Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 326(2): L125-L134, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38084404

RESUMEN

Cystic fibrosis-related diabetes (CFRD) affects 40%-50% of adults with CF and is associated with a decline in respiratory health. The microbial flora of the lung is known to change with the development of CF disease, but how CFRD affects the microbiome has not been described. We analyzed the microbiome in sputa from 14 people with CF, 14 with CFRD, and two who were classed as pre-CFRD by extracting DNA and amplifying the variable V3-V4 region of the microbial 16S ribosomal RNA gene by PCR. Sequences were analyzed and sources were identified to genus level. We found that the α-diversity of the microbiome using Shannon's diversity index was increased in CFRD compared with CF. Bray Curtis dissimilarity analysis showed that there was separation of the microbiomes in CF and CFRD sputa. The most abundant phyla identified in the sputum samples were Firmicutes and Proteobacteria, Actinobacteriota and Bacteroidota, and the ratio of Firmicutes/Bacteroidota was reduced in CFRD compared with CF. Pseudomonas, Azhorizophilus, Porphyromonas, and Actinobacillus were more abundant in CFRD compared with CF, whereas Staphylococcus was less abundant. The relative abundance of these genera did not correlate with age; some correlated with a decline in FEV1/FVC but all correlated with hemoglobin A1C (HbA1c) indicating that development of CFRD mediates further changes to the respiratory microbiome in CF.NEW & NOTEWORTHY Cystic fibrosis-related diabetes (CFRD) is associated with a decline in respiratory health. We show for the first time that there was a change in the sputum microbiome of people with CFRD compared with CF that correlated with markers of raised blood glucose.


Asunto(s)
Fibrosis Quística , Diabetes Mellitus , Microbiota , Adulto , Humanos , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Esputo , Pulmón/microbiología
2.
Am J Physiol Cell Physiol ; 324(1): C153-C166, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36409177

RESUMEN

New technologies such as single-cell RNA sequencing (scRNAseq) has enabled identification of the mRNA transcripts expressed by individual cells. This review provides insight from recent scRNAseq studies on the expression of glucose transporters in the epithelial cells of the airway epithelium from trachea to alveolus. The number of studies analyzed was limited, not all reported the full range of glucose transporters and there were differences between cells freshly isolated from the airways and those grown in vitro. Furthermore, glucose transporter mRNA transcripts were expressed at lower levels than other epithelial marker genes. Nevertheless, these studies highlighted that there were differences in cellular expression of glucose transporters. GLUT1 was the most abundant of the broadly expressed transporters that included GLUT8, 10, and 13. GLUT9 transcripts were more common in basal cells and GLUT12 in ionocytes/ciliated cells. In addition to alveolar cells, SGLT1 transcripts were present in secretory cells. GLUT3 mRNA transcripts were expressed in a cell cluster that expressed monocarboxylate (MCT2) transporters. Such distributions likely underlie cell-specific metabolic requirements to support proliferation, ion transport, mucous secretion, environment sensing, and airway glucose homeostasis. These studies have also highlighted the role of glucose transporters in the movement of dehydroascorbic acid/vitamin C/myoinositol/urate, which are factors important to the innate immune properties of the airways. Discrepancies remain between detection of mRNAs, protein, and function of glucose transporters in the lungs. However, collation of the data from further scRNAseq studies may provide a better consensus and understanding, supported by qPCR, immunohistochemistry, and functional experiments.


Asunto(s)
Células Epiteliales , Proteínas Facilitadoras del Transporte de la Glucosa , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Glucosa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo
3.
Am J Physiol Cell Physiol ; 321(6): C954-C963, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34613844

RESUMEN

Airway secretions contain many signaling molecules and peptides/proteins that are not found in airway surface liquid (ASL) generated by normal human bronchial epithelial cells (NHBEs) in vitro. These play a key role in innate defense and mediate communication between the epithelium, the immune cells, and the external environment. We investigated how culture of NHBE with apically applied secretions from healthy or diseased (cystic fibrosis, CF) lungs affected epithelial function with a view to providing better in vitro models of the in vivo environment. NHBEs from 6 to 8 different donors were cultured at air-liquid interface (ALI), with apically applied sputum from normal healthy donors (normal lung sputum; NLS) or CF donors (CFS) for 2-4 h, 48 h, or with sputum reapplied over 48 h. Proteomics analysis was carried out on the sputa and on the NHBE ASL before and after culture with sputa. Transepithelial electrical resistance (TEER), short circuit current (Isc), and changes to ASL height were measured. There were 71 proteins common to both sputa but not ASL. The protease:protease inhibitor balance was increased in CFS compared with NLS and ASL. Culture of NHBE with sputa for 48 h identified additional factors not present in NLS, CFS, or ASL alone. Culture with either NLS or CFS for 48 h increased cystic fibrosis transmembrane regulator (CFTR) activity, calcium-activated chloride channel (CaCC) activity, and changed ASL height. These data indicate that culture with healthy or disease sputum changes the proteomic profile of ASL and ion transport properties of NHBE and this may increase physiological relevance when using in vitro airway models.


Asunto(s)
Bronquios/metabolismo , Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteoma , Proteómica , Esputo/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Fibrosis Quística/diagnóstico , Impedancia Eléctrica , Humanos , Transporte Iónico , Factores de Tiempo
4.
Am J Physiol Cell Physiol ; 317(5): C983-C992, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31433692

RESUMEN

The airway epithelium maintains differential glucose concentrations between the airway surface liquid (ASL, ~0.4 mM) and the blood/interstitium (5-6 mM), which is important for defense against infection. Glucose primarily moves from the blood to the ASL via paracellular movement, down its concentration gradient, across the tight junctions. However, there is evidence that glucose can move transcellularly across epithelial cells. Using a Förster resonance energy transfer sensor for glucose, we investigated intracellular glucose concentrations in airway epithelial cells and the role of hexokinases in regulating intracellular glucose concentrations in normoglycemic and hyperglycemic conditions. Our findings indicated that in airway epithelial cells (H441 or primary human bronchial epithelial cells) exposed to 5 mM glucose (normoglycemia), intracellular glucose concentration is in the micromolar range. Inhibition of facilitative glucose transporters (GLUTs) with cytochalasin B reduced intracellular glucose concentration. When cells were exposed to 15 mM glucose (hyperglycemia), intracellular glucose concentration was reduced. Airway cells expressed hexokinases I, II, and III. Inhibition with 3-bromopyruvate decreased hexokinase activity by 25% and elevated intracellular glucose concentration, but levels remained in the micromolar range. Exposure to hyperglycemia increased glycolysis, glycogen, and sorbitol. Thus, glucose enters the airway cell via GLUTs and is then rapidly processed by hexokinase-dependent and hexokinase-independent metabolic pathways to maintain low intracellular glucose concentrations. We propose that this prevents transcellular transport and aids the removal of glucose from the ASL and that the main route of entry for glucose into the ASL is via the paracellular pathway.


Asunto(s)
Glucosa/metabolismo , Hexoquinasa/metabolismo , Hiperglucemia/metabolismo , Mucosa Respiratoria/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Hexoquinasa/antagonistas & inhibidores , Humanos , Piruvatos/farmacología , Mucosa Respiratoria/efectos de los fármacos
5.
J Cell Mol Med ; 23(1): 317-327, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30450773

RESUMEN

Airway epithelial tight junction (TJ) proteins form a resistive barrier to the external environment, however, during respiratory bacterial infection TJs become disrupted compromising barrier function. This promotes glucose flux/accumulation into the lumen which acts as a nutrient source for bacterial growth. Metformin used for the treatment of diabetes increases transepithelial resistance (TEER) and partially prevents the effect of bacteria but the mechanisms of action are unclear. We investigated the effect of metformin and Staphylococcus aureus on TJ proteins, zonula occludins (ZO)-1 and occludin in human airway epithelial cells (H441). We also explored the role of AMP-activated protein kinase (AMPK) and PKCζ in metformin-induced effects. Pretreatment with metformin prevented the S. aureus-induced changes in ZO-1 and occludin. Metformin also promoted increased abundance of full length over smaller cleaved occludin proteins. The nonspecific PKC inhibitor staurosporine reduced TEER but did not prevent the effect of metformin indicating that the pathway may involve atypical PKC isoforms. Investigation of TJ reassembly after calcium depletion showed that metformin increased TEER more rapidly and promoted the abundance and localization of occludin at the TJ. These effects were inhibited by the AMPK inhibitor, compound C and the PKCζ pseudosubstrate inhibitor (PSI). Metformin increased phosphorylation of occludin and acetyl-coA-carboxylase but only the former was prevented by PSI. This study demonstrates that metformin improves TJ barrier function by promoting the abundance and assembly of full length occludin at the TJ and that this process involves phosphorylation of the protein via an AMPK-PKCζ pathway.


Asunto(s)
Metformina/farmacología , Ocludina/metabolismo , Proteína Quinasa C/metabolismo , Staphylococcus aureus/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Línea Celular , Claudina-1/metabolismo , Células Epiteliales/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Fosforilación , Mucosa Respiratoria/citología , Mucosa Respiratoria/microbiología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidad , Proteínas de Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
6.
Thorax ; 73(9): 847-856, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29748250

RESUMEN

INTRODUCTION: Loss of the cystic fibrosis transmembrane conductance regulator in cystic fibrosis (CF) leads to hyperabsorption of sodium and fluid from the airway due to upregulation of the epithelial sodium channel (ENaC). Thickened mucus and depleted airway surface liquid (ASL) then lead to impaired mucociliary clearance. ENaC regulation is thus a promising target for CF therapy. Our aim was to develop siRNA nanocomplexes that mediate effective silencing of airway epithelial ENaC in vitro and in vivo with functional correction of epithelial ion and fluid transport. METHODS: We investigated translocation of nanocomplexes through mucus and their transfection efficiency in primary CF epithelial cells grown at air-liquid interface (ALI).Short interfering RNA (SiRNA)-mediated silencing was examined by quantitative RT-PCR and western analysis of ENaC. Transepithelial potential (Vt), short circuit current (Isc), ASL depth and ciliary beat frequency (CBF) were measured for functional analysis. Inflammation was analysed by histological analysis of normal mouse lung tissue sections. RESULTS: Nanocomplexes translocated more rapidly than siRNA alone through mucus. Transfections of primary CF epithelial cells with nanocomplexes targeting αENaC siRNA, reduced αENaC and ßENaC mRNA by 30%. Transfections reduced Vt, the amiloride-sensitive Isc and mucus protein concentration while increasing ASL depth and CBF to normal levels. A single dose of siRNA in mouse lung silenced ENaC by approximately 30%, which persisted for at least 7 days. Three doses of siRNA increased silencing to approximately 50%. CONCLUSION: Nanoparticle-mediated delivery of ENaCsiRNA to ALI cultures corrected aspects of the mucociliary defect in human CF cells and offers effective delivery and silencing in vivo.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/patología , Canales Epiteliales de Sodio/genética , Silenciador del Gen , ARN Interferente Pequeño , Transfección/métodos , Animales , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Humanos , Ratones , Nanopartículas
7.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L258-L267, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27979861

RESUMEN

Air-liquid interface (ALI) culture of primary airway epithelial cells enables mucociliary differentiation providing an in vitro model of the human airway, but their proliferative potential is limited. To extend proliferation, these cells were previously transduced with viral oncogenes or mouse Bmi-1 + hTERT, but the resultant cell lines did not undergo mucociliary differentiation. We hypothesized that use of human BMI-1 alone would increase the proliferative potential of bronchial epithelial cells while retaining their mucociliary differentiation potential. Cystic fibrosis (CF) and non-CF bronchial epithelial cells were transduced by lentivirus with BMI-1 and then their morphology, replication kinetics, and karyotype were assessed. When differentiated at ALI, mucin production, ciliary function, and transepithelial electrophysiology were measured. Finally, shRNA knockdown of DNAH5 in BMI-1 cells was used to model primary ciliary dyskinesia (PCD). BMI-1-transduced basal cells showed normal cell morphology, karyotype, and doubling times despite extensive passaging. The cell lines underwent mucociliary differentiation when cultured at ALI with abundant ciliation and production of the gel-forming mucins MUC5AC and MUC5B evident. Cilia displayed a normal beat frequency and 9+2 ultrastructure. Electrophysiological characteristics of BMI-1-transduced cells were similar to those of untransduced cells. shRNA knockdown of DNAH5 in BMI-1 cells produced immotile cilia and absence of DNAH5 in the ciliary axoneme as seen in cells from patients with PCD. BMI-1 delayed senescence in bronchial epithelial cells, increasing their proliferative potential but maintaining mucociliary differentiation at ALI. We have shown these cells are amenable to genetic manipulation and can be used to produce novel disease models for research and dissemination.


Asunto(s)
Bronquios/citología , Diferenciación Celular , Cilios/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Moco/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Animales , Dineínas Axonemales/metabolismo , Proliferación Celular , Forma de la Célula , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Dineínas/metabolismo , Impedancia Eléctrica , Fenómenos Electrofisiológicos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patología , Síndrome de Kartagener/fisiopatología , Cariotipificación , Ratones , Microtúbulos/metabolismo , Modelos Biológicos , Fenotipo , Transducción Genética
8.
J Cell Mol Med ; 20(4): 758-64, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26837005

RESUMEN

Lung disease and elevation of blood glucose are associated with increased glucose concentration in the airway surface liquid (ASL). Raised ASL glucose is associated with increased susceptibility to infection by respiratory pathogens including Staphylococcus aureus and Pseudomonas aeruginosa. We have previously shown that the anti-diabetes drug, metformin, reduces glucose-induced S. aureus growth across in vitro airway epithelial cultures. The aim of this study was to investigate whether metformin has the potential to reduce glucose-induced P. aeruginosa infections across airway epithelial (Calu-3) cultures by limiting glucose permeability. We also explored the effect of P. aeruginosa and metformin on airway epithelial barrier function by investigating changes in tight junction protein abundance. Apical P. aeruginosa growth increased with basolateral glucose concentration, reduced transepithelial electrical resistance (TEER) and increased paracellular glucose flux. Metformin pre-treatment of the epithelium inhibited the glucose-induced growth of P. aeruginosa, increased TEER and decreased glucose flux. Similar effects on bacterial growth and TEER were observed with the AMP activated protein kinase agonist, 5-aminoimidazole-4-carboxamide ribonucleotide. Interestingly, metformin was able to prevent the P. aeruginosa-induced reduction in the abundance of tight junction proteins, claudin-1 and occludin. Our study highlights the potential of metformin to reduce hyperglycaemia-induced P. aeruginosa growth through airway epithelial tight junction modulation, and that claudin-1 and occludin could be important targets to regulate glucose permeability across airway epithelia and supress bacterial growth. Further investigation into the mechanisms regulating metformin and P. aeruginosa action on airway epithelial tight junctions could yield new therapeutic targets to prevent/suppress hyperglycaemia-induced respiratory infections, avoiding the use of antibiotics.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Glucosa/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Metformina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Claudina-1/genética , Claudina-1/metabolismo , Técnicas de Cocultivo , Impedancia Eléctrica , Células Epiteliales/metabolismo , Expresión Génica/efectos de los fármacos , Glucosa/toxicidad , Humanos , Ocludina/genética , Ocludina/metabolismo , Permeabilidad/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Ribonucleótidos/farmacología , Uniones Estrechas/metabolismo
10.
J Cell Sci ; 126(Pt 9): 1942-51, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23447677

RESUMEN

Cellular directional migration in an electric field (galvanotaxis) is one of the mechanisms guiding cell movement in embryogenesis and in skin epidermal repair. The epithelial sodium channel (ENaC), in addition to its function of regulating sodium transport in kidney, has recently been found to modulate cell locomotory speed. Here we tested whether ENaC has an additional function of mediating the directional migration of galvanotaxis in keratinocytes. Genetic depletion of ENaC completely blocks only galvanotaxis and does not decrease migration speed. Overexpression of ENaC is sufficient to drive galvanotaxis in otherwise unresponsive cells. Pharmacologic blockade or maintenance of the open state of ENaC also decreases or increases, respectively, galvanotaxis, suggesting that the channel open state is responsible for the response. Stable lamellipodial extensions formed at the cathodal sides of wild-type cells at the start of galvanotaxis; these were absent in the ENaC knockout keratinocytes, suggesting that ENaC mediates galvanotaxis by generating stable lamellipodia that steer cell migration. We provide evidence that ENaC is required for directional migration of keratinocytes in an electric field, supporting a role for ENaC in skin wound healing.


Asunto(s)
Movimiento Celular , Canales Epiteliales de Sodio/metabolismo , Queratinocitos/metabolismo , Sodio/metabolismo , Animales , Canales Epiteliales de Sodio/genética , Técnicas de Silenciamiento del Gen , Humanos , Transporte Iónico/genética , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Piel/lesiones , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/genética
11.
Cell Mol Life Sci ; 71(23): 4665-73, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24810961

RESUMEN

Hyperglycaemia as a result of diabetes mellitus or acute illness is associated with increased susceptibility to respiratory infection with Staphylococcus aureus. Hyperglycaemia increases the concentration of glucose in airway surface liquid (ASL) and promotes the growth of S. aureus in vitro and in vivo. Whether elevation of other sugars in the blood, such as fructose, also results in increased concentrations in ASL is unknown and whether sugars in ASL are directly utilised by S. aureus for growth has not been investigated. We obtained mutant S. aureus JE2 strains with transposon disrupted sugar transport genes. NE768(fruA) exhibited restricted growth in 10 mM fructose. In H441 airway epithelial-bacterial co-culture, elevation of basolateral sugar concentration (5-20 mM) increased the apical growth of JE2. However, sugar-induced growth of NE768(fruA) was significantly less when basolateral fructose rather than glucose was elevated. This is the first experimental evidence to show that S. aureus directly utilises sugars present in the ASL for growth. Interestingly, JE2 growth was promoted less by glucose than fructose. Net transepithelial flux of D-glucose was lower than D-fructose. However, uptake of D-glucose was higher than D-fructose across both apical and basolateral membranes consistent with the presence of GLUT1/10 in the airway epithelium. Therefore, we propose that the preferential uptake of glucose (compared to fructose) limits its accumulation in ASL. Pre-treatment with metformin increased transepithelial resistance and reduced the sugar-dependent growth of S. aureus. Thus, epithelial paracellular permeability and glucose transport mechanisms are vital to maintain low glucose concentration in ASL and limit bacterial nutrient sources as a defence against infection.


Asunto(s)
Proteínas Bacterianas/genética , Células Epiteliales/metabolismo , Fructosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Staphylococcus aureus/crecimiento & desarrollo , Transporte Biológico , Línea Celular , Técnicas de Cocultivo , Elementos Transponibles de ADN , Células Epiteliales/microbiología , Eliminación de Gen , Humanos , Hiperglucemia/complicaciones , Mutación , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Staphylococcus aureus/genética
12.
Biochem J ; 464(2): 213-20, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25220254

RESUMEN

Both lung disease and elevation of blood glucose are associated with increased glucose concentration (from 0.4 to ~4.0 mM) in the airway surface liquid (ASL). This perturbation of ASL glucose makes the airway more susceptible to infection by respiratory pathogens. ASL is minute (~1 µl/cm(2)) and the measurement of glucose concentration in the small volume ASL is extremely difficult. Therefore, we sought to develop a fluorescent biosensor with sufficient sensitivity to determine glucose concentrations in ASL in situ. We coupled a range of environmentally sensitive fluorophores to mutated forms of a glucose/galactose-binding protein (GBP) including H152C and H152C/A213R and determined their equilibrium binding properties. Of these, GBP H152C/A213R-BADAN (Kd 0.86 ± 0.01 mM, Fmax/F0 3.6) was optimal for glucose sensing and in ASL increased fluorescence when basolateral glucose concentration was raised from 1 to 20 mM. Moreover, interpolation of the data showed that the glucose concentration in ASL was increased, with results similar to that using glucose oxidase analysis. The fluorescence of GBP H152C/A213R-BADAN in native ASL from human airway epithelial cultures in situ was significantly increased over time when basolateral glucose was increased from 5 to 20 mM. Overall our data indicate that this GBP is a useful tool to monitor glucose homoeostasis in the lung.


Asunto(s)
Técnicas Biosensibles/métodos , Glucemia/aislamiento & purificación , Proteínas de Unión al Calcio/química , Proteínas de Transporte de Monosacáridos/química , Proteínas de Unión Periplasmáticas/química , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Glucemia/química , Proteínas de Unión al Calcio/genética , Técnicas de Cultivo de Célula , Células Epiteliales/metabolismo , Colorantes Fluorescentes/química , Homeostasis , Humanos , Pulmón/citología , Pulmón/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Mutación , Proteínas de Unión Periplasmáticas/genética
13.
J Immunol ; 189(1): 373-80, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22623330

RESUMEN

The glucose concentration of the airway surface liquid (ASL) is much lower than that in blood and is tightly regulated by the airway epithelium. ASL glucose is elevated in patients with viral colds, cystic fibrosis, chronic obstructive pulmonary disease, and asthma. Elevated ASL glucose is also associated with increased incidence of respiratory infection. However, the mechanism by which ASL glucose increases under inflammatory conditions is unknown. The aim of this study was to investigate the effect of proinflammatory mediators (PIMs) on the mechanisms governing airway glucose homeostasis in polarized monolayers of human airway (H441) and primary human bronchial epithelial (HBE) cells. Monolayers were treated with TNF-α, IFN-γ, and LPS during 72 h. PIM treatment led to increase in ASL glucose concentration and significantly reduced H441 and HBE transepithelial resistance. This decline in transepithelial resistance was associated with an increase in paracellular permeability of glucose. Similar enhanced rates of paracellular glucose flux were also observed across excised trachea from LPS-treated mice. Interestingly, PIMs enhanced glucose uptake across the apical, but not the basolateral, membrane of H441 and HBE monolayers. This increase was predominantly via phloretin-sensitive glucose transporter (GLUT)-mediated uptake, which coincided with an increase in GLUT-2 and GLUT-10 abundance. In conclusion, exposure of airway epithelial monolayers to PIMs results in increased paracellular glucose flux, as well as apical GLUT-mediated glucose uptake. However, uptake was insufficient to limit glucose accumulation in ASL. To our knowledge, these data provide for the first time a mechanism to support clinical findings that ASL glucose concentration is increased in patients with airway inflammation.


Asunto(s)
Glucosa/metabolismo , Homeostasis/inmunología , Mediadores de Inflamación/farmacología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Animales , Transporte Biológico Activo/inmunología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Glucosa/biosíntesis , Glucosa/deficiencia , Proteínas Facilitadoras del Transporte de la Glucosa/biosíntesis , Transportador de Glucosa de Tipo 2/biosíntesis , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Mucosa Respiratoria/metabolismo , Propiedades de Superficie , Regulación hacia Arriba/inmunología
14.
Thorax ; 68(9): 835-45, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23709760

RESUMEN

BACKGROUND: Diabetes is a risk factor for respiratory infection, and hyperglycaemia is associated with increased glucose in airway surface liquid and risk of Staphylococcus aureus infection. OBJECTIVES: To investigate whether elevation of basolateral/blood glucose concentration promotes airway Staphylococcus aureus growth and whether pretreatment with the antidiabetic drug metformin affects this relationship. METHODS: Human airway epithelial cells grown at air-liquid interface (±18 h pre-treatment, 30 µM-1 mM metformin) were inoculated with 5×10(5) colony-forming units (CFU)/cm(2) S aureus 8325-4 or JE2 or Pseudomonas aeruginosa PA01 on the apical surface and incubated for 7 h. Wild-type C57BL/6 or db/db (leptin receptor-deficient) mice, 6-10 weeks old, were treated with intraperitoneal phosphate-buffered saline or 40 mg/kg metformin for 2 days before intranasal inoculation with 1×10(7) CFU S aureus. Mice were culled 24 h after infection and bronchoalveolar lavage fluid collected. RESULTS: Apical S aureus growth increased with basolateral glucose concentration in an in vitro airway epithelia-bacteria co-culture model. S aureus reduced transepithelial electrical resistance (RT) and increased paracellular glucose flux. Metformin inhibited the glucose-induced growth of S aureus, increased RT and decreased glucose flux. Diabetic (db/db) mice infected with S aureus exhibited a higher bacterial load in their airways than control mice after 2 days and metformin treatment reversed this effect. Metformin did not decrease blood glucose but reduced paracellular flux across ex vivo murine tracheas. CONCLUSIONS: Hyperglycaemia promotes respiratory S aureus infection, and metformin modifies glucose flux across the airway epithelium to limit hyperglycaemia-induced bacterial growth. Metformin might, therefore, be of additional benefit in the prevention and treatment of respiratory infection.


Asunto(s)
Carga Bacteriana/efectos de los fármacos , Glucemia/metabolismo , Células Epiteliales/metabolismo , Metformina/farmacología , Pseudomonas aeruginosa/crecimiento & desarrollo , Staphylococcus aureus/crecimiento & desarrollo , Animales , Glucemia/efectos de los fármacos , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Quimiocina CXCL9/metabolismo , Células Epiteliales/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hiperglucemia/sangre , Hiperglucemia/tratamiento farmacológico , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Sistema Respiratorio/metabolismo , Sistema Respiratorio/microbiología , Staphylococcus aureus/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
15.
PLoS One ; 18(7): e0287183, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37406028

RESUMEN

Airway diseases can disrupt tight junction proteins, compromising the epithelial barrier and making it more permeable to pathogens. In people with pulmonary disease who are prone to infection with Pseudomonas aeruginosa, pro-inflammatory leukotrienes are increased and anti-inflammatory lipoxins are decreased. Upregulation of lipoxins is effective in counteracting inflammation and infection. However, whether combining a lipoxin receptor agonist with a specific leukotriene A4 hydrolase (LTA4H) inhibitor could enhance these protective effects has not to our knowledge been investigated. Therefore, we explored the effect of lipoxin receptor agonist BML-111 and JNJ26993135 a specific LTA4H inhibitor that prevents the production of pro-inflammatory LTB4 on tight junction proteins disrupted by P. aeruginosa filtrate (PAF) in human airway epithelial cell lines H441 and 16HBE-14o. Pre-treatment with BML-111 prevented an increase in epithelial permeability induced by PAF and conserved ZO-1 and claudin-1 at the cell junctions. JNJ26993135 similarly prevented the increased permeability induced by PAF, restored ZO-1 and E-cadherin and reduced IL-8 but not IL-6. Cells pre-treated with BML-111 plus JNJ26993135 restored TEER and permeability, ZO-1 and claudin-1 to the cell junctions. Taken together, these data indicate that the combination of a lipoxin receptor agonist with a LTA4H inhibitor could provide a more potent therapy.


Asunto(s)
Lipoxinas , Uniones Estrechas , Humanos , Uniones Estrechas/metabolismo , Pseudomonas aeruginosa/metabolismo , Claudina-1/metabolismo , Células Epiteliales/metabolismo , Proteínas de Uniones Estrechas/metabolismo
16.
Cells ; 12(7)2023 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-37048070

RESUMEN

Primary human bronchial epithelial cultures (HBECs) are used to study airway physiology, disease, and drug development. HBECs often replicate human airway physiology/pathophysiology. Indeed, in the search for cystic fibrosis (CF) transmembrane conductance regulator (CFTR) therapies, HBECs were seen as the "gold standard" in preclinical studies. However, HBECs are not without their limitations: they are non-immortalized and the requirement for human donors, especially those with rare genetic mutations, can make HBECs expensive and/or difficult to source. For these reasons, researchers may opt to expand HBECs by passaging. This practice is common, but to date, there has not been a robust analysis of the impact of expanding HBECs on their phenotype. Here, we used functional studies of airway surface liquid (ASL) homeostasis, epithelial barrier properties, and RNA-seq and Western blotting to investigate HBEC changes over two passage cycles. We found that passaging impaired CFTR-mediated ASL secretion and led to a reduction in the plasma membrane expression of the epithelial sodium channel (ENaC) and CFTR. Passaging also resulted in an increase in transepithelial resistance and a decrease in epithelial water permeability. We then looked for changes at the mRNA level and found that passaging significantly affected 323 genes, including genes involved in inflammation, cell growth, and extracellular matrix remodeling. Collectively, these data highlight the potential for HBEC expansion to impact research findings.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Transporte Biológico , Transporte Iónico , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Mol Ther Methods Clin Dev ; 31: 101140, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38027060

RESUMEN

Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CFTR gene. The 10th most common mutation, c.3178-2477C>T (3849+10kb C>T), involves a cryptic, intronic splice site. This mutation was corrected in CF primary cells homozygous for this mutation by delivering pairs of guide RNAs (gRNAs) with Cas9 protein in ribonucleoprotein (RNP) complexes that introduce double-strand breaks to flanking sites to excise the 3849+10kb C>T mutation, followed by DNA repair by the non-homologous end-joining pathway, which functions in all cells of the airway epithelium. RNP complexes were delivered to CF basal epithelial cell by a non-viral, receptor-targeted nanocomplex comprising a formulation of targeting peptides and lipids. Canonical CFTR mRNA splicing was, thus, restored leading to the restoration of CFTR protein expression with concomitant restoration of electrophysiological function in airway epithelial air-liquid interface cultures. Off-target editing was not detected by Sanger sequencing of in silico-selected genomic sites with the highest sequence similarities to the gRNAs, although more sensitive unbiased whole genome sequencing methods would be required for possible translational developments. This approach could potentially be used to correct aberrant splicing signals in several other CF mutations and other genetic disorders where deep-intronic mutations are pathogenic.

18.
Eur Respir J ; 40(5): 1269-76, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22878875

RESUMEN

For over 50 years, glucose has been recognised to cross the lung epithelial barrier and be transported by lung epithelial cells. However, until recently, research into these processes focused on their effects on lung liquid volume. Here, we consider a newly identified role for pulmonary glucose transport in maintaining low airway surface liquid (ASL) glucose concentrations and propose that this contributes to lung defence against infection. Glucose diffuses into ASL via paracellular pathways at a rate determined by paracellular permeability and the transepithelial glucose gradient. Glucose is removed from ASL in proximal airways via facilitative glucose transporters, down a concentration gradient generated by intracellular glucose metabolism. In the distal lung, glucose transport via sodium-coupled glucose transporters predominates. These processes vary between species but universally maintain ASL glucose at 3-20-fold lower concentrations than plasma. ASL glucose concentrations are increased in respiratory disease and by hyperglycaemia. Elevated ASL glucose in intensive care patients was associated with increased Staphylococcus aureus infection. Diabetic patients with and without chronic lung disease are at increased risk of respiratory infection. Understanding of mechanisms underlying lung glucose homeostasis could identify new therapeutic targets for control of ASL glucose and prevention and treatment of lung infection.


Asunto(s)
Glucosa/metabolismo , Pulmón/metabolismo , Animales , Transporte Biológico , Epitelio/metabolismo , Humanos
19.
Pflugers Arch ; 462(3): 431-41, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21667229

RESUMEN

Using human H441 airway epithelial cells cultured at air-liquid interface (ALI), we have uniquely correlated the functional response to apical fluid volume expansion with the abundance and cleavage of endogenous α- and γENaC proteins in the apical membrane. Monolayers cultured at ALI rapidly elevated I (sc) when inserted into fluid-filled Ussing chambers. The increase in I (sc) was not significantly augmented by the apical addition of trypsin, and elevation was abolished by the protease inhibitor aprotinin and an inhibitor of the proprotein convertase, furin. These treatments also increased the IC50 amiloride indicating that the effect was via inhibition of highly Na⁺-selective ENaC channels. Apical fluid, 5-500 µl for 1 h in culture, increased the spontaneous starting I (sc) in a dose-dependent manner, whilst maximal fluid-induced I (sc) in the Ussing chamber was unchanged. Apical fluid expansion increased the abundance of 63-65-kDa αENaC proteins in the apical membrane. However, this could not be attributed to increased cleavage as protease inhibitors had no effect on the ratio of cleaved to non-cleaved (90 kDa) αENaC proteins. Instead, fluid expansion increased αENaC abundance in the membrane. In contrast, function correlated well with γENaC cleavage at known sites by furin and extracellular proteases. Interestingly, cleavage of γENaC was associated with increased retrieval from the membrane via the proteosomal pathway. Thus, the response to apical fluid volume expansion in H441 airway epithelial cells involves cleavage of γENaC, and changes in α- and γENaC protein abundance at the apical membrane.


Asunto(s)
Tamaño de la Célula , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Mucosa Respiratoria/citología , Sodio/metabolismo , Amilorida/farmacología , Animales , Línea Celular , Polaridad Celular , Electrofisiología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Humanos , Inhibidores de Proteasas/farmacología , Bloqueadores de los Canales de Sodio/farmacología
20.
Biosci Rep ; 41(6)2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34003249

RESUMEN

BACKGROUND: In the kidney glucose is freely filtered by the glomerulus and, mainly, reabsorbed by sodium glucose cotransporter 2 (SGLT2) expressed in the early proximal tubule. Human proximal tubule epithelial cells (PTECs) undergo pathological and fibrotic changes seen in diabetic kidney disease (DKD) in response to elevated glucose. We developed a specific in vitro model of DKD using primary human PTECs with exposure to high D-glucose and TGF-ß1 and propose a role for SGLT2 inhibition in regulating fibrosis. METHODS: Western blotting was performed to detect cellular and secreted proteins as well as phosphorylated intracellular signalling proteins. qPCR was used to detect CCN2 RNA. Gamma glutamyl transferase (GT) activity staining was performed to confirm PTEC phenotype. SGLT2 and ERK inhibition on high D-glucose, 25 mM, and TGF-ß1, 0.75 ng/ml, treated cells was explored using dapagliflozin and U0126, respectively. RESULTS: Only the combination of high D-glucose and TGF-ß1 treatment significantly up-regulated CCN2 RNA and protein expression. This increase was significantly ameliorated by dapagliflozin. High D-glucose treatment raised phospho ERK which was also inhibited by dapagliflozin. TGF-ß1 increased cellular phospho SSXS Smad3 serine 423 and 425, with and without high D-glucose. Glucose alone had no effect. Smad3 serine 204 phosphorylation was significantly raised by a combination of high D-glucose+TGF-ß1; this rise was significantly reduced by both SGLT2 and MEK inhibition. CONCLUSIONS: We show that high D-glucose and TGF-ß1 are both required for CCN2 expression. This treatment also caused Smad3 linker region phosphorylation. Both outcomes were inhibited by dapagliflozin. We have identified a novel SGLT2 -ERK mediated promotion of TGF-ß1/Smad3 signalling inducing a pro-fibrotic growth factor secretion. Our data evince support for substantial renoprotective benefits of SGLT2 inhibition in the diabetic kidney.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Glucosa/toxicidad , Glucósidos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Proteína Smad2/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Fosforilación , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA