Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 49(11): 6100-6113, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34107015

RESUMEN

Pulmonary diseases offer many targets for oligonucleotide therapeutics. However, effective delivery of oligonucleotides to the lung is challenging. For example, splicing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect a significant cohort of Cystic Fibrosis (CF) patients. These individuals could potentially benefit from treatment with splice switching oligonucleotides (SSOs) that can modulate splicing of CFTR and restore its activity. However, previous studies in cell culture used oligonucleotide transfection methods that cannot be safely translated in vivo. In this report, we demonstrate effective correction of a splicing mutation in the lung of a mouse model using SSOs. Moreover, we also demonstrate effective correction of a CFTR splicing mutation in a pre-clinical CF patient-derived cell model. We utilized a highly effective delivery strategy for oligonucleotides by combining peptide-morpholino (PPMO) SSOs with small molecules termed OECs. PPMOs distribute broadly into the lung and other tissues while OECs potentiate the effects of oligonucleotides by releasing them from endosomal entrapment. The combined PPMO plus OEC approach proved to be effective both in CF patient cells and in vivo in the mouse lung and thus may offer a path to the development of novel therapeutics for splicing mutations in CF and other lung diseases.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Pulmón/metabolismo , Morfolinos/administración & dosificación , Empalme del ARN , Animales , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Ratones , Mutación , Péptidos , Mucosa Respiratoria/metabolismo , Transfección
2.
Molecules ; 24(3)2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30736307

RESUMEN

Addition of small molecule Retro-1 has been described to enhance antisense and splice switching oligonucleotides. With the aim of assessing the effect of covalently linking Retro-1 to the biologically active oligonucleotide, three different derivatives of Retro-1 were prepared that incorporated a phosphoramidite group, a thiol or a 1,3-diene, respectively. Retro-1⁻oligonucleotide conjugates were assembled both on-resin (coupling of the phosphoramidite) and from reactions in solution (Michael-type thiol-maleimide reaction and Diels-Alder cycloaddition). Splice switching assays with the resulting conjugates showed that they were active but that they provided little advantage over the unconjugated oligonucleotide in the well-known HeLa Luc705 reporter system.


Asunto(s)
Oligonucleótidos/síntesis química , Oligonucleótidos/farmacología , Línea Celular Tumoral , Técnicas de Química Sintética , Cromatografía Líquida de Alta Presión , Humanos , Espectrometría de Masas , Estructura Molecular , Oligonucleótidos/química , Empalme del ARN/efectos de los fármacos , Relación Estructura-Actividad
3.
J Biomed Opt ; 29(4): 046004, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38690122

RESUMEN

Significance: Assessing the nanostructure of polymer solutions and biofluids is broadly useful for understanding drug delivery and disease progression and for monitoring therapy. Aim: Our objective is to quantify bronchial mucus solids concentration (wt. %) during hypertonic saline (HTS) treatment in vitro via nanostructurally constrained diffusion of gold nanorods (GNRs) monitored by polarization-sensitive optical coherence tomography (PS-OCT). Approach: Using PS-OCT, we quantified GNR translational (DT) and rotational (DR) diffusion coefficients within polyethylene oxide solutions (0 to 3 wt. %) and human bronchial epithelial cell (hBEC) mucus (0 to 6.4 wt. %). Interpolation of DT and DR data is used to develop an assay to quantify mucus concentration. The assay is demonstrated on the mucus layer of an air-liquid interface hBEC culture during HTS treatment. Results: In polymer solutions and mucus, DT and DR monotonically decrease with increasing concentration. DR is more sensitive than DT to changes above 1.5 wt. % of mucus and exhibits less intrasample variability. Mucus on HTS-treated hBEC cultures exhibits dynamic mixing from cilia. A region of hard-packed mucus is revealed by DR measurements. Conclusions: The extended dynamic range afforded by simultaneous measurement of DT and DR of GNRs using PS-OCT enables resolving concentration of the bronchial mucus layer over a range from healthy to disease in depth and time during HTS treatment in vitro.


Asunto(s)
Oro , Moco , Nanotubos , Tomografía de Coherencia Óptica , Tomografía de Coherencia Óptica/métodos , Humanos , Nanotubos/química , Oro/química , Moco/química , Moco/metabolismo , Difusión , Bronquios/diagnóstico por imagen , Células Epiteliales/química , Células Epiteliales/metabolismo , Solución Salina Hipertónica/farmacología , Solución Salina Hipertónica/química , Células Cultivadas
4.
Am J Physiol Cell Physiol ; 304(10): C976-84, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23467297

RESUMEN

Nucleotides within the airway surface liquid promote fluid secretion via activation of airway epithelial purinergic receptors. ATP is stored within and released from mucin granules as co-cargo with mucins, but the mechanism by which ATP, and potentially other nucleotides, enter the lumen of mucin granules is not known. We assessed the contribution of the recently identified SLC17A9 vesicle nucleotide transporter (VNUT) to the nucleotide availability within isolated mucin granules and further examined the involvement of VNUT in mucin granule secretion-associated nucleotide release. RT-PCR and Western blot analyses indicated that VNUT is abundantly expressed in airway epithelial goblet-like Calu-3 cells, migrating as a duplex with apparent mobility of 55 and 60 kDa. Subcellular fractionation studies indicated that VNUT55 was associated with high-density mucin granules, whereas VNUT60 was associated with low-density organelles. Immunofluorescence studies showed that recombinant VNUT localized to mucin granules and other organelles. Mucin granules isolated from VNUT short hairpin RNA-expressing cells exhibited a marked reduction of ATP, ADP, AMP, and UTP levels within granules. Ca(2+)-regulated vesicular ATP release was markedly reduced in these cells, but mucin secretion was not affected. These results suggest that VNUT is the relevant nucleotide transporter responsible for the uptake of cytosolic nucleotides into mucin granules. By controlling the entry of nucleotides into mucin granules, VNUT contributes to the release of purinergic signaling molecules necessary for the proper hydration of co-released mucins.


Asunto(s)
Células Caliciformes/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Nucleótidos/metabolismo , Sistema Respiratorio/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Adenosina Difosfato/biosíntesis , Adenosina Monofosfato/biosíntesis , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Transporte Biológico , Línea Celular , Gránulos Citoplasmáticos/metabolismo , Humanos , Mucinas/genética , Proteínas de Transporte de Nucleótidos/biosíntesis , ARN Interferente Pequeño , Vesículas Secretoras/metabolismo , Uridina Trifosfato/biosíntesis
5.
FASEB J ; 26(2): 533-45, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21990373

RESUMEN

Cigarette smoke (CS) exposure induces mucus obstruction and the development of chronic bronchitis (CB). While many of these responses are determined genetically, little is known about the effects CS can exert on pulmonary epithelia at the protein level. We, therefore, tested the hypothesis that CS exerts direct effects on the CFTR protein, which could impair airway hydration, leading to the mucus stasis characteristic of both cystic fibrosis and CB. In vivo and in vitro studies demonstrated that CS rapidly decreased CFTR activity, leading to airway surface liquid (ASL) volume depletion (i.e., dehydration). Further studies revealed that CS induced internalization of CFTR. Surprisingly, CS-internalized CFTR did not colocalize with lysosomal proteins. Instead, the bulk of CFTR shifted to a detergent-resistant fraction within the cell and colocalized with the intermediate filament vimentin, suggesting that CS induced CFTR movement into an aggresome-like, perinuclear compartment. To test whether airway dehydration could be reversed, we used hypertonic saline (HS) as an osmolyte to rehydrate ASL. HS restored ASL height in CS-exposed, dehydrated airway cultures. Similarly, inhaled HS restored mucus transport and increased clearance in patients with CB. Thus, we propose that CS exposure rapidly impairs CFTR function by internalizing CFTR, leading to ASL dehydration, which promotes mucus stasis and a failure of mucus clearance, leaving smokers at risk for developing CB. Furthermore, our data suggest that strategies to rehydrate airway surfaces may provide a novel form of therapy for patients with CB.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Pulmón/metabolismo , Fumar/efectos adversos , Fumar/metabolismo , Adulto , Anciano , Animales , Secuencia de Bases , Transporte Biológico Activo , Agua Corporal/metabolismo , Bronquitis Crónica/etiología , Bronquitis Crónica/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Cricetinae , Fibrosis Quística/etiología , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Cartilla de ADN/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Depuración Mucociliar , Mucosa Respiratoria/metabolismo , Solución Salina Hipertónica/farmacología , Humo/efectos adversos , Solubilidad
6.
Am J Physiol Cell Physiol ; 303(5): C490-8, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22673622

RESUMEN

The G(i)-coupled P2Y(14) receptor (P2Y(14)-R) is potently activated by UDP-sugars and UDP. Although P2Y(14)-R mRNA is prominently expressed in circulating neutrophils, the signaling pathways and functional responses associated with this receptor are undefined. In this study, we illustrate that incubation of isolated human neutrophils with UDP-glucose resulted in cytoskeleton rearrangement, change of cell shape, and enhanced cell migration. We also demonstrate that UDP-glucose promotes rapid, robust, and concentration-dependent activation of RhoA in these cells. Ecto-nucleotidases expressed on neutrophils rapidly hydrolyzed extracellular ATP, but incubation with UDP-glucose for up to 1 h resulted in negligible metabolism of the nucleotide-sugar. HL60 human promyelocytic leukemia cells do not express the P2Y(14)-R, but neutrophil differentiation of HL60 cells with DMSO resulted in markedly enhanced P2Y(14)-R expression. Accordingly, UDP-glucose, UDP-galactose, and UDP-N-acetylglucosamine promoted Rho activation in differentiated but not in undifferentiated HL60 cells. Stable expression of recombinant human P2Y(14)-R conferred UDP-sugar-promoted responses to undifferentiated HL60 cells. UDP-glucose-promoted RhoA activation also was accompanied by enhanced cell migration in differentiated HL60 cells, and these responses were blocked by Rho kinase inhibitors. These results support the notion that UDP-glucose is a stable and potent proinflammatory mediator that promotes P2Y(14)-R-mediated neutrophil motility via Rho/Rho kinase activation.


Asunto(s)
Quimiotaxis/fisiología , Neutrófilos/metabolismo , Receptores Purinérgicos P2/metabolismo , Transducción de Señal/fisiología , Uridina Difosfato Glucosa/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Forma de la Célula , Citoesqueleto , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/genética , Proteína de Unión al GTP rhoA/genética
7.
J Physiol ; 590(3): 545-62, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22144578

RESUMEN

Mucin secretion is an innate defence mechanism, which is noxiously upregulated in obstructive lung diseases (e.g. chronic obstructive pulmonary disease (COPD), cystic fibrosis and asthma). Mucin granule exocytosis is regulated by specific protein complexes, but the SNARE exocytotic core has not been defined in airway goblet cells. In this study, we identify VAMP8 as one of the SNAREs regulating mucin granule exocytosis. VAMP8 mRNA was present in human airway and lung epithelial cells, and deep-sequencing and expression analyses of airway epithelial cells revealed that VAMP8 transcripts were expressed at 10 times higher levels than other VAMP mRNAs. In human airway epithelial cell cultures and freshly excised tissues, VAMP8 immunolocalised mainly to goblet cell mucin granules. The function of VAMP8 in airway mucin secretion was tested by RNA interference techniques. Both VAMP8 short interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs) reduced mucin secretion induced by PAR agonists, neutrophil elastase and ATP in two airway epithelial cell culture models. Notably, basal (non-agonist elicited) mucin secretion was also reduced in these experiments. VAMP8 knockdown was also effective in decreasing mucin secretion in airway epithelial cell cultures with induced mucous metaplasia/mucin hypersecretion. Unlike VAMP8 silencing, knockdown of VAMP2 or VAMP3 did not affect mucin secretion. Importantly, in VAMP8 knock-out (KO) mice with IL-13-induced mucous metaplasia, mucin content in the bronchoalveolar lavage (BAL) and ATP-stimulated mucin secretion in the trachea were reduced compared to WT-matched littermates. Our data indicate that VAMP8 is an essential SNARE in airway mucin granule exocytosis. Reduction of VAMP8 activity/expression may provide a novel therapeutic target to ameliorate airway mucus obstruction in lung diseases.


Asunto(s)
Células Caliciformes/metabolismo , Mucinas/metabolismo , Proteínas R-SNARE/metabolismo , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Noqueados , Proteínas R-SNARE/deficiencia , Proteínas R-SNARE/genética , ARN Interferente Pequeño/genética
8.
J Biol Chem ; 286(30): 26277-86, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21606493

RESUMEN

ATP released from airway epithelial cells promotes purinergic receptor-regulated mucociliary clearance activities necessary for innate lung defense. Cell swelling-induced membrane stretch/strain is a common stimulus that promotes airway epithelial ATP release, but the mechanisms transducing cell swelling into ATP release are incompletely understood. Using knockdown and knockout approaches, we tested the hypothesis that pannexin 1 mediates ATP release from hypotonically swollen airway epithelia and investigated mechanisms regulating this activity. Well differentiated primary cultures of human bronchial epithelial cells subjected to hypotonic challenge exhibited enhanced ATP release, which was paralleled by the uptake of the pannexin probe propidium iodide. Both responses were reduced by pannexin 1 inhibitors and by knocking down pannexin 1. Importantly, hypotonicity-evoked ATP release from freshly excised tracheas and dye uptake in primary tracheal epithelial cells were impaired in pannexin 1 knockout mice. Hypotonicity-promoted ATP release and dye uptake in primary well differentiated human bronchial epithelial cells was accompanied by RhoA activation and myosin light chain phosphorylation and was reduced by the RhoA dominant negative mutant RhoA(T19N) and Rho and myosin light chain kinase inhibitors. ATP release and Rho activation were reduced by highly selective inhibitors of transient receptor potential vanilloid 4 (TRPV4). Lastly, knocking down TRPV4 impaired hypotonicity-evoked airway epithelial ATP release. Our data suggest that TRPV4 and Rho transduce cell membrane stretch/strain into pannexin 1-mediated ATP release in airway epithelia.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Pulmón/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Mucosa Respiratoria/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Adenosina Trifosfato/inmunología , Animales , Células Cultivadas , Conexinas/genética , Conexinas/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata/fisiología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Noqueados , Mutación Missense , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/inmunología , Cadenas Ligeras de Miosina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Fosforilación/fisiología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/inmunología , Canales Catiónicos TRPV/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/inmunología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/inmunología
9.
Subcell Biochem ; 55: 1-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21560042

RESUMEN

The purinergic events regulating the airways' innate defenses are initiated by the release of purines from the epithelium, which occurs constitutively and is enhanced by chemical or mechanical stimulation. While the external triggers have been reviewed exhaustively, this chapter focuses on current knowledge of the receptors and signaling cascades mediating nucleotide release. The list of secreted purines now includes ATP, ADP, AMP and nucleotide sugars, and involves at least three distinct mechanisms reflecting the complexity of airway epithelia. First, the constitutive mechanism involves ATP translocation to the ER/Golgi complex as energy source for protein folding, and fusion of Golgi-derived vesicles with the plasma membrane. Second, goblet cells package ATP with mucins into granules, which are discharged in response to P2Y(2)R activation and Ca(2+)-dependent signaling pathways. Finally, non-mucous cells support a regulated mechanism of ATP release involving protease activated receptor (PAR)-elicited G(12/13) activation, leading to the RhoGEF-mediated exchange of GDP for GTP on RhoA, and cytoskeleton rearrangement. Together, these pathways provide fine tuning of epithelial responses regulated by purinergic signaling events.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Humanos , Receptores Purinérgicos/metabolismo , Transducción de Señal
10.
Curr Opin Pharmacol ; 66: 102271, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988291

RESUMEN

In the clinically successful era of CFTR modulators and Theratyping, 10-20% of individuals with cystic fibrosis (CF) may develop disease due to CFTR mutations that remain undruggable. These individuals produce low levels of CFTR mRNA and/or not enough protein to be rescued with modulator drugs. Alternative therapeutic approaches to correct the CFTR defect at the mRNA level using nucleic acid technologies are currently feasible; e.g., oligonucleotides platforms, which are being rapidly developed to correct genetic disorders. Drug-like properties, great specificity, and predictable off-target effects by design make oligonucleotides a valuable approach with fewer clinical and ethical challenges than genomic editing strategies. Together with personalized and precision medicine approaches, oligonucleotides are ideal therapeutics to target CF-causing mutations that affect only a few individuals resilient to modulator therapies.


Asunto(s)
Fibrosis Quística , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Mutación , Oligonucleótidos/uso terapéutico , Medicina de Precisión , ARN Mensajero
11.
Am J Respir Cell Mol Biol ; 45(2): 253-60, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20935191

RESUMEN

Adenosine triphosphate (ATP) and its metabolite adenosine regulate airway mucociliary clearance via activation of purinoceptors. In this study, we investigated the contribution of goblet cells to airway epithelial ATP release. Primary human bronchial epithelial (HBE) cultures, typically dominated by ciliated cells, were induced to develop goblet cell metaplasia by infection with respiratory syncytial virus (RSV) or treatment with IL-13. Under resting conditions, goblet-cell metaplastic cultures displayed enhanced mucin secretion accompanied by increased rates of ATP release and mucosal surface adenosine accumulation as compared with nonmetaplastic control HBE cultures. Intracellular calcium chelation [1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetraacetoxymethyl ester] or disruption of the secretory pathways (nocodazole, brefeldin A, and N-ethylmaleimide) decreased mucin secretion and ATP release in goblet-cell metaplastic HBE cultures. Conversely, stimuli that triggered calcium-regulated mucin secretion (e.g., ionomycin or UTP) increased luminal ATP release and adenyl purine accumulation in control and goblet-cell metaplastic HBE cultures. Goblet cell-associated ATP release was not blocked by the connexin/pannexin hemichannel inhibitor carbenoxolone, suggesting direct nucleotide release from goblet cell vesicles rather than the hemichannel insertion. Collectively, our data demonstrate that nucleotide release is increased by goblet cell metaplasia, reflecting, at least in part, a mechanism tightly associated with goblet cell mucin secretion. Increased goblet cell nucleotide release and resultant adenosine accumulation provide compensatory mechanisms to hydrate mucins by paracrine stimulation of ciliated cell ion and water secretion and maintain mucociliary clearance, and to modulate inflammatory responses.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bronquios/metabolismo , Epitelio/metabolismo , Células Caliciformes/metabolismo , Células Caliciformes/patología , Metaplasia/metabolismo , Mucinas/metabolismo , Western Blotting , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/virología , Calcio/metabolismo , Células Cultivadas , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Ensayo de Inmunoadsorción Enzimática , Epitelio/efectos de los fármacos , Epitelio/virología , Etilmaleimida/farmacología , Exocitosis , Células Caliciformes/virología , Humanos , Técnicas para Inmunoenzimas , Interleucina-13/farmacología , Metaplasia/patología , Metaplasia/virología , ARN Mensajero/genética , Receptores Purinérgicos P2Y2/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/patogenicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Physiol ; 588(Pt 12): 2255-67, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20421285

RESUMEN

Purinergic regulation of airway innate defence activities is in part achieved by the release of nucleotides from epithelial cells. However, the mechanisms of airway epithelial nucleotide release are poorly understood. We have previously demonstrated that ATP is released from ionomycin-stimulated airway epithelial goblet cells coordinately with mucin exocytosis, suggesting that ATP is released as a co-cargo molecule from mucin-containing granules. We now demonstrate that protease-activated-receptor (PAR) agonists also stimulate the simultaneous release of mucins and ATP from airway epithelial cells. PAR-mediated mucin and ATP release were dependent on intracellular Ca(2+) and actin cytoskeleton reorganization since BAPTA AM, cytochalasin D, and inhibitors of Rho and myosin light chain kinases blocked both responses. To test the hypothesis that ATP is co-released with mucin from mucin granules, we measured the nucleotide composition of isolated mucin granules purified based on their MUC5AC and VAMP-8 content by density gradients. Mucin granules contained ATP, but the levels of ADP and AMP within granules exceeded by nearly 10-fold that of ATP. Consistent with this finding, apical secretions from PAR-stimulated cells contained relatively high levels of ADP/AMP, which could not be accounted for solely based on ATP release and hydrolysis. Thus, mucin granules contribute to ATP release and also are a source of extracellular ADP and AMP. Direct release of ADP/AMP from mucin granules is likely to provide a major source of airway surface adenosine to signal in a paracrine faction ciliated cell A(2b) receptors to activate ion/water secretion and appropriately hydrate goblet cell-released mucins.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Exocitosis , Células Caliciformes/metabolismo , Mucina 5AC/metabolismo , Receptores Proteinasa-Activados/metabolismo , Mucosa Respiratoria/metabolismo , Vesículas Secretoras/metabolismo , Citoesqueleto de Actina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Quelantes/farmacología , Exocitosis/efectos de los fármacos , Células Caliciformes/efectos de los fármacos , Humanos , Hidrólisis , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas R-SNARE/metabolismo , Receptores Proteinasa-Activados/agonistas , Mucosa Respiratoria/efectos de los fármacos , Vesículas Secretoras/efectos de los fármacos , Trombina/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
13.
J Clin Invest ; 117(2): 364-74, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17235394

RESUMEN

The role of the cystic fibrosis transmembrane conductance regulator (CFTR) as a cAMP-dependent chloride channel on the apical membrane of epithelia is well established. However, the processes by which CFTR is regulated on the cell surface are not clear. Here we report the identification of a protein-protein interaction between CFTR and the cytoskeletal filamin proteins. Using proteomic approaches, we identified filamins as proteins that associate with the extreme CFTR N terminus. Furthermore, we identified a disease-causing missense mutation in CFTR, serine 13 to phenylalanine (S13F), which disrupted this interaction. In cells, filamins tethered plasma membrane CFTR to the underlying actin network. This interaction stabilized CFTR at the cell surface and regulated the plasma membrane dynamics and confinement of the channel. In the absence of filamin binding, CFTR was internalized from the cell surface, where it prematurely accumulated in lysosomes and was ultimately degraded. Our data demonstrate what we believe to be a previously unrecognized role for the CFTR N terminus in the regulation of the plasma membrane stability and metabolic stability of CFTR. In addition, we elucidate the molecular defect associated with the S13F mutation.


Asunto(s)
Proteínas Contráctiles/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas de Microfilamentos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Línea Celular , Membrana Celular/metabolismo , Cricetinae , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Estabilidad de Medicamentos , Filaminas , Células HeLa , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Conformación Proteica , Proteómica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
14.
Am J Respir Crit Care Med ; 177(7): 730-42, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18079494

RESUMEN

RATIONALE: Chronic obstructive pulmonary disease is a leading cause of death worldwide, but its pathogenesis is not well understood. Previous studies have shown that airway surface dehydration in beta-epithelial Na(+) channel (betaENaC)-overexpressing mice caused a chronic lung disease with high neonatal pulmonary mortality and chronic bronchitis in adult survivors. OBJECTIVES: The aim of this study was to identify the initiating lesions and investigate the natural progression of lung disease caused by airway surface dehydration. METHODS: Lung morphology, gene expression, bronchoalveolar lavage, and lung mechanics were studied at different ages in betaENaC-overexpressing mice. MEASUREMENTS AND MAIN RESULTS: Mucus obstruction in betaENaC-overexpressing mice originated in the trachea in the first days of life and was associated with hypoxia, airway epithelial necrosis, and death. In surviving betaENaC-overexpressing mice, mucus obstruction extended into the lungs and was accompanied by goblet cell metaplasia, increased mucin expression, and airway inflammation with transient perinatal increases in tumor necrosis factor-alpha and macrophages, IL-13 and eosinophils, and persistent increases in keratinocyte-derived cytokine (KC), neutrophils, and chitinases in the lung. betaENaC-overexpressing mice also developed emphysema with increased lung volumes, distal airspace enlargement, and increased lung compliance. CONCLUSIONS: Our studies demonstrate that airway surface dehydration is sufficient to initiate persistent neutrophilic airway inflammation with chronic airways mucus obstruction and to cause transient eosinophilic airway inflammation and emphysema. These results suggest that deficient airway surface hydration may play a critical role in the pathogenesis of chronic obstructive pulmonary diseases of different etiologies and serve as a target for novel therapies.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Mucosa Respiratoria/metabolismo , Animales , Animales Recién Nacidos , Bronquitis Crónica/fisiopatología , Deshidratación , Progresión de la Enfermedad , Expresión Génica , Inflamación , Estudios Longitudinales , Ratones , Ratones Endogámicos , Depuración Mucociliar , Enfisema Pulmonar/fisiopatología
15.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30721150

RESUMEN

Airway mucin secretion is necessary for ciliary clearance of inhaled particles and pathogens but can be detrimental in pathologies such as asthma and cystic fibrosis. Exocytosis in mammals requires a Munc18 scaffolding protein, and airway secretory cells express all 3 Munc18 isoforms. Using conditional airway epithelial cell-deletant mice, we found that Munc18a has the major role in baseline mucin secretion, Munc18b has the major role in stimulated mucin secretion, and Munc18c does not function in mucin secretion. In an allergic asthma model, Munc18b deletion reduced airway mucus occlusion and airflow resistance. In a cystic fibrosis model, Munc18b deletion reduced airway mucus occlusion and emphysema. Munc18b deficiency in the airway epithelium did not result in any abnormalities of lung structure, particle clearance, inflammation, or bacterial infection. Our results show that regulated secretion in a polarized epithelial cell may involve more than one exocytic machine at the apical plasma membrane and that the protective roles of mucin secretion can be preserved while therapeutically targeting its pathologic roles.


Asunto(s)
Asma/metabolismo , Mucinas/metabolismo , Proteínas Munc18/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Exocitosis , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Munc18/genética , Mucosa Respiratoria/patología , Transcriptoma
16.
Mol Biol Cell ; 16(5): 2154-67, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15716351

RESUMEN

Previous studies in native tissues have produced conflicting data on the localization and metabolic fate of WT and deltaF508 cystic fibrosis transmembrane regulator (CFTR) in the lung. Combining immunocytochemical and biochemical studies utilizing new high-affinity CFTR mAbs with ion transport assays, we examined both 1) the cell type and region specific expression of CFTR in normal airways and 2) the metabolic fate of deltaF508 CFTR and associated ERM proteins in the cystic fibrosis lung. Studies of lungs from a large number of normal subjects revealed that WT CFTR protein localized to the apical membrane of ciliated cells within the superficial epithelium and gland ducts. In contrast, other cell types in the superficial, gland acinar, and alveolar epithelia expressed little WT CFTR protein. No deltaF508 CFTR mature protein or function could be detected in airway specimens freshly excised from a large number of deltaF508 homozygous subjects, despite an intact ERM complex. In sum, our data demonstrate that WT CFTR is predominantly expressed in ciliated cells, and deltaF508 CFTR pathogenesis in native tissues, like heterologous cells, reflects loss of normal protein processing.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Mutación , Sistema Respiratorio/metabolismo , Adolescente , Adulto , Bronquios/metabolismo , Niño , Preescolar , Cloruros/metabolismo , AMP Cíclico/metabolismo , Proteínas del Citoesqueleto , Epitelio/metabolismo , Expresión Génica , Homocigoto , Humanos , Inmunohistoquímica , Hibridación in Situ , Lactante , Pulmón/metabolismo , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Fosfoproteínas/metabolismo , Alveolos Pulmonares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Equilibrio Hidroelectrolítico
17.
ACS Chem Biol ; 12(8): 1999-2007, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28703575

RESUMEN

The pharmacological effectiveness of oligonucleotides has been hampered by their tendency to remain entrapped in endosomes, thus limiting their access to cytosolic or nuclear targets. We have previously reported a group of small molecules that enhance the effects of oligonucleotides by causing their release from endosomes. Here, we describe a second novel family of oligonucleotide enhancing compounds (OECs) that is chemically distinct from the compounds reported previously. We demonstrate that these molecules substantially augment the actions of splice switching oligonucleotides (SSOs) and antisense oligonucleotides (ASOs) in cell culture. We also find enhancement of SSO effects in a murine model. These new compounds act by increasing endosome permeability and causing partial release of entrapped oligonucleotides. While they also affect the permeability of lysosomes, they are clearly different from typical lysosomotropic agents. Current members of this compound family display a relatively narrow window between effective dose and toxic dose. Thus, further improvements are necessary before these agents can become suitable for therapeutic use.


Asunto(s)
Oligonucleótidos Antisentido/farmacología , Oligonucleótidos/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Sistemas de Liberación de Medicamentos , Células HeLa , Humanos , Lisosomas/efectos de los fármacos , Ratones , Microscopía Confocal , Oligonucleótidos/química , Oligonucleótidos Antisentido/química , Empalme del ARN
18.
Nat Commun ; 8(1): 1409, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29123085

RESUMEN

Modulation of airway surface liquid (ASL) pH has been proposed as a therapy for cystic fibrosis (CF). However, evidence that ASL pH is reduced in CF is limited and conflicting. The technical challenges associated with measuring ASL pH in vivo have precluded accurate measurements in humans. In order to address this deficiency, ASL pH was measured in vivo in children using a novel luminescent technology integrated with fibre-optic probes. Here we show that ASL pH in children with CF is similar to that of children without CF. Findings were supported by highly controlled direct pH measurements in primary human airway epithelial cell culture models, which also suggest that the potential ASL pH gradient produced by defective apical ion transport is balanced out by paracellular shunting of acid/base. Thus, reduced baseline ASL pH is unlikely to be an important pathobiological factor in early CF lung disease.


Asunto(s)
Fibrosis Quística/metabolismo , Mucosa Respiratoria/metabolismo , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/metabolismo , Bronquios/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Células Cultivadas , Niño , Preescolar , Fibrosis Quística/complicaciones , Fibrosis Quística/etiología , Femenino , Tecnología de Fibra Óptica , Colorantes Fluorescentes , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lactante , Masculino , Estudios Prospectivos
19.
Artículo en Inglés | MEDLINE | ID: mdl-27746581

RESUMEN

Mucus hydration (wt%) has become an increasingly useful metric in real-time assessment of respiratory health in diseases like cystic fibrosis and COPD, with higher wt% indicative of diseased states. However, available in vivo rheological techniques are lacking. Gold nanorods (GNRs) are attractive biological probes whose diffusion through tissue is sensitive to the correlation length of comprising biopolymers. Through employment of dynamic light scattering theory on OCT signals from GNRs, we find that weakly-constrained GNR diffusion predictably decreases with increasing wt% (more disease-like) mucus. Previously, we determined this method is robust against mucus transport on human bronchial epithelial (hBE) air-liquid interface cultures (R2=0.976). Here we introduce diffusion-sensitive OCT (DS-OCT), where we collect M-mode image ensembles, from which we derive depth- and temporally-resolved GNR diffusion rates. DS-OCT allows for real-time monitoring of changing GNR diffusion as a result of topically applied mucus-thinning agents, enabling monitoring of the dynamics of mucus hydration never before seen. Cultured human airway epithelial cells (Calu-3) with a layer of endogenous mucus were doped with topically deposited GNRs (80×22nm), and subsequently treated with hypertonic saline (HS) or isotonic saline (IS). DS-OCT provided imaging of the mucus thinning response up to a depth of 600µm with 4.65µm resolution, over a total of 8 minutes in increments of ≥3 seconds. For both IS and HS conditions, DS-OCT captured changes in the pattern of mucus hydration over time. DS-OCT opens a new window into understanding mechanisms of mucus thinning during treatment, enabling real-time efficacy feedback needed to optimize and tailor treatments for individual patients.

20.
Cold Spring Harb Perspect Med ; 2(9): a009589, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22951447

RESUMEN

Mucus pathology in cystic fibrosis (CF) has been known for as long as the disease has been recognized and is sometimes called mucoviscidosis. The disease is marked by mucus hyperproduction and plugging in many organs, which are usually most fatal in the airways of CF patients, once the problem of meconium ileus at birth is resolved. After the CF gene, CFTR, was cloned and its protein product identified as a cAMP-regulated Cl(-) channel, causal mechanisms underlying the strong mucus phenotype of the disease became obscure. Here we focus on mucin genes and polymeric mucin glycoproteins, examining their regulation and potential relationships to a dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR). Detailed examination of CFTR expression in organs and different cell types indicates that changes in CFTR expression do not always correlate with the severity of CF disease or mucus accumulation. Thus, the mucus hyperproduction that typifies CF does not appear to be a direct cause of a defective CFTR but, rather, to be a downstream consequence. In organs like the lung, up-regulation of mucin gene expression by inflammation results from chronic infection; however, in other instances and organs, the inflammation may have a non-infectious origin. The mucus plugging phenotype of the ß-subunit of the epithelial Na(+) channel (ßENaC)-overexpressing mouse is proving to be an archetypal example of this kind of inflammation, with a dehydrated airway surface/concentrated mucus gel apparently providing the inflammatory stimulus. Data indicate that the luminal HCO(3)(-) deficiency recently described for CF epithelia may also provide such a stimulus, perhaps by causing a mal-maturation of mucins as they are released onto luminal surfaces. In any event, the path between CFTR dysfunction and mucus hyperproduction has proven tortuous, and its unraveling continues to offer its own twists and turns, along with fascinating glimpses into biology.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Mucinas/genética , Moco/metabolismo , Obstrucción de las Vías Aéreas , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica/genética , Genitales/metabolismo , Glicoproteínas/análisis , Glicosilación , Humanos , Pulmón/metabolismo , Masculino , Ratones , Mucina 5AC/genética , Mucinas/biosíntesis , Mucinas/metabolismo , Moco/química , Moco/fisiología , Neumonía/metabolismo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA