Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Thorax ; 76(1): 73-82, 2021 01.
Article in English | MEDLINE | ID: mdl-33214245

ABSTRACT

INTRODUCTION: Fibroblastic foci represent the cardinal pathogenic lesion in idiopathic pulmonary fibrosis (IPF) and comprise activated fibroblasts and myofibroblasts, the key effector cells responsible for dysregulated extracellular matrix deposition in multiple fibrotic conditions. The aim of this study was to define the major transcriptional programmes involved in fibrogenesis in IPF by profiling unmanipulated myofibroblasts within fibrotic foci in situ by laser capture microdissection. METHODS: The challenges associated with deriving gene calls from low amounts of RNA and the absence of a meaningful comparator cell type were overcome by adopting novel data mining strategies and by using weighted gene co-expression network analysis (WGCNA), as well as an eigengene-based approach to identify transcriptional signatures, which correlate with fibrillar collagen gene expression. RESULTS: WGCNA identified prominent clusters of genes associated with cell cycle, inflammation/differentiation, translation and cytoskeleton/cell adhesion. Collagen eigengene analysis revealed that transforming growth factor ß1 (TGF-ß1), RhoA kinase and the TSC2/RHEB axis formed major signalling clusters associated with collagen gene expression. Functional studies using CRISPR-Cas9 gene-edited cells demonstrated a key role for the TSC2/RHEB axis in regulating TGF-ß1-induced mechanistic target of rapamycin complex 1 activation and collagen I deposition in mesenchymal cells reflecting IPF and other disease settings, including cancer-associated fibroblasts. CONCLUSION: These data provide strong support for the human tissue-based and bioinformatics approaches adopted to identify critical transcriptional nodes associated with the key pathogenic cell responsible for fibrogenesis in situ and further identify the TSC2/RHEB axis as a potential novel target for interfering with excessive matrix deposition in IPF and other fibrotic conditions.


Subject(s)
Gene Expression Regulation , Idiopathic Pulmonary Fibrosis/genetics , RNA/genetics , Transcriptome/genetics , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Signal Transduction , Up-Regulation
3.
Nat Commun ; 11(1): 4659, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938936

ABSTRACT

The αvß6 integrin plays a key role in the activation of transforming growth factor-ß (TGFß), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvß6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvß6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFß signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvß6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvß6, induces prolonged inhibition of TGFß signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy.


Subject(s)
Butyrates/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Integrins/antagonists & inhibitors , Naphthyridines/pharmacology , Pyrazoles/pharmacology , Pyrrolidines/pharmacology , Administration, Inhalation , Animals , Antigens, Neoplasm/metabolism , Bleomycin/toxicity , Butyrates/administration & dosage , Butyrates/metabolism , Butyrates/pharmacokinetics , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Integrins/metabolism , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Naphthyridines/administration & dosage , Naphthyridines/metabolism , Naphthyridines/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrrolidines/administration & dosage , Pyrrolidines/metabolism , Pyrrolidines/pharmacokinetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tomography, Emission-Computed, Single-Photon , Transforming Growth Factor beta/metabolism , Translational Research, Biomedical
4.
Proc Natl Acad Sci U S A ; 117(2): 1139-1147, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31879343

ABSTRACT

Pulmonary inflammatory responses lie under circadian control; however, the importance of circadian mechanisms in the underlying fibrotic phenotype is not understood. Here, we identify a striking change to these mechanisms resulting in a gain of amplitude and lack of synchrony within pulmonary fibrotic tissue. These changes result from an infiltration of mesenchymal cells, an important cell type in the pathogenesis of pulmonary fibrosis. Mutation of the core clock protein REVERBα in these cells exacerbated the development of bleomycin-induced fibrosis, whereas mutation of REVERBα in club or myeloid cells had no effect on the bleomycin phenotype. Knockdown of REVERBα revealed regulation of the little-understood transcription factor TBPL1. Both REVERBα and TBPL1 altered integrinß1 focal-adhesion formation, resulting in increased myofibroblast activation. The translational importance of our findings was established through analysis of 2 human cohorts. In the UK Biobank, circadian strain markers (sleep length, chronotype, and shift work) are associated with pulmonary fibrosis, making them risk factors. In a separate cohort, REVERBα expression was increased in human idiopathic pulmonary fibrosis (IPF) lung tissue. Pharmacological targeting of REVERBα inhibited myofibroblast activation in IPF fibroblasts and collagen secretion in organotypic cultures from IPF patients, thus suggesting that targeting of REVERBα could be a viable therapeutic approach.


Subject(s)
CLOCK Proteins/antagonists & inhibitors , Circadian Clocks/physiology , Fibroblasts/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/adverse effects , CLOCK Proteins/genetics , CLOCK Proteins/therapeutic use , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Idiopathic Pulmonary Fibrosis , Integrins , Lung/pathology , Male , Mesenchymal Stem Cells , Mice , Mice, Knockout , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , TATA Box Binding Protein-Like Proteins/metabolism , Transcriptome
5.
Sci Signal ; 12(582)2019 05 21.
Article in English | MEDLINE | ID: mdl-31113850

ABSTRACT

The differentiation of fibroblasts into a transient population of highly activated, extracellular matrix (ECM)-producing myofibroblasts at sites of tissue injury is critical for normal tissue repair. Excessive myofibroblast accumulation and persistence, often as a result of a failure to undergo apoptosis when tissue repair is complete, lead to pathological fibrosis and are also features of the stromal response in cancer. Myofibroblast differentiation is accompanied by changes in cellular metabolism, including increased glycolysis, to meet the biosynthetic demands of enhanced ECM production. Here, we showed that transforming growth factor-ß1 (TGF-ß1), the key pro-fibrotic cytokine implicated in multiple fibrotic conditions, increased the production of activating transcription factor 4 (ATF4), the transcriptional master regulator of amino acid metabolism, to supply glucose-derived glycine to meet the amino acid requirements associated with enhanced collagen production in response to myofibroblast differentiation. We further delineated the signaling pathways involved and showed that TGF-ß1-induced ATF4 production depended on cooperation between canonical TGF-ß1 signaling through Smad3 and activation of mechanistic target of rapamycin complex 1 (mTORC1) and its downstream target eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). ATF4, in turn, promoted the transcription of genes encoding enzymes of the de novo serine-glycine biosynthetic pathway and glucose transporter 1 (GLUT1). Our findings suggest that targeting the TGF-ß1-mTORC1-ATF4 axis may represent a novel therapeutic strategy for interfering with myofibroblast function in fibrosis and potentially in other conditions, including cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Collagen/biosynthesis , Glycine/biosynthesis , Mechanistic Target of Rapamycin Complex 1/metabolism , Serine/biosynthesis , Transforming Growth Factor beta1/pharmacology , Activating Transcription Factor 4/genetics , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Signal Transduction/drug effects
6.
Eur Respir J ; 53(3)2019 03.
Article in English | MEDLINE | ID: mdl-30765508

ABSTRACT

Phosphatidylinositol 3-kinases (PI3Ks) and mammalian target of rapamycin (mTOR) play a role in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Omipalisib (GSK2126458) is a potent inhibitor of PI3K/mTOR.A randomised, placebo-controlled, double-blind, repeat dose escalation, experimental medicine study of omipalisib in subjects with IPF was conducted (NCT01725139) to test safety, tolerability, pharmacokinetics and pharmacodynamics. Omipalisib was dosed at 0.25 mg, 1 mg and 2 mg twice daily for 8 days in four cohorts of four subjects randomised 3:1 to receive omipalisib or placebo (two cohorts received 2 mg twice daily).17 subjects with IPF were enrolled. The most common adverse event was diarrhoea, which was reported by four participants. Dose-related increases in insulin and glucose were observed. Pharmacokinetic analysis demonstrated that exposure in the blood predicts lung exposure. Exposure-dependent inhibition of phosphatidylinositol 3,4,5 trisphosphate and pAKT confirmed target engagement in blood and lungs. 18F-2-fluoro-2-deoxy-d-glucose(FDG)-positron emission tomography/computed tomography scans revealed an exposure-dependent reduction in 18F-FDG uptake in fibrotic areas of the lung, as measured by target-to-background, ratio thus confirming pharmacodynamic activity.This experimental medicine study demonstrates acceptable tolerability of omipalisib in subjects with IPF at exposures for which target engagement was confirmed both systemically and in the lungs.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Quinolines/administration & dosage , Sulfonamides/administration & dosage , Administration, Oral , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fluorodeoxyglucose F18 , Humans , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Lung/diagnostic imaging , Lung/pathology , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Positron Emission Tomography Computed Tomography , Pyridazines , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
7.
Nat Commun ; 10(1): 6, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30602778

ABSTRACT

Myofibroblasts are the key effector cells responsible for excessive extracellular matrix deposition in multiple fibrotic conditions, including idiopathic pulmonary fibrosis (IPF). The PI3K/Akt/mTOR axis has been implicated in fibrosis, with pan-PI3K/mTOR inhibition currently under clinical evaluation in IPF. Here we demonstrate that rapamycin-insensitive mTORC1 signaling via 4E-BP1 is a critical pathway for TGF-ß1 stimulated collagen synthesis in human lung fibroblasts, whereas canonical PI3K/Akt signaling is not required. The importance of mTORC1 signaling was confirmed by CRISPR-Cas9 gene editing in normal and IPF fibroblasts, as well as in lung cancer-associated fibroblasts, dermal fibroblasts and hepatic stellate cells. The inhibitory effect of ATP-competitive mTOR inhibition extended to other matrisome proteins implicated in the development of fibrosis and human disease relevance was demonstrated in live precision-cut IPF lung slices. Our data demonstrate that the mTORC1/4E-BP1 axis represents a critical signaling node during fibrogenesis with potential implications for the development of novel anti-fibrotic strategies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Collagen/biosynthesis , Fibroblasts/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Cell Cycle Proteins , Cell Line , Humans , Idiopathic Pulmonary Fibrosis/etiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases/metabolism
8.
Thorax ; 72(1): 66-73, 2017 01.
Article in English | MEDLINE | ID: mdl-27496101

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterised by pulmonary oedema, respiratory failure and severe inflammation. ARDS is further characterised by the recruitment of neutrophils into the lung interstitium and alveolar space. OBJECTIVES: The factors that regulate neutrophil infiltration into the inflamed lung and our understanding of the pathomechanisms in ARDS remain incomplete. This study aimed at determining the role of the chemokine (C-C motif) ligand (CCL)2 and CCL7 in ARDS. METHODS: CCL2 and CCL7 protein levels were measured in bronchoalveolar lavage (BAL) fluid obtained from lipopolysaccharide(LPS)-challenged human volunteers and two separate cohorts of patients with ARDS. Neutrophil chemotaxis to ARDS BAL fluid was evaluated and the contribution of each was assessed and compared with chemokine (C-X-C motif) ligand 8 (CXCL8). Chemokine receptor expression on neutrophils from blood or BAL fluid of patients with ARDS was analysed by flow cytometry. RESULTS: CCL2 and CCL7 were significantly elevated in BAL fluid recovered from LPS-challenged volunteers and patients with ARDS. BAL fluid from patients with ARDS was highly chemotactic for human neutrophils and neutralising either CCL2 or CCL7 attenuated the neutrophil chemotactic response. Moreover, CCL2 and CCL7 synergised with CXCL8 to promote neutrophil migration. Furthermore, neutrophils isolated from the blood or BAL fluid differentially regulated the cell surface expression of chemokine (C-X-C motif) receptor 1 and C-C chemokine receptor type 2 during ARDS. CONCLUSION: This study highlights important inflammatory chemokines involved in regulating neutrophil migration, which may have potential value as therapeutic targets for the treatment of ARDS.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemotaxis, Leukocyte , Interleukin-8/metabolism , Neutrophils/physiology , Respiratory Distress Syndrome/metabolism , Adult , Antibodies, Neutralizing/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL7/antagonists & inhibitors , Chemotaxis, Leukocyte/drug effects , Healthy Volunteers , Humans , Interleukin-8/antagonists & inhibitors , Lipopolysaccharides , Membrane Proteins/metabolism , Neutrophils/metabolism , Receptors, CCR2/metabolism , Receptors, Interleukin-8A/metabolism , Respiratory Distress Syndrome/immunology , Young Adult
10.
Thorax ; 71(8): 701-11, 2016 08.
Article in English | MEDLINE | ID: mdl-27103349

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal of all fibrotic conditions with no curative therapies. Common pathomechanisms between IPF and cancer are increasingly recognised, including dysfunctional pan-PI3 kinase (PI3K) signalling as a driver of aberrant proliferative responses. GSK2126458 is a novel, potent, PI3K/mammalian target of rapamycin (mTOR) inhibitor which has recently completed phase I trials in the oncology setting. Our aim was to establish a scientific and dosing framework for PI3K inhibition with this agent in IPF at a clinically developable dose. METHODS: We explored evidence for pathway signalling in IPF lung tissue and examined the potency of GSK2126458 in fibroblast functional assays and precision-cut IPF lung tissue. We further explored the potential of IPF patient-derived bronchoalveolar lavage (BAL) cells to serve as pharmacodynamic biosensors to monitor GSK2126458 target engagement within the lung. RESULTS: We provide evidence for PI3K pathway activation in fibrotic foci, the cardinal lesions in IPF. GSK2126458 inhibited PI3K signalling and functional responses in IPF-derived lung fibroblasts, inhibiting Akt phosphorylation in IPF lung tissue and BAL derived cells with comparable potency. Integration of these data with GSK2126458 pharmacokinetic data from clinical trials in cancer enabled modelling of an optimal dosing regimen for patients with IPF. CONCLUSIONS: Our data define PI3K as a promising therapeutic target in IPF and provide a scientific and dosing framework for progressing GSK2126458 to clinical testing in this disease setting. A proof-of-mechanism trial of this agent is currently underway. TRIAL REGISTRATION NUMBER: NCT01725139, pre-clinical.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Quinolines/therapeutic use , Sulfonamides/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Proliferation , Clinical Trials as Topic , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/pathology , Pyridazines , Signal Transduction , Treatment Outcome
11.
Dis Model Mech ; 8(9): 1129-39, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26138704

ABSTRACT

TGFß-ALK5 pro-fibrotic signalling and herpesvirus infections have been implicated in the pathogenesis and exacerbation of pulmonary fibrosis. In this study we addressed the role of TGFß-ALK5 signalling during the progression of fibrosis in a two-hit mouse model of murine γ-herpesvirus 68 (MHV-68) infection on the background of pre-existing bleomycin-induced pulmonary fibrosis. Assessment of total lung collagen levels in combination with ex vivo micro-computed tomography (µCT) analysis of whole lungs demonstrated that MHV-68 infection did not enhance lung collagen deposition in this two-hit model but led to a persistent and exacerbated inflammatory response. Moreover, µCT reconstruction and analysis of the two-hit model revealed distinguishing features of diffuse ground-glass opacities and consolidation superimposed on pre-existing fibrosis that were reminiscent of those observed in acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF). Virally-infected murine fibrotic lungs further displayed evidence of extensive inflammatory cell infiltration and increased levels of CCL2, TNFα, IL-1ß and IL-10. Blockade of TGFß-ALK5 signalling attenuated lung collagen accumulation in bleomycin-alone injured mice, but this anti-fibrotic effect was reduced in the presence of concomitant viral infection. In contrast, inhibition of TGFß-ALK5 signalling in virally-infected fibrotic lungs was associated with reduced inflammatory cell aggregates and increased levels of the antiviral cytokine IFNγ. These data reveal newly identified intricacies for the TGFß-ALK5 signalling axis in experimental lung fibrosis, with different outcomes in response to ALK5 inhibition depending on the presence of viral infection. These findings raise important considerations for the targeting of TGFß signalling responses in the context of pulmonary fibrosis.


Subject(s)
Gene Expression Regulation , Herpesviridae Infections/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Bleomycin/adverse effects , Chemokine CCL2/metabolism , Collagen/chemistry , Collagen/metabolism , Disease Models, Animal , Herpesviridae , Herpesviridae Infections/complications , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/complications , Inflammation , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lung/metabolism , Lung/virology , Male , Mice , Mice, Inbred C57BL , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography
12.
J Immunol ; 194(12): 6024-34, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25948816

ABSTRACT

Neutrophils are key effector cells of the innate immune response to pathogenic bacteria, but excessive neutrophilic inflammation can be associated with bystander tissue damage. The mechanisms responsible for neutrophil recruitment to the lungs during bacterial pneumonia are poorly defined. In this study, we focus on the potential role of the major high-affinity thrombin receptor, proteinase-activated receptor 1 (PAR-1), during the development of pneumonia to the common lung pathogen Streptococcus pneumoniae. Our studies demonstrate that neutrophils were indispensable for controlling S. pneumoniae outgrowth but contributed to alveolar barrier disruption. We further report that intra-alveolar coagulation (bronchoalveolar lavage fluid thrombin-antithrombin complex levels) and PAR-1 immunostaining were increased in this model of bacterial lung infection. Functional studies using the most clinically advanced PAR-1 antagonist, SCH530348, revealed a key contribution for PAR-1 signaling in influencing neutrophil recruitment to lung airspaces in response to both an invasive and noninvasive strain of S. pneumoniae (D39 and EF3030) but that PAR-1 antagonism did not impair the ability of the host to control bacterial outgrowth. PAR-1 antagonist treatment significantly decreased pulmonary levels of IL-1ß, CXCL1, CCL2, and CCL7 and attenuated alveolar leak. Ab neutralization studies further demonstrated a nonredundant role for IL-1ß, CXCL1, and CCL7 in mediating neutrophil recruitment in response to S. pneumoniae infection. Taken together, these data demonstrate a key role for PAR-1 during S. pneumoniae lung infection that is mediated, at least in part, by influencing multiple downstream inflammatory mediators.


Subject(s)
Neutrophils/immunology , Neutrophils/metabolism , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Receptor, PAR-1/metabolism , Animals , Blood Coagulation , Bronchoalveolar Lavage Fluid/immunology , Chemokines/metabolism , Chemotaxis/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Host-Pathogen Interactions/immunology , Inflammation Mediators/metabolism , Mice , Permeability , Pneumonia, Bacterial/blood , Pneumonia, Bacterial/pathology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/pathology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology , Receptor, PAR-1/antagonists & inhibitors , Streptococcus pneumoniae/immunology
13.
Ann Am Thorac Soc ; 12 Suppl 1: S16-20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25830828

ABSTRACT

The challenge facing many fibrotic lung diseases is that these conditions usually present late, often after several decades of repetitive alveolar epithelial injury, during which functional alveolar units are gradually obliterated and replaced with nonfunctional connective tissue. The resulting fibrosis is often progressive and, in the case of idiopathic pulmonary fibrosis (IPF), invariably leads to respiratory insufficiency and, ultimately, the premature death of affected individuals. Recent years have seen a greater appreciation of the relative importance of chronic inflammation as a driver of fibrotic responses. Current evidence suggests that IPF arises as a result of repetitive epithelial injury and a highly aberrant wound healing response in genetically susceptible and aged individuals. Nonspecific anti-inflammatory agents offer no clinical benefit, but the potential contribution of maladaptive immune responses in determining outcome is gaining increasing recognition. The importance of key differences in the tissue-regenerative potential in young versus aged individuals is also beginning to be more fully appreciated. Moreover, there is considerable overlap in the mechanisms underlying tissue repair and cancer, and patients with IPF are at heightened risk of developing lung cancer. Progressive fibrosis and cancer may therefore represent the extremes of a highly dysregulated tissue injury response. This brief review focuses on some of this evidence and on our current understanding of abnormal tissue repair responses after chronic epithelial injury in the specific context of IPF.


Subject(s)
Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/physiopathology , Pulmonary Alveoli/physiopathology , Humans , Lung Injury/therapy , Myofibroblasts/cytology , Signal Transduction
15.
Clin Sci (Lond) ; 128(4): 235-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25328010

ABSTRACT

The 2nd Cross Company Respiratory Symposium (CCRS), held in Horsham, U.K. in 2012, brought together representatives from across the pharmaceutical industry with expert academics, in the common interest of improving the design and translational predictiveness of in vivo models of respiratory disease. Organized by the respiratory representatives of the European Federation of Pharmaceutical Industries and Federations (EFPIA) group of companies involved in the EU-funded project (U-BIOPRED), the aim of the symposium was to identify state-of-the-art improvements in the utility and design of models of respiratory disease, with a view to improving their translational potential and reducing wasteful animal usage. The respiratory research and development community is responding to the challenge of improving translation in several ways: greater collaboration and open sharing of data, careful selection of the species, complexity and chronicity of the models, improved practices in preclinical research, continued refinement in models of respiratory diseases and their sub-types, greater understanding of the biology underlying human respiratory diseases and their sub-types, and finally greater use of human (and especially disease-relevant) cells, tissues and explants. The present review highlights these initiatives, combining lessons from the symposium and papers published in Clinical Science arising from the symposium, with critiques of the models currently used in the settings of asthma, idiopathic pulmonary fibrosis and COPD. The ultimate hope is that this will contribute to a more rational, efficient and sustainable development of a range of new treatments for respiratory diseases that continue to cause substantial morbidity and mortality across the world.


Subject(s)
Disease Models, Animal , Lung Diseases/pathology , Models, Biological , Translational Research, Biomedical , Animals , Drug Delivery Systems , Humans , Randomized Controlled Trials as Topic
17.
Am J Respir Cell Mol Biol ; 50(1): 144-57, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23972264

ABSTRACT

PAR1 plays a central role in mediating the interplay between coagulation and inflammation, but its role in regulating acute neutrophilic inflammation is unknown. We report that antagonism of PAR1 was highly effective at reducing acute neutrophil accumulation in a mouse model of LPS-induced lung inflammation. PAR1 antagonism also reduced alveolar-capillary barrier disruption in these mice. This protection was associated with a reduction in the expression of the chemokines, CCL2 and CCL7, but not the proinflammatory cytokines, TNF and IL-6, or the classic neutrophil chemoattractants, CXCL1 and CXCL2. Antibody neutralization of CCL2 and CCL7 significantly reduced LPS-induced total leukocyte and neutrophil accumulation, recovered from the bronchoalveolar lavage fluid of challenged mice. Immunohistochemical analysis revealed that CCL2 predominantly localized to alveolar macrophages and pulmonary epithelial cells, whereas CCL7 was restricted to the pulmonary epithelium. In keeping with these observations, the intranasal administration of recombinant CCL2 (rCCL2) and rCCL7 led to the accumulation of neutrophils within the lung airspaces of naive mice in the absence of any underlying inflammation. Flow cytometry analysis further demonstrated an increase in Ly6G(hi) neutrophils expressing the chemokine receptors, CCR1 and CCR2, isolated from mouse lungs compared with circulating neutrophils. Conversely, the expression of CXCR2 decreased on neutrophils isolated from the lung compared with circulating neutrophils. Furthermore, this switch in chemokine receptor expression was accentuated after acute LPS-induced lung inflammation. Collectively, these findings reveal a novel role for PAR1 and the chemokines, CCL2 and CCL7, during the early events of acute neutrophilic inflammation.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Neutrophils/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Receptor, PAR-1/metabolism , Animals , Chemokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Leukocytes/metabolism , Leukocytes/pathology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Inbred BALB C , Neutrophils/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Receptors, Chemokine/metabolism , Receptors, Interleukin-8B/metabolism
18.
Eur Respir J ; 42(6): 1633-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23520313

ABSTRACT

Research into the pathogenesis underlying the development of idiopathic pulmonary fibrosis is hampered by a repertoire of animal models that fail to recapitulate all the features of the human disease. Better use and understanding of what the animal models represent may improve clinical predictability. We interrogated ex vivo micro-computed tomography (CT) as a novel end-point measure in the mouse model of bleomycin-induced lung fibrosis (BILF), and to evaluate a therapeutic dosing regimen for preclinical drug evaluation. A detailed characterisation of BILF was performed using standard end-point measures (lung hydroxyproline and histology). High resolution micro-CT (∼13.7 µm voxel size) was evaluated for quantifying the extent and severity of lung fibrosis. The period from 14 to 28 days following bleomycin instillation represents progression of established fibrosis. A therapeutic dosing regimen during this period was validated using a transforming growth factor-ß receptor-1 kinase inhibitor, and micro-CT provided a highly sensitive and quantitative measure of fibrosis. Moreover, fibrotic lesions did not completely resolve, but instead persisted for ≥6 months following a single insult with bleomycin. Ex vivo micro-CT analysis of BILF allows robust evaluation of therapeutic dosing once fibrosis is already well established, requiring fewer mice than conventional biochemical end-points.


Subject(s)
Bleomycin/adverse effects , Drug Evaluation, Preclinical , Pulmonary Fibrosis/chemically induced , X-Ray Microtomography/methods , Animals , Chromatography, High Pressure Liquid , Collagen/analysis , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Imidazoles/chemistry , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinoxalines/chemistry , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Treatment Outcome
19.
Biochim Biophys Acta ; 1832(7): 1018-27, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23298546

ABSTRACT

Following tissue injury, a complex and coordinated wound healing response comprising coagulation, inflammation, fibroproliferation and tissue remodelling has evolved to nullify the impact of the original insult and reinstate the normal physiological function of the affected organ. Tissue fibrosis is thought to result from a dysregulated wound healing response as a result of continual local injury or impaired control mechanisms. Although the initial insult is highly variable for different organs, in most cases, uncontrolled or sustained activation of mesenchymal cells into highly synthetic myofibroblasts leads to the excessive deposition of extracellular matrix proteins and eventually loss of tissue function. Coagulation was originally thought to be an acute and transient response to tissue injury, responsible primarily for promoting haemostasis by initiating the formation of fibrin plugs to enmesh activated platelets within the walls of damaged blood vessels. However, the last 20years has seen a major re-evaluation of the role of the coagulation cascade following tissue injury and there is now mounting evidence that coagulation plays a critical role in orchestrating subsequent inflammatory and fibroproliferative responses during normal wound healing, as well as in a range of pathological contexts across all major organ systems. This review summarises our current understanding of the role of coagulation and coagulation initiated signalling in the response to tissue injury, as well as the contribution of uncontrolled coagulation to fibrosis of the lung, liver, kidney and heart. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.


Subject(s)
Blood Coagulation , Fibrosis , Fibrin/metabolism , Humans , Inflammation , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...