Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 13(2)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38397736

RESUMEN

We previously reported that maternal alcohol use increased the risk of sepsis in premature and term newborns. In the neonatal mouse, fetal ethanol (ETOH) exposure depleted the antioxidant glutathione (GSH), which promoted alveolar macrophage (AM) immunosuppression and respiratory syncytial virus (RSV) infections. In this study, we explored if oral liposomal GSH (LGSH) would attenuate oxidant stress and RSV infections in the ETOH-exposed mouse pups. C57BL/6 female mice were pair-fed a liquid diet with 25% of calories from ethanol or maltose-dextrin. Postnatal day 10 pups were randomized to intranasal saline, LGSH, and RSV. After 48 h, we assessed oxidant stress, AM immunosuppression, pulmonary RSV burden, and acute lung injury. Fetal ETOH exposure increased oxidant stress threefold, lung RSV burden twofold and acute lung injury threefold. AMs were immunosuppressed with decreased RSV clearance. However, LGSH treatments of the ETOH group normalized oxidant stress, AM immune phenotype, the RSV burden, and acute lung injury. These studies suggest that the oxidant stress caused by fetal ETOH exposure impaired AM clearance of infectious agents, thereby increasing the viral infection and acute lung injury. LGSH treatments reversed the oxidative stress and restored AM immune functions, which decreased the RSV infection and subsequent acute lung injury.

2.
Alcohol ; 80: 5-16, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30580016

RESUMEN

BACKGROUND: We previously reported that maternal alcohol use significantly increases the risk of sepsis in premature and term newborns. In the mouse, fetal ethanol exposure results in an immunosuppressed phenotype for the alveolar macrophage (AM) and decreases bacterial phagocytosis. In pregnant mice, ethanol decreased AM zinc homeostasis, which contributed to immunosuppression and impaired AM phagocytosis. In this study, we explored whether ethanol-induced zinc insufficiency extended to the pup AMs and contributed to immunosuppression and exacerbated viral lung infections. METHODS: C57BL/6 female mice were fed a liquid diet with 25% ethanol-derived calories or pair-fed a control diet with 25% of calories as maltose-dextrin. Some pup AMs were treated in vitro with zinc acetate before measuring zinc pools or transporter expression and bacteria phagocytosis. Some dams were fed additional zinc supplements in the ethanol or control diets, and then we assessed pup AM zinc pools, zinc transporters, and the immunosuppressant TGFß1. On postnatal day 10, some pups were given intranasal saline or respiratory syncytial virus (RSV), and then AM RSV phagocytosis and the RSV burden in the airway lining fluid were assessed. RESULTS: Fetal ethanol exposure decreased pup AM zinc pools, zinc transporter expression, and bacterial clearance, but in vitro zinc treatments reversed these alterations. In addition, the expected ethanol-induced increase in TGFß1 and immunosuppression were associated with decreased RSV phagocytosis and exacerbated RSV infections. However, additional maternal zinc supplements blocked the ethanol-induced perturbations in the pup AM zinc homeostasis and TGFß1 immunosuppression, thereby improving RSV phagocytosis and attenuating the RSV burden in the lung. CONCLUSION: These studies suggest that, despite normal maternal dietary zinc intake, in utero alcohol exposure results in zinc insufficiency, which contributes to compromised neonatal AM immune functions, thereby increasing the risk of bacterial and viral infections.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal/etiología , Macrófagos Alveolares/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/etiología , Zinc/deficiencia , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Trastornos del Espectro Alcohólico Fetal/inmunología , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Tolerancia Inmunológica , Macrófagos Alveolares/fisiología , Ratones , Ratones Endogámicos C57BL , Embarazo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/fisiopatología
3.
Physiol Rep ; 5(6)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28325787

RESUMEN

Our understanding of the intrinsic effects of cystic fibrosis (CF) transmembrane conductance regulator (cftr) deletion on resident neonatal alveolar macrophage (AM) remains limited. We previously demonstrated that diminished glutathione (GSH) or excessive AM transforming growth factor beta one (TGFß1) contributes to AM dysfunction in a variety of disease states. In this study, using a gut-corrected cftr neonatal knockout (KO) mouse model and a siRNA-manipulated macrophage-like cell line (THP-1 cell), we hypothesized (1) that cftr mutation alone increases neonatal AM oxidant stress and cellular TGFß1 signaling via altered GSH, thereby impairing cellular function, and (2) that exogenous GSH attenuates AM alterations and dysfunction in the KO AM In neonatal KO mice, the baseline bronchoalveolar lavage fluid demonstrated a near doubling in mixed disulfides (P ≤ 0.05) and oxidized GSSG (P ≤ 0.05) without concurrent inflammation compared to WT littermates. KO AM demonstrated diminished AM thiols (P ≤ 0.05), increased AM mitochondrial ROS (P ≤ 0.05), increased AM TGFß1 (P ≤ 0.05) with increased TGFß1 signaling (P ≤ 0.05), and impaired phagocytosis (P ≤ 0.05). KO AM mitochondrial ROS was modulated by exogenous GSH (P ≤ 0.05). Conversely, TGFß1 was reduced (P ≤ 0.05) and impaired phagocytosis was rescued (P ≤ 0.05) by exogenous GSH in the KO AM These results suggest that an altered neonatal AM phenotype may contribute to the initiation of lung inflammation/infection in the CF lung. Modulation of the AM in the neonatal CF lung may potentially alter progression of disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Glutatión/farmacología , Macrófagos Alveolares/metabolismo , Estrés Oxidativo/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Ratones , Ratones Endogámicos CFTR , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología
4.
Alcohol Alcohol ; 50(1): 30-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25371044

RESUMEN

AIMS: (a) Establish the minimum number of weeks of chronic ethanol ingestion needed to perturb zinc homeostasis, (b) Examine intracellular zinc status in the alveolar macrophages (AMs) when ethanol ingestion is combined with pregnancy, (c) Investigate whether in vitro zinc treatment reverses the effects of ethanol ingestion on the AM. METHODS: C57BL/6 female mice were fed a liquid diet (±25% ethanol-derived calories) during preconception and pregnancy. The control group was pair-fed to the ethanol group. In the isolated AMs, we measured intracellular AM zinc levels, zinc transporter expression, alternative activation and phagocytic index. Zinc acetate was added to some cells prior to analysis. RESULTS: Intracellular zinc levels in the AM decreased within 3 weeks of ethanol ingestion. After ethanol ingestion prior to and during pregnancy, zinc transporter expression and intracellular zinc levels were decreased in the AMs when compared with controls. Bacterial clearance was decreased because the AMs were alternatively activated. In vitro additions of zinc reversed these effects of ethanol. CONCLUSION: Ethanol ingestion prior to and during pregnancy perturbed AM zinc balance resulting in impaired bacterial clearance, but these effects were ameliorated by in vitro zinc treatments.


Asunto(s)
Etanol/efectos adversos , Macrófagos Alveolares/efectos de los fármacos , Zinc/deficiencia , Animales , Proteínas Portadoras/análisis , Proteínas Portadoras/biosíntesis , Femenino , Macrófagos Alveolares/química , Macrófagos Alveolares/fisiología , Ratones , Ratones Endogámicos C57BL , Fagocitosis/efectos de los fármacos , Embarazo , Complicaciones del Embarazo/inducido químicamente , Zinc/análisis
5.
Respir Res ; 15: 1, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24397246

RESUMEN

BACKGROUND: Analysis of exhaled breath condensates (EBC) is a non-invasive technique to evaluate biomarkers such as antioxidants in the pediatric population, but limited data exists of its use in intubated patients, particularly newborns. Currently, tracheal aspirate (TA) serves as the gold standard collection modality in critically ill newborns, but this method remains invasive. We tested the hypothesis that glutathione status would positively correlate between EBC and TA collections in intubated newborns in the Newborn Intensive Care Unit (NICU). We also hypothesized that these measurements would be associated with alveolar macrophage (AM) glutathione status in the newborn lung. METHODS: Reduced glutathione (rGSH), glutathione disulfide (GSSG), and total GSH (rGSH + (2 X GSSG)) were measured in sequential EBC and TA samples from 26 intubated newborns via high performance liquid chromatography (HPLC). Additionally, AM glutathione was evaluated via immunofluorescence. Pearson's correlation coefficient and associated 95% confidence intervals were used to quantify the associations between raw and urea-corrected concentrations in EBC and TA samples and AM staining. Statistical significance was defined as p ≤ 0.05 using two-tailed tests. The sample size was projected to allow for a correlation coefficient of 0.5, with 0.8 power and alpha of 0.05. RESULTS: EBC was obtainable from intubated newborns without adverse clinical events. EBC samples demonstrated moderate to strong positive correlations with TA samples in terms of rGSH, GSSG and total GSH. Positive correlations between the two sampling sites were observed in both raw and urea-corrected concentrations of rGSH, GSSG and total GSH. AM glutathione staining moderately correlated with GSSG and total GSH status in both the TA and EBC. CONCLUSIONS: GSH status in EBC samples of intubated newborns significantly correlated with the GSH status of the TA sample and was reflective of cellular GSH status in this cohort of neonatal patients. Non-invasive EBC sampling of intubated newborns holds promise for monitoring antioxidant status such as GSH in the premature lung. Further studies are necessary to evaluate the potential relationships between EBC biomarkers in the intubated premature newborn and respiratory morbidities.


Asunto(s)
Espiración/fisiología , Glutatión/análisis , Glutatión/metabolismo , Intubación Intratraqueal , Pulmón/química , Pulmón/metabolismo , Pruebas Respiratorias/métodos , Humanos , Recién Nacido , Intubación Intratraqueal/métodos , Proyectos Piloto
6.
Pediatr Res ; 72(2): 147-53, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22562289

RESUMEN

BACKGROUND: Maternal smoking in utero has been associated with adverse health outcomes including lower respiratory tract infections in infants and children, but the mechanisms underlying these associations continue to be investigated. We hypothesized that nicotine plays a significant role in mediating the effects of maternal tobacco smoke on the function of the neonatal alveolar macrophage (AM), the resident immune cell in the neonatal lung. METHODS: Primary AMs were isolated at postnatal day 7 from a murine model of in utero nicotine exposure. The murine AM cell line MH-S was used for additional in vitro studies. RESULTS: In utero nicotine increased interleukin-13 and transforming growth factor-ß1 (TGFß1) in the neonatal lung. Nicotine-exposed AMs demonstrated increased TGFß1 and increased markers of alternative activation with diminished phagocytic function. However, AMs from mice deficient in the α7 nicotinic acetylcholine receptor (α7 nAChR) had less TGFß1, reduced alternative activation, and improved phagocytic functioning despite similar in utero nicotine exposure. CONCLUSION: In utero nicotine exposure, mediated in part via the α7 nAChR, may increase the risk of lower respiratory tract infections in neonates by changing the resting state of AM toward alternative activation. These findings have important implications for immune responses in the nicotine-exposed neonatal lung.


Asunto(s)
Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Alveolos Pulmonares/citología , Receptores Nicotínicos/metabolismo , Fumar/efectos adversos , Animales , Western Blotting , Bungarotoxinas , Línea Celular , Femenino , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nicotina/administración & dosificación , Comunicación Paracrina/efectos de los fármacos , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Nicotínicos/genética , Receptor Nicotínico de Acetilcolina alfa 7
7.
Am J Physiol Lung Cell Mol Physiol ; 299(1): L8-16, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20382747

RESUMEN

We have previously demonstrated that fetal ethanol exposure deranges the function and viability of the neonatal alveolar macrophage. Although altered differentiation of the alveolar macrophage contributes to pulmonary disease states within the adult lung, the effects of fetal ethanol exposure on the normal differentiation of interstitial to alveolar macrophage in the newborn lung are unknown. In the current study, using a mouse model of fetal ethanol exposure, we hypothesized that altered terminal differentiation of the neonatal interstitial to alveolar macrophage contributes to the observed cellular dysfunction in the ethanol-exposed newborn mouse. Control alveolar macrophage differentiation was characterized by increased expression of CD32/CD11b (P < or = 0.05) and increased in vitro phagocytosis of Staphylococcus aureus (P < or = 0.05) compared with interstitial macrophage. After in utero ethanol exposure, both alveolar and interstitial macrophage lacked the acquisition of CD32/CD11b (P < or = 0.05) and displayed impaired in vitro phagocytosis (P < or = 0.05). Ethanol significantly increased transforming growth factor-beta(1) (TGF-beta(1)) in the bronchoalveolar lavage fluid (P < or = 0.05), as well as in both interstitial and alveolar macrophages (P < or = 0.05). Oxidant stress contributed to the ethanol-induced changes on the interstitial and alveolar cells, since maternal supplementation with the glutathione precursor S-adenosylmethionine during ethanol ingestion normalized CD32/CD11b (P < or = 0.05), phagocytosis (P < or = 0.05), and TGF-beta(1) in the bronchoalveolar lavage fluid and macrophages (P < or = 0.05). Contrary to our hypothesis, fetal ethanol exposure did not solely impair interstitial to alveolar macrophage differentiation. Rather, fetal ethanol exposure impaired both neonatal interstitial and alveolar macrophage phagocytic function and differentiation. Increased oxidant stress and elevated TGF-beta(1) contributed to the impaired differentiation of both interstitial and alveolar macrophage.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/fisiología , Efectos Tardíos de la Exposición Prenatal , Adulto , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos Alveolares/citología , Ratones , Ratones Endogámicos C57BL , Fagocitosis/fisiología , Embarazo
8.
Alcohol Clin Exp Res ; 33(2): 300-6, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19032578

RESUMEN

BACKGROUND: The effects of fetal alcohol exposure on the risks of neonatal lung injury and infection remain under investigation. The resident alveolar macrophage (AM) is the first line of immune defense against pulmonary infections. In utero ethanol (ETOH) exposure deranges the function of both premature and term guinea pig AM. We hypothesized that fetal ETOH exposure would increase the risk of pulmonary infection in vivo. METHODS: We developed a novel in vivo model of group B Streptococcus (GBS) pneumonia using our established guinea pig model of fetal ETOH exposure. Timed-pregnant guinea pigs were pair fed +/-ETOH and some were supplemented with the glutathione (GSH) precursor S-adenosyl-methionine (SAM-e). Term pups were given GBS intratracheally while some were pretreated with inhaled GSH prior to the experimental GBS. Neonatal lung and whole blood were evaluated for GBS while isolated AM were evaluated using fluorescent microscopy for GBS phagocytosis. RESULTS: Ethanol-exposed pups demonstrated increased lung infection and sepsis while AM phagocytosis of GBS was deficient compared with control. When SAM-e was added to the maternal diet containing ETOH, neonatal lung and systemic infection from GBS was attenuated and AM phagocytosis was improved. Inhaled GSH therapy prior to GBS similarly protected the ETOH-exposed pup from lung and systemic infection. CONCLUSIONS: In utero ETOH exposure impaired the neonatal lung's defense against experimental GBS, while maintaining GSH availability protected the ETOH-exposed lung. This study suggested that fetal alcohol exposure deranges the neonatal lung's defense against bacterial infection, and support further investigations into the potential therapeutic role for exogenous GSH to augment neonatal AM function.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Neumonía Bacteriana/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae , Animales , Animales Recién Nacidos , Antídotos/farmacocinética , Antídotos/farmacología , Peso Corporal/efectos de los fármacos , Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Etanol/antagonistas & inhibidores , Femenino , Glutatión/farmacocinética , Glutatión/farmacología , Cobayas , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Microscopía Confocal , Fagocitosis/efectos de los fármacos , Neumonía Bacteriana/patología , Embarazo , Pruebas de Función Respiratoria , S-Adenosilmetionina/administración & dosificación , S-Adenosilmetionina/farmacología , Sepsis/microbiología , Infecciones Estreptocócicas/patología
9.
Alcohol Clin Exp Res ; 31(2): 308-16, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17250624

RESUMEN

BACKGROUND: The effects of in utero alcohol exposure on the immune function of the newborn remain under investigation. Fetal ethanol (ETOH) exposure increases oxidative stress in the developing lung, in part due to decreased availability of the antioxidant glutathione (GSH). We have previously shown that in utero ETOH impairs alveolar macrophage phagocytosis and viability in the premature pup, while maintaining GSH availability with maternal supplementation of S-adenosyl-methionine (SAM) during ETOH ingestion improves macrophage function and viability. We hypothesized that dysfunction of the neonatal alveolar macrophage exposed to ETOH in utero would persist at term gestation. METHODS: Using a guinea-pig model of fetal ETOH exposure, timed-pregnant guinea-pigs were pair-fed ETOH+/-the GSH precursor SAM and the diet continued until spontaneous delivery. Term alveolar macrophages were evaluated using fluorescent microscopy for phagocytosis and apoptosis after in vitro incubation with Staphalococcus aureus. Using an in vivo model of intranasal Staph. aureus inoculation, the in vivo function of the term alveolar macrophage was also investigated using confocal fluorescent analysis. RESULTS: In utero ETOH exposure increased oxidant stress in the alveolar macrophage and decreased phagocytosis and viability in vitro and in vivo. Confocal analysis of phagocytosis in vivo demonstrated a marked impairment of internalization of the bacteria by the ETOH-exposed alveolar macrophage. The addition of SAM during maternal ETOH ingestion prevented loss of alveolar macrophage function and viability in vitro and in vivo. CONCLUSIONS: In utero ETOH exposure impairs alveolar macrophage function and viability in vitro and in vivo even at term gestation. The ETOH-induced changes in macrophage function and viability can be ablated with maternal SAM supplementation. Further investigations are required to identify the mechanisms of ETOH-induced derangement of phagocytosis in the neonatal alveolar macrophage and the clinical ramifications of altered immune function after in utero alcohol exposure for the newborn.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Macrófagos Alveolares/efectos de los fármacos , Exposición Materna/efectos adversos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Glutatión/metabolismo , Cobayas , Macrófagos Alveolares/citología , Macrófagos Alveolares/fisiología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Staphylococcus aureus/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 292(4): L824-32, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17122355

RESUMEN

We have previously demonstrated that chronic alcohol exposure decreases glutathione in the alveolar space. Although alcohol use is associated with decreased alveolar macrophage function, the mechanism by which alcohol impairs macrophage phagocytosis is unknown. In the current study, we examined the possibility that ethanol-induced alveolar macrophage dysfunction was secondary to decreased glutathione and subsequent chronic oxidative stress in the alveolar space. After 6 wk of ethanol ingestion, oxidant stress in the alveolar macrophages was evidenced by a 30-mV oxidation of the GSH/GSSG redox potential (P

Asunto(s)
Alcoholismo/fisiopatología , Glutatión/fisiología , Macrófagos Alveolares/fisiología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Homeostasis , Etiquetado Corte-Fin in Situ , Masculino , Microscopía Confocal , Estrés Oxidativo/fisiología , Fagocitosis/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Pediatr Res ; 57(1): 76-81, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15531743

RESUMEN

Immature function of the alveolar macrophage increases the risk of pulmonary infections in premature newborns. In utero alcohol increases fetal systemic oxidative stress. Because the premature lung is deficient in glutathione (GSH), we hypothesized that chronic in utero alcohol (ethanol) exposure exacerbates the oxidative stress within the developing lung, thereby impairing alveolar macrophage function. Additionally, we evaluated the effects of in vivo and in vitro GSH availability on ethanol-exposed macrophage function. Using a guinea pig model of chronic in utero ethanol exposure, fetal epithelial lining fluid (ELF) and alveolar macrophage GSH were decreased with increased markers of oxidative stress. Ethanol-exposed macrophage exhibited impaired phagocytosis and increased apoptosis compared with gestational control. When the GSH precursor S-adenosyl-methionine (SAM) was added to the maternal drinking water containing ethanol, fetal ELF and macrophage GSH were maintained and ELF oxidative stress diminished. In vivo maternal SAM therapy maintained macrophage phagocytosis and decreased apoptosis. In vitro GSH supplements also improved phagocytosis and viability in both premature and ethanol-exposed macrophage. This suggested that in utero ethanol impaired premature macrophage function and viability via decreased GSH availability. Furthermore, GSH supplementation during and after ethanol exposure improved fetal macrophage function and viability. These results add a new dimension to the detrimental effects of fetal alcohol exposure on the developing alveolar macrophage, raising the possibility of GSH therapy to augment premature alveolar macrophage function.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Etanol/efectos adversos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Exposición Materna , Animales , Apoptosis , Supervivencia Celular , Depresores del Sistema Nervioso Central/efectos adversos , Modelos Animales de Enfermedad , Epitelio/patología , Etanol/farmacología , Femenino , Glutatión/metabolismo , Cobayas , Etiquetado Corte-Fin in Situ , Peroxidación de Lípido , Macrófagos/metabolismo , Estrés Oxidativo , Fagocitosis , S-Adenosilmetionina/farmacología , Factores de Tiempo
12.
Alcohol ; 33(3): 191-7, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15596087

RESUMEN

Although pulmonary function is not altered, a history of alcohol abuse is an independent outcome variable in the development of acute respiratory distress syndrome. In the absence of cirrhosis, alcohol abuse decreased glutathione, the key antioxidant lining the alveolar space, by 80% and is associated with alveolar barrier leak. Neither the glutathione pool nor barrier leak was corrected by abstinence for 1 week. This aberrant glutathione homeostasis may contribute to enhanced alveolar permeability, thereby increasing susceptibility to the development of acute respiratory distress syndrome. In a rat model, chronic ingestion of ethanol decreased pulmonary glutathione concentration, increased alveolar barrier permeability, and increased the risk of acute lung injury. In alveolar type II cells, chronic ingestion of ethanol altered cellular functions such as decreased surfactant processing, decreased barrier integrity, and increased sensitivity to cytotoxin-induced apoptosis in vitro and in vivo. In alveolar macrophages, chronic ingestion of ethanol decreased phagocytosis of microorganisms and decreased cell viability, events that would increase the risk of pneumonia. A central role for glutathione availability was demonstrated by the normalization of cellular function and viability of type II cells and macrophages as well as decreased sensitivity to endotoxemia-induced acute lung injury when glutathione precursors were added to the ethanol diet. These results support the suggestion that chronic ingestion of ethanol increased the risk of acute lung injury not through ethanol per se but through the chronic oxidative stress that resulted from ethanol-induced glutathione depletion. Because chronic oxidative stress alters cellular functions and viability, the lung becomes more susceptible when a second hit such as sepsis occurs.


Asunto(s)
Alcoholismo/metabolismo , Alcoholismo/patología , Glutatión/fisiología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Alcoholismo/inmunología , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Glutatión/metabolismo , Humanos , Síndrome de Dificultad Respiratoria/inmunología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA