Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Respir Physiol Neurobiol ; 311: 104034, 2023 05.
Article in English | MEDLINE | ID: mdl-36792043

ABSTRACT

NaV channels expression have been reported in upper airways and tracheal smooth muscle cells controlling the generation and propagation of action potentials in the respiratory tract sensory neurons, but information about the presence of these proteins in the bronchioalveolar structures in human lungs was missing. The main objective covered in this work was to determine whether the NaV1.7 channels are expressed in lower airways, and to identify the cellular identities expressing these proteins. We detected high levels of the mRNA coding for NaV1.7 channels in isolated lung fibroblasts obtained from both normal lungs, and fibrotic lungs of patients with respiratory diseases. The protein was detected with two different antibodies in the bronchioalveolar tissue, alveolar endothelium, and capillary endothelium, in normal and pathologic lungs. These evidences are useful in the dissection of molecular mechanisms of pulmonary pathologies, and lead to consider the NaV1.7 channels as potential therapeutic targets for the treatment of pulmonary diseases.


Subject(s)
Lung , Sensory Receptor Cells , Humans , Action Potentials/physiology , Trachea
2.
Cells ; 11(18)2022 09 13.
Article in English | MEDLINE | ID: mdl-36139424

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant activation of the alveolar epithelium, the expansion of the fibroblast population, and the accumulation of extracellular matrix. Global gene expression of human lung fibroblasts stimulated with TGFß-1, a strong fibrotic mediator revealed the overexpression of ZNF365, a zinc finger protein implicated in cell cycle control and telomere stabilization. We evaluated the expression and localization of ZNF365 in IPF lungs and in the fibrotic response induced by bleomycin in WT and deficient mice of the orthologous gene Zfp365. In IPF, ZNF365 was overexpressed and localized in fibroblasts/myofibroblasts and alveolar epithelium. Bleomycin-induced lung fibrosis showed an upregulation of Zfp365 localized in lung epithelium and stromal cell populations. Zfp365 KO mice developed a significantly higher fibrotic response compared with WT mice by morphology and hydroxyproline content. Silencing ZNF365 in human lung fibroblasts and alveolar epithelial cells induced a significant reduction of growth rate and increased senescence markers, including Senescence Associated ß Galactosidase activity, p53, p21, and the histone variant γH2AX. Our findings demonstrate that ZNF365 is upregulated in IPF and experimental lung fibrosis and suggest a protective role since its absence increases experimental lung fibrosis mechanistically associated with the induction of cell senescence.


Subject(s)
DNA-Binding Proteins , Idiopathic Pulmonary Fibrosis , Transcription Factors , Animals , Bleomycin/toxicity , Cellular Senescence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibrosis , Histones , Humans , Hydroxyproline , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53 , beta-Galactosidase/metabolism
3.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012303

ABSTRACT

Double-stranded RNA adenosine deaminase 1 (ADAR1) is significantly down-regulated in fibroblasts derived from Idiopathic Pulmonary Fibrosis (IPF) patients, and its overexpression restored levels of miRNA-21, PELI1, and SPRY2. There are two ADAR1 isoforms in humans, ADAR1-p110 and ADAR1-p150, generated by an alternative promoter. Let-7d is considered an essential microRNA in Pulmonary Fibrosis (PF). In silico analysis revealed COL3A1 and SMAD2, proteins involved in the development of IPF, as Let-7d targets. We analyzed the role of ADAR1-p110 and ADAR1-p150 isoforms in the regulation of Let-7d maturation and the effect of this regulation on the expression of COL3A1 and SMAD2 in IPF fibroblast. We demonstrated that differential expression and subcellular distribution of ADAR1 isoforms in fibroblasts contribute to the up-regulation of pri-miR-Let-7d and down-regulation of mature Let-7d. Induction of overexpression of ADAR1 reestablishes the expression of pri-miR-Let-7d and Let-7d in lung fibroblasts. The reduction of mature Let-7d upregulates the expression of COL3A1 and SMAD2. Thus, ADAR1 isoforms and Let-7d could have a synergistic role in IPF, which is a promising explanation in the mechanisms of fibrosis development, and the regulation of both molecules could be used as a therapeutic approach in IPF.


Subject(s)
Adenosine Deaminase , Idiopathic Pulmonary Fibrosis , MicroRNAs , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins
4.
Front Immunol ; 13: 820347, 2022.
Article in English | MEDLINE | ID: mdl-35222396

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible lung disease of unknown etiology. Myofibroblasts are organized in peculiar subepithelial fibroblasts foci (FF), where they abnormally persist and exclude lymphocytes by unclear mechanisms. FF are the source of an excessive extracellular matrix, which results in progressive stiffening and destruction of the lung architecture. We hypothesized that the absence of T cells inside the FF could be related, at least partially, to an inefficient function of lymphocytes induced by IPF fibroblasts. Here, we evaluated the effect of a supernatant from IPF fibroblasts on T-cell apoptosis and migration capacity. Data showed that IPF fibroblasts secrete pro-apoptotic molecules (both from extrinsic and intrinsic pathways), generating a microenvironment that induces apoptosis of T cells at 3 h of culture, despite a weak anti-apoptotic profile exhibited by these T cells. At 24 h of culture, the supernatants from both IPF and control fibroblasts provoked T-cell death. However, at this time of culture, IPF fibroblasts caused a marked decrease in T-cell migration; in contrast, control lung fibroblasts induced an increase of T-cell migration. The reduction of T-cell migratory capacity provoked by IPF fibroblasts was associated with a negative regulation of RHOA and ROCK, two essential GTPases for migration, and was independent of the expression of chemokine receptors. In conclusion, our findings demonstrate that IPF fibroblasts/myofibroblasts induce apoptosis and affect T-cell migration, revealing a mechanism involved in the virtual absence of T lymphocytes inside the FF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Apoptosis , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Myofibroblasts/metabolism , T-Lymphocytes/metabolism
5.
Toxics ; 9(9)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34564378

ABSTRACT

Exposure to air pollutants in wildfire smoke and indoor pollution causes lung diseases. Short-term exposure to wood smoke (WS) is partially known to alter the expression of human matrix metalloproteinases (MMPs), inflammatory cytokines, and tissue inhibitors of metalloproteinases (TIMPs). Accordingly, we investigated the effect of exposing guinea pigs to WS for two and four three-hour periods on different days. The daily content of particles reported by indoor pollution was produced by 60 g of pinewood. We analyzed the cell profile and collagen content in bronchoalveolar lavages (BAL). The mRNA expression of pro-inflammatory cytokines, MMPs, and TIMPs was studied in lung tissue. Cytokines and gelatinolytic activity were analyzed in BAL and serum. The results showed that total cells, macrophages, neutrophils, and collagen increased in BAL, whereas neutrophils and lymphocytes decreased. TGF-ß1, TNF-α, IFN-γ, IL-1ß, IL-6, IL-8, MMP-2, MMP-9, TIMP-1, and TIMP-2 were upregulated in lungs, downregulating IL-12. TNF-α, IFN-γ, TGF-ß1, IL-1ß, IL-6, and IL-8 were increased in BAL and serum, decreasing IL-12. Gelatinase activity was increased in serum. Thus, guinea pigs exposed to short-term domestic doses of WS overexpressed pro-inflammatory cytokines, MMPs, and TIMPs. These results are similar to ECM remodeling and pulmonary and systemic inflammation reported in humans.

6.
Int J Toxicol ; 40(6): 506-516, 2021 12.
Article in English | MEDLINE | ID: mdl-34530646

ABSTRACT

Wood smoke (WS) contains many harmful compounds, including polycyclic aromatic hydrocarbons (PAHs). WS induces inflammation in the airways and lungs and can lead to the development of various acute and chronic respiratory diseases. Pulmonary fibroblasts are the main cells involved in the remodeling of the extracellular matrix (ECM) during the WS-induced inflammatory response. Although fibroblasts remain in a low proliferation state under physiological conditions, they actively participate in ECM remodeling during the inflammatory response in pathophysiological states. Consequently, we used normal human lung fibroblasts (NHLFs) to assess the potential effects of the PAHs-containing wood smoke extract (WSE) on the growth rate, total collagen synthesis, and the expression levels of collagen I and III, matrix metalloproteinase (MMP)-1, MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, and the transforming growth factor (TGF)-ß1. We also assessed MMPs activity. The results showed that WSE induced a trimodal behavior in the growth rate curves in NHLFs; the growth rate increased with 0.5-1 % WSE and decreased with 2.5% WSE, without causing cell damage; 5-20% WSE inhibited the growth and induced cell damage. After 3 hours of exposure, 2.5% WSE induced an increase in total collagen synthesis and upregulation of TGF-ß1, collagen I and III, MMP-1, TIMP-1, and TIMP-2 expression. However, MMP-2 expression was downregulated and MMP-9 was not expressed. The gelatinase activity of MMP-2 was also increased. These results suggest that WSE affects the ECM remodeling in NHLFs and indicate the potential involvement of PAHs in this process.


Subject(s)
Extracellular Matrix/drug effects , Fibroblasts/drug effects , Inflammation/chemically induced , Inflammation/physiopathology , Lung Diseases/chemically induced , Plant Extracts/adverse effects , Smoke/adverse effects , Cell Proliferation/drug effects , Humans , Magnoliopsida/chemistry , Wood/chemistry
7.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360637

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by increased activation of fibroblasts/myofibroblasts. Previous reports have shown that IPF fibroblasts are resistant to apoptosis, but the mechanisms remain unclear. Since inhibition of the mitochondrial permeability transition pore (mPTP) has been implicated in the resistance to apoptosis, in this study, we analyzed the role of mitochondrial function and the mPTP on the apoptosis resistance of IPF fibroblasts under basal conditions and after mitomycin C-induced apoptosis. We measured the release of cytochrome c, mPTP opening, mitochondrial calcium release, oxygen consumption, mitochondrial membrane potential, ADP/ATP ratio, ATP concentration, and mitochondrial morphology. We found that IPF fibroblasts were resistant to mitomycin C-induced apoptosis and that calcium, a well-established activator of mPTP, is decreased as well as the release of pro-apoptotic proteins such as cytochrome c. Likewise, IPF fibroblasts showed decreased mitochondrial function, while mPTP was less sensitive to ionomycin-induced opening. Although IPF fibroblasts did not present changes in the mitochondrial membrane potential, we found a fragmented mitochondrial network with scarce, thinned, and disordered mitochondria with reduced ATP levels. Our findings demonstrate that IPF fibroblasts are resistant to mitomycin C-induced apoptosis and that altered mPTP opening contributes to this resistance. In addition, IPF fibroblasts show mitochondrial dysfunction evidenced by a decrease in respiratory parameters.


Subject(s)
Apoptosis , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Adenosine Triphosphate/metabolism , Calcium/metabolism , Cytochromes c/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/pathology , Ionomycin , Mitochondria/pathology , Mitomycin , Oxygen/metabolism , Primary Cell Culture
8.
Biomolecules ; 11(3)2021 03 04.
Article in English | MEDLINE | ID: mdl-33806618

ABSTRACT

. In passages above ten and growing very actively, we observed that some human lung fibroblasts cultured under standard conditions were transformed into a lineage of epithelial-like cells (ELC). To systematically evaluate the possible mesenchymal-epithelial transition (MET) occurrence, fibroblasts were obtained from normal lungs and also from lungs affected by idiopathic interstitial diseases. When an unusual epithelial-like phenotypic change was observed, cultured cells were characterized by confocal immunofluorescence microscopy, immunoblotting, immunocytochemistry, cytofluorometry, gelatin zymography, RT-qPCR, and hybridization in a whole-transcript human microarray. Additionally, microvesicles fraction (MVs) from ELC and fibroblasts were used to induce MET, while the microRNAs (miRNAs) contained in the MVs were identified. Pattern-gene expression of the original fibroblasts and the derived ELC revealed profound changes, upregulating characteristic epithelial-cell genes and downregulating mesenchymal genes, with a marked increase of E-cadherin, cytokeratin, and ZO-1, and the loss of expression of α-SMA, collagen type I, and Thy-1 cell surface antigen (CD90). Fibroblasts, exposed to culture media or MVs from the ELC, acquired ELC phenotype. The miRNAs in MVs shown six expressed exclusively in fibroblasts, and three only in ELC; moreover, twelve miRNAs were differentially expressed between fibroblasts and ELC, all of them but one was overexpressed in fibroblasts. These findings suggest that the MET-like process can occur in human lung fibroblasts, either from normal or diseased lungs. However, the biological implication is unclear.


Subject(s)
Epithelial-Mesenchymal Transition , Fibroblasts/pathology , Lung Diseases, Interstitial/pathology , Lung/pathology , Actins/metabolism , Aged , Biomarkers/metabolism , Cell-Derived Microparticles/metabolism , Cells, Cultured , Cluster Analysis , Collagen Type I/metabolism , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/metabolism , Humans , Lung Diseases, Interstitial/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phenotype , Thy-1 Antigens/metabolism
9.
Eur Respir J ; 58(2)2021 08.
Article in English | MEDLINE | ID: mdl-33446609

ABSTRACT

BACKGROUND: Around 8-10% of individuals over 50 years of age present interstitial lung abnormalities (ILAs), but their risk factors are uncertain. METHODS: From 817 individuals recruited in our lung ageing programme at the Mexican National Institute of Respiratory Diseases, 80 (9.7%) showed ILAs and were compared with 564 individuals of the same cohort with normal high-resolution computed tomography to evaluate demographic and functional differences, and with 80 individuals randomly selected from the same cohort for biomarkers. We evaluated MUC5B variant rs35705950, telomere length, and serum levels of matrix metalloproteinase (MMP)-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-12, MMP-13, interleukin (IL)-6, surfactant protein (SP)-D, α-Klotho and resistin. RESULTS: Individuals with ILAs were usually males (p<0.005), older than controls (p<0.0001), smokers (p=0.01), with a greater frequency of MUC5B rs35705950 (OR 3.5, 95% CI 1.3-9.4; p=0.01), and reduced diffusing capacity of the lung for carbon monoxide and oxygen saturation. Resistin, IL-6, SP-D, MMP-1, MMP-7 and MMP-13 were significantly increased in individuals with ILAs. Resistin (12±5 versus 9±4 ng·mL-1; p=0.0005) and MMP-13 (357±143 versus 298±116 pg·mL-1; p=0.004) were the most increased biomarkers. On follow-up (24±18 months), 18 individuals showed progression which was associated with gastro-oesophageal reflux disease (OR 4.1, 95% CI 1.2-12.9; p=0.02) and in females with diabetes mellitus (OR 5.3, 95% CI 1.0-27.4; p=0.01). CONCLUSIONS: Around 10% of respiratory asymptomatic individuals enrolled in our lung ageing programme show ILAs. Increased serum concentrations of pro-inflammatory molecules and MMPs are associated with ILAs.


Subject(s)
Lung Diseases, Interstitial , Female , Humans , Lung/diagnostic imaging , Male , Matrix Metalloproteinase 7 , Mucin-5B , Risk Factors
10.
Int J Med Sci ; 17(15): 2357-2361, 2020.
Article in English | MEDLINE | ID: mdl-32922201

ABSTRACT

Idiopathic pulmonary fibrosis is a chronic and progressive disease of unknown cause. It is characterized by the aberrant activation of the bronchioalveolar epithelium, the formation of fibroblast foci and the excessive production extracellular matrix. The cellular and molecular mechanisms that contribute to the pathobiology of the disease are unclear. The CX3CL1-CX3CR1 axis regulates cellular responses that are known to be relevant in IPF, such as proliferation and collagen production. In this study, we characterize for the first time the expression of CX3CL1 and its receptor in lung tissue from patients with IPF; and its effect on collagen production in IPF fibroblasts. We found that CX3CL1-CX3CR1 axis has a modified expression in the lung tissue, importantly this axis is expressed on fibroblasts, and CX3CL1 decreased the collagen production in pulmonary fibroblasts derived from IPF patients.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Collagen/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , CX3C Chemokine Receptor 1/analysis , Cell Line , Cell Proliferation , Chemokine CX3CL1/analysis , Collagen/analysis , Extracellular Matrix/pathology , Fibroblasts/metabolism , Humans , Immunohistochemistry , Lung/cytology , Primary Cell Culture , Signal Transduction
11.
Lung ; 196(4): 393-400, 2018 08.
Article in English | MEDLINE | ID: mdl-29637273

ABSTRACT

INTRODUCTION: microRNAs (miRNAs) are small non-coding 1RNAs that post-transcriptionally regulate gene expression. Recent evidence shows that adenosine deaminases that act on RNA (ADAR) can edit miRNAs. miRNAs are involved in the development of different diseases, such as idiopathic pulmonary fibrosis (IPF). In IPF, about 40% of the miRNAs are differentially expressed with respect to controls. Among these miRNAs, miRNA-21 has been found over-expressed in IPF and its targets are anti-fibrosing molecules such as PELI1 and SPRY2. The objective of this study is to determine the role of ADAR1 and 2 on the expression of miRNA-21 in human lung fibroblasts trough quantification of gene expression, protein levels, and overexpression of ADAR1 and 2. METHODS: Six control and six fibrotic primary fibroblast cell cultures were used for RNA extraction, ADAR1, ADAR2, PELI1, SPRY2, miRNA-21, and pri-miRNA-21 expression was measured. Subsequently, two fibrotic fibroblast cultures were used for overexpression of ADAR1 and ADAR2, and they were stimulated with TGFß1. Real-time PCR and Western blot were performed. RESULTS: ADAR1 is significantly downregulated in IPF fibroblasts; the overexpression of ADAR1 and ADAR2 reestablishes the expression levels of miRNA-21, PELI1, and SPRY2 in fibroblasts of patients with IPF. CONCLUSION: These changes in the processing of miRNAs have great value in pathology diagnosis, including lung diseases, and play an important role in the understanding of molecular mechanisms involved in the development of different pathologies, as well as representing new therapeutic targets.


Subject(s)
Adenosine Deaminase/metabolism , Fibroblasts/enzymology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Case-Control Studies , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation, Enzymologic , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/drug effects , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Primary Cell Culture , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Transforming Growth Factor beta1/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
PLoS One ; 13(3): e0192963, 2018.
Article in English | MEDLINE | ID: mdl-29529050

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by epithelial cell activation, expansion of the fibroblast population and excessive extracellular matrix accumulation. The mechanisms are incompletely understood but evidence indicates that the deregulation of several proteases contributes to its pathogenesis. Transmembrane protease serine 4 (TMPRSS4) is a novel type II transmembrane serine protease that may promote migration and facilitate epithelial to mesenchymal transition (EMT), two critical processes in the pathogenesis of IPF. Thus, we hypothesized that over-expression of TMPRSS4 in the lung could promote the initiation and/or progression of IPF. In this study we first evaluated the expression and localization of TMPRSS4 in IPF lungs by real time PCR, western blot and immunohistochemistry. Then we examined the lung fibrotic response in wild-type and TMPRSS4 deficient mice using the bleomycin-induced lung injury model. We found that this protease is upregulated in IPF lungs, where was primarily expressed by epithelial and mast cells. Paralleling the findings in vivo, TMPRSS4 was expressed by alveolar and bronchial epithelial cells in vitro and unexpectedly, provoked an increase of E-cadherin. No expression was observed in normal human or IPF lung fibroblasts. The lung fibrotic response evaluated at 28 days after bleomycin injury was markedly attenuated in the haplodeficient and deficient TMPRSS4 mice. By morphology, a significant reduction of the fibrotic index was observed in KO and heterozygous mice which was confirmed by measurement of collagen content (hydroxyproline: WT: 164±21.1 µg/lung versus TMPRSS4 haploinsufficient: 110.2±14.3 µg/lung and TMPRSS4 deficient mice: 114.1±24.2 µg/lung (p<0.01). As in IPF, TMPRSS4 was also expressed in epithelial and mast cells. These findings indicate that TMPRSS4 is upregulated in IPF lungs and that may have a profibrotic role.


Subject(s)
Idiopathic Pulmonary Fibrosis/genetics , Lung Injury/genetics , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Up-Regulation , Animals , Bleomycin , Cell Line , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Serine Endopeptidases/analysis , Serine Endopeptidases/metabolism
13.
Am J Respir Cell Mol Biol ; 59(1): 65-76, 2018 07.
Article in English | MEDLINE | ID: mdl-29345973

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by the expansion of the myofibroblast population, excessive extracellular matrix accumulation, and destruction of the lung parenchyma. The R-spondin family (RSPO) comprises a group of proteins essential for development. Among them, RSPO2 is expressed primarily in the lungs, and its mutations cause severe defects in the respiratory tract. Interestingly, RSPO2 participates in the canonical Wingless/int1 pathway, a critical route in the pathogenesis of IPF. Thus, the aim of this study was to examine the expression and putative role of RSPO2 in this disease. We found that RSPO2 and its receptor leucine-rich G protein-coupled receptor 6 were upregulated in IPF lungs, where they localized primarily in fibroblasts and epithelial cells. Stimulation of IPF and normal lung fibroblasts with recombinant human RSPO2 resulted in the deregulation of numerous genes, although the transcriptional response was essentially distinct. In IPF fibroblasts, RSPO2 stimulation induced the up- or downregulation of several genes involved in the Wingless/int1 pathway (mainly from noncanonical signaling). In both normal and IPF fibroblasts, RSPO2 modifies the expression of genes implicated in several pathways, including the cell cycle and apoptosis. In accordance with gene expression, the stimulation of normal and IPF fibroblasts with RSPO2 significantly reduced cell proliferation and induced cell death. RSPO2 also inhibited collagen production and increased the expression of matrix metalloproteinase 1. Silencing RSPO2 with shRNA induced the opposite effects. Our findings demonstrate, for the first time to our knowledge, that RSPO2 is upregulated in IPF, where it appears to have an antifibrotic role.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Intercellular Signaling Peptides and Proteins/genetics , Up-Regulation/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Collagen/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Gene Silencing , Genome, Human , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lung/metabolism , Lung/pathology , Matrix Metalloproteinase 1/metabolism , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/pharmacology , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
14.
J Pathol ; 240(2): 197-210, 2016 10.
Article in English | MEDLINE | ID: mdl-27425145

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibroblast and myofibroblast proliferation, and extensive deposition of extracellular matrix (ECM). Fibroblast growth factor-1 (FGF-1) belongs to the FGF family and has been shown to inhibit fibroblast collagen production and differentiation into myofibroblasts, and revert epithelial-mesenchymal transition by inhibiting TGF-ß1 signalling pathways. However, the precise role of FGF-1 in pulmonary fibrosis has not yet been elucidated. In this study, we explore the mechanisms underlying the anti-fibrogenic effect of FGF-1 in pulmonary fibrosis in vitro and in vivo by prolonged transient overexpression of FGF-1 (AdFGF-1) and TGF-ß1 (AdTGF-ß1) using adenoviral vectors. In vivo, FGF-1 overexpression markedly attenuated TGF-ß1-induced pulmonary fibrosis in rat lungs when given both concomitantly, or delayed, by enhancing proliferation and hyperplasia of alveolar epithelial cells (AECs). AdFGF-1 also attenuated the TGF-ß1 signalling pathway and induced FGFR1 expression in AECs. In vitro, AdFGF-1 prevented the increase in α-SMA and the decrease in E-cadherin induced by AdTGF-ß1 in normal human lung fibroblasts, primary human pulmonary AECs, and A549 cells. Concomitantly, AdTGF-ß1-induced Smad2 phosphorylation was significantly reduced by AdFGF-1 in both cell types. AdFGF-1 also attenuated the increase in TGFßR1 protein and mRNA levels in fibroblasts. In AECs, AdFGF-1 decreased TGFßR1 protein by favouring TGFßR1 degradation through the caveolin-1/proteasome pathway. Furthermore, FGFR1 expression was increased in AECs, whereas it was decreased in fibroblasts. In serum of IPF patients, FGF-1 levels were increased compared to controls. Interestingly, FGF-1 expression was restricted to areas of AEC hyperplasia, but not α-SMA-positive areas in IPF lung tissue. Our results demonstrate that FGF-1 may have preventative and therapeutic effects on TGF-ß1-driven pulmonary fibrosis via inhibiting myofibroblast differentiation, inducing AEC proliferation, regulating TGF-ß1 signalling by controlling TGFßR1 expression and degradation, and regulating FGFR1 expression. Thus, modulating FGF-1 signalling represents a potential therapy for the treatment of pulmonary fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Epithelial Cells/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblasts/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/genetics , Animals , Cadherins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Epithelial Cells/pathology , Female , Fibroblast Growth Factor 1/metabolism , Fibroblasts/pathology , Humans , Phosphorylation , Proteasome Endopeptidase Complex , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Smad2 Protein/metabolism , Up-Regulation
15.
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
16.
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
17.
Lung ; 193(1): 25-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25537934

ABSTRACT

INTRODUCTION: Surfactant protein D (SP-D) plays an important role in the innate responses against pathogens and its production is altered in lung disorders. METHODS: We studied the circulating levels of SP-D in 37 patients with acute respiratory distress syndrome due to the A/H1N1 virus infection and in 40 healthy controls. Cox logistic regression models were constructed to explore the association of SP-D levels and risk of death. RESULTS: Mortality rate after a 28-day was 32.42 %. Significant higher levels of SP-D were detected in A/H1N1 patients with fatal outcome (p < 0.05). After adjusting for confounding variables, levels of SP-D ≥250 ng/mL were associated with increased the risk of death (HR = 8.27, 95 % CI 1.1-64.1, p = 0.043). CONCLUSIONS: Our results revealed that higher circulating levels of SP-D are associated with higher mortality risk in critically ill A/H1N1 patients. SP-D might be a predictive factor of poor outcomes in viral pneumonia.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/diagnosis , Pneumonia, Viral/diagnosis , Pulmonary Surfactant-Associated Protein D/blood , Respiratory Distress Syndrome/diagnosis , Adult , Biomarkers/blood , Case-Control Studies , Chi-Square Distribution , Critical Illness , Female , Hospital Mortality , Humans , Influenza, Human/blood , Influenza, Human/mortality , Influenza, Human/therapy , Influenza, Human/virology , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Multivariate Analysis , Pneumonia, Viral/blood , Pneumonia, Viral/mortality , Pneumonia, Viral/therapy , Pneumonia, Viral/virology , Prognosis , Proportional Hazards Models , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/virology , Risk Factors , Time Factors , Up-Regulation
18.
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
19.
Respir Res ; 15: 74, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980707

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) and C-reactive protein (CRP) are involved in chronic obstructive pulmonary disease (COPD) pathogenesis. The aim of the present work was to determine plasma concentrations of MMPs and CRP in COPD associated to biomass combustion exposure (BE) and tobacco smoking (TS). METHODS: Pulmonary function tests, plasma levels of MMP-1, MMP-7, MMP-9, MMP-9/TIMP-1 and CRP were measured in COPD associated to BE (n = 40) and TS (n =40) patients, and healthy non-smoking (NS) healthy women (controls, n = 40). RESULTS: Plasma levels of MMP-1, MMP-7, MMP-9, and MMP-9/TIMP-1 and CRP were higher in BE and TS than in the NS healthy women (p <0.01). An inverse correlation between MMP-1, MMP-7, MMP-9, MMP-9/TIMP-1 and CRP plasma concentrations and FEV1 was observed. CONCLUSIONS: Increase of MMPs and CRP plasma concentrations in BE suggests a systemic inflammatory phenomenon similar to that observed in COPD associated to tobacco smoking, which may also play a role in COPD pathogenesis.


Subject(s)
C-Reactive Protein/metabolism , Forced Expiratory Volume/physiology , Matrix Metalloproteinase 1/blood , Matrix Metalloproteinase 7/blood , Matrix Metalloproteinase 9/blood , Pulmonary Disease, Chronic Obstructive/blood , Smoking/blood , Aged , Aged, 80 and over , Biomarkers/blood , Biomass , Female , Fuel Oils/adverse effects , Humans , Middle Aged , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/epidemiology , Smoke/adverse effects , Smoking/adverse effects , Smoking/epidemiology
20.
Eur Respir J ; 42(5): 1309-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23143540

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an ageing-related lung disorder characterised by expansion of the myofibroblast population and aberrant lung remodelling. Dehydroepiandrosterone (DHEA), a steroid pro-hormone, decreases with age but an exaggerated decline has been associated with chronic degenerative diseases. We quantified the plasma levels of DHEA and its sulfated form (DHEA-S) in 137 IPF patients and 58 controls and examined the effects of DHEA on human lung fibroblasts. Plasma DHEA/DHEA-S was significantly decreased in male IPF patients (median (range) DHEA: 4.4 (0.2-29.2) versus 6.7 (2.1-15.2) ng · mL(-1), p<0.01; DHEA-S: 47 (15.0-211) versus 85.2 (37.6-247.0) µg · dL(-1), p<0.001), while in females only DHEA-S was significantly decreased (32.6 (15.0-303.0) versus 68.3 (16.4-171) µg · dL(-1), p<0.001). DHEA caused a decrease in fibroblast proliferation and an approximately two-fold increase in fibroblast apoptosis, probably through the intrinsic pathway with activation of caspase-9. This effect was accompanied by upregulation of several pro-apoptotic proteins (Bax and cyclin-dependent kinase-inhibitor CDNK1A) and downregulation of anti-apoptotic proteins, such as cellular inhibitor of apoptosis (c-IAP)1 and c-IAP2. DHEA also caused a significant decrease of transforming growth factor-ß1-induced collagen production and fibroblast to myofibroblast differentiation, and inhibited platelet-derived growth factor-induced fibroblast migration. These findings demonstrate a disproportionate decrease of DHEA/DHEA-S in IPF patients and indicate that this molecule has multiple antifibrotic properties.


Subject(s)
Dehydroepiandrosterone/blood , Dehydroepiandrosterone/pharmacology , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Aged , Apoptosis , Bronchoalveolar Lavage , Case-Control Studies , Caspase 9/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Female , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Male , Middle Aged , Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...