Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Respir Crit Care Med ; 207(2): 138-149, 2023 01 15.
Article in English | MEDLINE | ID: mdl-35972987

ABSTRACT

Rationale: High circulating galectin-3 is associated with poor outcomes in patients with coronavirus disease (COVID-19). We hypothesized that GB0139, a potent inhaled thiodigalactoside galectin-3 inhibitor with antiinflammatory and antifibrotic actions, would be safely and effectively delivered in COVID-19 pneumonitis. Objectives: Primary outcomes were safety and tolerability of inhaled GB0139 as an add-on therapy for patients hospitalized with COVID-19 pneumonitis. Methods: We present the findings of two arms of a phase Ib/IIa randomized controlled platform trial in hospitalized patients with confirmed COVID-19 pneumonitis. Patients received standard of care (SoC) or SoC plus 10 mg inhaled GB0139 twice daily for 48 hours, then once daily for up to 14 days or discharge. Measurements and Main Results: Data are reported from 41 patients, 20 of which were assigned randomly to receive GB0139. Primary outcomes: the GB0139 group experienced no treatment-related serious adverse events. Incidences of adverse events were similar between treatment arms (40 with GB0139 + SoC vs. 35 with SoC). Secondary outcomes: plasma GB0139 was measurable in all patients after inhaled exposure and demonstrated target engagement with decreased circulating galectin (overall treatment effect post-hoc analysis of covariance [ANCOVA] over days 2-7; P = 0.0099 vs. SoC). Plasma biomarkers associated with inflammation, fibrosis, coagulopathy, and major organ function were evaluated. Conclusions: In COVID-19 pneumonitis, inhaled GB0139 was well-tolerated and achieved clinically relevant plasma concentrations with target engagement. The data support larger clinical trials to determine clinical efficacy. Clinical trial registered with ClinicalTrials.gov (NCT04473053) and EudraCT (2020-002230-32).


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Galectin 3 , Inflammation , Treatment Outcome
2.
Am J Respir Crit Care Med ; 203(3): 296-306, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32809843

ABSTRACT

Rationale: To examine the potential of TLR9 (Toll-like receptor 9) activation to modulate the type 2 immune response in asthma.Objectives: To evaluate efficacy and safety of AZD1419, an inhaled TLR9 agonist, in a phase 2a, randomized, double-blind trial.Methods: Adult patients with asthma with a history of elevated eosinophils (>250 cells/µl) were randomized 1:1 to receive 13 once-weekly doses of inhaled AZD1419 (1, 4, or 8 mg; n = 40) or placebo (n = 41). Inhaled corticosteroids and long-acting ß2-agonist were tapered down and then discontinued. The last four doses of AZD1419 were given without maintenance medication, followed by a 40-week observation period. Primary endpoint was time to loss of asthma control (LOC).Measurements and Main Results: AZD1419 induced a T-helper cell type 1-type IFN response with a sustained reduction in markers of type 2 inflammation. However, there were no statistically significant differences between AZD1419 and placebo for time to LOC, proportion of patients with LOC, changes in Asthma Control Questionnaire-five-item version, exacerbations, reliever use, FEV1, peak expiratory flow, or fractional exhaled nitric oxide (FeNO). LOC was predicted by an early rise in FeNO in 63% of patients. Despite withdrawal of maintenance treatment, 24 patients completed the study without LOC; AZD1419 n = 11, placebo n = 13. Adverse events were balanced across groups, with no deaths or serious adverse events judged as causally related to AZD1419.Conclusions: AZD1419 was safe and well tolerated but did not lead to improved asthma control, despite reducing markers of type 2 inflammation. Results suggest that a novel accelerated step-down approach based on FeNO is possible for patients with well-controlled asthma.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Oligonucleotides/therapeutic use , Toll-Like Receptor 9/therapeutic use , Administration, Inhalation , Adult , Aged , Anti-Asthmatic Agents/administration & dosage , Asthma/immunology , Double-Blind Method , Female , Humans , Immunologic Factors/therapeutic use , Male , Middle Aged , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/drug effects , Treatment Outcome
3.
Eur Respir J ; 57(5)2021 05.
Article in English | MEDLINE | ID: mdl-33214209

ABSTRACT

Galectin (Gal)-3 is a profibrotic ß-galactoside-binding lectin that plays a key role in the pathogenesis of idiopathic pulmonary fibrosis (IPF) and IPF exacerbations. TD139 is a novel and potent small-molecule inhibitor of Gal-3.A randomised, double-blind, multicentre, placebo-controlled, phase 1/2a study was conducted to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of inhaled TD139 in 36 healthy subjects and 24 patients with IPF. Six dose cohorts of six healthy subjects were evaluated (4:2 TD139:placebo ratio) with single doses of TD139 (0.15-50 mg) and three dose cohorts of eight patients with IPF (5:3 TD139:placebo ratio) with once-daily doses of TD139 (0.3-10 mg) for 14 days.Inhaled TD139 was well tolerated with no significant treatment-related side-effects. TD139 was rapidly absorbed, with mean time taken to reach maximum plasma concentration (C max) values ranging from 0.6 to 3 h and a plasma half-life (T 1/2) of 8 h. The concentration of TD139 in the lung was >567-fold higher than in the blood, with systemic exposure predicting exposure in the target compartment. Gal-3 expression on alveolar macrophages was reduced in the 3 and 10 mg dose groups compared with placebo, with a concentration-dependent inhibition demonstrated. Inhibition of Gal-3 expression in the lung was associated with reductions in plasma biomarkers centrally relevant to IPF pathobiology (platelet-derived growth factor-BB, plasminogen activator inhibitor-1, Gal-3, CCL18 and YKL-40).TD139 is safe and well tolerated in healthy subjects and IPF patients. It was shown to suppress Gal-3 expression on bronchoalveolar lavage macrophages and, in a concerted fashion, decrease plasma biomarkers associated with IPF progression.


Subject(s)
Galectin 3 , Idiopathic Pulmonary Fibrosis , Double-Blind Method , Humans , Lung
4.
Am J Respir Crit Care Med ; 195(11): 1494-1508, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28199128

ABSTRACT

RATIONALE: Improving the early detection and chemoprevention of lung cancer are key to improving outcomes. The pathobiology of early squamous lung cancer is poorly understood. We have shown that amplification of sex-determining region Y-box 2 (SOX2) is an early and consistent event in the pathogenesis of this disease, but its functional oncogenic potential remains uncertain. We tested the impact of deregulated SOX2 expression in a novel organotypic system that recreates the molecular and microenvironmental context in which squamous carcinogenesis occurs. OBJECTIVES: (1) To develop an in vitro model of bronchial dysplasia that recapitulates key molecular and phenotypic characteristics of the human disease; (2) to test the hypothesis that SOX2 deregulation is a key early event in the pathogenesis of bronchial dysplasia; and (3) to use the model for studies on pathogenesis and chemoprevention. METHODS: We engineered the inducible activation of oncogenes in immortalized bronchial epithelial cells. We used three-dimensional tissue culture to build an organotypic model of bronchial dysplasia. MEASUREMENTS AND MAIN RESULTS: We recapitulated human bronchial dysplasia in vitro. SOX2 deregulation drives dysplasia, and loss of tumor promoter 53 is a cooperating genetic event that potentiates the dysplastic phenotype. Deregulated SOX2 alters critical genes implicated in hallmarks of cancer progression. Targeted inhibition of AKT prevents the initiation of the dysplastic phenotype. CONCLUSIONS: In the appropriate genetic and microenvironmental context, acute deregulation of SOX2 drives bronchial dysplasia. This confirms its oncogenic potential in human cells and affords novel insights into the impact of SOX2 deregulation. This model can be used to test therapeutic agents aimed at chemoprevention.


Subject(s)
Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/physiopathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/physiopathology , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , SOXB1 Transcription Factors/genetics , Cell Culture Techniques , Humans , Models, Biological
5.
Chembiochem ; 17(18): 1759-70, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27356186

ABSTRACT

Discovery of glycan-competitive galectin-3-binding compounds that attenuate lung fibrosis in a murine model and that block intracellular galectin-3 accumulation at damaged vesicles, hence revealing galectin-3-glycan interactions involved in fibrosis progression and in intracellular galectin-3 activities, is reported. 3,3'-Bis-(4-aryltriazol-1-yl)thiodigalactosides were synthesized and evaluated as antagonists of galectin-1, -2, -3, and -4 N-terminal, -4 C-terminal, -7 and -8 N-terminal, -9 N-terminal, and -9 C-terminal domains. Compounds displaying low-nanomolar affinities for galectins-1 and -3 were identified in a competitive fluorescence anisotropy assay. X-ray structural analysis of selected compounds in complex with galectin-3, together with galectin-3 mutant binding experiments, revealed that both the aryltriazolyl moieties and fluoro substituents on the compounds are involved in key interactions responsible for exceptional affinities towards galectin-3. The most potent galectin-3 antagonist was demonstrated to act in an assay monitoring galectin-3 accumulation upon amitriptyline-induced vesicle damage, visualizing a biochemically/medically relevant intracellular lectin-carbohydrate binding event and that it can be blocked by a small molecule. The same antagonist administered intratracheally attenuated bleomycin-induced pulmonary fibrosis in a mouse model with a dose/response profile comparing favorably with that of oral administration of the marketed antifibrotic compound pirfenidone.


Subject(s)
Bleomycin , Galectin 3/metabolism , Polysaccharides/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Thioglycosides/pharmacology , Administration, Oral , Animals , Binding Sites , Disease Models, Animal , Dose-Response Relationship, Drug , Galectin 3/administration & dosage , Galectin 3/chemistry , Mice , Molecular Conformation , Polysaccharides/analysis , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Structure-Activity Relationship , Thioglycosides/administration & dosage , Thioglycosides/chemistry , Thioglycosides/therapeutic use
6.
Gut ; 64(2): 312-21, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24837171

ABSTRACT

OBJECTIVE: Following chronic liver injury or when hepatocyte proliferation is impaired, ductular reactions containing hepatic progenitor cells (HPCs) appear in the periportal regions and can regenerate the liver parenchyma. HPCs exist in a niche composed of myofibroblasts, macrophages and laminin matrix. Galectin-3 (Gal-3) is a ß-galactoside-binding lectin that binds to laminin and is expressed in injured liver in mice and humans. DESIGN: We examined the role of Gal-3 in HPC activation. HPC activation was studied following dietary induced hepatocellular (choline-deficient ethionine-supplemented diet) and biliary (3,5-diethoxycarbonyl-1,4-dihydrocollidine supplemented diet) injury in wild type and Gal-3(-/-) mice. RESULTS: HPC proliferation was significantly reduced in Gal-3(-/-) mice. Gal-3(-/-) mice failed to form a HPC niche, with reduced laminin formation. HPCs isolated from wild type mice secrete Gal-3 which enhanced adhesion and proliferation of HPCs on laminin in an undifferentiated form. These effects were attenuated in Gal3(-/-) HPCs and in wild type HPCs treated with the Gal-3 inhibitor lactose. Gal-3(-/-) HPCs in vitro showed increased hepatocyte function and prematurely upregulated both biliary and hepatocyte differentiation markers and regulated cell cycle genes leading to arrest in G0/G1. CONCLUSIONS: We conclude that Gal-3 is required for the undifferentiated expansion of HPCs in their niche in injured liver.


Subject(s)
Galectin 3/physiology , Liver/injuries , Stem Cells/pathology , Animals , Cell Adhesion/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Diet/adverse effects , Galectin 3/biosynthesis , Galectin 3/deficiency , Hepatocytes/physiology , Humans , Laminin/metabolism , Liver/metabolism , Liver/pathology , Liver Regeneration/physiology , Macrophages/metabolism , Macrophages/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Stem Cell Niche/physiology , Stem Cells/metabolism , Stem Cells/physiology , Up-Regulation
7.
Acta Cardiol ; 69(6): 595-602, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25643429

ABSTRACT

OBJECTIVE: Galectin-3 is a pro-inflammatory, pro-fibrotic molecule implicated in the pathogenesis of heart failure, and associated with poor prognostic outcome. When measured following ST-elevation myocardial infarction (STEMI), a high plasma galectin-3 predicts greater 30-day morbidity and mortality, and increased heart failure incidence at a median of 2 years. This study aims to elucidate the temporal aspects of galectin-3 expression immediately post-STEMI and how expression relates to severity of myocardial injury. METHODS: Plasma galectin-3 levels were compared in 53 STEMI patients and 23 control patients with stable angina. Consecutive plasma galectin-3 levels, measured at a mean of 30 hours (sample A) and 54 hours (sample B) post pain, and analysis of galectin-3 vs time since onset of pain/time since reperfusion allowed assessment of temporal expression in STEMI patients. Myocardial injury markers included troponin and left ventricular ejection fraction (LVEF) at primary percutaneous coronary intervention (PCI). RESULTS: Circulating galectin-3 levels were significantly higher in STEMI patients than control patients when measured at a mean of 30 hours post pain (t = 2.72, df = 66, P = 0.008). However, levels had significantly decreased when measured 24 hours later (t = 2.13, df = 47, P = 0.039), with a negative linear relationship apparent between plasma galectin-3 levels and time since reperfusion on univariate analysis (OR = 0.871, 95% CI = 0.779-0.975, P = 0.021). A significantly lower circulating galectin-3 concentration was also found for sample A in those reperfused within 3 hours post-onset of pain (OR 0.045, 95% CI 0.003-0.669, P = 0.029). CONCLUSIONS: Plasma galectin-3 levels vary significantly following a STEMI over a short time period, in relation to timing of reperfusion.


Subject(s)
Galectin 3/blood , Myocardial Infarction/blood , Biomarkers/blood , Case-Control Studies , Electrocardiography , Female , Humans , Male , Middle Aged , Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Prognosis , Risk Factors , Time Factors , Troponin/blood , Ventricular Function, Left
8.
Glycobiology ; 23(6): 654-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23426722

ABSTRACT

Atherosclerosis is a major risk factor for cardiovascular disease (CVD) and stroke. Galectin-3 is a carbohydrate-binding lectin implicated in the pathophysiology of CVD and is highly expressed within atherosclerotic lesions in mice and humans. The object of this present study was to use genetic deletion and pharmacological inhibition in a well-characterized mouse model of atherosclerosis to determine the role of galectin-3 in plaque development. Apolipoprotein-E/galectin-3 knockout mice were generated and fed a high-cholesterol "western" diet. Galectin-3 deletion had no consistent effect on the serum lipid profile but halved atherosclerotic lesion formation in the thoracic aorta (57% reduction), the aortic arch (50% reduction) and the brachiocephalic arteries. The aortic plaques were smaller, with reduced lipid core and less collagen. In apolipoprotein E-deficient (ApoE(-/-)) mice, there was a switch from high inducible nitric oxide expression in early lesions (6 weeks) to arginase-1 expression in later lesions (20 weeks), which was reversed in ApoE(-/-)/gal-3(-/-) mice. Administration of modified citrus pectin, an inhibitor of galectin-3, during the latter stage of the disease reduced plaque volume. We conclude that inhibiting galectin-3 causes decreased atherosclerosis. Strategies to inhibit galectin-3 function may reduce plaque progression and potentially represent a novel therapeutic strategy in the treatment of atherosclerotic disease.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Galectin 3/antagonists & inhibitors , Pectins/pharmacology , Plaque, Atherosclerotic/prevention & control , Animals , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Arginase/metabolism , Arginine/metabolism , Atherosclerosis/blood , Cell Line , Cell Movement , Fatty Acids, Nonesterified/blood , Galectin 3/genetics , Galectin 3/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/pathology , Triglycerides/blood , Weight Gain
9.
Am J Respir Crit Care Med ; 185(5): 537-46, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22095546

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a chronic dysregulated response to alveolar epithelial injury with differentiation of epithelial cells and fibroblasts into matrix-secreting myofibroblasts resulting in lung scaring. The prognosis is poor and there are no effective therapies or reliable biomarkers. Galectin-3 is a ß-galactoside binding lectin that is highly expressed in fibrotic tissue of diverse etiologies. OBJECTIVES: To examine the role of galectin-3 in pulmonary fibrosis. METHODS: We used genetic deletion and pharmacologic inhibition in well-characterized murine models of lung fibrosis. Further mechanistic studies were performed in vitro and on samples from patients with IPF. MEASUREMENTS AND MAIN RESULTS: Transforming growth factor (TGF)-ß and bleomycin-induced lung fibrosis was dramatically reduced in mice deficient in galectin-3, manifest by reduced TGF-ß1-induced EMT and myofibroblast activation and collagen production. Galectin-3 reduced phosphorylation and nuclear translocation of ß-catenin but had no effect on Smad2/3 phosphorylation. A novel inhibitor of galectin-3, TD139, blocked TGF-ß-induced ß-catenin activation in vitro and in vivo and attenuated the late-stage progression of lung fibrosis after bleomycin. There was increased expression of galectin-3 in the bronchoalveolar lavage fluid and serum from patients with stable IPF compared with nonspecific interstitial pneumonitis and controls, which rose sharply during an acute exacerbation suggesting that galectin-3 may be a marker of active fibrosis in IPF and that strategies that block galectin-3 may be effective in treating acute fibrotic exacerbations of IPF. CONCLUSIONS: This study identifies galectin-3 as an important regulator of lung fibrosis and provides a proof of principle for galectin-3 inhibition as a potential novel therapeutic strategy for IPF.


Subject(s)
Galectin 3/physiology , Idiopathic Pulmonary Fibrosis/physiopathology , Transforming Growth Factor beta1/physiology , Animals , Bleomycin/toxicity , Collagen/biosynthesis , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/physiology , Fluorescent Antibody Technique , Galectin 3/antagonists & inhibitors , Gene Knockout Techniques , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism
10.
Immunol Rev ; 230(1): 160-71, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19594635

ABSTRACT

Galectin-3 is a beta-galactoside-binding animal lectin of approximately 30 kDa and is evolutionarily highly conserved. Galectin-3 is promiscuous, its localization within the tissue micro-environment may be extracellular, cytoplasmic, or nuclear, and it has a concentration-dependent ability to be monomeric or form oligomers. These properties impart great flexibility on galectin-3 as a specific regulator of many biological systems including inflammation. For example, in acute tissue damage galectin-3 is a key component in the host defense against microbes such as Streptococcus pneumoniae. However, if tissue injury becomes repetitive galectin-3 also appears to be intimately involved in the transition to chronic inflammation, facilitating the walling off of tissue injury with fibrogenesis and organ scarring. Therefore galectin-3 can be viewed as a regulatory molecule acting at various stages along the continuum from acute inflammation to chronic inflammation and tissue fibrogenesis. In this review, we examine the role of galectin-3 in inflammation, and discuss the manipulation of galectin-3 expression as a potentially novel therapeutic strategy in the treatment of a broad range of inflammatory diseases.


Subject(s)
Asthma/immunology , Fibroblasts/immunology , Fibrosis/immunology , Galectin 3/metabolism , Inflammation/immunology , Macrophages/immunology , Animals , Asthma/metabolism , Bacteria/immunology , Fibroblasts/metabolism , Fibrosis/metabolism , Fungi/immunology , Galectin 3/immunology , Humans , Immunity, Innate , Inflammation/metabolism , Inflammation/microbiology , Macrophages/metabolism
11.
Int J Cancer ; 131(6): E928-37, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22532287

ABSTRACT

Small cell lung cancer (SCLC) kills at least one person every 2 hr in the United Kingdom. Some patients do relatively well but most have rapidly progressive disease. There is no effective treatment and overall 2-year survival is less than 5%. Patients with SCLC have poorly understood local and systemic immune defects and can be immunocompromised. As CD4(+) T lymphocytes coordinate and regulate immunity, a better understanding of interactions between SCLC tumour cells and CD4(+) T cells may lead to effective molecular immunotherapy. We show that some, but not all, SCLC tumour cell lines secrete molecules that induce IL-10 secretion by and de novo differentiation of functional CD4(+)CD25(+)FOXP3(+)CD127(lo)Helios(-) regulatory T (Treg) cells in healthy blood lymphocytes. FOXP3(+) T cells were found in SCLC tumour biopsies, and patients with higher ratios of FOXP3(+) cells in tumour infiltrates have a worse survival rate. The inhibitory effect of SCLC tumour cells was not affected by blocking IL-10 receptor or TGF-ß signalling but was partially reversed by blocking IL-15, which is reported to be involved in human Treg cells induction. IL-15 was secreted by SCLC cells that inhibited CD4(+) T-cell proliferation and was present in SCLC biopsy tumour cells. These novel findings demonstrate that SCLC tumour cells can induce CD4(+) T-cell-mediated immunosuppression. This gives a potential mechanism by which SCLC tumour cells may downregulate local and systemic immune responses and contribute to poor patient survival. Our data suggest that IL-15 and Treg cells are potential new therapeutic targets to improve immune response and patient survival in SCLC.


Subject(s)
Carcinoma, Small Cell/immunology , Forkhead Transcription Factors/analysis , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/pathology , Cell Cycle , Cell Line, Tumor , Humans , Interleukin-10/physiology , Interleukin-15/physiology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphocyte Activation , Transforming Growth Factor beta/physiology
12.
J Cell Sci ; 123(Pt 1): 51-61, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20016066

ABSTRACT

The integrin family of heterodimeric cell-surface receptors are fundamental in cell-cell and cell-matrix adhesion. Changes to either integrin-ligand affinity or integrin gene expression are central to a variety of disease processes, including inflammation, cardiovascular disease and cancer. In screening for novel activators of integrin-ligand affinity we identified the previously uncharacterised multi-transmembrane domain protein Fam38A, located at the endoplasmic reticulum (ER). siRNA knockdown of Fam38A in epithelial cells inactivates endogenous beta1 integrin, reducing cell adhesion. Fam38A mediates integrin activation by recruiting the small GTPase R-Ras to the ER, which activates the calcium-activated protease calpain by increasing Ca(2+) release from cytoplasmic stores. Fam38A-induced integrin activation is blocked by inhibition of either R-Ras or calpain activity, or by siRNA knockdown of talin, a well-described calpain substrate. This highlights a novel mechanism for integrin activation by Fam38A, utilising calpain and R-Ras signalling from the ER. These data represent the first description of a novel spatial regulator of R-Ras, of an alternative integrin activation-suppression pathway based on direct relocalisation of R-Ras to the ER, and of a mechanism linking R-Ras and calpain signalling from the ER with modulation of integrin-ligand affinity.


Subject(s)
Epithelial Cells/metabolism , Integrin beta1/metabolism , Membrane Proteins/metabolism , Animals , CHO Cells , Calcium Signaling/genetics , Calpain/antagonists & inhibitors , Cell Adhesion/genetics , Cricetinae , Cricetulus , Endoplasmic Reticulum , Epithelial Cells/cytology , HeLa Cells , Humans , Integrin beta1/genetics , Ion Channels , Membrane Proteins/genetics , Protein Transport/genetics , RNA, Small Interfering/genetics , Talin/genetics , ras Proteins/antagonists & inhibitors
13.
Am J Respir Crit Care Med ; 184(5): 569-81, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21680953

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a devastating disease. Antiinflammatory therapies, including corticosteroids, are of no benefit. The role of monocytes and macrophages is therefore controversial. OBJECTIVES: To define the role of monocytes and macrophages during lung fibrogenesis and resolution, and explore the phenotype of the cells involved. METHODS: We used multiple in vivo depletional strategies, backed up by adoptive transfer techniques. Further studies were performed on samples from patients with IPF. MEASUREMENTS AND MAIN RESULTS: Depletion of lung macrophages during fibrogenesis reduced pulmonary fibrosis as measured by lung collagen (P = 0.0079); fibrosis score (P = 0.0051); and quantitative polymerase chain reaction for surrogate markers of fibrosis Col1 (P = 0.0083) and a-smooth muscle actin (P = 0.0349). There was an associated reduction in markers of the profibrotic alternative macrophage activation phenotype, Ym1 (P = 0.0179), and Arginase 1. The alternative macrophage marker CD163 was expressed on lung macrophages from patients with IPF. Depletion of Ly6Chi circulating monocytes reduced pulmonary fibrosis (P = 0.0052) and the number of Ym1- positive alternatively activated lung macrophages (P = 0.0310). Their adoptive transfer during fibrogenesis exacerbated fibrosis (P = 0.0304); however, adoptively transferred CD45.1 Ly6Chi cells were not found in the lungs of recipient CD45.2 mice. CONCLUSIONS: We demonstrate the importance of circulating monocytes and lung macrophages during pulmonary fibrosis, and emphasize the importance of the alternatively activated macrophage phenotype. We show that Ly6Chi monocytes facilitate the progression of pulmonary fibrosis, but are not obviously engrafted into lungs thereafter. Finally, we provide empirical data to suggest that macrophages may have a resolution-promoting role during the reversible phase of bleomycin-induced pulmonary fibrosis.


Subject(s)
Immunity, Cellular , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Monocytes/physiology , Pulmonary Fibrosis/etiology , Animals , Bleomycin/toxicity , Disease Models, Animal , Disease Progression , Female , Humans , Macrophage Activation/genetics , Macrophages, Alveolar/pathology , Mice , Phenotype , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
14.
Front Pharmacol ; 13: 949264, 2022.
Article in English | MEDLINE | ID: mdl-36003515

ABSTRACT

Rationale: Galectin-3 (Gal-3) drives fibrosis during chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Effective pharmacological therapies available for ALI are limited; identifying novel concepts in treatment is essential. GB0139 is a Gal-3 inhibitor currently under clinical investigation for the treatment of idiopathic pulmonary fibrosis. We investigate the role of Gal-3 in ALI and evaluate whether its inhibition with GB0139 offers a protective role. The effect of GB0139 on ALI was explored in vivo and in vitro. Methods: The pharmacokinetic profile of intra-tracheal (i.t.) GB0139 was investigated in C57BL/6 mice to support the daily dosing regimen. GB0139 (1-30 µg) was then assessed following acute i.t. lipopolysaccharide (LPS) and bleomycin administration. Histology, broncho-alveolar lavage fluid (BALf) analysis, and flow cytometric analysis of lung digests and BALf were performed. The impact of GB0139 on cell activation and apoptosis was determined in vitro using neutrophils and THP-1, A549 and Jurkat E6 cell lines. Results: GB0139 decreased inflammation severity via a reduction in neutrophil and macrophage recruitment and neutrophil activation. GB0139 reduced LPS-mediated increases in interleukin (IL)-6, tumor necrosis factor alpha (TNFα) and macrophage inflammatory protein-1-alpha. In vitro, GB0139 inhibited Gal-3-induced neutrophil activation, monocyte IL-8 secretion, T cell apoptosis and the upregulation of pro-inflammatory genes encoding for IL-8, TNFα, IL-6 in alveolar epithelial cells in response to mechanical stretch. Conclusion: These data indicate that Gal-3 adopts a pro-inflammatory role following the early stages of lung injury and supports the development of GB0139, as a potential treatment approach in ALI.

15.
J Immunol ; 182(5): 2795-807, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234174

ABSTRACT

Dendritic cells (DCs) have a central role in the development of adaptive immune responses, including antitumor immunity. Factors present in the tumor milieu can alter the maturation of DCs and inhibit their capacity to activate T cells. Using gene expression analysis, we found that human DCs increased the expression of TGF-beta1 transcripts following culture with human lung carcinoma cells (LCCs). These DCs produced increased amounts of TGF-beta1 protein compared with DCs not exposed to tumor cells. LCCs also decreased the expression of CD86 and HLA-DR by immature DCs. Furthermore, LCCs decreased CD86 expression and the production of TNF-alpha and IL-12 p70 by mature DCs. Moreover, LCCs also converted mature DCs into cells producing TGF-beta1. These TGF-beta1-producing DCs were poor at eliciting the activation of naive CD4(+) T cells and sustaining their proliferation and differentiation into Th1 (IFN-gamma(+)) effectors. Instead, TGF-beta1-producing DCs demonstrated an increased ability to generate CD4(+)CD25(+)Foxp3(+) regulatory T cells that suppress the proliferation of T lymphocytes. These results identify a novel mechanism by which the function of human DCs is altered by tumor cells and contributes to the evasion of the immune response.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , B7-2 Antigen/biosynthesis , B7-2 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/pathology , Down-Regulation/immunology , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/metabolism , Humans , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Lung Neoplasms/metabolism , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
16.
Respiration ; 81(2): 89-104, 2011.
Article in English | MEDLINE | ID: mdl-21311210

ABSTRACT

Chronic obstructive pulmonary disease (COPD) and lung cancer currently form the basis for an enormous disease burden in the developed world. As a result of changing smoking trends and tobacco use, regrettably, a similar picture is arising rapidly within the developing world. COPD is a recognised risk factor for lung cancer, and a significant proportion of patients diagnosed with lung cancer have COPD. An association between both conditions has long been suspected but has proven difficult to demonstrate thus far. However, the common factors between both conditions are now becoming apparent thanks to recent clinical and molecular advances. Abnormal regulation of the immune system and the establishment of chronic inflammation appear to be key events in this process. In addition, the complex interplay between genes and environment and the possibility of a genetic basis to lung cancer susceptibility in the context of COPD are becoming clearer concepts. As we begin to unravel the common pathways and molecules in the pathogenesis of both conditions, we may be able to not only identify novel strategies to prevent and treat COPD and lung cancer, but also recognise molecular markers to identify patients at high risk of developing lung cancer.


Subject(s)
Carcinoma/etiology , Lung Neoplasms/etiology , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Genetic Predisposition to Disease , Humans , Inflammation/complications
17.
Front Pharmacol ; 12: 715986, 2021.
Article in English | MEDLINE | ID: mdl-34526900

ABSTRACT

Rationale: Galectin-3 (Gal-3) is an immune regulator and an important driver of fibrosis in chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Previous work has shown that global deletion of galectin-3 reduces collagen deposition in a bleomycin-induced pulmonary fibrosis model (MacKinnon et al., Am. J. Respir. Crit. Care Med., 2012, 185, 537-46). An inhaled Gal-3 inhibitor, GB0139, is undergoing Phase II clinical development for idiopathic pulmonary fibrosis (IPF). This work aims to elucidate the role of Gal-3 in the myeloid and mesenchymal compartment on the development of acute and chronic lung injury. Methods: LgalS3 fl/fl mice were generated and crossed with mice expressing the myeloid (LysM) and mesenchymal (Pdgfrb) cre drivers to yield LysM-cre +/- /LgalS3 fl/fl and Pdgfrb-cre +/- /LgalS3 fl/fl mice. The response to acute (bleomycin or LPS) or chronic (bleomycin) lung injury was compared to globally deficient Gal-3 -/- mice. Results: Myeloid depletion of Gal-3 led to a significant reduction in Gal-3 expression in alveolar macrophages and neutrophils and a reduction in neutrophil recruitment into the interstitium but not into the alveolar space. The reduction in interstitial neutrophils corelated with decreased levels of pulmonary inflammation following acute bleomycin and LPS administration. In addition, myeloid deletion decreased Gal-3 levels in bronchoalveolar lavage (BAL) and reduced lung fibrosis induced by chronic bleomycin. In contrast, no differences in BAL Gal-3 levels or fibrosis were observed in Pdgfrb-cre +/- /LgalS3 fl/fl mice. Conclusions: Myeloid cell derived Galectin-3 drives acute and chronic lung inflammation and supports direct targeting of galectin-3 as an attractive new therapy for lung inflammation.

19.
Br Med Bull ; 95: 47-61, 2010.
Article in English | MEDLINE | ID: mdl-20643690

ABSTRACT

INTRODUCTION: Lung cancer is the commonest fatal malignancy in the developed world. Survival rates for lung cancer have not changed significantly over the past 30 years. Sources of data This report is a systematic review of the literature on our current understanding of lung cancer biology. Searches were carried out using PUBMED. 1990-2010. AREAS OF AGREEMENT: A concerted effort to reduce cigarette smoking and nicotine addiction is required. A better understanding of the biology of lung cancer will lead to the identification of earlier diagnostic markers and improved therapy. AREAS OF CONTROVERSY: How chronic inflammatory disorders such as COPD and lung fibrosis contribute to lung cancer development is incompletely understood. GROWING POINTS: Developing novel biological agents to target lung cancer. New microarray-based technologies provide new methods for predicting prognosis and response to treatment. AREAS TIMELY FOR DEVELOPING RESEARCH: Developing strategies to target lung cancer stem cells may provide a novel approach for treating drug resistant disease.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Bronchogenic/therapy , Lung Neoplasms/therapy , Signal Transduction/genetics , Smoking/adverse effects , Carcinoma, Bronchogenic/genetics , Cell Adhesion , Drug Resistance, Neoplasm/genetics , Humans , Lung Neoplasms/genetics
20.
Respir Med ; 173: 106175, 2020 11.
Article in English | MEDLINE | ID: mdl-33032168

ABSTRACT

BACKGROUND: Frequency of moderate and severe chronic obstructive pulmonary disease exacerbations is an important endpoint in clinical trials, but makes them large and lengthy when powered to evaluate it. We aimed to develop a composite endpoint (COPDCompEx) that could predict treatment effect on exacerbations, enabling the design of shorter early phase clinical trials requiring fewer patients. METHODS: In this post hoc analysis, data from 20 randomized controlled trials were used to develop and test COPDCompEx. Diary events were tested against predefined threshold values for peak expiratory flow, reliever medication use, and symptoms. A COPDCompEx event was defined as first occurrence of a diary event, a moderate or severe exacerbation, or a study dropout. Ratios of event frequency, treatment effect and future trial sample size were compared between COPDCompEx and moderate and severe exacerbations. FINDINGS: At 3 months, the proportion of patients experiencing COPDCompEx events increased over 3-fold versus exacerbations alone. All components contributed to COPDCompEx event rate. Treatment effects at 3 months were closely matched between COPDCompEx and exacerbations, and the large net gain in power substantially reduced the required sample size. INTERPRETATION: COPDCompEx may be used to predict treatment effect on moderate and severe exacerbations of chronic obstructive pulmonary disease. This may enable the design of shorter Phase 2 clinical trials requiring fewer patients when compared with current exacerbation studies, with exacerbations as a key Phase 3 endpoint. This would, therefore, allow more efficient decision-making with reduced burden and risk to study participants.


Subject(s)
Endpoint Determination/methods , Pulmonary Disease, Chronic Obstructive/drug therapy , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Disease Progression , Female , Humans , Male , Randomized Controlled Trials as Topic , Sample Size , Severity of Illness Index , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL