Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 291(16): 8414-27, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26907692

RESUMEN

The Hermansky Pudlak syndromes (HPS) constitute a family of disorders characterized by oculocutaneous albinism and bleeding diathesis, often associated with lethal lung fibrosis. HPS results from mutations in genes of membrane trafficking complexes that facilitate delivery of cargo to lysosome-related organelles. Among the affected lysosome-related organelles are lamellar bodies (LB) within alveolar type 2 cells (AT2) in which surfactant components are assembled, modified, and stored. AT2 from HPS patients and mouse models of HPS exhibit enlarged LB with increased phospholipid content, but the mechanism underlying these defects is unknown. We now show that AT2 in the pearl mouse model of HPS type 2 lacking the adaptor protein 3 complex (AP-3) fails to accumulate the soluble enzyme peroxiredoxin 6 (PRDX6) in LB. This defect reflects impaired AP-3-dependent trafficking of PRDX6 to LB, because pearl mouse AT2 cells harbor a normal total PRDX6 content. AP-3-dependent targeting of PRDX6 to LB requires the transmembrane protein LIMP-2/SCARB2, a known AP-3-dependent cargo protein that functions as a carrier for lysosomal proteins in other cell types. Depletion of LB PRDX6 in AP-3- or LIMP-2/SCARB2-deficient mice correlates with phospholipid accumulation in lamellar bodies and with defective intraluminal degradation of LB disaturated phosphatidylcholine. Furthermore, AP-3-dependent LB targeting is facilitated by protein/protein interaction between LIMP-2/SCARB2 and PRDX6 in vitro and in vivo Our data provide the first evidence for an AP-3-dependent cargo protein required for the maturation of LB in AT2 and suggest that the loss of PRDX6 activity contributes to the pathogenic changes in LB phospholipid homeostasis found HPS2 patients.


Asunto(s)
Complejo 3 de Proteína Adaptadora/metabolismo , Antígenos CD36/metabolismo , Síndrome de Hermanski-Pudlak/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Peroxiredoxina VI/metabolismo , Fosfatidilcolinas/metabolismo , Alveolos Pulmonares/metabolismo , Complejo 3 de Proteína Adaptadora/genética , Animales , Antígenos CD36/genética , Femenino , Síndrome de Hermanski-Pudlak/genética , Síndrome de Hermanski-Pudlak/patología , Proteínas de Membrana de los Lisosomas/genética , Masculino , Ratones , Peroxiredoxina VI/genética , Fosfatidilcolinas/genética , Alveolos Pulmonares/patología
2.
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.

3.
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
4.
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
5.
Cell Rep ; 7(2): 412-423, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24726356

RESUMEN

Spontaneous pneumothoraces due to lung cyst rupture afflict patients with the rare disease Birt-Hogg-Dubé (BHD) syndrome, which is caused by mutations of the tumor suppressor gene folliculin (FLCN). The underlying mechanism of the lung manifestations in BHD is unclear. We show that BHD lungs exhibit increased alveolar epithelial cell apoptosis and that Flcn deletion in mouse lung epithelium leads to cell apoptosis, alveolar enlargement, and an impairment of both epithelial barrier and overall lung function. We find that Flcn-null epithelial cell apoptosis is the result of impaired AMPK activation and increased cleaved caspase-3. AMPK activator LKB1 and E-cadherin are downregulated by Flcn loss and restored by its expression. Correspondingly, Flcn-null cell survival is rescued by the AMPK activator AICAR or constitutively active AMPK. AICAR also improves lung condition of Flcn(f/f):SP-C-Cre mice. Our data suggest that lung cysts in BHD may result from an underlying defect in alveolar epithelial cell survival, attributable to FLCN regulation of the E-cadherin-LKB1-AMPK axis.


Asunto(s)
Apoptosis , Síndrome de Birt-Hogg-Dubé/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Alveolos Pulmonares/metabolismo , Mucosa Respiratoria/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Síndrome de Birt-Hogg-Dubé/patología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Eliminación de Gen , Humanos , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Alveolos Pulmonares/patología , Ratas , Mucosa Respiratoria/patología , Proteínas Supresoras de Tumor/genética
6.
Autophagy ; 10(1): 45-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24275123

RESUMEN

Yeast Atg1 initiates autophagy in response to nutrient limitation. The Ulk gene family encompasses the mammalian orthologs of yeast ATG1. We created mice deficient for both Ulk1 and Ulk2 and found that the mice die within 24 h of birth. When found alive, pups exhibited signs of respiratory distress. Histological sections of lungs of the Ulk1/2 DKO pups showed reduced airspaces with thickened septae. A similar defect was seen in Atg5-deficient pups as both Ulk1/2 DKO and Atg5 KO lungs show numerous glycogen-laden alveolar type II cells by electron microscopy, PAS staining, and increased levels of glycogen in lung homogenates. No abnormalities were noted in expression of genes encoding surfactant proteins but the ability to incorporate exogenous choline into phosphatidylcholine, the major phospholipid component of surfactant, was increased in comparison to controls. Despite this, there was a trend for total phospholipid levels in lung tissue to be lower in Ulk1/2 DKO and Atg5 KO compared with controls. Autophagy was abundant in lung epithelial cells from wild-type mice, but lacking in Atg5 KO and Ulk1/2 DKO mice at P1. Analysis of the autophagy signaling pathway showed the existence of a negative feedback loop between the ULK1 and 2 and MTORC1 and 2, in lung tissue. In the absence of autophagy, alveolar epithelial cells are unable to mobilize internal glycogen stores independently of surfactant maturation. Together, the data suggested that autophagy plays a vital role in lung structural maturation in support of perinatal adaptation to air breathing.


Asunto(s)
Autofagia , Pulmón/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/ultraestructura , Animales , Animales Recién Nacidos , Proteína 5 Relacionada con la Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia , Glucógeno/metabolismo , Inmunohistoquímica , Pulmón/embriología , Pulmón/ultraestructura , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfolípidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Coloración y Etiquetado
7.
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
8.
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
9.
Nitric Oxide ; 25(3): 366-72, 2011 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-21871968

RESUMEN

Nitric oxide and secondary oxides of nitrogen react with unsaturated fatty acids such as linoleic acid to yield oxidized and nitrated products. Fatty acid nitroalkene derivatives, (e.g. nitrolinoleate [LNO(2)]) are produced by oxidative inflammatory reactions, detected clinically, display potent electrophilic reactivity and induce post-translational protein modifications that mediate adaptive inflammatory signaling responses. LNO(2) signaling was examined in lung epithelial cells because the alveolar compartment is a rich site for the transduction of redox and inflammatory reactions. LNO(2) did not directly induce Ca(2+) influx in cultured lung epithelial cells, but inhibited bradykinin-induced Ca(2+) influx in a cGMP-independent manner. In contrast, LNO(2) activated MAP kinase (Erk1/2) by a mechanism independent of bradykinin. It was hypothesized that these unique responses were transduced by activation of different protein kinase C isotypes, supported by the observation that LNO(2)-mediated inhibition of Ca(2+) influx was blocked by the non-selective PKC inhibitors chelerythine chloride and calphostin C, but not by the calcium dependent "classic" PKC inhibitor Gö6976. Western blot analysis showed that atypical PKCζ was activated by LNO(2) stimulation, with PKCζ and Erk activation also demonstrated in primary culture of human lung type II cells. Addition of pseudotypical PKCζ substrate peptide reversed LNO(2)-mediated induction of Ca(2+) influx and MAP kinase activation. Finally, the electrophilic nature of LNO(2) resulted in a novel mode of PKCζ activation, covalent adduction of the enzyme. In summary, LNO(2) mediated signaling in lung type II epithelial cells occurs via a unique pathway involving PKCζ.


Asunto(s)
Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Pulmón/citología , Proteína Quinasa C/metabolismo , Transducción de Señal , Alquenos/metabolismo , Humanos , Células Tumorales Cultivadas
10.
Traffic ; 12(9): 1196-210, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21707890

RESUMEN

Interstitial lung disease in both children and adults has been linked to mutations in the lung-specific surfactant protein C (SFTPC) gene. Among these, the missense mutation [isoleucine to threonine at codon 73 = human surfactant protein C (hSP-C(I73T) )] accounts for ∼30% of all described SFTPC mutations. We reported previously that unlike the BRICHOS misfolding SFTPC mutants, expression of hSP-C(I73T) induces lung remodeling and alveolar lipoproteinosis without a substantial Endoplasmic Reticulum (ER) stress response or ER-mediated intrinsic apoptosis. We show here that, in contrast to its wild-type counterpart that is directly routed to lysosomal-like organelles for processing, SP-C(I73T) is misdirected to the plasma membrane and subsequently internalized to the endocytic pathway via early endosomes, leading to the accumulation of abnormally processed proSP-C isoforms. Functionally, cells expressing hSP-C(I73T) demonstrated both impaired uptake and degradation of surfactant phospholipid, thus providing a molecular mechanism for the observed lipid accumulation in patients expressing hSP-C(I73T) through the disruption of normal phospholipid recycling. Our data provide evidence for a novel cellular mechanism for conformational protein-associated diseases and suggest a paradigm for mistargeted proteins involved in the disruption of the endosomal/lysosomal sorting machinery.


Asunto(s)
Endosomas/metabolismo , Enfermedades Pulmonares Intersticiales/genética , Mutación , Fosfolípidos/metabolismo , Proteína C Asociada a Surfactante Pulmonar/genética , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Adulto , Membrana Celular/metabolismo , Células Cultivadas , Niño , Endocitosis/fisiología , Retículo Endoplásmico/metabolismo , Endosomas/química , Humanos , Enfermedades Pulmonares Intersticiales/patología , Enfermedades Pulmonares Intersticiales/fisiopatología , Lisosomas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transferrina/metabolismo , Vesículas Transportadoras/metabolismo
11.
Am J Respir Crit Care Med ; 184(4): 449-58, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21616998

RESUMEN

RATIONALE: The pulmonary phenotype of Hermansky-Pudlak syndrome (HPS) in adults includes foamy alveolar type 2 cells, inflammation, and lung remodeling, but there is no information about ontogeny or early disease mediators. OBJECTIVES: To establish the ontogeny of HPS lung disease in an animal model, examine disease mediators, and relate them to patients with HPS1. METHODS: Mice with mutations in both HPS1/pale ear and HPS2/AP3B1/pearl (EPPE mice) were studied longitudinally. Total lung homogenate, lung tissue sections, and bronchoalveolar lavage (BAL) were examined for phospholipid, collagen, histology, cell counts, chemokines, surfactant protein D (SP-D), and S-nitrosylated SP-D. Isolated alveolar epithelial cells were examined for expression of inflammatory mediators, and chemotaxis assays were used to assess their importance. Pulmonary function test results and BAL from patients with HPS1 and normal volunteers were examined for clinical correlation. MEASUREMENTS AND MAIN RESULTS: EPPE mice develop increased total lung phospholipid, followed by a macrophage-predominant pulmonary inflammation, and lung remodeling including fibrosis. BAL fluid from EPPE animals exhibited early accumulation of both SP-D and S-nitrosylated SP-D. BAL fluid from patients with HPS1 exhibited similar changes in SP-D that correlated inversely with pulmonary function. Alveolar epithelial cells demonstrated expression of both monocyte chemotactic protein (MCP)-1 and inducible nitric oxide synthase in juvenile EPPE mice. Last, BAL from EPPE mice and patients with HPS1 enhanced migration of RAW267.4 cells, which was attenuated by immunodepletion of SP-D and MCP-1. CONCLUSIONS: Inflammation is initiated from the abnormal alveolar epithelial cells in HPS, and S-nitrosylated SP-D plays a significant role in amplifying pulmonary inflammation.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome de Hermanski-Pudlak , Ratones , Neumonía/etiología , Alveolos Pulmonares/fisiopatología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Mucosa Respiratoria/fisiopatología , Envejecimiento/metabolismo , Animales , Movimiento Celular , Quimiocina CCL2/metabolismo , Factores Quimiotácticos/metabolismo , Citocinas/metabolismo , Fibrosis , Síndrome de Hermanski-Pudlak/fisiopatología , Humanos , Pulmón/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Mutantes , Compuestos Nitrosos/metabolismo , Fosfolípidos/metabolismo , Alveolos Pulmonares/patología , Índice de Severidad de la Enfermedad , Factores de Tiempo
12.
J Immunol ; 186(5): 3197-205, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21282514

RESUMEN

CXCL5, a member of the CXC family of chemokines, contributes to neutrophil recruitment during lung inflammation, but its regulation is poorly understood. Because the T cell-derived cytokine IL-17A enhances host defense by triggering production of chemokines, particularly in combination with TNF-α, we hypothesized that IL-17A would enhance TNF-α-induced expression of CXCL5. Intratracheal coadministration of IL-17A and TNF-α in mice induced production of CXCL1, CXCL2, and CXCL5, which was associated with increased neutrophil influx in the lung at 8 and 24 h. The synergistic effects of TNF-α and IL17A were greatly attenuated in Cxcl5(-/-) mice at 24 h, but not 8 h, after exposure, a time when CXCL5 expression was at its peak in wild-type mice. Bone marrow chimeras produced using Cxcl5(-/-) donors and recipients demonstrated that lung-resident cells were the source of CXCL5. Using differentiated alveolar epithelial type II (ATII) cells derived from human fetal lung, we found that IL-17A enhanced TNF-α-induced CXCL5 transcription and stabilized TNF-α-induced CXCL5 transcripts. Whereas expression of CXCL5 required activation of NF-κB, IL-17A did not increase TNF-α-induced NF-κB activation. Apical costimulation of IL-17A and TNF-α provoked apical secretion of CXCL5 by human ATII cells in a transwell system, whereas basolateral costimulation led to both apical and basolateral secretion of CXCL5. The observation that human ATII cells secrete CXCL5 in a polarized fashion may represent a mechanism to recruit neutrophils in host defense in a fashion that discriminates the site of initial injury.


Asunto(s)
Quimiocina CXCL5/biosíntesis , Interleucina-17/fisiología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Células Cultivadas , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Quimiocina CXCL5/deficiencia , Quimiocina CXCL5/metabolismo , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-17/administración & dosificación , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Alveolos Pulmonares/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/biosíntesis
13.
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.

14.
J Histochem Cytochem ; 58(10): 891-901, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20566753

RESUMEN

The pulmonary alveolar epithelium is composed of two morphologically distinct cell types, type I (TI) and type II (TII) cells. Alveolar TII cells synthesize, secrete, and recycle surfactant components; contain ion transporters; and secrete immune effector molecules. In response to alveolar injury, TII cells have the capacity to act as progenitor cells, proliferating and transdifferentiating into TI cells. Although various proteins are associated with TII cells, a plasma membrane marker specific to human TII cells that would be useful for identification in tissue and for isolating this cell type has not been described previously. We devised a strategy to produce a monoclonal antibody (MAb) specific to the apical surface of human TII cells and developed an MAb that appears to be specific for human TII cells. The antibody recognizes a 280- to 300-kDa protein, HTII-280, which has the biochemical characteristics of an integral membrane protein. HTII-280 is detected by week 11 of gestation and is developmentally regulated. HTII-280 is useful for isolating human TII cells with purities and viabilities >95%. HTII-280 is likely to be a useful morphological and biochemical marker of human TII cells that may help to advance our understanding of various lung pathological conditions, including the origin and development of various lung tumors.


Asunto(s)
Proteínas de la Membrana/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/metabolismo , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Separación Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Lactante , Focalización Isoeléctrica , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Alveolos Pulmonares/embriología , Alveolos Pulmonares/crecimiento & desarrollo , Mucosa Respiratoria/metabolismo , Solubilidad
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA