Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Toxicol Appl Pharmacol ; 339: 133-142, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29180065

ABSTRACT

Exposure to supraphysiological concentrations of oxygen (hyperoxia) leads to bronchopulmonary dysplasia (BPD), one of the most common pulmonary morbidities in preterm neonates, which is more prevalent in males than females. Beta-naphthoflavone (BNF) is protective against hyperoxic lung injury in adult and neonatal wild type (WT) mice and in and mice lacking Cyp1a1gene. In this investigation, we tested the hypothesis that BNF treatment will attenuate neonatal hyperoxic lung injury in WT and Cyp1a2-/- mice, and elucidated the effect of sex-specific differences. Newborn WT or Cyp1a2-/- mice were treated with BNF (10mg/kg) or the vehicle corn oil (CO) i.p., from postnatal day (PND) 2 to 8 once every other day, while being maintained in room air or hyperoxia (85% O2) for 14days. Hyperoxia exposure lead to alveolar simplification and arrest in angiogenesis in WT as well as Cyp1a2-/- mice No significant differences were seen between WT and Cyp1a2-/- mice. Cyp1a2-/- female mice had better preservation of pulmonary angiogenesis at PND15 compared to similarly exposed males. BNF treatment attenuated lung injury and inflammation in both genotypes, and this was accompanied by a significant induction of hepatic and pulmonary CYP1A1 in WT but not in Cyp1a2-/- mice. BNF treatment increased NADPH quinone oxidoreductase (NQO1) mRNA levels in Cyp1a2-/- mouse livers compared to WT mice. These results suggest that BNF is protective in neonatal mice exposed to hyperoxia independent of CYP1A2 and this may entail the protective effect of phase II enzymes like NQO1.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Cytochrome P-450 CYP1A2/deficiency , Hyperoxia/drug therapy , Hyperoxia/metabolism , beta-Naphthoflavone/therapeutic use , Acute Lung Injury/genetics , Animals , Animals, Newborn , Cytochrome P-450 CYP1A2/genetics , Enzyme Inhibitors/therapeutic use , Female , Hyperoxia/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Treatment Outcome
2.
Toxicol Appl Pharmacol ; 272(2): 281-90, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23792423

ABSTRACT

Sex-specific differences in pulmonary morbidity in humans are well documented. Hyperoxia contributes to lung injury in experimental animals and humans. The mechanisms responsible for sex differences in the susceptibility towards hyperoxic lung injury remain largely unknown. In this investigation, we tested the hypothesis that mice will display sex-specific differences in hyperoxic lung injury. Eight week-old male and female mice (C57BL/6J) were exposed to 72 h of hyperoxia (FiO2>0.95). After exposure to hyperoxia, lung injury, levels of 8-iso-prostaglandin F2 alpha (8-iso-PGF 2α) (LC-MS/MS), apoptosis (TUNEL) and inflammatory markers (suspension bead array) were determined. Cytochrome P450 (CYP)1A expression in the lung was assessed using immunohistochemistry and western blotting. After exposure to hyperoxia, males showed greater lung injury, neutrophil infiltration and apoptosis, compared to air-breathing controls than females. Pulmonary 8-iso-PGF 2α levels were higher in males than females after hyperoxia exposure. Sexually dimorphic increases in levels of IL-6 (F>M) and VEGF (M>F) in the lungs were also observed. CYP1A1 expression in the lung was higher in female mice compared to males under hyperoxic conditions. Overall, our results support the hypothesis that male mice are more susceptible than females to hyperoxic lung injury and that differences in inflammatory and oxidative stress markers contribute to these sex-specific dimorphic effects. In conclusion, this paper describes the establishment of an animal model that shows sex differences in hyperoxic lung injury in a temporal manner and thus has important implications for lung diseases mediated by hyperoxia in humans.


Subject(s)
Acute Lung Injury/etiology , Hyperoxia/complications , Lung Injury/etiology , Sex Characteristics , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Apoptosis , Biomarkers/analysis , Blotting, Western , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Oxidative Stress
3.
Toxicol Appl Pharmacol ; 267(3): 209-17, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23337360

ABSTRACT

Hyperoxia contributes to the development of bronchopulmonary dysplasia (BPD) in premature infants. New BPD is characterized as having alveolar simplification. We reported previously that aryl hydrocarbon receptor (AhR) deficiency increased susceptibility to hyperoxic lung injury in adult mice, and this was associated with decreased expression of cytochrome P450 1A enzymes and increased lung inflammation. Whether AhR protects newborn mice against hyperoxia-induced alveolar simplification is unknown. Thus, we tested the hypothesis that decreased activation of the pulmonary AhR augments hyperoxia-induced alveolar simplification and lung inflammation in newborn mice. Experimental groups included one-day old wild type (WT) and AhR dysfunctional (AhRd) mice exposed to 21% O2 (air) or 85% O2 (hyperoxia) for 14 days. Exposure of newborn WT mice to hyperoxia resulted in increased protein, enzyme and mRNA expression of the AhR-regulated lung cytochrome P450 1A1, NAD(P)H quinone oxidoreductase-1, and microsomal glutathione S-transferase 1 enzymes, suggesting that hyperoxia increases activation of the pulmonary AhR. On the other hand, in the AhRd mice, hyperoxia induced the AhR-regulated enzymes to a lesser extent probably due to the dysfunctional AhR in these mice. Alveolar simplification and lung inflammation was increased in mice exposed to hyperoxia compared with those exposed to air, and AhRd mice were more susceptible to hyperoxia-induced alveolar simplification and lung inflammation compared with WT mice. These findings suggest that decreased activation of the pulmonary AhR in newborn AhRd mice augments hyperoxia-induced alveolar simplification and lung inflammation in these mice.


Subject(s)
Hyperoxia/metabolism , Oxygen/toxicity , Pulmonary Alveoli/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Animals , Animals, Newborn , Hyperoxia/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Pulmonary Alveoli/pathology , Random Allocation , Receptors, Aryl Hydrocarbon/physiology
5.
Biochem Biophys Res Commun ; 407(1): 79-85, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21362406

ABSTRACT

Supplemental oxygen administration is frequently administered to pre-term and term infants having pulmonary insufficiency. However, hyperoxia contributes to the development of bronchopulmonary dysplasia (BPD) in premature infants. Cytochrome P450 (CYP)A enzymes have been implicated in hyperoxic lung injury. In this study, we tested the hypothesis that hyperoxia induces CYP1A1 and 1A2 enzymes by transcriptional activation of the corresponding promoters in vivo, and transgenic mice expressing the human CYP1A1 or the mouse 1A2 promoter would be more susceptible to hyperoxic lung injury than wild type (WT) mice. Adult WT (CD-1) (12week-old) mice, transgenic mice carrying a 10kb human CYP1A1 promoter and the luciferase (luc) reporter gene (CYP1A1-luc), or mice expressing the mouse CYP1A2 promoter (CYP1A2-luc) were maintained in room air or exposed to hyperoxia for 24-72h. Hyperoxia exposure of CYP1A1-luc mice for 24 and 48h resulted in 2.5- and 1.25-fold increases, respectively, in signal intensities, compared to room air controls. By 72h, the induction had declined to control levels. CYP1A2-luc mice also showed enhanced luc expression after 24-48h, albeit to a lesser extent than those expressing the CYP1A1 promoter. Also, these mice showed decreased levels of endogenous CYP1A1 and 1A2 expression after prolonged hyperoxia, and were also more susceptible to lung injury than similarly exposed WT mice, with CYP1A2-luc mice showing the greatest injury. Our results support the hypothesis that hyperoxia induces CYP1A enzymes by transcriptional activation of its corresponding promoters, and that decreased endogenous expression of these enzymes contribute to the increased susceptibilities to hyperoxic lung injury in the transgenic animals. In summary, this is the first report providing direct evidence of hyperoxia-mediated induction of CYP1A1 and CYP1A2 expression in vivo by mechanisms entailing transcriptional activation of the corresponding promoters, a phenomenon that has implications for hyperoxic lung injury, as well as other pathologies caused by oxidative stress.


Subject(s)
Acute Lung Injury/enzymology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Hyperoxia/complications , Transcriptional Activation , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Genes, Reporter , Humans , Hyperoxia/enzymology , Hyperoxia/pathology , Luciferases/genetics , Mice , Mice, Transgenic , Oxygen/toxicity , Oxygen Inhalation Therapy/adverse effects , Promoter Regions, Genetic , Transcription, Genetic
6.
J Pharmacol Exp Ther ; 339(1): 106-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21768223

ABSTRACT

Hyperoxia contributes to lung injury in experimental animals and bronchopulmonary dysplasia (BPD) in preterm infants. Cytochrome P4501A (CYP1A) enzymes, which are regulated by the aryl hydrocarbon receptor (AhR), have been shown to attenuate hyperoxic lung injury in rodents. Omeprazole, a proton pump inhibitor, used in humans to treat gastric acid-related disorders, induces hepatic CYP1A in vitro. However, the mechanism by which omeprazole induces CYP1A and its impact on CYP1A expression in vivo and hyperoxic lung injury are unknown. Therefore, we tested the hypothesis that omeprazole attenuates hyperoxic lung injury in adult wild-type (WT) C57BL/6J mice by an AhR-mediated induction of pulmonary and hepatic CYP1A enzymes. Accordingly, we determined the effects of omeprazole on pulmonary and hepatic CYP1A expression and hyperoxic lung injury in adult WT and AhR dysfunctional (AhRd) mice. We found that omeprazole attenuated lung injury in WT mice. Attenuation of lung injury by omeprazole paralleled enhanced pulmonary CYP1A1 and hepatic CYP1A2 expression in the omeprazole-treated mice. On the other hand, omeprazole failed to enhance pulmonary CYP1A1 and hepatic CYP1A2 expression and protect against hyperoxic lung injury in AhRd mice. In conclusion, our results suggest that omeprazole attenuates hyperoxic lung injury in mice by AhR-mediated mechanisms, and this phenomenon is associated with induction of CYP1A enzymes. These studies have important implications for the prevention and/or treatment of hyperoxia-induced disorders such as BPD in infants and acute respiratory distress syndrome in older children and adults.


Subject(s)
Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Inhibitors/pharmacology , Hyperoxia/pathology , Omeprazole/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Animals , Blotting, Western , Chemokine CCL2/metabolism , Enzyme Induction , Immunohistochemistry , Liver/enzymology , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Microsomes/drug effects , Microsomes/metabolism , Neutrophil Infiltration/drug effects , Organ Size/drug effects , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Edema/pathology , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Toxicol Appl Pharmacol ; 256(2): 83-94, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21745492

ABSTRACT

Supplemental oxygen contributes to the development of bronchopulmonary dysplasia (BPD) in premature infants. In this investigation, we tested the hypothesis that prenatal treatment of pregnant mice (C57BL/6J) with the cytochrome P450 (CYP)1A1 inducer, ß-napthoflavone (BNF), will lead to attenuation of lung injury in newborns (delivered from these dams) exposed to hyperoxia by mechanisms entailing transplacental induction of hepatic and pulmonary CYP1A enzymes. Pregnant mice were administered the vehicle corn oil (CO) or BNF (40 mg/kg), i.p., once daily for 3 days on gestational days (17-19), and newborns delivered from the mothers were either maintained in room air or exposed to hyperoxia (>95% O(2)) for 1-5 days. After 3-5 days of hyperoxia, the lungs of CO-treated mice showed neutrophil infiltration, pulmonary edema, and perivascular inflammation. On the other hand, BNF-pretreated neonatal mice showed decreased susceptibility to hyperoxic lung injury. These mice displayed marked induction of ethoxyresorufin O-deethylase (EROD) (CYP1A1) and methoxyresorufin O-demethylase (MROD) (CYP1A2) activities, and levels of the corresponding apoproteins and mRNA levels until PND 3 in liver, while CYP1A1 expression alone was augmented in the lung. Prenatal BNF did not significantly alter gene expression of pulmonary NAD(P)H quinone reductase (NQO1). Hyperoxia for 24-72 h resulted in increased pulmonary levels of the F(2)-isoprostane 8-iso-PGF(2α), whose levels were decreased in mice prenatally exposed to BNF. In conclusion, our results suggest that prenatal BNF protects newborns against hyperoxic lung injury, presumably by detoxification of lipid hydroperoxides by CYP1A enzymes, a phenomenon that has implications for prevention of BPD in infants.


Subject(s)
Acute Lung Injury/prevention & control , Bronchopulmonary Dysplasia/prevention & control , Cytochrome P-450 CYP1A1/drug effects , Hyperoxia/drug therapy , beta-Naphthoflavone/pharmacology , Acute Lung Injury/pathology , Animals , Animals, Newborn , Blotting, Western , Bronchopulmonary Dysplasia/drug therapy , Cytochrome P-450 CYP1A1/biosynthesis , Enzyme Induction/drug effects , Female , Humans , Infant, Newborn , Infant, Premature , Lung/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , beta-Naphthoflavone/therapeutic use
8.
J Pharmacol Exp Ther ; 335(2): 369-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20732958

ABSTRACT

The cytochrome P4501A (CYP1A) enzymes play important roles in the metabolic activation and detoxification of numerous environmental carcinogens, including polycyclic aromatic hydrocarbons (PAHs). In this study, we tested the hypothesis that hepatic CYP1A2 differentially regulates mouse hepatic and pulmonary CYP1A1 expression and suppresses transcriptional activation of human CYP1A1 (hCYP1A1) promoter in response to 3-methylcholanthrene (MC) in vivo. Administration of wild-type (WT) (C57BL/6J) or Cyp1a2-null mice with a single dose of MC (100 µmol/kg i.p.) caused significant increases in hepatic CYP1A1/1A2 activities, apoprotein content, and mRNA levels 1 day after carcinogen withdrawal compared with vehicle-treated controls. The induction persisted in the WT, but not Cyp1a2-null, animals, for up to 15 days. In the lung, MC caused persistent CYP1A1 induction for up to 8 days in both genotypes, with Cyp1a2-null mice displaying a greater extent of CYP1A1 expression. It is noteworthy that MC caused significant augmentation of human CYP1A1 promoter activation in transgenic mice expressing the hCYP1A1 and the reporter luciferase gene on a Cyp1a2-null background, compared with transgenic mice on the WT background. In contrast, the mouse endogenous hepatic, but not pulmonary, persistent CYP1A1 expression was repressed by MC in the hCYP1A1-Cyp1a2-null mice. Liquid chromatography-mass spectrometry experiments showed that CYP1A2 catalyzed the formation of 1-hydroxy-3-MC and/or 2-hydroxy-3-MC, a metabolite that may contribute to the regulation of CYP1A1 expression. In conclusion, the results suggest that CYP1A2 plays a pivotal role in the regulation of hepatic and pulmonary CYP1A1 by PAHs, a phenomenon that potentially has important implications for PAH-mediated carcinogenesis.


Subject(s)
Carcinogens, Environmental/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Liver/drug effects , Lung/drug effects , Methylcholanthrene/toxicity , Transcriptional Activation/drug effects , Animals , Blotting, Western , Carcinogens, Environmental/pharmacokinetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2/physiology , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Liver/enzymology , Lung/enzymology , Methylcholanthrene/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
9.
Toxicol Appl Pharmacol ; 233(2): 169-78, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18824009

ABSTRACT

Supplemental oxygen, used to treat pulmonary insufficiency in newborns, contributes to the development of bronchopulmonary dysplasia (BPD). Cytochrome P4501A enzymes are induced by hyperoxia in animal models, but their role in human systems is unknown. Here we investigated the molecular mechanisms of induction of CYP1A1 by hyperoxia in human lung cell lines. Three human lung cell lines were exposed to hyperoxia (95% O2) for 0-72 h, and CYP1A1 activities, apoprotein contents, and mRNA levels were determined. Hyperoxia significantly induced CYP1A1 activity and protein contents (2-4 fold), and mRNA levels (30-40 fold) over control in each cell line. Transfection of a CYP1A1 promoter/luciferase reporter construct, followed by hyperoxia (4-72 h), showed marked (2-6 fold) induction of luciferase expression. EMSA and siRNA experiments strongly suggest that the Ah receptor (AHR) is involved in the hyperoxic induction of CYP1A1. MTT reduction assays showed attenuation of cell injury with the CYP1A1 inducer beta-naphthoflavone (BNF). Our results strongly suggest that hyperoxia transcriptionally activates CYP1A1 expression in human lung cell lines by AHR-dependent mechanisms, and that CYP1A1 induction is associated with decreased toxicity. This novel finding of induction of CYP1A1 in the absence of exogenous AHR ligands could lead to novel interventions in the treatment of BPD.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation, Enzymologic , Hyperoxia/complications , Receptors, Aryl Hydrocarbon/metabolism , Apoproteins/metabolism , Cell Line , Cytochrome P-450 CYP1A1/metabolism , Electrophoretic Mobility Shift Assay , Genes, Reporter , Humans , Luciferases/metabolism , Lung , Lung Injury/etiology , Lung Injury/physiopathology , Promoter Regions, Genetic , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic , Transfection
10.
Curr Opin Toxicol ; 7: 102-109, 2018 Feb.
Article in English | MEDLINE | ID: mdl-35784947

ABSTRACT

Oxygen supplementation has been used as a part of respiratory care for preterm and term newborns since the beginning of 19th century. Although oxygen administration can be life-saving, reactive oxygen species (ROS) and reactive nitrogen species (RNS) due to hyperoxia can have detrimental effects in the developing organs of the preterm infants, with both short and long term consequences. Oxygen toxicity on the immature tissues of preterm infants can contribute to the development of several diseases like retinopathy of prematurity (ROP) and bronchopulmonary dysplasia (BPD). The vascular development of human retina is completed at term, whereas the neural retina develops up to 5 years of age. Disruption of the normal retinal neurovascular growth is the pathognomonic feature of ROP, and can lead to vision threatening disease or even blindness. It is estimated that at least 100,000 infants all over the world will be blind every year due to ROP, which is the leading cause of blindness in children. In this review we will discuss the role of ROS and RNS in the development of ROP, and how through historical, epidemiological, and developmental aspects of this devastating disease, we can design future research for its prevention and treatment.

11.
Food Chem Toxicol ; 114: 23-33, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29432836

ABSTRACT

Quercetin (QU) is one of the most common flavonoids that are present in a wide variety of fruits, vegetables, and beverages. This compound possesses potent anti-inflammatory and anti-oxidant properties. Supplemental oxygen is routinely administered to premature infants with pulmonary insufficiency. However, hyperoxia is one of the major risk factors for the development of bronchopulmonary dysplasia (BPD), which is also termed chronic lung disease in premature infants. Currently, no preventive approaches have been reported against BPD. The treatment of BPD is notably limited to oxygen administration, ventilatory support, and steroids. Since QU has been shown to be effective in reducing inflammation and oxidative stress in various disease models, we hypothesized that the postnatal QU treatment of newborn mice will protect against hyperoxic lung injury by the upregulation of the phase I (CYP1A/B) and/or phase II, NADPH quinone reductase enzymes. Newborn C57BL/6J mice within 24 h of birth with the nursing dams were exposed to either 21% O2 (air) and/or 85% O2 (hyperoxia) for 7 days. The mice were treated, intraperitoneally (i.p.) once every other day with quercetin, at a concentration of 20 mg/kg, or saline alone from postnatal day (PND) 2-6. The mice were sacrificed on day 7, and lung and liver tissues were collected. The expression levels of CYP1A1, CYP1B1, NQO1 proteins and mRNA as well as the levels of MDA-protein adducts were analyzed in lung and liver tissues. The findings indicated that QU attenuated hyperoxia-mediated lung injury by reducing inflammation and improving alveolarization with decreased number of neutrophil and macrophage infiltration. The attenuation of this lung injury correlated with the upregulation of CYP1A1/CYP1B1/NQO1 mRNA, proteins and the down regulation of NF-kB levels and MDA-protein adducts in lung and liver tissues. The present study demonstrated the potential therapeutic value of quercetin in the prevention and/or treatment of BPD.


Subject(s)
Bronchopulmonary Dysplasia/drug therapy , Hyperoxia/drug therapy , Quercetin/administration & dosage , Animals , Animals, Newborn/metabolism , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Humans , Hyperoxia/genetics , Hyperoxia/metabolism , Infant, Newborn , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Oxygen/metabolism
12.
Toxicol Sci ; 165(2): 462-474, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29939353

ABSTRACT

Supplemental oxygen is a life-saving intervention administered to individuals suffering from respiratory distress, including adults with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Despite the clinical benefit, supplemental oxygen can create a hyperoxic environment that increases reactive oxygen species, oxidative stress, and lung injury. We have previously shown that cytochrome P450 (CYP)1A enzymes decrease susceptibility to hyperoxia-induced lung injury. In this investigation, we determined the role of CYP1B1 in hyperoxic lung injury in vivo. Eight- to ten-week old C57BL/6 wild type (WT) and Cyp1b1-/- mice were exposed to hyperoxia (>95% O2) for 24-72 h or maintained in room air (21% O2). Lung injury was assessed by histology and lung weight to body weight (LW/BW) ratios. Extent of inflammation was determined by assessing pulmonary neutrophil infiltration and cytokine levels. Lipid peroxidation markers were quantified by gas chromatography mass spectrometry, and oxidative DNA adducts were quantified by 32P-postlabeling as markers of oxidative stress. We found that Cyp1b1-/- mice displayed attenuation of lung weight and pulmonary edema, particularly after 48-72 h of hyperoxia compared with WT controls. Further, Cyp1b1-/- mice displayed decreased levels of pulmonary oxidative DNA adducts and pulmonary isofurans after 24 h of hyperoxia. Cyp1b1-/- mice also showed increased pulmonary CYP1A1 and 1A2 and mRNA expression. In summary, our results support the hypothesis that Cyp1b1-/- mice display decreased hyperoxic lung injury than wild type counterparts and that CYP1B1 may act as a pro-oxidant during hyperoxia exposure, contributing to increases in oxidative DNA damage and accumulation of lipid hydroperoxides.


Subject(s)
Acute Lung Injury/etiology , Cytochrome P-450 CYP1B1/genetics , DNA Damage , Hyperoxia/complications , Oxidative Stress , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Hyperoxia/enzymology , Hyperoxia/pathology , Lipid Peroxidation/genetics , Lipid Peroxides/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics
14.
Toxicol Sci ; 157(1): 260-271, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28201809

ABSTRACT

Prolonged hyperoxia contributes to bronchopulmonary dysplasia (BPD) in preterm infants. ß-Naphthoflavone (BNF) is a potent inducer of cytochrome P450 (CYP)1A enzymes, which have been implicated in hyperoxic injuries in adult mice. In this investigation, we tested the hypothesis that newborn mice lacking the Cyp1a1 gene would be more susceptible to hyperoxic lung injury than wild-type (WT) mice and that postnatal BNF treatment would rescue this phenotype by mechanisms involving CYP1A and/or NAD(P)H quinone oxidoreductase (NQO1) enzymes. Newborn WT or Cyp1a1-null mice were treated with BNF (10 mg/kg) or the vehicle corn oil (CO) i.p., from postnatal day (PND) 2 to 14 once every other day, while being maintained in room air or hyperoxia (85% O2) for 14 days. Both genotypes showed lung injury, inflammation, and alveolar simplification in hyperoxia, with Cyp1a1-null mice displaying increased susceptibility compared to WT mice. BNF treatment resulted in significant attenuation of lung injury and inflammation, with improved alveolarization in both WT and Cyp1a1-null mice. BNF exposed normoxic or hyperoxic WT mice showed increased expression of hepatic CYP1A1/1A2, pulmonary CYP1A1, and NQO1 expression at both mRNA and protein levels, compared with vehicle controls. However, BNF caused greater induction of hepatic CYP1A2 and pulmonary NQO1 enzymes in the Cyp1a1-null mice, suggesting that BNF protects against hyperoxic lung injury in WT and Cyp1a1-null mice through the induction of CYP1A and NQO1 enzymes. Further studies on the protective role of flavonoids against hyperoxic lung injury in newborns could lead to novel strategies for the prevention and/or treatment of BPD.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Cytochrome P-450 CYP1A1/genetics , Infant, Premature , Oxygen/administration & dosage , beta-Naphthoflavone/administration & dosage , Animals , Animals, Newborn , Blotting, Western , Bronchopulmonary Dysplasia/genetics , Genetic Predisposition to Disease , Humans , Infant, Newborn , Mice , Mice, Knockout , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/enzymology
15.
Toxicology ; 331: 14-23, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25703676

ABSTRACT

Sex-specific differences in pulmonary morbidity in adults and preterm infants are well documented. Hyperoxia contributes to lung injury in experimental animals and humans. Cytochrome P450 (CYP) 1A enzymes have been shown to play a mechanistic role in hyperoxic lung injury (HLI) in animal models. Whether CYP1A enzymes contribute to gender-specific differences in relation to HLI is unknown. In this investigation, we tested the hypothesis that mice will display gender-specific differences in HLI, and that this phenomenon will be altered in mice lacking the genes for Cyp1a1 or 1a2. Eight week-old male and female wild type (WT) (C57BL/6J) mice, Cyp1a1-/-, and Cyp1a2-/- mice were exposed to 72h of hyperoxia (FiO2>0.95). Lung injury and inflammation were assessed and pulmonary and hepatic CYP1A1 and CYP1A2 levels were quantified at the enzyme activity, protein and mRNA level. Upon exposure to hyperoxia, liver and lung microsomal proteins showed higher pulmonary CYP1A1 (apoprotein level and activity) in WT females compared to WT males and a greater induction in hepatic CYP1A2 mRNA levels and activity in WT females after hyperoxia exposure. The gender based female advantage was lost or reversed in Cyp1a1-/- and Cyp1a2-/- mice. These findings suggest an important role for CYP1A enzymes in the gender-specific modulation of hyperoxic lung injury.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Hyperoxia/complications , Liver/enzymology , Lung Injury/etiology , Lung/enzymology , Animals , Body Weight , Cytochrome P-450 CYP1A1/deficiency , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/deficiency , Cytochrome P-450 CYP1A2/genetics , Female , Gene Expression Regulation, Enzymologic , Lung/pathology , Lung Injury/enzymology , Lung Injury/genetics , Lung Injury/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Organ Size , RNA, Messenger/metabolism , Sex Factors , Time Factors
16.
Free Radic Biol Med ; 82: 147-59, 2015 May.
Article in English | MEDLINE | ID: mdl-25680282

ABSTRACT

Hyperoxia contributes to acute lung injury in diseases such as acute respiratory distress syndrome. Cytochrome P450 (CYP) 1A enzymes have been implicated in hyperoxic lung injury, but the mechanistic role of CYP1A2 in pulmonary injury is not known. We hypothesized that mice lacking the gene Cyp1a2 (which is predominantly expressed in the liver) will be more sensitive to lung injury and inflammation mediated by hyperoxia and that CYP1A2 will play a protective role by attenuating lipid peroxidation and oxidative stress in the lung. Eight- to ten-week-old WT (C57BL/6) or Cyp1a2(-/-) mice were exposed to hyperoxia (>95% O2) or maintained in room air for 24-72 h. Lung injury was assessed by determining the ratio of lung weight/body weight (LW/BW) and by histology. Extent of inflammation was determined by measuring the number of neutrophils in the lung as well as cytokine expression. The Cyp1a2(-/-) mice under hyperoxic conditions showed increased LW/BW ratios, lung injury, neutrophil infiltration, and IL-6 and TNF-α levels and augmented lipid peroxidation, as evidenced by increased formation of malondialdehyde- and 4-hydroxynonenal-protein adducts and pulmonary isofurans compared to WT mice. In vitro experiments showed that the F2-isoprostane PGF2-α is metabolized by CYP1A2 to a dinor metabolite, providing evidence for a catalytic role for CYP1A2 in the metabolism of F2-isoprostanes. In summary, our results support the hypothesis that hepatic CYP1A2 plays a critical role in the attenuation of hyperoxic lung injury by decreasing lipid peroxidation and oxidative stress in vivo.


Subject(s)
Cytochrome P-450 CYP1A2/genetics , Hyperoxia/metabolism , Interleukin-6/metabolism , Lung Injury/metabolism , Tumor Necrosis Factor-alpha/metabolism , Aldehydes/metabolism , Animals , Cytochrome P-450 CYP1A2/metabolism , Dinoprost/metabolism , F2-Isoprostanes/metabolism , Leukocyte Count , Lipid Peroxidation/physiology , Liver/metabolism , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils , Oxidative Stress/physiology
17.
PLoS One ; 9(7): e101581, 2014.
Article in English | MEDLINE | ID: mdl-25003466

ABSTRACT

Exposure to high concentration of oxygen (hyperoxia) leads to lung injury in experimental animal models and plays a role in the pathogenesis of diseases such as Acute Respiratory Distress Syndrome (ARDS) and Bronchopulmonary dysplasia (BPD) in humans. The mechanisms responsible for sex differences in the susceptibility towards hyperoxic lung injury remain largely unknown. The major goal of this study was to characterize the changes in the pulmonary transcriptome following hyperoxia exposure and further elucidate the sex-specific changes. Male and female (8-10 wk) wild type (WT) (C57BL/6J) mice were exposed to hyperoxia (FiO2>0.95) and gene expression in lung tissues was studied at 48 h. A combination of fold change ≥1.4 and false discovery rate (FDR)<5% was used to define differentially expressed genes (DEGs). Overrepresentation of gene ontology terms representing biological processes and signaling pathway impact analysis (SPIA) was performed. Comparison of DEG profiles identified 327 genes unique to females, 585 unique to males and 1882 common genes. The major new findings of this study are the identification of new candidate genes of interest and the sex-specific transcriptomic changes in hyperoxic lung injury. We also identified DEGs involved in signaling pathways like MAP kinase and NF-kappa B which may explain the differences in sex-specific susceptibility to hyperoxic lung injury. These findings highlight changes in the pulmonary transcriptome and sex-specific differences in hyperoxic lung injury, and suggest new pathways, whose components could serve as sex-specific biomarkers and possible therapeutic targets for acute lung injury (ALI)/acute respiratory distress (ARDS) in humans.


Subject(s)
Acute Lung Injury/etiology , Gene Expression Regulation , Hyperoxia/complications , Transcriptome , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Female , Gene Expression Profiling , Immunohistochemistry , Male , Mice , Reproducibility of Results , Sex Factors , Signal Transduction
18.
Toxicol Sci ; 141(1): 68-77, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24893714

ABSTRACT

Hyperoxia contributes to acute lung injury in diseases such as acute respiratory distress syndrome in adults and bronchopulmonary dysplasia in premature infants. Cytochrome P450 (CYP)1A1 has been shown to modulate hyperoxic lung injury. The mechanistic role(s) of CYP1A1 in hyperoxic lung injury in vivo is not known. In this investigation, we hypothesized that Cyp1a1(-/-) mice would be more susceptible to hyperoxic lung injury than wild-type (WT) mice, and that the protective role of CYP1A1 is in part due to CYP1A1-mediated decrease in the levels of reactive oxygen species-mediated lipid hydroperoxides, e.g., F2-isoprostanes/isofurans, leading to attenuation of oxidative damage. Eight- to ten-week-old male WT (C57BL/6J) or Cyp1a1(-/-) mice were exposed to hyperoxia (>95% O2) or room air for 24-72 h. The Cyp1a1(-/-) mice were more susceptible to oxygen-mediated lung damage and inflammation than WT mice, as evidenced by increased lung weight/body weight ratio, lung injury, neutrophil infiltration, and augmented expression of IL-6. Hyperoxia for 24-48 h induced CYP1A expression at the mRNA, protein, and enzyme levels in liver and lung of WT mice. Pulmonary F2-isoprostane and isofuran levels were elevated in WT mice after hyperoxia for 24 h. On the other hand, Cyp1a1(-/-) mice showed higher levels after 48-72 h of hyperoxia exposure compared to WT mice. Our results support the hypothesis that CYP1A1 protects against hyperoxic lung injury by decreasing oxidative stress. Future research could lead to the development of novel strategies for prevention and/or treatment of acute lung injury.


Subject(s)
Acute Lung Injury/genetics , Cytochrome P-450 CYP1A1/genetics , Hyperoxia/genetics , Oxidative Stress/genetics , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Blotting, Western , Body Weight , Hyperoxia/enzymology , Hyperoxia/pathology , Lipid Peroxidation/genetics , Lung/enzymology , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Organ Size
19.
Toxicol Lett ; 230(2): 322-32, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24657529

ABSTRACT

Maternal smoking is one of the risk factors for preterm birth and for the development of bronchopulmonary dysplasia (BPD). In this study, we tested the hypothesis that prenatal exposure of rats to benzo[a]pyrene (BP), a component of cigarette smoke, will result in increased susceptibility of newborns to oxygen-mediated lung injury and alveolar simplification, and that cytochrome P450 (CYP)1A and 1B1 enzymes and oxidative stress mechanistically contribute to this phenomenon. Timed pregnant Fisher 344 rats were administered BP (25 mg/kg) or the vehicle corn oil (CO) on gestational days 18, 19 and 20, and newborn rats were either maintained in room air or exposed to hyperoxia (85% O2) for 7 or 14 days. Hyperoxic newborn rats prenatally exposed to the vehicle CO showed lung injury and alveolar simplification, and inflammation, and these effects were potentiated in rats that were prenatally exposed to BP. Prenatal exposure to BP, followed by hyperoxia, also resulted in significant modulation of hepatic and pulmonary cytochrome P450 (CYP)1A and 1B1 enzymes at PND 7-14. These rats displayed significant oxidative stress in lungs at postnatal day (PND) 14, as evidenced by increased levels of the F2-isoprostane 8-iso-PGF2α. Furthermore, these animals showed BP-derived DNA adducts and oxidative DNA adducts in the lung. In conclusion, our results show increased susceptibility of newborns to oxygen-mediated lung injury and alveolar simplification following maternal exposure to BP, and our results suggest that modulation of CYP1A/1B1 enzymes, increases in oxidative stress, and BP-DNA adducts contributed to this phenomenon.


Subject(s)
Benzo(a)pyrene/toxicity , Fetus/drug effects , Hyperoxia/complications , Lung Injury/etiology , Pulmonary Alveoli/drug effects , Animals , Animals, Newborn , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , DNA Adducts/analysis , Female , Pregnancy , Rats , Rats, Inbred F344
20.
Free Radic Biol Med ; 51(10): 1910-7, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21906671

ABSTRACT

Hyperoxia contributes to the development of bronchopulmonary dysplasia in premature infants. Earlier we observed that aryl hydrocarbon receptor (AhR)-deficient mice are more susceptible to hyperoxic lung injury than AhR-sufficient mice, and this phenomenon was associated with a lack of expression of cytochrome P450 1A enzymes. Omeprazole, a proton pump inhibitor used in humans with gastric acid-related disorders, activates AhR in hepatocytes in vitro. However, the effects of omeprazole on AhR activation in the lungs and its impact on hyperoxia-induced reactive oxygen species (ROS) generation and inflammation are unknown. In this study, we tested the hypothesis that omeprazole attenuates hyperoxia-induced cytotoxicity, ROS generation, and expression of monocyte chemoattractant protein-1 (MCP-1) in human lung-derived H441 cells via AhR activation. Experimental groups included cells transfected with AhR small interfering RNA (siRNA). Hyperoxia resulted in significant increases in cytotoxicity, ROS generation, and MCP-1 production, which were significantly attenuated with the functional activation of AhR by omeprazole. The protective effects of omeprazole on cytotoxicity, ROS production, and MCP-1 production were lost in H441 cells whose AhR gene was silenced by AhR siRNA. These findings support the hypothesis that omeprazole protects against hyperoxic injury in vitro via AhR activation that is associated with decreased ROS generation and expression of MCP-1.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Epithelial Cells/drug effects , Hyperoxia/drug therapy , Omeprazole/administration & dosage , Receptors, Aryl Hydrocarbon/metabolism , Anti-Inflammatory Agents/adverse effects , Cell Line, Tumor , Cell Survival/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cytoprotection/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Hyperoxia/pathology , Hyperoxia/physiopathology , Lung/pathology , Lung Injury/prevention & control , Omeprazole/adverse effects , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/immunology
SELECTION OF CITATIONS
SEARCH DETAIL