Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 16(21): 2108-2118, 2017.
Article in English | MEDLINE | ID: mdl-28933587

ABSTRACT

It has become more and more evident that the BCL-2 family proteins mediate a wide range of non-apoptotic functions. The pro-apoptotic BAX protein has been reported in interphasic nuclei. Whether the nuclear form of BAX could be involved in non-apoptotic function is still unknown. Our study showed for the first time that BAX was associated with chromatin in vitro. Next, we used gain and loss of function approaches to decipher the potential role of nuclear BAX in non-apoptotic cells. In vitro, nuclear BAX promoted cell proliferation in lung epithelial cells and primary human lung fibroblasts by modulating CDKN1A expression. Interestingly, BAX occupancy of CDKN1A promoter was specifically enriched close to the transcription-starting site. Nuclear BAX also modulated the basal myofibroblastic differentiation and migration of primary human lung fibroblasts. Finally, BAX nuclear localization was associated in vivo with the remodelling of lung parenchyma during development, tumorigenesis as well as fibrosis compared to control adult human lungs. Hence, our study established for the first time, a strong link between the nuclear localization of the pro-apoptotic BAX protein and key basic cellular functions in the non-apoptotic setting.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Interphase , bcl-2-Associated X Protein/metabolism , Apoptosis/physiology , Cell Nucleus/metabolism , Fibroblasts/metabolism , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 313(5): L781-L795, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28729349

ABSTRACT

Fibroblast growth factor 9 (FGF9) is necessary for fetal lung development and is expressed by epithelium and mesothelium. We evaluated the role of FGF9 overexpression on adenoviral-induced pleural injury in vivo and determined the biological effects of FGF9 on mesothelial cells in vitro. We assessed the expression of FGF9 and FGF receptors by mesothelial cells in both human and mouse lungs. Intrapleural injection of an adenovirus expressing human FGF9 (AdFGF9) or a control adenovirus (AdCont) was performed. Mice were euthanized at days 3, 5, and 14 Expression of FGF9 and markers of inflammation and myofibroblastic differentiation was studied by qPCR and immunohistochemistry. In vitro, rat mesothelial cells were stimulated with FGF9 (20 ng/ml), and we assessed its effect on proliferation, survival, migration, and differentiation. FGF9 was expressed by mesothelial cells in human idiopathic pulmonary fibrosis. FGF receptors, mainly FGFR3, were expressed by mesothelial cells in vivo in humans and mice. AdCont instillation induced diffuse pleural thickening appearing at day 5, maximal at day 14 The altered pleura cells strongly expressed α-smooth muscle actin and collagen. AdFGF9 injection induced maximal FGF9 expression at day 5 that lasted until day 14 FGF9 overexpression prevented pleural thickening, collagen and fibronectin accumulation, and myofibroblastic differentiation of mesothelial cells. In vitro, FGF9 decreased mesothelial cell migration and inhibited the differentiating effect of transforming growth factor-ß1. We conclude that FGF9 has a potential antifibrotic effect on mesothelial cells.


Subject(s)
Adenoviridae/drug effects , Cell Movement/drug effects , Fibroblast Growth Factor 9/pharmacology , Idiopathic Pulmonary Fibrosis/virology , Lung/pathology , Animals , Cell Differentiation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelium/pathology , Epithelium/virology , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/virology , Mice, Inbred C57BL , Pleura/drug effects , Rats
3.
Respir Res ; 17: 36, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27044262

ABSTRACT

In human lung fibrotic lesions, fibroblasts were shown to be closely associated with immature dendritic cell (DC) accumulation. The aim of the present pilot study was to characterize the role of pulmonary fibroblasts on DC phenotype and function, using co-culture of lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) and from control patients, with a DC cell line MUTZ-3. We observed that co-culture of lung control and IPF fibroblasts with DCs reduced the expression of specific DC markers and down-regulated their T-cell stimulatory activity. This suggests that pulmonary fibroblasts might sustain chronic inflammation in the fibrotic lung by maintaining in situ a pool of immature DCs.


Subject(s)
Cell Communication/immunology , Dendritic Cells/cytology , Fibroblasts/cytology , Fibroblasts/immunology , Lung/cytology , Lung/immunology , Cell Proliferation/physiology , Cells, Cultured , Cytokines/immunology , Humans , In Vitro Techniques , Phenotype , Pilot Projects
4.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L615-29, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26773067

