Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 249, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431872

ABSTRACT

Airway mucus is essential for lung defense, but excessive mucus in asthma obstructs airflow, leading to severe and potentially fatal outcomes. Current asthma treatments have minimal effects on mucus, and the lack of therapeutic options stems from a poor understanding of mucus function and dysfunction at a molecular level and in vivo. Biophysical properties of mucus are controlled by mucin glycoproteins that polymerize covalently via disulfide bonds. Once secreted, mucin glycopolymers can aggregate, form plugs, and block airflow. Here we show that reducing mucin disulfide bonds disrupts mucus in human asthmatics and reverses pathological effects of mucus hypersecretion in a mouse allergic asthma model. In mice, inhaled mucolytic treatment loosens mucus mesh, enhances mucociliary clearance, and abolishes airway hyperreactivity (AHR) to the bronchoprovocative agent methacholine. AHR reversal is directly related to reduced mucus plugging. These findings establish grounds for developing treatments to inhibit effects of mucus hypersecretion in asthma.


Subject(s)
Disulfides/metabolism , Hypersensitivity/physiopathology , Lung/physiopathology , Mucus/metabolism , Adolescent , Adult , Animals , Asthma/metabolism , Asthma/physiopathology , Disease Models, Animal , Expectorants/pharmacology , Female , Glycoproteins/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged
2.
Sci Transl Med ; 11(486)2019 04 03.
Article in English | MEDLINE | ID: mdl-30944166

ABSTRACT

Although destructive airway disease is evident in young children with cystic fibrosis (CF), little is known about the nature of the early CF lung environment triggering the disease. To elucidate early CF pulmonary pathophysiology, we performed mucus, inflammation, metabolomic, and microbiome analyses on bronchoalveolar lavage fluid (BALF) from 46 preschool children with CF enrolled in the Australian Respiratory Early Surveillance Team for Cystic Fibrosis (AREST CF) program and 16 non-CF disease controls. Total airway mucins were elevated in CF compared to non-CF BALF irrespective of infection, and higher densities of mucus flakes containing mucin 5B and mucin 5AC were observed in samples from CF patients. Total mucins and mucus flakes correlated with inflammation, hypoxia, and oxidative stress. Many CF BALFs appeared sterile by culture and molecular analyses, whereas other samples exhibiting bacterial taxa associated with the oral cavity. Children without computed tomography-defined structural lung disease exhibited elevated BALF mucus flakes and neutrophils, but little/no bacterial infection. Although CF mucus flakes appeared "permanent" because they did not dissolve in dilute BALF matrix, they could be solubilized by a previously unidentified reducing agent (P2062), but not N-acetylcysteine or deoxyribonuclease. These findings indicate that early CF lung disease is characterized by an increased mucus burden and inflammatory markers without infection or structural lung disease and suggest that mucolytic and anti-inflammatory agents should be explored as preventive therapy.


Subject(s)
Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mucus/metabolism , Animals , Biomarkers/metabolism , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Inflammation/pathology , Lung/microbiology , Male , Microbiota , Sheep
3.
Am J Respir Crit Care Med ; 199(2): 171-180, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30212240

ABSTRACT

RATIONALE: Airways obstruction with thick, adherent mucus is a pathophysiologic and clinical feature of muco-obstructive respiratory diseases, including chronic obstructive pulmonary disease, asthma, and cystic fibrosis (CF). Mucins, the dominant biopolymer in mucus, organize into complex polymeric networks via the formation of covalent disulfide bonds, which govern the viscoelastic properties of the mucus gel. For decades, inhaled N-acetylcysteine (NAC) has been used as a mucolytic to reduce mucin disulfide bonds with little, if any, therapeutic effects. Improvement of mucolytic therapy requires the identification of NAC deficiencies and the development of compounds that overcome them. OBJECTIVES: Elucidate the pharmacological limitations of NAC and test a novel mucin-reducing agent, P3001, in preclinical settings. METHODS: The study used biochemical (e.g., Western blotting, mass spectrometry) and biophysical assays (e.g., microrheology/macrorheology, spinnability, mucus velocity measurements) to test compound efficacy and toxicity in in vitro and in vivo models and patient sputa. MEASUREMENTS AND MAIN RESULTS: Dithiothreitol and P3001 were directly compared with NAC in vitro and both exhibited superior reducing activities. In vivo, P3001 significantly decreased lung mucus burden in ßENaC-overexpressing mice, whereas NAC did not (n = 6-24 mice per group). In NAC-treated CF subjects (n = 5), aerosolized NAC was rapidly cleared from the lungs and did not alter sputum biophysical properties. In contrast, P3001 acted faster and at lower concentrations than did NAC, and it was more effective than DNase in CF sputum ex vivo. CONCLUSIONS: These results suggest that reducing the viscoelasticity of airway mucus is an achievable therapeutic goal with P3001 class mucolytic agents.


Subject(s)
Asthma/drug therapy , Cystic Fibrosis/drug therapy , Expectorants/therapeutic use , Mucociliary Clearance/drug effects , Mucus/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Acetylcysteine/therapeutic use , Animals , Asthma/physiopathology , Cystic Fibrosis/physiopathology , Disease Models, Animal , Dithiothreitol/therapeutic use , Humans , In Vitro Techniques , Male , Mice , Pulmonary Disease, Chronic Obstructive/physiopathology
4.
Nat Commun ; 9(1): 5363, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560893

ABSTRACT

The gain-of-function MUC5B promoter variant rs35705950 is the dominant risk factor for developing idiopathic pulmonary fibrosis (IPF). Here we show in humans that MUC5B, a mucin thought to be restricted to conducting airways, is co-expressed with surfactant protein C (SFTPC) in type 2 alveolar epithelia and in epithelial cells lining honeycomb cysts, indicating that cell types involved in lung fibrosis in distal airspace express MUC5B. In mice, we demonstrate that Muc5b concentration in bronchoalveolar epithelia is related to impaired mucociliary clearance (MCC) and to the extent and persistence of bleomycin-induced lung fibrosis. We also establish the ability of the mucolytic agent P-2119 to restore MCC and to suppress bleomycin-induced lung fibrosis in the setting of Muc5b overexpression. Our findings suggest that mucociliary dysfunction might play a causative role in bleomycin-induced pulmonary fibrosis in mice overexpressing Muc5b, and that MUC5B in distal airspaces is a potential therapeutic target in humans with IPF.


Subject(s)
Genetic Predisposition to Disease , Idiopathic Pulmonary Fibrosis/genetics , Mucin-5B/genetics , Mucin-5B/metabolism , Mucociliary Clearance/genetics , Respiratory Mucosa/pathology , Animals , Bleomycin/toxicity , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Expectorants/pharmacology , Expectorants/therapeutic use , Female , Gain of Function Mutation , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucociliary Clearance/drug effects , Promoter Regions, Genetic/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...