Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 51(4): 550-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24787463

RESUMEN

Claudins are a family of transmembrane proteins that are required for tight junction formation. Claudin (CLDN)-18.1, the only known lung-specific tight junction protein, is the most abundant claudin in alveolar epithelial type (AT) 1 cells, and is regulated by lung maturational agonists and inflammatory mediators. To determine the function of CLDN18 in the alveolar epithelium, CLDN18 knockout (KO) mice were generated and studied by histological, biochemical, and physiological approaches, in addition to whole-genome microarray. Alveolar epithelial barrier function was assessed after knockdown of CLDN18 in isolated lung cells. CLDN18 levels were measured by quantitative PCR in lung samples from fetal and postnatal human infants. We found that CLDN18 deficiency impaired alveolar epithelial barrier function in vivo and in vitro, with evidence of increased paracellular permeability and architectural distortion at AT1-AT1 cell junctions. Although CLDN18 KO mice were born without evidence of a lung abnormality, histological and gene expression analysis at Postnatal Day 3 and Week 4 identified impaired alveolarization. CLDN18 KO mice also had evidence of postnatal lung injury, including acquired AT1 cell damage. Human fetal lungs at 23-24 weeks gestational age, the highest-risk period for developing bronchopulmonary dysplasia, a disease of impaired alveolarization, had significantly lower CLDN18 expression relative to postnatal lungs. Thus, CLDN18 deficiency results in epithelial barrier dysfunction, injury, and impaired alveolarization in mice. Low expression of CLDN18 in human fetal lungs supports further investigation into a role for this tight junction protein in bronchopulmonary dysplasia.


Asunto(s)
Claudinas/deficiencia , Claudinas/metabolismo , Alveolos Pulmonares/metabolismo , Uniones Estrechas/metabolismo , Animales , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Claudinas/genética , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Humanos , Lactante , Recién Nacido , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Alveolos Pulmonares/embriología , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Factores de Riesgo , Uniones Estrechas/patología
2.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L970-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24142515

RESUMEN

The lipid transport protein, ABCA3, expressed in alveolar type 2 (AT2) cells, is critical for surfactant homeostasis. The first luminal loop of ABCA3 contains three putative N-linked glycosylation sites at residues 53, 124, and 140. A common cotranslational modification, N-linked glycosylation, is critical for the proper expression of glycoproteins by enhancing folding, trafficking, and stability through augmentation of the endoplasmic reticulum (ER) folding cycle. To understand its role in ABCA3 biosynthesis, we utilized EGFP-tagged fusion constructs with either wild-type or mutant ABCA3 cDNAs that contained glutamine for asparagine substitutions at the putative glycosylation motifs. In A549 cells, inhibition of glycosylation by tunicamycin increased the electrophoretic mobility (Mr) and reduced the expression level of wild-type ABCA3 in a dose-dependent manner. Fluorescence imaging of transiently transfected A549 or primary human AT2 cells showed that although single motif mutants exhibited a vesicular distribution pattern similar to wild-type ABCA3, mutation of N124 and N140 residues resulted in a shift toward an ER-predominant distribution. By immunoblotting, the N53 mutation exhibited no effect on either the Mr or ABCA3 expression level. In contrast, substitutions at N124 or N140, as well a N124/N140 double mutation, resulted in increased electrophoretic mobility indicative of a glycosylation deficiency accompanied by reduced overall expression levels. Diminished steady-state levels of glycan-deficient ABCA3 isoforms were rescued by treatment with the proteasome inhibitor MG132. These results suggest that cotranslational N-linked glycosylation at N124 and N140 is critical for ABCA3 stability, and its disruption results in protein destabilization and proteasomal degradation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Retículo Endoplásmico/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Células Cultivadas , Retículo Endoplásmico/genética , Glicosilación/efectos de los fármacos , Humanos , Mutación/genética , Complejo de la Endopetidasa Proteasomal/genética , Transporte de Proteínas/genética , Tunicamicina/farmacología
3.
Am J Physiol Lung Cell Mol Physiol ; 302(2): L216-25, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22037359

RESUMEN

Carcinoembryonic cell adhesion molecule 6 (CEACAM6) is a glycosylated, glycophosphatidylinositol-anchored protein expressed in epithelial cells of various primate tissues. It binds gram-negative bacteria and is overexpressed in human cancers. CEACAM6 is associated with lamellar bodies of cultured type II cells of human fetal lung and protects surfactant function in vitro. In this study, we characterized CEACAM6 expression in vivo in human lung. CEACAM6 was present in lung lavage of premature infants at birth and increased progressively in intubated infants with lung disease. Of surfactant-associated CEACAM6, ∼80% was the fully glycosylated, 90-kDa form that contains the glycophosphatidylinositol anchor, and the concentration (3.9% of phospholipid for adult lung) was comparable to that for surfactant proteins (SP)-A/B/C. We examined the affinity of CEACAM6 by purification of surfactant on density gradient centrifugation; concentrations of CEACAM6 and SP-B per phospholipid were unchanged, whereas levels of total protein and SP-A decreased by 60%. CEACAM6 mRNA content decreased progressively from upper trachea to peripheral fetal lung, whereas protein levels were similar in all regions of adult lung, suggesting proximal-to-distal developmental expression in lung epithelium. In adult lung, most type I cells and ∼50% of type II cells were immunopositive. We conclude that CEACAM6 is expressed by alveolar and airway epithelial cells of human lung and is secreted into lung-lining fluid, where fully glycosylated protein binds to surfactant. Production appears to be upregulated during neonatal lung disease, perhaps related to roles of CEACAM6 in surfactant function, cell proliferation, and innate immune defense.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Pulmón/metabolismo , Surfactantes Pulmonares/metabolismo , Adulto , Antígenos CD/química , Líquido del Lavado Bronquioalveolar/química , Adhesión Celular , Moléculas de Adhesión Celular/química , Células Epiteliales/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Glicosilación , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/metabolismo , Pulmón/citología , Pulmón/embriología , Enfermedades Pulmonares/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tráquea/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 299(1): L59-72, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20382748

RESUMEN

Neonatal chronic lung disease is characterized by failed formation of alveoli and capillaries, and excessive deposition of matrix elastin, which are linked to lengthy mechanical ventilation (MV) with O(2)-rich gas. Vitamin A supplementation has improved respiratory outcome of premature infants, but there is little information about the structural and molecular manifestations in the lung that occur with vitamin A treatment. We hypothesized that vitamin A supplementation during prolonged MV, without confounding by antenatal steroid treatment, would improve alveolar secondary septation, decrease thickness of the mesenchymal tissue cores between distal air space walls, and increase alveolar capillary growth. We further hypothesized that these structural advancements would be associated with modulated expression of tropoelastin and deposition of matrix elastin, phosphorylated Smad2 (pSmad2), cleaved caspase 3, proliferating cell nuclear antigen (PCNA), VEGF, VEGF-R2, and midkine in the parenchyma of the immature lung. Eight preterm lambs (125 days' gestation, term approximately 150 days) were managed by MV for 3 wk: four were treated with daily intramuscular Aquasol A (vitamin A), 5,000 IU/kg, starting at birth; four received vehicle alone. Postmortem lung assays included quantitative RT-PCR and in situ hybridization, immunoblot and immunohistochemistry, and morphometry and stereology. Daily vitamin A supplementation increased alveolar secondary septation, decreased thickness of the mesenchymal tissue cores between the distal air space walls, and increased alveolar capillary growth. Associated molecular changes were less tropoelastin mRNA expression, matrix elastin deposition, pSmad2, and PCNA protein localization in the mesenchymal tissue core of the distal air space walls. On the other hand, mRNA expression and protein abundance of VEGF, VEGF-R2, midkine, and cleaved caspase 3 were increased. We conclude that vitamin A treatment partially improves lung development in chronically ventilated preterm neonates by modulating expression of tropoelastin, deposition of elastin, and expression of vascular growth factors.


Asunto(s)
Enfermedades Pulmonares/dietoterapia , Enfermedades Pulmonares/fisiopatología , Enfermedades Pulmonares/veterinaria , Pulmón , Alveolos Pulmonares , Vitamina A , Vitaminas , Animales , Animales Recién Nacidos , Enfermedad Crónica , Suplementos Dietéticos , Elastina/genética , Elastina/metabolismo , Femenino , Edad Gestacional , Pulmón/efectos de los fármacos , Pulmón/crecimiento & desarrollo , Pulmón/patología , Enfermedades Pulmonares/patología , Embarazo , Nacimiento Prematuro , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/ultraestructura , Intercambio Gaseoso Pulmonar , Respiración Artificial , Ovinos , Tropoelastina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vitamina A/sangre , Vitamina A/farmacología , Vitamina A/uso terapéutico , Vitaminas/farmacología , Vitaminas/uso terapéutico
5.
Am J Physiol Lung Cell Mol Physiol ; 299(1): L36-50, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20382749

RESUMEN

Alveolar type II cells have multiple functions, including surfactant production and fluid clearance, which are critical for lung function. Differentiation of type II cells occurs in cultured fetal lung epithelial cells treated with dexamethasone plus cAMP and isobutylmethylxanthine (DCI) and involves increased expression of 388 genes. In this study, type II cells of human adult lung were isolated at approximately 95% purity, and gene expression was determined (Affymetrix) before and after culturing 5 days on collagen-coated dishes with or without DCI for the final 3 days. In freshly isolated cells, highly expressed genes included SFTPA/B/C, SCGB1A, IL8, CXCL2, and SFN in addition to ubiquitously expressed genes. Transcript abundance was correlated between fetal and adult cells (r = 0.88), with a subset of 187 genes primarily related to inflammation and immunity that were expressed >10-fold higher in adult cells. During control culture, expression increased for 8.1% of expressed genes and decreased for approximately 4% including 118 immune response and 10 surfactant-related genes. DCI treatment promoted lamellar body production and increased expression of approximately 3% of probed genes by > or =1.5-fold; 40% of these were also induced in fetal cells. Highly induced genes (> or =10-fold) included PGC, ZBTB16, DUOX1, PLUNC, CIT, and CRTAC1. Twenty-five induced genes, including six genes related to surfactant (SFTPA/B/C, PGC, CEBPD, and ADFP), also had decreased expression during control culture and thus are candidates for hormonal regulation in vivo. Our results further define the adult human type II cell molecular phenotype and demonstrate that a subset of genes remains hormone responsive in cultured adult cells.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica , Pulmón/citología , Adulto , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/farmacología , Dexametasona/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Humanos , Pulmón/fisiología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Ratas
6.
Pediatr Res ; 67(5): 521-5, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20098340

RESUMEN

Clinical trials demonstrated decreasing rates of bronchopulmonary dysplasia in preterm infants with hypoxic respiratory failure treated with inhaled nitric oxide (iNO). However, the molecular and biochemical effects of iNO on developing human fetal lungs remain vastly unknown. By using a well-characterized model of human fetal alveolar type II cells, we assessed the effects of iNO and hyperoxia, independently and concurrently, on NO-cGMP signaling pathway and differentiation. Exposure to iNO increased cGMP levels by 40-fold after 3 d and by 8-fold after 5 d despite constant expression of phosphodiesterase-5 (PDE5). The levels of cGMP declined significantly on exposure to iNO and hyperoxia at 3 and 5 d, although expression of soluble guanylyl cyclase (sGC) was sustained. Surfactant proteins B and C (SP-B, SP-C) and thyroid transcription factor (TTF)-1 mRNA levels increased in cells exposed to iNO in normoxia but not on exposure to iNO plus hyperoxia. Collectively, these data indicate an increase in type II cell markers when undifferentiated lung epithelial cells are exposed to iNO in room air. However, hyperoxia overrides these potentially beneficial effects of iNO despite sustained expression of sGC.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Diferenciación Celular , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Biomarcadores/metabolismo , Hipoxia de la Célula , Células Cultivadas , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Regulación de la Expresión Génica , Edad Gestacional , Guanilato Ciclasa/metabolismo , Humanos , Proteínas Nucleares/genética , Proteína B Asociada a Surfactante Pulmonar/genética , Proteína C Asociada a Surfactante Pulmonar/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Guanilil Ciclasa Soluble , Factor Nuclear Tiroideo 1 , Factores de Tiempo , Factores de Transcripción/genética
7.
Pediatr Res ; 67(6): 585-90, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20220547

RESUMEN

Stretch is an essential mechanism for lung growth and development. Animal models in which fetal lungs have been chronically over or underdistended demonstrate a disrupted mix of type II and type I cells, with static overdistention typically promoting a type I cell phenotype. The Rho GTPase family, key regulators of cytoskeletal signaling, are known to mediate cellular differentiation in response to stretch in other organs. Using a well-described model of alveolar epithelial cell differentiation and a validated stretch device, we investigated the effects of supraphysiologic stretch on human fetal lung alveolar epithelial cell phenotype. Static stretch applied to epithelial cells suppressed type II cell markers (SP-B and Pepsinogen C, PGC), and induced type I cell markers (Caveolin-1, Claudin 7 and Plasminogen Activator Inhibitor-1, PAI-1) as predicted. Static stretch was also associated with Rho A activation. Furthermore, the Rho kinase inhibitor Y27632 decreased Rho A activation and blunted the stretch-induced changes in alveolar epithelial cell marker expression. Together these data provide further evidence that mechanical stimulation of the cytoskeleton and Rho activation are key upstream events in mechanotransduction-associated alveolar epithelial cell differentiation.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Diferenciación Celular , Forma de la Célula , Mecanotransducción Celular , Proteína de Unión al GTP rhoA/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Amidas/farmacología , Biomarcadores/metabolismo , Caveolina 1/metabolismo , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Claudinas , Edad Gestacional , Humanos , Pulmón/embriología , Pulmón/enzimología , Mecanotransducción Celular/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Pepsinógeno C/metabolismo , Fenotipo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Piridinas/farmacología , Fibras de Estrés/metabolismo , Factores de Tiempo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
8.
Respir Res ; 10: 77, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19698107

RESUMEN

BACKGROUND: A precise balance exists between the actions of endogenous glucocorticoids (GC) and retinoids to promote normal lung development, in particular during alveolarization. The mechanisms controlling this balance are largely unknown, but recent evidence suggests that midkine (MK), a retinoic acid-regulated, pro-angiogenic growth factor, may function as a critical regulator. The purpose of this study was to examine regulation of MK by GC and RA during postnatal alveolar formation in rats. METHODS: Newborn rats were treated with dexamethasone (DEX) and/or all-trans-retinoic acid (RA) during the first two weeks of life. Lung morphology was assessed by light microscopy and radial alveolar counts. MK mRNA and protein expression in response to different treatment were determined by Northern and Western blots. In addition, MK protein expression in cultured human alveolar type 2-like cells treated with DEX and RA was also determined. RESULTS: Lung histology confirmed that DEX treatment inhibited and RA treatment stimulated alveolar formation, whereas concurrent administration of RA with DEX prevented the DEX effects. During normal development, MK expression was maximal during the period of alveolarization from postnatal day 5 (PN5) to PN15. DEX treatment of rat pups decreased, and RA treatment increased lung MK expression, whereas concurrent DEX+RA treatment prevented the DEX-induced decrease in MK expression. Using human alveolar type 2 (AT2)-like cells differentiated in culture, we confirmed that DEX and cAMP decreased, and RA increased MK expression. CONCLUSION: We conclude that MK is expressed by AT2 cells, and is differentially regulated by corticosteroid and retinoid treatment in a manner consistent with hormonal effects on alveolarization during postnatal lung development.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Citocinas/metabolismo , Dexametasona/farmacología , Células Epiteliales/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Tretinoina/farmacología , Factores de Edad , Proteínas Angiogénicas/genética , Animales , Animales Recién Nacidos , Northern Blotting , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Citocinas/genética , Células Epiteliales/metabolismo , Humanos , Midkina , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
9.
Biochim Biophys Acta ; 1575(1-3): 82-90, 2002 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-12020822

RESUMEN

Members of the CCAAT enhancer binding protein (C/EBP) transcription factor family were detected in fetal lung of both human and rat. In rat lung, the level of C/EBPs increased with time of gestation, peaking around birth. In adult rat lung, C/EBPs were localized to the alveolar type II cells. The effect of C/EBPs on pulmonary surfactant protein A (SP-A), which is also expressed late in gestation, was investigated. In contrast to control plasmids, C/EBP delta expressing plasmids reversed the action of a transcriptional silencer just upstream of the rat SP-A promoter. In order to test the effect of C/EBPs on endogenous SP-A gene expression, cells that express SP-A were exposed to a phosphorothioate-substituted, double-stranded oligonucleotide matching the consensus C/EBP binding site (decoy oligonucleotide) at concentrations from 0.5 to 10 microM for 72 h. A mutant oligonucleotide with an 8-base pair (bp) substitution served as a control. The decoy oligonucleotide reduced SP-A mRNA as much as 75% compared to a mutant oligonucleotide both in the human lung cell line, NCI-H441, and in primary human fetal alveolar type II cells. The data indicate that C/EBPs facilitate SP-A gene expression, possibly by overcoming transcriptional silencing.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteolípidos/genética , Surfactantes Pulmonares/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Pulmón/fisiología , Proteolípidos/biosíntesis , Proteína A Asociada a Surfactante Pulmonar , Proteínas Asociadas a Surfactante Pulmonar , Surfactantes Pulmonares/biosíntesis , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
J Histochem Cytochem ; 63(12): 908-21, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-26374831

RESUMEN

The membrane protein carcinoembryonic antigen cell adhesion molecule (CEACAM6) is expressed in the epithelium of various tissues, participating in innate immune defense, cell proliferation and differentiation, with overexpression in gastrointestinal tract, pancreatic and lung tumors. It is developmentally and hormonally regulated in fetal human lung, with an apparent increased production in preterm infants with respiratory failure. To further examine the expression and cell localization of CEACAM6, we performed immunohistochemical and biochemical studies in lung specimens from infants with and without chronic lung disease. CEACAM6 protein and mRNA were increased ~4-fold in lungs from infants with chronic lung disease as compared with controls. By immunostaining, CEACAM6 expression was markedly increased in the lung parenchyma of infants and children with a variety of chronic lung disorders, localizing to hyperplastic epithelial cells with a ~7-fold elevated proliferative rate by PCNA staining. Some of these cells also co-expressed membrane markers of both type I and type II cells, which is not observed in normal postnatal lung, suggesting they are transitional epithelial cells. We suggest that CEACAM6 is both a marker of lung epithelial progenitor cells and a contributor to the proliferative response after injury due to its anti-apoptotic and cell adhesive properties.


Asunto(s)
Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Enfermedades Pulmonares/genética , ARN Mensajero/genética , Insuficiencia Respiratoria/genética , Antígenos CD/metabolismo , Acuaporina 5/genética , Acuaporina 5/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Moléculas de Adhesión Celular/metabolismo , Enfermedad Crónica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Humanos , Lactante , Recién Nacido , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Surfactantes Pulmonares/metabolismo , ARN Mensajero/metabolismo , Insuficiencia Respiratoria/metabolismo , Insuficiencia Respiratoria/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Células Madre/metabolismo , Células Madre/patología , Factores de Transcripción
11.
Physiol Rep ; 3(12)2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26702074

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is expressed in the epithelium of various primate tissues, including lung airway and alveoli. In human lung, CEACAM6 is developmentally and hormonally regulated, protects surfactant function, has anti-apoptotic activity and is dysregulated in cancers. We hypothesized that alveolar CEACAM6 expression increases in lung injury and promotes cell proliferation during repair. Studies were performed in CEABAC transgenic mice-containing human CEACAM genes. The level of CEACAM6 in adult CEABAC lung was comparable to that in human infants; expression occurred in epithelium of airways and of some alveoli but rarely co-localized with markers of type I or type II cells. Ten days after bleomycin instillation, both the number of CEACAM6(+) cells and immunostaining intensity were elevated in injured lung areas, and there was increased co-localization with type I and II cell markers. To specifically address type II cells, we crossed CEABAC mice with animals expressing EGFP driven by the SP-C promoter. After bleomycin injury, partially flattened, elongated epithelial cells were observed that expressed type I cell markers and were primarily either EGFP(+) or CEACAM6(+). In cell cycle studies, mitosis was greater in CEACAM6(+) non-type II cells versus CEACAM6(+)/EGFP(+) cells. CEACAM6 epithelial expression was also increased after hyperoxic exposure and LPS instillation, suggesting a generalized response to acute lung injuries. We conclude that CEACAM6 expression is comparable in human lung and the CEABAC mouse. CEACAM6 in this model appears to be a marker of a progenitor cell population that contributes to alveolar epithelial cell replenishment after lung injury.

12.
Open Cell Dev Biol J ; 3: 1-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-25580166

RESUMEN

Inflammation is a major contributor to the pathogenesis of bronchopulmonary dysplasia (BPD). BPD is associated with prematurity of birth, sepsis, with increased production of both cytokines and nitric oxide, and with the shedding of bronchial epithelial cells. The pathological mechanisms involved in this disease remain unclear, in particular the role that epithelial maturity plays. The effects of pro-inflammatory cytokines upon immature and mature cells are examined within this study, using primary culture of human lung epithelial cells. Pro-inflammatory cytokines increase inducible nitric oxide synthase (iNOS) expression and raise NO production, irrespective of cellular maturity. Pre-incubation with 1400W, a specific iNOS inhibitor, abrogated pro-inflammatory cytokine-induced NO generation and apoptosis. However, immature fetal lung epithelial cells were uniquely sensitive to cellular injury in response to cytokine exposure. These observations suggest that pro-inflammatory cytokines, which are present within BPD, may cause apoptosis of lung epithelial cells via de novo generation of NO. Furthermore, the prematurity of lung epithelial cells may be a factor in free radical mediated pulmonary damage.

13.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L1019-30, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19329538

RESUMEN

Carcinoembryonic cell adhesion molecule 6 (CEACAM6) is a glycosylated, glycosylphosphatidylinositol (GPI)-anchored protein expressed in epithelial cells of various human tissues. It binds gram-negative bacteria and is overexpressed in cancers, where it is antiapoptotic and promotes metastases. To characterize CEACAM6 expression in developing lung, we cultured human fetal lung epithelial cells and examined responses to differentiation-promoting hormones, adenovirus expressing thyroid transcription factor-1 (TTF-1), and silencing of TTF-1 with small inhibitory RNA. Glucocorticoid and cAMP had additive stimulatory effects on CEACAM6 content, and combined treatment maximally increased transcription rate, mRNA, and protein approximately 10-fold. Knockdown of TTF-1 reduced hormone induction of CEACAM6 by 80%, and expression of recombinant TTF-1 increased CEACAM6 in a dose-dependent fashion. CEACAM6 content of lung tissue increased during the third trimester and postnatally. By immunostaining, CEACAM6 was present in fetal type II cells, but not mesenchymal cells, and localized to both the plasma membrane and within surfactant-containing lamellar bodies. CEACAM6 was secreted from cultured type II cells and was present in both surfactant and supernatant fractions of infant tracheal aspirates. In functional studies, CEACAM6 reduced inhibition of surfactant surface properties by proteins in vitro and blocked apoptosis of electroporated cultured cells. We conclude that CEACAM6 in fetal lung epithelial cells is developmentally and hormonally regulated and a target protein for TTF-1. Because CEACAM6 acts as an antiapoptotic factor and stabilizes surfactant function, in addition to a putative role in innate defense against bacteria, we propose that it is a multifunctional alveolar protein.


Asunto(s)
Antígenos CD/genética , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas Nucleares/metabolismo , Alveolos Pulmonares/fisiología , Mucosa Respiratoria/fisiología , Factores de Transcripción/metabolismo , Apoptosis/fisiología , Células Cultivadas , AMP Cíclico/farmacología , Sinergismo Farmacológico , Feto/citología , Proteínas Ligadas a GPI , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucocorticoides/farmacología , Humanos , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Alveolos Pulmonares/citología , Alveolos Pulmonares/embriología , Surfactantes Pulmonares/metabolismo , ARN Interferente Pequeño , Mucosa Respiratoria/citología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética
14.
Pediatr Pulmonol ; 43(7): 703-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18500734

RESUMEN

Bronchopulmonary dysplasia (BPD), a multifactorial disease of preterm neonates of complex etiology, is a significant problem within very low birth weight infants. Nitric oxide (NO) has been implicated in both the pathogenesis and as a potential therapeutic of this disease. At this time, there is little direct evidence of the changes in NO production and metabolism that occur within BPD in humans. Animal models have implied that reduced nitric oxide synthase (NOS) expression and NO production in the early stages of the disease may be critical factors. However, inflammation and hence iNOS expression, is also thought to play a role. In the present study we have utilized pathological samples to determine changes in the expression of NOS and NO metabolites within late stage BPD. It is our contention that within these samples iNOS expression is increased and associated with increased NO metabolite production. Mild immunostaining of all three nitric oxide synthase (NOS) enzymes (neuronal, inducible and endothelial) is observed in control lung with tight localization to the endothelium and epithelial airway. This tight localization was lost in samples from subjects with BPD. There was also a marked increase in iNOS expression throughout the lung tissue with strong coexistence with an epithelial cell marker cytokeratin. NO reaction products are altered with BPD as evidenced by increased S-nitrosothiol (SNO) and strong nitrotyrosine (NO(2)Y) imunoreactivity. This study demonstrates a strong correlation between products of NO reactivity and NOS localization in BPD.


Asunto(s)
Displasia Broncopulmonar/enzimología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/metabolismo , Biomarcadores , Células Epiteliales/enzimología , Femenino , Expresión Génica , Edad Gestacional , Humanos , Recién Nacido , Recién Nacido de muy Bajo Peso , Queratinas , Masculino
15.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1187-96, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18375741

RESUMEN

Chemokine receptors control several fundamental cellular processes in both hematopoietic and structural cells, including directed cell movement, i.e., chemotaxis, cell differentiation, and proliferation. We have previously demonstrated that CXCR3, the chemokine receptor expressed by Th1/Tc1 inflammatory cells present in the lung, is also expressed by human airway epithelial cells. In airway epithelial cells, activation of CXCR3 induces airway epithelial cell movement and proliferation, processes that underlie lung repair. The present study examined the expression and function of CXCR3 in human alveolar type II pneumocytes, whose destruction causes emphysema. CXCR3 was present in human fetal and adult type II pneumocytes as assessed by immunocytochemistry, immunohistochemistry, and Western blotting. CXCR3-A and -B splice variant mRNA was present constitutively in cultured type II cells, but levels of CXCR3-B greatly exceeded CXCR3-A mRNA. In cultured type II cells, I-TAC, IP-10, and Mig induced chemotaxis. Overexpression of CXCR3-A in the A549 pneumocyte cell line produced robust chemotactic responses to I-TAC and IP-10. In contrast, I-TAC did not induce chemotactic responses in CXCR3-B and mock-transfected cells. Finally, I-TAC increased cytosolic Ca(2+) and activated the extracellular signal-regulated kinase, p38, and phosphatidylinositol 3-kinase (PI 3-kinase)/protein kinase B kinases only in CXCR3-A-transfected cells. These data indicate that the CXCR3 receptor is expressed by human type II pneumocytes, and the CXCR3-A splice variant mediates chemotactic responses possibly through Ca(2+) activation of both mitogen-activated protein kinase and PI 3-kinase signaling pathways. Expression of CXCR3 in alveolar epithelial cells may be important in pneumocyte repair from injury.


Asunto(s)
Quimiotaxis/fisiología , Pulmón/citología , Neumonía/fisiopatología , Receptores CXCR3/fisiología , Adulto , Empalme Alternativo , Calcio/metabolismo , Línea Celular , Células Cultivadas , Dexametasona/farmacología , Humanos , Pulmón/embriología , Pulmón/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transfección , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 292(1): L249-57, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16997883

RESUMEN

In the fetal lung, endogenous transforming growth factor (TGF)-beta inhibits early morphogenesis and blocks hormone-induced type II cell differentiation. We hypothesized that endogenous TGF-beta inhibits type II cell differentiation and that the stimulatory effects of glucocorticoids result in part from suppression of TGF-beta. Epithelial cells were isolated from human fetal lung and cultured under defined conditions with and without dexamethasone plus cAMP to promote type II cell differentiation. Control cells produced TGF-beta, which was activated in part by alpha(V)beta(6)-integrin. Treatment with dexamethasone, but not cAMP, reduced TGF-beta1 and -beta2 transcripts and TGF-beta bioactivity in culture medium. To examine the effects of decreased TGF-beta in the absence of glucocorticoid, cells were treated with antibodies to TGF-beta and its receptors. By real-time RT-PCR, antibody blockade of TGF-beta reduced serpine1, a TGF-beta-inducible gene, and increased gene expression for sftpa, sftpb, sftpc, and titf1, mimicking the response to hormone treatment. By microarray analysis, 29 additional genes were induced by both TGF-beta antibody and hormone treatment, and 20 other genes were repressed by both treatments. For some genes, the fold response was comparable for antibody and hormone treatment. We conclude that endogenous TGF-beta suppresses expression of surfactant proteins and selected other type II cell genes in fetal lung, in part secondary to increased expression of titf1, and we propose that the mechanism of glucocorticoid-induced type II cell differentiation includes antagonism of TGF-beta gene suppression. Surfactant production during fetal development is likely influenced by relative levels of TGF-beta and glucocorticoids.


Asunto(s)
Glucocorticoides/farmacología , Pulmón/citología , Pulmón/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Dexametasona/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Feto/citología , Feto/metabolismo , Perfilación de la Expresión Génica , Humanos , Pulmón/metabolismo , Visón , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Transducción de Señal , Factores de Transcripción , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética
17.
Pediatr Res ; 61(4): 404-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17515862

RESUMEN

For alveolar type I cells, phenotype plasticity and physiology other than gas exchange await further clarification due to in vitro study difficulties in isolating and maintaining type I cells in primary culture. Using an established in vitro model of human fetal type II cells, in which the type II phenotype is induced and maintained by adding hormones, we assessed for transdifferentiation in culture toward a type I-like cell with hormone removal for up to 144 h, followed by electron microscopy, permeability studies, and RNA and protein analysis. Hormone withdrawal resulted in diminished type II cell characteristics, including decreased microvilli, lamellar bodies, and type II cell marker RNA and protein. There was a simultaneous increase in type I characteristics, including increased epithelial cell barrier function indicative of a tight monolayer and increased type I cell marker RNA and protein. Our results indicate that hormone removal from cultured human fetal type II cells results in transdifferentiation toward a type I-like cell. This model will be useful for continued in vitro studies of human fetal alveolar epithelial cell differentiation and phenotype plasticity.


Asunto(s)
Diferenciación Celular/fisiología , Feto/citología , Alveolos Pulmonares/citología , Mucosa Respiratoria/citología , Permeabilidad de la Membrana Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos
18.
Am J Respir Cell Mol Biol ; 36(2): 213-25, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16960125

RESUMEN

Thyroid transcription factor-1 (TTF-1, product of the Nkx2.1 gene) is essential for branching morphogenesis of the lung and enhances expression of surfactant proteins by alveolar type II cells. We investigated expression of two TTF-1 mRNA transcripts, generated by alternative start sites and coding for 42- and 46-kD protein isoforms in the mouse, during hormone-induced differentiation of human fetal lung type II cells in culture. Transcript for 42-kD TTF-1 was 20-fold more abundant than TTF-1(46) mRNA by RT-PCR. Only 42-kD protein was detected in lung cells, and its content increased during in vivo development and in response to in vitro glucocorticoid plus cAMP treatment. To examine TTF-1 target proteins, recombinant, phosphorylated TTF-1(42) was expressed in nuclei of cells by adenovirus transduction. By microarray analysis, 14 genes were comparably induced by recombinant TTF-1 (rTTF-1) and hormone treatment, and 9 additional hormone-responsive genes, including surfactant proteins-A/B/C, were partially induced by rTTF-1. The most highly (approximately 10-fold) TTF-1-induced genes were DC-LAMP (LAMP3) and CEACAM6 with induction confirmed by Western analysis and immunostaining. Treatment of cells with hormones plus small inhibitory RNA directed toward TTF-1 reduced TTF-1 content by approximately 50% and inhibited hormone induction of the 23 genes induced by rTTF-1. In addition, knockdown of TTF-1 inhibited 72 of 274 other genes induced by hormones. We conclude that 42-kD TTF-1 is required for induction of a subset of regulated genes during type II cell differentiation.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Alveolos Pulmonares/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adenoviridae , Animales , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Dexametasona/farmacología , Proteínas Ligadas a GPI , Regulación de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Células HeLa , Humanos , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Proteínas Nucleares/análisis , Biosíntesis de Proteínas/efectos de los fármacos , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/embriología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor Nuclear Tiroideo 1 , Factores de Transcripción/análisis , Transcripción Genética/efectos de los fármacos , Transducción Genética
19.
J Biol Chem ; 281(14): 9791-800, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16415354

RESUMEN

Members of the ATP binding cassette (ABC) protein superfamily actively transport a wide range of substrates across cell and intracellular membranes. Mutations in ABCA3, a member of the ABCA subfamily with unknown function, lead to fatal respiratory distress syndrome (RDS) in the newborn. Using cultured human lung cells, we found that recombinant wild-type hABCA3 localized to membranes of both lysosomes and lamellar bodies, which are the intracellular storage organelles for surfactant. In contrast, hABCA3 with mutations linked to RDS failed to target to lysosomes and remained in the endoplasmic reticulum as unprocessed forms. Treatment of those cells with the chemical chaperone sodium 4-phenylbutyrate could partially restore trafficking of mutant ABCA3 to lamellar body-like structures. Expression of recombinant ABCA3 in non-lung human embryonic kidney 293 cells induced formation of lamellar body-like vesicles that contained lipids. Small interfering RNA knockdown of endogenous hABCA3 in differentiating human fetal lung alveolar type II cells resulted in abnormal, lamellar bodies comparable with those observed in vivo with mutant ABCA3. Silencing of ABCA3 expression also reduced vesicular uptake of surfactant lipids phosphatidylcholine, sphingomyelin, and cholesterol but not phosphatidylethanolamine. We conclude that ABCA3 is required for lysosomal loading of phosphatidylcholine and conversion of lysosomes to lamellar body-like structures.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Fosfatidilcolinas/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/fisiopatología , Transportadoras de Casetes de Unión a ATP/biosíntesis , Técnicas de Cultivo de Célula , Regulación hacia Abajo , Retículo Endoplásmico , Feto , Silenciador del Gen , Proteínas Fluorescentes Verdes/análisis , Humanos , Recién Nacido , Riñón/citología , Riñón/embriología , Metabolismo de los Lípidos , Pulmón/citología , Lisosomas/química , Microscopía Confocal , Mutación , Transporte de Proteínas , Alveolos Pulmonares/citología , ARN Interferente Pequeño
20.
J Lipid Res ; 47(6): 1322-31, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16513897

RESUMEN

Maturation of fetal alveolar type II epithelial cells in utero is characterized by specific changes to lung surfactant phospholipids. Here, we quantified the effects of hormonal differentiation in vitro on the molecular specificity of cellular and secreted phospholipids from human fetal type II epithelial cells using electrospray ionization mass spectrometry. Differentiation, assessed by morphology and changes in gene expression, was accompanied by restricted and specific modifications to cell phospholipids, principally enrichments of shorter chain species of phosphatidylcholine (PC) and phosphatidylinositol, that were not observed in fetal lung fibroblasts. Treatment of differentiated epithelial cells with secretagogues stimulated the secretion of functional surfactant-containing surfactant proteins B and C (SP-B and SP-C). Secreted material was further enriched in this same set of phospholipid species but was characterized by increased contents of short-chain monounsaturated and disaturated species other than dipalmitoyl PC (PC16:0/16:0), principally palmitoylmyristoyl PC (PC16:0/14:0) and palmitoylpalmitoleoyl PC (PC16:0/16:1). Mixtures of these PC molecular species, phosphatidylglycerol, and SP-B and SP-C were functionally active and rapidly generated low surface tension on compression in a pulsating bubble surfactometer. These results suggest that hormonally differentiated human fetal type II cells do not select the molecular composition of surfactant phospholipid on the basis of saturation but, more likely, on the basis of acyl chain length.


Asunto(s)
Células Epiteliales/metabolismo , Metabolismo de los Lípidos , Fosfolípidos/metabolismo , Alveolos Pulmonares/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Diferenciación Celular , Células Cultivadas , Dexametasona/farmacología , Dimiristoilfosfatidilcolina/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Humanos , Fosfatidilcolinas/metabolismo , Fosfolípidos/química , Alveolos Pulmonares/citología , Alveolos Pulmonares/efectos de los fármacos , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Surfactantes Pulmonares/química , Tensión Superficial
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA