Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Clin Invest ; 129(2): 744-758, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640172

RESUMEN

Goblet cell metaplasia, a disabling hallmark of chronic lung disease, lacks curative treatments at present. To identify novel therapeutic targets for goblet cell metaplasia, we studied the transcriptional response profile of IL-13-exposed primary human airway epithelia in vitro and asthmatic airway epithelia in vivo. A perturbation-response profile connectivity approach identified geldanamycin, an inhibitor of heat shock protein 90 (HSP90) as a candidate therapeutic target. Our experiments confirmed that geldanamycin and other HSP90 inhibitors prevented IL-13-induced goblet cell metaplasia in vitro and in vivo. Geldanamycin also reverted established goblet cell metaplasia. Geldanamycin did not induce goblet cell death, nor did it solely block mucin synthesis or IL-13 receptor-proximal signaling. Geldanamycin affected the transcriptome of airway cells when exposed to IL-13, but not when exposed to vehicle. We hypothesized that the mechanism of action probably involves TGF-ß, ERBB, or EHF, which would predict that geldanamycin would also revert IL-17-induced goblet cell metaplasia, a prediction confirmed by our experiments. Our findings suggest that persistent airway goblet cell metaplasia requires HSP90 activity and that HSP90 inhibitors will revert goblet cell metaplasia, despite active upstream inflammatory signaling. Moreover, HSP90 inhibitors may be a therapeutic option for airway diseases with goblet cell metaplasia of unknown mechanism.


Asunto(s)
Benzoquinonas/efectos adversos , Células Caliciformes/metabolismo , Proteínas HSP90 de Choque Térmico , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Lactamas Macrocíclicas/efectos adversos , Transducción de Señal/efectos de los fármacos , Animales , Benzoquinonas/farmacología , Células Caliciformes/patología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas/farmacología , Masculino , Metaplasia , Ratones
2.
J Appl Physiol (1985) ; 123(3): 526-533, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28620056

RESUMEN

Mutations in the gene encoding the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) anion channel cause CF. The leading cause of death in the CF population is lung disease. Increasing evidence suggests that in utero airway development is CFTR-dependent and that developmental abnormalities may contribute to CF lung disease. However, relatively little is known about postnatal CF airway growth, largely because such studies are limited in humans. Therefore, we examined airway growth and lung volume in a porcine model of CF. We hypothesized that CF pigs would have abnormal postnatal airway growth. To test this hypothesis, we performed CT-based airway and lung volume measurements in 3-wk-old non-CF and CF pigs. We found that 3-wk-old CF pigs had tracheas of reduced caliber and irregular shape. Their bronchial lumens were reduced in size proximally but not distally, were irregularly shaped, and had reduced distensibility. Our data suggest that lack of CFTR results in aberrant postnatal airway growth and development, which could contribute to CF lung disease pathogenesis.NEW & NOTEWORTHY This CT scan-based study of airway morphometry in the cystic fibrosis (CF) postnatal period is unique, as analogous studies in humans are greatly limited for ethical and technical reasons. Findings such as reduced airway lumen area and irregular caliber suggest that airway growth and development are CF transmembrane conductance regulator-dependent and that airway growth defects may contribute to CF lung disease pathogenesis.


Asunto(s)
Bronquios/diagnóstico por imagen , Bronquios/crecimiento & desarrollo , Fibrosis Quística/diagnóstico por imagen , Tráquea/efectos de los fármacos , Tráquea/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Pulmón/diagnóstico por imagen , Pulmón/crecimiento & desarrollo , Porcinos
3.
JCI Insight ; 1(8)2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27390778

RESUMEN

In cystic fibrosis (CF), loss of CF transmembrane conductance regulator (CFTR) anion channel activity causes airway surface liquid (ASL) pH to become acidic, which impairs airway host defenses. One potential therapeutic approach is to correct the acidic pH in CF airways by aerosolizing HCO3- and/or nonbicarbonate pH buffers. Here, we show that raising ASL pH with inhaled HCO3- increased pH. However, the effect was transient, and pH returned to baseline values within 30 minutes. Tromethamine (Tham) is a buffer with a long serum half-life used as an i.v. formulation to treat metabolic acidosis. We found that Tham aerosols increased ASL pH in vivo for at least 2 hours and enhanced bacterial killing. Inhaled hypertonic saline (7% NaCl) is delivered to people with CF in an attempt to promote mucus clearance. Because an increased ionic strength inhibits ASL antimicrobial factors, we added Tham to hypertonic saline and applied it to CF sputum. We found that Tham alone and in combination with hypertonic saline increased pH and enhanced bacterial killing. These findings suggest that aerosolizing the HCO3--independent buffer Tham, either alone or in combination with hypertonic saline, might be of therapeutic benefit in CF airway disease.

4.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L670-9, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26801568

RESUMEN

While pathological and clinical data suggest that small airways are involved in early cystic fibrosis (CF) lung disease development, little is known about how the lack of cystic fibrosis transmembrane conductance regulator (CFTR) function contributes to disease pathogenesis in these small airways. Large and small airway epithelia are exposed to different airflow velocities, temperatures, humidity, and CO2 concentrations. The cellular composition of these two regions is different, and small airways lack submucosal glands. To better understand the ion transport properties and impacts of lack of CFTR function on host defense function in small airways, we adapted a novel protocol to isolate small airway epithelial cells from CF and non-CF pigs and established an organotypic culture model. Compared with non-CF large airways, non-CF small airway epithelia cultures had higher Cl(-) and bicarbonate (HCO3 (-)) short-circuit currents and higher airway surface liquid (ASL) pH under 5% CO2 conditions. CF small airway epithelia were characterized by minimal Cl(-) and HCO3 (-) transport and decreased ASL pH, and had impaired bacterial killing compared with non-CF small airways. In addition, CF small airway epithelia had a higher ASL viscosity than non-CF small airways. Thus, the activity of CFTR is higher in the small airways, where it plays a role in alkalinization of ASL, enhancement of antimicrobial activity, and lowering of mucus viscosity. These data provide insight to explain why the small airways are a susceptible site for the bacterial colonization.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Bicarbonatos/metabolismo , Fibrosis Quística/metabolismo , Células Epiteliales Alveolares/inmunología , Animales , Transporte Biológico , Células Cultivadas , Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Concentración de Iones de Hidrógeno , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Sus scrofa
5.
Clin Sci (Lond) ; 128(2): 131-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25142104

RESUMEN

Diabetes is a common and significant co-morbidity in cystic fibrosis (CF). The pathogenesis of cystic fibrosis related diabetes (CFRD) is incompletely understood. Because exocrine pancreatic disease is similar between humans and pigs with CF, the CF pig model has the potential to contribute significantly to the understanding of CFRD pathogenesis. We determined the structure of the endocrine pancreas in fetal, newborn and older CF and non-CF pigs and assessed endocrine pancreas function by intravenous glucose tolerance test (IV-GTT). In fetal pigs, pancreatic insulin and glucagon density was similar between CF and non-CF. In newborn and older pigs, the insulin and glucagon density was unchanged between CF and non-CF per total pancreatic area, but increased per remnant lobular tissue in CF reflecting exocrine pancreatic loss. Although fasting glucose levels were not different between CF and non-CF newborns, CF newborns demonstrated impaired glucose tolerance and increased glucose area under the curve during IV-GTT. Second phase insulin secretion responsiveness was impaired in CF newborn pigs and significantly lower than that observed in non-CF newborns. Older CF pigs had elevated random blood glucose levels compared with non-CF. In summary, glycaemic abnormalities and insulin secretion defects were present in newborn CF pigs and spontaneous hyperglycaemia developed over time. Functional changes in CF pig pancreas were not associated with a decline in islet cell mass. Our results suggest that functional islet abnormalities, independent of structural islet loss, contribute to the early pathogenesis of CFRD.


Asunto(s)
Glucemia , Fibrosis Quística/metabolismo , Diabetes Mellitus/metabolismo , Intolerancia a la Glucosa , Insulina/metabolismo , Animales , Fibrosis Quística/complicaciones , Fibrosis Quística/patología , Diabetes Mellitus/patología , Ensayo de Inmunoadsorción Enzimática , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiología , Páncreas/metabolismo , Páncreas/patología , Porcinos
6.
Am J Respir Crit Care Med ; 188(12): 1434-41, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24168209

RESUMEN

RATIONALE: Air trapping and airflow obstruction are being increasingly identified in infants with cystic fibrosis. These findings are commonly attributed to airway infection, inflammation, and mucus buildup. OBJECTIVES: To learn if air trapping and airflow obstruction are present before the onset of airway infection and inflammation in cystic fibrosis. METHODS: On the day they are born, piglets with cystic fibrosis lack airway infection and inflammation. Therefore, we used newborn wild-type piglets and piglets with cystic fibrosis to assess air trapping, airway size, and lung volume with inspiratory and expiratory X-ray computed tomography scans. Micro-computed tomography scanning was used to assess more distal airway sizes. Airway resistance was determined with a mechanical ventilator. Mean linear intercept and alveolar surface area were determined using stereologic methods. MEASUREMENTS AND MAIN RESULTS: On the day they were born, piglets with cystic fibrosis exhibited air trapping more frequently than wild-type piglets (75% vs. 12.5%, respectively). Moreover, newborn piglets with cystic fibrosis had increased airway resistance that was accompanied by luminal size reduction in the trachea, mainstem bronchi, and proximal airways. In contrast, mean linear intercept length, alveolar surface area, and lung volume were similar between both genotypes. CONCLUSIONS: The presence of air trapping, airflow obstruction, and airway size reduction in newborn piglets with cystic fibrosis before the onset of airway infection, inflammation, and mucus accumulation indicates that cystic fibrosis impacts airway development. Our findings suggest that early airflow obstruction and air trapping in infants with cystic fibrosis might, in part, be caused by congenital airway abnormalities.


Asunto(s)
Obstrucción de las Vías Aéreas/etiología , Fibrosis Quística/fisiopatología , Obstrucción de las Vías Aéreas/congénito , Obstrucción de las Vías Aéreas/diagnóstico por imagen , Obstrucción de las Vías Aéreas/patología , Resistencia de las Vías Respiratorias , Animales , Bronquios/patología , Bronquios/fisiopatología , Broncografía/métodos , Fibrosis Quística/diagnóstico por imagen , Fibrosis Quística/patología , Mediciones del Volumen Pulmonar , Tomografía Computarizada Multidetector , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Porcinos , Tráquea/diagnóstico por imagen , Tráquea/patología , Tráquea/fisiopatología
7.
J Clin Invest ; 123(6): 2685-93, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23676501

RESUMEN

Cystic fibrosis (CF) pigs develop disease with features remarkably similar to those in people with CF, including exocrine pancreatic destruction, focal biliary cirrhosis, micro-gallbladder, vas deferens loss, airway disease, and meconium ileus. Whereas meconium ileus occurs in 15% of babies with CF, the penetrance is 100% in newborn CF pigs. We hypothesized that transgenic expression of porcine CF transmembrane conductance regulator (pCFTR) cDNA under control of the intestinal fatty acid-binding protein (iFABP) promoter would alleviate the meconium ileus. We produced 5 CFTR-/-;TgFABP>pCFTR lines. In 3 lines, intestinal expression of CFTR at least partially restored CFTR-mediated anion transport and improved the intestinal phenotype. In contrast, these pigs still had pancreatic destruction, liver disease, and reduced weight gain, and within weeks of birth, they developed sinus and lung disease, the severity of which varied over time. These data indicate that expressing CFTR in intestine without pancreatic or hepatic correction is sufficient to rescue meconium ileus. Comparing CFTR expression in different lines revealed that approximately 20% of wild-type CFTR mRNA largely prevented meconium ileus. This model may be of value for understanding CF pathophysiology and testing new preventions and therapies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Fibrosis Quística/metabolismo , Ileus/metabolismo , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Expresión Génica , Humanos , Íleon/metabolismo , Íleon/patología , Ileus/patología , Recién Nacido , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Pulmón/patología , Masculino , Meconio/metabolismo , Páncreas/metabolismo , Páncreas/patología , Fenotipo , Regiones Promotoras Genéticas , Radiografía , Ratas , Sus scrofa , Tráquea/metabolismo , Tráquea/patología
8.
PLoS One ; 7(8): e43777, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952763

RESUMEN

Paraoxonases (PON) are a family of proteins (PON1, 2 and 3) with multiple enzymatic activities. PON1 interferes with homoserine lactone-mediated quorum sensing in bacteria and with reactive oxygen species (ROS) in humans and mice. PON1 gene mutations have been linked to multiple traits, including aging, and diseases of the cardiovascular, nervous and gastrointestinal system. The overlapping enzymatic activities in the PON family members and high linkage disequilibrium rates within their polymorphisms confound animal and human studies of PON1 function. In contrast, arthropods such as Drosophila melanogaster have no PON homologs, resulting in an ideal model to study interactions between PON genotype and host phenotypes. We hypothesized that expression of PON1 in D. melanogaster would alter ROS. We found that PON1 alters expression of multiple oxidative stress genes and decreases superoxide anion levels in normal and germ-free D. melanogaster. We also found differences in the composition of the gut microbiota, with a remarkable increase in levels of Lactobacillus plantarum and associated changes in expression of antimicrobial and cuticle-related genes. PON1 expression directly decreased superoxide anion levels and altered bacterial colonization of the gut and its gene expression profile, highlighting the complex nature of the interaction between host genotype and gut microbiota. We speculate that the interaction between some genotypes and human diseases may be mediated by the presence of certain gut bacteria that can induce specific immune responses in the gut and other host tissues.


Asunto(s)
Arildialquilfosfatasa/genética , Drosophila melanogaster/fisiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Superóxidos/metabolismo , Animales , Carga Bacteriana/genética , Femenino , Expresión Génica , Humanos , Lactobacillus/fisiología , Masculino , Metagenoma/genética , Estrés Oxidativo/genética , Simbiosis
9.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L152-60, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22637155

RESUMEN

A balance between alveolar liquid absorption and secretion is critical for maintaining optimal alveolar subphase liquid height and facilitating gas exchange in the alveolar space. However, the role of cystic fibrosis transmembrane regulator protein (CFTR) in this homeostatic process has remained elusive. Using a newly developed porcine model of cystic fibrosis, in which CFTR is absent, we investigated ion transport properties and alveolar liquid transport in isolated type II alveolar epithelial cells (T2AECs) cultured at the air-liquid interface. CFTR was distributed exclusively to the apical surface of cultured T2AECs. Alveolar epithelia from CFTR(-/-) pigs failed to increase liquid absorption in response to agents that increase cAMP, whereas cAMP-stimulated liquid absorption in CFTR(+/-) epithelia was similar to that in CFTR(+/+) epithelia. Expression of recombinant CFTR restored stimulated liquid absorption in CFTR(-/-) T2AECs but had no effect on CFTR(+/+) epithelia. In ex vivo studies of nonperfused lungs, stimulated liquid absorption was defective in CFTR(-/-) alveolar epithelia but similar between CFTR(+/+) and CFTR(+/-) epithelia. When epithelia were studied at the air-liquid interface, elevating cAMP levels increased subphase liquid height in CFTR(+/+) but not in CFTR(-/-) T2AECs. Our findings demonstrate that CFTR is required for maximal liquid absorption under cAMP stimulation, but it is not the rate-limiting factor. Furthermore, our data define a role for CFTR in liquid secretion by T2AECs. These insights may help to develop new treatment strategies for pulmonary edema and respiratory distress syndrome, diseases in which lung liquid transport is disrupted.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Agua Pulmonar Extravascular/metabolismo , Absorción , Células Epiteliales Alveolares/fisiología , Animales , Transporte Biológico , Polaridad Celular , Células Cultivadas , Cloruros/metabolismo , AMP Cíclico/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Impedancia Eléctrica , Femenino , Técnicas de Inactivación de Genes , Técnicas In Vitro , Masculino , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Tensión Superficial , Sus scrofa , Uniones Estrechas/metabolismo
10.
Sci Transl Med ; 3(74): 74ra24, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21411740

RESUMEN

Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. The most common CF-associated mutation is ΔF508, which deletes a phenylalanine in position 508. In vitro studies indicate that the resultant protein, CFTR-ΔF508, is misprocessed, although the in vivo consequences of this mutation remain uncertain. To better understand the effects of the ΔF508 mutation in vivo, we produced CFTR(ΔF508/ΔF508) pigs. Our biochemical, immunocytochemical, and electrophysiological data on CFTR-ΔF508 in newborn pigs paralleled in vitro predictions. They also indicated that CFTR(ΔF508/ΔF508) airway epithelia retain a small residual CFTR conductance, with maximal stimulation producing ~6% of wild-type function. Cyclic adenosine 3',5'-monophosphate (cAMP) agonists were less potent at stimulating current in CFTR(Δ)(F508/)(Δ)(F508) epithelia, suggesting that quantitative tests of maximal anion current may overestimate transport under physiological conditions. Despite residual CFTR function, four older CFTR(ΔF508/ΔF508) pigs developed lung disease similar to human CF. These results suggest that this limited CFTR activity is insufficient to prevent lung or gastrointestinal disease in CF pigs. These data also suggest that studies of recombinant CFTR-ΔF508 misprocessing predict in vivo behavior, which validates its use in biochemical and drug discovery experiments. These findings help elucidate the molecular pathogenesis of the common CF mutation and will guide strategies for developing new therapeutics.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/fisiopatología , Enfermedades Pulmonares/veterinaria , Mutación , Porcinos , Animales , Animales Recién Nacidos , Células Cultivadas , Progresión de la Enfermedad , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/fisiopatología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo
11.
PLoS One ; 6(1): e16166, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21311590

RESUMEN

Diabetes mellitus predisposes the host to bacterial infections. Moreover, hyperglycemia has been shown to be an independent risk factor for respiratory infections. The luminal surface of airway epithelia is covered by a thin layer of airway surface liquid (ASL) and is normally sterile despite constant exposure to bacteria. The balance between bacterial growth and killing in the airway determines the outcome of exposure to inhaled or aspirated bacteria: infection or sterility. We hypothesized that restriction of carbon sources--including glucose--in the ASL is required for sterility of the lungs. We found that airway epithelia deplete glucose from the ASL via a novel mechanism involving polarized expression of GLUT-1 and GLUT-10, intracellular glucose phosphorylation, and low relative paracellular glucose permeability in well-differentiated cultures of human airway epithelia and in segments of airway epithelia excised from human tracheas. Moreover, we found that increased glucose concentration in the ASL augments growth of P. aeruginosa in vitro and in the lungs of hyperglycemic ob/ob and db/db mice in vivo. In contrast, hyperglycemia had no effect on intrapulmonary bacterial growth of a P. aeruginosa mutant that is unable to utilize glucose as a carbon source. Our data suggest that depletion of glucose in the airway epithelial surface is a novel mechanism for innate immunity. This mechanism is important for sterility of the airways and has implications in hyperglycemia and conditions that result in disruption of the epithelial barrier in the lung.


Asunto(s)
Glucosa/deficiencia , Inmunidad Innata , Mucosa Respiratoria/microbiología , Animales , Líquidos Corporales , Células Cultivadas , Células Epiteliales/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Ratones , Pseudomonas aeruginosa , Mucosa Respiratoria/metabolismo , Sistema Respiratorio/microbiología , Esterilización
12.
Sci Transl Med ; 2(29): 29ra31, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20427821

RESUMEN

Lung disease causes most of the morbidity and mortality in cystic fibrosis (CF). Understanding the pathogenesis of this disease has been hindered, however, by the lack of an animal model with characteristic features of CF. To overcome this problem, we recently generated pigs with mutated CFTR genes. We now report that, within months of birth, CF pigs spontaneously developed hallmark features of CF lung disease, including airway inflammation, remodeling, mucus accumulation, and infection. Their lungs contained multiple bacterial species, suggesting that the lungs of CF pigs have a host defense defect against a wide spectrum of bacteria. In humans, the temporal and causal relations between inflammation and infection have remained uncertain. To investigate these processes, we studied newborn pigs. Their lungs showed no inflammation but were less often sterile than controls. Moreover, after introduction of bacteria into their lungs, pigs with CF failed to eradicate bacteria as effectively as wild-type pigs. These results suggest that impaired bacterial elimination is the pathogenic event that initiates a cascade of inflammation and pathology in CF lungs. Our finding that pigs with CF have a host defense defect against bacteria within hours of birth provides an opportunity to further investigate CF pathogenesis and to test therapeutic and preventive strategies that could be deployed before secondary consequences develop.


Asunto(s)
Fibrosis Quística/microbiología , Fibrosis Quística/patología , Pulmón/microbiología , Pulmón/patología , Porcinos/crecimiento & desarrollo , Porcinos/microbiología , Animales , Animales Recién Nacidos , Fibrosis Quística/complicaciones , Fibrosis Quística/diagnóstico por imagen , Modelos Animales de Enfermedad , Ileus/cirugía , Inflamación/complicaciones , Inflamación/patología , Pulmón/anomalías , Pulmón/diagnóstico por imagen , Meconio , Moco/metabolismo , Enfermedades Pancreáticas/patología , Radiografía Torácica , Análisis de Supervivencia , Factores de Tiempo
13.
Science ; 321(5897): 1837-41, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18818360

RESUMEN

Almost two decades after CFTR was identified as the gene responsible for cystic fibrosis (CF), we still lack answers to many questions about the pathogenesis of the disease, and it remains incurable. Mice with a disrupted CFTR gene have greatly facilitated CF studies, but the mutant mice do not develop the characteristic manifestations of human CF, including abnormalities of the pancreas, lung, intestine, liver, and other organs. Because pigs share many anatomical and physiological features with humans, we generated pigs with a targeted disruption of both CFTR alleles. Newborn pigs lacking CFTR exhibited defective chloride transport and developed meconium ileus, exocrine pancreatic destruction, and focal biliary cirrhosis, replicating abnormalities seen in newborn humans with CF. The pig model may provide opportunities to address persistent questions about CF pathogenesis and accelerate discovery of strategies for prevention and treatment.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística , Modelos Animales de Enfermedad , Porcinos , Animales , Animales Recién Nacidos , Cloruros/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/patología , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Femenino , Vesícula Biliar/patología , Ileus/patología , Ileus/fisiopatología , Intestinos/patología , Transporte Iónico , Hígado/patología , Cirrosis Hepática Biliar/patología , Pulmón/patología , Masculino , Páncreas Exocrino/patología , Recombinación Genética
14.
J Clin Invest ; 118(9): 3123-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18704198

RESUMEN

Pseudomonas aeruginosa uses quorum sensing, an interbacterial communication system, to regulate gene expression. The signaling molecule N-3-oxododecanoyl homoserine lactone (3OC12-HSL) is thought to play a central role in quorum sensing. Since 3OC12-HSL can be degraded by paraoxonase (PON) family members, we hypothesized that PONs regulate P. aeruginosa virulence in vivo. We chose Drosophila melanogaster as our model organism because it has been shown to be a tractable model for investigating host-pathogen interactions and lacks PONs. By using quorum-sensing-deficient P. aeruginosa, synthetic acyl-HSLs, and transgenic expression of human PON1, we investigated the role of 3OC12-HSL and PON1 on P. aeruginosa virulence. We found that P. aeruginosa virulence in flies was dependent upon 3OC12-HSL. PON1 transgenic flies expressed enzymatically active PON1 and thereby exhibited arylesterase activity and resistance to organophosphate toxicity. Moreover, PON1 flies were protected from P. aeruginosa lethality, and protection was dependent on the lactonase activity of PON1. Our findings show that PON1 can interfere with quorum sensing in vivo and provide insight into what we believe is a novel role for PON1 in the innate immune response to quorum-sensing-dependent pathogens. These results raise intriguing possibilities about human-pathogen interactions, including potential roles for PON1 as a modifier gene and for PON1 protein as a regulator of normal bacterial florae, a link between infection/inflammation and cardiovascular disease, and a potential therapeutic modality.


Asunto(s)
Arildialquilfosfatasa/biosíntesis , Arildialquilfosfatasa/genética , Regulación Enzimológica de la Expresión Génica , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Transgenes , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Inmunidad Innata , Inflamación , Modelos Biológicos , Modelos Genéticos , Mutación , Infecciones por Pseudomonas/mortalidad , Infecciones por Pseudomonas/prevención & control , Factores de Tiempo , Virulencia
15.
Am J Respir Cell Mol Biol ; 32(6): 548-52, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15746432

RESUMEN

Submucosal glands are abundant (approximately 1 gland/mm2) secretory structures in the tracheobronchial airways of the human lung. Because submucosal glands express antibacterial proteins, it has been proposed that they contribute to lung defense. However, this concept is challenged by the fact that mice do not have submucosal glands in their bronchial airways, yet are quite resistant to bacterial lung infection. The contribution of airway submucosal glands to host defense is also debated as a pathophysiologic component of cystic fibrosis lung disease. Here, we asked whether submucosal glands protect airways against bacterial infection. By comparing tracheal xenograft airways with and without glands, we found that the presence of glands enhanced bacterial killing in vivo and by airway secretions in vitro. Moreover, immunodepletion studies suggested that lysozyme is a major antibacterial component secreted by submucosal glands. These studies provide evidence that submucosal glands are a major source of antibacterials critical for maintaining sterile airways.


Asunto(s)
Infecciones Bacterianas/inmunología , Muramidasa/metabolismo , Mucosa Respiratoria/enzimología , Mucosa Respiratoria/inmunología , Tráquea/inmunología , Animales , Bronquios/enzimología , Bronquios/inmunología , Bronquios/metabolismo , Hurones , Ratas , Mucosa Respiratoria/metabolismo , Tráquea/enzimología , Tráquea/metabolismo , Trasplante Heterólogo
16.
Laryngoscope ; 114(11): 2021-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15510034

RESUMEN

OBJECTIVES: Factors that alter airway surface liquid (ASL) ionic concentrations may influence the course of sinusitis. Xylitol has been shown to effect ASL ionic composition in vitro and to reduce nasal bacterial carriage, otitis media, and dental caries in vivo. We examined the effect of xylitol on experimental sinusitis in the rabbit model. STUDY DESIGN: Prospective randomized controlled study of xylitol, saline, and Pseudomonas aeruginosa administration to the rabbit maxillary sinus. METHODS: P. aeruginosa was administered to the sinuses of 26 New Zealand white rabbits. Saline was placed in the left maxillary sinus and xylitol in the right. The rabbits were randomly assigned to one of three groups: one, simultaneous administration of bacteria and solutions with bacterial analysis at 20 minutes, 11 rabbits; two, preadministration of solutions 1 hour before bacterial infection with analysis at 20 minutes, 11 rabbits; three, established sinusitis, 4 rabbits had daily injections of solutions for 5 days starting 7 days after P. aeruginosa administration. RESULTS: In group 1, 6.96% of injected bacteria were retrieved on the left (saline), whereas 0.095% were retrieved on the right (xylitol) (P = .034). In group 2, 5.64% of inoculum was recovered from the left and 2.89% from the right (P = .188). Group 3 demonstrated evidence of sinusitis with recovery of noninoculate bacteria. with no difference between right and left. CONCLUSIONS: Xylitol reduces experimental sinusitis when administered simultaneously with bacteria. Its effect in established sinusitis is less clear. A role may exist for xylitol in nasal irrigation fluid in human disease.


Asunto(s)
Sinusitis Maxilar/tratamiento farmacológico , Sinusitis Maxilar/microbiología , Infecciones por Pseudomonas/tratamiento farmacológico , Xilitol/uso terapéutico , Animales , Incidencia , Estudios Prospectivos , Infecciones por Pseudomonas/microbiología , Conejos , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...