1
|
Guo H, Huang RR, Qu SS, Yao Y, Chen SH, Ding SL, Li YL. FAM134B deletion exacerbates apoptosis and epithelial-to-mesenchymal transition in rat lungs exposed to hyperoxia. iScience 2024; 27:110385. [PMID: 39092177 PMCID: PMC11292547 DOI: 10.1016/j.isci.2024.110385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/21/2023] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
Oxygen therapy is widely used in clinical practice; however, prolonged hyperoxia exposure may result in hyperoxic acute lung injury (HALI). In this study, we investigated the role of FAM134B in hyperoxia-induced apoptosis, cell proliferation, and epithelial-to-mesenchymal transition (EMT) using RLE-6TN cells and rat lungs. We also studied the effect of CeO2-NPs on RLE-6TN cells and lungs following hyperoxia exposure. FAM134B was inhibited in RLE-6TN cells and rat lungs following hyperoxia exposure. Overexpressing FAM134B promoted cell proliferation, and reduced EMT and apoptosis following hyperoxia exposure. FAM134B activation increased ER-phagy, decreased apoptosis, improved lung structure damage, and decreased collagen fiber deposition to limit lung injury. These effects could be reversed by PI3K/AKT pathway inhibitor LY294002. Additionally, CeO2-NPs protected RLE-6TN cells and lung damage following hyperoxia exposure by ameliorating impaired ER-phagy. Therefore, FAM134B restoration is a potential therapeutic target for the HALI. Moreover, CeO2-NPs can be used for the treatment of HALI.
Collapse
Affiliation(s)
- Hong Guo
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of Anesthesiology, Inner Mongolia Hospital of Peking University Cancer Hospital, Affiliated People's Hospital , Inner Mongolia Medical University, Hohhot 10020, China
| | - Rong-Rong Huang
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shan-Shan Qu
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Ying Yao
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Su-Heng Chen
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shao-Li Ding
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yu-Lan Li
- First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
2
|
Harris ZM, Sun Y, Joerns J, Clark B, Hu B, Korde A, Sharma L, Shin HJ, Manning EP, Placek L, Unutmaz D, Stanley G, Chun H, Sauler M, Rajagopalan G, Zhang X, Kang MJ, Koff JL. Epidermal Growth Factor Receptor Inhibition Is Protective in Hyperoxia-Induced Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9518592. [PMID: 36193076 PMCID: PMC9526641 DOI: 10.1155/2022/9518592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 01/01/2023]
Abstract
Aims Studies have linked severe hyperoxia, or prolonged exposure to very high oxygen levels, with worse clinical outcomes. This study investigated the role of epidermal growth factor receptor (EGFR) in hyperoxia-induced lung injury at very high oxygen levels (>95%). Results Effects of severe hyperoxia (100% oxygen) were studied in mice with genetically inhibited EGFR and wild-type littermates. Despite the established role of EGFR in lung repair, EGFR inhibition led to improved survival and reduced acute lung injury, which prompted an investigation into this protective mechanism. Endothelial EGFR genetic knockout did not confer protection. EGFR inhibition led to decreased levels of cleaved caspase-3 and poly (ADP-ribosyl) polymerase (PARP) and decreased terminal dUTP nick end labeling- (TUNEL-) positive staining in alveolar epithelial cells and reduced ERK activation, which suggested reduced apoptosis in vivo. EGFR inhibition decreased hyperoxia (95%)-induced apoptosis and ERK in murine alveolar epithelial cells in vitro, and CRISPR-mediated EGFR deletion reduced hyperoxia-induced apoptosis and ERK in human alveolar epithelial cells in vitro. Innovation. This work defines a protective role of EGFR inhibition to decrease apoptosis in lung injury induced by 100% oxygen. This further characterizes the complex role of EGFR in acute lung injury and outlines a novel hyperoxia-induced cell death pathway that warrants further study. Conclusion In conditions of severe hyperoxia (>95% for >24 h), EGFR inhibition led to improved survival, decreased lung injury, and reduced cell death. These findings further elucidate the complex role of EGFR in acute lung injury.
Collapse
Affiliation(s)
- Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - John Joerns
- Division of Pulmonary and Critical Care; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA 75390
| | - Brian Clark
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Asawari Korde
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Edward P. Manning
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Lindsey Placek
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyung Chun
- Section of Cardiovascular Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Govindarajan Rajagopalan
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| |
Collapse
|
3
|
Kundumani-Sridharan V, Subramani J, Raghavan S, Maiti GP, Owens C, Walker T, Wasnick J, Idell S, Das KC. Short-duration hyperoxia causes genotoxicity in mouse lungs: protection by volatile anesthetic isoflurane. Am J Physiol Lung Cell Mol Physiol 2019; 316:L903-L917. [PMID: 30810065 DOI: 10.1152/ajplung.00142.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High concentrations of oxygen (hyperoxia) are routinely used during anesthesia, and supplemental oxygen is also administered in connection with several other clinical conditions. Although prolonged hyperoxia is known to cause acute lung injury (ALI), whether short-duration hyperoxia causes lung toxicity remains unknown. We exposed mice to room air (RA or 21% O2) or 60% oxygen alone or in combination with 2% isoflurane for 2 h and determined the expression of oxidative stress marker genes, DNA damage and DNA repair genes, and expression of cell cycle regulatory proteins using quantitative PCR and Western analyses. Furthermore, we determined cellular apoptosis using TUNEL assay and assessed the DNA damage product 8-hydroxy-2'-deoxyguanosine (8-Oxo-dG) in the urine of 60% hyperoxia-exposed mice. Our study demonstrates that short-duration hyperoxia causes mitochondrial and nuclear DNA damage and that isoflurane abrogates this DNA damage and decreases apoptosis when used in conjunction with hyperoxia. In contrast, isoflurane mixed with RA caused significant 8-Oxo-dG accumulations in the mitochondria and nucleus. We further show that whereas NADPH oxidase is a major source of superoxide anion generated by isoflurane in normoxia, isoflurane inhibits superoxide generation in hyperoxia. Additionally, isoflurane also protected the mouse lungs against ALI (95% O2 for 36-h exposure). Our study established that short-duration hyperoxia causes genotoxicity in the lungs, which is abrogated when hyperoxia is used in conjunction with isoflurane, but isoflurane alone causes genotoxicity in the lung when delivered with ambient air.
Collapse
Affiliation(s)
| | - Jaganathan Subramani
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Somasundaram Raghavan
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Guru P Maiti
- Oklahoma Medical Research Foundation , Oklahoma City, Oklahoma
| | - Cade Owens
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Trevor Walker
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - John Wasnick
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| |
Collapse
|
4
|
Hyperoxia Disrupts Extracellular Signal-Regulated Kinases 1/2-Induced Angiogenesis in the Developing Lungs. Int J Mol Sci 2018; 19:ijms19051525. [PMID: 29783779 PMCID: PMC5983575 DOI: 10.3390/ijms19051525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/17/2022] Open
Abstract
Hyperoxia contributes to the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of infants that is characterized by interrupted alveologenesis. Disrupted angiogenesis inhibits alveologenesis, but the mechanisms of disrupted angiogenesis in the developing lungs are poorly understood. In pre-clinical BPD models, hyperoxia increases the expression of extracellular signal-regulated kinases (ERK) 1/2; however, its effects on the lung endothelial ERK1/2 signaling are unclear. Further, whether ERK1/2 activation promotes lung angiogenesis in infants is unknown. Hence, we tested the following hypotheses: (1) hyperoxia exposure will increase lung endothelial ERK1/2 signaling in neonatal C57BL/6J (WT) mice and in fetal human pulmonary artery endothelial cells (HPAECs); (2) ERK1/2 inhibition will disrupt angiogenesis in vitro by repressing cell cycle progression. In mice, hyperoxia exposure transiently increased lung endothelial ERK1/2 activation at one week of life, before inhibiting it at two weeks of life. Interestingly, hyperoxia-mediated decrease in ERK1/2 activation in mice was associated with decreased angiogenesis and increased endothelial cell apoptosis. Hyperoxia also transiently activated ERK1/2 in HPAECs. ERK1/2 inhibition disrupted angiogenesis in vitro, and these effects were associated with altered levels of proteins that modulate cell cycle progression. Collectively, these findings support our hypotheses, emphasizing that the ERK1/2 pathway is a potential therapeutic target for BPD infants with decreased lung vascularization.
Collapse
|
5
|
Recombinant CCN1 prevents hyperoxia-induced lung injury in neonatal rats. Pediatr Res 2017; 82:863-871. [PMID: 28700567 PMCID: PMC5874130 DOI: 10.1038/pr.2017.160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 05/20/2017] [Indexed: 01/19/2023]
Abstract
BackgroundCystein-rich protein 61 (Cyr61/CCN1) is a member of the CCN family of matricellular proteins that has an important role in tissue development and remodeling. However, the role of CCN1 in the pathogenesis of bronchopulmonary dysplasia (BPD) is unknown. Accordingly, we have investigated the effects of CCN1 on a hyperoxia-induced lung injury model in neonatal rats.MethodsIn experiment 1, newborn rats were randomized to room air (RA) or 85% oxygen (O2) for 7 or 14 days, and we assessed the expression of CCN1. In experiment 2, rat pups were exposed to RA or O2 and received placebo or recombinant CCN1 by daily intraperitoneal injection for 10 days. The effects of CCN1 on hyperoxia-induced lung inflammation, alveolar and vascular development, vascular remodeling, and right ventricular hypertrophy (RVH) were observed.ResultsIn experiment 1, hyperoxia downregulated CCN1 expression. In experiment 2, treatment with recombinant CCN1 significantly decreased macrophage and neutrophil infiltration, reduced inflammasome activation, increased alveolar and vascular development, and reduced vascular remodeling and RVH in the hyperoxic animals.ConclusionThese results demonstrate that hyperoxia-induced lung injury is associated with downregulated basal CCN1 expression, and treatment with CCN1 can largely reverse hyperoxic injury.
Collapse
|
6
|
Porzionato A, Sfriso MM, Mazzatenta A, Macchi V, De Caro R, Di Giulio C. Effects of hyperoxic exposure on signal transduction pathways in the lung. Respir Physiol Neurobiol 2015; 209:106-14. [DOI: 10.1016/j.resp.2014.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 12/18/2022]
|
7
|
Haugen M, Dammen R, Svejda B, Gustafsson BI, Pfragner R, Modlin I, Kidd M. Differential signal pathway activation and 5-HT function: the role of gut enterochromaffin cells as oxygen sensors. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1164-73. [PMID: 22936271 PMCID: PMC3517648 DOI: 10.1152/ajpgi.00027.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The chemomechanosensory function of the gut enterochromaffin (EC) cell enables it to respond to dietary agents and mechanical stretch. We hypothesized that the EC cell, which also sensed alterations in luminal or mucosal oxygen level, was physiologically sensitive to fluctuations in O(2). Given that low oxygen levels induce 5-HT production and secretion through a hypoxia inducible factor 1α (HIF-1α)-dependent pathway, we also hypothesized that increasing O(2) would reduce 5-HT production and secretion. Isolated normal EC cells as well as the well-characterized EC cell model KRJ-I were used to examine HIF signaling (luciferase-assays), hypoxia transcriptional response element (HRE)-mediated transcription (PCR), signaling pathways (Western blot), and 5-HT release (ELISA) during exposure to different oxygen levels. Normal EC cells and KRJ-I cells express HIF-1α, and transient transfection with Renilla luciferase under HRE control identified a hypoxia-mediated pathway in these cells. PCR confirmed activation of HIF-downstream targets, GLUT1, IGF2, and VEGF under reduced O(2) levels (0.5%). Reducing O(2) also elevated 5-HT secretion (2-3.2-fold) as well as protein levels of HIF-1α (1.7-3-fold). Increasing O(2) to 100% inhibited HRE-mediated signaling, transcription, reduced 5-HT secretion, and significantly lowered HIF-1α levels (∼75% of control). NF-κB signaling was also elevated during hypoxia (1.2-1.6-fold), but no significant changes were noted in PKA/cAMP. We concluded that gut EC cells are oxygen responsive, and alterations in O(2) levels differentially activate HIF-1α and tryptophan hydroxylase 1, as well as NF-κB signaling. This results in alterations in 5-HT production and secretion and identifies that the chemomechanosensory role of EC cells extends to oxygen sensing.
Collapse
Affiliation(s)
- Martin Haugen
- 1Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut;
| | - Rikard Dammen
- 1Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut;
| | - Bernhard Svejda
- 1Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut;
| | - Bjorn I. Gustafsson
- 2Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway;
| | - Roswitha Pfragner
- 3Institute of Pathophysiology and Immunology, Centre for Molecular Medicine, Graz, Austria
| | - Irvin Modlin
- 1Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut;
| | - Mark Kidd
- 1Gastrointestinal Pathobiology Research Group, Yale University School of Medicine, New Haven, Connecticut;
| |
Collapse
|
8
|
Xu A, Prophete C, Chen LC, Emala CW, Cohen MD. Interactive effect of cigarette smoke extract and world trade center dust particles on airway cell cytotoxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2011; 74:887-902. [PMID: 21623534 DOI: 10.1080/15287394.2011.573719] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Rescue workers and residents exposed to the environment surrounding the collapse of the World Trade Center (WTC) on September 11, 2001, have suffered a disproportionate incidence of chronic lung disease attributed to the inhalation of airborne dust. To date, the pathophysiology of this lung disease is poorly understood. The aim of this study was to examine whether airborne dust contaminants recovered from the surrounding area 24-48 h after the collapse of the WTC demonstrate direct cytotoxicity to two airway cell types that were most directly exposed to inhaled dust, airway epithelial and smooth muscle cells. It was also of interest to determine whether the presence of these dusts could modulate the effects of cigarette smoke on these cell types in that some of the individuals who responded to the collapse site were also smokers. Human cultured airway epithelial (BEAS-2B) cells were exposed to 10% cigarette smoke extract (CSE), WTC dust particles (10-53 μm; 0.01-0.5 μg/μl), or a combination of the two for 2-24 h. Cell viability was measured by determining mitochondrial integrity (MTT assays) and apoptosis (poly-ADP-ribose polymerase [PARP] immunoblotting). Conditioned cell culture media recovered from the CSE- and/or WTC dust-exposed BEAS-2B cells were then applied to cultured human airway smooth muscle cells that were subsequently assayed for mitochondrial integrity and their ability to synthesize cyclic AMP (a regulator of airway smooth muscle constriction). BEAS-2B cells underwent necrotic cell death following exposure to WTC dust or CSE for 2-24 h without evidence of apoptosis. Smooth muscle cells demonstrated cellular toxicity and enhanced cyclic AMP synthesis following exposure to conditioned media from WTC- or CSE-exposed epithelial cells. These acute toxicity assays of WTC dust and CSE offer insights into lung cell toxicity that may contribute to the pathophysiology of chronic lung disease in workers and residents exposed to WTC dust. These studies clearly showed that WTC dust (at least the supercoarse particle fraction) or CSE alone exerted direct adverse effects on airway epithelial and smooth muscle cells, and altered the signaling properties of airway smooth muscle cells. In addition the combination of CSE and WTC exerted an interactive effect on cell toxicity. It remains to be determined whether these initial cell death events might account, in part, for the chronic lung effects associated with WTC dust exposure among First Responders and others.
Collapse
Affiliation(s)
- Alice Xu
- Columbia University, New York, New York, USA
| | | | | | | | | |
Collapse
|
9
|
Chen Y, Chang L, Li W, Rong Z, Liu W, Shan R, Pan R. Thioredoxin protects fetal type II epithelial cells from hyperoxia-induced injury. Pediatr Pulmonol 2010; 45:1192-200. [PMID: 20812253 DOI: 10.1002/ppul.21307] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 04/04/2010] [Accepted: 04/05/2010] [Indexed: 11/06/2022]
Abstract
Oxygen toxicity is known to be one of the major contributors to bronchopulmonary dysplasia, a chronic lung disease in premature infants. Thioredoxin (Trx) is an antioxidant that prevents oxidative stress-induced cell death, suggesting a potential therapeutic role in bronchopulmonary dysplasia. The aim of this study was to determine the role of Trx in the pathogenesis of hyperoxia-induced alveolar epithelial cell injury. Alveolar type II epithelial cells from fetal rat lung were exposed to hyperoxia in vitro in the presence or absence of recombinant human Trx (rhTrx 2 µg/ml). Cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. Apoptosis and levels of reactive oxygen species (ROS) were measured by flow cytometry. Activation of mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase-Akt (PI3K-Akt) pathways were detected by Western blotting. We also investigated the effects of rhTrx on the following antioxidants (superoxide dismutase, catalase, and glutathione peroxidase). Trx significantly reduced hyperoxia-induced cell death and increased cell viability. In addition, ROS generation in type II cells was inhibited by rhTrx under hyperoxic conditions. We demonstrated that rhTrx protected type II cells against hyperoxic injury via sustaining the extracellular signal regulated kinase and PI3K activation, and decreasing of c-Jun N-terminal protein kinase and p38 activation. The results also showed manganese superoxide dismutase and glutathione peroxidase activities were increased by rhTrx in type II cells exposed to hyperoxia.Taken together, these results demonstrate that rhTrx administration markedly attenuates hyperoxia-induced type II cell injury through reduction of ROS generation, elevation of antioxidant activities and regulation of both MAPK and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Yan Chen
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Waldow T, Witt W, Ulmer A, Janke A, Alexiou K, Matschke K. Preconditioning by inhaled nitric oxide prevents hyperoxic and ischemia/reperfusion injury in rat lungs. Pulm Pharmacol Ther 2008; 21:418-29. [PMID: 18453045 DOI: 10.1016/j.pupt.2007.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Since the generation of nitric oxide (NO) is an essential step in the trigger phase of ischemic preconditioning, short-term inhalation of NO before ischemia should ameliorate ischemia/reperfusion (I/R) injury of the lung. We tested this hypothesis in high oxygen (>99%) ventilated rats in order to additionally evaluate compatibility of NO and exposure to hyperoxia. Male adult Sprague-Dawley rats inhaled NO (15 ppm, 10 min) before the left lung hilum was clamped for 1 h, and the reperfusion phase was observed for 4 h (NO group). Animals in the I/R group underwent the same treatment, but without NO inhalation. A third group without I/R served as time-matched controls. Animals in the I/R group showed severe I/R injury in terms of arterial pO2 (apO2), which was reduced to 22% of surgical controls (SCs) at time point 30 min reperfusion, and increased endothelial permeability (Evans blue procedure). The pretreatment with NO attenuated these effects. The pO2 after 4 h reperfusion was still 3.0-fold higher in the NO group compared to I/R. In contrast, the I/R- and hyperoxia-induced invasion of leukocytes, as determined by measuring myeloperoxidase (MPO) activity, was not affected by NO. These data were correlated with the activity of major cellular signaling pathways by measuring the phosphorylation at activating and inhibitory sites of extracellular-signal regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38, protein kinase B (AKT), and glycogen synthase kinase 3beta (GSK-3beta), and by determination of cGMP in plasma and lung tissue. Inhalation of NO partly prevented the loss of activation by I/R and hyperoxic ventilation of ERK, JNK, and AKT, and it reduced the I/R-induced activation of GSK-3beta. The level of cGMP in plasma and lung tissue was increased in the NO group after 4 h reperfusion. In conclusion, application of inhaled NO in the preconditioning mode prevented I/R injury in the rat lung without interfering effects of hyperoxic ventilation. The effects of NO on cellular signaling pathways resemble mechanisms of ischemic preconditioning, but further studies have to evaluate the physiological relevance of these results.
Collapse
Affiliation(s)
- Thomas Waldow
- Clinic for Cardiac Surgery, University Hospital Dresden, Fetscherstr. 76, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
11
|
Monick MM, Powers LS, Barrett CW, Hinde S, Ashare A, Groskreutz DJ, Nyunoya T, Coleman M, Spitz DR, Hunninghake GW. Constitutive ERK MAPK activity regulates macrophage ATP production and mitochondrial integrity. THE JOURNAL OF IMMUNOLOGY 2008; 180:7485-96. [PMID: 18490749 DOI: 10.4049/jimmunol.180.11.7485] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A unique feature of human alveolar macrophages is their prolonged survival in the face of a stressful environment. We have shown previously that the ERK MAPK is constitutively active in these cells and is important in prolonging cell survival. This study examines the role of the ERK pathway in maintaining mitochondrial energy production. The data demonstrate that ATP levels in alveolar macrophages depend on intact mitochondria and optimal functioning of the electron transport chain. Significant levels of MEK and ERK localize to the mitochondria and inhibition of ERK activity induces an early and profound depletion in cellular ATP coincident with a loss of mitochondrial transmembrane potential. The effect of ERK suppression on ATP levels was specific, since it did not occur with PI3K/Akt, p38, or JNK suppression. ERK inhibition led to cytosolic release of mitochondrial proteins and caspase activation. Both ERK inhibition and mitochondrial blockers induced loss of plasma membrane permeability and cell death. The cell death induced by ERK inhibition had hallmarks of both apoptotic (caspase activation) and necrotic (ATP loss) cell death. By blocking ERK inhibition-induced reactive oxygen species, caspase activation was prevented, although necrotic pathways continued to induce cell death. This suggests that mitochondrial dysfunction caused by ERK inhibition generates both apoptotic and necrotic cell death-inducing pathways. As a composite, these data demonstrate a novel mitochondrial role for ERK in maintaining mitochondrial membrane potential and ATP production in human alveolar macrophages.
Collapse
Affiliation(s)
- Martha M Monick
- Department of Medicine, University of Iowa Carver College of Medicine and Veterans Administration Medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Mao Q, Gundavarapu S, Patel C, Tsai A, Luks FI, De Paepe ME. The Fas system confers protection against alveolar disruption in hyperoxia-exposed newborn mice. Am J Respir Cell Mol Biol 2008; 39:717-29. [PMID: 18587053 DOI: 10.1165/rcmb.2008-0052oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The functional significance of the Fas/Fas-ligand (FasL) system in hyperoxia-induced lung injury and alveolar disruption in newborn lungs in vivo remains undetermined. To assess the role of the Fas/FasL system, we compared the effects of hyperoxia (95% O2 from birth to Postnatal Day [P]7) in Fas-deficient lpr mice and wild-type mice. Alveolar disruption was more severe in hyperoxic lpr mice than in wild-type mice. In addition, a transient alveolarization defect was noted in normoxic lpr mice. Hyperoxia induced marked up-regulation of pulmonary Fas expression in wild-type mice, as well as elevated mRNA levels of pro-apoptotic Bax, Bad, and Bak. Pulmonary apoptotic activity was similar in hyperoxic wild-type and lpr mice. In contrast, lung growth and proliferation, assessed by stereologic volumetry and Ki67 proliferation studies, were significantly higher in hyperoxic wild-type mice compared with lpr mice, suggesting the Fas/FasL system has a pro-proliferative role in hyperoxic conditions. Levels of the prosurvival MAPkinase, pERK1/2, were significantly higher in hyperoxic wild-type mice compared with lpr mice, while pAkt levels were similar. These data suggest that the primary role of the Fas/FasL system in hyperoxic newborn lungs is pro-proliferative, rather than pro-apoptotic, and likely mediated through a Fas-ERK1/2 pathway. Fas-induced proliferation and lung growth in hyperoxic newborn lungs may counteract, in part, the detrimental effects of apoptosis mediated by non-Fas pathways, such as pro-apoptotic Bax/Bcl-2 family members. The capacity of the Fas/FasL signaling pathway to mediate protective rather than destructive functions in hyperoxic newborn lungs highlights the versatility of this complex pathway.
Collapse
Affiliation(s)
- Quanfu Mao
- Department of Pathology, Women and Infants Hospital, Providence, Rhode Island 02905, USA
| | | | | | | | | | | |
Collapse
|
13
|
Wang X, Wang Y, Kim HP, Choi AMK, Ryter SW. FLIP inhibits endothelial cell apoptosis during hyperoxia by suppressing Bax. Free Radic Biol Med 2007; 42:1599-609. [PMID: 17448907 DOI: 10.1016/j.freeradbiomed.2007.02.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 02/16/2007] [Accepted: 02/20/2007] [Indexed: 02/08/2023]
Abstract
High oxygen tension (hyperoxia) causes pulmonary cell death, involving apoptosis, necrosis, or mixed death phenotypes, though the underlying mechanisms remain unclear. In mouse lung endothelial cells (MLEC) hyperoxia activates both extrinsic (Fas-dependent) and intrinsic (mitochondria-dependent) apoptotic pathways. We examined the hypothesis that FLIP, an inhibitor of caspase-8, can protect endothelial cells against the lethal effects of hyperoxia. Hyperoxia caused the time-dependent downregulation of FLIP in MLEC. Overexpression of FLIP attenuated intracellular reactive oxygen species generation during hyperoxia exposure, by downregulating extracellular-regulated kinase-1/2 activation and p47(phox) expression. FLIP prevented hyperoxia-induced trafficking of the death-inducing signal complex from the Golgi apparatus to the plasma membrane. Furthermore, FLIP blocked the activations of caspase-8/Bid, caspases -3/-9, and inhibited the mitochondrial translocation and activation of Bax, resulting in protection against hyperoxia-induced cell death. Under normoxic conditions, FLIP expression increased the phosphorylation of p38 mitogen-activated protein kinase leading to increased phosphorylation of Bax during hyperoxic stress. Furthermore, FLIP expression markedly inhibited protein kinase C activation and expression of distinct protein kinase C isoforms (alpha, eta, and zeta), and stabilized an interaction of PKC with Bax. In conclusion, FLIP exerted novel inhibitory effects on extrinsic and intrinsic apoptotic pathways, which significantly protected endothelial cells from the lethal effects of hyperoxia.
Collapse
Affiliation(s)
- Xue Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, 3459 Fifth Avenue, MUH 628NW, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
14
|
Zaher TE, Miller EJ, Morrow DMP, Javdan M, Mantell LL. Hyperoxia-induced signal transduction pathways in pulmonary epithelial cells. Free Radic Biol Med 2007; 42:897-908. [PMID: 17349918 PMCID: PMC1876680 DOI: 10.1016/j.freeradbiomed.2007.01.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 01/05/2007] [Accepted: 01/08/2007] [Indexed: 10/23/2022]
Abstract
Mechanical ventilation with hyperoxia is necessary to treat critically ill patients. However, prolonged exposure to hyperoxia leads to the generation of excessive reactive oxygen species (ROS), which can cause acute inflammatory lung injury. One of the major effects of hyperoxia is the injury and death of pulmonary epithelium, which is accompanied by increased levels of pulmonary proinflammatory cytokines and excessive leukocyte infiltration. A thorough understanding of the signaling pathways leading to pulmonary epithelial cell injury/death may provide some insights into the pathogenesis of hyperoxia-induced acute inflammatory lung injury. This review focuses on epithelial responses to hyperoxia and some of the major factors regulating pathways to epithelial cell injury/death, and proinflammatory responses on exposure to hyperoxia. We discuss in detail some of the most interesting players, such as NF-kappaB, that can modulate both proinflammatory responses and cell injury/death of lung epithelial cells. A better appreciation for the functions of these factors will no doubt help us to delineate the pathways to hyperoxic cell death and proinflammatory responses.
Collapse
Affiliation(s)
- Tahereh E. Zaher
- Department of Pharmaceutical Sciences, St. John’s University College of Pharmacy, Queens, NY 11439
- Cardiopulmonary Research, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030
| | - Edmund J. Miller
- Surgercal Immunology, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030
| | - Dympna M. P. Morrow
- Cardiopulmonary Research, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030
| | - Mohammad Javdan
- Cardiopulmonary Research, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, St. John’s University College of Pharmacy, Queens, NY 11439
- Cardiopulmonary Research, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030
- *Correspondence author: Lin L. Mantell, Department of Pharmaceutical Sciences, St. John’s University College of Pharmacy, 108/SB28 St. Albert Hall, 8000 Utopia Parkway, Queens, New York 11439, Tel: 718-990-5933, Fax: 718-990-1877,
| |
Collapse
|
15
|
Xu D, Guthrie JR, Mabry S, Sack TM, Truog WE. Mitochondrial aldehyde dehydrogenase attenuates hyperoxia-induced cell death through activation of ERK/MAPK and PI3K-Akt pathways in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2006; 291:L966-75. [PMID: 16782756 DOI: 10.1152/ajplung.00045.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oxygen toxicity is one of the major risk factors in the development of the chronic lung disease or bronchopulmonary dysplasia in premature infants. Using proteomic analysis, we discovered that mitochondrial aldehyde dehydrogenase (mtALDH or ALDH2) was downregulated in neonatal rat lung after hyperoxic exposure. To study the role of mtALDH in hyperoxic lung injury, we overexpressed mtALDH in human lung epithelial cells (A549) and found that mtALDH significantly reduced hyperoxia-induced cell death. Compared with control cells (Neo-A549), the necrotic cell death in mtALDH-overexpressing cells (mtALDH-A549) decreased from 25.3 to 6.5%, 50.5 to 9.1%, and 52.4 to 15.1% after 24-, 48-, and 72-h hyperoxic exposure, respectively. The levels of intracellular and mitochondria-derived reactive oxygen species (ROS) in mtALDH-A549 cells after hyperoxic exposure were significantly lowered compared with Neo-A549 cells. mtALDH overexpression significantly stimulated extracellular signal-regulated kinase (ERK) phosphorylation under normoxic and hyperoxic conditions. Inhibition of ERK phosphorylation partially eliminated the protective effect of mtALDH in hyperoxia-induced cell death, suggesting ERK activation by mtALDH conferred cellular resistance to hyperoxia. mtALDH overexpression augmented Akt phosphorylation and maintained the total Akt level in mtALDH-A549 cells under normoxic and hyperoxic conditions. Inhibition of phosphatidylinositol 3-kinase (PI3K) activation by LY294002 in mtALDH-A549 cells significantly increased necrotic cell death after hyperoxic exposure, indicating that PI3K-Akt activation by mtALDH played an important role in cell survival after hyperoxia. Taken together, these data demonstrate that mtALDH overexpression attenuates hyperoxia-induced cell death in lung epithelial cells through reduction of ROS, activation of ERK/MAPK, and PI3K-Akt cell survival signaling pathways.
Collapse
Affiliation(s)
- Dong Xu
- Neonatology Research Laboratory, Children's Mercy Hospital, Pediatric Research Center, 4th Floor, 2401 Gillham Rd., Kansas City, MO 64108, USA.
| | | | | | | | | |
Collapse
|
16
|
Kuwahara I, Lillehoj EP, Lu W, Singh IS, Isohama Y, Miyata T, Kim KC. Neutrophil elastase induces IL-8 gene transcription and protein release through p38/NF-{kappa}B activation via EGFR transactivation in a lung epithelial cell line. Am J Physiol Lung Cell Mol Physiol 2006; 291:L407-16. [PMID: 16632517 DOI: 10.1152/ajplung.00471.2005] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In this study, we investigated the regulation and mechanism of IL-8 expression by A549 human lung carcinoma cells treated with neutrophil elastase (NE). NE-treated cells exhibited significantly higher IL-8 protein levels in culture media compared with cells treated with vehicle alone. Blocking of gene transcription with actinomycin D suggested that NE stimulated IL-8 synthesis via increased mRNA expression, which was verified by real-time RT-PCR. NE activated the IL-8 promoter but did not alter the stability of its mRNA, confirming that the protease induced IL-8 synthesis through increased gene transcription. The results from the use of chemical inhibitors and mutant gene constructs against various signal transduction components seem to suggest the linear signaling pathway involving the activation of PKC-delta --> dual oxidase 1 --> reactive oxygen species --> TNF-alpha-converting enzyme --> EGF receptor --> p38 --> NF-kappaB for NE-activated IL-8 gene expression. A NF-kappaB potential binding site, located between nucleotides -82 and -69 of the IL-8 promoter, was identified as necessary for NE-induced IL-8 transcription. We conclude that NE increases IL-8 transcription through p38/NF-kappaB activation via EGFR transactivation.
Collapse
Affiliation(s)
- Ippei Kuwahara
- Respiratory Immunology and Asthma Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Ahmad A, Ahmad S, Chang LY, Schaack J, White CW. Endothelial Akt activation by hyperoxia: role in cell survival. Free Radic Biol Med 2006; 40:1108-18. [PMID: 16545678 DOI: 10.1016/j.freeradbiomed.2005.10.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Revised: 10/14/2005] [Accepted: 10/18/2005] [Indexed: 10/25/2022]
Abstract
High oxygen concentrations (hyperoxia), often required in the treatment of preterm infants and critically ill patients, cause lung injury, targeting especially the endothelium. Exposure of primary human lung microvascular endothelial cells (HLMVEC) to hyperoxia caused transient Akt activation after 60 min, as determined by Western blot analysis of phosphorylated Ser 473 of Akt. Akt phosphorylation was also increased after 24 h of hyperoxic exposure, which declined at 48 h. Adenoviral (Ad)-mediated expression of constitutively active myrAkt protected HLMVEC against hyperoxic injury. Cell death due to hyperoxia (95% O2, 8 days), which was primarily necrotic, was substantial in control and Ad-LacZ-transduced cells, but was diminished by almost half in myrAkt-transduced cells. Hyperoxia caused increased cellular glucose consumption, an effect that was amplified in cells transduced with myrAkt compared to the LacZ-transduced or the nontransduced controls. Increased glucose consumption in myrAkt-expressing cells was accompanied by increased phosphorylation of mTOR and p70 S6-kinase. Rapamycin treatment decreased glucose consumption in myrAkt-transduced cells to levels comparable to those in control and LacZ-transduced cells exposed to hyperoxia. Ultrastructural morphometric analyses demonstrated that mitochondria and endoplasmic reticulum were less swollen in myrAkt cells relative to controls exposed to hyperoxia. These studies demonstrate that early activation of Akt occurs in hyperoxia in HLMVEC. That this event is a beneficial response is suggested by the finding that constitutive activation of Akt protects against hyperoxic stress, at least in part, by maintaining mitochondrial integrity.
Collapse
Affiliation(s)
- Aftab Ahmad
- Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson Street, Denver, CO 80206, USA
| | | | | | | | | |
Collapse
|
18
|
Abstract
Oxidative injury to the lung is associated with widespread injury to the alveolar epithelium, which can be fatal unless the process is controlled and repaired. Keratinocyte growth factor (KGF), a member of the fibroblast growth factor family, has been shown to protect the lung from a variety of oxidative insults. The mechanism(s) underlying the protective effects of KGF in lung injury is being investigated in many laboratories. Although KGF has potent mitogenic effects on epithelial cells, the proliferative effect of KGF was shown to be abolished in oxygen-breathing animals, but KGF was still able to inhibit alveolar damage. This demonstrates that the protective effect of KGF cannot simply be explained by the ability of KGF to stimulate type II cell proliferation. To identify the mechanisms involved in the protective effects of KGF, we used an inducible lung-specific transgenic approach to overexpress KGF in murine lungs, since constitutive overexpression of KGF in the mouse affects lung development. The transgenic system allowed us to identify the pro-survival Akt pathway as an important mediator of the protective effects of KGF both in vivo and in vitro. In addition, use of a yeast two-hybrid system led to the identification two proteins p90RSK and PAK4 that associate with the KGF receptor and are important for the protective functions of KGF. Experiments are underway to determine whether the different pathways triggered by KGF all converge on the Akt pathway, or whether they independently induce protective mechanisms that along with Akt are crucial for cell survival.
Collapse
Affiliation(s)
- Prabir Ray
- University of Pittsburgh School of Medicine, Department of Medicine/Pulmonary Division, Pittsburgh, PA 15213, USA.
| |
Collapse
|
19
|
Current World Literature. Curr Opin Allergy Clin Immunol 2006; 6:67-9. [PMID: 16505615 DOI: 10.1097/01.all.0000202355.95779.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Pagano A, Pitteloud C, Reverdin C, Métrailler-Ruchonnet I, Donati Y, Barazzone Argiroffo C. Poly(ADP-ribose)polymerase Activation Mediates Lung Epithelial Cell DeathIn Vitrobut Is Not Essential in Hyperoxia-Induced Lung Injury. Am J Respir Cell Mol Biol 2005; 33:555-64. [PMID: 16151053 DOI: 10.1165/rcmb.2004-0361oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
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.
Collapse
Affiliation(s)
- Alessandra Pagano
- Departments of Pediatrics and Pathology, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
21
|
Jin Y, Kim HP, Ifedigbo E, Lau LF, Choi AMK. Cyr61 Protects against Hyperoxia-Induced Cell Death via Akt Pathway in Pulmonary Epithelial Cells. Am J Respir Cell Mol Biol 2005; 33:297-302. [PMID: 15961723 DOI: 10.1165/rcmb.2005-0144oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We have used gene expression profiling approaches to identify new molecular targets in various models of lung injury and human lung diseases. Among the many genes that are significantly induced in these studies, cysteine-rich61 (Cyr61) consistently ranks as one of the most significant genes. Here, we use the well-established model of hyperoxia to better understand the function of Cyr61 in acute lung injury. Cyr61, a stress-related immediate-early response gene, has known diverse functions involving angiogenesis, tumorigenesis, and wound repair. It belongs to the newly discovered "CCN" family containing six growth and regulatory factors. We showed that hyperoxia induces Cyr61 expression in a variety of pulmonary cells and in lung tissue in vivo. Loss of function studies, by suppressing Cyr61 expression by siRNA, accelerated lung epithelial cell death after hyperoxia. Gain of function studies, by overexpressing Cyr61, significantly conferred increased resistance to hyperoxia-induced cell death. Moreover, cells overexpressing Cyr61 induce Akt activation. Inhibition of Akt by siRNA abrogated the protective effects of Cyr61-overexpressing cells in response to hyperoxia. Taken together, our data demonstrate that Cyr61 expression provides cytoprotection in hyperoxia-induced pulmonary epithelial cell death and that this effect was in part mediated via the Akt signaling pathway.
Collapse
Affiliation(s)
- Yang Jin
- Division of Pulmonary, Allergy and Critical Care Medicine, 628 NW MUH, University of Pittsburgh Medical Center, 3459 5th Ave., Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|