51
|
Gefitinib Inhibits Bleomycin-Induced Pulmonary Fibrosis via Alleviating the Oxidative Damage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8249693. [PMID: 29849916 PMCID: PMC5924979 DOI: 10.1155/2018/8249693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/24/2017] [Accepted: 11/26/2017] [Indexed: 01/09/2023]
Abstract
Pulmonary fibrosis (PF) is a life-threatening interstitial lung disease. In this study, we tried to reveal the model of action between high-mobility group box 1 (HMGB1) and α-smooth muscle actin (α-SMA) and the protective role of gefitinib in pulmonary fibrosis induced by the administration of bleomycin aerosol in mice. For the mechanism study, lung tissues were harvested two weeks after modeling to detect the coexpression of HMGB1 and α-SMA by immunohistochemistry and immunofluorescence staining. Protein-DNA interactions were analyzed using a pulldown assay to study the relationship between HMGB1 and α-SMA. For the gefitinib treatment study, the mice were divided into three groups: phosphate-buffered saline (PBS) control group, PBS-treated PF group, and gefitinib-treated PF group. Gavage of gefitinib or PBS (20 mg/kg/day) was performed after bleomycin treatment for two weeks until the mice were sacrificed. Lung and blood samples were collected to assess the histological changes, oxidative stress, and expression of NOXs, HMGB1, EGFR, MAPKs, AP-1, and NF-κB to determine the curative effect and related molecular mechanisms. The results revealed the high coexpression of α-SMA and HMGB1 in some interstitial cells in the fibrotic lung. The DNA-protein pulldown analysis proved that HMGB34367 acted as a novel transcriptional factor for the α-SMA promoter and participated in the eventual development of pulmonary fibrosis. Second, gefitinib could significantly decrease lung fibrotic changes and the level of MDA and recover the T-AOC level. Meanwhile, gefitinib could also reduce the NOX1/2/4, HMGB1, p-EGFR, p-ERK, p-JNK, p-P38, p-NF-κB, p-c-Jun, and p-c-Fos expression levels in fibrotic lungs. The present study suggested that gefitinib could alleviate lung fibrosis through the HMGB1/NOXs-ROS/EGFR-MAPKs-AP-1/NF-κB signal in bleomycin-induced pulmonary fibrosis.
Collapse
|
52
|
Sharma M, Afolayan AJ. Redox Signaling and Persistent Pulmonary Hypertension of the Newborn. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:277-287. [PMID: 29047092 DOI: 10.1007/978-3-319-63245-2_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Reactive oxygen species (ROS) are redox-signaling molecules that are critically involved in regulating endothelial cell functions, host defense, aging, and cellular adaptation. Mitochondria are the major sources of ROS and important sources of redox signaling in pulmonary circulation. It is becoming increasingly evident that increased mitochondrial oxidative stress and aberrant signaling through redox-sensitive pathways play a direct causative role in the pathogenesis of many cardiopulmonary disorders including persistent pulmonary hypertension of the newborn (PPHN). This chapter highlights redox signaling in endothelial cells, antioxidant defense mechanism, cell responses to oxidative stress, and their contributions to disease pathogenesis.
Collapse
Affiliation(s)
- Megha Sharma
- Assistant Professor of Pediatrics, 999 N92nd Street, CCC suite 410, Milwaukee, WI, 53226, USA
| | - Adeleye J Afolayan
- Assistant Professor of Pediatrics, 999 N92nd Street, CCC suite 410, Milwaukee, WI, 53226, USA.
| |
Collapse
|
53
|
Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-mediated vascular barrier injury: Role of neutrophil extracellular traps. Microcirculation 2018; 24. [PMID: 28120468 DOI: 10.1111/micc.12352] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
Neutrophils play an essential role in host defense against infection or injury. While neutrophil activation is necessary for pathogen clearance and tissue repair, a hyperactive response can lead to tissue damage and microcirculatory disorders, a process involving complex neutrophil-endothelium cross talk. This review highlights recent research findings about neutrophil-mediated signaling and structural changes, including those induced by neutrophil extracellular traps, which ultimately lead to vascular barrier injury.
Collapse
Affiliation(s)
- Jamie E Meegan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Danielle C Coleman
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
54
|
The Role of Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Lung Architecture Remodeling. Antioxidants (Basel) 2017; 6:antiox6040104. [PMID: 29257052 PMCID: PMC5745514 DOI: 10.3390/antiox6040104] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic lung disorders, such as pulmonary artery hypertension (PAH), chronic obstructive pulmonary disease (COPD), asthma and neonatal bronchopulmonary dysplasia (BPD), are characterized by airway and/or vascular remodeling. Despite differences in the pathology, reactive oxygen species (ROS) have been highlighted as a critical contributor to the initiation and development of airway and vascular remodeling. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox) appear to play a pivotal role in lung signaling, leading to marked changes in pulmonary airway and vascular cell phenotypes, including proliferation, hypertrophy and apoptosis. In this review, we summarized the current literature regarding the role of Nox in the airway and vascular remodeling.
Collapse
|
55
|
Dietz RM, Wright CJ. Oxidative stress diseases unique to the perinatal period: A window into the developing innate immune response. Am J Reprod Immunol 2017; 79:e12787. [PMID: 29194835 DOI: 10.1111/aji.12787] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022] Open
Abstract
The innate immune system has evolved to play an integral role in the normally developing lung and brain. However, in response to oxidative stress, innate immunity, mediated by specific cellular and molecular programs and signaling, contributes to pathology in these same organ systems. Despite opposing drivers of oxidative stress, namely hyperoxia in neonatal lung injury and hypoxia/ischemia in neonatal brain injury, similar pathways-including toll-like receptors, NFκB and MAPK cascades-have been implicated in tissue damage. In this review, we consider recent insights into the innate immune response to oxidative stress in both neonatal and adult models to better understand hyperoxic lung injury and hypoxic-ischemic brain injury across development and aging. These insights support the development of targeted immunotherapeutic strategies to address the challenge of harnessing the innate immune system in oxidative stress diseases of the neonate.
Collapse
Affiliation(s)
- Robert M Dietz
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
56
|
Zhao M, Tang S, Xin J, Wei Y, Liu D. Reactive oxygen species induce injury of the intestinal epithelium during hyperoxia. Int J Mol Med 2017; 41:322-330. [PMID: 29138796 PMCID: PMC5746288 DOI: 10.3892/ijmm.2017.3247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Long-term therapeutic hyperoxia may exert serious toxic effects on intestinal epithelial cells in vitro and in vivo. The aim of the present study was to investigate the cause of this intestinal injury under conditions of hyperoxia. Caco-2 cells were treated with different concentrations of hydrogen peroxide (H2O2) and 85% hyperoxia for 24 h. higher rates of injury of Caco-2 cells were observed in the hyperoxia and H2O2 groups compared with the control group. The reactive oxygen species (ROS) level of the hyperoxia group was significantly higher compared with that of the 400 µM H2O2 group. The protein and gene levels of RelA, RelB, hypoxia‑inducible factor-1α, tumor necrosis factor-α and apoptosis signal‑regulating kinase 1 were significantly higher in the hyperoxia and H2O2 groups compared with those in the control group. In conclusion, during hyperoxia, intestinal epithelial cells were destroyed and the levels of ROS were increased. Therefore, ROS may play an important role in intestinal injury in a hyperoxic environment.
Collapse
Affiliation(s)
- Min Zhao
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shimiao Tang
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Junchi Xin
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Yingliang Wei
- Department of Orthopaedic Surgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Dongyan Liu
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
57
|
Zhao M, Tang S, Xin J, Liu D. Influence of reactive oxygen species on secretory component in the intestinal epithelium during hyperoxia. Exp Ther Med 2017; 14:4033-4040. [PMID: 29075338 PMCID: PMC5648505 DOI: 10.3892/etm.2017.5027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 05/19/2017] [Indexed: 01/23/2023] Open
Abstract
Redox imbalance is established in various human diseases. Treatment of intestinal epithelial cells with hyperoxia for a prolonged period of time may cause serious effects on redox balance. Secretory component (SC) protein is secreted by intestinal epithelial cells, and has a vital role in mucosal immune systems and intestinal defense. The present study aimed to investigate the influence of reactive oxygen species (ROS) on intestinal epithelial cells and intestinal epithelial SC protein under hyperoxic conditions. Caco-2 cells were treated with increasing concentrations of hydrogen peroxide (H2O2) or 85% O2 (hyperoxia) for 24 h. Flow cytometry, immunohistochemistry staining, western blot analysis and reverse transcription-quantitative polymerase chain reaction were performed to detect the expression levels of SC protein. Significantly increased apoptosis and mortality rates were observed in hyperoxia- and H2O2-treated Caco-2 cells, as compared with the untreated control cells (P<0.05). Protein and mRNA expression levels of SC were significantly increased in hyperoxia- and H2O2-treated groups, as compared with the control group (P<0.05). During hyperoxia, intestinal epithelial cells were destroyed and ROS levels increased. Therefore, the results of the present study suggested that ROS might have an important role in intestinal injury in hyperoxic environments.
Collapse
Affiliation(s)
- Min Zhao
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Shimiao Tang
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Junchi Xin
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Dongyan Liu
- Medical Research Center, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
58
|
Campochiaro PA, Mir TA. The mechanism of cone cell death in Retinitis Pigmentosa. Prog Retin Eye Res 2017; 62:24-37. [PMID: 28962928 DOI: 10.1016/j.preteyeres.2017.08.004] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/31/2017] [Accepted: 08/31/2017] [Indexed: 12/16/2022]
Abstract
Retinitis Pigmentosa (RP) is a group of diseases in which one of a large number of mutations causes death of rod photoreceptors. After rods die, cone photoreceptors slowly degenerate in a characteristic pattern. The mechanism of rod cell death varies depending upon the gene that is mutated and the rate that rods degenerate is an important prognostic feature, because cones do not begin to degenerate until almost all rods have been eliminated. Rod cell death causes night blindness, but visual disability and blindness result from cone degeneration and therefore it is critical to determine the mechanisms by which it occurs. The death of rods reduces oxygen consumption resulting in high tissue levels of oxygen in the outer retina. The excess oxygen stimulates superoxide radical production by mismatches in the electron transport chain in mitochondria and by stimulation of NADPH oxidase activity in cytoplasm. The high levels of superoxide radicals overwhelm the antioxidant defense system and generate more reactive species including peroxynitrite which is extremely damaging and difficult to detoxify. This results in progressive oxidative damage in cones which contributes to cone cell death and loss of function because drugs or gene transfer that reduce oxidative stress promote cone survival and maintenance of function. Compared with aqueous humor samples from control patients, those from patients with RP show significant elevation of carbonyl content on proteins indicating oxidative damage and a reduction in the ratio of reduced to oxidized glutathione indicating depletion of a major component of the antioxidant defense system from ongoing oxidative stress. The first step in clinical trials will be to identify doses of therapeutic agents that reverse these biomarkers of disease to assist in design of much longer trials with functional and anatomic endpoints.
Collapse
Affiliation(s)
- Peter A Campochiaro
- Departments of Ophthalmology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Tahreem A Mir
- Departments of Ophthalmology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
59
|
Teixeira G, Szyndralewiez C, Molango S, Carnesecchi S, Heitz F, Wiesel P, Wood JM. Therapeutic potential of NADPH oxidase 1/4 inhibitors. Br J Pharmacol 2017; 174:1647-1669. [PMID: 27273790 PMCID: PMC5446584 DOI: 10.1111/bph.13532] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/16/2022] Open
Abstract
The NADPH oxidase (NOX) family of enzymes produces ROS as their sole function and is becoming recognized as key modulators of signal transduction pathways with a physiological role under acute stress and a pathological role after excessive activation under chronic stress. The seven isoforms differ in their regulation, tissue and subcellular localization and ROS products. The most studied are NOX1, 2 and 4. Genetic deletion of NOX1 and 4, in contrast to NOX2, has revealed no significant spontaneous pathologies and a pathogenic relevance of both NOX1 and 4 across multiple organs in a wide range of diseases and in particular inflammatory and fibrotic diseases. This has stimulated interest in NOX inhibitors for therapeutic application. GKT136901 and GKT137831 are two structurally related compounds demonstrating a preferential inhibition of NOX1 and 4 that have suitable properties for in vivo studies and have consequently been evaluated across a range of disease models and compared with gene deletion. In contrast to gene deletion, these inhibitors do not completely suppress ROS production, maintaining some basal level of ROS. Despite this and consistent with most gene deletion studies, these inhibitors are well tolerated and slow or prevent disease progression in a range of models of chronic inflammatory and fibrotic diseases by modulating common signal transduction pathways. Clinical trials in patients with GKT137831 have demonstrated excellent tolerability and reduction of various markers of chronic inflammation. NOX1/4 inhibition may provide a safe and effective therapeutic strategy for a range of inflammatory and fibrotic diseases. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- G Teixeira
- Evotec International GmbHGoettingenGermany
| | | | - S Molango
- Genkyotex SAPlan les OuatesSwitzerland
| | | | - F Heitz
- Genkyotex SAPlan les OuatesSwitzerland
| | - P Wiesel
- Genkyotex SAPlan les OuatesSwitzerland
| | | |
Collapse
|
60
|
Hydrogen Sulfide Prevents Formation of Reactive Oxygen Species through PI3K/Akt Signaling and Limits Ventilator-Induced Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3715037. [PMID: 28250891 PMCID: PMC5307128 DOI: 10.1155/2017/3715037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/05/2016] [Accepted: 01/09/2017] [Indexed: 11/19/2022]
Abstract
The development of ventilator-induced lung injury (VILI) is still a major problem in mechanically ventilated patients. Low dose inhalation of hydrogen sulfide (H2S) during mechanical ventilation has been proven to prevent lung damage by limiting inflammatory responses in rodent models. However, the capacity of H2S to affect oxidative processes in VILI and its underlying molecular signaling pathways remains elusive. In the present study we show that ventilation with moderate tidal volumes of 12 ml/kg for 6 h led to an excessive formation of reactive oxygen species (ROS) in mice lungs which was prevented by supplemental inhalation of 80 parts per million of H2S. In addition, phosphorylation of the signaling protein Akt was induced by H2S. In contrast, inhibition of Akt by LY294002 during ventilation reestablished lung damage, neutrophil influx, and proinflammatory cytokine release despite the presence of H2S. Moreover, the ability of H2S to induce the antioxidant glutathione and to prevent ROS production was reversed in the presence of the Akt inhibitor. Here, we provide the first evidence that H2S-mediated Akt activation is a key step in protection against VILI, suggesting that Akt signaling limits not only inflammatory but also detrimental oxidative processes that promote the development of lung injury.
Collapse
|
61
|
Kellner M, Noonepalle S, Lu Q, Srivastava A, Zemskov E, Black SM. ROS Signaling in the Pathogenesis of Acute Lung Injury (ALI) and Acute Respiratory Distress Syndrome (ARDS). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:105-137. [PMID: 29047084 PMCID: PMC7120947 DOI: 10.1007/978-3-319-63245-2_8] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The generation of reactive oxygen species (ROS) plays an important role for the maintenance of cellular processes and functions in the body. However, the excessive generation of oxygen radicals under pathological conditions such as acute lung injury (ALI) and its most severe form acute respiratory distress syndrome (ARDS) leads to increased endothelial permeability. Within this hallmark of ALI and ARDS, vascular microvessels lose their junctional integrity and show increased myosin contractions that promote the migration of polymorphonuclear leukocytes (PMNs) and the transition of solutes and fluids in the alveolar lumen. These processes all have a redox component, and this chapter focuses on the role played by ROS during the development of ALI/ARDS. We discuss the origins of ROS within the cell, cellular defense mechanisms against oxidative damage, the role of ROS in the development of endothelial permeability, and potential therapies targeted at oxidative stress.
Collapse
Affiliation(s)
- Manuela Kellner
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Satish Noonepalle
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Qing Lu
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Anup Srivastava
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Evgeny Zemskov
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA
| | - Stephen M Black
- Department of Medicine, Center for Lung Vascular Pathobiology, University of Arizona, 1501 N Campbell Ave., Tucson, AZ, 85719, USA.
| |
Collapse
|
62
|
Sepehrvand N, Ezekowitz JA. Oxygen Therapy in Patients With Acute Heart Failure. JACC-HEART FAILURE 2016; 4:783-790. [DOI: 10.1016/j.jchf.2016.03.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 02/26/2016] [Accepted: 03/03/2016] [Indexed: 01/11/2023]
|
63
|
Molecular mechanisms underlying hyperoxia acute lung injury. Respir Med 2016; 119:23-28. [DOI: 10.1016/j.rmed.2016.08.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
|
64
|
Harijith A, Pendyala S, Ebenezer DL, Ha AW, Fu P, Wang YT, Ma K, Toth PT, Berdyshev EV, Kanteti P, Natarajan V. Hyperoxia-induced p47phox activation and ROS generation is mediated through S1P transporter Spns2, and S1P/S1P1&2 signaling axis in lung endothelium. Am J Physiol Lung Cell Mol Physiol 2016; 311:L337-51. [PMID: 27343196 DOI: 10.1152/ajplung.00447.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Hyperoxia-induced lung injury adversely affects ICU patients and neonates on ventilator assisted breathing. The underlying culprit appears to be reactive oxygen species (ROS)-induced lung damage. The major contributor of hyperoxia-induced ROS is activation of the multiprotein enzyme complex NADPH oxidase. Sphingosine-1-phosphate (S1P) signaling is known to be involved in hyperoxia-mediated ROS generation; however, the mechanism(s) of S1P-induced NADPH oxidase activation is unclear. Here, we investigated various steps in the S1P signaling pathway mediating ROS production in response to hyperoxia in lung endothelium. Of the two closely related sphingosine kinases (SphKs)1 and 2, which synthesize S1P from sphingosine, only Sphk1(-/-) mice conferred protection against hyperoxia-induced lung injury. S1P is metabolized predominantly by S1P lyase and partial deletion of Sgpl1 (Sgpl1(+/-)) in mice accentuated lung injury. Hyperoxia stimulated S1P accumulation in human lung microvascular endothelial cells (HLMVECs), and downregulation of S1P transporter spinster homolog 2 (Spns2) or S1P receptors S1P1&2, but not S1P3, using specific siRNA attenuated hyperoxia-induced p47(phox) translocation to cell periphery and ROS generation in HLMVECs. These results suggest a role for Spns2 and S1P1&2 in hyperoxia-mediated ROS generation. In addition, p47(phox) (phox:phagocyte oxidase) activation and ROS generation was also reduced by PF543, a specific SphK1 inhibitor in HLMVECs. Our data indicate a novel role for Spns2 and S1P1&2 in the activation of p47(phox) and production of ROS involved in hyperoxia-mediated lung injury in neonatal and adult mice.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, National Jewish Health, Denver, Colorado; Department of Pharmacology, National Jewish Health, Denver, Colorado;
| | - Srikanth Pendyala
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - David L Ebenezer
- Department of Biochemistry & Molecular Genetics, National Jewish Health, Denver, Colorado
| | - Alison W Ha
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Panfeng Fu
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - Yue-Ting Wang
- Department of Medicinal Chemistry, National Jewish Health, Denver, Colorado
| | - Ke Ma
- Department of Pathology, National Jewish Health, Denver, Colorado
| | - Peter T Toth
- Department of Pathology, National Jewish Health, Denver, Colorado
| | | | - Prasad Kanteti
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - Viswanathan Natarajan
- Department of Pharmacology, National Jewish Health, Denver, Colorado; Department of Biochemistry & Molecular Genetics, National Jewish Health, Denver, Colorado; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
65
|
Lung extracellular matrix and redox regulation. Redox Biol 2016; 8:305-15. [PMID: 26938939 PMCID: PMC4777985 DOI: 10.1016/j.redox.2016.02.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 12/28/2022] Open
Abstract
Pulmonary fibrosis affects millions worldwide and, even though there has been a significant investment in understanding the processes involved in wound healing and maladaptive repair, a complete understanding of the mechanisms responsible for lung fibrogenesis eludes us, and interventions capable of reversing or halting disease progression are not available. Pulmonary fibrosis is characterized by the excessive expression and uncontrolled deposition of extracellular matrix (ECM) proteins resulting in erosion of the tissue structure. Initially considered an 'end-stage' process elicited after injury, these events are now considered pathogenic and are believed to contribute to the course of the disease. By interacting with integrins capable of signal transduction and by influencing tissue mechanics, ECM proteins modulate processes ranging from cell adhesion and migration to differentiation and growth factor expression. In doing so, ECM proteins help orchestrate complex developmental processes and maintain tissue homeostasis. However, poorly controlled deposition of ECM proteins promotes inflammation, fibroproliferation, and aberrant differentiation of cells, and has been implicated in the pathogenesis of pulmonary fibrosis, atherosclerosis and cancer. Considering their vital functions, ECM proteins are the target of investigation, and oxidation-reduction (redox) reactions have emerged as important regulators of the ECM. Oxidative stress invariably accompanies lung disease and promotes ECM expression directly or through the overproduction of pro-fibrotic growth factors, while affecting integrin binding and activation. In vitro and in vivo investigations point to redox reactions as targets for intervention in pulmonary fibrosis and related disorders, but studies in humans have been disappointing probably due to the narrow impact of the interventions tested, and our poor understanding of the factors that regulate these complex reactions. This review is not meant to provide a comprehensive review of this field, but rather to highlight what has been learned and to raise interest in this area in need of much attention.
Collapse
|
66
|
Diebold BA, Smith SM, Li Y, Lambeth JD. NOX2 As a Target for Drug Development: Indications, Possible Complications, and Progress. Antioxid Redox Signal 2015; 23:375-405. [PMID: 24512192 PMCID: PMC4545678 DOI: 10.1089/ars.2014.5862] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/08/2014] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE NOX2 is important for host defense, and yet is implicated in a large number of diseases in which inflammation plays a role in pathogenesis. These include acute and chronic lung inflammatory diseases, stroke, traumatic brain injury, and neurodegenerative diseases, including Alzheimer's and Parkinson's Diseases. RECENT ADVANCES Recent drug development programs have targeted several NOX isoforms that are implicated in a variety of diseases. The focus has been primarily on NOX4 and NOX1 rather than on NOX2, due, in part, to concerns about possible immunosuppressive side effects. Nevertheless, NOX2 clearly contributes to the pathogenesis of many inflammatory diseases, and its inhibition is predicted to provide a novel therapeutic approach. CRITICAL ISSUES Possible side effects that might arise from targeting NOX2 are discussed, including the possibility that such inhibition will contribute to increased infections and/or autoimmune disorders. The state of the field with regard to existing NOX2 inhibitors and targeted development of novel inhibitors is also summarized. FUTURE DIRECTIONS NOX2 inhibitors show particular promise for the treatment of inflammatory diseases, both acute and chronic. Theoretical side effects include pro-inflammatory and autoimmune complications and should be considered in any therapeutic program, but in our opinion, available data do not indicate that they are sufficiently likely to eliminate NOX2 as a drug target, particularly when weighed against the seriousness of many NOX2-related indications. Model studies demonstrating efficacy with minimal side effects are needed to encourage future development of NOX2 inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- Becky A. Diebold
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M.E. Smith
- Department of Biology and Physics, Kennesaw State University, Kennesaw, Georgia
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - J. David Lambeth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
67
|
Castillo RL, Carrasco Loza R, Romero-Dapueto C. Pathophysiological Approaches of Acute Respiratory Distress syndrome: Novel Bases for Study of Lung Injury. Open Respir Med J 2015; 9:83-91. [PMID: 26312099 PMCID: PMC4541465 DOI: 10.2174/1874306401509010083] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 04/16/2015] [Accepted: 04/16/2015] [Indexed: 12/22/2022] Open
Abstract
Experimental approaches have been implemented to research the lung damage related-mechanism. These models show in animals pathophysiological events for acute respiratory distress syndrome (ARDS), such as neutrophil activation, reactive oxygen species burst, pulmonary vascular hypertension, exudative edema, and other events associated with organ dysfunction. Moreover, these approaches have not reproduced the clinical features of lung damage. Lung inflammation is a relevant event in the develop of ARDS as component of the host immune response to various stimuli, such as cytokines, antigens and endotoxins. In patients surviving at the local inflammatory states, transition from injury to resolution is an active mechanism regulated by the immuno-inflammatory signaling pathways. Indeed, inflammatory process is regulated by the dynamics of cell populations that migrate to the lung, such as neutrophils and on the other hand, the role of the modulation of transcription factors and reactive oxygen species (ROS) sources, such as nuclear factor kappaB and NADPH oxidase. These experimental animal models reproduce key components of the injury and resolution phases of human ALI/ARDS and provide a methodology to explore mechanisms and potential new therapies.
Collapse
Affiliation(s)
- R L Castillo
- Programa de Fisiopatología, Facultad de Medicina, Universidad de Chile, Chile
| | - R Carrasco Loza
- Departamento de Medicina, Hospital del Salvador, Santiago, Chile; Laboratorio de Investigación Biomédica, Hospital del Salvador, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - C Romero-Dapueto
- Servicio de Medicina Física y Rehabilitación, Clínica Alemana de Santiago, Santiago, Chile
| |
Collapse
|
68
|
Datta A, Kim GA, Taylor JM, Gugino SF, Farrow KN, Schumacker PT, Berkelhamer SK. Mouse lung development and NOX1 induction during hyperoxia are developmentally regulated and mitochondrial ROS dependent. Am J Physiol Lung Cell Mol Physiol 2015; 309:L369-77. [PMID: 26092998 DOI: 10.1152/ajplung.00176.2014] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 06/16/2015] [Indexed: 01/08/2023] Open
Abstract
Animal models demonstrate that exposure to supraphysiological oxygen during the neonatal period compromises both lung and pulmonary vascular development, resulting in a phenotype comparable to bronchopulmonary dysplasia (BPD). Our prior work in murine models identified postnatal maturation of antioxidant enzyme capacities as well as developmental regulation of mitochondrial oxidative stress in hyperoxia. We hypothesize that consequences of hyperoxia may also be developmentally regulated and mitochondrial reactive oxygen species (ROS) dependent. To determine whether age of exposure impacts the effect of hyperoxia, neonatal mice were placed in 75% oxygen for 72 h at either postnatal day 0 (early postnatal) or day 4 (late postnatal). Mice exposed to early, but not late, postnatal hyperoxia demonstrated decreased alveolarization and septation, increased muscularization of resistance pulmonary arteries, and right ventricular hypertrophy (RVH) compared with normoxic controls. Treatment with a mitochondria-specific antioxidant, (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mitoTEMPO), during early postnatal hyperoxia protected against compromised alveolarization and RVH. In addition, early, but not late, postnatal hyperoxia resulted in induction of NOX1 expression that was mitochondrial ROS dependent. Because early, but not late, exposure resulted in compromised lung and cardiovascular development, we conclude that the consequences of hyperoxia are developmentally regulated and decrease with age. Attenuated disease in mitoTEMPO-treated mice implicates mitochondrial ROS in the pathophysiology of neonatal hyperoxic lung injury, with potential for amplification of ROS signaling through NOX1 induction. Furthermore, it suggests a potential role for targeted antioxidant therapy in the prevention or treatment of BPD.
Collapse
Affiliation(s)
- Ankur Datta
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Gina A Kim
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Joann M Taylor
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Sylvia F Gugino
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Kathryn N Farrow
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Paul T Schumacker
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
69
|
Sela M, Tirza G, Ravid O, Volovitz I, Solodeev I, Friedman O, Zipori D, Gur E, Krelin Y, Shani N. NOX1-induced accumulation of reactive oxygen species in abdominal fat-derived mesenchymal stromal cells impinges on long-term proliferation. Cell Death Dis 2015; 6:e1728. [PMID: 25880095 PMCID: PMC4650551 DOI: 10.1038/cddis.2015.84] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 01/22/2015] [Accepted: 02/16/2015] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and can be derived from different adult tissues including fat. Our repeated attempts to produce long-term proliferative cultures of rat abdominal adipose stem cells (aASCs) under normal oxygen concentration (21%) were unsuccessful. We set to examine the events controlling this cytostasis of aASCs and found that it resulted from overproduction of reactive oxygen species (ROS) that led to apoptosis. ROS overproduction in aASCs was accompanied by increased expression of NOX1 but not of NOX2 or NOX4. NOX family members are an important source of intracellular ROS pointing to NOX1 involvement in ROS accumulation. This was verified when aASCs that were grown under 3% oxygen conditions expanded long term, displaying reduced NOX1 expression and decreased ROS accumulation. NOX1 involvement in aASC cytostasis was reaffirmed when cells that were expanded under normoxic conditions in the presence of a specific NOX1 inhibitor, ML171, demonstrated reduced ROS accumulation, reduced apoptosis and long-term expansion. aASC expansion arrest was accompanied also by a weak fat differentiation and migratory potential, which was enhanced by NOX1 inhibition. This suggests an inhibitory role for NOX1-induced ROS overproduction on aASCs, their fat differentiation and migratory potential. In contrast to aASCs, similar cells produced from subcutaneous fat were easily expanded in normoxic cultures, exhibiting low ROS concentrations, a low number of apoptotic cells and improved fat differentiation and migration. Taken together, our results show, for the first time, that NOX1-induced ROS accumulation halts ASC expansion and reduces their differentiation and migratory potential under normoxic conditions. Importantly, this phenotype comprises a tissue-specific signature as it was evident in aASCs but not in subcutaneous ASCs. NOX-induced ROS accumulation and cytokine production by fat are part of the metabolic syndrome. The similarity of this phenomenon to aASC phenotype may indicate that they arise from similar molecular mechanisms.
Collapse
Affiliation(s)
- M Sela
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - G Tirza
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - O Ravid
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - I Volovitz
- The Neurosurgery Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - I Solodeev
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - O Friedman
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - D Zipori
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - E Gur
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Y Krelin
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - N Shani
- The Plastic Surgery Department Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
70
|
Wu CY, Bermúdez-Humarán LG, Yue F, Li M, Zhang LP. Intranasal administration with recombinant Lactococcus lactis expressing heme oxygenase-1 reduces hyperoxia-induced lung inflammation in rat pups. Biotechnol Lett 2015; 37:1203-11. [DOI: 10.1007/s10529-015-1795-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 02/17/2015] [Indexed: 01/07/2023]
|
71
|
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: 1.7] [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]
|
72
|
Abstract
Determining the role of NADPH oxidases in the context of virus infection is an emerging area of research and our knowledge is still sparse. The expression of various isoforms of NOX/DUOX (NADPH oxidase/dual oxidase) in the epithelial cells (ECs) lining the respiratory tract renders them primary sites from which to orchestrate the host defence against respiratory viruses. Accumulating evidence reveals distinct facets of the involvement of NOX/DUOX in host antiviral and pro-inflammatory responses and in the control of the epithelial barrier integrity, with individual isoforms mediating co-operative, but surprisingly also opposing, functions. Although in vivo studies in mice are in line with some of these observations, a complete understanding of the specific functions of epithelial NOX/DUOX awaits lung epithelial-specific conditional knockout mice. The goal of the present review is to summarize our current knowledge of the role of individual NOX/DUOX isoforms expressed in the lung epithelium in the context of respiratory virus infections so as to highlight potential opportunities for therapeutic intervention.
Collapse
|
73
|
Benipal B, Feinstein SI, Chatterjee S, Dodia C, Fisher AB. Inhibition of the phospholipase A2 activity of peroxiredoxin 6 prevents lung damage with exposure to hyperoxia. Redox Biol 2015; 4:321-7. [PMID: 25637741 PMCID: PMC4803794 DOI: 10.1016/j.redox.2015.01.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 01/08/2015] [Accepted: 01/10/2015] [Indexed: 11/16/2022] Open
Abstract
Lung injury associated with hyperoxia reflects in part the secondary effects of pulmonary inflammation and the associated production of reactive oxygen species due to activation of NADPH oxidase, type 2 (NOX2). Activation of NOX2 requires the phospholipase A2 (PLA2) activity of peroxiredoxin 6 (Prdx6). Therefore, we evaluated whether blocking Prdx6 PLA2 activity using the inhibitor MJ33 would be protective in a mouse model of acute lung injury resulting from hyperoxic exposure. Mice were treated with an intraperitoneal injection of MJ33 (2.5nmol/g body weight) at the start of exposure (zero time) and at 48h during continuous exposure to 100% O2 for 80h. Treatment with MJ33 reduced the number of neutrophils and the protein content in the fluid obtained by bronchoalveolar lavage, inhibited the increase in lipid peroxidation products in lung tissue, decreased the number of apoptotic cells in the lung, and decreased the perivascular edema associated with the 80h exposure to hyperoxia. Thus, blocking Prdx6 PLA2 activity by MJ33 significantly protected lungs against damage from hyperoxia, presumably by preventing the activation of NOX2 and the amplification of lung injury associated with inflammation. These findings demonstrate that MJ33, a potent inhibitor of Prdx6 PLA2 activity, can protect mouse lungs against the manifestations of acute lung injury due to oxidative stress.
Collapse
Affiliation(s)
- Bavneet Benipal
- Institute for Environmental Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sheldon I Feinstein
- Institute for Environmental Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shampa Chatterjee
- Institute for Environmental Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandra Dodia
- Institute for Environmental Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aron B Fisher
- Institute for Environmental Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
74
|
Trocme C, Deffert C, Cachat J, Donati Y, Tissot C, Papacatzis S, Braunersreuther V, Pache JC, Krause KH, Holmdahl R, Barazzone-Argiroffo C, Carnesecchi S. Macrophage-specific NOX2 contributes to the development of lung emphysema through modulation of SIRT1/MMP-9 pathways. J Pathol 2014; 235:65-78. [PMID: 25116588 PMCID: PMC4280678 DOI: 10.1002/path.4423] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 12/19/2022]
Abstract
Reactive oxygen species (ROS) participate in the pathogenesis of emphysema. Among ROS-producing enzymes, NOX NADPH oxidases are thought to be responsible for tissue injury associated with several lung pathologies. To determine whether NOX2 and/or NOX1 participate in the development of emphysema, their expression patterns were first studied by immunohistochemistry in the lungs of emphysematous patients. Subsequently, we investigated their contribution to elastase-induced emphysema using NOX2- and NOX1-deficient mice. In human lung, NOX2 was mainly detected in macrophages of control and emphysematous lungs, while NOX1 was expressed in alveolar epithelium and bronchial cells. We observed an elevated number of NOX2-positive cells in human emphysematous lungs, as well as increased NOX2 and NOX1 mRNA expression in mouse lungs following elastase exposure. Elastase-induced alveolar airspace enlargement and elastin degradation were prevented in NOX2-deficient mice, but not in NOX1-deficient mice. This protection was independent of inflammation and correlated with reduced ROS production. Concomitantly, an elevation of sirtuin 1 (SIRT1) level and a decrease of matrix metalloproteinase-9 (MMP-9) expression and activity were observed in alveolar macrophages and neutrophils. We addressed the specific role of macrophage-restricted functional NOX2 in elastase-induced lung emphysema using Ncf1 mutant mice and Ncf1 macrophage rescue mice (Ncf1 mutant mice with transgenic expression of Ncf1 only in CD68-positive mononuclear phagocytes; the MN mouse). Compared to WT mice, the lack of functional NOX2 led to decreased elastase-induced ROS production and protected against emphysema. In contrast, ROS production was restored specifically in macrophages from Ncf1 rescue mice and contributes to emphysema. Taken together, our results demonstrate that NOX2 is involved in the pathogenesis of human emphysema and macrophage-specific NOX2 participates in elastase-induced emphysema through the involvement of SIRT1/MMP-9 pathways in mice.
Collapse
Affiliation(s)
- Candice Trocme
- Laboratory of Protein and Enzyme Biochemistry, University Hospital, Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Kim MJ, Ryu JC, Kwon Y, Lee S, Bae YS, Yoon JH, Ryu JH. Dual oxidase 2 in lung epithelia is essential for hyperoxia-induced acute lung injury in mice. Antioxid Redox Signal 2014; 21:1803-18. [PMID: 24766345 PMCID: PMC4203470 DOI: 10.1089/ars.2013.5677] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS Acute lung injury (ALI) induced by excessive hyperoxia has been employed as a model of oxidative stress imitating acute respiratory distress syndrome. Under hyperoxic conditions, overloading quantities of reactive oxygen species (ROS) are generated in both lung epithelial and endothelial cells, leading to ALI. Some NADPH oxidase (NOX) family enzymes are responsible for hyperoxia-induced ROS generation in lung epithelial and endothelial cells. However, the molecular mechanisms of ROS production in type II alveolar epithelial cells (AECs) and ALI induced by hyperoxia are poorly understood. RESULTS In this study, we show that dual oxidase 2 (DUOX2) is a key NOX enzyme that affects hyperoxia-induced ROS production, particularly in type II AECs, leading to lung injury. In DUOX2 mutant mice (DUOX2(thyd/thyd)) or mice in which DUOX2 expression is knocked down in the lungs, hyperoxia-induced ALI was significantly lower than in wild-type (WT) mice. DUOX2 was mainly expressed in type II AECs, but not endothelial cells, and hyperoxia-induced ROS production was markedly reduced in primary type II AECs isolated from DUOX2(thyd/thyd) mice. Furthermore, DUOX2-generated ROS are responsible for caspase-mediated cell death, inducing ERK and JNK phophorylation in type II AECs. INNOVATION To date, no role for DUOX2 has been defined in hyperoxia-mediated ALI despite it being a NOX homologue and major ROS source in lung epithelium. CONCLUSION Here, we present the novel finding that DUOX2-generated ROS induce AEC death, leading to hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Min-Ji Kim
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Chan Ryu
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Younghee Kwon
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Suhee Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Yun Soo Bae
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Joo-Heon Yoon
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Hwan Ryu
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
76
|
Zanetti F, Giacomello M, Donati Y, Carnesecchi S, Frieden M, Barazzone-Argiroffo C. Nicotine mediates oxidative stress and apoptosis through cross talk between NOX1 and Bcl-2 in lung epithelial cells. Free Radic Biol Med 2014; 76:173-84. [PMID: 25151121 DOI: 10.1016/j.freeradbiomed.2014.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/18/2014] [Accepted: 08/01/2014] [Indexed: 02/07/2023]
Abstract
Nicotine contributes to the onset and progression of several pulmonary diseases. Among the various pathophysiological mechanisms triggered by nicotine, oxidative stress and cell death are reported in several cell types. We found that chronic exposure to nicotine (48h) induced NOX1-dependent oxidative stress and apoptosis in primary pulmonary cells. In murine (MLE-12) and human (BEAS-2B) lung epithelial cell lines, nicotine acted as a sensitizer to cell death and synergistically enhanced apoptosis when cells were concomitantly exposed to hyperoxia. The precise signaling pathway was investigated in MLE-12 cells in which NOX1 was abrogated by a specific inhibitor or stably silenced by shRNA. In the early phase of exposure (1h), nicotine mediated intracellular Ca(2+) fluxes and activation of protein kinase C, which in its turn activated NOX1, leading to cellular and mitochondrial oxidative stress. The latter triggered the intrinsic apoptotic machinery by modulating the expression of Bcl-2 and Bax. Overexpression of Bcl-2 completely prevented nicotine's detrimental effects, suggesting Bcl-2as a downstream key regulator in nicotine/NOX1-induced cell damage. These results suggest that NOX1 is a major contributor to the generation of intracellular oxidative stress induced by nicotine and might be an important molecule to target in nicotine-related lung pathologies.
Collapse
Affiliation(s)
- Filippo Zanetti
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Marta Giacomello
- Venetian Institute of Molecular Medicine, University of Padova, Padova, Italy
| | - Yves Donati
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Stephanie Carnesecchi
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Constance Barazzone-Argiroffo
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
77
|
Wedgwood S, Steinhorn RH. Role of reactive oxygen species in neonatal pulmonary vascular disease. Antioxid Redox Signal 2014; 21:1926-42. [PMID: 24350610 PMCID: PMC4202910 DOI: 10.1089/ars.2013.5785] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Abnormal lung development in the perinatal period can result in severe neonatal complications, including persistent pulmonary hypertension (PH) of the newborn and bronchopulmonary dysplasia. Reactive oxygen species (ROS) play a substantive role in the development of PH associated with these diseases. ROS impair the normal pulmonary artery (PA) relaxation in response to vasodilators, and ROS are also implicated in pulmonary arterial remodeling, both of which can increase the severity of PH. RECENT ADVANCES PA ROS levels are elevated when endogenous ROS-generating enzymes are activated and/or when endogenous ROS scavengers are inactivated. Animal models have provided valuable insights into ROS generators and scavengers that are dysregulated in different forms of neonatal PH, thus identifying potential therapeutic targets. CRITICAL ISSUES General antioxidant therapy has proved ineffective in reversing PH, suggesting that it is necessary to target specific signaling pathways for successful therapy. FUTURE DIRECTIONS Development of novel selective pharmacologic inhibitors along with nonantioxidant therapies may improve the treatment outcomes of patients with PH, while further investigation of the underlying mechanisms may enable earlier detection of the disease.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, University of California Davis Medical Center , Sacramento, California
| | | |
Collapse
|
78
|
Wu F, Szczepaniak WS, Shiva S, Liu H, Wang Y, Wang L, Wang Y, Kelley EE, Chen AF, Gladwin MT, McVerry BJ. Nox2-dependent glutathionylation of endothelial NOS leads to uncoupled superoxide production and endothelial barrier dysfunction in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 307:L987-97. [PMID: 25326583 DOI: 10.1152/ajplung.00063.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microvascular barrier integrity is dependent on bioavailable nitric oxide (NO) produced locally by endothelial NO synthase (eNOS). Under conditions of limited substrate or cofactor availability or by enzymatic modification, eNOS may become uncoupled, producing superoxide in lieu of NO. This study was designed to investigate how eNOS-dependent superoxide production contributes to endothelial barrier dysfunction in inflammatory lung injury and its regulation. C57BL/6J mice were challenged with intratracheal LPS. Bronchoalveolar lavage fluid was analyzed for protein accumulation, and lung tissue homogenate was assayed for endothelial NOS content and function. Human lung microvascular endothelial cell (HLMVEC) monolayers were exposed to LPS in vitro, and barrier integrity and superoxide production were measured. Biopterin species were quantified, and coimmunoprecipitation (Co-IP) assays were performed to identify protein interactions with eNOS that putatively drive uncoupling. Mice exposed to LPS demonstrated eNOS-dependent increased alveolar permeability without evidence for altered canonical NO signaling. LPS-induced superoxide production and permeability in HLMVEC were inhibited by the NOS inhibitor nitro-l-arginine methyl ester, eNOS-targeted siRNA, the eNOS cofactor tetrahydrobiopterin, and superoxide dismutase. Co-IP indicated that LPS stimulated the association of eNOS with NADPH oxidase 2 (Nox2), which correlated with augmented eNOS S-glutathionylation both in vitro and in vivo. In vitro, Nox2-specific inhibition prevented LPS-induced eNOS modification and increases in both superoxide production and permeability. These data indicate that eNOS uncoupling contributes to superoxide production and barrier dysfunction in the lung microvasculature after exposure to LPS. Furthermore, the results implicate Nox2-mediated eNOS-S-glutathionylation as a mechanism underlying LPS-induced eNOS uncoupling in the lung microvasculature.
Collapse
Affiliation(s)
- Feng Wu
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - William S Szczepaniak
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Pharmacology, Pittsburgh, Pennsylvania
| | - Huanbo Liu
- University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Yinna Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ling Wang
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Ying Wang
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania
| | - Eric E Kelley
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Anesthesiology, Pittsburgh, Pennsylvania
| | - Alex F Chen
- University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine Department of Surgery, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Bryan J McVerry
- University of Pittsburgh School of Medicine Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania; University of Pittsburgh Vascular Medicine Institute, Pittsburgh, Pennsylvania;
| |
Collapse
|
79
|
Liu YY, Li LF, Fu JY, Kao KC, Huang CC, Chien Y, Liao YW, Chiou SH, Chang YL. Induced pluripotent stem cell therapy ameliorates hyperoxia-augmented ventilator-induced lung injury through suppressing the Src pathway. PLoS One 2014; 9:e109953. [PMID: 25310015 PMCID: PMC4195701 DOI: 10.1371/journal.pone.0109953] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/12/2014] [Indexed: 01/20/2023] Open
Abstract
Background High tidal volume (VT) mechanical ventilation (MV) can induce the recruitment of neutrophils, release of inflammatory cytokines and free radicals, and disruption of alveolar epithelial and endothelial barriers. It is proposed to be the triggering factor that initiates ventilator-induced lung injury (VILI) and concomitant hyperoxia further aggravates the progression of VILI. The Src protein tyrosine kinase (PTK) family is one of the most critical families to intracellular signal transduction related to acute inflammatory responses. The anti-inflammatory abilities of induced pluripotent stem cells (iPSCs) have been shown to improve acute lung injuries (ALIs); however, the mechanisms regulating the interactions between MV, hyperoxia, and iPSCs have not been fully elucidated. In this study, we hypothesize that Src PTK plays a critical role in the regulation of oxidants and inflammation-induced VILI during hyperoxia. iPSC therapy can ameliorate acute hyperoxic VILI by suppressing the Src pathway. Methods Male C57BL/6 mice, either wild-type or Src-deficient, aged between 2 and 3 months were exposed to high VT (30 mL/kg) ventilation with or without hyperoxia for 1 to 4 h after the administration of Oct4/Sox2/Parp1 iPSCs at a dose of 5×107 cells/kg of mouse. Nonventilated mice were used for the control groups. Results High VT ventilation during hyperoxia further aggravated VILI, as demonstrated by the increases in microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 (MIP-2) and plasminogen activator inhibitor-1 (PAI-1) production, Src activation, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and malaldehyde (MDA) level. Administering iPSCs attenuated ALI induced by MV during hyperoxia, which benefited from the suppression of Src activation, oxidative stress, acute inflammation, and apoptosis, as indicated by the Src-deficient mice. Conclusion The data suggest that iPSC-based therapy is capable of partially suppressing acute inflammatory and oxidant responses that occur during hyperoxia-augmented VILI through the inhibition of Src-dependent signaling pathway.
Collapse
Affiliation(s)
- Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail: (LFL); (YLC)
| | - Jui-Ying Fu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chin Kao
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chung-Chi Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yueh Chien
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Wen Liao
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Lih Chang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (LFL); (YLC)
| |
Collapse
|
80
|
Vlahos R, Selemidis S. NADPH Oxidases as Novel Pharmacologic Targets against Influenza A Virus Infection. Mol Pharmacol 2014; 86:747-59. [DOI: 10.1124/mol.114.095216] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
81
|
Huang B, Li Q, Xu S, Tian M, Zhen X, Bi Y, Xu F. Substance P protects against hyperoxic-induced lung injury in neonatal rats. Exp Lung Res 2014; 41:12-20. [DOI: 10.3109/01902148.2014.959140] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
82
|
Czövek D, Peták F, Donati Y, Belin X, Pache JC, Barazzone Argiroffo C, Habre W. Prevention of hyperoxia-induced bronchial hyperreactivity by sildenafil and vasoactive intestinal peptide: impact of preserved lung function and structure. Respir Res 2014; 15:81. [PMID: 25117627 PMCID: PMC4261898 DOI: 10.1186/1465-9921-15-81] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 07/02/2014] [Indexed: 01/10/2023] Open
Abstract
Objective Hyperoxia exposure leads to the development of lung injury and bronchial hyperreactivity (BHR) via involvement of nitric oxide (NO) pathway. We aimed at characterizing whether the stimulation of the NO pathway by sildenafil or vasoactive intestinal peptide (VIP) is able to prevent the hyperoxia-induced development of BHR. The respective roles of the preserved lung volume and alveolar architecture, the anti-inflammatory and anti-apoptotic potentials of these treatments in the diminished lung responsiveness were also characterized. Materials and methods Immature (28-day-old) rats were exposed for 72 hours to room air (Group C), hyperoxia (>95%, Group HC), or hyperoxia with the concomitant administration of vasoactive intestinal peptide (VIP, Group HV) or sildenafil (Group HS). Following exposure, the end-expiratory lung volume (EELV) was assessed plethysmographically. Airway and respiratory tissue mechanics were measured under baseline conditions and following incremental doses of methacholine to assess BHR. Inflammation was assessed by analyzing the bronchoalveolar lavage fluid (BALF), while biochemical and histological analyses were used to characterize the apoptotic and structural changes in the lungs. Results The BHR, the increased EELV, the aberrant alveolarization, and the infiltration of inflammatory cells into the BALF that developed in Group HC were all suppressed significantly by VIP or sildenafil treatment. The number of apoptotic cells increased significantly in Group HC, with no evidence of statistically significant effects on this adverse change in Groups HS and HV. Conclusions These findings suggest that stimulating the NO pathway by sildenafil and VIP exert their beneficial effect against hyperoxia-induced BHR via preserving normal EELV, inhibiting airway inflammation and preserving the physiological lung structure, whereas the antiapoptotic potential of these treatments were not apparent in this process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Walid Habre
- Department of Anesthesiology, Anesthesiological Investigation Unit, Pharmacology and Intensive Care, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
83
|
Choi DH, Lee KH, Kim JH, Seo JH, Kim HY, Shin CY, Han JS, Han SH, Kim YS, Lee J. NADPH oxidase 1, a novel molecular source of ROS in hippocampal neuronal death in vascular dementia. Antioxid Redox Signal 2014; 21:533-50. [PMID: 24294978 PMCID: PMC4086030 DOI: 10.1089/ars.2012.5129] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
AIMS Chronic cerebral hypoperfusion (CCH) is a common pathological factor that contributes to neurodegenerative diseases such as vascular dementia (VaD). Although oxidative stress has been strongly implicated in the pathogenesis of VaD, the molecular mechanism underlying the selective vulnerability of hippocampal neurons to oxidative damage remains unknown. We assessed whether the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) complex, a specialized superoxide generation system, plays a role in VaD by permanent ligation of bilateral common carotid arteries in rats. RESULTS Male Wistar rats (10 weeks of age) were subjected to bilateral occlusion of the common carotid arteries (two-vessel occlusion [2VO]). Nox1 expression gradually increased in hippocampal neurons, starting at 1 week after 2VO and for approximately 15 weeks after 2VO. The levels of superoxide, DNA oxidation, and neuronal death in the CA1 subfield of the hippocampus, as well as consequential cognitive impairment, were increased in 2VO rats. Both inhibition of Nox by apocynin, a putative Nox inhibitor, and adeno-associated virus-mediated Nox1 knockdown significantly reduced 2VO-induced reactive oxygen species generation, oxidative DNA damage, hippocampal neuronal degeneration, and cognitive impairment. INNOVATION AND CONCLUSION We provided evidence that neuronal Nox1 is activated in the hippocampus under CCH, causing oxidative stress and consequential hippocampal neuronal death and cognitive impairment. This evidence implies that Nox1-mediated oxidative stress plays an important role in neuronal cell death and cognitive dysfunction in VaD. Nox1 may serve as a potential therapeutic target for VaD.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
- Department of Medical Science, School of Medicine, Konkuk University, Seoul, Korea
| | - Kyoung-Hee Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Ji-Hye Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Ju-Ha Seo
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Hahn Young Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Chan Young Shin
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Jung-Soo Han
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Seol-Heui Han
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Yoon-Seong Kim
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Jongmin Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
- Rehabilitation Medicine, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
84
|
Perez M, Wedgwood S, Lakshminrusimha S, Farrow KN, Steinhorn RH. Hydrocortisone normalizes phosphodiesterase-5 activity in pulmonary artery smooth muscle cells from lambs with persistent pulmonary hypertension of the newborn. Pulm Circ 2014; 4:71-81. [PMID: 25006423 DOI: 10.1086/674903] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 11/09/2013] [Indexed: 12/15/2022] Open
Abstract
Phosphodiesterase-5 (PDE5) is the primary phosphodiesterase in the pulmonary vasculature. It degrades cyclic guanosine monophosphate (cGMP) and inhibits cGMP-mediated vasorelaxation. We previously reported that hydrocortisone treatment decreased hyperoxia-induced PDE5 activity and markers of oxidative stress in lambs with persistent pulmonary hypertension of the newborn (PPHN) ventilated with 100% O2. The objective of our study was to determine the molecular mechanism by which hydrocortisone downregulates PDE5 and oxidative stress in fetal pulmonary artery smooth muscle cells (FPASMCs) from PPHN lambs. PPHN FPASMC were incubated for 24 hours in either 21% or 95% O2. Some cells were treated with 100 nM hydrocortisone and/or ±1 μM helenalin, an inhibitor of nuclear factor κ B (NFκB), a redox-sensitive transcription factor. Exposure to hyperoxia led to increased PDE5 activity, oxidative stress, and NFκB activity. Pretreatment of PPHN FPASMC with hydrocortisone normalized PDE5 activity, decreased cytosolic oxidative stress, increased expression of extracellular superoxide dismutase and NFκB inhibitory protein, and decreased NFκB activity. Similarly, treatment with NFκB inhibitor, helenalin, decreased PDE5 activity. These data suggest that hyperoxia activates NFκB, which in turn induces PDE5 activity in PPHN FPASMC, whereas treatment with hydrocortisone attenuates these changes by blocking reactive oxygen species-induced NFκB activity.
Collapse
Affiliation(s)
- Marta Perez
- Department of Pediatrics, Northwestern University, Chicago, Illinois, USA
| | - Stephen Wedgwood
- Department of Pediatrics, University of California Davis Medical Center, Sacramento, California, USA
| | | | - Kathryn N Farrow
- Department of Pediatrics, Northwestern University, Chicago, Illinois, USA
| | - Robin H Steinhorn
- Department of Pediatrics, University of California Davis Medical Center, Sacramento, California, USA
| |
Collapse
|
85
|
Yang G, Wright CJ, Hinson MD, Fernando AP, Sengupta S, Biswas C, La P, Dennery PA. Oxidative stress and inflammation modulate Rev-erbα signaling in the neonatal lung and affect circadian rhythmicity. Antioxid Redox Signal 2014; 21:17-32. [PMID: 24252172 PMCID: PMC4048579 DOI: 10.1089/ars.2013.5539] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS The response to oxidative stress and inflammation varies with diurnal rhythms. Nevertheless, it is not known whether circadian genes are regulated by these stimuli. We evaluated whether Rev-erbα, a key circadian gene, was regulated by oxidative stress and/or inflammation in vitro and in a mouse model. RESULTS A unique sequence consisting of overlapping AP-1 and nuclear factor kappa B (NFκB) consensus sequences was identified on the mouse Rev-erbα promoter. This sequence mediates Rev-erbα promoter activity and transcription in response to oxidative stress and inflammation. This region serves as an NrF2 platform both to receive oxidative stress signals and to activate Rev-erbα, as well as an NFκB-binding site to repress Rev-erbα with inflammatory stimuli. The amplitude of the rhythmicity of Rev-erbα was altered by pre-exposure to hyperoxia or disruption of NFκB in a cell culture model of circadian simulation. Oxidative stress overcame the inhibitory effect of NFκB binding on Rev-erbα transcription. This was confirmed in neonatal mice exposed to hyperoxia, where hyperoxia-induced lung Rev-erbα transcription was further increased with NFκB disruption. Interestingly, this effect was not observed in similarly exposed adult mice. INNOVATION These data provide novel mechanistic insights into how key circadian genes are regulated by oxidative stress and inflammation in the neonatal lung. CONCLUSION Rev-erbα transcription and circadian oscillation are susceptible to oxidative stress and inflammation in the neonate. Due to Rev-erbα's role in cellular metabolism, this could contribute to lung cellular function and injury from inflammation and oxidative stress.
Collapse
Affiliation(s)
- Guang Yang
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Clyde J. Wright
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | - Maurice D. Hinson
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Amal P. Fernando
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shaon Sengupta
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Chhanda Biswas
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ping La
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Phyllis A. Dennery
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
86
|
Bernard K, Hecker L, Luckhardt TR, Cheng G, Thannickal VJ. NADPH oxidases in lung health and disease. Antioxid Redox Signal 2014; 20:2838-53. [PMID: 24093231 PMCID: PMC4026303 DOI: 10.1089/ars.2013.5608] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The evolution of the lungs and circulatory systems in vertebrates ensured the availability of molecular oxygen (O2; dioxygen) for aerobic cellular metabolism of internal organs in large animals. O2 serves as the physiologic terminal acceptor of mitochondrial electron transfer and of the NADPH oxidase (Nox) family of oxidoreductases to generate primarily water and reactive oxygen species (ROS), respectively. RECENT ADVANCES The purposeful generation of ROS by Nox family enzymes suggests important roles in normal physiology and adaptation, most notably in host defense against invading pathogens and in cellular signaling. CRITICAL ISSUES However, there is emerging evidence that, in the context of chronic stress and/or aging, Nox enzymes contribute to the pathogenesis of a number of lung diseases. FUTURE DIRECTIONS Here, we review evolving functions of Nox enzymes in normal lung physiology and emerging pathophysiologic roles in lung disease.
Collapse
Affiliation(s)
- Karen Bernard
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | | | |
Collapse
|
87
|
Sliman SM, Patel RB, Cruff JP, Kotha SR, Newland CA, Schrader CA, Sherwani SI, Gurney TO, Magalang UJ, Parinandi NL. Adiponectin protects against hyperoxic lung injury and vascular leak. Cell Biochem Biophys 2014; 67:399-414. [PMID: 22183615 DOI: 10.1007/s12013-011-9330-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adiponectin (Ad), an adipokine exclusively secreted by the adipose tissue, has emerged as a paracrine metabolic regulator as well as a protectant against oxidative stress. Pharmacological approaches of protecting against clinical hyperoxic lung injury during oxygen therapy/treatment are limited. We have previously reported that Ad inhibits the NADPH oxidase-catalyzed formation of superoxide from molecular oxygen in human neutrophils. Based on this premise, we conducted studies to determine whether (i) exogenous Ad would protect against the hyperoxia-induced barrier dysfunction in the lung endothelial cells (ECs) in vitro, and (ii) endogenously synthesized Ad would protect against hyperoxic lung injury in wild-type (WT) and Ad-overexpressing transgenic (AdTg) mice in vivo. The results demonstrated that exogenous Ad protected against the hyperoxia-induced oxidative stress, loss of glutathione (GSH), cytoskeletal reorganization, barrier dysfunction, and leak in the lung ECs in vitro. Furthermore, the hyperoxia-induced lung injury, vascular leak, and lipid peroxidation were significantly attenuated in AdTg mice in vivo. Also, AdTg mice exhibited elevated levels of total thiols and GSH in the lungs as compared with WT mice. For the first time, our studies demonstrated that Ad protected against the hyperoxia-induced lung damage apparently through attenuation of oxidative stress and modulation of thiol-redox status.
Collapse
Affiliation(s)
- Sean M Sliman
- Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Dorothy M. Davis Heart & Lung Research Institute, Department of Internal Medicine, The Ohio State University College of Medicine, 473 W. 12th Avenue, Columbus, OH, 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal 2014; 20:1126-67. [PMID: 23991888 PMCID: PMC3929010 DOI: 10.1089/ars.2012.5149] [Citation(s) in RCA: 2940] [Impact Index Per Article: 267.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract Reactive oxygen species (ROS) are key signaling molecules that play an important role in the progression of inflammatory disorders. An enhanced ROS generation by polymorphonuclear neutrophils (PMNs) at the site of inflammation causes endothelial dysfunction and tissue injury. The vascular endothelium plays an important role in passage of macromolecules and inflammatory cells from the blood to tissue. Under the inflammatory conditions, oxidative stress produced by PMNs leads to the opening of inter-endothelial junctions and promotes the migration of inflammatory cells across the endothelial barrier. The migrated inflammatory cells not only help in the clearance of pathogens and foreign particles but also lead to tissue injury. The current review compiles the past and current research in the area of inflammation with particular emphasis on oxidative stress-mediated signaling mechanisms that are involved in inflammation and tissue injury.
Collapse
Affiliation(s)
- Manish Mittal
- 1 Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois
| | | | | | | | | |
Collapse
|
89
|
NADPH oxidase 1 and its derived reactive oxygen species mediated tissue injury and repair. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:282854. [PMID: 24669283 PMCID: PMC3942082 DOI: 10.1155/2014/282854] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 12/11/2013] [Accepted: 12/17/2013] [Indexed: 01/22/2023]
Abstract
Reactive oxygen species are mostly viewed to cause oxidative damage to various cells and induce organ dysfunction after ischemia-reperfusion injury. However, they are also considered as crucial molecules for cellular signal transduction in biology. NADPH oxidase, whose only function is reactive oxygen species production, has been extensively investigated in many cell types especially phagocytes. The deficiency of NADPH oxidase extends the process of inflammation and delays tissue repair, which causes chronic granulomatous disease in patients. NADPH oxidase 1, one member of the NADPH oxidase family, is not only constitutively expressed in a variety of tissues, but also induced to increase expression in both mRNA and protein levels under many circumstances. NADPH oxidase 1 and its derived reactive oxygen species are suggested to be able to regulate inflammation reaction, cell proliferation and migration, and extracellular matrix synthesis, which contribute to the processes of tissue injury and repair.
Collapse
|
90
|
Carnesecchi S, Dunand-Sauthier I, Zanetti F, Singovski G, Deffert C, Donati Y, Cagarelli T, Pache JC, Krause KH, Reith W, Barazzone-Argiroffo C. NOX1 is responsible for cell death through STAT3 activation in hyperoxia and is associated with the pathogenesis of acute respiratory distress syndrome. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:537-551. [PMID: 24551274 PMCID: PMC3925898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 12/21/2013] [Indexed: 06/03/2023]
Abstract
Reactive oxygen species (ROS) contribute to alveolar cell death in acute respiratory distress syndrome (ARDS) and we previously demonstrated that NOX1-derived ROS contributed to hyperoxia-induced alveolar cell death in mice. The study investigates whether NOX1 expression is modulated in epithelial cells concomitantly to cell death and associated to STAT3 signaling in the exudative phase of ARDS. In addition, the role of STAT3 activation in NOX1-dependent epithelial cell death was confirmed by using a lung epithelial cell line and in mice exposed to hyperoxia. NOX1 expression, cell death and STAT3 staining were evaluated in the lungs of control and ARDS patients by immunohistochemistry. In parallel, a stable NOX1-silenced murine epithelial cell line (MLE12) and NOX1-deficient mice were used to characterize signalling pathways. In the present study, we show that NOX1 is detected in alveolar epithelial cells of ARDS patients in the exudative stage. In addition, increased alveolar epithelial cell death and phosphorylated STAT3 are observed in ARDS patients and associated with NOX1 expression. Phosphorylated STAT3 is also correlated with TUNEL staining. We also confirmed that NOX1-dependent STAT3 activation participates to alveolar epithelial cell death. Silencing and acute inhibition of NOX1 in MLE12 led to decreased cell death and cleaved-caspase 3 induced by hyperoxia. Additionally, hyperoxia-induced STAT3 phosphorylation is dependent on NOX1 expression and associated with cell death in MLE12 and mice. This study demonstrates that NOX1 is involved in human ARDS pathophysiology and is responsible for the damage occurring in alveolar epithelial cells at least in part via STAT3 signalling pathways.
Collapse
Affiliation(s)
- Stephanie Carnesecchi
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | | | - Filippo Zanetti
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Grigory Singovski
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Christine Deffert
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Yves Donati
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Thomas Cagarelli
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Jean-Claude Pache
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Walter Reith
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Constance Barazzone-Argiroffo
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| |
Collapse
|
91
|
Abstract
Oxidants play an important role in homeostatic function, but excessive oxidant generation has an adverse effect on health. The manipulation of Reactive Oxygen Species (ROS) can have a beneficial effect on various lung pathologies. However indiscriminate uses of anti-oxidant strategies have not demonstrated any consistent benefit and may be harmful. Here we propose that nuanced strategies are needed to modulate the oxidant system to obtain a beneficial result in the lung diseases such as Acute Lung Injury (ALI) and Chronic Obstructive Pulmonary Disease (COPD). We identify novel areas of lung oxidant responses that may yield fruitful therapies in the future.
Collapse
Affiliation(s)
- Praveen Mannam
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anup Srivastava
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Patty J Lee
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
92
|
Veit F, Pak O, Egemnazarov B, Roth M, Kosanovic D, Seimetz M, Sommer N, Ghofrani HA, Seeger W, Grimminger F, Brandes RP, Schermuly RT, Weissmann N. Function of NADPH oxidase 1 in pulmonary arterial smooth muscle cells after monocrotaline-induced pulmonary vascular remodeling. Antioxid Redox Signal 2013; 19:2213-31. [PMID: 23706097 DOI: 10.1089/ars.2012.4904] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIMS Chronic hypoxia induces pulmonary hypertension (PH) that is concomitant with pulmonary vascular remodeling. Reactive oxygen species (ROS) are thought to play a major role in this. Recent findings suggest that ROS production by NADPH oxidase 4 (Nox4) is important in this remodeling. We investigated whether ROS production by Nox is also important in an inflammatory model of monocrotaline (MCT)-induced PH. We examined ROS production, their possible sources, and their impact on the function of pulmonary arterial smooth muscle cells (PASMC) isolated from MCT-treated and healthy rats. RESULTS MCT-PASMC showed increased intracellular superoxide production, migration, and proliferation compared with healthy controls due to increased Nox1 expression. A comparison of PASMC from MCT- and nontreated rats revealed an up-regulation of Sod2, Nrf2, cyclin D1, and matrix metalloproteinase-9 (MMP-9) as well as an increased phosphorylation of cofilin and extracellular signal-regulated kinases (Erk). Expression of Sod2, Nrf2, and cyclin D1 and phosphorylation of cofilin and Erk were Nox1 dependent. INNOVATION The role of ROS in PH is not fully understood. Mitochondria and Nox have been suggested as sources of altered ROS generation in PH, yet it remains unclear whether increased or decreased ROS contributes to the development of PH. Our studies provide evidence that for different triggers of PH, different Nox isoforms regulate proliferation and migration of PASMC. CONCLUSION In contrast to hypoxia-induced PH, Nox1 but not Nox4 is responsible for pathophysiological proliferation and migration of PASMC in an inflammatory model of MCT-induced PH via increased superoxide production. Thus, different Nox isoforms may be targeted in different forms of PH.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellencecluster Cardio-Pulmonary System (ECCPS), German Lung Center (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen , Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Abstract
Acute lung injury (ALI) and its more severe form of clinical manifestation, the acute respiratory distress syndrome is associated with significant dysfunction in air exchange due to inflammation of the lung parenchyma. Several factors contribute to the inflammatory process, including hypoxia (inadequate oxygen), hyperoxia (higher than normal partial pressure of oxygen), inflammatory mediators (such as cytokines), infections (viral and bacterial), and environmental conditions (such as cigarette smoke or noxious gases). However, studies over the past several decades suggest that oxidants formed in the various cells of the lung including endothelial, alveolar, and epithelial cells as well as lung macrophages and neutrophils in response to the factors mentioned above mediate the pathogenesis of ALI. Oxidants modify cellular proteins, lipids, carbohydrates, and DNA to cause their aberrant function. For example, oxidation of lipids changes membrane permeability. Interestingly, recent studies also suggest that spatially and temporally regulated production of oxidants plays an important role antimicrobial defense and immunomodulatory function (such as transcription factor activation). To counteract the oxidants an arsenal of antioxidants exists in the lung to maintain the redox status, but when overwhelmed tissue injury and exacerbation of inflammation occurs. We present below the current understanding of the pathogenesis of oxidant-mediated ALI.
Collapse
Affiliation(s)
- J Vidya Sarma
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | |
Collapse
|
94
|
Parker JC. Acute lung injury and pulmonary vascular permeability: use of transgenic models. Compr Physiol 2013; 1:835-82. [PMID: 23737205 DOI: 10.1002/cphy.c100013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute lung injury is a general term that describes injurious conditions that can range from mild interstitial edema to massive inflammatory tissue destruction. This review will cover theoretical considerations and quantitative and semi-quantitative methods for assessing edema formation and increased vascular permeability during lung injury. Pulmonary edema can be quantitated directly using gravimetric methods, or indirectly by descriptive microscopy, quantitative morphometric microscopy, altered lung mechanics, high-resolution computed tomography, magnetic resonance imaging, positron emission tomography, or x-ray films. Lung vascular permeability to fluid can be evaluated by measuring the filtration coefficient (Kf) and permeability to solutes evaluated from their blood to lung clearances. Albumin clearances can then be used to calculate specific permeability-surface area products (PS) and reflection coefficients (σ). These methods as applied to a wide variety of transgenic mice subjected to acute lung injury by hyperoxic exposure, sepsis, ischemia-reperfusion, acid aspiration, oleic acid infusion, repeated lung lavage, and bleomycin are reviewed. These commonly used animal models simulate features of the acute respiratory distress syndrome, and the preparation of genetically modified mice and their use for defining specific pathways in these disease models are outlined. Although the initiating events differ widely, many of the subsequent inflammatory processes causing lung injury and increased vascular permeability are surprisingly similar for many etiologies.
Collapse
Affiliation(s)
- James C Parker
- Department of Physiology, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
95
|
Wollen EJ, Sejersted Y, Wright MS, Bik-Multanowski M, Madetko-Talowska A, Günther CC, Nygård S, Kwinta P, Pietrzyk JJ, Saugstad OD. Transcriptome profiling of the newborn mouse lung after hypoxia and reoxygenation: hyperoxic reoxygenation affects mTOR signaling pathway, DNA repair, and JNK-pathway regulation. Pediatr Res 2013; 74:536-44. [PMID: 23999071 DOI: 10.1038/pr.2013.140] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 04/01/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND The use of oxygen in acute treatment of asphyxiated term newborns is associated with increased mortality. It is unclear how hyperoxic reoxygenation after hypoxia affects transcriptional changes in the newborn lung. METHODS On postnatal day 7, C57BL/6 mice (n = 62) were randomized to 120-min hypoxia (fraction of inspired oxygen (FiO2) 0.08) or normoxia. The hypoxia group was further randomized to reoxygenation for 30 min with FiO2 0.21, 0.40, 0.60, or 1.00, and the normoxia group to FiO2 0.21 or 1.00. Transcriptome profiling was performed on homogenized lung tissue using the Affymetrix 750k expression array, and validation was carried out by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS The hypoxia-reoxygenation model induced hypoxia-inducible factor 1 (HIF-1) targets like Vegfc, Adm, and Aqp1. In total, ~70% of the significantly differentially expressed genes were detected in the two high hyperoxic groups (FiO2 0.60 and 1.00). Reoxygenation with 100% oxygen after hypoxia uniquely upregulated Gadd45g, Dusp1, Peg3, and Tgm2. Pathway analysis identified mammalian target of rapamycin (mTOR) signaling pathway, DNA repair, c-jun N-terminal kinase (JNK)-pathway regulation, and cell cycle after hyperoxic reoxygenation was applied. CONCLUSION Acute hypoxia induces HIF-1 targets independent of the reoxygenation regime applied. Hyperoxic reoxygenation affects pathways regulating cell growth and survival. DNA-damage-responsive genes are restricted to reoxygenation with 100% oxygen.
Collapse
Affiliation(s)
- Embjørg J Wollen
- Department of Pediatric Research, Women and Children's Division, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Luong KVQ, Nguyen LTH. Beneficial role of vitamin D3 in the prevention of certain respiratory diseases. Ther Adv Respir Dis 2013; 7:327-50. [PMID: 24056290 DOI: 10.1177/1753465813503029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There is evidence of aberrations in the vitamin D-endocrine system in subjects with respiratory diseases. Vitamin D deficiency is highly prevalent in patients with respiratory diseases, and patients who receive vitamin D have significantly larger improvements in inspiratory muscle strength and maximal oxygen uptake. Studies have provided an opportunity to determine which proteins link vitamin D to respiratory pathology, including the major histocompatibility complex class II molecules, vitamin D receptor, vitamin D-binding protein, chromosome P450, Toll-like receptors, poly(ADP-ribose) polymerase-1, and the reduced form of nicotinamide adenine dinucleotide phosphate. Vitamin D also exerts its effect on respiratory diseases through cell signaling mechanisms, including matrix metalloproteinases, mitogen-activated protein kinase pathways, the Wnt/β-catenin signaling pathway, prostaglandins, reactive oxygen species, and nitric oxide synthase. In conclusion, vitamin D plays a significant role in respiratory diseases. The best form of vitamin D for use in the treatment of respiratory diseases is calcitriol because it is the active metabolite of vitamin D3 and modulates inflammatory cytokine expression. Further investigation of calcitriol in respiratory diseases is needed.
Collapse
Affiliation(s)
- Khanh Vinh Quoc Luong
- Vietnamese American Medical Research Foundation, 14971 Brookhurst Street, Westminster, CA 92683, USA
| | | |
Collapse
|
97
|
Berkelhamer SK, Kim GA, Radder JE, Wedgwood S, Czech L, Steinhorn RH, Schumacker PT. Developmental differences in hyperoxia-induced oxidative stress and cellular responses in the murine lung. Free Radic Biol Med 2013; 61:51-60. [PMID: 23499839 PMCID: PMC3723750 DOI: 10.1016/j.freeradbiomed.2013.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 02/24/2013] [Accepted: 03/04/2013] [Indexed: 01/11/2023]
Abstract
Exposure of newborn mice to high inspired oxygen elicits a distinct phenotype of compromised alveolar and vascular development, although lethality during long-term exposure is lower in newborns compared to adults. As the effects of hyperoxia are mediated by excessive reactive oxygen species (ROS) generation, we hypothesized that newborn mice may exhibit enhanced expression of antioxidant defenses or attenuated ROS generation compared with adults. We measured subcellular oxidant responses to acute hyperoxia in lung slices and alveolar epithelial cells at varying time points during postnatal murine lung development. Oxidant stress was assessed using RoGFP, a ratiometric protein thiol redox sensor, targeted to the cytosol or the mitochondrial matrix. In contrast to newborn resistance to oxygen-induced mortality, cells of lung slices from younger mice demonstrated exaggerated mitochondrial matrix oxidant stress compared to adults, whereas oxidant stress responses in the cytosol were absent. Cell death in lung slices from newborn mice exposed to 48h of hyperoxia was also greater than for adults. Consistent with these findings, expression of antioxidant enzymes in newborn lungs was lower than in adults, and induction of antioxidant levels and activity during 24h of in vivo exposure was absent. However, expression of the reactive oxygen species-generating enzyme NADPH oxidase 1 was increased with hyperoxic exposure in the young but not the adult lung. Collectively, these results suggest that the greater lethality in adult animals may be more likely attributed to processes such as inflammation than to differences in antioxidant defenses. Therapies for neonatal and adult oxidative lung injury should therefore consider and address developmental differences in oxidative stress responses.
Collapse
Affiliation(s)
- Sara K. Berkelhamer
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| | - Gina A. Kim
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| | - Josiah E. Radder
- Department of Pulmonary and Critical Care Medicine, 240 E. Huron Ave, McGaw Mezzanine, Northwestern University, Chicago, IL. 60611 USA
| | - Stephen Wedgwood
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| | - Lyubov Czech
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| | - Robin H. Steinhorn
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| | - Paul T. Schumacker
- Department of Pediatrics, 310 E. Superior St, Morton Building, Northwestern University, Chicago IL. 60611 USA
| |
Collapse
|
98
|
Budinger GRS, Mutlu GM. Balancing the risks and benefits of oxygen therapy in critically III adults. Chest 2013; 143:1151-1162. [PMID: 23546490 DOI: 10.1378/chest.12-1215] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oxygen therapy is an integral part of the treatment of critically ill patients. Maintenance of adequate oxygen delivery to vital organs often requires the administration of supplemental oxygen, sometimes at high concentrations. Although oxygen therapy is lifesaving, it may be associated with deleterious effects when administered for prolonged periods at high concentrations. Here, we review the recent advances in our understanding of the molecular responses to hypoxia and high levels of oxygen and review the current guidelines for oxygen therapy in critically ill patients.
Collapse
Affiliation(s)
- G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.
| | - Gökhan M Mutlu
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
99
|
Yen FL, Tsai MH, Yang CM, Liang CJ, Lin CC, Chiang YC, Lee HC, Ko HH, Lee CW. Curcumin nanoparticles ameliorate ICAM-1 expression in TNF-α-treated lung epithelial cells through p47 (phox) and MAPKs/AP-1 pathways. PLoS One 2013; 8:e63845. [PMID: 23671702 PMCID: PMC3650060 DOI: 10.1371/journal.pone.0063845] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 04/09/2013] [Indexed: 12/31/2022] Open
Abstract
Upregulation of intercellular adhesion molecule-1 (ICAM-1) involves adhesions between both circulating and resident leukocytes and the human lung epithelial cells during lung inflammatory reactions. We have previously demonstrated that curcumin-loaded polyvinylpyrrolidone nanoparticles (CURN) improve the anti-inflammatory and anti-oxidative properties of curcumin in hepatocytes. In this study, we focused on the effects of CURN on the expression of ICAM-1 in TNF-α-treated lung epithelial cells and compared these to the effects of curcumin water preparation (CURH). TNF-αinduced ICAM-1 expression, ROS production, and cell-cell adhesion were significantly attenuated by the pretreatment with antioxidants (DPI, APO, or NAC) and CURN, but not by CURH, as revealed by western blot analysis, RT-PCR, promoter assay, and ROS detection and adhesion assay. In addition, treatment of TNF-α-treated cells with CURN and antioxidants also resulted in an inhibition of activation of p47 (phox) and phosphorylation of MAPKs, as compared to that using CURH. Our findings also suggest that phosphorylation of MAPKs may eventually lead to the activation of transcription factors. We also observed that the effects of TNF-α treatment for 30 min, which includes a significant increase in the binding activity of AP-1 and phosphorylation of c-jun and c-fos genes, were reduced by CURN treatment. In vivo studies have revealed that CURN improved the anti-inflammation activities of CURH in the lung epithelial cells of TNF-α-treated mice. Our results indicate that curcumin-loaded polyvinylpyrrolidone nanoparticles may potentially serve as an anti-inflammatory drug for the treatment of respiratory diseases.
Collapse
Affiliation(s)
- Feng-Lin Yen
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Horng Tsai
- Division of Neonatology and Pediatric Hematology/Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chan-Jung Liang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Ching Lin
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Chang Chiang
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung, Taiwan
- China Medical University, Taichung, Taiwan
| | - Hui-Chun Lee
- Division of Basic Medical Sciences, Department of Nursing, and Chronic Diseases and Health Promotion Research Center, Chang Gung Institute of Technology, Chia-Yi, Taiwan
| | - Horng-Huey Ko
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiang-Wen Lee
- Division of Basic Medical Sciences, Department of Nursing, and Chronic Diseases and Health Promotion Research Center, Chang Gung Institute of Technology, Chia-Yi, Taiwan
| |
Collapse
|
100
|
Selemidis S, Seow HJ, Broughton BRS, Vinh A, Bozinovski S, Sobey CG, Drummond GR, Vlahos R. Nox1 oxidase suppresses influenza a virus-induced lung inflammation and oxidative stress. PLoS One 2013; 8:e60792. [PMID: 23577160 PMCID: PMC3620107 DOI: 10.1371/journal.pone.0060792] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 03/02/2013] [Indexed: 12/15/2022] Open
Abstract
Influenza A virus infection is an ongoing clinical problem and thus, there is an urgent need to understand the mechanisms that regulate the lung inflammation in order to unravel novel generic pharmacological strategies. Evidence indicates that the Nox2-containing NADPH oxidase enzyme promotes influenza A virus-induced lung oxidative stress, inflammation and dysfunction via ROS generation. In addition, lung epithelial and endothelial cells express the Nox1 isoform of NADPH oxidase, placing this enzyme at key sites to regulate influenza A virus-induced lung inflammation. The aim of this study was to investigate whether Nox1 oxidase regulates the inflammatory response and the oxidative stress to influenza infection in vivo in mice. Male WT and Nox1-deficient (Nox1−/y) mice were infected with the moderately pathogenic HkX-31 (H3N2, 1×104 PFU) influenza A virus for analysis of bodyweight, airways inflammation, oxidative stress, viral titre, lung histopathology, and cytokine/chemokine expression at 3 and 7 days post infection. HkX-31 virus infection of Nox1−/y mice resulted in significantly greater: loss of bodyweight (Day 3); BALF neutrophilia, peri-bronchial, peri-vascular and alveolar inflammation; Nox2-dependent inflammatory cell ROS production and peri-bronchial, epithelial and endothelial oxidative stress. The expression of pro-inflammatory cytokines including CCL2, CCL3, CXCL2, IL-1β, IL-6, GM-CSF and TNF-α was higher in Nox1−/y lungs compared to WT mice at Day 3, however, the expression of CCL2, CCL3, CXCL2, IFN-γ and the anti-inflammatory cytokine IL-10 were lower in lungs of Nox1−/y mice vs. WT mice at Day 7. Lung viral titre, and airways infiltration of active CD8+ and CD4+ T lymphocytes, and of Tregs were similar between WT and Nox1−/y mice. In conclusion, Nox1 oxidase suppresses influenza A virus induced lung inflammation and oxidative stress in mice particularly at the early phases of the infection. Nox1 and Nox2 oxidases appear to have opposing roles in the regulation of inflammation caused by influenza A viruses.
Collapse
Affiliation(s)
- Stavros Selemidis
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
- * E-mail: (SS); (RV)
| | - Huei Jiunn Seow
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Antony Vinh
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Steven Bozinovski
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Grant R. Drummond
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Ross Vlahos
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (SS); (RV)
| |
Collapse
|