Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Front Immunol ; 11: 595282, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33224150

RESUMEN

Background: Selenium (Se) levels decrease in the circulation during acute inflammatory states and sepsis, and are inversely associated with morbidity and mortality. A more specific understanding of where selenoproteins and Se processing are compromised during insult is needed. We investigated the acute signaling response in selenoenzymes and Se processing machinery in multiple organs after innate immune activation in response to systemic lipopolysaccharide (LPS). Methods: Wild type (WT) adult male C57/B6 mice were exposed to LPS (5 mg/kg, intraperitoneal). Blood, liver, lung, kidney and spleen were collected from control mice as well as 2, 4, 8, and 24 h after LPS. Plasma Se concentration was determined by ICP-MS. Liver, lung, kidney and spleen were evaluated for mRNA and protein content of selenoenzymes and proteins required to process Se. Results: After 8 h of endotoxemia, plasma levels of Se and the Se transporter protein, SELENOP were significantly decreased. Consistent with this timing, the transcription and protein content of several hepatic selenoenzymes, including SELENOP, glutathione peroxidase 1 and 4 were significantly decreased. Furthermore, hepatic transcription and protein content of factors required for the Se processing, including selenophosphate synthetase 2 (Sps2), phosphoseryl tRNA kinase (Pstk), selenocysteine synthase (SepsecS), and selenocysteine lyase (Scly) were significantly decreased. Significant LPS-induced downregulation of these key selenium processing enzymes was observed in isolated hepatocytes. In contrast to the acute and dynamic changes observed in the liver, selenoenzymes did not decrease in the lung, kidney or spleen. Conclusion: Hepatic selenoenzyme production and Se processing factors decreased after endotoxemia. This was temporally associated with decreased circulating Se. In contrast to these active changes in the regulation of Se processing in the liver, selenoenzymes did not decrease in the lung, kidney or spleen. These findings highlight the need to further study the impact of innate immune challenges on Se processing in the liver and the impact of targeted therapeutic Se replacement strategies during innate immune challenge.


Asunto(s)
Endotoxemia/inmunología , Hígado/inmunología , Selenoproteínas/inmunología , Animales , Endotoxemia/sangre , Glutatión Peroxidasa , Hepatocitos , Riñón/inmunología , Pulmón/inmunología , Masculino , Ratones Endogámicos C57BL , Selenio/sangre , Bazo/inmunología
2.
Life Sci ; 259: 118285, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32798556

RESUMEN

AIMS: Interleukin-1ß (IL-1ß) contributes to the development of bronchopulmonary dysplasia (BPD). Thioredoxin reductase-1 (Txnrd1) inhibition activates nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent responses. Txnrd1 activity is selenium (Se) dependent and Se deficiency is common in prematurity. Auranofin (AFN), a Txnrd1 inhibitor, decreases IL-1ß levels and increases Nrf2 activation in lipopolysaccharide (LPS) treated alveolar macrophages. In lung epithelia, AFN-induced Nrf2 activation is Se dependent. We tested the hypothesis that the effects of Txnrd1 inhibition in alveolar macrophages are Se dependent. MAIN METHODS: To establish Se sufficient (Se+) and deficient (Se-) conditions, alveolar (MH-S) macrophages were cultured in 2.5% fetal bovine serum (FBS) ± 25 nM Na2SeO3. Se- (2.5% FBS) and Se+ (2.5% FBS + 25 nM Na2SeO3) cells were cultured in the presence or absence of 0.05 µg/mL LPS and/or 0.5 µM AFN. Nrf2 activation was determined by measuring NADPH quinone oxidoreductase-1 (Nqo1) and glutathione levels. IL-1ß mRNA (Il1b) and protein levels were measured using qRT-PCR and ELISA. Data were analyzed by ANOVA followed by Tukey's post-hoc. KEY FINDINGS: We detected an independent effect of AFN, but not LPS, on Nqo1 expression and GSH levels in Se+ and Se- cells. LPS significantly increased Il1b and IL-1ß levels in both groups. AFN-mediated attenuation of this effect was not impacted by Se status. SIGNIFICANCE: The beneficial effects of Txnrd1 inhibition in alveolar macrophages are Se-independent and therefore unlikely to be diminished by clinical Se deficiency.


Asunto(s)
Auranofina/farmacología , Macrófagos Alveolares/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Auranofina/metabolismo , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/fisiopatología , Glutatión/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Macrófagos/metabolismo , Macrófagos Alveolares/fisiología , Ratones , Cultivo Primario de Células , Selenio/metabolismo , Selenio/farmacología , Tiorredoxina Reductasa 1/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/metabolismo
3.
Free Radic Biol Med ; 142: 138-145, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30769161

RESUMEN

Preterm birth is a primary cause of worldwide childhood mortality. Bronchopulmonary dysplasia, characterized by impaired alveolar and lung vascular development, affects 25-50% of extremely low birth weight (BW; <1 kg) infants. Abnormalities in lung function persist into childhood in affected infants and are second only to asthma in terms of childhood respiratory disease healthcare costs. While advances in the medical care of preterm infants have reduced mortality, the incidence of BPD has not decreased in the past 10 years. Reactive oxygen intermediates play a key role in the development of lung disease but, despite promising preclinical therapies, antioxidants have failed to translate into meaningful clinical interventions to decrease the incidence of lung disease in premature infants. In this review we will summarize the state of the art research developments in regards to antioxidants and premature lung disease and discuss the limitations of antioxidant therapies in order to more fully comprehend the reasons why therapeutic antioxidant administration failed to prevent BPD. Finally we will review promising therapeutic strategies and targets.


Asunto(s)
Antioxidantes/uso terapéutico , Displasia Broncopulmonar/terapia , Suplementos Dietéticos , Factor 2 Relacionado con NF-E2/agonistas , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Tiorredoxinas/agonistas , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/fisiopatología , Niño , Glutatión/administración & dosificación , Glutatión/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Unidades de Cuidado Intensivo Neonatal , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Medicina de Precisión/métodos , Especies Reactivas de Oxígeno/metabolismo , Selenio/administración & dosificación , Selenio/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
4.
Redox Biol ; 19: 331-338, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30212802

RESUMEN

The trace element selenium (Se) contributes to redox signaling, antioxidant defense, and immune responses in critically ill neonatal and adult patients. Se is required for the synthesis and function of selenoenzymes including thioredoxin (Trx) reductase-1 (TXNRD1) and glutathione peroxidases (GPx). We have previously identified TXNRD1, primarily expressed by airway epithelia, as a promising therapeutic target to prevent lung injury, likely via nuclear factor E2-related factor 2 (Nrf2)-dependent mechanisms. The present studies utilized the TXNRD1 inhibitor auranofin (AFN) to test the hypothesis that Se positively influences Nrf2 activation and selenoenzyme responses in lung epithelial cells. Murine transformed Club cells (mtCCs) were supplemented with 0, 10, 25, or 100 nM Na2SeO3 to create a range of Se conditions and were cultured in the presence or absence of 0.5 µM AFN. TXNRD1 and GPX2 protein expression and enzymatic activity were significantly greater upon Se supplementation (p < 0.05). AFN treatment (0.5 µM AFN for 1 h) significantly inhibited TXNRD1 but not GPx activity (p < 0.001). Recovery of TXNRD1 activity following AFN treatment was significantly enhanced by Se supplementation (p < 0.041). Finally, AFN-induced Nrf2 transcriptional activation was significantly greater in mtCCs supplemented in 25 or 100 nM Na2SeO3 when compared to non-supplemented controls (p < 0.05). Our novel studies indicate that Se levels positively influence Nrf2 activation and selenoenzyme responses following TXNRD1 inhibition. These data suggest that Se status significantly influences physiologic responses to TXNRD1 inhibitors. In conclusion, correction of clinical Se deficiency, if present, will be necessary for optimal therapeutic effectiveness of TXNRD1 inhibitors in the prevention of lung disease.


Asunto(s)
Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Selenio/farmacología , Tiorredoxina Reductasa 1/antagonistas & inhibidores , Animales , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glutatión Peroxidasa/metabolismo , Pulmón/citología , Pulmón/metabolismo , Ratones , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Tiorredoxina Reductasa 1/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 309(5): L441-8, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26138643

RESUMEN

We have previously shown that an adverse perinatal environment significantly alters lung growth and development and results in persistently altered cardiopulmonary physiology in adulthood. Our model of maternal LPS treatment followed by 14 days of neonatal hyperoxia exposure causes severe pulmonary disease characterized by permanent decreases in alveolarization and diffuse interstitial fibrosis. The current investigations tested the hypothesis that dysregulation of Notch signaling pathways contributes to the permanently altered lung phenotype in our model and that the improvements we have observed previously with maternal docosahexaenoic acid (DHA) supplementation are mediated through normalization of Notch-related protein expression. Results indicated that inflammation (IL-6 levels) and oxidation (F2a-isoprostanes) persisted through 8 wk of life in mice exposed to LPS/O2 perinatally. These changes were attenuated by maternal DHA supplementation. Modest but inconsistent differences were observed in Notch-pathway proteins Jagged 1, DLL 1, PEN2, and presenilin-2. We detected substantial increases in markers of apoptosis including PARP-1, APAF-1, caspase-9, BCL2, and HMGB1, and these increases were attenuated in mice that were nursed by DHA-supplemented dams during the perinatal period. Although Notch signaling is not significantly altered at 8 wk of age in mice with perinatal exposure to LPS/O2, our findings indicate that persistent apoptosis continues to occur at 8 wk of age. We speculate that ongoing apoptosis may contribute to persistently altered lung development and may further enhance susceptibility to additional pulmonary disease. Finally, we found that maternal DHA supplementation prevented sustained inflammation, oxidation, and apoptosis in our model.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Inflamación/tratamiento farmacológico , Enfermedades Pulmonares/tratamiento farmacológico , Pulmón/patología , Receptores Notch/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Hipoxia de la Célula/fisiología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/uso terapéutico , Femenino , Proteína HMGB1/metabolismo , Hiperoxia/patología , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , Proteína Jagged-1 , Lipopolisacáridos , Enfermedades Pulmonares/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Estrés Oxidativo/efectos de los fármacos , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos
6.
J Nutr ; 144(3): 258-66, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24453131

RESUMEN

The preterm infant is often exposed to maternal and neonatal inflammatory stimuli and is born with immature lungs, resulting in a need for oxygen therapy. Nutritional intervention with docosahexaenoic acid (DHA; 6.3 g/kg of diet) has been shown to attenuate inflammation in various human diseases. Previous studies demonstrated that maternal DHA supplementation during late gestation and lactation attenuated hyperoxic lung injury in newborn mouse pups. In the present studies, we tested the hypothesis that DHA supplementation to the dam would reduce hyperoxic lung injury and growth deficits in a more severe model of systemic maternal inflammation, including lipopolysaccharide (LPS) and neonatal hyperoxia exposure. On embryonic day 16, dams were placed on DHA (6.3 g DHA/kg diet) or control diets and injected with saline or LPS. Diets were maintained through weaning. At birth, pups were placed in room air or hyperoxia for 14 d. Improvements in birth weight (P < 0.01), alveolarization (P ≤ 0.01), and pulmonary function (P ≤ 0.03) at 2 and 8 wk of age were observed in pups exposed to perinatal inflammation and born to DHA-supplemented dams compared with control diet-exposed pups. These improvements were associated with decreases in tissue macrophage numbers (P < 0.01), monocyte chemoattractant protein-1 expression (P ≤ 0.05), and decreases in soluble receptor for advanced glycation end products concentrations (P < 0.01) at 2 and 8 wk. Furthermore, DHA supplementation attenuated pulmonary fibrosis, which was associated with the reduction of matrix metalloproteinases 2, 3, and 8 (P ≤ 0.03) and collagen mRNA (P ≤ 0.05), and decreased collagen (P < 0.01) and vimentin (P ≤ 0.03) protein concentrations. In a model of severe inflammation, maternal DHA supplementation lessened inflammation and improved lung growth in the offspring. Maternal supplementation with DHA may be a therapeutic strategy to reduce neonatal inflammation.


Asunto(s)
Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Retardo del Crecimiento Fetal/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Animales Recién Nacidos , Peso al Nacer , Quimiocina CCL2/metabolismo , Dieta , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Hiperoxia/tratamiento farmacológico , Lipopolisacáridos/efectos adversos , Pulmón/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 8 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Fosforilación , ARN Mensajero/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
7.
J Nutr ; 141(2): 214-22, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21178083

RESUMEN

DHA is a long-chain fatty acid that has potent antiinflammatory properties. Whereas maternal DHA dietary supplementation has been shown to improve cognitive development in infants fed DHA-supplemented milk, the antiinflammatory effects of maternal DHA supplementation on the developing fetus and neonate have not been extensively explored. Pregnant C3H/HeN dams were fed purified control or DHA-supplemented diets (~0.25% of total fat) at embryonic d 16 and consumed these diets throughout the study. At birth, the nursing mouse pups were placed in room air (RA; 21% O(2)) or >95% O(2) (hyperoxia) for up to 7 d. These studies tested the hypothesis that maternal DHA supplementation would decrease inflammation and improve alveolarization in the lungs of newborn mouse pups exposed to hyperoxia. Survival, inflammatory responses, and lung growth were compared among control diet/RA, DHA/RA, control/O(2), and DHA/O(2) pups. There were fewer neutrophils and macrophages in lung tissues from pups nursed by DHA-supplemented dams than in those nursed by dams fed the control diet at 7 d of hyperoxia exposure (P < 0.015). Although differences due to hyperoxia exposure were observed, maternal diet did not affect keratinocyte-derived chemokine, macrophage inflammatory protein-2, IL-1ß, or TNFα mRNA levels in pup tissues. Hyperoxia also induced NF-κB activity, but maternal diet did not affect NF-κB or PPARγ activities. In mice, DHA supplementation decreases leukocyte infiltration in the offspring exposed to hyperoxia, suggesting a potential role for DHA supplementation as a therapy to reduce inflammation in preterm infants.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ácidos Docosahexaenoicos/uso terapéutico , Hiperoxia , Pulmón/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neumonía/tratamiento farmacológico , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Recuento de Células , Grasas de la Dieta/administración & dosificación , Suplementos Dietéticos , Ácidos Docosahexaenoicos/farmacología , Femenino , Pulmón/inmunología , Pulmón/fisiología , Macrófagos , Ratones , Ratones Endogámicos C3H , Neutrófilos , Fagocitos , Neumonía/etiología , Neumonía/inmunología , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal/inmunología , Alveolos Pulmonares/efectos de los fármacos
8.
Exp Lung Res ; 37(3): 155-61, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21128861

RESUMEN

The aims of this study were to test the hypothesis that mice expressing mitochondrially targeted human glutathione reductase (GR) driven by a surfactant protein C promoter ((spc-mt)hGR) are functionally riboflavin deficient and that this deficiency exacerbates hyperoxic lung injury. The authors further hypothesized that dietary supplementation with riboflavin (FADH) will improve the bioactivity of GR, thus enhancing resistance to hyperoxic lung injury. Transgenic (mt-spc)hGR mice and their nontransgenic littermates were fed control or riboflavin-supplemented diets upon weaning. At 6 weeks of age the mice were exposed to either room air (RA) or >95% O(2) for up to 84 hours. GR activities (with and without exogenous FADH) and GR protein levels were measured in lung tissue homogenates. Glutathione (GSH) and glutathione disulfide (GSSG) concentrations were assayed to identify changes in GR activity in vivo. Lung injury was assessed by right lung to body weight ratios and bronchoalveolar lavage protein concentrations. The data showed that enhanced GR activity in the mitochondria of lung type II cells does not protect adult mice from hyperoxic lung injury. Furthermore, the addition of riboflavin to the diets of (spc-mt)hGR mice neither enhances GR activities nor offers protection from hyperoxic lung injury. The results indicated that modulation of mitochondrial GR activity in lung type II cells is not an effective therapy to minimize hyperoxic lung injury.


Asunto(s)
Glutatión Reductasa/metabolismo , Hiperoxia/prevención & control , Lesión Pulmonar/prevención & control , Riboflavina/administración & dosificación , Animales , Glutatión/metabolismo , Glutatión Reductasa/genética , Humanos , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lesión Pulmonar/complicaciones , Lesión Pulmonar/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteína C Asociada a Surfactante Pulmonar/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deficiencia de Riboflavina/complicaciones , Deficiencia de Riboflavina/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA