Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34548396

RESUMEN

Elevated levels of MUC5AC, one of the major gel-forming mucins in the lungs, are closely associated with chronic obstructive lung diseases such as chronic bronchitis and asthma. It is not known, however, how the structure and/or gel-making properties of MUC5AC contribute to innate lung defense in health and drive the formation of stagnant mucus in disease. To understand this, here we studied the biophysical properties and macromolecular assembly of MUC5AC compared to MUC5B. To study each native mucin, we used Calu3 monomucin cultures that produced MUC5AC or MUC5B. To understand the macromolecular assembly of MUC5AC through N-terminal oligomerization, we expressed a recombinant whole N-terminal domain (5ACNT). Scanning electron microscopy and atomic force microscopy imaging indicated that the two mucins formed distinct networks on epithelial and experimental surfaces; MUC5B formed linear, infrequently branched multimers, whereas MUC5AC formed tightly organized networks with a high degree of branching. Quartz crystal microbalance-dissipation monitoring experiments indicated that MUC5AC bound significantly more to hydrophobic surfaces and was stiffer and more viscoelastic as compared to MUC5B. Light scattering analysis determined that 5ACNT primarily forms disulfide-linked covalent dimers and higher-order oligomers (i.e., trimers and tetramers). Selective proteolytic digestion of the central glycosylated region of the full-length molecule confirmed that MUC5AC forms dimers and higher-order oligomers through its N terminus. Collectively, the distinct N-terminal organization of MUC5AC may explain the more adhesive and unique viscoelastic properties of branched, highly networked MUC5AC gels. These properties may generate insight into why/how MUC5AC forms a static, "tethered" mucus layer in chronic muco-obstructive lung diseases.


Asunto(s)
Células Epiteliales/metabolismo , Mucina 5AC/química , Mucina 5AC/metabolismo , Mucina 5B/química , Mucina 5B/metabolismo , Mucosa Respiratoria/metabolismo , Células Cultivadas , Células Epiteliales/citología , Humanos , Mucosa Respiratoria/citología
2.
Glycobiology ; 31(4): 436-443, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33083824

RESUMEN

Understanding the basic elements of the airway mucosal surfaces and how they form a functional barrier is essential in understanding disease initiation, progression, pathogenesis and ultimately treating chronic lung diseases. Using primary airway epithelial cell cultures, atomic force microscopy (AFM), multiangle light scattering and quartz crystal micro balance with dissipation monitoring techniques, here we report that the membrane bound mucins (MBMs) found in the periciliary layer (PCL) of the airway surface are densely decorated with keratan sulfate (KS). AFM and immunoblotting show that the KS sidechains can be removed enzymatically with keratanase II (KII) treatment, and the antibody accessibility for B2729 (MUC1), MUCH4 (MUC4) and OC125 (MUC16) was substantially enhanced. Light scattering analysis confirmed that KII treatment removed ~40% of the mass from the mucin fractions. Surface binding experiments indicated that MBMs were able to pack into a tighter conformation following KS removal, suggesting that negatively charged KS sidechains play a role in mucin-mucin repulsion and contribute to "space filling" in the PCL. We also observed that soluble filtrate from the common airway pathogen Pseudomonas aeruginosa is capable of stripping KS from MBMs. Altogether, our findings indicate that KS glycosylation of MBMs may play an important role in the integrity of the airway mucosal barrier and its compromise in disease.


Asunto(s)
Sulfato de Queratano , Mucinas , Glicosilación , Sulfato de Queratano/metabolismo , Pulmón/metabolismo , Mucinas/metabolismo
3.
Am J Respir Cell Mol Biol ; 63(6): 767-779, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32877614

RESUMEN

Smoking remains a leading cause of preventable morbidity and mortality worldwide. Despite a downward trend in cigarette use, less-regulated tobacco products, such as cigarillos, which are often flavored to appeal to specific demographics, such as younger people, are becoming increasingly popular. Cigar/cigarillo smoking has been considered a safer alternative to cigarettes; however, the health risks associated with cigar in comparison with cigarette smoking are not well understood. To address this knowledge gap, we characterized the effects of multiple brands of cigarillos on the airway epithelium using ex vivo and in vivo models. To analyze these effects, we assessed the cellular viability and integrity of smoke-exposed primary airway cell cultures. We also investigated the protein compositions of apical secretions from cigarillo-exposed airway epithelial cultures and BAL fluid of cigarillo-exposed mice through label-free quantitative proteomics and determined the chemical composition of smoke collected from the investigated cigarillo products. We found that cigarillo smoke exerts similar or greater effects than cigarette smoke in terms of reduced cell viability; altered protein levels, including those of innate immune proteins; induced oxidative-stress markers; and greater nicotine delivery to cells. The analysis of the chemical composition of the investigated cigarillo products revealed differences that might be linked to the differential effects of these products on cell viability and protein abundance profiles, which have been associated with a range of health risks in the context of airway biology. These findings contradict the assumption that cigarillos might be safer and less harmful than cigarettes. Instead, our results indicate that cigarillo smoke is associated with equal or greater health risks and the same or increased airway toxicity compared with cigarette smoke.


Asunto(s)
Epitelio/efectos de los fármacos , Epitelio/metabolismo , Nicotina/farmacología , Sistema Respiratorio/metabolismo , Animales , Fumar Cigarrillos/efectos adversos , Aromatizantes/farmacología , Humanos , Ratones Endogámicos C57BL , Sistema Respiratorio/efectos de los fármacos , Fumar/efectos adversos , Nicotiana/efectos adversos , Productos de Tabaco/efectos adversos
4.
Am J Respir Cell Mol Biol ; 60(2): 209-220, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30230353

RESUMEN

Airway epithelium structure/function can be altered by local inflammatory/immune signals, and this process is called epithelial remodeling. The mechanism by which this innate response is regulated, which causes mucin/mucus overproduction, is largely unknown. Exosomes are nanovesicles that can be secreted and internalized by cells to transport cellular cargo, such as proteins, lipids, and miRNA. The objective of this study was to understand the role exosomes play in airway remodeling through cell-cell communication. We used two different human airway cell cultures: primary human tracheobronchial (HTBE) cells, and a cultured airway epithelial cell line (Calu-3). After intercellular exosomal transfer, comprehensive proteomic and genomic characterization of cell secretions and exosomes was performed. Quantitative proteomics and exosomal miRNA analysis profiles indicated that the two cell types are fundamentally distinct. HTBE cell secretions were typically dominated by fundamental innate/protective proteins, including mucin MUC5B, and Calu-3 cell secretions were dominated by pathology-associated proteins, including mucin MUC5AC. After exosomal transfer/intake, approximately 20% of proteins, including MUC5AC and MUC5B, were significantly altered in HTBE secretions. After exosome transfer, approximately 90 miRNAs (∼4%) were upregulated in HTBE exosomes, whereas Calu-3 exosomes exhibited a preserved miRNA profile. Together, our data suggest that the transfer of exosomal cargo between airway epithelial cells significantly alters the qualitative and quantitative profiles of airway secretions, including mucin hypersecretion, and the miRNA cargo of exosomes in target cells. This finding indicates that cellular information can be carried between airway epithelial cells via exosomes, which may play an important role in airway biology and epithelial remodeling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Bronquios/citología , Comunicación Celular/fisiología , Células Epiteliales/metabolismo , Vesículas Extracelulares/metabolismo , Línea Celular , Células Cultivadas , Medios de Cultivo/metabolismo , Células Epiteliales/citología , Exosomas/metabolismo , Vesículas Extracelulares/genética , Expresión Génica , Humanos , MicroARNs , Mucina 5AC/metabolismo , Mucina 5B/metabolismo , Proteínas/análisis , Proteínas/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L910-L918, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30211652

RESUMEN

Mucociliary clearance (MCC) plays an essential role in maintaining airway sterility and health. Conversely, mucociliary dysfunction is implicated across many airway obstructive diseases. Understanding the necessary requirements for successful MCC is imperative to establish the pathology of disease, as well as to develop therapeutic strategies. Although postural, that is, gravitational, drainage is used clinically to aid mucus clearance, it is ignored in both animal and cell culture models of MCC. In this study, we develop a novel mucus clearance assay that enables the first particle image velocimetry of human bronchial epithelial cell cultures tilted relative to the gravitational field. This tilting system makes it possible to observe drainage of the airway surface liquid and, thus, reveals the effect gravity has on mucociliary clearance. First, we use this assay to demonstrate that beating cilia alone cannot transport buffer upward against gravity. Next, we show the same cilia successfully transporting mucus upward. These results indicate that the biophysical and biochemical properties of mucus enable vertical clearance and that current assay systems are not equipped to determine which properties are required for physiologically relevant vertical mucociliary clearance.


Asunto(s)
Depuración Mucociliar/fisiología , Mucosa Respiratoria/fisiología , Células Cultivadas , Cilios/fisiología , Drenaje/métodos , Células Epiteliales/fisiología , Humanos , Reología/métodos
6.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L192-L205, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982737

RESUMEN

In cystic fibrosis (CF) lungs, epithelial Na+ channel (ENaC) hyperactivity causes a reduction in airway surface liquid volume, leading to decreased mucocilliary clearance, chronic bacterial infection, and lung damage. Inhibition of ENaC is an attractive therapeutic option. However, ENaC antagonists have failed clinically because of off-target effects in the kidney. The S18 peptide is a naturally occurring short palate lung and nasal epithelial clone 1 (SPLUNC1)-derived ENaC antagonist that restores airway surface liquid height for up to 24 h in CF human bronchial epithelial cultures. However, its efficacy and safety in vivo are unknown. To interrogate the potential clinical efficacy of S18, we assessed its safety and efficacy using human airway cultures and animal models. S18-mucus interactions were tested using superresolution microscopy, quartz crystal microbalance with dissipation, and confocal microscopy. Human and murine airway cultures were used to measure airway surface liquid height. Off-target effects were assessed in conscious mice and anesthetized rats. Morbidity and mortality were assessed in the ß-ENaC-transgenic (Tg) mouse model. Restoration of normal mucus clearance was measured in cystic fibrosis transmembrane conductance regulator inhibitor 172 [CFTR(inh)-172]-challenged sheep. We found that S18 does not interact with mucus and rapidly penetrated dehydrated CF mucus. Compared with amiloride, an early generation ENaC antagonist, S18 displayed a superior ability to slow airway surface liquid absorption, reverse CFTR(inh)-172-induced reduction of mucus transport, and reduce morbidity and mortality in the ß-ENaC-Tg mouse, all without inducing any detectable signs of renal toxicity. These data suggest that S18 is the first naturally occurring ENaC antagonist to show improved preclinical efficacy in animal models of CF with no signs of renal toxicity.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Glicoproteínas/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Péptidos/farmacología , Fosfoproteínas/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Animales , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Transporte Iónico , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Masculino , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/metabolismo
7.
Methods Mol Biol ; 2763: 51-59, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38347399

RESUMEN

Membrane-bound mucins constitute a large portion of the periciliary layer of lung epithelial surfaces, and thus play an important role in many aspects of innate defense. The biophysical and biochemical properties of the membrane-bound mucins have important implications for mucociliary clearance, viral penetration, and potential therapeutics delivered to the airway surface. Hence, isolating them and determining these properties is important in understanding airways disease and ultimately in developing treatments. Here, we describe a method using isopycnic centrifugation to enrich and isolate shed membrane-bound mucins from the washings of human bronchial epithelial cell cultures.


Asunto(s)
Células Epiteliales , Mucinas , Humanos , Mucinas/metabolismo , Células Epiteliales/metabolismo , Membranas/metabolismo , Pulmón/metabolismo
8.
Methods Mol Biol ; 2763: 361-371, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38347426

RESUMEN

Mucin networks serve as the structural scaffold of mucus and play a significant role in determining its biophysical properties. Thus, characterizing the organization, macromolecular structure, and interactions within these networks is a key step in understanding the parameters that govern mucus functionality in both health and disease. Atomic force microscopy (AFM) is uniquely suited to study mucin networks; AFM can clearly resolve nanometer-sized features, does not require fixation or metallization, and can be performed in air or aqueous solutions. In this chapter we describe protocols to image mucin networks using AFM. First, we describe two protocols to enrich and isolate mucin samples in preparation for AFM imaging. Next, we detail a protocol to deposit the samples onto a mica substrate. Finally, we give general tips to optimize and troubleshoot AFM imaging of mucin networks.


Asunto(s)
Mucinas , Microscopía de Fuerza Atómica/métodos , Estructura Molecular
9.
mBio ; 15(6): e0345123, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38651896

RESUMEN

The airway milieu of individuals with muco-obstructive airway diseases (MADs) is defined by the accumulation of dehydrated mucus due to hyperabsorption of airway surface liquid and defective mucociliary clearance. Pathological mucus becomes progressively more viscous with age and disease severity due to the concentration and overproduction of mucin and accumulation of host-derived extracellular DNA (eDNA). Respiratory mucus of MADs provides a niche for recurrent and persistent colonization by respiratory pathogens, including Pseudomonas aeruginosa, which is responsible for the majority of morbidity and mortality in MADs. Despite high concentration inhaled antibiotic therapies and the absence of antibiotic resistance, antipseudomonal treatment failure in MADs remains a significant clinical challenge. Understanding the drivers of antibiotic tolerance is essential for developing more effective treatments that eradicate persistent infections. The complex and dynamic environment of diseased airways makes it difficult to model antibiotic efficacy in vitro. We aimed to understand how mucin and eDNA concentrations, the two dominant polymers in respiratory mucus, alter the antibiotic tolerance of P. aeruginosa. Our results demonstrate that polymer concentration and molecular weight affect P. aeruginosa survival post antibiotic challenge. Polymer-driven antibiotic tolerance was not explicitly associated with reduced antibiotic diffusion. Lastly, we established a robust and standardized in vitro model for recapitulating the ex vivo antibiotic tolerance of P. aeruginosa observed in expectorated sputum across age, underlying MAD etiology, and disease severity, which revealed the inherent variability in intrinsic antibiotic tolerance of host-evolved P. aeruginosa populations. IMPORTANCE: Antibiotic treatment failure in Pseudomonas aeruginosa chronic lung infections is associated with increased morbidity and mortality, illustrating the clinical challenge of bacterial infection control. Understanding the underlying infection environment, as well as the host and bacterial factors driving antibiotic tolerance and the ability to accurately recapitulate these factors in vitro, is crucial for improving antibiotic treatment outcomes. Here, we demonstrate that increasing concentration and molecular weight of mucin and host eDNA drive increased antibiotic tolerance to tobramycin. Through systematic testing and modeling, we identified a biologically relevant in vitro condition that recapitulates antibiotic tolerance observed in ex vivo treated sputum. Ultimately, this study revealed a dominant effect of in vivo evolved bacterial populations in defining inter-subject ex vivo antibiotic tolerance and establishes a robust and translatable in vitro model for therapeutic development.


Asunto(s)
Antibacterianos , Moco , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Moco/microbiología , Moco/metabolismo , Humanos , Mucinas/metabolismo , Farmacorresistencia Bacteriana , Polímeros/metabolismo , Infección Persistente/microbiología , Pulmón/microbiología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Adaptación Fisiológica
10.
bioRxiv ; 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38187602

RESUMEN

The airway milieu of individuals with muco-obstructive airway diseases (MADs) is defined by the accumulation of dehydrated mucus due to hyperabsorption of airway surface liquid and defective mucociliary clearance. Pathological mucus becomes progressively more viscous with age and disease severity due to the concentration and overproduction of mucin and accumulation of host-derived extracellular DNA (eDNA). Respiratory mucus of MADs provides a niche for recurrent and persistent colonization by respiratory pathogens, including Pseudomonas aeruginosa , which is responsible for the majority of morbidity and mortality in MADs. Despite high concentration inhaled antibiotic therapies and the absence of antibiotic resistance, antipseudomonal treatment failure in MADs remains a significant clinical challenge. Understanding the drivers of antibiotic recalcitrance is essential for developing more effective treatments that eradicate persistent infections. The complex and dynamic environment of diseased airways makes it difficult to model antibiotic efficacy in vitro . We aimed to understand how mucin and eDNA concentrations, the two dominant polymers in respiratory mucus, alter the antibiotic tolerance of P. aeruginosa . Our results demonstrate that polymer concentration and molecular weight affect P. aeruginosa survival post antibiotic challenge. Polymer-driven antibiotic tolerance was not explicitly associated with reduced antibiotic diffusion. Lastly, we established a robust and standardized in vitro model for recapitulating the ex vivo antibiotic tolerance of P. aeruginosa observed in expectorated sputum across age, underlying MAD etiology, and disease severity, which revealed the inherent variability in intrinsic antibiotic tolerance of host-evolved P. aeruginosa populations. Importance: Antibiotic treatment failure in Pseudomonas aeruginosa chronic lung infections is associated with increased morbidity and mortality, illustrating the clinical challenge of bacterial infection control. Understanding the underlying infection environment, as well as the host and bacterial factors driving antibiotic tolerance and the ability to accurately recapitulate these factors in vitro , is crucial for improving antibiotic treatment outcomes. Here, we demonstrate that increasing concentration and molecular weight of mucin and host eDNA drive increased antibiotic tolerance to tobramycin. Through systematic testing and modeling, we identified a biologically relevant in vitro condition that recapitulates antibiotic tolerance observed in ex vivo treated sputum. Ultimately, this study revealed a dominant effect of in vivo evolved bacterial populations in defining inter-subject ex vivo antibiotic tolerance and establishes a robust and translatable in vitro model for therapeutic development.

11.
Mucosal Immunol ; 9(6): 1442-1454, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27072609

RESUMEN

Chronic lung diseases such as cystic fibrosis, chronic bronchitis, and asthma are characterized by hypersecretion and poor clearance of mucus, which are associated with poor prognosis and mortality. Little is known about the relationship between the biophysical properties of mucus and its molecular composition. The mucins MUC5B and MUC5AC are traditionally believed to generate the characteristic biophysical properties of airway mucus. However, the contribution of hundreds of globular proteins to the biophysical properties of mucus is not clear. Approximately one-third of the total mucus proteome comprises distinct, multi-protein complexes centered around airway mucins. These complexes constitute a discrete entity we call the "mucin interactome". The data suggest that while the majority of these proteins interact with mucins via electrostatic and weak interactions, some interact through very strong hydrophobic and/or covalent interactions. Using reagents that interfere with protein-protein interactions, the complexes can be disassembled, and mucus rheology can be dramatically altered. Using MUC5B-glutathione S-transferase (GST) and MUC5B-galectin-3 as a representative of these interactions, we provide evidence that individual mucin protein interactions can alter the biophysical properties of mucus and modulate the biological function of the protein. We propose that the key mechano- and bio-active functions of mucus depend on the dynamic interactions between mucins and globular proteins. These observations challenge the paradigm that mucins are the only molecules that confer biophysical properties of mucus. These observations may ultimately lead to a greater understanding of the system and guide the development of strategies for more effective interventions using better therapeutic agents.


Asunto(s)
Proteínas Portadoras/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Mucinas/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Proteoma , Proteómica/métodos
12.
Proc SPIE Int Soc Opt Eng ; 7554: 755420, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-22299066

RESUMEN

Cystic fibrosis (CF) is a genetic defect in the cystic fibrosis transmembrane conductance regulator protein and is the most common life-limiting genetic condition affecting the Caucasian population. It is an autosomal recessive, monogenic inherited disorder characterized by failure of airway host defense against bacterial infection, which results in bronchiectasis, the breakdown of airway wall extracellular matrix (ECM). In this study, we show that the in vitro models consisting of human tracheo-bronchial-epithelial (hBE) cells grown on porous supports with embedded magnetic nanoparticles (MNPs) at an air-liquid interface are suitable for long term, non-invasive assessment of ECM remodeling using magnetomotive optical coherence elastography (MMOCE). The morphology of ex vivo CF and normal lung tissues using OCT and correlative study with histology is also examined. We also demonstrate a quantitative measure of normal and CF airway elasticity using MMOCE. The improved understanding of pathologic changes in CF lung structure and function and the novel method of longitudinal in vitro ECM assessment demonstrated in this study may lead to new in vivo imaging and elastography methods to monitor disease progression and treatment in cystic fibrosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA