Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Pediatr Pulmonol ; 58(8): 2212-2218, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37171089

RESUMEN

This review outlines some of the major contributions to Neonatal Pulmonology published in 2022 in Pediatric Pulmonology in the areas of lung ultrasound, prevention and treatment of bronchopulmonary dysplasia, and pulmonary function outcomes of neonatal lung disease.


Asunto(s)
Displasia Broncopulmonar , Neumología , Recién Nacido , Niño , Humanos , Displasia Broncopulmonar/terapia , Displasia Broncopulmonar/prevención & control , Pulmón/diagnóstico por imagen
2.
Pediatr Pulmonol ; 56(12): 3577-3579, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34379366

RESUMEN

Pediatric Pulmonology publishes original research, reviews, and case reports related to a wide range of children's respiratory disorders. This review summarizes the past year's publications in the topic area of neonatal pulmonology, in the context of selected literature from other journals relevant to the discipline.


Asunto(s)
Neumología , Enfermedades Respiratorias , Niño , Humanos , Recién Nacido
4.
Pediatr Pulmonol ; 54(6): 765-769, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30809976

RESUMEN

Pediatric Pulmonology publishes original research, reviews and case reports related to a wide range of children's respiratory disorders. This review (Part 2 of a 4-part series) summarizes the past year's publications in the topic area of neonatal lung diseases, in the context of selected literature from other journals relevant to the discipline.


Asunto(s)
Enfermedades del Prematuro , Enfermedades Respiratorias , Biomarcadores , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/fisiopatología , Enfermedades del Prematuro/prevención & control , Neumología , Enfermedades Respiratorias/fisiopatología , Enfermedades Respiratorias/prevención & control
5.
Pediatr Pulmonol ; 53(8): 1147-1151, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29737005

RESUMEN

The articles published in 2017 in topic areas relevant to neonatal pulmonology are reviewed in Part 2 of the Year-in-Review.


Asunto(s)
Neonatología , Neumología , Displasia Broncopulmonar , Humanos , Recién Nacido , Pulmón/embriología
6.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L208-L216, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27913427

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic lung injury characterized by impaired alveologenesis that may persist into adulthood. Rat models of BPD using varying degrees of hyperoxia to produce injury either cause early mortality or spontaneously recover following removal of the inciting stimulus, thus limiting clinical relevance. We sought to refine an established rat model induced by exposure to 60% O2 from birth by following hyperoxia with intermittent hypoxia (IH). Rats exposed from birth to air or 60% O2 until day 14 were recovered in air with or without IH (FIO2 = 0.10 for 10 min every 6 h) until day 28 Animals exposed to 60% O2 and recovered in air had no evidence of abnormal lung morphology on day 28 or at 10-12 wk. In contrast, 60% O2-exposed animals recovered in IH had persistently increased mean chord length, more dysmorphic septal crests, and fewer peripheral arteries. Recovery in IH also increased pulmonary vascular resistance, Fulton index, and arterial wall thickness. IH-mediated abnormalities in lung structure (but not pulmonary hypertension) persisted when reexamined at 10-12 wk, accompanied by increased pulmonary vascular reactivity and decreased exercise tolerance. Increased mean chord length secondary to IH was prevented by treatment with a peroxynitrite decomposition catalyst [5,10,15,20-Tetrakis(4-sulfonatophenyl)-21H,23H-porphyrin iron (III) chloride, 30 mg/kg/day, days 14-28], an effect accompanied by fewer inflammatory cells. We conclude that IH during recovery from hyperoxia-induced injury prevents recovery of alveologenesis and leads to changes in lung and pulmonary vascular function lasting into adulthood, thus more closely mimicking contemporary BPD.


Asunto(s)
Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/patología , Hiperoxia/complicaciones , Hipoxia/complicaciones , Lesión Pulmonar/complicaciones , Alveolos Pulmonares/crecimiento & desarrollo , Alveolos Pulmonares/patología , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Catálisis , Modelos Animales de Enfermedad , Femenino , Hiperoxia/patología , Hipertensión Pulmonar/complicaciones , Hipoxia/patología , Lesión Pulmonar/patología , Masculino , Metaloporfirinas/farmacología , Ácido Peroxinitroso/metabolismo , Condicionamiento Físico Animal , Neumonía/complicaciones , Ratas Sprague-Dawley
7.
Pediatr Pulmonol ; 51(10): 1048-1056, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26918397

RESUMEN

Bronchopulmonary dysplasia (BPD) is an inflammatory lung disorder common in premature infants who undergo mechanical ventilation with supplemental oxygen. Inhaled nitric oxide (iNO) has been used to prevent experimental and clinical BPD. Earlier studies showed that NO effects in alveolar epithelial cells (AEC) are mediated by S-nitrosothiol uptake via L-type amino acid transporter-1 (LAT1). Because LAT1 expression could influence the efficacy of iNO therapy, we sought to determine whether pulmonary LAT1 expression is altered in preterm baboons with experimental BPD and in human newborns susceptible to developing BPD. Using fixed lung obtained from 125 d to 140 d gestation baboon models of BPD, LAT1 immunostaining was measured in control and BPD animals. In adult controls and in 140 d gestational controls (GC), LAT1 was expressed in both type I and type II AECs. In 140 d BPD lungs, LAT1 expression density in alveolar tissue was decreased. In 125 d GC baboons, LAT1 immunostaining was largely confined to cuboidal AECs, whereas animals given 14 d of mechanical ventilation exhibited diminished alveolar septal LAT1 Labeling. The pattern in adult human donor lung was comparable to that observed in adult baboons. LAT1 was expressed in lungs obtained from some but not all very premature newborns at autopsy. In human and baboon lung, adult and newborn, pulmonary vascular cells expressed LAT1. In summary, LAT1 is expressed in AECs and pulmonary vascular cells in baboons and humans. Experimental BPD in premature baboons decreases pulmonary LAT1 expression and alters its spatial localization. Heterogeneity of functional LAT1 could affect S-nitrosothiol importation, which could impair iNO therapy. Pediatr Pulmonol. 2016;51:1048-1056. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Pulmón/metabolismo , Adulto , Animales , Humanos , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Papio , Respiración Artificial
8.
Bioorg Med Chem Lett ; 25(18): 3793-7, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26248802

RESUMEN

The chemokine receptors CXCR1 and CXCR2 are important pharmaceutical targets due to their key roles in inflammatory diseases and cancer progression. We have previously identified 2-[5-(4-fluoro-phenylcarbamoyl)-pyridin-2-ylsulfanylmethyl]-phenylboronic acid (SX-517) and 6-(2-boronic acid-5-trifluoromethoxy-benzylsulfanyl)-N-(4-fluoro-phenyl)-nicotinamide (SX-576) as potent non-competitive boronic acid-containing CXCR1/2 antagonists. Herein we report the synthesis and evaluation of aminopyridine and aminopyrimidine analogs of SX-517 and SX-576, identifying (2-{(benzyl)[(5-boronic acid-2-pyridyl)methyl]amino}-5-pyrimidinyl)(4-fluorophenylamino)formaldehyde as a potent chemokine antagonist with improved aqueous solubility and oral bioavailability.


Asunto(s)
Ácidos Borónicos/farmacología , Niacinamida/análogos & derivados , Receptores de Interleucina-8A/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Administración Oral , Disponibilidad Biológica , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Niacinamida/administración & dosificación , Niacinamida/química , Niacinamida/farmacología , Solubilidad , Relación Estructura-Actividad , Agua/química
9.
Antioxid Redox Signal ; 21(13): 1823-36, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25089378

RESUMEN

AIMS: The aims of this study were to determine hyperoxia effects on S-nitrosothiol (SNO) accumulation and L-type amino acid transporter 1 (LAT1) expression/function in alveolar epithelium and to determine whether hyperoxia impairs exogenous nitric oxide (NO) treatment effects in alveolar epithelium through effects on LAT1 expression and/or function. RESULTS: SNO accumulation in vitro and in vivo after NO treatment was dependent on the LAT1 system transport. Hyperoxia (60% or 90%) impaired NO effects on SNO accumulation and soluble guanylyl cyclase activation in proportion to the magnitude of hyperoxia and the duration of exposure, up to 12 h, in type I-like (R3/1) and type II-like (L2) rat and human (A549) alveolar epithelial cells. LAT function, determined by sodium-independent (3)H-leucine uptake, was impaired in a parallel manner. Hyperoxia impaired LAT1 expression in alveolar epithelial cells, determined by immunoblots and immunofluorescence, and in newborn rats exposed to 60% O2 for 4 days, determined by immunohistochemistry. INNOVATION: Despite significant preclinical evidence, inhaled NO has shown disappointing limitations in clinical applications. Our studies suggest an important explanation: oxidative stress, a common feature of diseases in which therapeutic NO would be considered, impairs LAT1 expression and function, blocking a major route for inhaled NO (iNO) action, that is, the uptake of S-nitrosocysteine via LAT1. CONCLUSIONS: SNO uptake after NO treatment is dependent on LAT1. Hyperoxia impairs SNO uptake and NO effects during NO exposure and impairs LAT system function and LAT1 expression. Effects on SNO formation and transport must be considered for rational optimization of NO-based therapeutics.


Asunto(s)
Células Epiteliales/metabolismo , Hiperoxia/patología , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Óxido Nítrico/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Transporte Biológico/fisiología , Línea Celular Tumoral , Cisteína/análogos & derivados , Cisteína/metabolismo , Células Epiteliales/patología , Guanilato Ciclasa/metabolismo , Humanos , Hiperoxia/metabolismo , Leucina/metabolismo , Masculino , Estrés Oxidativo/fisiología , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo , S-Nitrosotioles/metabolismo , Guanilil Ciclasa Soluble
10.
J Expo Sci Environ Epidemiol ; 23(1): 32-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22805991

RESUMEN

Adverse birth outcomes are associated with exposure to air pollution during pregnancy. Road proximity is a simple, widely available metric for capturing local variation in exposure to traffic-related air pollution. We characterized maternal exposure to traffic-related air pollution during pregnancy using residential proximity to major roadways among 2004-2008 singleton births in NC. Controlling for maternal race, age, education, nativity, marital status, and tobacco use, and season of birth, parity, infant sex, and Census tract-level urbanization and income, we evaluated the association between road proximity and pregnancy outcomes using generalized linear mixed models with a random intercept for each Census tract. Birth weight, birth weight percentile for gestational age, gestational hypertension, and small-for-gestational age were not associated with road proximity; however, women residing within 250 m of a major roadway were at 3-5% increased odds of low birth weight, preterm birth, and late preterm birth compared with women residing beyond 250 m (P<0.05). Our analyses demonstrate an association between proximity to major roadways and pregnancy outcomes using a large sample. Road proximity may represent a relatively straightforward method for assessing maternal risk from exposure to traffic-related air pollution, with results offering guidance for studies that can more accurately characterize air pollution exposures.


Asunto(s)
Exposición a Riesgos Ambientales , Resultado del Embarazo , Trasplante , Femenino , Humanos , North Carolina , Embarazo
11.
Am J Respir Cell Mol Biol ; 48(2): 230-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23239496

RESUMEN

The pharmacological effects of nitric oxide (NO) administered as a gas are dependent on the conversion to S-nitrosocysteine, and as such are largely mediated by the L-type amino-acid transporters (LATs) in several cell types. The dipeptide transporter PEPT2 has been proposed as a second route for S-nitrosothiol (SNO) transport, but this has never been demonstrated. Because NO governs important immune functions in alveolar macrophages, we exposed rat alveolar macrophages (primary and NR8383 cells) to NO gas at the air-liquid interface ± LPS stimulation in the presence of PEPT2 substrate Cys-Gly (or the LAT substrate L-Cys) ± transporter competitors. We found that SNO uptake and NO-dependent actions, such as the activation of soluble guanylyl cyclase (sGC), the augmentation of sGC-dependent filamentous actin (F-actin) polymerization, phagocytosis, and the inhibition of NF-κB activation, were significantly augmented by the addition of Cys-Gly in a manner dependent on PEPT2 transport. We found parallel (and greater) effects that were dependent on LAT transport. The contribution of cystine/cysteine shuttling via system x cystine transporter (xCT) to SNO uptake was relatively minor. The observed effects were unaffected by NO synthase inhibition. The NO gas treatment of alveolar macrophages increased SNO uptake, the activation of sGC, F-actin polymerization, and phagocytosis, and inhibited NF-κB activation, in a manner dependent on SNO transport via PEPT2, as well as via LAT.


Asunto(s)
Macrófagos Alveolares/efectos de los fármacos , Óxido Nítrico/farmacología , S-Nitrosotioles/metabolismo , Simportadores/fisiología , Animales , Transporte Biológico , Células Cultivadas , Macrófagos Alveolares/metabolismo , Ratas
12.
PLoS One ; 7(7): e40789, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815821

RESUMEN

Acute exacerbations of pulmonary fibrosis are characterized by rapid decrements in lung function. Environmental factors that may contribute to acute exacerbations remain poorly understood. We have previously demonstrated that exposure to inhaled lipopolysaccharide (LPS) induces expression of genes associated with fibrosis. To address whether exposure to LPS could exacerbate fibrosis, we exposed male C57BL/6 mice to crystalline silica, or vehicle, followed 28 days later by LPS or saline inhalation. We observed that mice receiving both silica and LPS had significantly more total inflammatory cells, more whole lung lavage MCP-1, MIP-2, KC and IL-1ß, more evidence of oxidative stress and more total lung hydroxyproline than mice receiving either LPS alone, or silica alone. Blocking oxidative stress with N-acetylcysteine attenuated whole lung inflammation but had no effect on total lung hydroxyproline. These observations suggest that exposure to innate immune stimuli, such as LPS in the environment, may exacerbate stable pulmonary fibrosis via mechanisms that are independent of inflammation and oxidative stress.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Acetilcisteína/farmacología , Administración por Inhalación , Animales , Lavado Broncoalveolar , Citocinas/metabolismo , Agua Potable , Hidroxiprolina/metabolismo , Inflamación/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Carbonilación Proteica/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio
13.
Am J Respir Cell Mol Biol ; 47(1): 37-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22323364

RESUMEN

Inhaled nitric oxide (iNO) is used to treat pulmonary hypertension and is being investigated for prevention of bronchopulmonary dysplasia in neonates. Extrapulmonary effects of iNO are widely recognized, but the underlying chemistry and pharmacology are poorly understood. Growing evidence suggests that, in addition to acting via diffusion, NO can be converted into nitrosants capable of reacting with endogenous L-cysteine (L-Cys) in the alveolar lining fluid, forming S-nitrosothiol (SNO)-L-cysteine (CSNO). CSNO can then enter cells via the type L amino acid transporter (LAT). To determine the influence of LAT and supplemental L-Cys on the functional activity of iNO and transpulmonary movement of SNOs or other related species, we exposed C57Bl6 mice to nebulized L-Cys or D-cysteine (D-Cys) and/or LAT competitors. Isolated lungs were then perfused with physiologic buffer while effluent was collected to assay perfusate SNOs. Nebulized L-Cys, but not D-Cys, augmented the iNO-induced increase in circulating SNOs in the effluent without altering iNO-induced pulmonary vasodilation. Addition to the perfusate of either L-leucine (L-Leu) or 2-amino-2-norborane carboxylic acid, two distinct LAT competitors, inhibited appearance in the perfusate of SNOs in L-Cys-exposed lungs; a higher concentration of L-Leu significantly inhibited the iNO-induced pulmonary vasodilation as well as SNO accumulation. We conclude that iNO-induced pulmonary vasodilation and the transpulmonary movement of iNO-derived SNOs are mediated in part by formation of extracellular CSNO, uptake by alveolar epithelial LAT, and/or export by LAT from the pulmonary endothelium into the circulation. Therapies that exploit and optimize LAT-dependent SNO transport might improve the efficacy of and clinical outcomes with NO-based therapy by improving systemic SNO delivery.


Asunto(s)
Pulmón/irrigación sanguínea , Pulmón/metabolismo , Óxido Nítrico/farmacología , S-Nitrosotioles/metabolismo , Vasodilatación , Administración por Inhalación , Aminoácidos Cíclicos/farmacología , Animales , Transporte Biológico Activo , Cisteína/administración & dosificación , Cisteína/farmacología , Femenino , Hipertensión Pulmonar/tratamiento farmacológico , Leucina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/administración & dosificación , Óxido Nítrico/uso terapéutico , Vasodilatación/efectos de los fármacos
14.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L327-33, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21724860

RESUMEN

Inducible nitric oxide synthase (NOS2) expression is increased in the airway epithelium in acute inflammatory disorders although the physiological impact remains unclear. We have previously shown that NOS2 inhibits NF-κB (p50-p65) activation in respiratory epithelial cells by inducing S-nitrosylation of the p65 monomer (SNO-p65). In addition, we have demonstrated that mouse lung SNO-p65 levels are acutely depleted in a lipopolysaccharide (LPS) model of lung injury and that augmenting SNO-p65 levels before LPS treatment results in decreased airway epithelial NF-κB activation, airway inflammation, and lung injury. We now show that aerosolized LPS induces NOS2 expression in the respiratory epithelium concomitant with an increase in lung SNO-p65 levels and a decrease in airway NF-κB activity. Genetic deletion of NOS2 results in an absence of SNO-p65 formation, persistent NF-κB activity in the respiratory epithelium, and prolonged airway inflammation. These results indicate that a primary function of LPS-induced NOS2 expression in the respiratory epithelium is to modulate the inflammatory response through deactivation of NF-κB via S-nitrosylation of p65, thereby counteracting the initial stimulus-coupled denitrosylation.


Asunto(s)
Óxido Nítrico Sintasa de Tipo II/fisiología , Mucosa Respiratoria/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Inflamación/inducido químicamente , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis
15.
Exp Lung Res ; 37(1): 10-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21077778

RESUMEN

The objective of this study was to determine whether overexpression of human extracellular superoxide dismutase (hEC-SOD) can preserve nitric oxide (NO) bioavailability. In vitro studies examined the transient expression of hEC-SOD in mouse epithelial (C10) cells and its effect on extracellular accumulation of NO, intracellular cyclic guanosine monophosphate (cGMP), and nuclear factor kappa B (NF-κB) activation under normal and oxidative stress conditions. In vivo, newborn rabbits were treated with a plasmid containing hEC-SOD cDNA or vehicle plasmid alone, followed by exposure to hyperoxia (Fio2 = 95% for 7 days). A third group was raised under normoxic conditions. cGMP and NF-κB activation were studied. There was significantly higher NO accumulation in cells expressing hEC-SOD exposed to oxidative stress compared with nontransfected cells. Accumulation of cGMP was significantly higher in cells expressing hEC-SOD. Oxidative stress induced NF-κB activation, which was abrogated by hEC-SOD expression. In vivo, there was significantly higher cGMP accumulation in transfected neonatal rabbit lung tissue at 3 and 7 days of hyperoxic exposure. Immunostaining for NF-κB, showed a marked increase in NF-κB concentration in nontreated neonatal rabbit lung tissue compared to transfected neonatal lung with hEC-SOD and the control air group. These results show that transient EC-SOD overexpression maintains NO bioavailability, which directly leads to maintenance of cGMP activity and reduction of NF-κB activation under oxidative stress.


Asunto(s)
Células Epiteliales/enzimología , Hiperoxia/enzimología , Lesión Pulmonar/prevención & control , Pulmón/enzimología , Óxido Nítrico/metabolismo , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Animales , Animales Recién Nacidos , Disponibilidad Biológica , Línea Celular , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Hiperoxia/complicaciones , Hiperoxia/genética , Hiperoxia/patología , Pulmón/patología , Lesión Pulmonar/enzimología , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , FN-kappa B/metabolismo , Conejos , Superóxido Dismutasa/genética , Factores de Tiempo , Transfección , Regulación hacia Arriba
16.
Free Radic Biol Med ; 49(2): 294-300, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20423728

RESUMEN

The pathway by which inhaled NO gas enters pulmonary alveolar epithelial cells has not been directly tested. Although the expected mechanism is diffusion, another route is the formation of S-nitroso-L-cysteine, which then enters the cell through the L-type amino acid transporter (LAT). To determine if NO gas also enters alveolar epithelium this way, we exposed alveolar epithelial-rat type I, type II, L2, R3/1, and human A549-cells to NO gas at the air liquid interface in the presence of L- and D-cysteine+/-LAT competitors. NO gas exposure concentration dependently increased intracellular NO and S-nitrosothiol levels in the presence of L- but not D-cysteine, which was inhibited by LAT competitors, and was inversely proportional to diffusion distance. The effect of L-cysteine on NO uptake was also concentration dependent. Without preincubation with L-cysteine, NO uptake was significantly reduced. We found similar effects using ethyl nitrite gas in place of NO. Exposure to either gas induced activation of soluble guanylyl cylase in a parallel manner, consistent with LAT dependence. We conclude that NO gas uptake by alveolar epithelium achieves NO-based signaling predominantly by forming extracellular S-nitroso-L-cysteine that is taken up through LAT, rather than by diffusion. Augmenting extracellular S-nitroso-L-cysteine formation may augment pharmacological actions of inhaled NO gas.


Asunto(s)
Sistema de Transporte de Aminoácidos y+L/metabolismo , Transporte Biológico/efectos de los fármacos , Epitelio/metabolismo , Guanilato Ciclasa/metabolismo , Proteínas de Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Sistema de Transporte de Aminoácidos y+L/antagonistas & inhibidores , Animales , Unión Competitiva , Cisteína/análogos & derivados , Cisteína/metabolismo , Cisteína/farmacología , Difusión , Epitelio/patología , Guanilato Ciclasa/genética , Humanos , Leucina/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Óxido Nítrico/farmacología , Alveolos Pulmonares/patología , Ratas , S-Nitrosotioles/metabolismo , Estereoisomerismo
17.
Am J Respir Crit Care Med ; 180(12): 1218-26, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19762564

RESUMEN

RATIONALE: Epidemiologic studies implicate air pollutant exposure during pregnancy as a risk factor for wheezing in offspring. Ozone exposure is linked to exacerbations of wheezing in children. OBJECTIVES: To determine if maternal pulmonary exposure to traffic-related particles during pregnancy augments ozone-induced airway hyperresponsiveness in offspring. METHODS: C57BL6 time-mated mice were given NIST SRM#1648 (particulate matter [PM]) 0.48 mg, saline vehicle, or no treatment by tracheal insufflation twice weekly for 3 weeks. PM exposure augmented maternal lung inflammation and placental TNF-alpha, Keratinocyte-derived cytokine (KC), and IL-6 (measured at gestation Day 18). After parturition, dams and litters were exposed to air or ozone 1 ppm 3 h/d, every other day, thrice weekly for 4 weeks. Respiratory system resistance in pups was measured at baseline and after administration of nebulized methacholine. MEASUREMENTS AND MAIN RESULTS: Ozone increased airway hyperresponsiveness, but the increase was greatest in pups born to PM-treated dams. Whole-lung TNF-alpha, IL-1beta, KC, IL-6, and MCP-1 were increased in ozone-treated pups, with the greatest increase in pups born to dams given PM. Airway epithelial mucous metaplasia estimated by periodic acid-Schiff Alcian blue staining was increased in ozone-exposed pups born to PM-treated dams. Alveolar development, determined by morphometry, and airway smooth muscle bulk, estimated using alpha-actin histochemistry, were unaffected by prenatal or postnatal treatment. CONCLUSIONS: Maternal pulmonary exposure to PM during pregnancy augments placental cytokine expression and postnatal ozone-induced pulmonary inflammatory cytokine responses and ozone-induced airway hyperresponsiveness without altering airway structure.


Asunto(s)
Hiperreactividad Bronquial/inducido químicamente , Exposición Materna , Oxidantes Fotoquímicos/toxicidad , Ozono/toxicidad , Material Particulado/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire , Análisis de Varianza , Animales , Hiperreactividad Bronquial/fisiopatología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Inflamación/fisiopatología , Exposición por Inhalación , Pulmón/fisiopatología , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , Pruebas de Función Respiratoria
18.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L134-42, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19411313

RESUMEN

Hyperoxia disrupts postnatal lung development in part through inducing inflammation. To determine the contribution of leukocyte-derived reactive oxygen species, we exposed newborn wild-type and NADPH oxidase p47(phox) subunit null (p47(phox-/-)) mice to air or acute hyperoxia (95% O(2)) for up to 11 days. Hyperoxia-induced pulmonary neutrophil influx was similar in wild-type and p47(-/-) mice at postnatal days (P) 7 and 11. Macrophages were decreased in wild-type hyperoxia-exposed mice compared with p47(phox-/-) mice at P11. Hyperoxia impaired type II alveolar epithelial cell and bronchiolar epithelial cell proliferation, but depression of type II cell proliferation was significantly less in p47(-/-) mice at P3 and P7, when inflammation was minimal. We found reciprocal results for the expression of the cell cycle inhibitor p21(cip/waf) in type II cells, which was induced in 95% O(2)-exposed wild-type mice, but significantly less in p47(phox-/-) littermates at P7. Despite partial preservation of type II cell proliferation, deletion of p47(phox) did not prevent the major adverse effects of hyperoxia on alveolar development estimated by morphometry at P11, but hyperoxia impairment of elastin deposition at alveolar septal crests was significantly worse in wild-type vs. p47(phox-/-) mice at P11. Since we found that p47(phox) is expressed in a subset of alveolar epithelial cells, its deletion may protect postnatal type II alveolar epithelial proliferation from hyperoxia through effects on epithelial as well as phagocyte-generated superoxide.


Asunto(s)
Células Epiteliales/enzimología , Células Epiteliales/patología , Hiperoxia/enzimología , Hiperoxia/patología , NADPH Oxidasas/metabolismo , Alveolos Pulmonares/enzimología , Alveolos Pulmonares/patología , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Aire , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Líquido del Lavado Bronquioalveolar , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/efectos de los fármacos , Eliminación de Gen , Ratones , Oxígeno/farmacología , Neumonía/enzimología , Neumonía/patología , Transporte de Proteínas/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/crecimiento & desarrollo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Análisis de Supervivencia , Tirosina/análogos & derivados , Tirosina/metabolismo
19.
Pediatr Res ; 66(2): 121-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19390491

RESUMEN

Reactive oxygen species (ROS) serve as cell signaling molecules for normal biologic processes. However, the generation of ROS can also provoke damage to multiple cellular organelles and processes, which can ultimately disrupt normal physiology. An imbalance between the production of ROS and the antioxidant defenses that protect cells has been implicated in the pathogenesis of a variety of diseases, such as cancer, asthma, pulmonary hypertension, and retinopathy. The nature of the injury will ultimately depend on specific molecular interactions, cellular locations, and timing of the insult. This review will outline the origins of endogenous and exogenously generated ROS. The molecular, cellular, pathologic, and physiologic targets will then be discussed with a particular emphasis on aspects relevant to child development. Finally, antioxidant defenses that scavenge ROS and mitigate associated toxicities will be presented, with a discussion of potential therapeutic approaches for the prevention and/or treatment of human diseases using enzymatic and nonenzymatic antioxidants.


Asunto(s)
Oxígeno/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Humanos , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/toxicidad , Mucosa Respiratoria/metabolismo , Transducción de Señal/fisiología , Superóxido Dismutasa/metabolismo
20.
Am J Respir Crit Care Med ; 180(1): 11-8, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19324975

RESUMEN

RATIONALE: S-Nitrosothiols (SNO) inhibit immune activation of the respiratory epithelium and airway SNO levels are decreased in inflammatory lung disease. Ethyl nitrite (ENO) is a gas with chemical properties favoring SNO formation. Augmentation of airway SNO by inhaled ENO treatment may decrease lung inflammation and subsequent injury by inhibiting activation of the airway epithelium. OBJECTIVES: To determine the effect of inhaled ENO on airway SNO levels and LPS-induced lung inflammation/injury. METHODS: Mice were treated overnight with inhaled ENO (10 ppm) or air, followed immediately by exposure to aerosolized LPS or saline. Parameters of inflammation and lung injury were quantified 1 hour after completion of the aerosol exposure and correlated to lung airway and tissue SNO levels. MEASUREMENTS AND MAIN RESULTS: Aerosolized LPS induced a decrease in airway and lung tissue SNO levels including S-nitrosylated NF-kappaB. The decrease in lung SNO was associated with an increase in lung NF-kappaB activity, cytokine/chemokine expression (keratinocyte-derived chemokine, tumor necrosis factor-alpha, and IL-6), airway neutrophil influx, and worsened lung compliance. Pretreatment with inhaled ENO restored airway SNO levels and reduced LPS-mediated NF-kappaB activation thereby inhibiting the downstream inflammatory response and preserving lung compliance. CONCLUSIONS: Airway SNO serves an antiinflammatory role in the lung. Inhaled ENO can be used to augment airway SNO and protect from LPS-induced acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/administración & dosificación , Nitritos/administración & dosificación , Lesión Pulmonar Aguda/inmunología , Administración por Inhalación , Animales , Masculino , Ratones , S-Nitrosotioles/inmunología , S-Nitrosotioles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA