Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Am J Respir Cell Mol Biol ; 59(1): 77-86, 2018 07.
Article in English | MEDLINE | ID: mdl-29373068

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive aging-associated disease of unknown etiology. A growing body of evidence indicates that aberrant activated alveolar epithelial cells induce the expansion and activation of the fibroblast population, leading to the destruction of the lung architecture. Some matrix metalloproteinases (MMPs) are upregulated in IPF, indicating that they may be important in the pathogenesis and/or progression of IPF. In the present study, we examined the expression of MMP28 in this disease and evaluated its functional effects in two alveolar epithelial cell lines and in human primary bronchial epithelial cells. We found that the enzyme is expressed in bronchial (apical and cytoplasmic localization) and alveolar (cytoplasmic and nuclear localization) epithelial cells in two different groups of patients with IPF. In vitro MMP28 epithelial silencing decreased the proliferation rate and delayed wound closing, whereas overexpression showed opposite effects, protecting from apoptosis and enhanced epithelial-mesenchymal transition. Our findings demonstrate that MMP28 is upregulated in epithelial cells from IPF lungs, where it may play a role in increasing the proliferative and migratory phenotype in a catalysis-dependent manner.


Subject(s)
Cell Nucleus/metabolism , Epithelium/metabolism , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Matrix Metalloproteinases, Secreted/genetics , Pulmonary Alveoli/pathology , Up-Regulation/genetics , A549 Cells , Animals , Apoptosis , Biocatalysis , Cell Movement , Cell Proliferation , Cytoprotection , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Epithelium/pathology , Gene Silencing , Humans , Matrix Metalloproteinases, Secreted/metabolism , Protein Transport , Rats
15.
Am J Respir Cell Mol Biol ; 58(4): 461-470, 2018 04.
Article in English | MEDLINE | ID: mdl-29115860

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by excessive deposition of extracellular matrix (ECM) in the lung parenchyma. The abnormal ECM deposition slowly overtakes normal lung tissue, disturbing gas exchange and leading to respiratory failure and death. ECM cross-linking and subsequent stiffening is thought to be a major contributor of disease progression and also promotes the activation of transforming growth factor (TGF)-ß1, one of the main profibrotic growth factors. Lysyl oxidase-like (LOXL) 1 belongs to the cross-linking enzyme family and has been shown to be up-regulated in active fibrotic regions of bleomycin-treated mice and patients with IPF. We demonstrate in this study that LOXL1-deficient mice are protected from experimental lung fibrosis induced by overexpression of TGF-ß1 using adenoviral (Ad) gene transfer (AdTGF-ß1). The lack of LOXL1 prevented accumulation of insoluble cross-linked collagen in the lungs, and therefore limited lung stiffness after AdTGF-ß1. In addition, we applied mechanical stretch to lung slices from LOXL1+/+ and LOXL1-/- mice treated with AdTGF-ß1. Lung stiffness (Young's modulus) of LOXL1-/- lung slices was significantly lower compared with LOXL1+/+ lung slices. Moreover, the release of activated TGF-ß1 after mechanical stretch was significantly lower in LOXL1-/- mice compared with LOXL1+/+ mice after AdTGF-ß1. These data support the concept that cross-linking enzyme inhibition represents an interesting therapeutic target for drug development in IPF.


Subject(s)
Adenoviridae/genetics , Amino Acid Oxidoreductases/deficiency , Collagen/metabolism , Gene Transfer Techniques , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/enzymology , Transforming Growth Factor beta1/genetics , Adenoviridae/metabolism , Amino Acid Oxidoreductases/genetics , Animals , Disease Models, Animal , Elastic Modulus , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Lung/physiopathology , Lung Compliance , Mechanotransduction, Cellular , Mice, Knockout , Pulmonary Stretch Receptors/metabolism , Transforming Growth Factor beta1/biosynthesis , Up-Regulation
16.
Am J Respir Cell Mol Biol ; 58(4): 449-460, 2018 04.
Article in English | MEDLINE | ID: mdl-29053336

ABSTRACT

Selective repression of the antifibrotic gene CXCL10 contributes to tissue remodeling in idiopathic pulmonary fibrosis (IPF). We have previously reported that histone deacetylation and histone H3 lysine 9 (H3K9) methylation are involved in CXCL10 repression. In this study, we explored the role of H3K27 methylation and the interplay between the two histone lysine methyltransferases enhancer of zest homolog 2 (EZH2) and G9a in CXCL10 repression in IPF. By applying chromatin immunoprecipitation, Re-ChIP, and proximity ligation assays, we demonstrated that, like G9a-mediated H3K9 methylation, EZH2-mediated histone H3 lysine 27 trimethylation (H3K27me3) was significantly enriched at the CXCL10 promoter in fibroblasts from IPF lungs (F-IPF) compared with fibroblasts from nonfibrotic lungs, and we also found that EZH2 and G9a physically interacted with each other. EZH2 knockdown reduced not only EZH2 and H3K27me3 but also G9a and H3K9me3, and G9a knockdown reduced not only G9 and H3K9me3 but also EZH2 and H3K27me3. Depletion and inhibition of EZH2 and G9a also reversed histone deacetylation and restored CXCL10 expression in F-IPF. Furthermore, treatment of fibroblasts from nonfibrotic lungs with the profibrotic cytokine transforming growth factor-ß1 increased EZH2, G9a, H3K27me3, H3K9me3, and histone deacetylation at the CXCL10 promoter, similar to that observed in F-IPF, which was correlated with CXCL10 repression and was prevented by EZH2 and G9a knockdown. These findings suggest that a novel and functionally interdependent interplay between EZH2 and G9a regulates histone methylation-mediated epigenetic repression of the antifibrotic CXCL10 gene in IPF. This interdependent interplay may prove to be a target for epigenetic intervention to restore the expression of CXCL10 and other antifibrotic genes in IPF.


Subject(s)
Chemokine CXCL10/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Fibroblasts/enzymology , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , Case-Control Studies , Cells, Cultured , Chemokine CXCL10/genetics , DNA Methylation , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenetic Repression , Fibroblasts/drug effects , Fibroblasts/pathology , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/pathology , Promoter Regions, Genetic , Signal Transduction , Transforming Growth Factor beta1/pharmacology
17.
Am J Respir Cell Mol Biol ; 58(5): 585-593, 2018 05.
Article in English | MEDLINE | ID: mdl-29019702

ABSTRACT

Organ fibrosis, including idiopathic pulmonary fibrosis, is associated with significant morbidity and mortality. Because currently available therapies have limited effect, there is a need to better understand the mechanisms by which organ fibrosis occurs. We have recently reported that transforming growth factor (TGF)-ß, a key cytokine that promotes fibrogenesis, induces the expression of the enzymes of the de novo serine and glycine synthesis pathway in human lung fibroblasts, and that phosphoglycerate dehydrogenase (PHGDH; the first and rate-limiting enzyme of the pathway) is required to promote collagen protein synthesis downstream of TGF-ß. In this study, we investigated whether inhibition of de novo serine and glycine synthesis attenuates lung fibrosis in vivo. We found that TGF-ß induces mRNA and protein expression of PHGDH in murine fibroblasts. Similarly, intratracheal administration of bleomycin resulted in increased expression of PHGDH in mouse lungs, localized to fibrotic regions. Using a newly developed small molecule inhibitor of PHGDH (NCT-503), we tested whether pharmacologic inhibition of PHGDH could inhibit fibrogenesis both in vitro and in vivo. Treatment of murine and human lung fibroblasts with NCT-503 decreased TGF-ß-induced collagen protein synthesis. Mice treated with the PHGDH inhibitor beginning 7 days after intratracheal instillation of bleomycin had attenuation of lung fibrosis. These results indicate that the de novo serine and glycine synthesis pathway is necessary for TGF-ß-induced collagen synthesis and bleomycin-induced pulmonary fibrosis. PHGDH and other enzymes in the de novo serine and glycine synthesis pathway may be a therapeutic target for treatment of fibrotic diseases, including idiopathic pulmonary fibrosis.


Subject(s)
Airway Remodeling/drug effects , Bleomycin , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/drug effects , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Animals , Collagen/metabolism , Disease Models, Animal , Fibroblasts/enzymology , Fibroblasts/pathology , Glycine/metabolism , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Phosphoglycerate Dehydrogenase/metabolism , Serine/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology
18.
Respir Res ; 19(1): 24, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29409529

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal fibrotic disorder, with no curative therapies. The signal transducer and activator of transcription 3 (STAT3) protein is activated in lung fibroblasts and alveolar type II cells (ATII), thereby contributing to lung fibrosis in IPF. Although activation of Janus kinase 2 (JAK2) has been implicated in proliferative disorders, its role in IPF is unknown. The aim of this study was to analyze JAK2 activation in IPF, and to determine whether JAK2/STAT3 inhibition is a potential therapeutic strategy for this disease. METHODS AND RESULTS: JAK2/p-JAK2 and STAT3/pSTAT3 expression was evaluated using quantitative real time-PCR, western blotting, and immunohistochemistry. Compared to human healthy lung tissue (n = 10) both proteins were upregulated in the lung tissue of IPF patients (n = 12). Stimulating primary ATII and lung fibroblasts with transforming growth factor beta 1 or interleukin (IL)-6/IL-13 activated JAK2 and STAT3, inducing epithelial to mesenchymal and fibroblast to myofibroblast transitions. Dual p-JAK2/p-STAT3 inhibition with JSI-124 or silencing of JAK2 and STAT3 genes suppressed ATII and the fibroblast to myofibroblast transition, with greater effects than the sum of those obtained using JAK2 or STAT3 inhibitors individually. Dual rather than single inhibition was also more effective for inhibiting fibroblast migration, preventing increases in fibroblast senescence and Bcl-2 expression, and ameliorating impaired autophagy. In rats administered JSI-124, a dual inhibitor of p-JAK2/p-STAT3, at a dose of 1 mg/kg/day, bleomycin-induced lung fibrosis was reduced and collagen deposition in the lung was inhibited, as were JAK2 and STAT3 activation and several markers of fibrosis, autophagy, senescence, and anti-apoptosis. CONCLUSIONS: JAK2 and STAT3 are activated in IPF, and their dual inhibition may be an attractive strategy for treating this disease.


Subject(s)
Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , A549 Cells , Adult , Aged , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Janus Kinase 2/antagonists & inhibitors , Male , Middle Aged , Rats , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/pharmacology
19.
Lung ; 196(4): 393-400, 2018 08.
Article in English | MEDLINE | ID: mdl-29637273

ABSTRACT

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


Subject(s)
Adenosine Deaminase/metabolism , Fibroblasts/enzymology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/enzymology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Case-Control Studies , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation, Enzymologic , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/drug effects , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Primary Cell Culture , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Transforming Growth Factor beta1/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Pharmacol Rev ; 67(1): 103-17, 2015.
Article in English | MEDLINE | ID: mdl-25395505

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung scarring, short median survival, and limited therapeutic options, creating great need for new pharmacologic therapies. IPF is thought to result from repetitive environmental injury to the lung epithelium, in the context of aberrant host wound healing responses. Tissue responses to injury fundamentally involve reorganization of the actin cytoskeleton of participating cells, including epithelial cells, fibroblasts, endothelial cells, and macrophages. Actin filament assembly and actomyosin contraction are directed by the Rho-associated coiled-coil forming protein kinase (ROCK) family of serine/threonine kinases (ROCK1 and ROCK2). As would therefore be expected, lung ROCK activation has been demonstrated in humans with IPF and in animal models of this disease. ROCK inhibitors can prevent fibrosis in these models, and more importantly, induce the regression of already established fibrosis. Here we review ROCK structure and function, upstream activators and downstream targets of ROCKs in pulmonary fibrosis, contributions of ROCKs to profibrotic cellular responses to lung injury, ROCK inhibitors and their efficacy in animal models of pulmonary fibrosis, and potential toxicities of ROCK inhibitors in humans, as well as involvement of ROCKs in fibrosis in other organs. As we discuss, ROCK activation is required for multiple profibrotic responses, in the lung and multiple other organs, suggesting ROCK participation in fundamental pathways that contribute to the pathogenesis of a broad array of fibrotic diseases. Multiple lines of evidence therefore indicate that ROCK inhibition has great potential to be a powerful therapeutic tool in the treatment of fibrosis, both in the lung and beyond.


Subject(s)
Drug Design , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors , Animals , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Lung/enzymology , Lung/pathology , Protein Conformation , Protein Kinase Inhibitors/adverse effects , Structure-Activity Relationship , rho-Associated Kinases/chemistry , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL