Abstract
High concentrations of oxygen (hyperoxia) are routinely used during anesthesia, and supplemental oxygen is also administered in connection with several other clinical conditions. Although prolonged hyperoxia is known to cause acute lung injury (ALI), whether short-duration hyperoxia causes lung toxicity remains unknown. We exposed mice to room air (RA or 21% O2) or 60% oxygen alone or in combination with 2% isoflurane for 2 h and determined the expression of oxidative stress marker genes, DNA damage and DNA repair genes, and expression of cell cycle regulatory proteins using quantitative PCR and Western analyses. Furthermore, we determined cellular apoptosis using TUNEL assay and assessed the DNA damage product 8-hydroxy-2′-deoxyguanosine (8-Oxo-dG) in the urine of 60% hyperoxia-exposed mice. Our study demonstrates that short-duration hyperoxia causes mitochondrial and nuclear DNA damage and that isoflurane abrogates this DNA damage and decreases apoptosis when used in conjunction with hyperoxia. In contrast, isoflurane mixed with RA caused significant 8-Oxo-dG accumulations in the mitochondria and nucleus. We further show that whereas NADPH oxidase is a major source of superoxide anion generated by isoflurane in normoxia, isoflurane inhibits superoxide generation in hyperoxia. Additionally, isoflurane also protected the mouse lungs against ALI (95% O2 for 36-h exposure). Our study established that short-duration hyperoxia causes genotoxicity in the lungs, which is abrogated when hyperoxia is used in conjunction with isoflurane, but isoflurane alone causes genotoxicity in the lung when delivered with ambient air.
Keywords: DNA damage, DNA repair, hyperoxia, isoflurane, lung
INTRODUCTION
Oxygen therapy is a common medical necessity that has significant negative side effects (11, 54, 55). For example, in addition to direct pulmonary toxicity, hyperoxia has been implicated in the progression of several pulmonary disorders, including bronchopulmonary dysplasia (5, 58), acute respiratory distress syndrome (9, 23, 35), persistent pulmonary hypertension of the newborn (1, 50), and lung fibrosis (20). In addition to its routine use during anesthesia, oxygen is also administered in connection with several other clinical conditions (28, 43). Additionally, pure oxygen is often delivered to patients before specific procedures (e.g., intubation, extubation, and endoscopy) because of the potential for brief episodes of airway loss and transient hypoxemia (43). Breathing 100% oxygen before performance of these procedures increases the time that apnea can be tolerated should misadventure prevent immediate establishment of a patent airway. Considering its wide application, it is difficult to eliminate high oxygen concentrations from clinical use. On the contrary, it would be highly desirable to co-deliver oxygen with a gas that can prevent or eliminate the toxic effects of high oxygen exposure. Whereas prolonged breathing of oxygen at high concentrations is known to cause lung damage in animal experiments (22, 36, 42), the effects of short-duration exposure to hyperoxia on lung injury remains unknown. Almost all animal or cell culture experiments use 24 to 96 h of 90%–95% oxygen exposure to induce lung injury or to induce cell death in cultured cells (36). Thus, there is a paucity of studies that address the effect of short-duration hyperoxia exposure on lung injury. Because hyperoxia is used for short duration in several clinical situations, we investigated whether short-duration hyperoxia causes pulmonary oxygen toxicity.
Isoflurane is a halogenated volatile anesthetic that is widely used perioperatively. Although published results show both beneficial and detrimental effects of isoflurane on murine lung inflammation and sepsis, the role of isoflurane on hyperoxia-induced lung injury remains, to our knowledge, unexplored. For example, a recent publication indicates that isoflurane protects against endotoxin-mediated lung injury by preserving epithelial tight junction integrity (24) and in inflammation during sepsis (34). Isoflurane has also been shown to decrease apoptosis of endothelial cells exposed to tumor necrosis factor alpha (TNFα) (4). However, other findings indicate that isoflurane produces reactive oxygen species (ROS), which are critical for preconditioning against lipopolysaccharide (LPS)-mediated acute lung injury (ALI) (10). Although isoflurane is almost always used in combination with high oxygen concentrations for short durations (1–5 h or more) during surgery, the detrimental effects of short-duration hyperoxia on lung and the effect of isoflurane on possible lung injury caused by short-duration hyperoxia exposure remain unknown. Furthermore, the effect of isoflurane on hyperoxia-induced ALI has never been investigated, although several studies have reported the beneficial effect of isoflurane or isoflurane plus oxygen in alleviation of lung injury and inflammation caused by endotoxin or sepsis (3, 32, 34).
It has been established that elevated levels of ROS are generated in response to hyperoxia in the mitochondria and by NADPH oxidase (Nox) activation (15, 16, 18, 57). Since ROS can induce DNA lesions, as well as damage proteins and lipids, it is believed that the extensive damage to lung cells and lung architecture occurs as a consequence of hyperoxic insult. Mechanistically, it has been demonstrated that hyperoxic conditions contribute to lung injury and its progression as a consequence of enhanced DNA damage (6, 45, 49, 60). The cellular response to DNA damage is a complex process involving a number of proteins that recognize the process and initiate a signaling cascade, which results in the activation, or inhibition, of various effector molecules required for regulating DNA repair, cellular proliferation, and apoptosis (2, 45). Base excision repair (BER) is a major pathway involved in the repair of oxidative DNA damage caused by hyperoxia. Overexpression of apurinic endonuclease 1 (APE1) or Oxo guanine glycosylase (Ogg1) has been shown to protect cells against hyperoxia-mediated lung injury (59). Both of these enzymes belong to the BER class that removes nucleotide bases that have been damaged by oxidative stress (59).
In this report, we investigated the effect of short-duration (2 h) hyperoxia (60%) on cultured mouse alveolar type II cells (MLE-12) and primary type I, type II, and endothelial cells isolated from murine lung. In addition, we also utilized human primary microvascular endothelial cells from lung (HMVEC-L) and the mouse lung in vivo for these studies. We demonstrate that short-duration hyperoxia causes increased production of O2·− by mitochondria and NADPH oxidases that causes pulmonary genotoxicity. In contrast, isoflurane delivered with hyperoxia decreases O2·− generation and consequently alleviates oxygen toxicity. However, isoflurane alone produces significant O2·− via NADPH oxidase. Isoflurane in normoxia causes significant lung genotoxicity and apoptosis of lung cells. Furthermore, we show that lung injury due to prolonged inhalation of hyperoxia is significantly reduced when delivered with low concentrations of isoflurane.
MATERIALS AND METHODS
Cell culture and oxygen exposure.
MLE-12 cells were cultured in HITES (hydrocortisone, insulin, transferrin, estradiol and selenium) medium (14) supplemented with 2% fetal bovine serum. Actively growing MLE-12 cells were exposed to normoxia [21% oxygen, room air (RA)] or hyperoxia (60% oxygen) along with or without 2% isoflurane for 2 or 16 h in a Modular Incubator Chamber (Billups-Rothenberg, San Diego, CA). HMVEC-L were cultured in endothelial basal medium (EBM) with growth supplements (Lonza).
Antibodies.
Anti-p53 (sc-6243), anti-Cdc25A (sc-97), anti-Chk1 (sc-8408), anti-cyclin B1 (sc-7393), anti-Ape1 (sc-17774), anti-SPC (sc-7706), and anti-AQP5 (sc-28628) antibodies were purchased from Santa Cruz Biotechnology (Dallas, TX). 8-Hydroxy-2′-deoxyguanosine (8-Oxo-dG) antibodies (MAB 3560) were obtained from Millipore (Burlington, MA). Anti-phospho p53 (Ser15) antibody (cat. no. 9284), anti-phospho Cdc25C (Ser216) antibody (cat. no. 9528), anti-phospho Chk1 (Ser345) antibody (cat. no. 2341), anti-phospho ERK1/2 (Thr202/Tyr204) antibody (cat. no. 9101), and anti-p21 antibody (cat. no. 2947) were procured from Cell Signaling Technology (Danvers, MA). Anti-Tom20 antibody (ab-186734), anti-Ogg1 antibody (ab-135940), and anti-NOX4 antibody (ab-133303) were purchased from Abcam (Cambridge, MA).
Exposure of mice to hyperoxia and isoflurane.
C57BL/6J male mice (12–14 wk old) were purchased from Charles River Laboratories and used in this study. All procedures were approved by the Institutional Animal Care and Use Committee at the Texas Tech University Health Sciences Center, Lubbock, and University of Texas Health Science Center at Tyler and were consistent with the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health. Mice (n = 5 to 6 per group) were exposed to 21% O2, 60% O2, 21% O2 plus 2% isoflurane, or 60% O2 plus 2% isoflurane for 2 h (2 l/min). The lower and higher dosages of isoflurane (0.25%) were used to demonstrate the effect of non-anesthetic versus anesthetic dose of isoflurane on genotoxic effects of hyperoxia. An isoflurane vaporizer was used to mix oxygen and isoflurane. In some cases, mice were allowed to recover under normoxic conditions for up to 24 h after isoflurane exposure. When we exposed mice to isoflurane or isoflurane plus 60% oxygen, mouse body temperature (monitored by rectal probe) dropped to 29°C by 30 min. We provided heat supplementation by warming base by a heat pad [Homeothermic Blanket Systems with Flexible Probe 507222-F (Harvard Apparatus, Holliston, MA) for monitoring the temperature] and utilizing a lamp, and with this arrangement the temperature was maintained between 35.9°C and 36.5°C. At the end of these treatments, both anesthetized and non-anesthetized control groups of mice were euthanized by cervical dislocation followed by thoracotomy. The lungs were perfused with PBS via right ventricular puncture, dissected out, and processed immediately.
Isolation of pulmonary epithelial and endothelial cells.
Endothelial cells from mouse lungs were isolated as described by Sobczak et al. (52). In brief, 6–8-week-old mice were anesthetized with ketamine (100 mg/kg) and xylazine (10 mg/kg), and after opening of the thoracic cavity, lungs were perfused from the right ventricle, excised, and placed in DMEM containing 1% penicillin/streptomycin. Lung lobes were minced into small pieces and digested in DMEM containing 1 mg/ml collagenase II from Worthington for 45 min. Then the mixture was triturated by passing through a 16-guage cannula and filtered with a 70-μm strainer. After centrifugation, the pellet was dissolved in DMEM, and endothelial cells were selected by incubating with anti-platelet endothelial cell adhesion molecule antibodies (BD Biosciences)-bound sheep anti-rat Dynabeads (Invitrogen) for 30 min, washed, and trypsinized to detach cells from Dynabeads. Finally, the cells were suspended in EBM-2 medium and plated in gelatin-coated plates. Type II alveolar epithelial cells from mouse lungs were isolated as described in Ref. 16. Briefly, lungs were teased apart after immersing in 1.5 ml dispase for 45 min, filtered through 100- and 40-µM nylon filters, and seeded onto anti-CD45 and anti-CD32 coated 100-mm dishes. Supernatant containing type II alveolar cells was suspended in DMEM with 10% FBS and seeded onto six-well plates or in cover glass for microscopy.
PCR array analysis.
Mice were exposed to hyperoxia with or without 2% isoflurane, and after appropriate length of treatment or recovery, they were euthanized and the lungs were isolated. The lungs were perfused with nuclease-free PBS and preserved in RNALater (Ambion). RNA was isolated from the lung tissue using Trizol Reagent, and equal amounts of RNA were subjected to reverse transcription using an RT2 First Strand Kit (Qiagen). Gene expression analysis was performed by RT2 Profiler PCR Array (Mouse Oxidative Stress and Antioxidant Defense, PAMM-065-ZC, and Mouse DNA Damage Signaling Pathway, PAMM-029-ZC) following the manufacturer’s protocol.
Western blotting.
Protein extracts were prepared from appropriately treated MLE-12 cells or lungs using radioimmunoprecipitation assay buffer. Minced lung pieces were homogenized in cold radioimmunoprecipitation assay buffer in a glass dounce homogenizer. The homogenates were centrifuged at 15,000 g for 15 min at 4°C. Protein concentrations were determined with the BCA protein assay kit (Pierce Chemical, Rockford, IL). Protein extracts were analyzed for Western blotting using their specific antibodies as previously published (18, 39).
Immunofluorescence microscopy.
MLE-12 cells, primary type II cells, or primary endothelial cells were cultured on glass coverslips and exposed to hyperoxia with or without 2% isoflurane. The cells were then washed, fixed, permeabilized, and probed with rabbit anti-8-Oxo-dG and mouse anti-Tom 20 antibodies followed by Alexa Fluor 488- and Alexa Fluor 568-conjugated donkey anti-rabbit and anti-mouse secondary antibodies. Cells were counterstained with DAPI. A stack of fluorescent images was obtained along the z-axis at 60-nm intervals via a ×63/1.40 numerical aperture objective using a Zeiss Axio Imager Z2 upright fluorescent microscope and deconvolved using AxioVision 4.9 software. Lung tissue sections were deparaffinized, hydrated, permeabilized, blocked, and immunostained with anti-8-Oxo-dG and mouse anti-Tom 20 antibodies. A stack of fluorescent images was obtained along the z-axis at 100-nm intervals via a ×40/1.40 numerical aperture objective and was deconvolved.
Hematoxylin-eosin staining.
C57BL6 mice (12–14 wk) were exposed to 60% oxygen with or without 2% isoflurane to study the effect of short-duration hyperoxia on mouse lung injury. We also exposed mice to either 95% oxygen or 95% oxygen with 0.25% isoflurane for 36 h to study the effect of isoflurane on ALI in prolonged hyperoxia. After the exposure, mice were euthanized, and the lungs were flushed with warm PBS via the right ventricle. After blood was removed from the lung, the entire organ was inflated via instillation of 4% paraformaldehyde through the trachea for 12 h. The inflated lung was removed en bloc and immersed in this fixative. Fixed lung was paraffin mounted and sectioned using a microtome. Lungs sections were stained with hematoxylin and eosin. The stained sections were observed under a light microscope for assessment of lung injury indices (41). Lung injury score was implemented as suggested by an official American Thoracic Society Workshop Report for histological evidence of lung injury as published (41). In brief, the score criteria are as follows: A, neutrophils in alveolar space; B, neutrophils in interstitial space; C, hyaline membranes; E, alveolar septal thickening. Each of these indicators was scored as per Table-1 of the lung injury scoring system (41). The score was calculated as number of each alphabet/field and number of field × 100. The injury score is reported as a continuous value between 0 and 1 (41).
8-Oxo-dG ELISA.
Urine was collected from mice exposed to normoxia, normoxia plus isoflurane, hyperoxia, and hyperoxia plus isoflurane. For urine collection, individual mice were allowed to urinate on a clear polyethylene surface, and the urine was aspirated by a micropipette into microcentrifuge tubes. The level of 8-Oxo-dG in the urine of mice was determined using an HT 8-oxo-dG ELISA Kit (R & D Systems, cat. no. 4380-096-K) following supplier’s instructions.
TUNEL assay.
Type 2 epithelial cells and endothelial cells from mouse lung (12–14 wk) were isolated and cultured on glass coverslips and exposed to 21% O2 or 60% O2 with/without 2% isoflurane for 24 h. At the end of the exposure period, cells were fixed and permeabilized, and apoptotic type 2 and endothelial cells were labeled with TdT-mediated fluorescein-dUTP-X nick end labeling (TUNEL) kit (cat. no. 11684795910, In Situ Cell Death Detection Kit, Sigma). Cells were also counterstained with DAPI. Immunofluorescence images were obtained using Zeiss LSM880 fluorescent confocal microscope via ×20 objective. TUNEL-positive type 2 and endothelial nuclei were counted using Zeiss Zen 2.3 software. Deparaffinized and hydrated lung sections were subjected to TUNEL assay using In Situ Cell Death Detection Kit from Roche Applied Science (cat. no. 11684795910) following the supplier's protocol.
Flow cytometry and annexin V apoptosis assay.
MLE-12 cells or HMVEC-Ls were exposed to 21% or 60% O2 with or without 2% isoflurane for 2 h or 24 h, and then the cells were trypsinized using TrypLE reagent. The collected cells were washed and incubated with 1X Annexin V binding buffer containing Annexin V-FITC (cat. no. 556547, BD Bioscience) as per manufacturer instructions. After incubation, the cells were washed with 1X binding buffer and subjected to FACS analysis using BD FACS CALIBUR, and the results were analyzed using FlowJo software.
Detection of superoxide anion by electron paramagnetic resonance spectrometry.
Superoxide formation in MLE-12 cells was detected by electron paramagnetic resonance (EPR) spectroscopy using spin probe 1-hydroxy-3-methoxycarbonyl-2, 2,5,5-tetramethylpyrrolidine hydrochloride (CMH). After appropriate treatments, MLE-12 cells were resuspended in Krebs-Henseleit Buffer (118 mM NaCl, 4.7 mM KCl, 1.2 mM MgSO4, 1.25 mM CaCl2, 1.2 mM KH2PO4, 25 mM NaHCO3, 11 mM glucose, pH 7.4) and incubated with 0.1 mM CMH in the presence of 25 µM desferoxamine and 2.5 µM diethyldithiocarbamate at 37°C. At the end of incubation period, the reaction mix was loaded in an EPR capillary tube, and the generated superoxide was detected as paramagnetic nitroxide radical (CM·) using a Bruker EMX Nano spectrometer at room temperature. Mitochondrial superoxide formation in MLE-12 cells was detected by EPR spectroscopy using spin probe 1-hydroxy-4-[2-(triphenylphosphonio)-acetamido]-2,2,6,6-tetramethylpiperidine (mitoTEMPO-H). After appropriate treatments, MLE-12 cells were resuspended in Krebs-Henseleit Buffer and incubated with 50 µM mitoTEMPO-H at 37°C for 15 min. Superoxide generated by MLE-12 cells was detected as paramagnetic nitroxide radical using Bruker EMX Nano spectrometer at room temperature. EPR spectra were acquired under following scan conditions: microwave frequency, 9.63 GHz; power, 0.31 mW; attenuation 25 dB; modulation frequency, 100 kHz; modulation amplitude, 1.00 G; sweep time, 60 s; time constant, 1.28 s; receiver gain, 40 dB; magnetic field, 3400–3460 G.
Statistical analysis.
Data are expressed as the mean ± SEM. Multiple experimental groups are compared by analysis of variance with Tukey’s posttest comparison of means using GraphPad Prism software. The Student’s t-test was used when comparing means from two experimental groups. A value of P ≤ 0.05 was considered statistically significant. Unless otherwise stated, the figures represent a minimum of three mice (n = 3) that were used to determine mean values and other analyses.
RESULTS
Exposure of MLE-12 cells to 60% oxygen for 24 h causes mitochondrial and nuclear DNA damage.
We exposed MLE-12 cells to 60% oxygen for 2 h and measured the level of the oxidative DNA damage product 8-Oxo-dG (44) using immunofluorescence. However, we did not observe 8-Oxo-dG, as there was no fluorescence in these cells (Fig. 1A). Next, we increased the exposure to 24 h, and as shown in Fig. 1, B and C, this treatment caused significant mitochondrial DNA damage, as detected by co-localization of mitochondrial membrane protein Tom-20 and 8-Oxo-dG. Additionally, 8-Oxo-dG accumulations were also found in the nucleus, demonstrating nuclear DNA damage as well (Fig. 1B). In contrast, when oxygen and isoflurane were used in combination, the level of 8-Oxo-dG was significantly decreased (Fig. 1B). Interestingly, isoflurane in normoxia caused significant mitochondrial and nuclear DNA damage as visualized by accumulation of 8-Oxo-dG in the nucleus and mitochondria (Fig. 1B). However, isoflurane in combination with hyperoxia caused significant reduction in 8-Oxo-dG accumulations in both nucleus and mitochondria. Collectively, our data presented in Fig. 1 show that in cultured cells 60% O2 exposure for 2 h does not cause detectable mitochondrial or nuclear DNA damage, but exposure to higher duration (>16 h) of 60% O2 induces significant DNA damage. Furthermore, our data also show that isoflurane is able to abrogate DNA damage induced by prolonged exposure to hyperoxia, but isoflurane with normoxia causes significant nuclear and mitochondrial DNA damage.
Mice exposed to 60% oxygen for 2 h show significant mitochondrial 8-Oxo-dG accumulations in the lung, but not in the presence of isoflurane.
Our cell culture data show that mouse alveolar epithelial cells (MLE-12) are resistant to induction of DNA damage after 2 h of 60% O2 exposure. Therefore, we sought to determine whether in in vivo the lung would sustain any DNA damage due to 2-h exposure to 60% O2, as most lung injury studies using hyperoxia use >60% oxygen exposure for 48–96 h (16, 17, 46). We determined whether short-term 60% O2 exposure damages lung DNA in vivo by analyzing the levels of 8-Oxo-dG in the lung following 2 h of exposure. As shown in Fig. 2A, high levels of 8-Oxo-dG were observed in the lungs of mice exposed to 60% O2 for 2 h (Fig. 2A, top left, and Fig. 2B, left), indicating significant mitochondrial DNA damage in the lungs of these animals. We also exposed mice to 2 h of 60% oxygen and recovered them for 24 h in RA. As shown in Fig. 2C, the level of 8-Oxo-dG was decreased in 60%-oxygen-exposed animals after 24-h recovery in RA. In contrast, mice breathing 60% oxygen along with 2% isoflurane had significantly lower levels of 8-Oxo-dG (Fig. 2, B and C), indicating that this anesthetic protects against pulmonary DNA damage caused by short-duration hyperoxic exposure.
We next examined whether 8-Oxo-dG detected in the lungs in hyperoxia appears in the urine of mice exposed to high levels of oxygen, as 8-Oxo-dG is known to be excreted in the urine in response to DNA damage by other agents (12, 25, 31). As shown in Fig. 2D, urinary levels of 8-Oxo-dG (as determined by ELISA) were significantly increased 4 h posttreatment. When isoflurane was used in conjunction with oxygen, the level of 8-Oxo-dG was decreased to baseline levels, indicating significant protection by the anesthetic. Additionally, after 24 h of recovery in normoxia, the level of 8-Oxo-dG was decreased in animals exposed to 60% oxygen. However, there was no change in the levels of 8-Oxo-dG in animals exposed to hyperoxia plus isoflurane.
Isoflurane prevents apoptosis of pulmonary cells exposed to hyperoxia.
One of the consequences of DNA damage is the induction of DNA repair. If the damage is extensive, then the process of energy-dependent apoptosis ensures that the damaged cells are eliminated to prevent deleterious mutations. Accordingly, we determined whether short-duration hyperoxia-induced DNA damage signal culminates in the apoptosis of lung cells in vivo. As shown in Fig. 3, A and B, significant pulmonary apoptosis occurred in mice lung exposed to 60% O2 for 2 h. However, apoptosis was significantly decreased when isoflurane was co-delivered along with the high concentrations of oxygen. Next, we allowed mice to recover for 24 h in RA following 2-h exposure to 60% oxygen alone or oxygen plus isoflurane. As shown in Fig. 3, A and B, the oxygen only group showed a high percentage of cells with TUNEL-positive staining after 24 h of recovery. However, the isoflurane plus oxygen group demonstrated a significantly lower percentage of TUNEL-positive staining that was not significantly different than the lungs of mice in normoxia. We further determined specific cell types of lung that are associated with enhanced apoptosis due to exposure to 24-h hyperoxia. Isolated type II cells from lungs of mice exposed to 60% O2 showed TUNEL-positive nuclei demonstrating significant apoptosis, but when isoflurane was mixed with hyperoxia, the level of apoptosis was significantly decreased (Fig. 3, C and D). Furthermore, isoflurane in RA also caused significant apoptosis, which is in agreement with our MLE-12 cells exposed to 16 h with isoflurane in RA (Fig. 1B). In contrast to type II cells, the endothelial cells from lung did not show significant apoptosis in response to hyperoxia. Additionally, these cells also did not show enhanced apoptosis when exposed to isoflurane in normoxia (Fig. 3, E and F). Next, we determined the effect of hyperoxia and isoflurane on lung type II and I cells in situ in lung sections. As shown in Fig. 3, G and H, both type I and type II cells underwent apoptosis in hyperoxia and also in normoxia with isoflurane, but the magnitude of apoptosis was higher in type I cells compared with type II cells. Isoflurane in combination with 60% O2 significantly decreased apoptosis caused by hyperoxia alone in both cell types (Fig. 3, G and H). We also performed annexin V labeling and performed apoptosis assay of MLE-12 and HMVEC-L using flow cytometry. As shown in Fig. 4A, isoflurane in normoxia caused significant apoptosis of MLE-12 cells similar to that obtained with 60% O2 hyperoxia. Isoflurane significantly decreased apoptosis of MLE-12 cells in hyperoxia. We also evaluated the apoptosis of endothelial cells using this method, and as shown in Fig. 4B, endothelial cells did not show significant apoptosis. Isoflurane alone in normoxia also failed to show significant apoptosis in contrast to MLE-12 cells.
Isoflurane induces O2·− production with ambient air but inhibits O2·− generation when mixed with hyperoxia.
Several studies indicate that isoflurane causes ROS generation (10, 24, 48); however, other studies demonstrate that isoflurane provides protection against sepsis or LPS-dependent cytotoxicity. Thus, the role of isoflurane as an inducer or inhibitor of ROS such as O2·− has not yet been established. We determined the effect of isoflurane on O2·− generation in the presence of air or hyperoxia using EPR spectrometry. As shown in Fig. 5, A and B, isoflurane mixed with air (or 21% oxygen) produced significant levels of O2·− in MLE-12 cells. In contrast, when it was mixed with 60% oxygen, the level of O2·− was significantly decreased (Fig. 5, A and B), suggesting that isoflurane can be an O2·− generating agent when mixed with air but scavenges hyperoxia-dependent O2·− production. Furthermore, our data in Fig. 5, A and B show that diphenyliodonium (DPI), a nonselective NADPH oxidase (Nox) inhibitor significantly inhibited O2·− generation by hyperoxia; however, this inhibition was not complete, suggesting that mitochondria could account for DPI-resistant O2·− generation in hyperoxia. However, when isoflurane was mixed with 60% oxygen in the presence of DPI, complete inhibition of O2·− generation was achieved (Fig. 5, A and B). Next, we determined the effect of DPI on isoflurane-mediated O2·− release in normoxia. As shown in Fig. 5, A and B, DPI almost completely inhibited the CMH signal, indicating NADPH oxidase is a major source of O2·− by isoflurane. Taken together, these experiments show that Nox is a major source of O2·− in normoxia by isoflurane.
Isoflurane decreases mitochondrial O2·− production in hyperoxia but does not increase mitochondrial O2·− generation in normoxia.
We further determined whether mitochondria are a significant source of O2·− for 2-h 60% O2 exposure and whether isoflurane would decrease this O2·−. As shown in Fig. 5, C and D, mitoTEMPO spectra (a mitochondrial superoxide anion spin probe) were observed in 60% O2 exposure in MLE-12 cells (Fig. 5, C and D). This spectrum was decreased to a nonsignificant extent by DPI, indicating that Nox is not the source of O2·− in hyperoxia as expected. However, isoflurane significantly decreased the mitoTEMPO spectra, indicating significant decrease in mitochondrial O2·− in the presence of isoflurane in hyperoxia (Fig. 5, C and D). There was no increase in mitoTEMPO signal in normoxia with isoflurane, indicating that isoflurane predominantly generates O2·− from Nox in normoxia.
Isoflurane diminishes DNA damage signaling caused by short-term hyperoxia.
We previously established that prolonged hyperoxia activates checkpoint kinase 1 and the tumor-suppressor protein p53 as a consequence of DNA damage (17, 38). Here, we determined whether short duration of hyperoxia would also initiate gene expression associated with DNA damage signaling in the lungs of mice. As shown in Fig. 6A, several DNA damage response genes associated with cell cycle arrest and the DNA damage response were significantly increased in response to short-duration hyperoxia exposure in mice lungs. These genes include RAD9, a homolog of (S. pombe) Rad9A, Ligase I (LigA), RAD51 homolog (S. cerevisiae) (Rad51), Fanconi anemia complementation group G (Fancg), X-ray repair complementing defective repair in Chinese hamster cells 2 (Xrcc2), Replication protein A1 (Apa1), Fanconi anemia complementation group D2 (Fancd2), Hus1 homolog (S. pombe) (Hus1), MutL homolog 3 (E. coli) (Mlh3), Cell division cycle 25 homolog A (S. pombe) (Cdc25A), RAD1 homolog (S. pombe) (Rad1), Fanconi anemia complementation group A (Fanca), Ataxia telangiectasia and rad3 related (ATR), and Checkpoint kinase 1 homolog (S. pombe) (Chek1). Expression of all of these genes remained at basal level when isoflurane was used in combination with hyperoxia, indicating that isoflurane alleviates the activation of DNA damage signaling by protecting DNA damage in the lung due to short-duration hyperoxia.
As shown in Fig. 6, B and C, the expression of the p53 protein, as well as the occurrence of activating phosphorylation on Ser15, was dramatically increased during short exposures to 60% O2, which points to a DNA damage and repair response (18). We also found that the expression of p21, a target gene of the p53 transcription factor, is increased (Fig. 6B) as a consequence of 60% O2 exposure, indicating cell cycle arrest and also transcriptional activity of p53 (37). Furthermore, other G2/M (growth phase 2/metaphase) proteins, such as Cdc25C and cyclin B1 levels (Fig. 6, B and C), were increased in animals experiencing short-term hyperoxia. However, inclusion of 2% isoflurane with high oxygen treatment prevented the increases in all of these DNA damage response proteins, confirming the protective effect of isoflurane against DNA damage that occurs in response to hyperoxia.
Short-duration hyperoxia provokes the expression of Nox components, p47phox, and p22phox but not when isoflurane is co-administered.
We next sought to determine the activation of oxidative stress response genes in response to short-duration hyperoxia and the effect of isoflurane on the activation of these genes. We utilized a PCR microarray analysis of oxidative stress response genes following 2-h exposures to oxygen or oxygen plus isoflurane. As shown in Fig. 7A, the expression of p22phox (a component of NADPH oxidase) was increased ~14-fold, and p47phox (another component of NADPH oxidase) was increased more than 25-fold in response to 60% O2. Thus, the expression of either of these genes may account for increased superoxide anion (O2·−) generation in response to high levels of oxygen. Interestingly, isoflurane markedly decreased the expression of all of these genes activated in hyperoxia, any of which could significantly contribute to the lung damage via generation of high levels of O2·−. Collectively, the results of this experiment indicate that p47phox and p22phox could be sources of O2·− production that occur in response to short duration of hyperoxia. Our findings also indicate that isoflurane has a significant impact in diminishing the expression of these genes. Since p22phox and p47phox are components of Nox proteins, we determined the expression of Nox4 in the lung tissue of mice exposed to normoxia or hyperoxia in the presence or absence of isoflurane. Our data presented in Fig. 7, B and C show that isoflurane in combination with hyperoxia caused significant increase in Nox4 expression, suggesting isoflurane may induce the production of H2O2 via Nox4 in hyperoxia, as activated Nox4 is known to produce H2O2 (27). However, Nox4 expression was increased in isoflurane plus 60% oxygen exposed animals. We also tested two other isoforms of Nox mRNA by RT-PCR, such as Duox1 and Duox2, and hyperoxia-induced Duox2 mRNA, but in the presence of isoflurane, this increase was diminished (Fig. 7, D and E).
Mice breathing isoflurane in combination with 60% O2 show increased expression of pulmonary BER genes.
We analyzed the expression levels of DNA damage response genes during hyperoxia and changes in them when the treatment included isoflurane. When we measured the fold increase of these genes, we found that several of them are upregulated in response to the inclusion of isoflurane in a 2-h exposure to 60% O2 (Fig. 8A) Furthermore, we found that most of these DNA repair genes fell into the BER pathway (44). For example, Gadd45α, Fen1, and XPC are associated with the BER pathway (19, 21, 51). In addition, Ppp1r15α is also known to participate in DNA repair pathways (44). Taken together, our microarray study shows that isoflurane may induce DNA repair pathways, in particular those involved in BER. We determined the expression of Ogg1 and APE1 in lungs of mice following 2 h of exposure to 60% O2 in the presence or absence of isoflurane. As shown in Fig. 8, B and C, the expression of Ogg1 and APE1 was decreased in lungs exposed to 60% oxygen. When isoflurane was mixed with the oxygen, the expression of Ogg1 was maintained, demonstrating that this major BER enzyme is rescued by isoflurane. Collectively, our data show that isoflurane may be involved in improving DNA repair, specifically via BER, when used in combination with hyperoxia.
Isoflurane inhibits the phosphorylation of ERK.
Elevated concentrations of oxygen are known to cause ERK activation (7, 8, 56). Because ROS are known to cause ERK phosphorylation (i.e., activation), we next determined the effect of isoflurane on ERK phosphorylation. As shown in Fig. 9, A and B, we found ERK was activated when mice were exposed to high oxygen for short periods. However, isoflurane, inhibited hyperoxia-induced ERK phosphorylation but increased the expression of ERK protein.
Isoflurane inhibits lung injury in mice exposed to hyperoxia.
Prolonged breathing of hyperoxia is known to cause lung damage in mice and humans. Therefore, we tested whether the protective role of isoflurane in lung injury could extend to ALI induced by longer duration and higher concentration of oxygen. As shown in Fig. 10, A and B, markers associated with lung injury were significantly higher in mice exposed to 36-h hyperoxia (41). However, in the presence of isoflurane, the expression of these markers was significantly lower (Fig. 10, A and B). These data suggest that isoflurane could also provide significant protection against deleterious effects of prolonged oxygen exposure in the lung.
DISCUSSION
In this report, we present evidence that mice exposed to 60% oxygen (relevant as a carrier gas in anesthesia) for 2 h experience mitochondrial and nuclear DNA damage, and when isoflurane is used in conjunction with hyperoxia, the DNA damage is abrogated. Our findings suggest that cell culture studies are not responsive to short-duration of hyperoxia-mediated DNA damage, in contrast to in vivo studies with exposure of mice to 2-h 60% oxygen that causes significant DNA damage. Furthermore, these studies show, for the first time, that significant DNA damage occurs in the lungs in response to short-duration hyperoxia. Additionally, we have shown that isoflurane in normoxia generates O2·− via activation of NADPH oxidases; however, isoflurane does not produce O2·− via a mitochondrial pathway. Interestingly, isoflurane inhibits hyperoxia-mediated O2·− production. We also demonstrated that isoflurane inhibits the expression of p47phox and p22phox (components of NADPH oxidase), which are otherwise induced in mice inhaling 60% oxygen for 2 h. Furthermore, the expression of Ogg1, Gadd34, and Gadd45 (all of which are involved in DNA repair) are increased in the presence of isoflurane plus oxygen, but not in only oxygen, demonstrating that the anesthetic induces DNA repair mechanisms. Furthermore, using a prolonged ALI model, we show that isoflurane significantly protects against ALI. Collectively, these novel data point to a previously unrecognized role for short-duration hyperoxia as a genotoxic stress and the beneficial role of isoflurane in protecting against the DNA damage and ALI caused by hyperoxia.
Several studies indicate that isoflurane causes ROS generation (48), and other studies demonstrate that lung injury caused by LPS or sepsis is prevented by isoflurane. Thus, a clear mechanism of action for this anesthetic has not yet been established. We believe that these contradictory findings are due to the way isoflurane was administered to mice. There are instances in which isoflurane was co-delivered with high oxygen concentrations and others in which it was applied with ambient air. For example, in several animal investigations in which isoflurane was used as a preconditioning agent, we noted that isoflurane was used with ambient air (33, 40, 48, 53). These findings demonstrate that isoflurane generates low levels of O2·−, which is crucial for its function as a preconditioning agent. On the other hand, when isoflurane was applied in combination with high concentrations of oxygen, it protected against lung injury and inflammation induced by either LPS or TNFα. Our current finding provides a compelling explanation of this property of isoflurane. Isoflurane in combination with normoxia produces O2·− by activating NADPH oxidase but inhibits O2·− generation when it is delivered with hyperoxia.
Tissue hypoxia during anesthesia administration is the primary reason that high concentrations of oxygen are routinely used perioperatively (43). However, prolonged breathing of oxygen at high concentrations is known to cause lung damage in animal experiments. By contrast, the effect of short-duration hyperoxia on lung biology has not been previously investigated. Our data further establish that short-term exposure to 60% oxygen causes significant DNA damage in mitochondria and the nucleus, to the point that damage products appear in the urine of mice. This establishes that short-duration hyperoxia is a substantial genotoxic stress in the lung. Surprisingly, when we delivered isoflurane in a hyperoxic gas mixture, we did not detect markers of DNA damage either in the lung or in cultured cells. Consequently, this establishes a strong protective role for this anesthetic during periods of hyperoxia.
Our findings also indicate that hyperoxia induces significant release of 8-Oxo-dG in the urine of mice after 4 h of exposure, indicating oxidative DNA damage and BER. 8-Oxo-dG is primarily repaired by Ogg1, which recognizes the lesion and excises it as free 8-Oxo-dG (13). Accumulation of 8-Oxo-dG is linked to various pathologies, including cancer and aging. Furthermore, free 8-Oxo-dG could pair with adenosine, causing a GC→TA mutation that is frequently encountered as a deleterious consequence of oxidized DNA (47). It has also been shown that accumulated 8-Oxo-dG interacts with Ogg1 and activates Ras and Rac1 GTPases, resulting in an intracellular signaling cascade. Addition of 8-Oxo-dG to cells has been shown to cause oxidative stress (29). Therefore, we speculated that 8-Oxo-dG accumulation in response to hyperoxia generates oxidative stress via expression and activation of p22phox and p47phox. Indeed, our data establish that p22phox and p47phox mRNA levels are significantly decreased when isoflurane is administered in combination with high oxygen tension. These results demonstrate that isoflurane inhibits 8-Oxo-dG-dependent signaling, leading to decreased transcription of these NADPH oxidase components. Thus, since hyperoxia is known to induce NADPH oxidase, it is likely that hyperoxia-stimulated O2·− generation in the mitochondria activates p22phox and p47phox (30), resulting in further increases in O2·− levels by NADPH oxidase. It is also already known that 8-Oxo-dG can directly activate NADPH oxidase when it is artificially introduced into the lungs of mice (29). This suggests that 8-Oxo-dG accumulation caused by short-duration high oxygen exposures would comprise a major mechanism of increased oxidative stress in the lung. Interestingly, we detected a significant level of 8-Oxo-dG in the urine of mice, which suggests that this marker could potentially be used to monitor the extent of DNA damage in humans exposed to high oxygen concentrations for short durations.
We observed a significant increase in DNA damage response genes and proteins due to short-duration of 60% oxygen exposure in mice lung but not when isoflurane was included with oxygen. These data suggest, for the first time, that short-duration hyperoxia causes significant DNA damage. By extrapolation, these exposures could potentially be deleterious to human lungs and be ameliorated by supplemental isoflurane. Since these genes are involved in DNA repair, cell cycle arrest or induction of apoptosis administration of hyperoxia without isoflurane could induce significant mutagenesis of lung DNA with potential for development of cancer or other diseases associated with genotoxic stress. Exposure of cells to isoflurane combined with ambient air caused increase in p53 (and pSer15), Cdc25C, and cyclin B1 expression, demonstrating that the anesthetic alone could cause activation of DNA damage response proteins when administered with air. These data are in agreement with our current finding that isoflurane in ambient air generates O2·− via NADPH oxidase and with other studies demonstrating increased ROS generation by isoflurane when administered to mice or cells with ambient air (10, 33, 40).
Thus, the administration of isoflurane in conjunction with oxygen could potentially alter clinical outcomes, in which the anesthetic could protect against lung injury. Hyperoxia provides a significant source of O2·−, as mitochondria generate high levels of O2·− in high concentrations of oxygen (26). Isoflurane could potentially act as a scavenger of O2·− in a hyperoxic condition. Collectively, our findings suggest that isoflurane inhibits DNA damage signaling in response to short periods of hyperoxia and could thereby protect the lung against arrested cell growth or apoptosis and consequently lung injury in ALI.
GRANTS
The work presented here is supported by National Heart, Lung, and Blood Institute Grant HL 1 R01 130061 to K. Das.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
K.D. conceived and designed research; V.K.-S., J.S., S.R., G.P.M., C.O., T.W., and K.D. performed experiments; V.K.-S., J.S., C.O., and K.D. analyzed data; V.K.-S., J.S., and K.D. interpreted results of experiments; V.K.-S., J.S., and K.D. prepared figures; V.K.-S. and K.D. drafted manuscript; J.W., S.I., and K.D. edited and revised manuscript; V.K.-S. and K.D. approved final version of manuscript.
REFERENCES
- 1.Abman SH, Warady BA, Lum GM, Koops BL. Systemic hypertension in infants with bronchopulmonary dysplasia. J Pediatr 104: 928–931, 1984. doi: 10.1016/S0022-3476(84)80501-6. [DOI] [PubMed] [Google Scholar]
- 2.Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15: 2177–2196, 2001. doi: 10.1101/gad.914401. [DOI] [PubMed] [Google Scholar]
- 3.Anderson SL, Duke-Novakovski T, Singh B. The immune response to anesthesia: part 1. Vet Anaesth Analg 41: 113–126, 2014. doi: 10.1111/vaa.12125. [DOI] [PubMed] [Google Scholar]
- 4.Bakar AM, Park SW, Kim M, Lee HT. Isoflurane protects against human endothelial cell apoptosis by inducing sphingosine kinase-1 via ERK MAPK. Int J Mol Sci 13: 977–993, 2012. doi: 10.3390/ijms13010977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Balasubramaniam V, Mervis CF, Maxey AM, Markham NE, Abman SH. Hyperoxia reduces bone marrow, circulating, and lung endothelial progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 292: L1073–L1084, 2007. doi: 10.1152/ajplung.00347.2006. [DOI] [PubMed] [Google Scholar]
- 6.Buckley S, Barsky L, Driscoll B, Weinberg K, Anderson KD, Warburton D. Apoptosis and DNA damage in type 2 alveolar epithelial cells cultured from hyperoxic rats. Am J Physiol Lung Cell Mol Physiol 274: L714–L720, 1998. doi: 10.1152/ajplung.1998.274.5.L714. [DOI] [PubMed] [Google Scholar]
- 7.Buckley S, Driscoll B, Barsky L, Weinberg K, Anderson K, Warburton D. ERK activation protects against DNA damage and apoptosis in hyperoxic rat AEC2. Am J Physiol Lung Cell Mol Physiol 277: L159–L166, 1999. doi: 10.1152/ajplung.1999.277.1.L159. [DOI] [PubMed] [Google Scholar]
- 8.Carnesecchi S, Deffert C, Pagano A, Garrido-Urbani S, Métrailler-Ruchonnet I, Schäppi M, Donati Y, Matthay MA, Krause KH, Barazzone Argiroffo C. NADPH oxidase-1 plays a crucial role in hyperoxia-induced acute lung injury in mice. Am J Respir Crit Care Med 180: 972–981, 2009. doi: 10.1164/rccm.200902-0296OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Christofidou-Solomidou M, Muzykantov VR. Antioxidant strategies in respiratory medicine. Treat Respir Med 5: 47–78, 2006. doi: 10.2165/00151829-200605010-00004. [DOI] [PubMed] [Google Scholar]
- 10.Chung IS, Kim JA, Kim JA, Choi HS, Lee JJ, Yang M, Ahn HJ, Lee SM. Reactive oxygen species by isoflurane mediates inhibition of nuclear factor κB activation in lipopolysaccharide-induced acute inflammation of the lung. Anesth Analg 116: 327–335, 2013. doi: 10.1213/ANE.0b013e31827aec06. [DOI] [PubMed] [Google Scholar]
- 11.Coalson JJ, Kuehl TJ, Escobedo MB, Hilliard JL, Smith F, Meredith K, Null DM Jr, Walsh W, Johnson D, Robotham JL. A baboon model of bronchopulmonary dysplasia. II. Pathologic features. Exp Mol Pathol 37: 335–350, 1982. doi: 10.1016/0014-4800(82)90046-6. [DOI] [PubMed] [Google Scholar]
- 12.Cooke MS, Henderson PT, Evans MD. Sources of extracellular, oxidatively-modified DNA lesions: implications for their measurement in urine. J Clin Biochem Nutr 45: 255–270, 2009. doi: 10.3164/jcbn.SR09-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dantzer F, Luna L, Bjørås M, Seeberg E. Human OGG1 undergoes serine phosphorylation and associates with the nuclear matrix and mitotic chromatin in vivo. Nucleic Acids Res 30: 2349–2357, 2002. doi: 10.1093/nar/30.11.2349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Das KC. Hyperoxia decreases glycolytic capacity, glycolytic reserve and oxidative phosphorylation in MLE-12 cells and inhibits complex I and II function, but not complex IV in isolated mouse lung mitochondria. PLoS One 8: e73358, 2013. doi: 10.1371/journal.pone.0073358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Das KC. Thioredoxin system in premature and newborn biology. Antioxid Redox Signal 6: 177–184, 2004. doi: 10.1089/152308604771978480. [DOI] [PubMed] [Google Scholar]
- 16.Das KC. Thioredoxin-deficient mice, a novel phenotype sensitive to ambient air and hypersensitive to hyperoxia-induced lung injury. Am J Physiol Lung Cell Mol Physiol 308: L429–L442, 2015. doi: 10.1152/ajplung.00285.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Das KC, Dashnamoorthy R. Hyperoxia activates the ATR-Chk1 pathway and phosphorylates p53 at multiple sites. Am J Physiol Lung Cell Mol Physiol 286: L87–L97, 2004. doi: 10.1152/ajplung.00203.2002. [DOI] [PubMed] [Google Scholar]
- 18.Das KC, Wasnick JD. Biphasic response of checkpoint control proteins in hyperoxia: exposure to lower levels of oxygen induces genome maintenance genes in experimental baboon BPD. Mol Cell Biochem 395: 187–198, 2014. doi: 10.1007/s11010-014-2124-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.David SS, O’Shea VL, Kundu S. Base-excision repair of oxidative DNA damage. Nature 447: 941–950, 2007. doi: 10.1038/nature05978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Day BJ. Antioxidants as potential therapeutics for lung fibrosis. Antioxid Redox Signal 10: 355–370, 2008. doi: 10.1089/ars.2007.1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.de Melo JT, de Souza Timoteo AR, Lajus TB, Brandão JA, de Souza-Pinto NC, Menck CF, Campalans A, Radicella JP, Vessoni AT, Muotri AR, Agnez-Lima LF. XPC deficiency is related to APE1 and OGG1 expression and function. Mutat Res 784-785: 25–33, 2016. doi: 10.1016/j.mrfmmm.2016.01.004. [DOI] [PubMed] [Google Scholar]
- 22.Dias-Freitas F, Metelo-Coimbra C, Roncon-Albuquerque R Jr. Molecular mechanisms underlying hyperoxia acute lung injury. Respir Med 119: 23–28, 2016. doi: 10.1016/j.rmed.2016.08.010. [DOI] [PubMed] [Google Scholar]
- 23.Dos Santos CC. Hyperoxic acute lung injury and ventilator-induced/associated lung injury: new insights into intracellular signaling pathways. Crit Care 11: 126, 2007. doi: 10.1186/cc5733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Englert JA, Macias AA, Amador-Munoz D, Pinilla Vera M, Isabelle C, Guan J, Magaoay B, Suarez Velandia M, Coronata A, Lee A, Fredenburgh LE, Culley DJ, Crosby G, Baron RM. Isoflurane ameliorates acute lung injury by preserving epithelial tight junction integrity. Anesthesiology 123: 377–388, 2015. doi: 10.1097/ALN.0000000000000742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Evans MD, Olinski R, Loft S, Cooke MS; European Standards Committee on Urinary (DNA) Lesion Analysis . Toward consensus in the analysis of urinary 8-oxo-7,8-dihydro-2′-deoxyguanosine as a noninvasive biomarker of oxidative stress. FASEB J 24: 1249–1260, 2010. doi: 10.1096/fj.09-147124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Freeman BA, Crapo JD. Hyperoxia increases oxygen radical production in rat lungs and lung mitochondria. J Biol Chem 256: 10986–10992, 1981. [PubMed] [Google Scholar]
- 27.Gordillo G, Fang H, Park H, Roy S. Nox-4-dependent nuclear H2O2 drives DNA oxidation resulting in 8-OHdG as urinary biomarker and hemangioendothelioma formation. Antioxid Redox Signal 12: 933–943, 2010. doi: 10.1089/ars.2009.2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Habre W, Peták F. Perioperative use of oxygen: variabilities across age. Br J Anaesth 113, Suppl 2: ii26–ii36, 2014. doi: 10.1093/bja/aeu380. [DOI] [PubMed] [Google Scholar]
- 29.Hajas G, Bacsi A, Aguilera-Aguirre L, Hegde ML, Tapas KH, Sur S, Radak Z, Ba X, Boldogh I. 8-Oxoguanine DNA glycosylase-1 links DNA repair to cellular signaling via the activation of the small GTPase Rac1. Free Radic Biol Med 61: 384–394, 2013. doi: 10.1016/j.freeradbiomed.2013.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.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 311: L337–L351, 2016. doi: 10.1152/ajplung.00447.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Harman SM, Liang L, Tsitouras PD, Gucciardo F, Heward CB, Reaven PD, Ping W, Ahmed A, Cutler RG. Urinary excretion of three nucleic acid oxidation adducts and isoprostane F(2)alpha measured by liquid chromatography-mass spectrometry in smokers, ex-smokers, and nonsmokers. Free Radic Biol Med 35: 1301–1309, 2003. doi: 10.1016/j.freeradbiomed.2003.07.003. [DOI] [PubMed] [Google Scholar]
- 32.Hatakeyama N, Matsuda N. Alert cell strategy: mechanisms of inflammatory response and organ protection. Curr Pharm Des 20: 5766–5778, 2014. doi: 10.2174/138161282036140912122809. [DOI] [PubMed] [Google Scholar]
- 33.Hirata N, Shim YH, Pravdic D, Lohr NL, Pratt PF Jr, Weihrauch D, Kersten JR, Warltier DC, Bosnjak ZJ, Bienengraeber M. Isoflurane differentially modulates mitochondrial reactive oxygen species production via forward versus reverse electron transport flow: implications for preconditioning. Anesthesiology 115: 531–540, 2011. doi: 10.1097/ALN.0b013e31822a2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Huang Y, Wang XX, Sun DD, Zhang ZX, Yang WW, Shao T, Han H, Zhang EF, Pu ZS, Hou ZX, Dong HL, Xiong LZ, Hou LC. Sub-anesthesia dose of isoflurane in 60% oxygen reduces inflammatory responses in experimental sepsis models. Chin Med J (Engl) 130: 840–853, 2017. doi: 10.4103/0366-6999.202734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ingbar DH. Mechanisms of repair and remodeling following acute lung injury. Clin Chest Med 21: 589–616, 2000. doi: 10.1016/S0272-5231(05)70168-4. [DOI] [PubMed] [Google Scholar]
- 36.Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respir Care 58: 123–141, 2013. doi: 10.4187/respcare.01963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kastan MB, Lim DS. The many substrates and functions of ATM. Nat Rev Mol Cell Biol 1: 179–186, 2000. doi: 10.1038/35043058. [DOI] [PubMed] [Google Scholar]
- 38.Kulkarni A, Das KC. Differential roles of ATR and ATM in p53, Chk1, and histone H2AX phosphorylation in response to hyperoxia: ATR-dependent ATM activation. Am J Physiol Lung Cell Mol Physiol 294: L998–L1006, 2008. doi: 10.1152/ajplung.00004.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kundumani-Sridharan V, Subramani J, Das KC. Thioredoxin activates MKK4-NFκB pathway in a redox-dependent manner to control manganese superoxide dismutase gene expression in endothelial cells. J Biol Chem 290: 17505–17519, 2015. doi: 10.1074/jbc.M115.660365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ludwig LM, Tanaka K, Eells JT, Weihrauch D, Pagel PS, Kersten JR, Warltier DC. Preconditioning by isoflurane is mediated by reactive oxygen species generated from mitochondrial electron transport chain complex III. Anesth Analg 99: 1308–1315, 2004. doi: 10.1213/01.ANE.0000134804.09484.5D. [DOI] [PubMed] [Google Scholar]
- 41.Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, Kuebler WM; Acute Lung Injury in Animals Study Group . An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol 44: 725–738, 2011. doi: 10.1165/rcmb.2009-0210ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Matute-Bello G, Frevert CW, Martin TR. Animal models of acute lung injury. Am J Physiol Lung Cell Mol Physiol 295: L379–L399, 2008. doi: 10.1152/ajplung.00010.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Meyhoff CS, Staehr AK, Rasmussen LS. Rational use of oxygen in medical disease and anesthesia. Curr Opin Anaesthesiol 25: 363–370, 2012. doi: 10.1097/ACO.0b013e328352b402. [DOI] [PubMed] [Google Scholar]
- 44.Mitra S, Izumi T, Boldogh I, Bhakat KK, Hill JW, Hazra TK. Choreography of oxidative damage repair in mammalian genomes. Free Radic Biol Med 33: 15–28, 2002. doi: 10.1016/S0891-5849(02)00819-5. [DOI] [PubMed] [Google Scholar]
- 45.O’Reilly MA. DNA damage and cell cycle checkpoints in hyperoxic lung injury: braking to facilitate repair. Am J Physiol Lung Cell Mol Physiol 281: L291–L305, 2001. doi: 10.1152/ajplung.2001.281.2.L291. [DOI] [PubMed] [Google Scholar]
- 46.O’Reilly MA, Staversky RJ, Finkelstein JN, Keng PC. Activation of the G2 cell cycle checkpoint enhances survival of epithelial cells exposed to hyperoxia. Am J Physiol Lung Cell Mol Physiol 284: L368–L375, 2003. doi: 10.1152/ajplung.00299.2002. [DOI] [PubMed] [Google Scholar]
- 47.Radak Z, Boldogh I. 8-Oxo-7,8-dihydroguanine: links to gene expression, aging, and defense against oxidative stress. Free Radic Biol Med 49: 587–596, 2010. doi: 10.1016/j.freeradbiomed.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Riess ML, Kevin LG, McCormick J, Jiang MT, Rhodes SS, Stowe DF. Anesthetic preconditioning: the role of free radicals in sevoflurane-induced attenuation of mitochondrial electron transport in Guinea pig isolated hearts. Anesth Analg 100: 46–53, 2005. doi: 10.1213/01.ANE.0000139346.76784.72. [DOI] [PubMed] [Google Scholar]
- 49.Roper JM, Mazzatti DJ, Watkins RH, Maniscalco WM, Keng PC, O’Reilly MA. In vivo exposure to hyperoxia induces DNA damage in a population of alveolar type II epithelial cells. Am J Physiol Lung Cell Mol Physiol 286: L1045–L1054, 2004. doi: 10.1152/ajplung.00376.2003. [DOI] [PubMed] [Google Scholar]
- 50.Rosenberg AA, Murdaugh E, White CW. The role of oxygen free radicals in postasphyxia cerebral hypoperfusion in newborn lambs. Pediatr Res 26: 215–219, 1989. doi: 10.1203/00006450-198909000-00012. [DOI] [PubMed] [Google Scholar]
- 51.Shafirovich V, Kropachev K, Anderson T, Liu Z, Kolbanovskiy M, Martin BD, Sugden K, Shim Y, Chen X, Min JH, Geacintov NE. Base and nucleotide excision repair of oxidatively generated guanine lesions in DNA. J Biol Chem 291: 5309–5319, 2016. doi: 10.1074/jbc.M115.693218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sobczak M, Dargatz J, Chrzanowska-Wodnicka M. Isolation and culture of pulmonary endothelial cells from neonatal mice. J Vis Exp 46: 2316, 2010. doi: 10.3791/2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tanaka K, Weihrauch D, Ludwig LM, Kersten JR, Pagel PS, Warltier DC. Mitochondrial adenosine triphosphate-regulated potassium channel opening acts as a trigger for isoflurane-induced preconditioning by generating reactive oxygen species. Anesthesiology 98: 935–943, 2003. doi: 10.1097/00000542-200304000-00021. [DOI] [PubMed] [Google Scholar]
- 54.Tang PS, Mura M, Seth R, Liu M. Acute lung injury and cell death: how many ways can cells die? Am J Physiol Lung Cell Mol Physiol 294: L632–L641, 2008. doi: 10.1152/ajplung.00262.2007. [DOI] [PubMed] [Google Scholar]
- 55.Tateda K, Deng JC, Moore TA, Newstead MW, Paine R III, Kobayashi N, Yamaguchi K, Standiford TJ. Hyperoxia mediates acute lung injury and increased lethality in murine Legionella pneumonia: the role of apoptosis. J Immunol 170: 4209–4216, 2003. doi: 10.4049/jimmunol.170.8.4209. [DOI] [PubMed] [Google Scholar]
- 56.Truong SV, Monick MM, Yarovinsky TO, Powers LS, Nyunoya T, Hunninghake GW. Extracellular signal-regulated kinase activation delays hyperoxia-induced epithelial cell death in conditions of Akt downregulation. Am J Respir Cell Mol Biol 31: 611–618, 2004. doi: 10.1165/rcmb.2004-0141OC. [DOI] [PubMed] [Google Scholar]
- 57.Usatyuk PV, Romer LH, He D, Parinandi NL, Kleinberg ME, Zhan S, Jacobson JR, Dudek SM, Pendyala S, Garcia JG, Natarajan V. Regulation of hyperoxia-induced NADPH oxidase activation in human lung endothelial cells by the actin cytoskeleton and cortactin. J Biol Chem 282: 23284–23295, 2007. doi: 10.1074/jbc.M700535200. [DOI] [PubMed] [Google Scholar]
- 58.Walsh MC, Szefler S, Davis J, Allen M, Van Marter L, Abman S, Blackmon L, Jobe A. Summary proceedings from the bronchopulmonary dysplasia group. Pediatrics 117, Suppl 1: S52–S56, 2006. doi: 10.1542/peds.2005-0620I. [DOI] [PubMed] [Google Scholar]
- 59.Wu M, He YH, Kobune M, Xu Y, Kelley MR, Martin WJ II. Protection of human lung cells against hyperoxia using the DNA base excision repair genes hOgg1 and Fpg. Am J Respir Crit Care Med 166: 192–199, 2002. doi: 10.1164/rccm.200112-130OC. [DOI] [PubMed] [Google Scholar]
- 60.Yee M, Vitiello PF, Roper JM, Staversky RJ, Wright TW, McGrath-Morrow SA, Maniscalco WM, Finkelstein JN, O’Reilly MA. Type II epithelial cells are critical target for hyperoxia-mediated impairment of postnatal lung development. Am J Physiol Lung Cell Mol Physiol 291: L1101–L1111, 2006. doi: 10.1152/ajplung.00126.2006. [DOI] [PubMed] [Google Scholar]