Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Biomolecules ; 12(2)2022 02 09.
Article in English | MEDLINE | ID: mdl-35204783

ABSTRACT

Abnormalities in airway epithelia and lung parenchyma are found in Atp8b1 mutant mice, which develop pulmonary fibrosis after hyperoxic insult. Microarray and ingenuity pathway analysis (IPA) show numerous transcripts involved in ciliogenesis are downregulated in 14-month (14 M) -old Atp8b1 mouse lung compared with wild-type C57BL/6. Lung epithelium of Atp8b1 mice demonstrate apical abnormalities of ciliated and club cells in the bronchial epithelium on transmission electron microscopy (TEM). Matrix metalloproteinase 7 (MMP7) regulates of ciliogenesis and is a biomarker for idiopathic pulmonary fibrosis (IPF) in humans. Mmp7 transcript and protein expression are significantly upregulated in 14 M Atp8b1 mutant mouse lung. MMP7 expression is also increased in bronchoalveolar lavage fluid (BAL). Immunohistochemistry is localized MMP7 to bronchial epithelial cells in the Atp8b1 mutant. In conclusion, MMP7 is upregulated in the aged Atp8b1 mouse model, which displays abnormal ciliated cell and club cell morphology. This mouse model can facilitate the exploration of the role of MMP7 in epithelial integrity and ciliogenesis in IPF. The Atp8b1 mutant mouse is proposed as a model for IPF.


Subject(s)
Adenosine Triphosphatases , Idiopathic Pulmonary Fibrosis , Matrix Metalloproteinase 7 , Phospholipid Transfer Proteins , Adenosine Triphosphatases/metabolism , Animals , Bronchoalveolar Lavage Fluid , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Mice , Mice, Inbred C57BL , Phospholipid Transfer Proteins/metabolism
2.
Cells ; 10(11)2021 10 21.
Article in English | MEDLINE | ID: mdl-34831059

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive terminal lung disease, and therapies aim to block fibrosis. Fibroblast proliferation is controlled by C/EBP-ß, microRNA cluster 17-92 (miR17-92), and Erk1/2 mitogen-activated protein kinase. This study assessed the role of miR17-92 in IPF-fibroblast proliferation and its modification by treprostinil. Fibroblasts were isolated from eight IPF patients, five interstitial lung fibrosis patients, and seven control lungs. Fibroblasts were stimulated with TGF-ß1 over 24 h. The miR17-92 expression was analyzed by RT-qPCR, and protein expression by Western blotting. TGF-ß1 upregulated C/EBP-ß in all fibroblasts, which was reduced by treprostinil in control-fibroblasts, but not in IPF-fibroblasts. Compared to controls, the guide strands miR-19a-3p, miR-19b-3p, miR-20a-5p, and miR-92a-3p, as well as the passenger strands miR-17-3p, miR-18-3p, miR-19a-1-5p, and miR-92a-5p were significantly increased in IPF-fibroblasts. In controls, TGF-ß1 and treprostinil significantly reduced specific miR17-92 members. IPF-fibroblast proliferation was inhibited by treprostinil through increased expression of the Erk1/2 inhibitor DUSP1. These data suggest that proliferation control via miR17-92 and C/EBP-ß is disrupted in IPF-fibroblasts. Therefore, the inhibition of early stages of signaling cascades or specific mitogen receptors might be less effective. However, the increased proliferation is sensitive to Erk1/2 inhibition by treprostinil-induced DUSP1.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Epoprostenol/analogs & derivatives , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Epoprostenol/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Male , Middle Aged , Transforming Growth Factor beta1/metabolism
3.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34204949

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is one of the most symptomatic progressive fibrotic lung diseases, in which patients have an extremely poor prognosis. Therefore, understanding the precise molecular mechanisms underlying pulmonary fibrosis is necessary for the development of new therapeutic options. Stress-activated protein kinases (SAPKs), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38) are ubiquitously expressed in various types of cells and activated in response to cellular environmental stresses, including inflammatory and apoptotic stimuli. Type II alveolar epithelial cells, fibroblasts, and macrophages are known to participate in the progression of pulmonary fibrosis. SAPKs can control fibrogenesis by regulating the cellular processes and molecular functions in various types of lung cells (including cells of the epithelium, interstitial connective tissue, blood vessels, and hematopoietic and lymphoid tissue), all aspects of which remain to be elucidated. We recently reported that the stepwise elevation of intrinsic p38 signaling in the lungs is correlated with a worsening severity of bleomycin-induced fibrosis, indicating an importance of this pathway in the progression of pulmonary fibrosis. In addition, a transcriptome analysis of RNA-sequencing data from this unique model demonstrated that several lines of mechanisms are involved in the pathogenesis of pulmonary fibrosis, which provides a basis for further studies. Here, we review the accumulating evidence for the spatial and temporal roles of SAPKs in pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/genetics , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase 4/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Blood Vessels/enzymology , Blood Vessels/growth & development , Fibroblasts/enzymology , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Lung/embryology , Lung/pathology , MAP Kinase Signaling System/genetics , Macrophages/enzymology
4.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070506

ABSTRACT

Concentration of hyaluronic acid (HA) in the lungs increases in idiopathic pulmonary fibrosis (IPF). HA is involved in the organization of fibrin, fibronectin, and collagen. HA has been proposed to be a biomarker of fibrosis and a potential target for antifibrotic therapy. Hyaluronidase (HD) breaks down HA into fragments, but is a subject of rapid hydrolysis. A conjugate of poloxamer hyaluronidase (pHD) was prepared using protein immobilization with ionizing radiation. In a model of bleomycin-induced pulmonary fibrosis, pHD decreased the level of tissue IL-1ß and TGF-ß, prevented the infiltration of the lung parenchyma by CD16+ cells, and reduced perivascular and peribronchial inflammation. Simultaneously, a decrease in the concentrations of HA, hydroxyproline, collagen 1, total soluble collagen, and the area of connective tissue in the lungs was observed. The effects of pHD were significantly stronger compared to native HD which can be attributed to the higher stability of pHD. Additional spiperone administration increased the anti-inflammatory and antifibrotic effects of pHD and accelerated the regeneration of the damaged lung. The potentiating effects of spiperone can be explained by the disruption of the dopamine-induced mobilization and migration of fibroblast progenitor cells into the lungs and differentiation of lung mesenchymal stem cells (MSC) into cells of stromal lines. Thus, a combination of pHD and spiperone may represent a promising approach for the treatment of IPF and lung regeneration.


Subject(s)
Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Lung/drug effects , Spiperone/pharmacology , Animals , Cell Differentiation/drug effects , Collagen Type I/metabolism , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/pharmacokinetics , Hydroxyproline/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/enzymology , Inflammation/metabolism , Interleukin-1beta/metabolism , Keratins/metabolism , Lung/enzymology , Lung/metabolism , Lung/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Poloxamer/chemistry , Receptors, IgG/metabolism , Spiperone/administration & dosage , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
J Biol Chem ; 297(1): 100810, 2021 07.
Article in English | MEDLINE | ID: mdl-34023385

ABSTRACT

Pulmonary fibrosis is a progressive lung disease often occurring secondary to environmental exposure. Asbestos exposure is an important environmental mediator of lung fibrosis and remains a significant cause of disease despite strict regulations to limit exposure. Lung macrophages play an integral role in the pathogenesis of fibrosis induced by asbestos (asbestosis), in part by generating reactive oxygen species (ROS) and promoting resistance to apoptosis. However, the mechanism by which macrophages acquire apoptosis resistance is not known. Here, we confirm that macrophages isolated from asbestosis subjects are resistant to apoptosis and show they are associated with enhanced mitochondrial content of NADPH oxidase 4 (NOX4), which generates mitochondrial ROS generation. Similar results were seen in chrysotile-exposed WT mice, while macrophages from Nox4-/- mice showed increased apoptosis. NOX4 regulated apoptosis resistance by activating Akt1-mediated Bcl-2-associated death phosphorylation. Demonstrating the importance of NOX4-mediated apoptosis resistance in fibrotic remodeling, mice harboring a conditional deletion of Nox4 in monocyte-derived macrophages exhibited increased apoptosis and were protected from pulmonary fibrosis. Moreover, resolution occurred when Nox4 was deleted in monocyte-derived macrophages in mice with established fibrosis. These observations suggest that NOX4 regulates apoptosis resistance in monocyte-derived macrophages and contributes to the pathogenesis of pulmonary fibrosis. Targeting NOX4-mediated apoptosis resistance in monocyte-derived macrophages may provide a novel therapeutic target to protect against the development and/or progression of pulmonary fibrosis.


Subject(s)
Apoptosis , Disease Progression , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Macrophages/enzymology , Macrophages/pathology , NADPH Oxidase 4/metabolism , Animals , Cell Line , Female , Lung/pathology , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Biological , Monocytes/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , bcl-Associated Death Protein/metabolism
6.
Curr Opin Pulm Med ; 27(1): 54-60, 2021 01.
Article in English | MEDLINE | ID: mdl-33065600

ABSTRACT

PURPOSE OF REVIEW: Matrix metalloproteinases (MMPs) are a family of over 20 zinc-dependent proteases with different biological and pathological activities, and many have been implicated in several diseases. Although nonselective MMP inhibitors are known to induce serious side-effects, targeting individual MMPs may offer a safer therapeutic potential for several diseases. Hence, we provide a concise overview on MMP-12, given its association with pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis, and other progressive pulmonary fibrosis (PPF), which may also occur in coronavirus disease 2019. RECENT FINDINGS: In asthma, COPD, and PPF, increased MMP-12 levels have been associated with inflammation and/or structural changes within the lungs and negatively correlated with functional parameters. Increased pulmonary MMP-12 levels and MMP-12 gene expression have been related to disease severity in asthma and COPD. Targeting MMP-12 showed potential in animal models of pulmonary diseases but human data are still very scarce. SUMMARY: Although there may be a potential role of MMP-12 in asthma, COPD and PPF, several pathophysiological aspects await elucidation. Targeting MMP-12 may provide further insights into MMP-12 related mechanisms and how this translates into clinical outcomes; this warrants further research.


Subject(s)
Asthma/enzymology , COVID-19/enzymology , Idiopathic Pulmonary Fibrosis/enzymology , Matrix Metalloproteinase 12/metabolism , Pulmonary Disease, Chronic Obstructive/enzymology , Animals , Asthma/drug therapy , Asthma/etiology , Asthma/physiopathology , Biomarkers/metabolism , COVID-19/etiology , COVID-19/physiopathology , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/physiopathology , Matrix Metalloproteinase Inhibitors/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/physiopathology , COVID-19 Drug Treatment
7.
Int J Mol Sci ; 21(19)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977539

ABSTRACT

Autotaxin (ATX) is a secreted glycoprotein, widely present in biological fluids, largely responsible for extracellular lysophosphatidic acid (LPA) production. LPA is a bioactive growth-factor-like lysophospholipid that exerts pleiotropic effects in almost all cell types, exerted through at least six G-protein-coupled receptors (LPAR1-6). Increased ATX expression has been detected in different chronic inflammatory diseases, while genetic or pharmacological studies have established ATX as a promising therapeutic target, exemplified by the ongoing phase III clinical trial for idiopathic pulmonary fibrosis. In this report, we employed an in silico drug discovery workflow, aiming at the identification of structurally novel series of ATX inhibitors that would be amenable to further optimization. Towards this end, a virtual screening protocol was applied involving the search into molecular databases for new small molecules potentially binding to ATX. The crystal structure of ATX in complex with a known inhibitor (HA-155) was used as a molecular model docking reference, yielding a priority list of 30 small molecule ATX inhibitors, validated by a well-established enzymatic assay of ATX activity. The two most potent, novel and structurally different compounds were further structurally optimized by deploying further in silico tools, resulting to the overall identification of six new ATX inhibitors that belong to distinct chemical classes than existing inhibitors, expanding the arsenal of chemical scaffolds and allowing further rational design.


Subject(s)
Databases, Protein , Enzyme Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Small Molecule Libraries , Animals , Chronic Disease , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/enzymology , Inflammation/drug therapy , Inflammation/enzymology , Structure-Activity Relationship
8.
Sci Rep ; 10(1): 11657, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669620

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by interstitial remodeling and pulmonary dysfunction. The etiology of IPF is not completely understood but involves pathologic inflammation and subsequent failure to resolve fibrosis in response to epithelial injury. Treatments for IPF are limited to anti-inflammatory and immunomodulatory agents, which are only partially effective. Prostaglandin E2 (PGE2) disrupts TGFß signaling and suppresses myofibroblast differentiation, however practical strategies to raise tissue PGE2 during IPF have been limited. We previously described the discovery of a small molecule, (+)SW033291, that binds with high affinity to the PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) and increases PGE2 levels. Here we evaluated pulmonary 15-PGDH expression and activity and tested whether pharmacologic 15-PGDH inhibition (PGDHi) is protective in a mouse model of bleomycin-induced pulmonary fibrosis (PF). Long-term PGDHi was well-tolerated, reduced the severity of pulmonary fibrotic lesions and extracellular matrix remodeling, and improved pulmonary function in bleomycin-treated mice. Moreover, PGDHi attenuated both acute inflammation and weight loss, and decreased mortality. Endothelial cells and macrophages are likely targets as these cell types highly expressed 15-PGDH. In conclusion, PGDHi ameliorates inflammatory pathology and fibrosis in murine PF, and may have clinical utility to treat human disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Idiopathic Pulmonary Fibrosis/drug therapy , Pyridines/pharmacology , Thiophenes/pharmacology , Animals , Bleomycin/administration & dosage , Body Weight/drug effects , Dinoprostone/agonists , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Extracellular Matrix/drug effects , Extracellular Matrix/enzymology , Female , Gene Expression , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/mortality , Inflammation , Lung/drug effects , Lung/enzymology , Lung/pathology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Respiratory Function Tests , Survival Analysis
9.
Am J Respir Cell Mol Biol ; 62(6): 699-708, 2020 06.
Article in English | MEDLINE | ID: mdl-32208980

ABSTRACT

The mTOR pathway is one of the key signal cascades in the pathogenesis of idiopathic pulmonary fibrosis. Previous studies have mainly focused on this pathway in the fibroblasts and/or myofibroblasts, but not in the epithelial cells. In this study, we sought to investigate the role of the mTOR pathway in lung epithelial cells in lung fibrosis. Using Sftpc-mTORSL1+IT transgenic mice, in which active mTOR is conditionally expressed in lung epithelial cells, we assessed the effects of chronically activated mTOR in lung epithelial cells on lung phenotypes as well as bleomycin-induced lung fibrosis. Furthermore, we isolated alveolar epithelial cell type 2 from mice and performed RNA sequencing. Sftpc-mTORSL1+IT transgenic mice had no obvious abnormal findings, but, after bleomycin administration, showed more severe fibrotic changes and lower lung compliance than control mice. RNA sequencing revealed Angptl4 (angiopoietin-like protein 4) as a candidate downstream gene of the mTOR pathway. In vitro studies revealed that ANGPTL4, as well as mTOR, promoted tight junction vulnerability and epithelial-mesenchymal transition. mTOR activation in lung epithelial cells promoted lung fibrosis and the expression of ANGPTL4, a novel downstream target of the mTOR pathway, which could be related to the etiology of fibrosis.


Subject(s)
Alveolar Epithelial Cells/enzymology , Epithelial-Mesenchymal Transition/physiology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , TOR Serine-Threonine Kinases/physiology , A549 Cells , Alveolar Epithelial Cells/pathology , Angiopoietin-Like Protein 4/biosynthesis , Angiopoietin-Like Protein 4/genetics , Animals , Bleomycin/toxicity , Caveolin 1/biosynthesis , Caveolin 1/genetics , Enzyme Activation , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Male , Mice , Mice, Transgenic , Phenotype , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-1 Protein/genetics
10.
Clin Sci (Lond) ; 134(7): 889-905, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32219338

ABSTRACT

Senescence and mitochondrial stress are mutually reinforcing age-related processes that contribute to idiopathic pulmonary fibrosis (IPF); a lethal disease that manifests primarily in the elderly. Whilst evidence is accumulating that GMP-AMP synthase (cGAS) is crucial in perpetuating senescence by binding damaged DNA released into the cytosol, its role in IPF is not known. The present study examines the contributions of cGAS and self DNA to the senescence of lung fibroblasts from IPF patients (IPF-LFs) and age-matched controls (Ctrl-LFs). cGAS immunoreactivity was observed in regions of fibrosis associated with fibroblasts in lung tissue of IPF patients. Pharmacological inhibition of cGAS or its knockdown by silencing RNA (siRNA) diminished the escalation of IPF-LF senescence in culture over 7 days as measured by decreased p21 and p16 expression, histone 2AXγ phosphorylation and/or IL-6 production (P < 0.05, n = 5-8). The targeting of cGAS also attenuated etoposide-induced senescence in Ctrl-LFs (P < 0.05, n = 5-8). Levels of mitochondrial DNA (mDNA) detected by qPCR in the cytosol and medium of IPF-LFs or senescence-induced Ctrl-LFs were higher than Ctrl-LFs at baseline (P < 0.05, n = 5-7). The addition of DNAse I (100 U/ml) deaccelerated IPF-LF senescence (P < 0.05, n = 5), whereas ectopic mDNA or the induction of endogenous mDNA release augmented Ctrl-LF senescence in a cGAS-dependent manner (P < 0.05, n = 5). In conclusion, we provide evidence that cGAS reinforces lung fibroblast senescence involving damaged self DNA. The targeting of cGAS to supress senescent-like responses may have potential important therapeutic implications in the treatment of IPF.


Subject(s)
Cell Proliferation , Cellular Senescence , DNA, Mitochondrial/metabolism , Fibroblasts/enzymology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , Nucleotidyltransferases/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Histones/metabolism , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/drug effects , Lung/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/genetics , Paracrine Communication , Phosphorylation , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
11.
J Allergy Clin Immunol ; 145(3): 818-833.e11, 2020 03.
Article in English | MEDLINE | ID: mdl-31812575

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a disease with high 5-year mortality and few therapeutic options. Prostaglandin (PG) E2 exhibits antifibrotic properties and is reduced in bronchoalveolar lavage from patients with IPF. 15-Prostaglandin dehydrogenase (15-PGDH) is the key enzyme in PGE2 metabolism under the control of TGF-ß and microRNA 218. OBJECTIVE: We sought to investigate the expression of 15-PGDH in IPF and the therapeutic potential of a specific inhibitor of this enzyme in a mouse model and human tissue. METHODS: In vitro studies, including fibrocyte differentiation, regulation of 15-PGDH, RT-PCR, and Western blot, were performed using peripheral blood from healthy donors and patients with IPF and A549 cells. Immunohistochemistry, immunofluorescence, 15-PGDH activity assays, and in situ hybridization as well as ex vivo IPF tissue culture experiments were done using healthy donor and IPF lungs. Therapeutic effects of 15-PGDH inhibition were studied in the bleomycin mouse model of pulmonary fibrosis. RESULTS: We demonstrate that 15-PGDH shows areas of increased expression in patients with IPF. Inhibition of this enzyme increases PGE2 levels and reduces collagen production in IPF precision cut lung slices and in the bleomycin model. Inhibitor-treated mice show amelioration of lung function, decreased alveolar epithelial cell apoptosis, and fibroblast proliferation. Pulmonary fibrocyte accumulation is also decreased by inhibitor treatment in mice, similar to PGE2 that inhibits fibrocyte differentiation from blood of healthy donors and patients with IPF. Finally, microRNA 218-5p, which is downregulated in patients with IPF, suppressed 15-PGDH expression in vivo and in vitro. CONCLUSIONS: These findings highlight the role of 15-PGDH in IPF and suggest 15-PGDH inhibition as a promising therapeutic approach.


Subject(s)
Hydroxyprostaglandin Dehydrogenases/metabolism , Idiopathic Pulmonary Fibrosis/enzymology , MicroRNAs/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Dinoprostone/metabolism , Eicosanoids/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/pathology , Mice , Pyridines/pharmacology , Thiophenes/pharmacology
12.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31868203

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal and chronic disease with a high rate of infection and mortality; however, its etiology and pathogenesis remain unclear. Studies have revealed that epithelial-mesenchymal transition (EMT) is a crucial cellular event in IPF. Here, we identified that the pulmonary fibrosis inducer bleomycin simultaneously increased the expression of bFGF and TGF-ß1 and inhibited epithelial-specific regulatory protein (ESRP1) expression in vivo and in vitro. In addition, in vitro experiments showed that bFGF and TGF-ß1 down-regulated the expression of ESRP1 and that silencing ESRP1 promoted EMT in A549 cells. Notably, we determined that bFGF activates PI3K/Akt signaling, and treatment with the PI3K/Akt inhibitor LY294002 inhibited bleomycin-induced cell morphology changes and EMT. In addition, the effects of LY294002 on bleomycin-induced EMT were inhibited by ESRP1 silencing in A549 cells. Taken together, these findings suggest that bleomycin induced EMT through down-regulating ESRP1 by simultaneously increasing bFGF and TGF-ß1 in pulmonary fibrosis. Additionally, our findings indicated that bFGF inhibits ESRP1 by activating PI3K/Akt signaling.


Subject(s)
Bleomycin , Epithelial-Mesenchymal Transition , Fibroblast Growth Factor 2/metabolism , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , Phosphatidylinositol 3-Kinase/metabolism , RNA-Binding Proteins/metabolism , A549 Cells , Animals , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/pathology , Male , Mice , RNA-Binding Proteins/genetics , Signal Transduction , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
13.
Anal Chem ; 91(23): 14873-14878, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31670506

ABSTRACT

Fibroblast activation protein-α (FAP), as a crucial member of cell surface glycoprotein, highly expresses in reactive fibroblasts of tumors and several fibrosis diseases. It is a potential target for drug design and also reported as a prodrug strategy to increase the therapeutic window of some anticancer agents. In this work, we developed the first bioluminogenic probe for FAP with a limit-of-detection of 0.254 ng/mL, which could be applied to evaluate the FAP inhibitors in vitro. The experiments of transgenic mice and tumor-bearing nude mice validated our probe 1 could reflect the endogenous FAP level in vivo. Furthermore, this probe was successfully used to reflect FAP up-regulation in the lung homogenates of the bleomycin-induced idiopathic pulmonary fibrosis mice.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/diagnostic imaging , Diagnostic Imaging/methods , Gelatinases/genetics , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Membrane Proteins/genetics , Molecular Probes/pharmacokinetics , Serine Endopeptidases/genetics , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Bleomycin/administration & dosage , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Endopeptidases , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Gelatinases/antagonists & inhibitors , Gelatinases/metabolism , Gene Expression , Heterografts , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Limit of Detection , Luminescent Measurements , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Nude , Mice, Transgenic , Molecular Probes/chemical synthesis , Serine Endopeptidases/metabolism
14.
Genes Dev ; 33(19-20): 1381-1396, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31488579

ABSTRACT

Short telomere syndromes manifest as familial idiopathic pulmonary fibrosis; they are the most common premature aging disorders. We used genome-wide linkage to identify heterozygous loss of function of ZCCHC8, a zinc-knuckle containing protein, as a cause of autosomal dominant pulmonary fibrosis. ZCCHC8 associated with TR and was required for telomerase function. In ZCCHC8 knockout cells and in mutation carriers, genomically extended telomerase RNA (TR) accumulated at the expense of mature TR, consistent with a role for ZCCHC8 in mediating TR 3' end targeting to the nuclear RNA exosome. We generated Zcchc8-null mice and found that heterozygotes, similar to human mutation carriers, had TR insufficiency but an otherwise preserved transcriptome. In contrast, Zcchc8-/- mice developed progressive and fatal neurodevelopmental pathology with features of a ciliopathy. The Zcchc8-/- brain transcriptome was highly dysregulated, showing accumulation and 3' end misprocessing of other low-abundance RNAs, including those encoding cilia components as well as the intronless replication-dependent histones. Our data identify a novel cause of human short telomere syndromes-familial pulmonary fibrosis and uncover nuclear exosome targeting as an essential 3' end maturation mechanism that vertebrate TR shares with replication-dependent histones.


Subject(s)
Carrier Proteins/genetics , Idiopathic Pulmonary Fibrosis/genetics , Loss of Function Mutation , Nuclear Proteins/genetics , RNA/metabolism , Telomerase/metabolism , Animals , Brain/enzymology , Brain/physiopathology , Cell Line , Cilia/genetics , Female , Genetic Linkage , HCT116 Cells , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/physiopathology , Male , Mice , Mice, Knockout , Neurodevelopmental Disorders/genetics , Pedigree , RNA Processing, Post-Transcriptional/genetics , Telomere Shortening/genetics
15.
Clin Sci (Lond) ; 133(12): 1309-1320, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31217321

ABSTRACT

Nintedanib, a Food and Drug Administration-approved drug for the treatment of patients with idiopathic pulmonary fibrosis (IPK), inhibits both tyrosine kinase receptors and non-receptor kinases, and block activation of platelet-derived growth factor receptors, fibroblast growth factor receptor, vascular endothelial growth factor receptors, and Src family kinases. Preclinical and clinical studies have revealed the potent anti-fibrotic effect of nintedanib in IPK in human and animal models. Recent preclinical studies have also demonstrated the inhibitory effect of nintedanib on the development and progression of tissue fibrosis in other organs, including liver, kidney, and skin. The anti-fibrotic actions of nintedanib occur through a number of mechanisms, including blocking differentiation of fibroblasts to myofibroblasts, inhibition of epithelial-mesenchymal transition, and suppression of inflammation and angiogenesis. In this article, we summarize the mechanisms and efficacy of nintedanib in the treatment of fibrotic diseases in animal models and clinical trials, provide an update on recent advances in the development of other novel antifibrotic agents in preclinical and clinical study, and offer our perspective about the possible clinical application of these agents in fibrotic diseases.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/therapeutic use , Kidney Diseases/drug therapy , Kidney/drug effects , Liver Cirrhosis/drug therapy , Liver/drug effects , Lung/drug effects , Protein Kinase Inhibitors/therapeutic use , Animals , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Indoles/adverse effects , Kidney/enzymology , Kidney/pathology , Kidney Diseases/enzymology , Kidney Diseases/pathology , Liver/enzymology , Liver/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Lung/enzymology , Lung/pathology , Molecular Targeted Therapy , Protein Kinase Inhibitors/adverse effects , Signal Transduction
16.
Int J Mol Med ; 44(1): 273-280, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115510

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by progressive lung scarring and excessive extracellular matrix depositon. When stimulated, alveolar epithelial cells (AECs) are aberrantly activated, the expression of profibrotic molecules is enhanced, and lung fibrosis is promoted, but the mechanism for this is unclear. It has been reported that a downregulation of the Na,K­ATPase ß1 subunit in renal epithelial cells is involved in renal fibrosis development, but the role of this protein in lung fibrosis remains unknown. In the present study, the expression of the Na,K­ATPase ß1 subunit was revealed to be markedly decreased in AECs of patients with IPF and a bleomycin­induced pulmonary fibrosis mouse model. Treatment with transforming growth factor ß­1 led to significantly downregulation of the Na,K­ATPase ß1 subunit in lung adenocarcioma A549 cells. Furthermore, the knockdown of the Na,K­ATPase ß1 subunit in A549 cells resulted in the upregulation of profibrotic molecules, activation of the neurogenic locus notch homolog protein 1 and extracellular signal­regulated kinase 1/2 signaling pathways and induction of endoplasmic reticulum stress. These findings reveal that the downregulation of the Na,K­ATPase ß1 subunit enhances the expression of profibrotic molecules in AECs and may contribute to IPF pathogenesis.


Subject(s)
Alveolar Epithelial Cells/enzymology , Down-Regulation , Gene Expression Regulation, Enzymologic , Idiopathic Pulmonary Fibrosis/enzymology , MAP Kinase Signaling System , Sodium-Potassium-Exchanging ATPase/biosynthesis , A549 Cells , Adult , Alveolar Epithelial Cells/pathology , Animals , Endoplasmic Reticulum Stress , Humans , Idiopathic Pulmonary Fibrosis/pathology , Male , Mice , Middle Aged
17.
Int J Cancer ; 145(11): 3064-3077, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31032902

ABSTRACT

Myofibroblasts are a population of highly contractile fibroblasts that express and require the activity of the transcription factor Snail1. Cancer-associated fibroblasts (CAFs) correlate with low survival of cancer patients when present in the stroma of primary tumors. Remarkably, the presence of myofibroblastic CAFs (which express Snail1) creates mechanical properties in the tumor microenvironment that support metastasis. However, therapeutic blockage of fibroblast activity in patients with cancer is a double-edged sword, as normal fibroblast activities often restrict tumor cell invasion. We used fibroblasts depleted of Snail1 or protein arginine methyltransferases 1 and 4 (PRMT1/-4) to identify specific epigenetic modifications induced by TGFß/Snail1. Furthermore, we analyzed the in vivo efficiency of methyltransferase inhibitors using mouse models of wound healing and metastasis, as well as fibroblasts isolated from patients with idiopathic pulmonary fibrosis (IPF). Mechanistically, TGFß-induced Snail1 promotes the epigenetic mark of asymmetrically dimethylated arginine. Critically, we found that inhibitors of methyltransferases prevent myofibroblast activity (but not regular fibroblast activity) in the extracellular matrix, both in cell culture and in vivo. In a mouse breast cancer model, the inhibitor sinefungin reduces both the myofibroblast activity in the tumor stroma and the metastatic burden in the lung. Two distinct inhibitors effectively blocked the exacerbated myofibroblast activity of patient-derived IPF fibroblasts. Our data reveal epigenetic regulation of myofibroblast transdifferentiation in both wound healing and in disease (fibrosis and breast cancer). Thus, methyltransferase inhibitors are good candidates as therapeutic reagents for these diseases.


Subject(s)
Breast Neoplasms/drug therapy , Enzyme Inhibitors/administration & dosage , Idiopathic Pulmonary Fibrosis/drug therapy , Lung Neoplasms/secondary , Methyltransferases/antagonists & inhibitors , Myofibroblasts/drug effects , Snail Family Transcription Factors/genetics , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Breast Neoplasms/enzymology , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Female , Gene Deletion , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Methyltransferases/genetics , Mice , Myofibroblasts/cytology , Myofibroblasts/enzymology , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
18.
Biomed Pharmacother ; 112: 108669, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30784938

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal lung disease with a poor prognosis and limited treatment options. The incidence of IPF increases with age, and the mechanisms related to aging such as cellular senescence have been strongly implicated in disease pathology. Therefore, a better understanding of fibroblasts senescence might provide a new therapeutic strategy to prevent and treat pulmonary fibrosis. In this study, we aimed to explore the effects of citrus alkaline extracts (CAE) on the fibroblasts senescence, and elucidate the underlying mechanism to ameliorate pulmonary fibrosis. We demonstrated that CAE mitigated the collagen deposition by the initial early treatment, suggesting a potential preventive effect of CAE on pulmonary fibrosis. The expression of senescence biomarkers P16INK4a and P21, concomitant with down-regulation of the myofibroblasts marker α-SMA, and the number of senescence-associated ß-galactosidase (SA-ß-Gal) positive cells were decreased by CAE treatment, indicating a significant inhibitory effect of CAE on fibroblast senescence. Additionally, CAE down-regulated the expression of the senescence-associated secretory phenotype (SASP) in etoposide-induced senescent fibroblasts. Further studies indicated that COX-2 activation was required for CAE to inhibit the lung fibroblast senescence through a P53-dependent pathway. Results showed that the anti-senescence effect of CAE was abrogated when COX-2 was knocked down or inhibited by COX-2 inhibitor NS-398 or indomethacin in lung fibroblasts. Meanwhile, the anti-fibrotic and anti-senescence effect of CAE were abolished due to disruption of COX-2 in vivo. Collectively, our results provided a novel insight into the potential mechanism of CAE to inhibit the fibroblasts activation through preventing cellular senescence.


Subject(s)
Cellular Senescence/drug effects , Citrus , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Plant Extracts/therapeutic use , Animals , Cells, Cultured , Cellular Senescence/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibroblasts/enzymology , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Mice , Mice, Inbred C57BL , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Random Allocation
19.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L175-L186, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30358439

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fibroproliferative lung disease, and fibroblast-myofibroblast differentiation (FMD) is thought to be a key event in the pathogenesis of IPF. Histone deacetylase-8 (HDAC8) has been shown to associate with α-smooth muscle actin (α-SMA; a marker of FMD) and regulates cell contractility in vascular smooth muscle cells. However, the role of HDAC8 in FMD or pulmonary fibrosis has never been reported. This study investigated the role of HDAC8 in pulmonary fibrosis with a focus on FMD. We observed that HDAC8 expression was increased in IPF lung tissue as well as transforming growth factor (TGF)ß1-treated normal human lung fibroblasts (NHLFs). Immunoprecipitation experiments revealed that HDAC8 was associated with α-SMA in TGFß1-treated NHLFs. HDAC8 inhibition with NCC170 (HDAC8-selective inhibitor) repressed TGFß1-induced fibroblast contraction and α-SMA protein expression in NHLFs cultured in collagen gels. HDAC8 inhibition with HDAC8 siRNA also repressed TGFß1-induced expression of profibrotic molecules such as fibronectin and increased expression of antifibrotic molecules such as peroxisome proliferator-activated receptor-γ (PPARγ). Chromatin immunoprecipitation quantitative PCR using an antibody against H3K27ac (histone H3 acetylated at lysine 27; a known HDAC8 substrate and a marker for active enhancers) suggested that HDAC8 inhibition with NCC170 ameliorated TGFß1-induced loss of H3K27ac at the PPARγ gene enhancer. Furthermore, NCC170 treatment significantly decreased fibrosis measured by Ashcroft score as well as expression of type 1 collagen and fibronectin in bleomycin-treated mouse lungs. These data suggest that HDAC8 contributes to pulmonary fibrosis and that there is a therapeutic potential for HDAC8 inhibitors to treat IPF as well as other fibrotic lung diseases.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Myofibroblasts/enzymology , Repressor Proteins/antagonists & inhibitors , Acetylation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylases/biosynthesis , Histones/metabolism , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myofibroblasts/pathology , PPAR gamma/metabolism , Repressor Proteins/biosynthesis , Transforming Growth Factor beta1/metabolism
20.
Mol Biol Cell ; 29(18): 2165-2175, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29995590

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an incurable disease of the lung that is characterized by excessive deposition of extracellular matrix (ECM), resulting in disruption of normal lung function. The signals regulating fibrosis include both transforming growth factor beta (TGF-ß) and tissue rigidity and a major signaling pathway implicated in fibrosis involves activation of the GTPase RhoA. During studies exploring how elevated RhoA activity is sustained in IPF, we discovered that not only is RhoA activated by profibrotic stimuli but also that the expression of Rnd3, a major antagonist of RhoA activity, and the activity of p190RhoGAP (p190), a Rnd3 effector, are both suppressed in IPF fibroblasts. Restoration of Rnd3 levels in IPF fibroblasts results in an increase in p190 activity, a decrease in RhoA activity and a decrease in the overall fibrotic phenotype. We also find that treatment with IPF drugs nintedanib and pirfenidone decreases the fibrotic phenotype and RhoA activity through up-regulation of Rnd3 expression and p190 activity. These data provide evidence for a pathway in IPF where fibroblasts down-regulate Rnd3 levels and p190 activity to enhance RhoA activity and drive the fibrotic phenotype.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line , Down-Regulation , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Indoles/pharmacology , Phenotype , Pyridones/pharmacology , Signal Transduction , Transforming Growth Factor beta , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...