Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L312-22, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20382751

ABSTRACT

Bcl-2 is an anti-apoptotic molecule preventing oxidative stress damage and cell death. We have previously shown that Bcl-2 is able to prevent hyperoxia-induced cell death when overexpressed in a murine fibrosarcoma cell line L929. We hypothesized that its specific overexpression in pulmonary epithelial type II cells could prevent hyperoxia-induced lung injury by protecting the epithelial side of the alveolo-capillary barrier. In the present work, we first showed that in vitro Bcl-2 can rescue murine pulmonary epithelial cells (MLE12) from oxygen-induced cell apoptosis, as shown by analysis of LDH release, annexin V/propidium staining, and caspase-3 activity. We then generated transgenic mice overexpressing specifically Bcl-2 in lung epithelial type II cells under surfactant protein C (SP-C) promoter (Tg-Bcl-2) and exposed them to hyperoxia. Bcl-2 did not hinder hyperoxia-induced mitochondria and DNA oxidative damage of type II cell in vivo. Accordingly, lung damage was identical in both Tg-Bcl-2 and littermate mice strains, as measured by lung weight, bronchoalveolar lavage, and protein content. Nevertheless, we observed a significant lower number of TUNEL-positive cells in type II cells isolated from Tg-Bcl-2 mice exposed to hyperoxia compared with cells isolated from littermate mice. In summary, these results show that although Bcl-2 overexpression is able to prevent hyperoxia-induced cell death at single cell level in vitro and ex vivo, it is not sufficient to prevent cell death of parenchymal cells and to protect the lung from acute damage in mice.


Subject(s)
Acute Lung Injury/prevention & control , Epithelial Cells/metabolism , Hyperoxia/complications , Hyperoxia/metabolism , Lung/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Acute Lung Injury/etiology , Animals , Apoptosis , Cell Death , Cells, Cultured , DNA Damage , Epithelial Cells/classification , Hyperoxia/pathology , Hyperoxia/physiopathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation
2.
Am J Respir Crit Care Med ; 180(10): 972-81, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19661248

ABSTRACT

RATIONALE: Hyperoxia-induced acute lung injury has been used for many years as a model of oxidative stress mimicking clinical acute lung injury and the acute respiratory distress syndrome. Excess quantities of reactive oxygen species (ROS) are responsible for oxidative stress-induced lung injury. ROS are produced by mitochondrial chain transport, but also by NADPH oxidase (NOX) family members. Although NOX1 and NOX2 are expressed in the lungs, their precise function has not been determined until now. OBJECTIVES: To determine whether NOX1 and NOX2 contribute in vivo to hyperoxia-induced acute lung injury. METHODS: Wild-type and NOX1- and NOX2-deficient mice, as well as primary lung epithelial and endothelial cells, were exposed to room air or 100% O(2) for 72 hours. MEASUREMENTS AND MAIN RESULTS: Lung injury was significantly prevented in NOX1-deficient mice, but not in NOX2-deficient mice. Hyperoxia-dependent ROS production was strongly reduced in lung sections, in isolated epithelial type II cells, and lung endothelial cells from NOX1-deficient mice. Concomitantly, lung cell death in situ and in primary cells was markedly decreased in NOX1-deficient mice. In wild-type mice, hyperoxia led to phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), two mitogen-activated protein kinases involved in cell death signaling, and to caspase-3 activation. In NOX1-deficient mice, JNK phosphorylation was blunted, and ERK phosphorylation and caspase-3 activation were decreased. CONCLUSIONS: NOX1 is an important contributor to ROS production and cell death of the alveolocapillary barrier during hyperoxia and is an upstream actor in oxidative stress-induced acute lung injury involving JNK and ERK pathways in mice.


Subject(s)
Hypoxia/complications , Lung Injury/enzymology , NADPH Oxidases/physiology , Animals , Cell Death/physiology , Endothelium/cytology , Epithelial Cells/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/cytology , Lung Injury/etiology , Mice , Mice, Inbred C57BL , NADPH Oxidases/deficiency , Phosphorylation , Reactive Oxygen Species/metabolism
3.
Free Radic Biol Med ; 42(7): 1062-74, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17349933

ABSTRACT

Bcl-2 is an antiapoptotic molecule that prevents oxidative stress damage and cell death. We investigated the possible protective mechanisms mediated by Bcl-2 during hyperoxia-induced cell death in L929 cells. In these cells, hyperoxia promoted apoptosis without DNA fragmentation. Overexpression of Bcl-2 significantly protected cells from oxygen-induced apoptosis, as shown by measurement of lactate dehydrogenase release, quantification of apoptotic nuclei, and detection of Annexin-V-positive cells. Bcl-2 partially prevented mitochondrial damage and interfered with the mitochondrial proapoptotic signaling pathway: it reduced Bax translocation to mitochondria, decreased the release of cytochrome c, and inhibited caspase 3 activation. However, treatment with the caspase inhibitor Z-VAD.fmk failed to rescue the cells from death, indicating that protection provided by Bcl-2 was due not only to caspase inhibition. Bcl-2 also prevented the release of mitochondrial apoptotic inducing factor, a mediator of caspase-independent apoptosis, correlating with the absence of oligonucleosomal DNA fragmentation. In addition, Bcl-2-overexpressing cells showed significantly higher intracellular amounts of glutathione after 72 h of oxygen exposure. In conclusion, our results demonstrate that the overexpression of Bcl-2 is able to prevent hyperoxia-induced cell death, by affecting mitochondria-dependent apoptotic pathways and increasing intracellular antioxidant compounds.


Subject(s)
Apoptosis/physiology , Hyperoxia/physiopathology , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Caspase Inhibitors , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , Glutathione/metabolism , Immunohistochemistry , Mice , Microscopy, Electron , Reactive Oxygen Species/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L619-29, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17575013

ABSTRACT

Oxygen-based therapies expose lung to elevated levels of ROS and induce lung cell damage and inflammation. Injured cells are replaced through increased proliferation and differentiation of epithelial cells and fibroblasts. Failure to modulate these processes leads to excessive cell proliferation, collagen deposition, fibrosis, and chronic lung disease. Poly(ADP-ribose) polymerase-1 (PARP-1) is activated in response to DNA damage and participates in DNA repair, genomic integrity, and cell death. In this study, we evaluated the role of PARP-1 in lung repair during recovery after acute hyperoxia exposure. We exposed PARP-1 -/- and wild-type mice for 64 h to 100% hyperoxia and let them recover in air for 5-21 days. PARP-1-deficient mice exhibited significantly higher lung cell hyperplasia and proliferation than PARP-1 +/+ animals after 5 and 10 days of recovery. This was accompanied by an increased inflammatory response in PARP-1 -/- compared with wild-type animals, characterized by neutrophil infiltration and increased IL-6 levels in bronchoalveolar lavages. These lesions were reversible, since the extent of the hyperplastic regions was reduced after 21 days of recovery and did not result in fibrosis. In vitro, lung primary fibroblasts derived from PARP-1 -/- mice showed a higher proliferative response than PARP-1 +/+ cells during air recovery after hyperoxia-induced growth arrest. Altogether, these results reveal an essential role of PARP-1 in the control of cell repair and tissue remodeling after hyperoxia-induced lung injury.


Subject(s)
Hyperoxia/pathology , Lung/enzymology , Lung/pathology , Poly(ADP-ribose) Polymerases/metabolism , Wound Healing , Air , Animals , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/pathology , Fibrosis , Hyperoxia/chemically induced , Hyperplasia , Immunohistochemistry , Inflammation , Lung/cytology , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/deficiency
5.
Am J Respir Cell Mol Biol ; 33(6): 555-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16151053

ABSTRACT

Hyperoxia induces extensive DNA damage and lung cell death by apoptotic and nonapoptotic pathways. We analyzed the regulation of Poly(ADP-ribose)polymerase-1 (PARP-1), a nuclear enzyme activated by DNA damage, and its relation to cell death during hyperoxia in vitro and in vivo. In lung epithelial-derived A549 cells, which are known to die by necrosis when exposed to oxygen, a minimal amount of PARP-1 was cleaved, correlating with the absence of active caspase-3. Conversely, in primary lung fibroblasts, which die mainly by apoptosis, the complete cleavage of PARP-1 was concomitant to the induction of active caspase-3, as assessed by Western blot and caspase activity. Blockade of caspase activity by Z-VAD reduced the amount of cleaved PARP-1 in fibroblasts. Hyperoxia induced PARP activity in both cell types, as revealed by poly-ADP-ribose accumulation. In A549 cells, the final outcome of necrosis was dependent on PARP activity because it was prevented by the PARP inhibitor 3-aminobenzamide. In contrast, apoptosis of lung fibroblasts was not sensitive to 3-aminobenzamide and was not affected by PARP-1 deletion. In vivo, despite evidence of PARP activation in hyperoxia-exposed mouse lungs, absence of PARP-1 did not change the extent of lung damage, arguing for redundant oxidative stress-induced cell death pathways.


Subject(s)
Apoptosis , Epithelial Cells/pathology , Hyperoxia/enzymology , Lung/pathology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Benzamides/pharmacology , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation , Epithelial Cells/enzymology , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL