Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Alcohol Clin Exp Res ; 46(12): 2214-2224, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36281822

RESUMEN

BACKGROUND: Alcohol impairs pulmonary innate immune function and is associated with an increased risk of tuberculosis (TB). Toll-like receptor 2 (TLR2) is a pattern recognition receptor on alveolar macrophages that recognizes Mycobacterium tuberculosis (Mtb). The expression of TLR2 depends, in part, on granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling. Given our prior work demonstrating the suppression of GM-CSF signaling following chronic alcohol ingestion, we hypothesized that alcohol impairs TLR2 expression via the suppression of GM-CSF and thereby reduces the ability of the macrophage to recognize and phagocytose Mtb. METHODS: Primary alveolar macrophages were isolated from control-fed and alcohol-fed rats. Prior to cell isolation, some alcohol-fed rats were treated with intranasal GM-CSF and then endotracheally inoculated with an attenuated strain of Mtb. Primary macrophages were then isolated and immunofluorescence was used to determine phagocytic efficiency and TLR2 expression in the presence and absence of GM-CSF treatment and phagocytic efficiency in the presence and absence of TLR2 neutralization. RESULTS: TLR2 expression and phagocytosis of Mtb were significantly lower in the alveolar macrophages of alcohol-fed rats than control-fed rats. In parallel, blocking TLR2 signaling recapitulated this decreased phagocytosis of Mtb. In contrast, intranasal GM-CSF treatment restored TLR2 expression and Mtb phagocytosis in the alveolar macrophages of alcohol-fed rats to levels comparable to those of control-fed rats. CONCLUSIONS: Chronic alcohol ingestion reduces TLR2 protein expression and phagocytosis of Mtb, likely due to impaired GM-CSF signaling. GM-CSF restores membrane-bound TLR2 expression and phagocytic function.


Asunto(s)
Etanol , Macrófagos Alveolares , Mycobacterium tuberculosis , Fagocitosis , Receptor Toll-Like 2 , Animales , Ratas , Etanol/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Mycobacterium tuberculosis/metabolismo , Receptor Toll-Like 2/metabolismo , Fagocitosis/efectos de los fármacos
2.
Am J Physiol Cell Physiol ; 317(2): C390-C397, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091144

RESUMEN

Chronic HIV infection causes redox stress and increases the risk of acute and chronic lung injury, even when individuals are adherent to antiretroviral therapy. HIV-1 transgene expression in rats inhibits nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which regulates antioxidant defenses and alveolar epithelial cell (AEC) barrier function, but the mechanism is unknown. In this study, we present novel evidence that these pathological effects of HIV are mediated by microRNA-144 (miR-144). HIV-1 transgene expression in vivo increases the expression of miR-144 in the alveolar epithelium, and this can be replicated by direct exposure of naïve primary AECs to either Tat or gp120 ex vivo. Further, treating naïve primary AECs with a miR-144 mimic decreased the expression and activity of Nrf2 and inhibited their barrier formation. In contrast, treatment with a miR-144 antagomir increased the expression and activity of Nrf2 and improved barrier function in primary AECs isolated from HIV-1 transgenic rats. Importantly, either delivering the miR-144 antagomir intratracheally, or directly activating Nrf2 by dietary treatment with PB123, increased Nrf2 expression and barrier formation in HIV-1 transgenic rat AECs. This study provides new experimental evidence that HIV-induced inhibition of Nrf2 and consequent AEC barrier dysfunction are mediated via miR-144, and that these pathophysiological effects can be mitigated in vivo by either directly antagonizing miR-144 or activating Nrf2. Our findings suggest that targeting the inhibition of Nrf2 in individuals living with HIV could enhance their lung health and decrease the lung-specific morbidity and mortality that persists despite antiretroviral therapy.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , MicroARNs/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/virología , Animales , Antagomirs/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Interacciones Huésped-Patógeno , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/genética , Ratas Endogámicas F344 , Ratas Transgénicas , Transducción de Señal
3.
Alcohol Clin Exp Res ; 43(7): 1427-1438, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31081931

RESUMEN

BACKGROUND: Alcohol exposure induces TGFß1 and renders the lung susceptible to injury and disrepair. We determined that TGFß1 regulates myofibroblast differentiation through the loss of Thy-1 expression and consequent induction of α-SMA. TGFß1 is important for T helper 17 (Th17) differentiation and IL-17 secretion, which in turn participates in tissue repair. We hypothesized that alcohol induces Th17 differentiation via TGFß1 and that IL-17 produced by these cells contributes to the development of profibrotic lung myofibroblasts. METHODS: Primary lung fibroblasts (PLFs) were treated with alcohol, TGFß1, and IL-17 and then analyzed for Thy-1 expression and cell morphology. Naïve and Th17-polarized CD4+ T cells were exposed to alcohol and assessed for IL-17 expression. CD4+ T cells from alcohol-fed mice were analyzed for Th17 and IL-17 expression. Lungs of control-fed, bleomycin-treated and alcohol-fed, bleomycin-treated mice were analyzed for IL-17 protein expression. RESULTS: Alcohol-treated PLFs expressed lower levels of Thy-1 than untreated cells. TGFß1 or IL-17 exposure suppressed PLF Thy-1 expression. When administered together, TGFß1 and IL-17 additively down-regulated Thy-1 expression. Exposure of naïve and Th17-polarized CD4+ T cells to alcohol induced the Th17 phenotype and augmented their production of IL-17. CD4+ Th17+ levels are elevated in the peripheral compartment but not in the lungs of alcohol-fed animals. Treatment of the PLFs with IL-17 and alcohol induced α-SMA expression. Induction of α-SMA and myofibroblast morphology by IL-17 occurred selectively in a Thy-1- fibroblast subpopulation. Chronic alcohol ingestion augmented lung-specific IL-17 expression following bleomycin-induced lung injury. CONCLUSIONS: Alcohol exposure skews T cells toward a Th17 immune response that in turn primes the lung for fibroproliferative disrepair through loss of Thy-1 expression and induction of myofibroblast differentiation. These effects suggest that IL-17 and TGFß1 contribute to fibroproliferative disrepair in the lung and targeting these proteins could limit morbidity and mortality following lung injury in alcoholic individuals.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Fibroblastos/efectos de los fármacos , Interleucina-17/biosíntesis , Pulmón/patología , Miofibroblastos/efectos de los fármacos , Antígenos Thy-1/biosíntesis , Antígenos Thy-1/genética , Actinas/biosíntesis , Actinas/genética , Animales , Recuento de Linfocito CD4 , Transdiferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Pulmón/efectos de los fármacos , Linfotoxina-alfa/biosíntesis , Ratones , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos
4.
Am J Respir Cell Mol Biol ; 56(5): 563-567, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27911588

RESUMEN

The advent of antiretroviral therapy has transformed infection by the type 1 human immunodeficiency virus (HIV) from a rapidly fatal disease to a chronic illness with excellent long-term survival rates. Although HIV primarily targets the adaptive arm of host immunity, it simultaneously impacts the innate immune system, and has profound implications for lung health, even when viral suppression is achieved with antiretroviral therapy. The lung has evolved a unique array of innate immune defenses, and the pathophysiological interactions between HIV and the pulmonary innate immune system deserve particular attention. In this review, we discuss work that elucidates how the components of innate immunity both respond to and are perturbed by infection with HIV.


Asunto(s)
VIH/inmunología , Inmunidad Innata/inmunología , Humanos , Pulmón/inmunología , Modelos Biológicos
5.
Alcohol Clin Exp Res ; 40(9): 1846-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27436123

RESUMEN

BACKGROUND: Alcohol abuse, which impairs antioxidant defenses and promotes acute lung injury, increases Nrf2 nuclear translocation but nevertheless inhibits its activation of the antioxidant response element (ARE). Thioredoxin-1 (Trx1) is required for optimal Nrf2 binding and activation of the ARE, and we hypothesized that its inhibition contributes to impaired Nrf2-ARE signaling in the alcoholic lung. METHODS: Lung tissue and primary lung fibroblasts (PLFs) were isolated from C57/BL6 wild-type (WT) and transgenic mice overexpressing the human Trx1 gene with a nuclear localizing sequence (NLS-Tg); some mice consumed alcohol in water prior to lung tissue and PLF isolation; in some mice, acute lung injury was induced with intratracheal bleomycin. In other experiments, PLFs were isolated from WT and NLS-Tg mice and then exposed to alcohol. Finally, PLF isolated from WT mice were transfected with Trx1 expression vector containing either a cytosolic localized sequence (NES) or a nuclear localized sequence (NLS) prior to alcohol exposure. RESULTS: Alcohol treatment in vivo or in vitro decreased Trx1 expression, and bleomycin-treated alcohol-fed mice had fibrotic disrepair in their lungs. In parallel, whereas alcohol exposure in vitro increased TGFß1 expression and decreased Nrf2-ARE activity in PLF from WT mice, these effects were not observed in PLF from NLS-Tg mice. Finally, selective overexpression of Trx1 in the nucleus but not in the cytosol preserved Nrf2-ARE activity during alcohol exposure. CONCLUSIONS: Although alcohol-induced redox stress actually promotes Nrf2 nuclear translocation, the coincident suppression of Trx1 impairs Nrf2-ARE activity within the nuclear compartment. Nuclear overexpression of Trx1 restored Nrf2-ARE activity and attenuated alcohol-induced TGFß1 expression and alcohol-induced exaggerate response to bleomycin-induced acute lung injury.


Asunto(s)
Núcleo Celular/metabolismo , Etanol/administración & dosificación , Factor 2 Relacionado con NF-E2/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal/fisiología , Tiorredoxinas/biosíntesis , Animales , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/prevención & control , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 308(10): L1086-93, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25840997

RESUMEN

Alveolar macrophage (AM) immune function depends on the activation of the transcription factor PU.1 by granulocyte macrophage colony-stimulating factor. We have determined that chronic alcohol ingestion dampens PU.1 signaling via an unknown zinc-dependent mechanism; specifically, although PU.1 is not known to be a zinc-dependent transcription factor, zinc treatment reversed alcohol-mediated dampening of PU.1 signaling. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a zinc-dependent basic leucine zipper protein essential for antioxidant defenses, is also impaired by chronic alcohol ingestion and enhanced by zinc treatment. We hypothesized that the response of PU.1 to zinc treatment may result from the action of Nrf2 on PU.1. We first performed Nrf2/PU.1 protein coimmunoprecipitation on a rat AM cell line (NR8383) and found no evidence of protein-protein interactions. We then found evidence of increased Nrf2 binding to the PU.1 promoter region by chromatin immunoprecipitation. We next activated Nrf2 using either sulforaphane or an overexpression vector and inhibited Nrf2 with silencing RNA to determine whether Nrf2 could actively regulate PU.1. Nrf2 activation increased protein expression of both factors as well as gene expression of their respective downstream effectors, NAD(P)H dehydrogenase[quinone] 1 (NQO1) and cluster of differentiation antigen-14 (CD14). In contrast, Nrf2 silencing decreased the expression of both proteins, as well as gene expression of their effectors. Activating and inhibiting Nrf2 in primary rat AMs resulted in similar effects. Taken together, these findings suggest that Nrf2 regulates the expression and activity of PU.1 and that antioxidant response and immune activation are coordinately regulated within the AM.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos Alveolares/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Elementos de Respuesta , Transactivadores/biosíntesis , Animales , Línea Celular , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Macrófagos Alveolares/citología , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/genética , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Endogámicas F344 , Transactivadores/genética , Zinc/farmacología
7.
Am J Physiol Lung Cell Mol Physiol ; 308(12): L1212-23, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25888574

RESUMEN

Lung barrier dysfunction is a cardinal feature of the acute respiratory distress syndrome (ARDS). Alcohol abuse, which increases the risk of ARDS two- to fourfold, induces transforming growth factor (TGF)-ß1, which increases epithelial permeability and impairs granulocyte/macrophage colony-stimulating factor (GM-CSF)-dependent barrier integrity in experimental models. We hypothesized that the relative balance of GM-CSF and TGF-ß1 signaling regulates lung epithelial barrier function. GM-CSF and TGF-ß1 were tested separately and simultaneously for their effects on lung epithelial cell barrier function in vitro. TGF-ß1 alone caused an ∼ 25% decrease in transepithelial resistance (TER), increased paracellular flux, and was associated with projections perpendicular to tight junctions ("spikes") containing claudin-18 that colocalized with F-actin. In contrast, GM-CSF treatment induced an ∼ 20% increase in TER, decreased paracellular flux, and showed decreased colocalization of spike-associated claudin-18 with F-actin. When simultaneously administered to lung epithelial cells, GM-CSF antagonized the effects of TGF-ß1 on epithelial barrier function in cultured cells. Given this, GM-CSF and TGF-ß1 levels were measured in bronchoalveolar lavage (BAL) fluid from patients with ventilator-associated pneumonia and correlated with markers for pulmonary edema and patient outcome. In patient BAL fluid, protein markers of lung barrier dysfunction, serum α2-macroglobulin, and IgM levels were increased at lower ratios of GM-CSF/TGF-ß1. Critically, patients who survived had significantly higher GM-CSF/TGF-ß1 ratios than nonsurviving patients. This study provides experimental and clinical evidence that the relative balance between GM-CSF and TGF-ß1 signaling is a key regulator of lung epithelial barrier function. The GM-CSF/TGF-ß1 ratio in BAL fluid may provide a concentration-independent biomarker that can predict patient outcomes in ARDS.


Asunto(s)
Células Epiteliales/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Pulmón/metabolismo , Fenómenos Fisiológicos Respiratorios , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Pulmón/citología , Masculino , Ratas , Ratas Sprague-Dawley
8.
Alcohol Clin Exp Res ; 38(2): 336-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24033682

RESUMEN

BACKGROUND: Alcohol abuse increases the risk for acute lung injury (ALI). In both experimental models and in clinical studies, chronic alcohol ingestion causes airway oxidative stress and glutathione depletion and increases the expression of transforming growth factor beta-1 (TGFß1), a potent inducer of fibrosis, in the lung. Therefore, we hypothesized that alcohol ingestion could promote aberrant fibrosis following experimental ALI and that treatment with the glutathione precursor s-adenosylmethionine (SAMe) could mitigate these effects. METHODS: Three-month-old C57BL/6 mice were fed standard chow ± alcohol (20% v/v) in their drinking water for 8 weeks and ±SAMe (4% w/v) during the last 4 weeks. ALI was induced by intratracheal instillation of bleomycin (2.5 units/kg), and lungs were assessed histologically at 7 and 14 days for fibrosis and at 14 days for the expression of extracellular matrix proteins and TGFß1. RESULTS: Alcohol ingestion had no apparent effect on lung inflammation at 7 days, but at 14 days after bleomycin treatment, it increased lung tissue collagen deposition, hydroxyproline content, and the release of activated TGFß1 into the airway. In contrast, SAMe supplementation completely mitigated alcohol-induced priming of these aberrant fibrotic changes through decreased TGFß1 expression in the lung. In parallel, SAMe decreased alcohol-induced TGFß1 and Smad3 mRNA expressions by lung fibroblasts in vitro. CONCLUSIONS: These new experimental findings demonstrate that chronic alcohol ingestion renders the experimental mouse lung susceptible to fibrosis following bleomycin-induced ALI, and that these effects are likely driven by alcohol-mediated oxidative stress and its induction and activation of TGFß1.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Fibrosis Pulmonar/inducido químicamente , Actinas/biosíntesis , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Bleomicina/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Dieta , Ensayo de Inmunoadsorción Enzimática , Etanol/antagonistas & inhibidores , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Hidroxiprolina/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neumonía/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , ARN Mensajero/biosíntesis , ARN Mensajero/genética , S-Adenosilmetionina/farmacología , Factor de Crecimiento Transformador beta1/biosíntesis
9.
Alcohol Clin Exp Res ; 38(11): 2731-42, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25421510

RESUMEN

BACKGROUND: Chronic alcohol ingestion induces the expression of transforming growth factor beta-1(TGFß1), inhibits nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated activation of the antioxidant response element (ARE), depletes alveolar glutathione pools, and potentiates acute lung injury. In this study, we examined the mechanistic relationship between TGFß1 and Nrf2-ARE signaling in the experimental alcoholic lung. METHODS: Wild-type mice were treated ± alcohol in drinking water for 8 weeks and their lungs were assessed for Nrf2 expression. In parallel, mouse lung fibroblasts were cultured ± alcohol and treated ± sulforaphane (SFP; an activator of Nrf2), ±TGFß1, ±TGFß1 neutralizing antibody, and/or ±activin receptor-like kinase 5 inhibitors (to block TGß1 receptor signaling) and then analyzed for the expression of Nrf2, Kelch-like ECH-associated protein 1 (Keap1) and TGFß1, Nrf2-ARE activity, and the expression of the Nrf2-ARE-dependent antioxidants glutathione s-transferase theta 2 (GSTT2) and glutamate-cysteine ligase catalytic subunit (GCLC). Finally, silencing RNA (siRNA) of Nrf2 was then performed prior to alcohol exposure and subsequent analysis of TGFß1 expression. RESULTS: Alcohol treatment in vivo or in vitro decreased Nrf2 expression in murine whole lung and lung fibroblasts, respectively. In parallel, alcohol exposure in vitro decreased Keap1 gene and protein expression in lung fibroblasts. Furthermore, alcohol exposure increased TGFß1 expression but decreased Nrf2-ARE activity and expression of the ARE-dependent genes for GSTT2 and GCLC. These effects of alcohol were prevented by treatment with SFP; in contrast, Nrf2 SiRNA expression exacerbated alcohol-induced TGFß1 expression. Finally, TGFß1 treatment directly suppressed Nrf2-ARE activity whereas blocking TGFß1 signaling attenuated alcohol-induced suppression of Nrf2-ARE activity. CONCLUSIONS: Alcohol-induced oxidative stress is mediated by TGFß1, which suppresses Nrf2-ARE-dependent expression of antioxidant defenses and creates a vicious cycle that feeds back to further increase TGFß1 expression. These effects of alcohol can be mitigated by activation of Nrf2, suggesting a potential therapy in individuals at risk for lung injury due to alcohol abuse.


Asunto(s)
Etanol/toxicidad , Fibroblastos/metabolismo , Pulmón/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Factor de Crecimiento Transformador beta1/fisiología , Animales , Línea Celular Transformada , Fibroblastos/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Células 3T3 NIH , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Factor de Crecimiento Transformador beta1/farmacología
10.
Am J Respir Crit Care Med ; 188(6): 716-23, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23805851

RESUMEN

RATIONALE: Alcohol use disorders cause oxidative stress in the lower airways and increase susceptibility to pneumonia and lung injury. Currently, no therapeutic options exist to mitigate the pulmonary consequences of alcoholism. OBJECTIVES: We recently determined in an animal model that alcohol ingestion impairs pulmonary zinc metabolism and causes alveolar macrophage immune dysfunction. The objective of this research is to determine the effects of alcoholism on zinc bioavailability and alveolar macrophage function in human subjects. METHODS: We recruited otherwise healthy alcoholics (n = 17) and matched control subjects (n = 17) who underwent bronchoscopy for isolation of alveolar macrophages, which were analyzed for intracellular zinc, phagocytic function, and surface expression of granulocyte-macrophage colony-stimulating factor receptor; all three of these indices are decreased in experimental models. MEASUREMENTS AND MAIN RESULTS: Alcoholic subjects had normal serum zinc, but significantly decreased alveolar macrophage intracellular zinc levels (adjusted means [SE], 718 [41] vs. 948 [25] RFU/cell; P < 0.0001); bacterial phagocytosis (adjusted means [SE], 1,027 [48] vs. 1,509 [76] RFU/cell; P < 0.0001); and expression of granulocyte-macrophage colony-stimulating factor receptor ß subunit (adjusted means [SE], 1,471 [42] vs. 2,114 [35] RFU/cell; P < 0.0001]. Treating alveolar macrophages with zinc acetate and glutathione in vitro increased intracellular zinc levels and improved their phagocytic function. CONCLUSIONS: These novel clinical findings provide evidence that alcohol abuse is associated with significant zinc deficiency and immune dysfunction within the alveolar space and suggest that dietary supplementation with zinc and glutathione precursors could enhance airway innate immunity and decrease the risk for pneumonia or lung injury in these vulnerable individuals.


Asunto(s)
Alcoholismo/complicaciones , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Enfermedades del Sistema Inmune/inducido químicamente , Macrófagos Alveolares/metabolismo , Zinc/deficiencia , Adolescente , Adulto , Alcoholismo/inmunología , Alcoholismo/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , Broncoscopía/métodos , Etanol/efectos adversos , Etanol/inmunología , Etanol/metabolismo , Femenino , Humanos , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/metabolismo , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Macrófagos Alveolares/inmunología , Masculino , Persona de Mediana Edad , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Adulto Joven , Zinc/inmunología , Zinc/metabolismo
11.
Am J Respir Cell Mol Biol ; 48(4): 511-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23306837

RESUMEN

Excessive alcohol use increases the risk of acute lung injury and pneumonia. Chronic alcohol ingestion causes oxidative stress within the alveolar space, including near depletion of glutathione (GSH), which impairs alveolar epithelial and macrophage function, in experimental animals and human subjects. However, the fundamental mechanism(s) by which alcohol induces such profound lung oxidative stress is unknown. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a redox-sensitive master transcription factor that regulates activation of the antioxidant response element (ARE). As the alveolar epithelium controls GSH levels within the alveolar space, we hypothesized that alcohol also decreases Nrf2 expression and/or activation within the alveolar epithelium. In this study, we determined that alcohol ingestion in vivo or direct alcohol exposure in vitro down-regulated the Nrf2-ARE pathway in lung epithelial cells, decreased the expression of antioxidant genes, and lowered intracellular GSH levels. RNA silencing of Nrf2 gene expression in alveolar epithelial cells in vitro decreased expression of these same antioxidant genes, and likewise lowered intracellular GSH levels, findings that mirrored the effects of alcohol. In contrast, treating alcohol-exposed alveolar epithelial cells in vitro with the Nrf2 activator, sulforaphane, preserved Nrf2 expression, ARE activation, intracellular GSH levels, and epithelial barrier function. These new experimental findings implicate down-regulation of the Nrf2-ARE signaling pathway as a fundamental mechanism by which alcohol causes profound oxidative stress and alveolar epithelial dysfunction, and suggest that treatments, such as sulforaphane, that activate this pathway could mitigate the pathophysiological consequences of alcohol on the lung and other organs.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Consumo de Bebidas Alcohólicas/efectos adversos , Barrera Alveolocapilar/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Estrés Oxidativo , Elementos de Respuesta , Transducción de Señal , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Animales , Anticarcinógenos/farmacología , Barrera Alveolocapilar/patología , Línea Celular , Depresores del Sistema Nervioso Central/efectos adversos , Depresores del Sistema Nervioso Central/farmacología , Etanol/efectos adversos , Etanol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/genética , Glutatión/metabolismo , Humanos , Isotiocianatos , Masculino , Factor 2 Relacionado con NF-E2/genética , Ratas , Ratas Sprague-Dawley , Sulfóxidos , Tiocianatos/farmacología
12.
Am J Physiol Lung Cell Mol Physiol ; 305(3): L267-77, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23748533

RESUMEN

The master transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) regulates the expression of antioxidant and phase II-metabolizing enzymes by activating the antioxidant response element (ARE) and thereby protects cells and tissues from oxidative stress. Pulmonary complications remain the leading cause of death in human immunodeficiency virus (HIV)-1-infected individuals, who display systemic oxidative stress and glutathione deficiency that can be modeled in transgenic rats where HIV-1-related viral proteins decrease glutathione levels and cause epithelial barrier dysfunction within the alveolar space by as yet unknown mechanisms. We hypothesized that HIV-1-related proteins inhibit Nrf2-mediated antioxidant defenses and thereby disrupt the normally tight alveolar epithelial barrier. Nrf2 RNA silencing dampened Nrf2/ARE activity, decreased the expression of the tight junction proteins zonula occludens-1, occludin, and claudin-18, increased paracellular permeability of alveolar epithelial monolayers derived from wild-type rats, and therefore reproduced the effects of HIV-1 transgene expression on the epithelial barrier that we had previously described. In contrast, upregulating Nrf2 activity, either by plasmid-mediated overexpression or treatment with the Nrf2 activator sulforaphane, increased the expression of ARE-dependent antioxidants, including NAD(P)H dehydrogenase, quinone 1 and glutathione, improved the expression of tight junction proteins, and restored the ability to form tight barriers in alveolar epithelial cells from HIV-1 transgenic rats. Taken together, these new findings argue that HIV-1-related proteins downregulate Nrf2 expression and/or activity within the alveolar epithelium, which in turn impairs antioxidant defenses and barrier function, thereby rendering the lung susceptible to oxidative stress and injury. Furthermore, this study suggests that activating the Nrf2/ARE pathway with the dietary supplement sulforaphane could augment antioxidant defenses and lung health in HIV-1-infected individuals.


Asunto(s)
Elementos de Respuesta Antioxidante/fisiología , VIH-1/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Alveolos Pulmonares/metabolismo , Animales , Anticarcinógenos/farmacología , Células Cultivadas , Claudinas/metabolismo , Regulación hacia Abajo , Glutatión/análisis , Glutatión/biosíntesis , Isotiocianatos , NAD(P)H Deshidrogenasa (Quinona)/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Ocludina/metabolismo , Quinonas/metabolismo , Interferencia de ARN , ARN Mensajero , Ratas , Ratas Transgénicas , Sulfóxidos , Tiocianatos/farmacología , Proteínas de Uniones Estrechas/biosíntesis , Proteína de la Zonula Occludens-1/metabolismo
13.
Respir Res ; 14: 39, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23547562

RESUMEN

BACKGROUND: Chronic alcohol abuse causes oxidative stress and impairs alveolar epithelial barrier integrity, thereby rendering the lung susceptible to acute edematous injury. Experimentally, alcohol-induced oxidative stress increases the expression of transforming growth factor ß1 (TGFß1) in the lung; however, we do not know the precise contribution of various alveolar cells in this process. In the present study, we focused on cell-cell interactions between alveolar macrophages and epithelial cells and the potential mechanisms by which TGFß1 may become activated in the alveolar space of the alcoholic lung. METHODS: Primary alveolar macrophages and epithelial cells were isolated from control- and alcohol-fed Sprague-Dawley rats. Expression of TGFß1 and the epithelial integrin αvß6 were examined by real time PCR and either immunocytochemistry or flow cytometry. Alveolar epithelial cells were cultured on transwell supports in the presence of macrophage cell lysate from control- or alcohol-fed rats or in the presence of viable macrophages ± alcohol. Epithelial barrier function was assessed by transepithelial resistance (TER) and paracellular flux of Texas Red dextran. RESULTS: TGFß1 expression was increased in alveolar macrophages from alcohol-fed rats, and TGFß1 protein was predominantly membrane-bound. Importantly, alveolar macrophage cellular lysate from alcohol-fed rats decreased TER and increased paracellular dextran flux in primary alveolar epithelial cell monolayers as compared to the lysates from control-fed rats. Alcohol-induced epithelial barrier dysfunction was prevented by anti-TGFß1 antibody treatment, indicating the presence of bioactive TGFß1 in the macrophage lysate. In addition, co-culturing macrophages and epithelial cells in the presence of alcohol decreased epithelial barrier function, which also was prevented by anti-TGFß1 and anti-αvß6 treatment. In parallel, chronic alcohol ingestion in vivo, or direct treatment with active TGFß1 in vitro, increased the expression of αvß6 integrin, which is known to activate TGFß1, in alveolar epithelial cells. CONCLUSIONS: Taken together, these data suggest that interactions between alveolar epithelial cells and macrophages contribute to the alcohol-mediated disruption of epithelial barrier function via the expression and activation of TGFß1 at points of cell-cell contact.


Asunto(s)
Comunicación Celular , Células Epiteliales/metabolismo , Etanol/toxicidad , Macrófagos/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Administración Oral , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Etanol/administración & dosificación , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/fisiología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
14.
Alcohol Clin Exp Res ; 37(3): 361-71, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23013362

RESUMEN

BACKGROUND: Chronic alcohol ingestion alters the dynamic balance between granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor beta1 (TGFß1) signaling within the alveolar space and, in parallel, impairs alveolar macrophage and epithelial cell function by inhibiting expression of the zinc importer ZIP4 and decreasing zinc bioavailability in the alveolar compartment. As the transcription factor Krüppel-like factor 4 (KLF4 ) binds to ZIP4 , we hypothesized that alcohol exposure and consequent perturbations in GM-CSF and TGFß1 signaling could decrease cellular KLF4 expression and/or binding as a mechanism by which it inhibits ZIP4 expression and decreases cellular zinc levels. METHODS AND RESULTS: Alcohol exposure in vitro or chronic ingestion in vivo decreased KLF4 expression in alveolar macrophages and epithelial cells. Treatment with GM-CSF or TGFß1 showed an enhancing or dampening effect on KLF4 expression and binding, respectively. Further, treatment of a rat alveolar macrophage cell line with alcohol in vitro for 4 weeks decreased the expression of the zinc transporters ZIP4 and ZNT1, and of the zinc storage protein metallothionein 1. In parallel, treating these macrophages with KLF4 siRNA decreased ZIP4 expression and decreased cellular zinc and phagocytic capacity to levels equivalent to those following alcohol exposure. In epithelial monolayers, transepithelial electrical resistance (TER) was significantly decreased by alcohol ingestion as compared with control diets, and it was restored by in vitro GM-CSF treatment. In contrast, in vitro TGFß1 treatment of the epithelial monolayers from control-fed rats significantly decreased TER as compared with untreated control monolayers. CONCLUSIONS: Taken together, these results suggest that within the alveolar space, chronic alcohol exposure decreases KLF4 and ZIP4 expression and consequently decreases zinc transport into cells, which, in turn, impairs their function. Furthermore, the dynamic decrease in the relative influence of GM-CSF versus TGFß1 could mediate the zinc deficiency and consequent cellular dysfunction that characterize the "alcoholic lung" phenotype.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Proteínas de Transporte de Catión/antagonistas & inhibidores , Regulación hacia Abajo/genética , Líquido Intracelular/metabolismo , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Dedos de Zinc/genética , Zinc/metabolismo , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/patología , Animales , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Línea Celular , Células Cultivadas , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos Alveolares/patología , Masculino , Ratas , Ratas Sprague-Dawley
15.
Alcohol Clin Exp Res ; 37(6): 914-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23421903

RESUMEN

BACKGROUND: Chronic ethanol (EtOH) abuse in humans is known to independently increase the incidence of and mortality due to acute lung injury in at-risk individuals. However, the mechanisms by which EtOH affects lung cells remain incompletely elucidated. In earlier work, we reported that EtOH increased the expression in lung fibroblasts of fibronectin, a matrix glycoprotein implicated in lung injury and repair. This effect was blocked by α-bungarotoxin, a neurotoxin that binds certain nicotinic acetylcholine receptors (nAChRs) thereby implicating nAChRs in this process. Here, we examine the identity of these receptors. METHODS: Mouse lung fibroblasts were stimulated with EtOH (60 mM) or acetylcholine (100 to 500 µM) and evaluated for the expression of fibronectin and nAChRs. Inhibitors to nAChRs or the antioxidant N-acetyl cysteine (NAC) were used to assess changes in fibronectin expression. Animals exposed to EtOH for up to 6 weeks were used to evaluate the expression of nAChRs in vivo. RESULTS: First, in EtOH-treated fibroblasts, we observed increased expression of α4 and α9 nAChR subunits. Second, we found that acetylcholine, a natural ligand for nAChRs, mimicked the effects of EtOH. Dihydro-ß-erythroidin hydrobromide, a competitive inhibitor of α4 nAChR, blocked the increase in fibronectin expression and cell proliferation. Furthermore, EtOH-induced fibronectin expression was inhibited in cells silenced for α4 nAChR. However, EtOH-treated cells showed increased α-bungarotoxin binding suggesting that α4 nAChR mediates the effects of EtOH via a ligand-independent pathway. Knowing there are several important cysteine residues near the ligand-binding site of α4 nAChRs, we tested the antioxidant NAC and found that it too blocked the induction of fibronectin expression by EtOH. Also, fibroblasts exposed to oxidant stress showed increased fibronectin expression that was blocked with α-bungarotoxin. Finally, we showed increased expression of α4 nAChRs in the lung tissue of mice and rats exposed to EtOH suggesting a role for these receptors in vivo. CONCLUSIONS: Altogether, our observations suggest that α4 nAChRs serve as sensors for EtOH-induced oxidant stress in lung fibroblasts, thereby revealing a new mechanism by which EtOH may affect lung cells and tissue remodeling and pointing to nAChRs as potential targets for intervention.


Asunto(s)
Etanol/farmacología , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Pulmón/citología , Receptores Nicotínicos/metabolismo , Acetilcolina/farmacología , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Fibronectinas/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Estrés Oxidativo , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/fisiología
16.
Proc Natl Acad Sci U S A ; 107(31): 13848-53, 2010 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-20643942

RESUMEN

Osteoporosis and bone fractures are increasingly recognized complications of HIV-1 infection. Although antiretroviral therapy itself has complex effects on bone turnover, it is now evident that the majority of HIV-infected individuals already exhibit reduced bone mineral density before therapy. The mechanisms responsible are likely multifactorial and have been difficult to delineate in humans. The HIV-1 transgenic rat recapitulates many key features of human AIDS. We now demonstrate that, like their human counterparts, HIV-1 transgenic rats undergo severe osteoclastic bone resorption, a consequence of an imbalance in the ratio of receptor activator of NF-kappaB ligand, the key osteoclastogenic cytokine, to that of its physiological decoy receptor osteoprotegerin. This imbalance stemmed from a switch in production of osteoprotegerin to that of receptor activator of NF-kappaB ligand by B cells, and was further compounded by a significantly elevated number of osteoclast precursors. With the advancing age of individuals living with HIV/AIDS, low bone mineral density associated with HIV infection is likely to collide with the pathophysiology of skeletal aging, leading to increased fracture risk. Understanding the mechanisms driving bone loss in HIV-infected individuals will be critical to developing effective therapeutic strategies.


Asunto(s)
Resorción Ósea/inmunología , VIH-1/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Densidad Ósea , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Masculino , Ligando RANK/biosíntesis , Ratas , Ratas Endogámicas F344 , Ratas Transgénicas
17.
Alcohol Clin Exp Res (Hoboken) ; 47(10): 1839-1849, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37864530

RESUMEN

BACKGROUND: Chronic alcohol ingestion predisposes to lung injury and disrepair during sepsis. Our previous studies outlined roles for transforming growth factor-beta 1 (TGFß1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in epithelial barrier homeostasis and how alcohol perturbs their expression and signaling. Here we hypothesize that ethanol-exposed lung fibroblasts (LF) are a source of dysregulated TGFß1 and GM-CSF and thereby alter airway epithelial barrier function. METHODS: Human or rat LF were cultured ± ethanol for 2 weeks and then co-cultured with human or rat airway epithelial cells (AEC) seeded on Transwell permeable supports. In selected groups, a TGFß1 receptor type 1 (TGFßR1) inhibitor (SB431542) or a TGFß1 neutralizing antibody was applied. Transepithelial electrical resistance (TER) was measured prior to co-culture and on day 5 of co-culture. AEC were then analyzed for the expression of selected tight junction and mesenchymal proteins, and transwell membranes were analyzed by immunofluorescence microscopy for ZO-1 expression and localization. TGFß1 and GM-CSF levels in conditioned media from the co-cultures were quantified by ELISA. RESULTS: AEC co-cultured with ethanol-exposed LF (ELF) showed a significant reduction in TER and corresponding decreases in ZO-1 expression, whereas collagen type 1A1 and α-smooth muscle actin protein expression were increased. In parallel, in conditioned media from the ELF + AEC co-cultures, activated TGFß1 levels increased and GM-CSF levels decreased. Notably, all the effects of ELF on the AEC were prevented by blocking TGFß1 activity. CONCLUSIONS: Prior ethanol exposure to LF induces barrier dysfunction in naive AEC in a paracrine fashion through activation of TGFß1 signaling and suppression of GM-CSF. These experimental findings provide a potential mechanism by which chronic alcohol ingestion impairs airway epithelial integrity and renders individuals susceptible to lung injury.

18.
Shock ; 59(4): 612-620, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36640152

RESUMEN

ABSTRACT: Increased epithelial permeability in sepsis is mediated via disruptions in tight junctions, which are closely associated with the perijunctional actin-myosin ring. Genetic deletion of myosin light chain kinase (MLCK) reverses sepsis-induced intestinal hyperpermeability and improves survival in a murine model of intra-abdominal sepsis. In an attempt to determine the generalizability of these findings, this study measured the impact of MLCK deletion on survival and potential associated mechanisms following pneumonia-induced sepsis. MLCK -/- and wild-type mice underwent intratracheal injection of Pseudomonas aeruginosa . Unexpectedly, survival was significantly worse in MLCK -/- mice than wild-type mice. This was associated with increased permeability to Evans blue dye in bronchoalveolar lavage fluid but not in tissue homogenate, suggesting increased alveolar epithelial leak. In addition, bacterial burden was increased in bronchoalveolar lavage fluid. Cytokine array using whole-lung homogenate demonstrated increases in multiple proinflammatory and anti-inflammatory cytokines in knockout mice. These local pulmonary changes were associated with systemic inflammation with increased serum levels of IL-6 and IL-10 and a marked increase in bacteremia in MLCK -/- mice. Increased numbers of both bulk and memory CD4 + T cells were identified in the spleens of knockout mice, with increased early and late activation. These results demonstrate that genetic deletion of MLCK unexpectedly increases mortality in pulmonary sepsis, associated with worsened alveolar epithelial leak and both local and systemic inflammation. This suggests that caution is required in targeting MLCK for therapeutic gain in sepsis.


Asunto(s)
Pulmón , Quinasa de Cadena Ligera de Miosina , Neumonía , Sepsis , Animales , Ratones , Citocinas , Inflamación , Mucosa Intestinal , Pulmón/metabolismo , Pulmón/patología , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/genética , Permeabilidad , Neumonía/complicaciones , Sepsis/patología , Uniones Estrechas/fisiología
19.
Alcohol Clin Exp Res ; 35(10): 1866-75, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21569054

RESUMEN

BACKGROUND: Alcohol abuse and HIV-1 infection frequently coexist, and these individuals are at high risk for serious lung infections and respiratory failure. Although alcohol ingestion and HIV-1 transgene expression have been shown to independently cause oxidative stress and disrupt alveolar epithelial barrier function in experimental models, their interactive effects have not been examined. METHODS AND RESULTS: In this study, we determined that chronic alcohol ingestion (12 weeks) exacerbated the already significant defects in alveolar epithelial paracellular permeability and lung liquid clearance in HIV-1 transgenic rats. Further, immunocytochemical analyses of tight junction protein expression in primary alveolar epithelial cells showed that occludin and zonula occludens-1 localization within the plasma membrane was more disrupted than in either condition alone, consistent with the observed defects in epithelial barrier function. Interestingly, expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), the transcription factor required to activate the antioxidant-response element, was decreased in primary alveolar epithelial cells isolated from HIV-1 transgenic rats. In parallel, exposing lung epithelial cells in vitro to either alcohol or the HIV-related protein gp120 also decreased Nrf2 expression. Importantly, treatment with procysteine, which increases thiol antioxidants including glutathione, improved tight junction protein localization in the plasma membrane and restored alveolar epithelial barrier function in alcohol-fed HIV-1 transgenic rats. CONCLUSIONS: These results provide novel evidence that HIV-related proteins and alcohol together causes more barrier dysfunction in the lung epithelium than either stress alone. However, these significant effects on the alveolar barrier can be mitigated by augmenting the thiol antioxidant pool, a strategy with potential clinical applications in subjects who are highly vulnerable to lung disease because of coexistent alcohol abuse and HIV infection.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Infecciones por VIH/patología , VIH-1 , Pulmón/efectos de los fármacos , Alcoholismo/metabolismo , Alcoholismo/patología , Alcoholismo/fisiopatología , Animales , Antioxidantes/fisiología , Depresores del Sistema Nervioso Central/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Comorbilidad , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Epiteliales/patología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Epitelio/fisiopatología , Etanol/metabolismo , Etanol/farmacología , Glutatión/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/fisiopatología , Masculino , Proteínas de la Membrana/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/metabolismo , Ocludina , Ácido Pirrolidona Carboxílico/farmacología , Ratas , Ratas Endogámicas F344 , Ratas Transgénicas , Tiazolidinas/farmacología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Factores de Tiempo
20.
Alcohol Clin Exp Res ; 35(8): 1519-28, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21447000

RESUMEN

BACKGROUND: Chronic alcohol abuse causes oxidative stress, impairs alveolar macrophage immune function, and increases the risk of pneumonia and acute lung injury. Recently we determined that chronic alcohol ingestion in rats decreases zinc levels and macrophage function in the alveolar space; provocative findings in that zinc is essential for normal immune and antioxidant defenses. Alveolar macrophage immune function depends on stimulation by granulocyte/monocyte colony-stimulating factor, which signals via the transcription factor PU.1. In parallel, the antioxidant response element signals via the transcription factor Nrf2. However, the role of zinc bioavailability on these signaling pathways within the alveolar space is unknown. METHODS: To determine the efficacy of dietary zinc supplementation on lung bacterial clearance and oxidative stress, we tested 3 different groups of rats: control-fed, alcohol-fed, and alcohol-fed with zinc supplementation. Rats were then inoculated with intratracheal Klebsiella pneumoniae, and lung bacterial clearance was determined 24 hours later. Isolated alveolar macrophages were isolated from uninfected animals and evaluated for oxidative stress and signaling through PU.1 and Nrf2. RESULTS: Alcohol-fed rats had a 5-fold decrease in lung bacterial clearance compared to control-fed rats. Dietary zinc supplementation of alcohol-fed rats normalized bacterial clearance and mitigated oxidative stress in the alveolar space, as reflected by the relative balance of the thiol redox pair cysteine and cystine, and increased nuclear binding of both PU.1 and Nrf2 in alveolar macrophages from alcohol-fed rats. CONCLUSIONS: Dietary zinc supplementation prevents alcohol-induced alveolar macrophage immune dysfunction and oxidative stress in a relevant experimental model, suggesting that such a strategy could decrease the risk of pneumonia and lung injury in individuals with alcohol use disorders.


Asunto(s)
Macrófagos Alveolares , Factor 2 Relacionado con NF-E2 , Proteínas Proto-Oncogénicas , Oligoelementos , Transactivadores , Zinc , Animales , Masculino , Ratas , Alcoholismo/metabolismo , Alcoholismo/fisiopatología , Modelos Animales de Enfermedad , Etanol , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Infecciones por Klebsiella/metabolismo , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Pulmón/inmunología , Pulmón/fisiopatología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factores de Tiempo , Oligoelementos/farmacología , Oligoelementos/uso terapéutico , Transactivadores/metabolismo , Zinc/farmacología , Zinc/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA