Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Lancet Respir Med ; 11(6): 550-562, 2023 06.
Article in English | MEDLINE | ID: mdl-36842446

ABSTRACT

BACKGROUND: Elexacaftor-tezacaftor-ivacaftor has been shown to be safe and efficacious in people with cystic fibrosis and at least one F508del allele. Our aim was to identify a novel cystic fibrosis transmembrane conductance regulator (CFTR) modulator combination capable of further increasing CFTR-mediated chloride transport, with the potential for once-daily dosing. METHODS: We conducted two phase 2 clinical trials to assess the safety and efficacy of a once-daily combination of vanzacaftor-tezacaftor-deutivacaftor in participants with cystic fibrosis who were aged 18 years or older. A phase 2 randomised, double-blind, active-controlled study (VX18-561-101; April 17, 2019, to Aug 20, 2020) was carried out to compare deutivacaftor monotherapy with ivacaftor monotherapy in participants with CFTR gating mutations, following a 4-week ivacaftor monotherapy run-in period. Participants were randomly assigned to receive either ivacaftor 150 mg every 12 h, deutivacaftor 25 mg once daily, deutivacaftor 50 mg once daily, deutivacaftor 150 mg once daily, or deutivacaftor 250 mg once daily in a 1:1:2:2:2 ratio. The primary endpoint was absolute change in ppFEV1 from baseline at week 12. A phase 2 randomised, double-blind, controlled, proof-of-concept study of vanzacaftor-tezacaftor-deutivacaftor (VX18-121-101; April 30, 2019, to Dec 10, 2019) was conducted in participants with cystic fibrosis and heterozygous for F508del and a minimal function mutation (F/MF genotypes) or homozygous for F508del (F/F genotype). Participants with F/MF genotypes were randomly assigned 1:2:2:1 to receive either 5 mg, 10 mg, or 20 mg of vanzacaftor in combination with tezacaftor-deutivacaftor or a triple placebo for 4 weeks, and participants with the F/F genotype were randomly assigned 2:1 to receive either vanzacaftor (20 mg)-tezacaftor-deutivacaftor or tezacaftor-ivacaftor active control for 4 weeks, following a 4-week tezacaftor-ivacaftor run-in period. Primary endpoints for part 1 and part 2 were safety and tolerability and absolute change in ppFEV1 from baseline to day 29. Secondary efficacy endpoints were absolute change from baseline at day 29 in sweat chloride concentrations and Cystic Fibrosis Questionnaire-Revised (CFQ-R) respiratory domain score. These clinical trials are registered with ClinicalTrials.gov, NCT03911713 and NCT03912233, and are complete. FINDINGS: In study VX18-561-101, participants treated with deutivacaftor 150 mg once daily (n=23) or deutivacaftor 250 mg once daily (n=24) had mean absolute changes in ppFEV1 of 3·1 percentage points (95% CI -0·8 to 7·0) and 2·7 percentage points (-1·0 to 6·5) from baseline at week 12, respectively, versus -0·8 percentage points (-6·2 to 4·7) with ivacaftor 150 mg every 12 h (n=11); the deutivacaftor safety profile was consistent with the established safety profile of ivacaftor 150 mg every 12 h. In study VX18-121-101, participants with F/MF genotypes treated with vanzacaftor (5 mg)-tezacaftor-deutivacaftor (n=9), vanzacaftor (10 mg)-tezacaftor-deutivacaftor (n=19), vanzacaftor (20 mg)-tezacaftor-deutivacaftor (n=20), and placebo (n=10) had mean changes relative to baseline at day 29 in ppFEV1 of 4·6 percentage points (-1·3 to 10·6), 14·2 percentage points (10·0 to 18·4), 9·8 percentage points (5·7 to 13·8), and 1·9 percentage points (-4·1 to 8·0), respectively, in sweat chloride concentration of -42·8 mmol/L (-51·7 to -34·0), -45·8 mmol/L (95% CI -51·9 to -39·7), -49·5 mmol/L (-55·9 to -43·1), and 2·3 mmol/L (-7·0 to 11·6), respectively, and in CFQ-R respiratory domain score of 17·6 points (3·5 to 31·6), 21·2 points (11·9 to 30·6), 29·8 points (21·0 to 38·7), and 3·3 points (-10·1 to 16·6), respectively. Participants with the F/F genotype treated with vanzacaftor (20 mg)-tezacaftor-deutivacaftor (n=18) and tezacaftor-ivacaftor (n=10) had mean changes relative to baseline (taking tezacaftor-ivacaftor) at day 29 in ppFEV1 of 15·9 percentage points (11·3 to 20·6) and -0·1 percentage points (-6·4 to 6·1), respectively, in sweat chloride concentration of -45·5 mmol/L (-49·7 to -41·3) and -2·6 mmol/L (-8·2 to 3·1), respectively, and in CFQ-R respiratory domain score of 19·4 points (95% CI 10·5 to 28·3) and -5·0 points (-16·9 to 7·0), respectively. The most common adverse events overall were cough, increased sputum, and headache. One participant in the vanzacaftor-tezacaftor-deutivacaftor group had a serious adverse event of infective pulmonary exacerbation and another participant had a serious rash event that led to treatment discontinuation. For most participants, adverse events were mild or moderate in severity. INTERPRETATION: Once-daily dosing with vanzacaftor-tezacaftor-deutivacaftor was safe and well tolerated and improved lung function, respiratory symptoms, and CFTR function. These results support the continued investigation of vanzacaftor-tezacaftor-deutivacaftor in phase 3 clinical trials compared with elexacaftor-tezacaftor-ivacaftor. FUNDING: Vertex Pharmaceuticals.


Subject(s)
Cystic Fibrosis , Humans , Adult , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Chlorides , Forced Expiratory Volume , Aminophenols/adverse effects , Benzodioxoles/therapeutic use , Mutation , Double-Blind Method , Chloride Channel Agonists/therapeutic use
2.
Am J Respir Cell Mol Biol ; 68(2): 176-185, 2023 02.
Article in English | MEDLINE | ID: mdl-36174229

ABSTRACT

Tissue availability remains an important limitation of single-cell genomic technologies for investigating cellular heterogeneity in human health and disease. BAL represents a minimally invasive approach to assessing an individual's lung cellular environment for diagnosis and research. However, the lack of high-quality, healthy lung reference data is a major obstacle to using single-cell approaches to study a plethora of lung diseases. Here, we performed single-cell RNA sequencing on over 40,000 cells isolated from the BAL of four healthy volunteers. Of the six cell types or lineages we identified, macrophages were consistently the most numerous across individuals. Our analysis confirmed the expression of marker genes defining cell types despite background signals because of the ambient RNA found in many single-cell studies. We assessed the variability of gene expression across macrophages and defined a distinct subpopulation of cells expressing a set of genes associated with Macrophage Inflammatory Protein 1 (MIP-1). RNA in situ hybridization and reanalysis of published lung single-cell data validated the presence of this macrophage subpopulation. Thus, our study characterizes lung macrophage heterogeneity in healthy individuals and provides a valuable resource for future studies to understand the lung environment in health and disease.


Subject(s)
Macrophage Inflammatory Proteins , Macrophages , Humans , Macrophage Inflammatory Proteins/genetics , Bronchoalveolar Lavage Fluid , Healthy Volunteers , RNA
3.
Lancet Respir Med ; 10(3): 267-277, 2022 03.
Article in English | MEDLINE | ID: mdl-34942085

ABSTRACT

BACKGROUND: Elexacaftor plus tezacaftor plus ivacaftor is a triple-combination cystic fibrosis transmembrane conductance regulator (CFTR) modulator regimen shown to be generally safe and efficacious in people with cystic fibrosis aged 12 years or older with at least one F508del-CFTR allele. We aimed to assess the magnitude and durability of the clinical effects of this triple combination regimen in people with cystic fibrosis homozygous for the F508del-CFTR mutation. METHODS: We conducted a multicentre, randomised, double-blind, active-controlled, phase 3b trial of elexacaftor plus tezacaftor plus ivacaftor at 35 medical centres in Australia, Belgium, Germany, and the UK. Eligible participants were those with cystic fibrosis homozygous for the F508del-CFTR mutation, aged 12 years or older with stable disease, and with a percent predicted FEV1 of 40-90% inclusive. After a 4-week run-in period, in which participants received tezacaftor 100 mg orally once daily and ivacaftor 150 mg orally every 12 h, participants were randomly assigned (1:1) to receive 24 weeks of either elexacaftor 200 mg orally once daily plus tezacaftor 100 mg orally once daily plus ivacaftor 150 mg orally every 12 h (elexacaftor plus tezacaftor plus ivacaftor group) or tezacaftor 100 mg orally once daily plus ivacaftor 150 mg orally every 12 h (tezacaftor plus ivacaftor group). Randomisation was stratified by percent predicted FEV1, age at screening visit, and whether the participant was receiving CFTR modulators at the time of the screening visit. Patients, investigators, and sponsor's study execution team were masked to treatment assignment. The primary endpoint was the absolute change in Cystic Fibrosis Questionnaire-Revised (CFQ-R) respiratory domain score from baseline (ie, at the end of the tezacaftor plus ivacaftor run-in period) up to and including week 24. The key secondary endpoint was the absolute change from baseline in percent predicted FEV1 up to and including week 24; other secondary endpoints were the absolute change from baseline in sweat chloride concentrations up to and including week 24, and safety and tolerability. All endpoints were assessed in all randomised patients who had received at least one dose of their assigned regimen. This study is registered with ClinicalTrials.gov, NCT04105972. FINDINGS: Between Oct 3, 2019, and July 24, 2020, 176 participants were enrolled. Following the 4-week tezacaftor plus ivacaftor run-in period, 175 participants were randomly assigned (87 to the elexacaftor plus tezacaftor plus ivacaftor group and 88 to the tezacaftor plus ivacaftor group) and dosed in the treatment period. From baseline up to and including week 24, the mean CFQ-R respiratory domain score increased by 17·1 points (95% CI 14·1 to 20·1) in the elexacaftor plus tezacaftor plus ivacaftor group and by 1·2 points (-1·7 to 4·2) in the tezacaftor plus ivacaftor group (least squares mean treatment difference 15·9 points [95% CI 11·7 to 20·1], p<0·0001), the mean percent predicted FEV1 increased by 11·2 percentage points (95% CI 9·8 to 12·6) in the elexacaftor plus tezacaftor plus ivacaftor group and by 1·0 percentage points (-0·4 to 2·4) in the tezacaftor plus ivacaftor group (least squares mean treatment difference 10·2 percentage points [8·2 to 12·1], p<0·0001), and the mean sweat chloride concentration decreased by 46·2 mmol/L (95% CI 43·7 to 48·7) in the elexacaftor plus tezacaftor plus ivacaftor group and by 3·4 mmol/L (1·0 to 5·8) in the tezacaftor plus ivacaftor group (least squares mean treatment difference -42·8 mmol/L [-46·2 to -39·3], nominal p<0·0001). Most participants (70 [80%] in the elexacaftor plus tezacaftor plus ivacaftor group and 74 [84%] in the tezacaftor plus ivacaftor group) had adverse events that were mild or moderate in severity; serious adverse events occurred in five (6%) of 87 participants in the elexacaftor plus tezacaftor plus ivacaftor group and 14 (16%) of 88 participants in the tezacaftor plus ivacaftor group. One (1%) participant in the elexacaftor plus tezacaftor plus ivacaftor group discontinued treatment due to an adverse event of anxiety and depression. Two (2%) participants in the tezacaftor plus ivacaftor group discontinued treatment due to adverse events of psychotic disorder (n=1) and obsessive-compulsive disorder (n=1). INTERPRETATION: The elexacaftor plus tezacaftor plus ivacaftor regimen was safe and well tolerated, and led to significant and clinically meaningful improvements in respiratory-related quality of life and lung function, as well as improved CFTR function, changes that were durable over 24 weeks and superior to those seen with tezacaftor plus ivacaftor in this patient population. FUNDING: Vertex Pharmaceuticals.


Subject(s)
Cystic Fibrosis , Aminophenols/therapeutic use , Benzodioxoles/therapeutic use , Child , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Double-Blind Method , Humans , Indoles , Mutation , Pyrazoles , Pyridines , Pyrrolidines , Quality of Life , Quinolones
4.
Int J Mol Sci ; 21(16)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764262

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic disease for which novel approaches are urgently required. We reported increased sphingosine kinase 1 (SPHK1) in IPF lungs and that SPHK1 inhibition using genetic and pharmacologic approaches reduces murine bleomycin-induced pulmonary fibrosis. We determined whether PF543, a specific SPHK1 inhibitor post bleomycin or asbestos challenge mitigates lung fibrosis by reducing mitochondrial (mt) DNA damage and pro-fibrotic monocyte recruitment-both are implicated in the pathobiology of pulmonary fibrosis. Bleomycin (1.5 U/kg), crocidolite asbestos (100 µg/50 µL) or controls was intratracheally instilled in Wild-Type (C57Bl6) mice. PF543 (1 mg/kg) or vehicle was intraperitoneally injected once every two days from day 7-21 following bleomycin and day 14-21 or day 30-60 following asbestos. PF543 reduced bleomycin- and asbestos-induced pulmonary fibrosis at both time points as well as lung expression of profibrotic markers, lung mtDNA damage, and fibrogenic monocyte recruitment. In contrast to human lung fibroblasts, asbestos augmented lung epithelial cell (MLE) mtDNA damage and PF543 was protective. Post-exposure PF543 mitigates pulmonary fibrosis in part by reducing lung epithelial cell mtDNA damage and monocyte recruitment. We reason that SPHK1 signaling may be an innovative therapeutic target for managing patients with IPF and other forms of lung fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Idiopathic Pulmonary Fibrosis/drug therapy , Methanol/analogs & derivatives , Pulmonary Fibrosis/drug therapy , Pyrrolidines/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Alveolar Epithelial Cells/drug effects , Animals , Asbestos/toxicity , Bleomycin/pharmacology , DNA Damage/drug effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/metabolism , Methanol/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Monocytes/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Sulfones
5.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L1084-L1096, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32209025

ABSTRACT

Alveolar epithelial cell (AEC) apoptosis, arising from mitochondrial dysfunction and mitophagy defects, is important in mediating idiopathic pulmonary fibrosis (IPF). Our group established a role for the mitochondrial (mt) DNA base excision repair enzyme, 8-oxoguanine-DNA glycosylase 1 (mtOGG1), in preventing oxidant-induced AEC mtDNA damage and apoptosis and showed that OGG1-deficient mice have increased lung fibrosis. Herein, we determined whether mice overexpressing the mtOGG1 transgene (mtOgg1tg) are protected against lung fibrosis and whether AEC mtOGG1 preservation of mtDNA integrity mitigates phosphatase and tensin homolog-induced putative kinase 1 (PINK1) deficiency and apoptosis. Compared with wild type (WT), mtOgg1tg mice have diminished asbestos- and bleomycin-induced pulmonary fibrosis that was accompanied by reduced lung and AEC mtDNA damage and apoptosis. Asbestos and H2O2 promote the MLE-12 cell PINK1 deficiency, as assessed by reductions in the expression of PINK1 mRNA and mitochondrial protein expression. Compared with WT, Pink1-knockout (Pink1-KO) mice are more susceptible to asbestos-induced lung fibrosis and have increased lung and alveolar type II (AT2) cell mtDNA damage and apoptosis. AT2 cells from Pink1-KO mice and PINK1-silenced (siRNA) MLE-12 cells have increased mtDNA damage that is augmented by oxidative stress. Interestingly, mtOGG1 overexpression attenuates oxidant-induced MLE-12 cell mtDNA damage and apoptosis despite PINK1 silencing. mtDNA damage is increased in the lungs of patients with IPF as compared with controls. Collectively, these findings suggest that mtOGG1 maintenance of AEC mtDNA is crucial for preventing PINK1 deficiency that promotes apoptosis and lung fibrosis. Given the key role of AEC apoptosis in pulmonary fibrosis, strategies aimed at preserving AT2 cell mtDNA integrity may be an innovative target.


Subject(s)
Alveolar Epithelial Cells/drug effects , Asbestosis/genetics , DNA Glycosylases/genetics , Lung/drug effects , Mitochondria/drug effects , Protein Kinases/genetics , Pulmonary Fibrosis/genetics , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Asbestos/administration & dosage , Asbestosis/etiology , Asbestosis/metabolism , Asbestosis/pathology , Bleomycin/administration & dosage , DNA Damage , DNA Glycosylases/deficiency , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Gene Expression Regulation , Hydrogen Peroxide/pharmacology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Primary Cell Culture , Protein Kinases/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Titanium/administration & dosage
6.
Front Immunol ; 10: 1813, 2019.
Article in English | MEDLINE | ID: mdl-31417574

ABSTRACT

Macrophages are often viewed through the lens of their core functions, but recent transcriptomic studies reveal them to be largely distinct across tissue types. While these differences appear to be shaped by their local environment, the key signals that drive these transcriptional differences remain unclear. Since Wnt signaling plays established roles in cell fate decisions, and tissue patterning during development and tissue repair after injury, we consider evidence that Wnt signals both target and are affected by macrophage functions. We propose that the Wnt gradients present in developing and adult tissues effectively shape macrophage fates and phenotypes. We also highlight evidence that macrophages, through an ability to dispatch Wnt signals, may couple tissue debridement and matrix remodeling with stem cell activation and tissue repair.


Subject(s)
Cell Differentiation/immunology , Macrophages/immunology , Stem Cells/immunology , Wnt Signaling Pathway/immunology , Animals , Humans , beta Catenin/immunology
7.
Respir Med Case Rep ; 26: 304-306, 2019.
Article in English | MEDLINE | ID: mdl-30886821

ABSTRACT

BACKGROUND: Mirabegron, a ß3 adrenergic receptor agonist, is FDA approved for treatment of overactive bladder. Approved in 2012 in the US, there have been no reports of any effects of mirabegron on pulmonary function. CASE PRESENTATION: We report the case of a 65 year old male with a history of Parkinson's disease, OSA, and aspiration pneumonia presenting with subacute worsening dyspnea and found to have worsening restrictive ventilatory defect with a pattern consistent with neuromuscular weakness. After recalling that initiation of mirabegron correlated with onset of his worsening symptoms, the patient decided to perform a trial period off the drug. He subsequently reported prompt improvement in his respiratory symptoms, which was confirmed objectively by pulmonary function tests. In this case, mirabegron was temporally associated with subacute worsening of the patient's pulmonary restrictive physiology, with subsequent resolution after discontinuation of the medication. CONCLUSIONS: The mechanism of this adverse effect is unknown, but we speculate that this effect may be potentially mediated by the effect of ß3 adrenergic receptor agonism on skeletal muscle, in this case in a patient with pre-existing neuromuscular disease. Careful assessment of patients who develop shortness of breath while on mirabegron should include an assessment for restrictive lung disease secondary neuromuscular dysfunction. Additional study is needed of the effects of ß3 agonism on skeletal muscle.

8.
Am J Respir Crit Care Med ; 199(12): 1517-1536, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30554520

ABSTRACT

Rationale: The contributions of diverse cell populations in the human lung to pulmonary fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal changes within individual cell populations during pulmonary fibrosis that are important for disease pathogenesis. Objectives: To determine whether single-cell RNA sequencing can reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other cell types in lung tissue from subjects with pulmonary fibrosis compared with control subjects. Methods: We performed single-cell RNA sequencing on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and on one bronchoscopic cryobiospy sample from a patient with idiopathic pulmonary fibrosis. We validated these data using in situ RNA hybridization, immunohistochemistry, and bulk RNA-sequencing on flow-sorted cells from 22 additional subjects. Measurements and Main Results: We identified a distinct, novel population of profibrotic alveolar macrophages exclusively in patients with fibrosis. Within epithelial cells, the expression of genes involved in Wnt secretion and response was restricted to nonoverlapping cells. We identified rare cell populations including airway stem cells and senescent cells emerging during pulmonary fibrosis. We developed a web-based tool to explore these data. Conclusions: We generated a single-cell atlas of pulmonary fibrosis. Using this atlas, we demonstrated heterogeneity within alveolar macrophages and epithelial cells from subjects with pulmonary fibrosis. These results support the feasibility of discovery-based approaches using next-generation sequencing technologies to identify signaling pathways for targeting in the development of personalized therapies for patients with pulmonary fibrosis.


Subject(s)
Cells, Cultured/pathology , Epithelial Cells/pathology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Sequence Analysis, RNA , Stem Cells/pathology , Transcriptome , Animals , Disease Models, Animal , Female , Humans , Male
9.
J Exp Med ; 214(8): 2387-2404, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28694385

ABSTRACT

Little is known about the relative importance of monocyte and tissue-resident macrophages in the development of lung fibrosis. We show that specific genetic deletion of monocyte-derived alveolar macrophages after their recruitment to the lung ameliorated lung fibrosis, whereas tissue-resident alveolar macrophages did not contribute to fibrosis. Using transcriptomic profiling of flow-sorted cells, we found that monocyte to alveolar macrophage differentiation unfolds continuously over the course of fibrosis and its resolution. During the fibrotic phase, monocyte-derived alveolar macrophages differ significantly from tissue-resident alveolar macrophages in their expression of profibrotic genes. A population of monocyte-derived alveolar macrophages persisted in the lung for one year after the resolution of fibrosis, where they became increasingly similar to tissue-resident alveolar macrophages. Human homologues of profibrotic genes expressed by mouse monocyte-derived alveolar macrophages during fibrosis were up-regulated in human alveolar macrophages from fibrotic compared with normal lungs. Our findings suggest that selectively targeting alveolar macrophage differentiation within the lung may ameliorate fibrosis without the adverse consequences associated with global monocyte or tissue-resident alveolar macrophage depletion.


Subject(s)
Lung/pathology , Macrophages, Alveolar/pathology , Animals , Cell Differentiation , Fibrosis , Humans , Lung/cytology , Mice , Monocytes/pathology
10.
Am J Respir Cell Mol Biol ; 56(2): 191-201, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27668462

ABSTRACT

Previous studies established that attenuating Wnt/ß-catenin signaling limits lung fibrosis in the bleomycin mouse model of this disease, but the contribution of this pathway to distinct lung cell phenotypes relevant to tissue repair and fibrosis remains incompletely understood. Using microarray analysis, we found that bleomycin-injured lungs from mice that lack the Wnt coreceptor low density lipoprotein receptor-related protein 5 (Lrp5) and exhibit reduced fibrosis showed enrichment for pathways related to extracellular matrix processing, immunity, and lymphocyte proliferation, suggesting the contribution of an immune-matrix remodeling axis relevant to fibrosis. Activation of ß-catenin signaling was seen in lung macrophages using the ß-catenin reporter mouse, Axin2+/LacZ. Analysis of lung immune cells by flow cytometry after bleomycin administration revealed that Lrp5-/- lungs contained significantly fewer Siglec Flow alveolar macrophages, a cell type previously implicated as positive effectors of fibrosis. Macrophage-specific deletion of ß-catenin in CD11ccre;ß-cateninflox mice did not prevent development of bleomycin-induced fibrosis but facilitated its resolution by 8 weeks. In a nonresolving model of fibrosis, intratracheal administration of asbestos in Lrp5-/- mice also did not prevent the development of fibrosis but hindered the progression of fibrosis in asbestos-treated Lrp5-/- lungs, phenocopying the findings in bleomycin-treated CD11ccre;ß-cateninflox mice. Activation of ß-catenin signaling using lithium chloride resulted in worsened fibrosis in wild-type mice, further supporting that the effects of loss of Lrp5 are directly mediated by Wnt/ß-catenin signaling. Together, these data suggest that lung myeloid cells are responsive to Lrp5/ß-catenin signaling, leading to differentiation of an alveolar macrophage subtype that antagonizes the resolution of lung fibrosis.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Lung/pathology , Macrophages/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Bleomycin , Cell Differentiation , Extracellular Matrix/metabolism , Immunity , Macrophage Activation , Macrophages/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/pathology , Pulmonary Fibrosis/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
11.
Methods Mol Biol ; 1481: 49-60, 2016.
Article in English | MEDLINE | ID: mdl-27590151

ABSTRACT

ß-catenin (CTNNB1) is a dual-function cell-cell adhesion/transcriptional co-activator protein and an essential transducer of canonical Wnt signals. Although a number of established techniques and reagents are available to quantify the nuclear signaling activity of ß-catenin (e.g., TCF-dependent reporter assays, nuclear accumulation of ß-catenin, and generation of N-terminally hypophosphorylated ß-catenin), there are cell-type and context-dependent limitations of these methods. Since the posttranscriptional stabilization of ß-catenin outside of the cadherin complex appears universally required for ß-catenin signaling, the following method allows for simple assessment of the cadherin-free fraction of ß-catenin in cells, using a GST-tagged form of ICAT (Inhibitor of ß-Catenin and Tcf) as an affinity matrix. This method is more sensitive and quantitative than immunofluorescence and may be useful in studies that implicate TCF-independent signaling events.


Subject(s)
Cell Nucleus/chemistry , Molecular Biology/methods , Transcription, Genetic , beta Catenin/isolation & purification , Cadherins/chemistry , Cadherins/genetics , Cell Adhesion/genetics , Wnt Proteins/chemistry , Wnt Proteins/isolation & purification , Wnt Signaling Pathway , beta Catenin/genetics
12.
Biochem Biophys Res Commun ; 470(3): 606-612, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26797284

ABSTRACT

Adiponectin is a pleiotropic adipokine implicated in obesity, metabolic syndrome and cardiovascular disease. Recent studies have identified adiponectin as a negative regulator of tissue fibrosis. Wnt/ß-catenin signaling has also been implicated in metabolic syndrome and can promote tissue fibrosis, but the extent to which adiponectin cross-regulates Wnt/ß-catenin signaling is unknown. Using primary human dermal fibroblasts and recombinant purified proteins, we show that adiponectin can limit ß-catenin accumulation and downstream gene activation by inhibiting Lrp6 phosphorylation, a key activation step in canonical Wnt signaling. Inhibition of Wnt3a-mediated Lrp6 phospho-activation is relatively rapid (e.g., by 30 min), and is not dependent on established adiponectin G-protein coupled receptors, AdipoR1 and R2, suggesting a more direct relationship to Lrp6 signaling. In contrast, the ability of adiponectin to limit Wnt-induced and baseline collagen production in fibroblasts requires AdipoR1/R2. These results suggest the possibility that the pleiotropic effects of adiponectin may be mediated through distinct cell surface receptor complexes. Accordingly, we propose that the anti-fibrotic activity of adiponectin may be mediated through AdipoR1/R2 receptors, while the ability of adiponectin to inhibit Lrp6 phospho-activation may be relevant to other recently established roles for Lrp6 signaling in glucose metabolism and metabolic syndrome.


Subject(s)
Adiponectin/administration & dosage , Fibroblasts/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Humans , Phosphorylation , Skin/cytology , Skin/drug effects , Skin/metabolism , Wnt Signaling Pathway/drug effects
13.
Nat Commun ; 6: 6574, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25762200

ABSTRACT

Activation of the NLRP3 inflammasome and subsequent maturation of IL-1ß have been implicated in acute lung injury (ALI), resulting in inflammation and fibrosis. We investigated the role of vimentin, a type III intermediate filament, in this process using three well-characterized murine models of ALI known to require NLRP3 inflammasome activation. We demonstrate that central pathophysiologic events in ALI (inflammation, IL-1ß levels, endothelial and alveolar epithelial barrier permeability, remodelling and fibrosis) are attenuated in the lungs of Vim(-/-) mice challenged with LPS, bleomycin and asbestos. Bone marrow chimeric mice lacking vimentin have reduced IL-1ß levels and attenuated lung injury and fibrosis following bleomycin exposure. Furthermore, decreased active caspase-1 and IL-1ß levels are observed in vitro in Vim(-/-) and vimentin-knockdown macrophages. Importantly, we show direct protein-protein interaction between NLRP3 and vimentin. This study provides insights into lung inflammation and fibrosis and suggests that vimentin may be a key regulator of the NLRP3 inflammasome.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Vimentin/metabolism , Acute Lung Injury/metabolism , Animals , Bleomycin/chemistry , Bone Marrow Cells/cytology , Bronchoalveolar Lavage Fluid , Cell Line , Cell Proliferation , Female , Fibrosis , Interleukin-1beta/metabolism , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Microscopy, Atomic Force , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Interaction Mapping
14.
Am J Respir Crit Care Med ; 190(2): 185-95, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24921217

ABSTRACT

RATIONALE: Wnt/ß-catenin signaling has been implicated in lung fibrosis, but how this occurs and whether expression changes in Wnt pathway components predict disease progression is unknown. OBJECTIVES: To determine whether the Wnt coreceptor Lrp5 drives pulmonary fibrosis in mice and is predictive of disease severity in humans. METHODS: We examined mice with impaired Wnt signaling caused by loss of the Wnt coreceptor Lrp5 in models of lung fibrosis induced by bleomycin or an adenovirus encoding an active form of transforming growth factor (TGF)-ß. We also analyzed gene expression in peripheral blood mononuclear cells (PBMC) from patients with idiopathic pulmonary fibrosis (IPF). MEASUREMENTS AND MAIN RESULTS: In patients with IPF, analysis of peripheral blood mononuclear cells revealed that elevation of positive regulators, Lrp5 and 6, was independently associated with disease progression. LRP5 was also associated with disease severity at presentation in an additional cohort of patients with IPF. Lrp5 null mice were protected against bleomycin-induced pulmonary fibrosis, an effect that was phenocopied by direct inhibition of ß-catenin signaling by the small molecular inhibitor of ß-catenin responsive transcription. Transplantation of Lrp5 null bone marrow cells into wild-type mice did not limit fibrosis. Instead, Lrp5 loss was associated with reduced TGF-ß production by alveolar type 2 cells and leukocytes. Consistent with a role of Lrp5 in the activation of TGF-ß, Lrp5 null mice were not protected against lung fibrosis induced by TGF-ß. CONCLUSIONS: We show that the Wnt coreceptor, Lrp5, is a genetic driver of lung fibrosis in mice and a marker of disease progression and severity in humans with IPF. Evidence that TGF-ß signaling can override a loss in Lrp5 has implications for patient selection and timing of Wnt pathway inhibitors in lung fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Aged , Animals , Biomarkers/metabolism , Disease Progression , Female , Humans , Idiopathic Pulmonary Fibrosis/etiology , Leukocytes, Mononuclear/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Prospective Studies , Severity of Illness Index , Signal Transduction , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Am J Respir Cell Mol Biol ; 49(6): 892-901, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23885834

ABSTRACT

Asbestos exposure results in pulmonary fibrosis (asbestosis) and malignancies (bronchogenic lung cancer and mesothelioma) by mechanisms that are not fully understood. Alveolar epithelial cell (AEC) apoptosis is important in the development of pulmonary fibrosis after exposure to an array of toxins, including asbestos. An endoplasmic reticulum (ER) stress response and mitochondria-regulated (intrinsic) apoptosis occur in AECs of patients with idiopathic pulmonary fibrosis, a disease with similarities to asbestosis. Asbestos induces AEC intrinsic apoptosis, but the role of the ER is unclear. The objective of this study was to determine whether asbestos causes an AEC ER stress response that promotes apoptosis. Using human A549 and rat primary isolated alveolar type II cells, amosite asbestos fibers increased AEC mRNA and protein expression of ER stress proteins involved in the unfolded protein response, such as inositol-requiring kinase (IRE) 1 and X-box-binding protein-1, as well as ER Ca²(2+) release ,as assessed by a FURA-2 assay. Eukarion-134, a superoxide dismutase/catalase mimetic, as well as overexpression of Bcl-XL in A549 cells each attenuate asbestos-induced AEC ER stress (IRE-1 and X-box-binding protein-1 protein expression; ER Ca²(2+) release) and apoptosis. Thapsigargin, a known ER stress inducer, augments AEC apoptosis, and eukarion-134 or Bcl-XL overexpression are protective. Finally, 4-phenylbutyric acid, a chemical chaperone that attenuates ER stress, blocks asbestos- and thapsigargin-induced AEC IRE-1 protein expression, but does not reduce ER Ca²(2+) release or apoptosis. These results show that asbestos triggers an AEC ER stress response and subsequent intrinsic apoptosis that is mediated in part by ER Ca²(2+) release.


Subject(s)
Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Apoptosis/drug effects , Asbestos, Amosite/toxicity , Endoplasmic Reticulum Stress/drug effects , Alveolar Epithelial Cells/physiology , Animals , Antioxidants/pharmacology , Apoptosis/physiology , Calcium Signaling/drug effects , Cell Line , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organometallic Compounds/pharmacology , Phenylbutyrates/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Regulatory Factor X Transcription Factors , Salicylates/pharmacology , Thapsigargin/pharmacology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
17.
Arthritis Rheum ; 64(8): 2734-45, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22328118

ABSTRACT

OBJECTIVE: Fibrosis in human diseases and animal models is associated with aberrant Wnt/ß-catenin pathway activation. The aim of this study was to characterize the regulation, activity, mechanism of action, and significance of Wnt/ß-catenin signaling in the context of systemic sclerosis (SSc). METHODS: The expression of Wnt signaling pathway components in SSc skin biopsy specimens was analyzed. The regulation of profibrotic responses by canonical Wnt/ß-catenin was examined in explanted human mesenchymal cells. Fibrotic responses were studied using proliferation, migration, and gel contraction assays. The cell fate specification of subcutaneous preadipocytes by canonical Wnt signaling was evaluated. RESULTS: Analysis of published genome-wide expression data revealed elevated expression of the Wnt receptor FZD2 and the Wnt target LEF1 and decreased expression of Wnt antagonists DKK2 and WIF1 in skin biopsy specimens from subsets of patients with diffuse cutaneous SSc compared to the other distinct subsets. Immunohistochemical analysis showed increased nuclear ß-catenin expression in these biopsy specimens. In vitro, Wnt-3a induced ß-catenin activation, stimulated fibroblast proliferation and migration, collagen gel contraction, and myofibroblast differentiation, and enhanced profibrotic gene expression. Genetic and pharmacologic approaches were used to demonstrate that these profibrotic responses involved autocrine transforming growth factor ß signaling via Smads. In contrast, in explanted subcutaneous preadipocytes, Wnt-3a repressed adipogenesis and promoted myofibroblast differentiation. CONCLUSION: Canonical Wnt signaling was hyperactivated in SSc skin biopsy specimens. In explanted mesenchymal cells, Wnt-3a stimulated fibrogenic responses while suppressing adipogenesis. Taken together, these results indicate that Wnts have potent profibrotic effects, and that canonical Wnt signaling plays an important role in the pathogenesis of fibrosis and lipoatrophy in SSc.


Subject(s)
Mesoderm/metabolism , Mesoderm/pathology , Scleroderma, Systemic/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adult , Biopsy , Case-Control Studies , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Frizzled Receptors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Mesoderm/drug effects , Repressor Proteins/metabolism , Scleroderma, Systemic/pathology , Skin/metabolism , Skin/pathology , Wnt3A Protein/pharmacology
18.
Thorax ; 67(2): 139-46, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21921091

ABSTRACT

BACKGROUND: The development of organ fibrosis after injury requires activation of transforming growth factor ß(1) which regulates the transcription of profibrotic genes. The systemic administration of a proteasomal inhibitor has been reported to prevent the development of fibrosis in the liver, kidney and bone marrow. It is hypothesised that proteasomal inhibition would prevent lung and skin fibrosis after injury by inhibiting TGF-ß(1)-mediated transcription. METHODS: Bortezomib, a small molecule proteasome inhibitor in widespread clinical use, was administered to mice beginning 7 days after the intratracheal or intradermal administration of bleomycin and lung and skin fibrosis was measured after 21 or 40 days, respectively. To examine the mechanism of this protection, bortezomib was administered to primary normal lung fibroblasts and primary lung and skin fibroblasts obtained from patients with idiopathic pulmonary fibrosis and scleroderma, respectively. RESULTS: Bortezomib promoted normal repair and prevented lung and skin fibrosis when administered beginning 7 days after the initiation of bleomycin. In primary human lung fibroblasts from normal individuals and patients with idiopathic pulmonary fibrosis and in skin fibroblasts from a patient with scleroderma, bortezomib inhibited TGF-ß(1)-mediated target gene expression by inhibiting transcription induced by activated Smads. An increase in the abundance and activity of the nuclear hormone receptor PPARγ, a repressor of Smad-mediated transcription, contributed to this response. CONCLUSIONS: Proteasomal inhibition prevents lung and skin fibrosis after injury in part by increasing the abundance and activity of PPARγ. Proteasomal inhibition may offer a novel therapeutic alternative in patients with dysregulated tissue repair and fibrosis.


Subject(s)
Boronic Acids/therapeutic use , Proteasome Inhibitors , Pulmonary Fibrosis/prevention & control , Pyrazines/therapeutic use , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Autocrine Communication/drug effects , Bleomycin , Boronic Acids/pharmacology , Bortezomib , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , PPAR gamma/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pyrazines/pharmacology , Scleroderma, Systemic/pathology , Signal Transduction/drug effects , Skin/pathology , Transforming Growth Factor beta1/metabolism
19.
Curr Opin Rheumatol ; 23(6): 562-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21885974

ABSTRACT

PURPOSE OF REVIEW: The Wnt/ß-catenin signaling pathway plays a critical role in development and adult tissue homeostasis. Recent investigations implicate Wnt/ß-catenin signaling in abnormal wound repair and fibrogenesis. The purpose of this review is to highlight recent key studies that support a role for Wnt/ß-catenin signaling in fibrosis. RECENT FINDINGS: Studies of patients with fibrotic diseases have demonstrated changes in components of the Wnt/ß-catenin pathway. In animal models, perturbations in Wnt/ß-catenin signaling appear to aggravate or ameliorate markers of injury and fibrosis in a variety of different tissues. Studies also suggest that fibroblasts from different tissue sources may have markedly divergent responses to Wnt/ß-catenin signaling. Cross-talk between Wnt/ß-catenin and transforming growth factor-ß pathways is complex and context-dependent, and may promote fibrogenesis through coregulation of fibrogenic gene targets. High throughput screening has identified several novel chemical inhibitors of Wnt/ß-catenin signaling that may be of therapeutic potential. SUMMARY: Wnt/ß-catenin signaling appears important in normal wound healing and its sustained activation is associated with fibrogenesis. The mechanism by which Wnt/ß-catenin signaling may modify the response to injury is cell-type and context-dependent. Better understanding of this signaling pathway may provide a promising new therapeutic approach for human fibrotic diseases.


Subject(s)
Fibrosis/etiology , beta Catenin/metabolism , Animals , Fibrosis/metabolism , Fibrosis/therapy , Glycoproteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Signal Transduction , Skin/metabolism , Wnt Signaling Pathway , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...