selective pharmacological inhibitors showed that TGF-beta1 induces EMT through Smad pathway, while reversion by FGF-1 occurs through MAPK/ERK kinase pathway, resulting in ERK-1 phosphorylation and Smad2 dephosphorylation. These findings indicate that TGF-beta1-induced EMT is reversed by FGF-1 and suggest therapeutic approaches to target this process in IPF.


Subject(s)
Cell Transdifferentiation/drug effects , Epithelial Cells/physiology , Fibroblast Growth Factor 1/pharmacology , Mesoderm/physiology , Transforming Growth Factor beta1/pharmacology , Animals , Cells, Cultured , Epithelial Cells/drug effects , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/pathology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mesoderm/drug effects , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase Kinases/physiology , Phosphorylation , Rats , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors
18.
Respir Med ; 104(6): 889-94, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20153157

ABSTRACT

Hypersensitivity Pneumonitis (HP) is a lung inflammatory disorder caused by inhalation of organic particles by a susceptible host. However, only a small proportion of individuals exposed to HP-associated antigens develop the disease, suggesting that additional host/environmental factors may play a role. We have previously found that genetic susceptibility associated to the major histocompatibility complex (MHC) plays an important role in this disease. The low molecular weight proteosome (LMP, currently named PSMB) genes code for subunits of the proteosome, a multimeric enzymatic complex that degrades proteins into peptides in order to be presented in the MHC class I pathway. We hypothesized that polymorphisms in PSMB8 or PSMB9 genes could be involved in the susceptibility to HP. Thus, in this study we analyzed the polymorphic site at amino acid position 60 (Arg/His) of the fourth exon in the PSMB9 gene and the amino acid position 49 (Gln/Lys) in the second exon of PSMB8 gene in 50 Mexican patients with HP and 50 healthy ethnically matched controls. PSMB typing was performed using polymerase chain reaction-restriction fragment length polymorphisms (PCR-RFLP). Our results demonstrated that HP patients had a significant increase of the PSMB8 KQ genotype frequency (OR = 7.25, CI = 2.61-21.3; p = 0.000034). No differences were found in the distribution of PSMB9 alleles/genotypes. However, PSMB9-RH/PSMB8 KQ haplotype was significantly increased in HP patients (OR = 6.77, CI = 1.34-65.31, p < 0.02). These findings suggest that PSMB8 KQ genotype could increase the risk to develop hypersensitivity pneumonitis.


Subject(s)
Alveolitis, Extrinsic Allergic/genetics , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Haplotypes/genetics , Polymorphism, Genetic/genetics , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/physiopathology , Female , Genotype , Humans , Male , Mexico
19.
Rev. Inst. Nac. Enfermedades Respir ; 18(3): 187-194, jul.-sep. 2005. ilus, tab
Article in Spanish | LILACS | ID: lil-632563

ABSTRACT

Introducción: La fibrosis pulmonar idiopática (FPI) es una neumopatía crónica de difícil diagnóstico que puede confundirse con otras enfermedades pulmonares intersticiales difusas (EPID), por lo que es importante buscar nuevos métodos auxiliares para su diagnóstico. La osteopontina (OPN) es una glicoproteína multifuncional con efecto estimulante sobre los fibroblastos y la síntesis de matriz extracelular. Dado su posible papel profibrosante, consideramos de interés evaluar su concentración en el lavado bronquioalveolar (LBA) de pacientes con FPI y compararla con otras EPID. Material y métodos: La OPN se cuantificó por ELISA en 25 pacientes con FPI, 10 con neumonía intersticial no específica (NINE), 16 con neumonitis por hipersensibilidad (NH) y 10 controles sanos. Resultados: La concentración de OPN en pacientes con FPI fue significativamente mayor que en pacientes con NH y sanos (p < 0.0001) así como en pacientes con NINE (p < 0.05). Esta prueba mostró una sensibilidad de 80% y especificidad de 75% para el diagnóstico de FPI cuando el logaritmo de la concentración de OPN es de > 2.0. No hubo correlación entre los niveles de OPN con las pruebas de función pulmonar. Es interesante observar una asociación significativa entre los niveles de OPN y la presencia de hipocratismo digital (RM: 6.69; IC 1.25-10.76; p < 0.05). Conclusiones: La OPN se encuentra elevada en el LBA de pacientes con FPI por lo que podría considerarse como una prueba auxiliar en el diagnóstico diferencial de las EPID. Adicionalmente, nuestros hallazgos sugieren que la OPN puede estar implicada en la patogénesis del hipocratismo digital.


Background: Idiopathic pulmonary fibrosis (IPF) is a chronic pulmonary disease of difficult diagnosis that could be confused with other interstitial lung diseases (ILD). In this context, it is important to seek new methods which can be helpful in the diagnosis of this disease. Osteopontin (OPN) is a multifunctional glycopro-tein that has a stimulating effect over fibroblasts and extracellular matrix accumulation. Because it has a possible pro-fibrosing role, we decided to measure its concentration in bronchioalveolar lavage (BAL) from patients with IPF and to compare it with other ILDs. Methods: OPN concentration was measured by ELISA method in 25 IPF patients, 10 patients with non-specific interstitial pneumonia (NSIP), 16 patients with hypersensitivity pneumonitis (HP) and 10 healthy controls. Results: OPN levels in IPF patients were significantly increased compared with patients with HP and healthy controls (p < 0.0001) as with NSIP patients (p< 0.05). This test showed an 80% sensitivity and 75% specificity when the OPN concentration logarithm is > 2.0. There was no correlation between OPN levels and pulmonary function tests. Interestingly, we observed a significant association between OPN levels and finger clubbing (OR: 6.69; Cl: 1.25-10.76; p < 0.05). Conclusions: IPF patients showed increased OPN levels; this test might be considered as an ancillary tool in the differential diagnosis of ILD. Additionally, our findings suggest that OPN could be implicated in the pathogenesis of clubbing.

SELECTION OF CITATIONS
SEARCH DETAIL
...