Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pulm Med ; 2011: 918036, 2011.
Article in English | MEDLINE | ID: mdl-21876799

ABSTRACT

WE COMPARED LUNG DELIVERY METHODS OF RECOMBINANT ADENOVIRUS (RAD): (1) rAd suspended in saline, (2) rAd suspended in saline followed by a pulse-chase of a perfluorochemical (PFC) liquid mixture, and (3) a PFC-rAd suspension. Cell uptake, distribution, and temporal expression of rAd were examined using A549 cells, a murine model using luciferase bioluminescence, and histological analyses. Relative to saline, a 4X increase in transduction efficiency was observed in A549 cells exposed to PFC-rAd for 2-4 h. rAd transgene expression was improved in alveolar epithelial cells, and the level and distribution of luciferase expression when delivered in PFC-rAd suspensions consistently peaked at 24 h. These results demonstrate that PFC-rAd suspensions improve distribution and enhance rAd-mediated gene expression which has important implications in improving lung function by gene therapy.

2.
Free Radic Biol Med ; 45(8): 1143-9, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18692129

ABSTRACT

Exposure of lung epithelial cells to hyperoxia results in the generation of excess reactive oxygen species (ROS), cell damage, and production of proinflammatory cytokines (interleukin-8; IL-8). Although activation of the NF-kappaB and c-Jun N-terminal kinase (JNK)/activator protein (AP)-1 transcription pathways occurs in hyperoxia, it is unclear whether activation of the AP-1 pathway has a direct impact on IL-8 production and whether overexpression of superoxide dismutase (SOD) can mitigate these proinflammatory processes. A549 cells were exposed to 95% O(2), and ROS production, AP-1 activation, and IL-8 levels were determined. Experimental groups included cells transduced with a recombinant adenovirus encoding CuZnSOD or MnSOD (two- to threefold increased activity) or transfected with a JNK1 small interfering RNA (RNAi). Hyperoxia resulted in significant increases in ROS generation, AP-1 activation, and IL-8 production, which were significantly attenuated by overexpression of either MnSOD or CuZnSOD. JNK1 RNAi also moderated IL-8 induction. The data indicate that activation of JNK1/AP-1 and subsequent IL-8 induction in hyperoxia are mediated by intracellular ROS, with SOD having significant protective effects.


Subject(s)
Hyperoxia/metabolism , Interleukin-8/metabolism , Oxidative Stress/physiology , Superoxide Dismutase/metabolism , Transcription Factor AP-1/metabolism , Blotting, Western , Cell Line, Tumor , Enzyme Activation/physiology , Epithelial Cells/metabolism , Humans , Lung/metabolism , Reactive Oxygen Species/metabolism , Transfection
3.
Free Radic Biol Med ; 42(10): 1517-23, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17448898

ABSTRACT

Hyperoxia and pulmonary infections are well known to increase the risk of acute and chronic lung injury in newborn infants, but it is not clear whether hyperoxia directly increases the risk of pneumonia. The purpose of this study was to examine: (1) the effects of hyperoxia and antioxidant enzymes on inflammation and bacterial clearance in mononuclear cells and (2) developmental differences between adult and neonatal mononuclear cells in response to hyperoxia. Mouse macrophages were exposed to either room air or 95% O2 for 24 h and then incubated with Pseudomonas aeruginosa. After 1 h, bacterial adherence, phagocytosis, and macrophage inflammatory protein (MIP)-1alpha production were analyzed. Bacterial adherence increased 5.8-fold (p < 0.0001), phagocytosis decreased 60% (p < 0.05), and MIP-1alpha production increased 49% (p < 0.05) in response to hyperoxia. Overexpression of MnSOD or catalase significantly decreased bacterial adherence by 30.5%, but only MnSOD significantly improved bacterial phagocytosis and attenuated MIP-1alpha production. When monocytes from newborns and adults were exposed to hyperoxia, phagocytosis was impaired in both groups. However, adult monocytes were significantly more impaired than neonatal monocytes. Data indicate that hyperoxia significantly increases bacterial adherence while impairing function of mononuclear cells, with adult cells being more impaired than neonatal cells. MnSOD reduces bacterial adherence and inflammation and improves bacterial phagocytosis in mononuclear cells in response to hyperoxia, which should minimize the development of oxidant-induced lung injury as well as reducing nosocomial infections.


Subject(s)
Antioxidants/metabolism , Hyperoxia/immunology , Macrophages/immunology , Oxidoreductases/metabolism , Phagocytosis , Pseudomonas aeruginosa/immunology , Animals , Cell Adhesion , Chemokine CCL3 , Chemokine CCL4 , Macrophage Inflammatory Proteins/metabolism , Macrophages/enzymology , Macrophages/microbiology , Mice , Superoxide Dismutase/metabolism
4.
Pediatr Res ; 60(1): 65-70, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16690961

ABSTRACT

Previous studies suggest acute lung injury (ALI) in premature newborns is associated with relative deficiency of antioxidant enzymes that may be ameliorated by recombinant human superoxide dismutase (rhSOD). Perfluorochemicals (PFCs) are distributed homogeneously and support gas exchange in diseased lungs. We investigated whether PFCs could provide an effective delivery system for rhSOD. Juvenile rabbits were lung-lavaged, treated with surfactant, and randomized: group I: fluorescently labeled rhSOD (5 mg/kg in 2 mL/kg saline); group II: fluorescently labeled rhSOD (5 mg/kg in 18 mL/kg PFC). Animals were ventilated with oxygen for 4 h; the lungs were harvested for analysis of SOD distribution and oxidative injury. Cardiopulmonary indices remained stable and similar between groups. Qualitative assessment (QA) showed a more homogeneous lung SOD distribution in group II and a better histologic profile. QA of lung SOD distribution showed significant increase in SOD concentrations in group II (7.37 +/- 1.54 microg/mg protein) compared with group I (1.65 +/- 0.23 microg/mg protein). Oxidative injury as assessed by normalized protein carbonyl was 149.1 +/- 26.8% SEM in group II compared with 200.5 +/- 7.3% SEM in group I. Plasma SOD was significantly higher in group II. Administration of rhSOD with or without PFCs does not compromise cardiovascular function or impede lung recovery after ALI. PFCs enhance rhSOD delivery to the lungs by 400% while decreasing lung oxidative damage by 25% compared with rhSOD alone. These data suggest that PFCs optimize lung rhSOD delivery and might enhance the beneficial effects of rhSOD in preventing acute and chronic lung injury.


Subject(s)
Animals, Newborn/metabolism , Fluorocarbons/pharmacology , Lung/metabolism , Recombinant Proteins/pharmacokinetics , Superoxide Dismutase/pharmacokinetics , Animals , Biological Transport/drug effects , Drug Delivery Systems , Lung/enzymology , Lung/physiopathology , Lung Diseases/enzymology , Lung Diseases/therapy , Oxidative Stress/physiology , Protein Carbonylation , Pulmonary Gas Exchange/drug effects , Rabbits , Random Allocation , Superoxide Dismutase/analysis , Superoxide Dismutase/blood
5.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L978-86, 2006 May.
Article in English | MEDLINE | ID: mdl-16387755

ABSTRACT

Reactive oxygen species (ROS) can cause cell injury and death via mitochondrial-dependent pathways, and supplementation with antioxidants has been shown to ameliorate these processes. The c-Jun NH(2)-terminal kinase (JNK) pathway has been shown to play a critical role in ROS-induced cell death. To determine if targeting catalase (CAT) to the mitochondria provides better protection than cytosolic expression against H(2)O(2)-induced injury, the following two approaches were taken: 1) adenoviral-mediated transduction was performed using cytosolic (CCAT) or mitochondrial (MCAT) CAT cDNAs and 2) stable cell lines were generated overexpressing CAT in mitochondria (n = 3). Cells were exposed to 250 microM H(2)O(2), and cell survival, mitochondrial function, cytochrome c release, and JNK activity were analyzed. Although all viral transduced cells had a transient twofold increase in CAT activity, MCAT cells had significantly higher survival rates, the best mitochondrial function, and lowest JNK activity compared with CCAT and LacZ controls. The improved protection with MCAT was observed in primary type II lung epithelial cells and in transformed lung epithelial cells. In the three stable cell lines, cell survival directly correlated with extent of mitochondrial localization (r = 0.60572, P < 0.05) and not overall CAT activity (r = -0.45501, P < 0.05). Data indicate that targeting of antioxidants directly to the mitochondria is more effective in protecting lung epithelial cells against ROS-induced injury. This has important implications in antioxidant supplementation trials to prevent ROS-induced lung injury in critically ill patients.


Subject(s)
Catalase/metabolism , Cell Death/drug effects , Hydrogen Peroxide/toxicity , Lung/enzymology , Mitochondria/enzymology , Respiratory Mucosa/enzymology , Animals , Catalase/genetics , Cytosol/enzymology , Genetic Vectors , Humans , Lung/drug effects , Lung/pathology , Male , Mitochondria/drug effects , Mitochondria/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology
6.
Am J Physiol Lung Cell Mol Physiol ; 288(4): L718-26, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15579623

ABSTRACT

Prolonged exposure to supraphysiological oxygen concentrations results in the generation of reactive oxygen species, which can cause significant lung injury in critically ill patients. Supplementation with human recombinant antioxidant enzymes (AOE) may mitigate hyperoxic lung injury, but it is unclear which combination and concentration will optimally protect pulmonary epithelial cells. First, stable cell lines were generated in alveolar epithelial cells (MLE12) overexpressing one or more of the following AOE: Mn superoxide dismutase (MnSOD), CuZnSOD, or glutathione peroxidase 1. Next, A549 cells were transduced with 50-300 particles/cell of recombinant adenovirus containing either LacZ or each of the three AOE (alone or in combination). Cells were then exposed to 95% O(2) for up to 3 days, with cell number and viability determined daily. Overexpression of either MnSOD (primarily mitochondrial) or CuZnSOD (primarily cytosolic) reversed the growth inhibitory effects of hyperoxia within the first 48 h of exposure, resulting in a significant increase in viable cells (P < 0.05), with 1.5- to 3-fold increases in activity providing optimal protection. Protection from mitochondrial oxidation was confirmed by assessing aconitase activity, which was significantly improved in cells overexpressing MnSOD (P < 0.05). Data indicate that optimal protection from hyperoxic injury occurs in cells coexpressing MnSOD and glutathione peroxidase 1, with prevention of mitochondrial oxidation being a critical factor. This has important implications for clinical trials in preterm infants receiving SOD supplementation to prevent acute and chronic lung injury.


Subject(s)
Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/physiology , Glutathione Peroxidase/genetics , Hyperoxia/physiopathology , Lung/cytology , Pulmonary Alveoli/enzymology , Superoxide Dismutase/genetics , Transgenes/physiology , Aconitate Hydratase/metabolism , Adenoviridae/genetics , Antioxidants/metabolism , Cell Proliferation , HeLa Cells , Humans , Hydrogen Peroxide , Lung/enzymology , Mitochondria/metabolism , Recombinant Proteins/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 287(6): L1199-206, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15286004

ABSTRACT

Bacterial infection of the tracheobronchial tree is a frequent, serious complication in patients receiving treatment with oxygen and mechanical ventilation, resulting in increased morbidity and mortality. Using human airway epithelial cell culture models, we examined the effect of hyperoxia on bacterial adherence and the expression of interleukin-8 (IL-8), an important mediator involved in the inflammatory process. A 24-h exposure to 95% O(2) increased Pseudomonas aeruginosa (PA) adherence 57% in A549 cells (P < 0.01) and 115% in 16HBE cells (P < 0.01) but had little effect on Staphylococcus aureus (SA) adherence. Exposure to hyperoxia, followed by a 1-h incubation with SA, further enhanced PA adherence (P < 0.01), suggesting that hyperoxia and SA colonization may enhance the susceptibility of lung epithelial cells to gram-negative infections. IL-8 expression was also increased in cells exposed to both hyperoxia and PA. Stable or transient overexpression of manganese superoxide dismutase reduced both basal and stimulated levels of PA adherence and IL-8 levels in response to exposure to either hyperoxia or PA. These data indicate that hyperoxia increases susceptibility to infection and that the pathways are mediated by reactive oxygen species. Therapeutic intervention strategies designed to prevent accumulation of intracellular reactive oxygen species may reduce opportunistic pulmonary infections.


Subject(s)
Bacterial Adhesion/physiology , Interleukin-8/genetics , Pseudomonas aeruginosa/physiology , Respiratory Mucosa/physiology , Superoxide Dismutase/metabolism , Adenocarcinoma , Cell Line, Tumor , Humans , Hyperoxia , Mitochondria/enzymology , Oxidoreductases/metabolism , Recombinant Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Staphylococcus aureus/physiology , Superoxide Dismutase/genetics , Transfection
8.
Am J Respir Cell Mol Biol ; 29(6): 779-83, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12842852

ABSTRACT

Oxidant insults can lead to apoptotic and nonapoptotic cell death. Lung epithelial cells exposed to high levels of oxygen do not die via apoptosis, but through a much slower, morphologically distinct process involving cell and nuclear swelling. In contrast, H2O2 induces a rapid apoptotic cell death. We first assessed the effect of oxidant exposure on activator protein-1 (c-Jun and Fos) and c-Jun N-terminal kinase (JNK) regulation in MLE12 cells. Both oxidants induced c-Jun and Fos expression, albeit with a different pattern of regulation-hyperoxia (95% O2) induced a biphasic response, whereas H2O2 (500 microM) induced a sustained response. We then examined the role of JNK by Western blot, JNK activity assay, and a pull-down assay and observed an identical pattern of regulation. To assess whether JNK functions in a pro-death or pro-survival capacity, we generated stable cell lines that constitutively express a dominant-negative mutation of JNK resulting in significant inhibition of JNK activity. Inhibition of the JNK pathway in this manner prevented hyperoxic and H2O2-induced cell death. These results demonstrate that hyperoxic cell death is pathway-driven and that both modes of death involve the JNK signaling pathway.


Subject(s)
Cell Survival/physiology , Hydrogen Peroxide/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Oxidants/metabolism , Animals , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinases/genetics , Oxygen/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transcription Factor AP-1/metabolism
9.
Pediatr Res ; 54(4): 509-15, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12815115

ABSTRACT

Despite the widespread use of exogenous surfactant, acute and chronic lung injury continues to be a major cause of morbidity in preterm infants. CC10 is a protein produced by Clara cells that inhibits phospholipase A2 and has anti-inflammatory and antifibrotic properties. We studied whether intratracheal (IT) recombinant human Clara cell protein (rhCC10) could safely minimize lung injury in a newborn piglet model of acute lung injury. Twenty-nine newborn piglets were given Survanta and then ventilated for 48 h receiving the following: room air (group 1); 100% O2 (group 2); or 100% O2 and 25, 5, or 1 mg/kg (groups 3, 4, and 5, respectively) of IT rhCC10 (diluted to 2 mL/kg with saline) at time 0. Laboratory studies, oxygen ratios, static pressure-volume curves, bronchoalveolar lavage (for inflammatory markers), and histologic analyses were performed over the 48-h study period. Pulmonary compliance and oxygenation were significantly improved in animals receiving 5 mg/kg IT rhCC10 compared with room air and 100% O2 controls (p < 0.004 and p < 0.05, respectively, ANOVA). Reductions in inflammatory markers were seen in animals receiving rhCC10, although changes did not reach statistical significance. No significant toxicity was noted. rhCC10 appeared safe and improved pulmonary function in this newborn piglet model of hyperoxic lung injury. We speculate that rhCC10 may represent a promising therapy for the prevention of lung injury in preterm infants.


Subject(s)
Animals, Newborn , Enzyme Inhibitors/therapeutic use , Recombinant Proteins/therapeutic use , Respiratory Distress Syndrome/drug therapy , Swine , Administration, Inhalation , Animals , Biological Products/administration & dosage , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Humans , Infant, Newborn , Lung Compliance , Recombinant Proteins/administration & dosage , Respiratory Physiological Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL
...