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by an accumulation of extracellular matrix proteins and fibroblasts in the distal airways. Key developmental lung signaling pathways are reactivated in IPF. For instance, fibroblast growth factor 9 (FGF9) and FGF18, involved in epithelial-mesenchymal interactions, are critical for lung development. We evaluated the expression of FGF9, FGF18, and FGF receptors (FGFRs) in lung tissue from controls and IPF patients and assessed their effect on proliferation, survival, migration, and differentiation of control and IPF human lung fibroblasts (HLFs). FGF9, FGF18, and all FGFRs were present in the remodeled alveolar epithelium close to the fibroblast foci in IPF lungs. FGFR3 was generally detected in fibroblast foci by immunohistochemistry. In vitro, HLFs mainly expressed mesenchyme-associated FGFR isoforms (FGFR1c and FGFR3c) and FGFR4. FGF9 did not affect fibroblast proliferation, whereas FGF18 inhibited cell growth in control fibroblasts. FGF9 and FGF18 decreased Fas-ligand-induced apoptosis in control but not in IPF fibroblasts. FGF9 prevented transforming growth factor ß1-induced myofibroblast differentiation. FGF9 and FGF18 increased the migratory capacities of HLF, and FGF9 actively modulated matrix metalloproteinase activity. In addition, FGFR3 inhibition by small interfering RNA impacted p-ERK activation by FGF9 and FGF18 and their effects on differentiation and migration. These results identify FGF9 as an antiapoptotic and promigratory growth factor on HLF, maintaining fibroblasts in an undifferentiated state. The biological effects of FGF9 and FGF18 were partially driven by FGFR3. FGF18 was a less potent molecule. Both growth factors likely contribute to the fibrotic process in vivo.


Subject(s)
Fibroblast Growth Factor 9/physiology , Fibroblast Growth Factors/physiology , Myofibroblasts/physiology , Receptors, Fibroblast Growth Factor/metabolism , Aged , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis , Lung/metabolism , Lung/pathology , Middle Aged
5.
J Immunol ; 195(6): 2788-96, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26268659

ABSTRACT

Epithelial to mesenchymal cell transition (EMT), whereby fully differentiated epithelial cells transition to a mesenchymal phenotype, has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). CXCR3 and its ligands are recognized to play a protective role in pulmonary fibrosis. In this study, we investigated the presence and extent of EMT and CXCR3 expression in human IPF surgical lung biopsies and assessed whether CXCR3 and its ligand CXCL9 modulate EMT in alveolar epithelial cells. Coexpression of the epithelial marker thyroid transcription factor-1 and the mesenchymal marker α-smooth muscle actin and CXCR3 expression was examined by immunohistochemical staining of IPF surgical lung biopsies. Epithelial and mesenchymal marker expression was examined by quantitative real-time PCR, Western blotting, and immunofluorescence in human alveolar epithelial (A549) cells treated with TGF-ß1 and CXCL9, with Smad2, Smad3, and Smad7 expression and cellular localization examined by Western blotting. We found that significantly more cells were undergoing EMT in fibrotic versus normal areas of lung in IPF surgical lung biopsy samples. CXCR3 was expressed by type II pneumocytes and fibroblasts in fibrotic areas in close proximity to cells undergoing EMT. In vitro, CXCL9 abrogated TGF-ß1-induced EMT. A decrease in TGF-ß1-induced phosphorylation of Smad2 and Smad3 occurred with CXCL9 treatment. This was associated with increased shuttling of Smad7 from the nucleus to the cytoplasm where it inhibits Smad phosphorylation. This suggests a role for EMT in the pathogenesis of IPF and provides a novel mechanism for the inhibitory effects of CXCL9 on TGF-ß1-induced EMT.


Subject(s)
Chemokine CXCL9/metabolism , Epithelial-Mesenchymal Transition/physiology , Idiopathic Pulmonary Fibrosis/pathology , Respiratory Mucosa/metabolism , Transforming Growth Factor beta1/metabolism , Actins/biosynthesis , Biomarkers/metabolism , Cell Line , Chemokine CXCL9/pharmacology , Epithelial Cells/metabolism , Humans , Nuclear Proteins/biosynthesis , Phosphorylation , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/metabolism , Respiratory Mucosa/cytology , Smad2 Protein/biosynthesis , Smad3 Protein/biosynthesis , Smad7 Protein/biosynthesis , Thyroid Nuclear Factor 1 , Transcription Factors/biosynthesis , Transforming Growth Factor beta1/pharmacology
6.
Am J Respir Cell Mol Biol ; 52(2): 244-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25032514

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by myofibroblast proliferation and extracellular-matrix accumulation. IPF typically starts in subpleural lung regions, and recent studies suggest that pleural mesothelial cells play a role in the onset of the disease. The transition of mesothelial cells into myofibroblasts (mesothelio-mesenchymal transition) is induced by the profibrotic cytokine, transforming growth factor (TGF)-ß1, and is thought to play a role in the development and progression of IPF. The Mothers Against Decapentaplegic homolog (Smad)-dependent pathway is the main TGF-ß1 pathway involved in fibrosis. αB-crystallin is constitutively expressed in the lungs, and is inducible by stress, acts as a chaperon, and is known to play a role in cell cytoskeleton architecture. We recently showed that the lack of αB-crystallin hampered TGF-ß1 signaling by favoring Smad4 monoubiquitination and nuclear export. We demonstrate here, for the first time, that αB-crystallin is strongly overexpressed in the pleura of fibrotic lungs from patients with IPF and in rodent models of pleural/subpleural fibrosis. αB-crystallin-deficient mice are protected from pleural/subpleural fibrosis induced by the transient adenoviral-mediated overexpression of TGF-ß1 or the intrapleural injection of bleomycin combined with carbon particles. We show that αB-crystallin inhibition hampers Smad4 nuclear localization in pleural mesothelial cells and the consequent characteristics of mesothelio-mesenchymal transition. αB-crystallin-deficient mesothelial cells fail to acquire the properties of myofibroblasts, thus limiting their migration in vivo and the progression of fibrosis in the lung parenchyma. In conclusion, our work demonstrates that αB-crystallin may be a key target for the development of specific drugs in the treatment of IPF.


Subject(s)
Bleomycin/pharmacology , Crystallins/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Myofibroblasts/drug effects , Pleura/drug effects , Animals , Cytoskeleton/drug effects , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Idiopathic Pulmonary Fibrosis/pathology , Mice , Mice, Knockout , Pleura/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
7.
Lab Invest ; 94(11): 1237-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199049

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic diffuse lung disease characterized by an accumulation of excess fibrous material in the lung. Protease nexin-1 (PN-1) is a tissue serpin produced by many cell types, including lung fibroblasts. PN-1 is capable of regulating proteases of both coagulation and fibrinolysis systems, by inhibiting, respectively, thrombin and plasminergic enzymes. PN-1 is thus a good candidate for regulating tissue remodeling occurring during IPF. We demonstrated a significant increase of PN-1 expression in lung tissue extracts, lung fibroblasts and bronchoalveolar lavage fluids of patients with IPF. The increase of PN-1 expression was reproduced after stimulation of control lung fibroblasts by transforming growth factor-ß, a major pro-fibrotic cytokine involved in IPF. Another serpin, plasminogen activator inhibitor-1 (PAI-1) is also overexpressed in fibrotic fibroblasts. Unlike PAI-1, cell-bound PN-1 as well as secreted PN-1 from IPF and stimulated fibroblasts were shown to inhibit efficiently thrombin activity, indicating that both serpins should exhibit complementary roles in IPF pathogenesis, via their different preferential antiprotease activities. Moreover, we observed that overexpression of PN-1 induced by transfection of control fibroblasts led to increased fibronectin expression, whereas PN-1 silencing induced in fibrotic fibroblasts led to decreased fibronectin expression. Overexpression of PN-1 lacking either its antiprotease activity or its binding capacity to glycosaminoglycans had no effect on fibronectin expression. These novel findings suggest that modulation of PN-1 expression in lung fibroblasts may also have a role in the development of IPF by directly influencing the expression of extracellular matrix proteins. Our data provide new insights into the role of PN-1 in the poorly understood pathological processes involved in IPF and could therefore give rise to new therapeutic approaches.


Subject(s)
Fibroblasts/enzymology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , Serpin E2/metabolism , Case-Control Studies , Fibronectins/metabolism , Humans , Thrombin/metabolism , Transforming Growth Factor beta/metabolism
8.
J Pathol ; 232(4): 458-72, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24307592

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by the proliferation of myofibroblasts and the accumulation of extracellular matrix (ECM) in the lungs. TGF-ß1 is the major profibrotic cytokine involved in IPF and is responsible for myofibroblast proliferation and differentiation and ECM synthesis. αB-crystallin is constitutively expressed in the lungs and is inducible by stress, acts as a chaperone and is known to play a role in cell cytoskeleton architecture homeostasis. The role of αB-crystallin in fibrogenesis remains unknown. The principal signalling pathway involved in this process is the Smad-dependent pathway. We demonstrate here that αB-crystallin is strongly expressed in fibrotic lung tissue from IPF patients and in vivo rodent models of pulmonary fibrosis. We also show that αB-crystallin-deficient mice are protected from bleomycin-induced fibrosis. Similar protection from fibrosis was observed in αB-crystallin KO mice after transient adenoviral-mediated over-expression of IL-1ß or TGF-ß1. We show in vitro in primary epithelial cells and fibroblasts that αB-crystallin increases the nuclear localization of Smad4, thereby enhancing the TGF-ß1-Smad pathway and the consequent activation of TGF-ß1 downstream genes. αB-crystallin over-expression disrupts Smad4 mono-ubiquitination by interacting with its E3-ubiquitin ligase, TIF1γ, thus limiting its nuclear export. Conversely, in the absence of αB-crystallin, TIF1γ can freely interact with Smad4. Consequently, Smad4 mono-ubiquitination and nuclear export are favoured and thus TGF-ß1-Smad4 pro-fibrotic activity is inhibited. This study demonstrates that αB-crystallin may be a key target for the development of specific drugs in the treatment of IPF or other fibrotic diseases.


Subject(s)
Cell Nucleus/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Smad4 Protein/metabolism , alpha-Crystallin B Chain/metabolism , Active Transport, Cell Nucleus , Animals , Bleomycin , Cell Nucleus/pathology , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/prevention & control , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung/pathology , Mice , Mice, 129 Strain , Mice, Knockout , RNA Interference , Rats, Sprague-Dawley , Transcription Factors/metabolism , Transfection , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , alpha-Crystallin B Chain/genetics
9.
PLoS One ; 8(1): e53736, 2013.
Article in English | MEDLINE | ID: mdl-23341987

ABSTRACT

BACKGROUND: Fibrocytes are circulating precursors for fibroblasts. Blood fibrocytes are increased in patients with idiopathic pulmonary fibrosis (IPF). The aim of this study was to determine whether alveolar fibrocytes are detected in broncho-alveolar lavage (BAL), to identify their prognostic value, and their potential association with culture of fibroblasts from BAL. METHODS: We quantified fibrocytes in BAL from 26 patients with IPF, 9 patients with Systemic Sclerosis(SSc)-interstitial lung disease (ILD), and 11 controls. BAL cells were cultured to isolate alveolar fibroblasts. RESULTS: Fibrocytes were detected in BAL in 14/26 IPF (54%) and 5/9 SSc patients (55%), and never in controls. Fibrocytes were in median 2.5% [0.4-19.7] and 3.0% [2.7-3.7] of BAL cells in IPF and SSc-ILD patients respectively. In IPF patients, the number of alveolar fibrocytes was correlated with the number of alveolar macrophages and was associated with a less severe disease but not with a better outcome. Fibroblasts were cultured from BAL in 12/26 IPF (46%), 5/9 SSc-ILD (65%) and never in controls. The detection of BAL fibrocytes did not predict a positive culture of fibroblasts. CONCLUSION: Fibrocytes were detected in BAL fluid in about half of the patients with IPF and SSc-ILD. Their number was associated with less severe disease in IPF patients and did not associate with the capacity to grow fibroblasts from BAL fluid.


Subject(s)
Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Pulmonary Alveoli/pathology , Scleroderma, Systemic/pathology , Aged , Aged, 80 and over , Bronchoalveolar Lavage , Cell Count , Female , Humans , Idiopathic Pulmonary Fibrosis/therapy , Male , Middle Aged , Scleroderma, Systemic/therapy , Time Factors
10.
FASEB J ; 27(4): 1549-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23288928

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by myofibroblast proliferation. Transition of epithelial/mesothelial cells into myofibroblasts [epithelial-to-mesenchymal transition (EMT)] occurs under the influence of transforming growth factor (TGF)-ß1, with Snail being a major transcription factor. We study here the role of the heat-shock protein HSP27 in fibrogenesis and EMT. In vitro, we have up- and down-modulated HSP27 expression in mesothelial and epithelial cell lines and studied the expression of different EMT markers induced by TGF-ß1. In vivo, we inhibited HSP27 with the antisense oligonucleotide OGX-427 (in phase II clinical trials as anticancer agent) in our rat subpleural/pulmonary fibrosis models. We demonstrate that HSP27 is strongly expressed during the fibrotic process in patients with IPF and in different in vivo models. We showed that HSP27 binds to and stabilizes Snail and consequently induces EMT. Conversely, HSP27 knockdown leads to Snail proteasomal degradation, thus inhibiting TGF-ß1-induced EMT. Inhibition of HSP27 with OGX-427 efficiently blocks EMT and fibrosis development. Controls in vivo were an empty adenovirus that did not induce fibrosis and a control antisense oligonucleotide. The present work opens the possibility of a new therapeutic use for HSP27 inhibitors against IPF, for which there is no conclusively effective treatment.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , HSP27 Heat-Shock Proteins/antagonists & inhibitors , Snails/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cadherins/metabolism , Cell Line , Epithelial Cells/metabolism , Fibrosis/metabolism , Humans , Oligonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Thionucleotides/pharmacology , Transcription Factors/metabolism
11.
Antioxid Redox Signal ; 18(1): 66-79, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22703534

ABSTRACT

AIMS: Oxidants have been implicated in the pathophysiology of idiopathic pulmonary fibrosis (IPF), especially in myofibroblastic differentiation. We aimed at testing the hypothesis that nuclear factor erythroid 2-related factor 2 (Nrf2), the main regulator of endogenous antioxidant enzymes, is involved in fibrogenesis via myofibroblastic differentiation. Fibroblasts were cultured from the lungs of eight controls and eight IPF patients. Oxidants-antioxidants balance, nuclear Nrf2 expression, and fibroblast phenotype (α-smooth muscle actin and collagen I expression, proliferation, migration, and contraction) were studied under basal conditions and after Nrf2 knockdown or activation by Nrf2 or Keap1 siRNA transfection. The effects of sulforaphane (SFN), an Nrf2 activator, on the fibroblast phenotype were tested under basal and pro-fibrosis conditions (transforming growth factor ß [TGF-ß]). RESULTS: Decreased Nrf2 expression was associated with a myofibroblast phenotype in IPF compared with control fibroblasts. Nrf2 knockdown induced oxidative stress and myofibroblastic differentiation in control fibroblasts. Conversely, Nrf2 activation increased antioxidant defences and myofibroblastic dedifferentation in IPF fibroblasts. SFN treatment decreased oxidants, and induced Nrf2 expression, antioxidants, and myofibroblastic dedifferentiation in IPF fibroblasts. SFN inhibited TGF-ß profibrotic deleterious effects in IPF and control fibroblasts and restored antioxidant defences. Nrf2 knockdown abolished SFN antifibrosis effects, suggesting that they were Nrf2 mediated. INNOVATION AND CONCLUSION: Our findings confirm that decreased nuclear Nrf2 plays a role in myofibroblastic differentiation and that SFN induces human pulmonary fibroblast dedifferentiation in vitro via Nrf2 activation. Thus, Nrf2 could be a novel therapeutic target in IPF.


Subject(s)
Cell Dedifferentiation , Cell Nucleus/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Aldehydes/metabolism , Animals , Becaplermin , Cells, Cultured , Collagen Type I/metabolism , Epoxide Hydrolases/metabolism , Gene Knockdown Techniques , Heme Oxygenase-1/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Isothiocyanates , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/physiology , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/physiology , Oxidative Stress , Phenotype , Proto-Oncogene Proteins c-sis/physiology , RNA, Small Interfering/genetics , Sulfoxides , Thiocyanates/pharmacology , Transforming Growth Factor beta/physiology
12.
Am J Pathol ; 181(6): 2126-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23031257

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown cause. Key signaling developmental pathways are aberrantly expressed in IPF. The hedgehog pathway plays a key role during fetal lung development and may be involved in lung fibrogenesis. We determined the expression pattern of several Sonic hedgehog (SHH) pathway members in normal and IPF human lung biopsies and primary fibroblasts. The effect of hedgehog pathway inhibition was assayed by lung fibroblast proliferation and differentiation with and without transforming growth factor (TGF)-ß1. We showed that the hedgehog pathway was reactivated in the IPF lung. Importantly, we deciphered the cross talk between the hedgehog and TGF-ß pathway in human lung fibroblasts. TGF-ß1 modulated the expression of key components of the hedgehog pathway independent of Smoothened, the obligatory signal transducer of the pathway. Smoothened was required for TGF-ß1-induced myofibroblastic differentiation of control fibroblasts, but differentiation of IPF fibroblasts was partially resistant to Smoothened inhibition. Furthermore, functional hedgehog pathway machinery from the primary cilium, as well as GLI-dependent transcription in the nucleus, was required for the TGF-ß1 effects on normal and IPF fibroblasts during myofibroblastic differentiation. These data identify the GLI transcription factors as potential therapeutic targets in lung fibrosis.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cilia/drug effects , Cilia/pathology , Female , Gene Expression Regulation/drug effects , Hedgehog Proteins/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunohistochemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Middle Aged , Models, Biological , Myofibroblasts/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Veratrum Alkaloids/pharmacology
13.
Am J Respir Crit Care Med ; 183(6): 759-66, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20935114

ABSTRACT

RATIONALE: Injury to alveolar epithelial cells is central to the pathophysiology of idiopathic pulmonary fibrosis (IPF). An abnormal autoimmune response directed against antigens of the alveolar epithelium may contribute to the disease. OBJECTIVES: To detect circulating autoantibodies (autoAbs) directed against epithelial structures. METHODS: We performed immunoblot by separating human placental amnion extract or alveolar epithelial cell (A549 cell line) proteins on polyacrylamide gels, blotting on nitrocellulose membranes, and incubating with serum from patients with IPF (n = 40) or healthy subjects (n = 40). Proteomic analysis and mass spectrometry characterized the target protein. Inhibition experiments performed with the correspondent recombinant protein confirmed our results. MEASUREMENTS AND MAIN RESULTS: We identified IgG autoAbs recognizing a 200-kD protein in the serum of patients with IPF. Proteomic analysis identified this protein as human periplakin (PPL), a component of desmosomes. Anti-PPL Abs were found by immunoblot in both serum and bronchoalveolar lavage in patients with IPF: 16/40 (40%) of them were positive versus none of the control subjects. Immunohistochemistry revealed that PPL was strongly expressed in bronchial and alveolar epithelium, but that PPL exhibited changes in intracellular localization among normal and fibrotic alveolar epithelium. In an alveolar epithelial wound repair assay, an anti-PPL IgG decreased cell migration. Recombinant PPL induced bronchoalveolar lavage T lymphocyte proliferation. Patients with IPF with anti-PPL Abs had a more severe respiratory disease, despite no difference in survival. CONCLUSIONS: We found a new circulating autoAb directed against PPL in patients with IPF, associated with a more severe disease.


Subject(s)
Autoantibodies/blood , Autoimmunity , Idiopathic Pulmonary Fibrosis/immunology , Immunoglobulin G/blood , Plakins/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Female , Humans , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/physiopathology , Male , Middle Aged , Respiratory Mucosa/immunology , Severity of Illness Index
14.
PLoS One ; 5(5): e10886, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20526373

ABSTRACT

BACKGROUND: Few data concern the pathophysiology of primary spontaneous pneumothorax (PSP), which is associated with alveolar hypoxia/reoxygenation. This study tested the hypothesis that PSP is associated with oxidative stress in lung macrophages. We analysed expression of the oxidative stress marker 4-HNE; the antioxidant and anti-inflammatory proteins heme oxygenase-1 (HO-1), biliverdin reductase (BVR) and heavy chain of ferritin (H-ferritin); and the transcription factors controlling their expression Nrf2 and HIF-1alpha, in lung samples from smoker and nonsmoker patients with PSP (PSP-S and PSP-NS), cigarette smoke being a risk factor of recurrence of the disease. METHODOLOGY/PRINCIPAL FINDINGS: mRNA was assessed by RT-PCR and proteins by western blot, immunohistochemistry and confocal laser analysis. 4-HNE, HO-1, BVR and H-ferritin were increased in macrophages from PSP-S as compared to PSP-NS and controls (C). HO-1 increase was associated with increased expression of HIF-1alpha mRNA and protein in alveolar macrophages in PSP-S patients, whereas Nrf2 was not modified. To understand the regulation of HO-1, BVR and H-ferritin, THP-1 macrophages were exposed to conditions mimicking conditions in C, PSP-S and PSP-NS patients: cigarette smoke condensate (CS) or air exposure followed or not by hypoxia/reoxygenation. Silencing RNA experiments confirmed that HIF-1alpha nuclear translocation was responsible for HO-1, BVR and H-ferritin induction mediated by CS and hypoxia/reoxygenation. CONCLUSIONS/SIGNIFICANCE: PSP in smokers is associated with lung macrophage oxidative stress. The response to this condition involves HIF-1alpha-mediated induction of HO-1, BVR and H-ferritin.


Subject(s)
Apoferritins/metabolism , Heme Oxygenase-1/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/enzymology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pneumothorax/enzymology , Smoking/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoferritins/genetics , Biopsy , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Hypoxia , Cell Line , Enzyme Induction , Eosinophil Major Basic Protein , Gene Expression Regulation , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/pathology , Macrophages/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Oxygen/metabolism , Pneumothorax/pathology , Proteoglycans/genetics , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
15.
Am J Respir Crit Care Med ; 182(3): 385-95, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20395561

ABSTRACT

RATIONALE: Lung dendritic cells (DCs) have been shown to accumulate in human fibrotic lung disease, but little is known concerning a role for DCs in the pathogenesis of fibrotic lung. OBJECTIVES: To characterize lung DCs in an in vivo model of bleomycin-induced pulmonary fibrosis in mice. METHODS: We characterized the kinetics and activation of pulmonary DCs during the course of bleomycin-induced lung injury by flow cytometry on lung single-cell suspensions. We also characterized the lymphocytes accumulating in bleomycin lung and the chemokines susceptible to favor the recruitment of immune cells. MEASUREMENTS AND MAIN RESULTS: We show, for the first time, that increased numbers of CD11c(+)/major histocompatibility complex class II(+) DCs, including CD11b(hi) monocyte-derived inflammatory DCs, infiltrate the lung of treated animals during the fibrotic phase of the response to bleomycin. These DCs are mature DCs expressing CD40, CD86, and CD83. They are associated with increased numbers of recently activated memory T cells expressing CD44, CD40L, and CD28, suggesting that fully mature DCs and Ag-experienced T cells can drive an efficient effector immune response within bleomycin lung. Most importantly, when DCs are inactivated with VAG539, a recently described new immunomodulator, VAG539 treatment attenuates the hallmarks of bleomycin lung injury. CONCLUSIONS: These findings identify lung DCs as key proinflammatory cells potentially able to sustain pulmonary inflammation and fibrosis in the bleomycin model.


Subject(s)
Dendritic Cells/metabolism , Lung/pathology , Pulmonary Fibrosis/pathology , Animals , Antigens, CD/metabolism , Bleomycin/pharmacology , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid/cytology , Chemokines/metabolism , Disease Models, Animal , Flow Cytometry , Immunologic Factors/pharmacology , Lung/metabolism , Major Histocompatibility Complex/physiology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , T-Lymphocytes/metabolism
16.
Am J Respir Cell Mol Biol ; 42(3): 286-93, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19448157

ABSTRACT

Hepatocyte growth factor (HGF) is a growth factor for alveolar epithelial cells. Activation of pro-HGF to HGF is regulated by the HGF activator (HGFA), a serine protease, and a specific inhibitor (HGFA inhibitor-1, HAI-1). An imbalance in the HGFA/HAI-1 system might contribute to lung fibrosis. Pro-HGF activation capacity from bronchoalveolar lavage (BAL) fluid was evaluated 3, 7, and 14 days after the intratracheal bleomycin injection (Bleo) in mice with or without thrombin. BAL fluid from naïve mice was used as control. HGFA and HAI-1 mRNA were evaluated by QPCR in the whole lung or by Western blot in BAL fluid. BAL fluid from control mice and Bleo mice activated pro-HGF in vitro at a similar degree. Thrombin accelerated proHGF activation by Bleo BAL on Day 3 and Day 7, but not on Day 14, or in control BAL. Incubation of pro-HGF with BAL from Bleo Day 3 and Day 7 mice increased phosphorylation of HGFR on A549 cells. Thrombin-induced pro-HGF activation was inhibited by an anti-HGFA antibody and accelerated by an anti-HAI-1 antibody. Active HGFA was not detected in control BAL and was strongly induced in Bleo BAL. HGFA concentrations were higher on Day 3 and Day 7 than on Day 14. HAI-1 was detected at low levels in control BAL and increased strongly by Day 3 with stable concentrations until Day 14. By demonstrating an imbalance between HGFA and HAI-1 expression in BAL fluid, our results highlight a defective thrombin-dependent proHGF activation system at the fibrotic phase of bleomycin-induced pulmonary fibrosis.


Subject(s)
Bleomycin , Hepatocyte Growth Factor/metabolism , Protein Precursors/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Antibodies, Neutralizing/immunology , Bronchoalveolar Lavage Fluid , Cell Line , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Lung/metabolism , Lung/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Proteinase Inhibitory Proteins, Secretory , Proto-Oncogene Proteins c-met/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism
17.
PLoS One ; 4(5): e5585, 2009.
Article in English | MEDLINE | ID: mdl-19440310

ABSTRACT

BACKGROUND: Fibrosis, which is characterized by the pathological accumulation of collagen, is recognized as an important feature of many chronic diseases, and as such, constitutes an enormous health burden. We need non-invasive specific methods for the early diagnosis and follow-up of fibrosis in various disorders. Collagen targeting molecules are therefore of interest for potential in vivo imaging of fibrosis. In this study, we developed a collagen-specific probe using a new approach that takes advantage of the inherent specificity of Glycoprotein VI (GPVI), the main platelet receptor for collagens I and III. METHODOLOGY/PRINCIPAL FINDINGS: An anti-GPVI antibody that neutralizes collagen-binding was used to screen a bacterial random peptide library. A cyclic motif was identified, and the corresponding peptide (designated collagelin) was synthesized. Solid-phase binding assays and histochemical analysis showed that collagelin specifically bound to collagen (Kd 10(-7) M) in vitro, and labelled collagen fibers ex vivo on sections of rat aorta and rat tail. Collagelin is therefore a new specific probe for collagen. The suitability of collagelin as an in vivo probe was tested in a rat model of healed myocardial infarctions (MI). Injecting Tc-99m-labelled collagelin and scintigraphic imaging showed that uptake of the probe occurred in the cardiac area of rats with MI, but not in controls. Post mortem autoradiography and histological analysis of heart sections showed that the labeled areas coincided with fibrosis. Scintigraphic molecular imaging with collagelin provides high resolution, and good contrast between the fibrotic scars and healthy tissues. The capacity of collagelin to image fibrosis in vivo was confirmed in a mouse model of lung fibrosis. CONCLUSION/SIGNIFICANCE: Collagelin is a new collagen-targeting agent which may be useful for non-invasive detection of fibrosis in a broad spectrum of diseases.


Subject(s)
Diagnostic Imaging/methods , Fibrosis/pathology , Peptides/chemistry , Peptides/metabolism , Platelet Membrane Glycoproteins/chemistry , Radionuclide Imaging/methods , Animals , Aorta/metabolism , Blotting, Western , Fibrosis/metabolism , In Vitro Techniques , Peptides/chemical synthesis , Rats , Tail/metabolism
18.
Am J Physiol Lung Cell Mol Physiol ; 294(2): L334-43, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18065658

ABSTRACT

The mechanisms of pulmonary repair in acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) are poorly known. Hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF) are key factors involved in alveolar epithelial repair, present in the bronchoalveolar lavage fluid (BALF) from patients with ALI/ARDS. The role of BALF mediators in their production remains to be determined. We evaluated the overall effect of BALF from 52 patients (27 ventilated patients with ALI/ARDS, 10 ventilated patients without ALI, and 15 nonventilated control patients) on HGF and KGF synthesis by lung fibroblasts. Fibroblasts were cultured in the presence of BALF. HGF and KGF protein secretion was measured using ELISA, and mRNA expression was evaluated using quantitative real-time RT-PCR. Only BALF from ALI/ARDS patients upregulated both HGF and KGF mRNA expression and protein synthesis (+271 and +146% for HGF and KGF, respectively). BALF-induced HGF synthesis from ALI/ARDS patients was higher than that from ventilated patients without ALI (P < 0.05). HGF secretion was correlated with BALF IL-1beta levels (rho = 0.62, P < 0.001) and BALF IL-1beta/IL-1 receptor antagonist ratio (rho = 0.54, P < 0.007) in the ALI/ARDS group. An anti-IL-1beta antibody partially (>50%) inhibited the BALF-induced HGF and PGE(2) secretion, whereas NS-398, a specific cyclooxygenase-2 (COX-2) inhibitor, completely inhibited it. Anti-IL-1beta antibodies as well as NS-398 reversed the COX-2 upregulation induced by BALF. Therefore, IL-1beta is a main BALF mediator involved in HGF secretion, which is mediated through a PGE(2)/COX-2-dependent mechanism. BALF mediators may participate in vivo in the production of HGF and KGF by lung fibroblasts during ALI/ARDS.


Subject(s)
Fibroblasts/metabolism , Hepatocyte Growth Factor/metabolism , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Antibodies/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Female , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation/drug effects , Hepatocyte Growth Factor/genetics , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Distress Syndrome/enzymology
19.
Am J Respir Crit Care Med ; 176(10): 1007-14, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17717200

ABSTRACT

RATIONALE: There is growing evidence that resident cells, such as fibroblasts and epithelial cells, can drive the persistent accumulation of dendritic cells (DCs) in chronically inflamed tissue, leading to the organization and the maintenance of ectopic lymphoid aggregates. This phenomenon, occurring through a chemokine-mediated retention mechanism, has been documented in various disorders, but not in fibrotic interstitial lung disorders in which the presence of organized lymphoid follicles has been documented. OBJECTIVES: To characterize the distribution of DCs in fibrotic lung, and to analyze the expression of the main chemokines known to regulate DC recruitment. METHODS: Lung resection tissue (lungs with idiopathic pulmonary fibrosis; n = 12; lungs with nonspecific interstitial pneumonia, n = 5; control lungs, n = 5) was snap-frozen for subsequent immunohistochemical techniques on serial sections and reverse transcriptase-polymerase chain reaction analysis. MEASUREMENTS AND MAIN RESULTS: Results were similar in idiopathic pulmonary fibrosis and nonspecific interstitial pneumonia lungs, which were heavily infiltrated by immature DCs in established fibrosis and in areas of epithelial hyperplasia. Altered epithelial cells and fibroblasts, particularly in fibroblastic foci, frankly expressed all chemokines (CCL19, CCL20, CCL22, and CXCL12) susceptible to favor the recruitment of immune cells. Lymphoid follicles were infiltrated by maturing DCs, which could originate from the pool of DCs accumulating in their vicinity. CONCLUSIONS: These findings suggest that resident cells in pulmonary fibrosis can sustain chronic inflammation by driving the accumulation of DCs with the potential to mature locally within ectopic lymphoid follicles. Future strategies should consider DCs or chemokines as therapeutic targets in the treatment of pulmonary fibrosis.


Subject(s)
Chemokines, CC/metabolism , Dendritic Cells/physiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Adult , Aged , Case-Control Studies , Cell Count , Cell Movement , Epithelial Cells/physiology , Female , Fibroblasts/physiology , Humans , Male , Middle Aged
20.
Microsc Res Tech ; 70(2): 162-70, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17177275

ABSTRACT

The organization of collagen during fibrotic processes is poorly characterized because of the lack of appropriate methodologies. Here we show that multimodal multiphoton microscopy provides novel insights into lung fibrosis. We characterize normal and fibrotic pulmonary tissue in the bleomycin model, and show that second-harmonic generation by fibrillar collagen reveals the micrometer-scale three-dimensional spatial distribution of the fibrosis. We find that combined two-photon excited fluorescence and second-harmonic imaging of unstained lung tissue allows separating the inflammatory and fibrotic steps in this pathology, underlining characteristic features of fibroblastic foci in human Idiopathic Pulmonary Fibrosis samples. Finally, we propose phenomenological scores of lung fibrosis and we show that they unambiguously sort out control and treated mice, with a better sensitivity and reproducibility in the subpleural region. These results should be readily generalized to other organs, as an accurate method to assess extracellular matrix remodeling during fibrosis.


Subject(s)
Collagen/ultrastructure , Extracellular Matrix/ultrastructure , Lung/pathology , Microscopy, Fluorescence, Multiphoton/methods , Pulmonary Fibrosis/pathology , Animals , Bleomycin , Humans , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Pleura/pathology , Pulmonary Fibrosis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...