Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Antioxid Redox Signal ; 21(13): 1823-36, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25089378

ABSTRACT

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


Subject(s)
Epithelial Cells/metabolism , Hyperoxia/pathology , Large Neutral Amino Acid-Transporter 1/metabolism , Nitric Oxide/metabolism , Pulmonary Alveoli/metabolism , Animals , Biological Transport/physiology , Cell Line, Tumor , Cysteine/analogs & derivatives , Cysteine/metabolism , Epithelial Cells/pathology , Guanylate Cyclase/metabolism , Humans , Hyperoxia/metabolism , Leucine/metabolism , Male , Oxidative Stress/physiology , Pulmonary Alveoli/pathology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , S-Nitrosothiols/metabolism , Soluble Guanylyl Cyclase
2.
Am J Respir Cell Mol Biol ; 47(1): 37-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22323364

ABSTRACT

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


Subject(s)
Lung/blood supply , Lung/metabolism , Nitric Oxide/pharmacology , S-Nitrosothiols/metabolism , Vasodilation , Administration, Inhalation , Amino Acids, Cyclic/pharmacology , Animals , Biological Transport, Active , Cysteine/administration & dosage , Cysteine/pharmacology , Female , Hypertension, Pulmonary/drug therapy , Leucine/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...