Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 339
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L29-L40, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33026236

ABSTRACT

Pulmonary fibrosis is one of the important causes of morbidity and mortality in fibroproliferative disorders such as systemic sclerosis (SSc) and idiopathic pulmonary fibrosis (IPF). Lysyl oxidase (LOX) is a copper-dependent amine oxidase whose primary function is the covalent crosslinking of collagens in the extracellular matrix (ECM). We investigated the role of LOX in the pathophysiology of SSc. LOX mRNA and protein levels were increased in lung fibroblasts of SSc patients compared with healthy controls and IPF patients. In vivo, bleomycin induced LOX mRNA expression in lung tissues, and LOX activity increased in the circulation of mice with pulmonary fibrosis, suggesting that circulating LOX parallels levels in lung tissues. Circulating levels of LOX were reduced upon amelioration of fibrosis with an antifibrotic peptide. LOX induced ECM production at the transcriptional level in lung fibroblasts, human lungs, and human skin maintained in organ culture. In vivo, LOX synergistically exacerbated fibrosis in bleomycin-treated mice. Further, LOX increased the production of interleukin (IL)-6, and the increase was mediated by LOX-induced c-Fos expression, the nuclear localization of c-Fos, and its engagement with the IL-6 promoter region. Our findings demonstrate that LOX expression and activity correlate with fibrosis in vitro, ex vivo, and in vivo. LOX induced ECM production via upregulation of IL-6 and nuclear localization of c-Fos. Thus, LOX has a direct pathogenic role in SSc-associated fibrosis that is independent of its crosslinking function. Our findings also suggest that measuring circulating LOX levels and activity can be used for monitoring response to antifibrotic therapy.


Subject(s)
Extracellular Matrix/pathology , Lung/pathology , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Case-Control Studies , Extracellular Matrix/enzymology , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Interleukin-6/metabolism , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Protein-Lysine 6-Oxidase/genetics , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/etiology , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/etiology
2.
J Cell Sci ; 132(20)2019 10 18.
Article in English | MEDLINE | ID: mdl-31527052

ABSTRACT

Tissue fibrosis is a chronic disease driven by persistent fibroblast activation that has recently been linked to epigenetic modifications. Here, we screened a small library of epigenetic small-molecule modulators to identify compounds capable of inhibiting or reversing TGFß-mediated fibroblast activation. We identified pracinostat, an HDAC inhibitor, as a potent attenuator of lung fibroblast activation and confirmed its efficacy in patient-derived fibroblasts isolated from fibrotic lung tissue. Mechanistically, we found that HDAC-dependent transcriptional repression was an early and essential event in TGFß-mediated fibroblast activation. Treatment of lung fibroblasts with pracinostat broadly attenuated TGFß-mediated epigenetic repression and promoted fibroblast quiescence. We confirmed a specific role for HDAC-dependent histone deacetylation in the promoter region of the anti-fibrotic gene PPARGC1A (PGC1α) in response to TGFß stimulation. Finally, we identified HDAC7 as a key factor whose siRNA-mediated knockdown attenuates fibroblast activation without altering global histone acetylation. Together, these results provide novel mechanistic insight into the essential role HDACs play in TGFß-mediated fibroblast activation via targeted gene repression.


Subject(s)
Down-Regulation/drug effects , Fibroblasts/enzymology , Histone Deacetylases/metabolism , Lung/enzymology , Pulmonary Fibrosis/enzymology , Transforming Growth Factor beta/pharmacology , Cell Line , Fibroblasts/pathology , Histone Deacetylases/genetics , Humans , Lung/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Promoter Regions, Genetic , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology
3.
J Vasc Res ; 58(4): 237-251, 2021.
Article in English | MEDLINE | ID: mdl-33910208

ABSTRACT

INTRODUCTION: Pulmonary hypertension is characterized by vasoconstriction and remodeling of pulmonary arteries, leading to right ventricular hypertrophy and failure. We have previously found upregulation of transglutaminase 2 (TG2) in the right ventricle of chronic hypoxic rats. The hypothesis of the present study was that treatment with the transglutaminase inhibitor, cystamine, would inhibit the development of pulmonary arterial remodeling, pulmonary hypertension, and right ventricular hypertrophy. METHODS: Effect of cystamine on transamidase activity was investigated in tissue homogenates. Wistar rats were exposed to chronic hypoxia and treated with vehicle, cystamine (40 mg/kg/day in mini-osmotic pumps), sildenafil (25 mg/kg/day), or the combination for 2 weeks. RESULTS: Cystamine concentration-dependently inhibited TG2 transamidase activity in liver and lung homogenates. In contrast to cystamine, sildenafil reduced right ventricular systolic pressure and hypertrophy and decreased pulmonary vascular resistance and muscularization in chronic hypoxic rats. Fibrosis in the lung tissue decreased in chronic hypoxic rats treated with cystamine. TG2 expression was similar in the right ventricle and lung tissue of drug and vehicle-treated hypoxic rats. DISCUSSION/CONCLUSIONS: Cystamine inhibited TG2 transamidase activity, but cystamine failed to prevent pulmonary hypertension, right ventricular hypertrophy, and pulmonary arterial muscularization in the chronic hypoxic rat.


Subject(s)
Arterial Pressure/drug effects , Cystamine/pharmacology , Enzyme Inhibitors/pharmacology , Hypertension, Pulmonary/prevention & control , Hypoxia/drug therapy , Protein Glutamine gamma Glutamyltransferase 2/antagonists & inhibitors , Pulmonary Artery/drug effects , Animals , Disease Models, Animal , Female , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/complications , Hypoxia/enzymology , Hypoxia/physiopathology , Male , Mice, Inbred C57BL , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/physiopathology , Pulmonary Fibrosis/prevention & control , Rats, Wistar , Vascular Remodeling/drug effects , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
4.
Toxicol Appl Pharmacol ; 432: 115757, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34673086

ABSTRACT

An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1ß, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-ß1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-ß1/SMAD pathways.


Subject(s)
HMGB1 Protein/metabolism , Lung/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Pneumonia/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/enzymology , Receptor for Advanced Glycation End Products/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Arsenites , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Lung/ultrastructure , Male , Phosphorylation , Pneumonia/chemically induced , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats, Wistar , Signal Transduction , Sodium Compounds
5.
Biochem Biophys Res Commun ; 532(4): 598-604, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32900484

ABSTRACT

OBJECTIVE: Pulmonary fibrosis is a fatal interstitial lung disease that is characterized by excessive accumulation of extracellular matrix (ECM) and remodeling of lung. The precise mechanisms underlying pulmonary fibrosis still remain unclear. In the current study, we aimed to investigate the alteration and function of serine (or cysteine) peptidase inhibitor, clade A, member 3 N (Serpina3n) in pulmonary fibrotic models and explore the potential mechanisms. METHODS: We induced pulmonary fibrosis in mice by silica and bleomycin respectively and determined Serpina3n in lung tissues, and then verified the expression of Serpina3n and its correlation with pulmonary fibrosis at seven time points in a bleomycin longstanding model. Moreover, adeno-associated virus type 9 (AAV9)-mediated Serpina3n knockdown was used to treat pulmonary fibrosis in the bleomycin model, whose possible mechanisms would be preliminarily explored by detecting chymotrypsin C as an example. RESULTS: Serpina3n was up-regulated significantly in lungs of both models at mRNA and protein levels relative to control. Notably, the expression of Serpina3n peaked during the 3rd week and then decreased until nearly normal levels during the 10th week, which was closely related to fibrotic procession in bleomycin-treated mice. AAV-mediated Serpina3n knockdown in the lung tissues alleviated bleomycin-induced fibrotic symptoms at various levels and disinhibit chymotrypsin C. CONCLUSIONS: Our study revealed that Serpina3n is a critical regulator in pulmonary fibrosis and suggested Serpina3n inhibition as a potential therapeutic strategy in chronic pulmonary injuries.


Subject(s)
Acute-Phase Proteins/physiology , Pulmonary Fibrosis/metabolism , Serpins/physiology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Bleomycin , Chymotrypsin/metabolism , Gene Knockdown Techniques , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Serpins/genetics , Serpins/metabolism , Up-Regulation
6.
Mol Cell Biochem ; 469(1-2): 159-167, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32350681

ABSTRACT

The present study investigated the therapeutic effect of curcumin on bleomycin (BLM)-induced alterations in glycoprotein components in the fibrotic lungs. Analysis of the bronchoalveolar lavage fluid (BALF) demonstrated increased fibronectin content at 3, 5, 7, and 14 days after BLM administration. Similarly, lung tissue fibronectin content revealed a progressive increase at various times (days 3, 5, 7, 14, and 28) during the development of lung fibrosis. In addition, alveolar macrophage release of fibronectin was also elevated in BLM-treated rats. Analysis of carbohydrate moieties of glycoproteins revealed an increase in total hexose, fucose, sialic acid and hexosamine levels at 7, 14, and 28 days after BLM treatment. Furthermore, the activities of lung glycosidases such as N-acetyl-ß-D-glucosaminidase, ß-glucosidase, ß-galactosidase, and ß-fucosidase in the fibrotic rats were elevated. Importantly, curcumin significantly inhibited the BLM-induced increases in BALF and lung fibronectin levels. Treatment of BLM rats with curcumin dramatically suppressed alveolar macrophage release of fibronectin. Curcumin also inhibited the increases in complex carbohydrates and glycosidases in the fibrotic lungs. These findings suggest that BLM-induced lung fibrosis is associated with accumulation of glycoproteins, and curcumin has the ability to suppress the enhanced deposition of glycoproteins in the fibrotic lung.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Curcumin/pharmacology , Glycoproteins/analysis , Pulmonary Fibrosis/drug therapy , Acetylglucosaminidase/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Bleomycin/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Fibronectins/metabolism , Fucose/analysis , Glycoproteins/metabolism , Hexosamines/analysis , Hexoses/analysis , Macrophages, Alveolar/drug effects , Male , N-Acetylneuraminic Acid/analysis , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/metabolism , Rats , Rats, Wistar , beta-Galactosidase/metabolism , beta-Glucosidase/metabolism
7.
Exp Lung Res ; 46(3-4): 75-80, 2020.
Article in English | MEDLINE | ID: mdl-32102576

ABSTRACT

Purpose: We previously found extensive desialylation of glycoconjugates and upregulation of the sialidase enzyme NEU3 in fibrotic lesions in human and mouse lungs. However, studies using microarray analysis of whole lung tissue mRNA and single cell RNA-seq found no significant difference in levels of NEU3 mRNA between IPF patients and controls. This study aimed to elucidate how NEU3 was upregulated in fibrotic lungs.Materials and methods: Transforming growth factor-ß1 (TGF-ß1), a key driver of fibrosis, was added to A549 human alveolar basal epithelial adenocarcinoma cells and human small airway epithelial cells (HSAEpC). NEU3 expression in A549 cells and HSAEpC was detected by immunofluorescence staining. NEU3 translation and degradation were assessed by polysome profiling (polysomes efficiently translate mRNAs; monosomes poorly translate mRNAs) and cycloheximide chase after treating cells with or without TGF-ß1 for 48 h.Results: TGF-ß1 increased NEU3 expression and secretion in A549 cells and HSAEpC but did not change total (nuclear + cytosolic) NEU3 mRNA levels. TGF-ß1 decreased the degradation rate of NEU3 in A549 cells. TGF-ß1 decreased NEU3 mRNA levels in monosomes and increased NEU3 mRNA level in polysomes.Conclusion: TGF-ß1 upregulates levels of NEU3 in epithelial cells by both decreasing NEU3 degradation and by increasing the translation of NEU3 mRNA, explaining the apparent paradox of high levels of NEU3 protein in pulmonary fibrosis without a concomitant increase in the expression of NEU3 mRNA.


Subject(s)
Neuraminidase/metabolism , Pulmonary Fibrosis/etiology , Transforming Growth Factor beta1/metabolism , A549 Cells , Epithelial Cells/enzymology , Humans , Pulmonary Fibrosis/enzymology
8.
Immunopharmacol Immunotoxicol ; 42(5): 436-444, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32762376

ABSTRACT

BACKGROUND: Bleomycin (BLM) is a chemotherapy drug used to treat cancer, one of which side effects is that it can lead to pulmonary fibrosis (PF). Atractylenoide III (AtrIII), derived from the dried roots of rhizoma atractylodis of compositae, is one of the main active substances of rhizoma atractylodis. It has anti-inflammatory, anti-tumor and other effects. This study aimed to investigate whether AtrIII alleviated BLM-induced PF and oxidative stress in rats through the nuclear factor erythroid-2-related factor 2/NQO1,NAD(P)H:quinine oxidoreductase 1/Heme oxygenase-1 (Nrf2/NQO1/HO-1) pathway. METHODS: A BLM-induced pulmonary fibrosis model in SD rats was established. The respiratory dynamics were evaluated by using Wholebody flow-through plethysmography. Lung injury and pulmonary fibrosis were observed by Hematoxylin-eosin (HE) and Masson staining. Apoptosis was assay by Tunel assay. Inflammatory factors were detected with commercial kits. Expression of mRNAs and proteins were detected by RT-qPCR and Western blot, respectively. RESULTS: AtrIII (1.2, 2.4 mg/kg) improved the lung injury and lung function in the BLM-induced Sprague-Dawley (SD) rats. AtrIII reduced the apoptosis rate and protein expression of Caspase-3 and Caspase-9. AtrIII (1.2, 2.4 mg/kg) decrease the pulmonary fibrosis damage and protein expression transforming growth factor-ß (TGF-ß) and α-smooth muscle actin (α-SMA). AtrIII also down-regulated the levels of interleukin 6 (IL-6), inductible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α), while up-regulated the level of IL-10 in peripheral blood serum. Moreover, AtrIII (1.2, 2.4 mg/kg) increased the activity of superoxide dismutase (SOD) and glutathione (GSH), while decreased the malondialdehyde (MDA) content and lactate dehydrogenase (LDH) activity. AtrIII (1.2, 2.4 mg/kg) increased the levels of Nrf2, NQO1 and HO-1. In addition, AtrIII reversed the effects of Nrf2 interference on pulmonary fibrosis damage, decreased SOD and GSH activity, and increased MDA content. CONCLUSION: AtrIII could attenuate the pulmonary fibrosis and reliev oxidative stress through the Nrf2/NQO1/ HO-1 pathway.


Subject(s)
Antioxidants/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Lactones/pharmacology , Lung/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Pulmonary Fibrosis/prevention & control , Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Bleomycin , Disease Models, Animal , Lung/enzymology , Lung/pathology , Male , NF-E2-Related Factor 2/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Rats, Sprague-Dawley , Signal Transduction
9.
Am J Respir Cell Mol Biol ; 61(4): 492-500, 2019 10.
Article in English | MEDLINE | ID: mdl-30943369

ABSTRACT

It has been increasingly recognized lately that aberrant cellular metabolism plays an important role in the pathogenesis of pulmonary fibrosis. In our previous systemic studies, we found that human lung myofibroblasts undergo glutaminolytic reprogramming, which is mediated by an increased expression of glutaminase (Gls) 1. We showed that augmented glutaminolysis critically regulates collagen production by promoting its stabilization in human lung myofibroblasts. Our study indicates that lung fibroblast Gls1 is a promising therapeutic target for this disease. In this investigation, we primarily focused on delineating the in vivo role of fibroblast Gls1 in mouse models of pulmonary fibrosis and determining the efficacy of Gls1 inhibition in treating this pathology. We now show that fibroblast Gls1 is upregulated in fibrotic mouse lungs. We present evidence that mice with ablation of fibroblast Gls1 are protected from bleomycin-induced lung fibrosis. We show that the Gls1 inhibitor, CB-839, is therapeutically efficacious in treating both bleomycin- and transforming growth factor-ß1-induced pulmonary fibrosis. Our study has thus established a solid rationale for advancing Gls1 inhibitors, particularly CB-839, to the next stage of testing in the treatment of this disease.


Subject(s)
Fibroblasts/enzymology , Glutaminase/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Animals , Benzeneacetamides/pharmacology , Bleomycin/toxicity , Cell Line , Collagen/biosynthesis , Enzyme Induction , Glutaminase/genetics , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Thiadiazoles/pharmacology , Transforming Growth Factor beta1/pharmacology
10.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L961-L976, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30785343

ABSTRACT

Matrix metalloprotease 13 (MMP13) deficiency in pulmonary fibrosis has described contradictory phenotypes on inflammatory and fibrotic responses after lung injury, and its role during lung fibrosis resolution is still undefined. MMP13 has been considered the main collagenase in rodents, and the remodeling of fibrillar collagen is widely attributed to the action of this enzyme. In this study we aimed to explore the role of MMP13 during lung fibrosis progression and resolution. Lung fibrosis was induced by intratracheal instillation, and inflammatory, fibrotic, and resolution stages were evaluated in Mmp13-null and wild-type (WT) mice. Bronchoalveolar lavage fluid was taken for cytokine array analysis and activity of gelatinases. Our results showed that MMP13 is upregulated mainly during two stages after lung injury, inflammation and resolution of fibrosis, and it is mainly expressed by alveolar and interstitial macrophages. Mmp13-null mice exhibited more extensive inflammation at 7 days after bleomycin treatment, and it was characterized by increased macrophage infiltration and significant alterations in proinflammatory cytokines. We also documented that Mmp13-deficient mice experienced more severe and prolonged lung fibrosis compared with WT mice. Delayed resolution in Mmp13-deficient lungs was characterized by a decreased overall collagenolytic activity and persistent fibrotic foci associated with emphysema-like areas. Together, our findings indicate that MMP13 plays an antifibrotic role and its activity is crucial in lung repair and restoration of tissue integrity during fibrosis resolution.


Subject(s)
Bleomycin/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 13 , Pulmonary Fibrosis , Up-Regulation/drug effects , Animals , Bleomycin/pharmacology , Bronchoalveolar Lavage , Cytokines/genetics , Cytokines/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Mice , Mice, Mutant Strains , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology
11.
Toxicol Appl Pharmacol ; 366: 10-16, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30653976

ABSTRACT

Idiopathic pulmonary fibrosis is a pathological result of dysfunctional repair response to tissue injury, leading to chronically impaired gas exchange and death. Macrophages are believed to be critical in this disease pathogenesis; However, the exact mechanisms remain enigmatic. Here, we demonstrated that macrophages might contribute to pulmonary fibrosis at the early stage because the aggregation of macrophages appeared earlier than epithelial-mesenchymal transition and fibrosis in mouse and rat experimental models of pulmonary fibrosis. It has been found that macrophages could promote epithelial-mesenchymal transition of alveolar epithelial cells and fibroblast migration in co-culture models between macrophages and alveolar epithelial cells/fibroblasts. Importantly, we used protein micro array to analyze the cytokines that were altered after bleomycin treatment. Only thymic stromal lymphopoietin and matrix metalloproteinase 9 were significantly increased. We further confirmed that TSLP participated in the macrophage-induced epithelial-mesenchymal transition of alveolar epithelial cells using a TSLP recombinant protein. MMP9 was also involved in macrophage-induced fibroblast migration, which can be reversed by an inhibitor of MMP9. Collectively, these findings explained the underlying mechanisms of macrophage-promoted pulmonary fibrosis.


Subject(s)
Bleomycin , Cytokines/metabolism , Fibroblasts/enzymology , Lung/enzymology , Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 9/metabolism , Pulmonary Fibrosis/enzymology , Animals , Cell Line , Cell Movement , Coculture Techniques , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Fibroblasts/pathology , Lung/pathology , Macrophages, Alveolar/pathology , Mice, Inbred ICR , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats, Wistar , Signal Transduction , Time Factors , Thymic Stromal Lymphopoietin
12.
FASEB J ; 32(3): 1250-1264, 2018 03.
Article in English | MEDLINE | ID: mdl-29122847

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by the deposition of excessive extracellular matrix and the destruction of lung parenchyma, resulting from an aberrant wound-healing response. Although IPF is often associated with an imbalance in protease activity, the mechanisms underlying the sustained repair mechanisms are not fully understood. Here, we addressed the role of the recently identified, membrane-anchored serine protease human airway trypsin-like protease (HAT). In the present study, we show that both HAT expression and activity were up-regulated in human IPF specimens. Next, adenoviral overexpression of HAT before bleomycin challenge attenuated lung injury as well as extracellular matrix deposition in the bleomycin-induced pulmonary fibrosis model. In vitro, HAT prevented specific fibrosis-associated responses in primary human pulmonary fibroblasts and induced the expression of mediators associated with the prostaglandin E2 pathway. Altogether, our findings suggested that HAT could have a protective role in IPF and other fibrotic lung disorders.-Menou, A., Flajolet, P., Duitmen, J., Justet, A., Moog, S., Jaillet, M., Tabèze, L., Solhonne, B., Garnier, M., Mal, H., Mordant, P., Castier, Y., Cazes, A., Sallenave, J.-M., Mailleux, A. A., Crestani, B. Human airway trypsin-like protease exerts potent, antifibrotic action in pulmonary fibrosis.


Subject(s)
Lung Injury/prevention & control , Pulmonary Fibrosis/prevention & control , Serine Endopeptidases/administration & dosage , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Lung/drug effects , Lung/enzymology , Lung/pathology , Lung Injury/chemically induced , Lung Injury/enzymology , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Serine Endopeptidases/metabolism , Signal Transduction
13.
Am J Respir Cell Mol Biol ; 58(1): 28-39, 2018 01.
Article in English | MEDLINE | ID: mdl-28800254

ABSTRACT

Pulmonary fibrosis is the leading cause of death in systemic sclerosis (SSc). Sirtuin1 (SIRT1) is a deacetylase with known antiinflammatory and antifibrotic activity in the liver, kidney, and skin. The role of SIRT1 in SSc-related pulmonary fibrosis is unknown. In the present work, we determined that the expression of SIRT1 in peripheral blood mononuclear cells of patients with SSc with pulmonary fibrosis is lower than that in patients with SSc without pulmonary fibrosis. In in vivo studies of bleomycin-induced lung fibrosis in mice, SIRT1 activation with resveratrol reduced collagen production when it was administered either prophylactically during the inflammatory stage or after the development of fibrosis. Furthermore, SIRT1 activation or overexpression inhibited tumor necrosis factor-α-induced inflammatory responses in vitro in human fetal lung fibroblasts, depletion of SIRT1 in fibroblasts enhanced inflammation, and these effects were related to changes in the acetylation of NF-κB. In addition, SIRT1 activation or exogenous overexpression inhibited collagen production in vitro, and these manipulations also inhibited fibrosis via inactivation of transforming growth factor-ß/mothers against decapentaplegic homolog and mammalian target of rapamycin signaling. Taken together, our results show that a loss of SIRT1 may participate in the pathogenesis of SSc-related pulmonary fibrosis, and that SIRT1 activation is an effective treatment for both the early (inflammatory) and late (fibrotic) stages of pulmonary fibrosis. Thus, SIRT1 may be a promising therapeutic target in the management of SSc-related pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , Scleroderma, Systemic , Sirtuin 1/metabolism , Animals , Cell Line , Enzyme Activation , Female , Humans , Male , Mice , NF-kappa B/metabolism , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/prevention & control , Scleroderma, Systemic/complications , Scleroderma, Systemic/enzymology , Scleroderma, Systemic/prevention & control , Tumor Necrosis Factor-alpha/metabolism
14.
Am J Respir Cell Mol Biol ; 58(4): 471-481, 2018 04.
Article in English | MEDLINE | ID: mdl-29211497

ABSTRACT

Pulmonary fibrosis is thought to result from dysregulated wound repair after repetitive lung injury. Many cellular responses to injury involve rearrangements of the actin cytoskeleton mediated by the two isoforms of the Rho-associated coiled-coil-forming protein kinase (ROCK), ROCK1 and ROCK2. In addition, profibrotic mediators such as transforming growth factor-ß, thrombin, and lysophosphatidic acid act through receptors that activate ROCK. Inhibition of ROCK activation may be a potent therapeutic strategy for human pulmonary fibrosis. Pharmacological inhibition of ROCK using nonselective ROCK inhibitors has been shown to prevent fibrosis in animal models; however, the specific roles of each ROCK isoform are poorly understood. Furthermore, the pleiotropic effects of this kinase have raised concerns about on-target adverse effects of ROCK inhibition such as hypotension. Selective inhibition of one isoform might be a better-tolerated strategy. In the present study, we used a genetic approach to determine the roles of ROCK1 and ROCK2 in a mouse model of bleomycin-induced pulmonary fibrosis. Using ROCK1- or ROCK2-haploinsufficient mice, we found that reduced expression of either ROCK1 or ROCK2 was sufficient to protect them from bleomycin-induced pulmonary fibrosis. In addition, we found that both isoforms contribute to the profibrotic responses of epithelial cells, endothelial cells, and fibroblasts. Interestingly, ROCK1- and ROCK2-haploinsufficient mice exhibited similar protection from bleomycin-induced vascular leak, myofibroblast differentiation, and fibrosis; however, ROCK1-haploinsufficient mice demonstrated greater attenuation of epithelial cell apoptosis. These findings suggest that selective inhibition of either ROCK isoform has the potential to be an effective therapeutic strategy for pulmonary fibrosis.


Subject(s)
Fibroblasts/enzymology , Lung/enzymology , Pulmonary Fibrosis/prevention & control , rho-Associated Kinases/metabolism , Animals , Apoptosis , Bleomycin , Capillary Permeability , Cell Differentiation , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Fibroblasts/pathology , Haploinsufficiency , Humans , Lung/pathology , Mice, Knockout , Myofibroblasts/enzymology , Myofibroblasts/pathology , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , rho-Associated Kinases/deficiency , rho-Associated Kinases/genetics
15.
Toxicol Appl Pharmacol ; 348: 43-53, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29673857

ABSTRACT

Lung epithelial cells are the first cell-type to come in contact with hazardous dust materials. Upon deposition, they invoke complex reactions in attempt to eradicate particles from the airways, and repair damage. The cell surface is composed of a heterogeneous network of matrix proteins and proteoglycans, which act as scaffold and control cell-signaling networks. These functions are controlled, in part, by the sulfation patterns of heparin-sulfate proteoglycans (HSPGs), which are enzymatically regulated. Although there is evidence of altered HSPG-sulfation in idiopathic pulmonary fibrosis (IPF), this is not investigated in silicosis. Our previous studies revealed down-regulation of Sulfatase-1 (SULF1) in human bronchial epithelial cells (BECs) by crystalline silica (CS). In this study, CS-induced down-regulation of SULF1, and increases in Sulfated-HSPGs, were determined in human BECs, and in rat lungs. By siRNA and plasmid transfection techniques the effects of SULF1 expression on silica-induced fibrogenic and proliferative gene expression were determined. These studies confirmed down-regulation of SULF1 and subsequent increases in sulfated-HSPGs in vitro. Moreover, short-term exposure of rats to CS resulted in similar changes in vivo. Conversely, effects were reversed after long term CS exposure of rats. SULF1 knockdown, and overexpression alleviated and exacerbated silica-induced decrease in cell viability, respectively. Furthermore, overexpression of SULF1 promoted silica-induced proliferative and fibrogenic gene expression, and collagen production. These findings demonstrate that the HSPG modification enzyme SULF1 and HSPG sulfation are altered by CS in vitro and in vivo. Furthermore, these changes may contribute to CS-induced lung pathogenicity by affecting injury tolerance, hyperproliferation, and fibrotic effects.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Lung/drug effects , Pulmonary Fibrosis/chemically induced , Silicon Dioxide/toxicity , Silicosis/etiology , Sulfotransferases/metabolism , Animals , Cell Line , Collagen/metabolism , Crystallization , Down-Regulation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Heparin/analogs & derivatives , Heparin/metabolism , Humans , Lung/enzymology , Lung/pathology , Proteoglycans/metabolism , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Rats, Wistar , Signal Transduction/drug effects , Silicon Dioxide/chemistry , Silicosis/enzymology , Silicosis/genetics , Silicosis/pathology , Sulfotransferases/genetics , Time Factors
16.
FASEB J ; 31(6): 2477-2491, 2017 06.
Article in English | MEDLINE | ID: mdl-28251930

ABSTRACT

In pulmonary fibrosis, an inflammatory reaction and differentiation of myofibroblasts culminate in pathologic deposition of collagen. Amine oxidase copper containing-3 (AOC3) is a cell-surface-expressed oxidase that regulates leukocyte extravasation. Here we analyzed the potential role of AOC3 using gene-modified and inhibitor-treated mice in a bleomycin-induced pulmonary fibrosis model. Inflammation and fibrosis of lungs were assessed by histologic, flow cytometric, and quantitative PCR analysis. AOC3-deficient mice showed a 30-50% reduction in fibrosis, collagen synthesis, numbers of myofibroblasts, and accumulation of CD4+ lymphocytes, NK T cells, macrophages, and type 2 innate lymphoid cells compared with wild-type control mice. AOC3-knock-in mice, which express a catalytically inactive form of AOC3, were also protected from lung fibrosis. In wild-type mice, a small-molecule AOC3 inhibitor treatment reduced leukocyte infiltration, myofibroblast differentiation, and fibrotic injury both in prophylactic and early therapeutic settings by about 50% but was unable to reverse the established fibrosis. AOC3 was also induced in myofibroblasts in human idiopathic pulmonary fibrosis. Thus, the oxidase activity of AOC3 contributes to the development of lung fibrosis mainly by regulating the accumulation of pathogenic leukocyte subtypes, which drive the fibrotic response.-Marttila-Ichihara, F., Elima, K., Auvinen, K., Veres, T. Z., Rantakari, P., Weston, C., Miyasaka, M., Adams, D., Jalkanen, S., Salmi, M. Amine oxidase activity regulates the development of pulmonary fibrosis.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Pulmonary Fibrosis/enzymology , Amine Oxidase (Copper-Containing)/genetics , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Carboxylic Acids , Cell Adhesion Molecules/genetics , Gene Expression Regulation, Enzymologic/physiology , Humans , Lung/enzymology , Lung/pathology , Lymphocytes/physiology , Mice , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pyrrolidines
17.
Am J Respir Cell Mol Biol ; 56(3): 362-371, 2017 03.
Article in English | MEDLINE | ID: mdl-27805412

ABSTRACT

Loss of extracellular superoxide dismutase 3 (SOD3) contributes to inflammatory and fibrotic lung diseases. The human SOD3 R213G polymorphism decreases matrix binding, redistributing SOD3 from the lung to extracellular fluids, and protects against LPS-induced alveolar inflammation. We used R213G mice expressing a naturally occurring single-nucleotide polymorphism, rs1799895, within the heparin-binding domain of SOD3, which results in an amino acid substitution at position 213 to test the hypothesis that the redistribution of SOD3 into the extracellular fluids would impart protection against bleomycin-induced lung fibrosis and secondary pulmonary hypertension (PH). In R213G mice, SOD3 content and activity was increased in extracellular fluids and decreased in lung at baseline, with greater increases in bronchoalveolar lavage fluid (BALF) SOD3 compared with wild-type mice 3 days after bleomycin. R213G mice developed less fibrosis based on pulmonary mechanics, fibrosis scoring, collagen quantification, and gene expression at 21 days, and less PH by right ventricular systolic pressure and pulmonary arteriole medial wall thickening at 28 days. In wild-type mice, macrophages, lymphocytes, neutrophils, proinflammatory cytokines, and protein increased in BALF on Day 7 and/or 21. In R213G mice, total BALF cell counts increased on Day 7 but resolved by 21 days. At 1 or 3 days, BALF pro- and antiinflammatory cytokines and BALF protein were higher in R213G mice, resolving by 21 days. We conclude that the redistribution of SOD3 as a result of the R213G single-nucleotide polymorphism protects mice from bleomycin-induced fibrosis and secondary PH by improved resolution of alveolar inflammation.


Subject(s)
Pneumonia/complications , Pneumonia/genetics , Polymorphism, Single Nucleotide/genetics , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/genetics , Superoxide Dismutase/genetics , Alveolar Epithelial Cells/metabolism , Animals , Bleomycin , Bronchoalveolar Lavage Fluid , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Mice, Inbred C57BL , Models, Biological , Pneumonia/blood , Pneumonia/enzymology , Pneumonia/physiopathology , Pulmonary Fibrosis/blood , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/physiopathology , Superoxide Dismutase/blood , Vascular Remodeling
18.
Am J Respir Cell Mol Biol ; 56(6): 772-783, 2017 06.
Article in English | MEDLINE | ID: mdl-28225294

ABSTRACT

Fibrosis is a major cause of progressive organ dysfunction in several chronic pulmonary diseases. Rho-associated coiled-coil forming kinase (ROCK) has been shown to be involved in myofibroblast differentiation driven by altered matrix stiffness in a fibrotic state. There are two known ROCK isoforms in humans, ROCK1 and ROCK2, but the specific role of each isoform in myofibroblast differentiation in lung fibrosis remains unknown. To study this, we developed a gelatin methacryloyl hydrogel-based culture system with different stiffness levels relevant to healthy and fibrotic lungs. We have shown that stiff matrix, but not soft matrix, can induce myofibroblast differentiation with high smooth muscle actin isoform (αSMA) expression. Furthermore, our data confirmed that the inhibition of ROCK signaling by a pharmacological inhibitor (i.e., Y27632) attenuates stiffness-induced αSMA expression and fiber assembly in myofibroblasts. To assess the role of ROCK isoforms in this process, we used short interfering RNA to knock down the expression of each isoform. Our data showed that knocking down either ROCK1 or ROCK2 did not result in a reduction in αSMA expression in myofibroblasts on stiff matrix, as opposed to soft matrix, where αSMA expression was reduced significantly. Paradoxically, on stiff matrix, the absence of one isoform (particularly ROCK2) exaggerated αSMA expression and led to thick fiber assembly. Moreover, complete loss of αSMA fiber assembly was seen only in the absence of both ROCK isoforms, suggesting that both isoforms are implicated in this process. Overall, our results indicate the differential role of ROCK isoforms in myofibroblast differentiation on soft and stiff matrices.


Subject(s)
Cell Differentiation , Myofibroblasts/enzymology , Myofibroblasts/pathology , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Stress, Mechanical , rho-Associated Kinases/metabolism , Actins/metabolism , Amides/pharmacology , Biomechanical Phenomena/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gelatin/pharmacology , Gene Silencing/drug effects , Humans , Hydrogels/pharmacology , Isoenzymes/metabolism , Methacrylates/pharmacology , Polymerization/drug effects , Pyridines/pharmacology , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , Substrate Specificity/drug effects , Tissue Scaffolds/chemistry , Trans-Activators/metabolism
19.
J Cell Mol Med ; 21(2): 324-335, 2017 02.
Article in English | MEDLINE | ID: mdl-27704718

ABSTRACT

Idiopathic pulmonary fibrosis is a severe disease characterized by excessive myofibroblast proliferation, extracellular matrix and fibrils deposition, remodelling of lung parenchyma and pulmonary insufficiency. Drugs able to reduce disease progression are available, but therapeutic results are unsatisfactory; new and safe treatments are urgently needed. Poly(ADP-ribose) polymerases-1 (PARP-1) is an abundant nuclear enzyme involved in key biological processes: DNA repair, gene expression control, and cell survival or death. In liver and heart, PARP-1 activity facilitates oxidative damage, collagen deposition and fibrosis development. In this study, we investigated the effects of HYDAMTIQ, a potent PARP-1 inhibitor, in a murine model of lung fibrosis. We evaluated the role of PARP on transforming growth factor-ß (TGF-ß) expression and TGF-ß/SMAD signalling pathway in lungs. Mice were intratracheally injected with bleomycin and then treated with either vehicle or different doses of HYDAMTIQ for 21 days. Airway resistance to inflation and lung static compliance, markers of lung stiffness, were assayed. Histochemical and biochemical parameters to evaluate TGF-ß/SMAD signalling pathway with alpha-smooth muscle actin (αSMA) deposition and the levels of a number of inflammatory markers (tumour necrosis factor-α, interleukin-1ß, iNOS and COX-2) were performed. Bleomycin administration increased lung stiffness. It also increased lung PARP activity, TGF-ß levels, pSMAD3 expression, αSMA deposition and content of inflammatory markers. HYDAMTIQ attenuated all the above-mentioned physiological, biochemical and histopathological markers. Our findings support the proposal that PARP inhibitors could have a therapeutic potential in reducing the progression of signs and symptoms of the disease by decreasing TGF-ß expression and the TGF-ß/SMAD transduction pathway.


Subject(s)
Isoquinolines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/enzymology , Signal Transduction/drug effects , Smad Proteins/metabolism , Thiophenes/therapeutic use , Transforming Growth Factor beta/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Bleomycin , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Hydroxyproline/metabolism , Inflammation Mediators/metabolism , Isoquinolines/pharmacology , Lung/pathology , Lung/physiopathology , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Pulmonary Fibrosis/chemically induced , Thiophenes/pharmacology
20.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L722-L730, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28283477

ABSTRACT

Progressive pulmonary fibrosis is a devastating consequence of many acute and chronic insults to the lung. Lung injury leads to alveolar epithelial cell (AEC) death, destruction of the basement membrane, and activation of transforming growth factor-ß (TGF-ß). There is subsequent resolution of the injury and a coordinated and concurrent initiation of fibrosis. Both of these processes may involve activation of similar intracellular signaling pathways regulated in part by dynamic changes to the extracellular matrix. Matrix signaling can augment the profibrotic fibroblast response to TGF-ß. However, similar matrix/integrin signaling pathways may also be involved in the inhibition of ongoing TGF-ß-induced AEC apoptosis. Focal adhesion kinase (FAK) is an integrin-associated signaling molecule expressed by many cell types. We used mice with AEC-specific FAK deletion to isolate the epithelial aspect of integrin signaling in the bleomycin model of lung injury and fibrosis. Mice with AEC-specific deletion of FAK did not exhibit spontaneous lung injury but did have significantly greater terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling-positive cells (18.6 vs. 7.1) per ×200 field, greater bronchoalveolar lavage protein (3.2 vs. 1.8 mg/ml), and significantly greater death (77 vs. 19%) after bleomycin injury compared with littermate control mice. Within primary AECs, activated FAK directly associates with caspase-8 and inhibits activation of the caspase cascade resulting in less apoptosis in response to TGF-ß. Our studies support a model in which dynamic changes to the extracellular matrix after injury promote fibroblast activation and inhibition of epithelial cell apoptosis in response to TGF-ß through FAK activation potentially complicating attempts to nonspecifically target this pathway for antifibrotic therapy.


Subject(s)
Epithelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lung Injury/enzymology , Lung/pathology , Pulmonary Fibrosis/enzymology , Signal Transduction , Animals , Apoptosis , Bleomycin , Caspase 8/metabolism , Enzyme Activation , Epithelial Cells/pathology , Gene Deletion , Lung Injury/complications , Lung Injury/pathology , Mice , Models, Biological , Organ Specificity , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Protein C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL