Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 211: 35-46, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38081439

ABSTRACT

Pregnant women exposed to polycyclic aromatic hydrocarbons (PAHs) are at increased risk for premature delivery. Premature infants often require supplemental oxygen, a known risk factor for bronchopulmonary dysplasia (BPD). Cytochrome P450 (CYP) enzymes have been implicated in hyperoxic lung injury. We hypothesize that prenatal PAH exposure exacerbates oxygen-mediated lung injury in neonatal mice, and that this effect is differentially altered in mice lacking the gene for (Cyp)1a1, 1a2, or 1b1. Timed pregnant wild type (WT) (C57BL/6J) mice were orally administered a PAH mixture of benzo[a]pyrene (BP) and benzo[b]fluoranthene (BbF) or the vehicle corn oil (CO) once daily on gestational days 16-19, and the dose response on postnatal lung injury was examined. In addition, timed pregnant mice with one of four genotypes, WT, Cyp1a1-null, Cyp1a2-null, and Cyp1b1-null, were treated orally with CO or PAH on gestational days 16-19 and exposed to hyperoxia or room air for 14 days. Lung injury was assessed on PND15 by radial alveolar count (RAC) and mean linear intercept (MLI) Gene expression of DNA repair genes in lung and liver were measured. Results showed that neonatal hyperoxic lung injury is augmented by prenatal PAH exposure in a dose-dependent manner. This effect was differentially altered in the Cyp-null mice, with Cyp1a2-null showing the greatest extent of lung injury. We concluded that newborn mice exposed to PAH in utero had more significant lung injury in response to hyperoxia than non-PAH exposed pups, and that CYP1A1 and CYP1A2 are protective against lung injury while CYP1B1 augments lung injury.


Subject(s)
Hyperoxia , Lung Injury , Polycyclic Aromatic Hydrocarbons , Prenatal Exposure Delayed Effects , Humans , Infant, Newborn , Female , Animals , Mice , Pregnancy , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Lung Injury/chemically induced , Hyperoxia/complications , Hyperoxia/genetics , Polycyclic Aromatic Hydrocarbons/toxicity , Polycyclic Aromatic Hydrocarbons/metabolism , Mice, Inbred C57BL , Lung/metabolism , Cytochrome P-450 Enzyme System , Oxygen , Mice, Knockout
3.
Oxid Med Cell Longev ; 2021: 5544600, 2021.
Article in English | MEDLINE | ID: mdl-34691356

ABSTRACT

Supplemental oxygen administration is frequently used in premature infants and adults with pulmonary insufficiency. NADPH quinone oxidoreductase (NQO1) protects cells from oxidative injury by decreasing reactive oxygen species (ROS). In this investigation, we tested the hypothesis that overexpression of NQO1 in BEAS-2B cells will mitigate cell injury and oxidative DNA damage caused by hyperoxia and that A-1221C single nucleotide polymorphism (SNP) in the NQO1 promoter would display altered susceptibility to hyperoxia-mediated toxicity. Using stable transfected BEAS-2B cells, we demonstrated that hyperoxia decreased cell viability in control cells (Ctr), but this effect was differentially mitigated in cells overexpressing NQO1 under the regulation of the CMV viral promoter, the wild-type NQO1 promoter (NQO1-NQO1), or the NQO1 promoter carrying the SNP. Interestingly, hyperoxia decreased the formation of bulky oxidative DNA adducts or 8-hydroxy-2'-deoxyguanosine (8-OHdG) in Ctr cells. qPCR studies showed that mRNA levels of CYP1A1 and NQO1 were inversely related to DNA adduct formation, suggesting the protective role of these enzymes against oxidative DNA injury. In SiRNA experiments entailing the NQO1-NQO1 promoter, hyperoxia caused decreased cell viability, and this effect was potentiated in cells treated with CYP1A1 siRNA. We also found that hyperoxia caused a marked induction of DNA repair genes DDB2 and XPC in Ctr cells, supporting the idea that hyperoxia in part caused attenuation of bulky oxidative DNA lesions by enhancing nucleotide excision repair (NER) pathways. In summary, our data support a protective role for human NQO1 against oxygen-mediated toxicity and oxidative DNA lesions in human pulmonary cells, and protection against toxicity was partially lost in SNP cells. Moreover, we also demonstrate a novel protective role for CYP1A1 in the attenuation of oxidative cells and DNA injury. Future studies on the mechanisms of attenuation of oxidative injury by NQO1 should help in developing novel approaches for the prevention/treatment of ARDS in humans.


Subject(s)
Lung/metabolism , Lung/physiopathology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress , Humans , Lung/pathology
4.
Expert Opin Drug Metab Toxicol ; 17(2): 171-178, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33215946

ABSTRACT

INTRODUCTION: Hyperoxic lung injury is a condition that can occur in patients in need of supplemental oxygen, such as premature infants with bronchopulmonary dysplasia or adults with acute respiratory distress syndrome. Cytochrome P450 (CYP) enzymes play critical roles in the metabolism of endogenous and exogenous compounds. AREAS COVERED: Through their complex pathways, some subfamilies of these enzymes may contribute to or protect against hyperoxic lung injury. Oxidative stress from reactive oxygen species (ROS) production is most likely a major contributor of hyperoxic lung injury. CYP1A enzymes have been shown to protect against hyperoxic lung injury while CYP1B enzymes seem to contribute to it. CYP2J2 enzymes help protect against hyperoxic lung injury by triggering EET production, thereby, increasing antioxidant enzymes. The metabolism of arachidonic acid to ω-terminal hydroxyeicosatetraenoic acid (20-HETEs) by CYP4A and CYP4F enzymes could impact hyperoxic lung injury via the vasodilating effects of 20-HETE. CYP2E1 and CYP2A enzymes may contribute to the oxidative stress in the lungs caused by ethanol- and nicotine-metabolism, respectively. EXPERT OPINION: Overall, the CYP enzymes, depending upon the isoform, play a contributory or protective role in hyperoxic lung injury, and are, therefore, ideal candidates for developing drugs that can treat oxygen-mediated lung injury.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hyperoxia/complications , Lung Injury/etiology , Adult , Animals , Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/physiopathology , Humans , Hyperoxia/enzymology , Infant, Newborn , Infant, Premature , Lung Injury/enzymology , Lung Injury/physiopathology , Oxidative Stress/physiology , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/physiopathology
5.
Curr Opin Toxicol ; 20-21: 77-84, 2020.
Article in English | MEDLINE | ID: mdl-33283080

ABSTRACT

Cytochrome P4501A (CYP1A) enzymes play important roles in xenobiotic and endobiotic metabolism. Due to uncoupling reactions during the enzymatic cycle, CYP1A enzymes can release reactive oxidative species (ROS) in the form of superoxide radical, hydrogen peroxide, hydroxyl radical etc. An imbalance between production of free radicals and the ability of antioxidants to detoxify the free radicals can lead to accumulation of ROS, which in turn can lead to oxidative stress. Oxidative stress can lead to inflammation and toxicity, which in turn can cause human diseases such as bronchopulmonary disease (BPD), ARDS, renal hypertension, etc. CYP1A enzymes, depending on the organ system, they either contribute or protect against oxidative injury. Thus, they have dual roles in regard to oxidative stress. This review presents an overview of the mechanistic relationship between CYP1A enzymes and oxidative stress in relation to various diseases in different organs (e.g., liver, lungs, heart, kidneys, and reproductive organs).

6.
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
7.
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
8.
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
9.
Biochem Biophys Res Commun ; 495(1): 408-413, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29101037

ABSTRACT

Hyperoxia contributes to the development of bronchopulmonary dysplasia (BPD) in premature infants. In this study, we tested the hypothesis that newborn transgenic mice carrying the human CYP1A1-Luc promoter will display transcriptional activation of the human CYP1A1 promoter in vivo upon exposure to hyperoxia, and that these mice will be less susceptible to hyperoxic lung injury and alveolar simplification than similarly exposed wild type (WT) mice. Newborn WT (CD-1) or transgenic mice carrying a 13.2 kb human CYP1A1 promoter and the luciferase (Luc) reporter gene (CYP1A1-luc) were maintained in room air or exposed to hyperoxia (85% O2) for 7-14 days. Hyperoxia exposure of CYP1A1-Luc mice for 7 and 14 days resulted in 4- and 30-fold increases, respectively, in hepatic Luc (CYP1A1) expression, compared to room air controls. In lung, hyperoxia caused a 2-fold induction of reporter Luc at 7 days, but the induction declined after 14 days. The newborn CYP1A1-Luc mice were less susceptible to lung injury and alveolar simplification than similarly exposed wild type (WT) CD-1 mice. Also, the CYP1A1-Luc mice showed increased levels of hepatic and pulmonary CYP1A1 expression and hepatic CYP1A2 activity after hyperoxia exposure. Hyperoxia also increased NADP(H) quinone reductase (NQO1) pulmonary gene expression in both CD-1 and CYP1A1-Luc mice at both time points, but this was more pronounced in the latter at 14 days. Our results support the hypothesis that hyperoxia activates the human CYP1A1 promoter in newborn mice, and that increased endogenous expression of CYP1A1 and NADP(H) quinone reductase (NQO1) contributes to the decreased susceptibilities to hyperoxic lung injury in the transgenic animals. This is the first report providing evidence of hyperoxia-mediated transcriptional activation of the human CYP1A1 promoter in newborn mice, and this in conjunction with decreased lung injury, suggests that these phenomena have important implications for BPD.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Hyperoxia/complications , Hyperoxia/genetics , Lung Injury/etiology , Lung Injury/genetics , Lung/pathology , Transcriptional Activation , Animals , Animals, Newborn , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Lung/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Transgenic , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxygen/metabolism , Promoter Regions, Genetic
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.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L991-L1005, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28818871

ABSTRACT

Bronchopulmonary dysplasia (BPD) is characterized by impaired alveolar secondary septation and vascular growth. Exposure to high concentrations of oxygen (hyperoxia) contributes to the development of BPD. The male sex is considered an independent risk factor for the development of BPD. The reasons underlying sexually dimorphic outcomes in premature neonates are not known. We hypothesized that sex-specific modulation of biological processes in the lung under hyperoxic conditions contributes to sex-based differences. Neonatal male and female mice (C57BL/6) were exposed to hyperoxia [95% [Formula: see text], postnatal day (PND) 1-5: saccular stage of lung development] and euthanized on PND 7 or 21. Pulmonary gene expression was studied using RNA-Seq on the Illumina HiSeq 2500 platform. Analysis of the pulmonary transcriptome revealed differential sex-specific modulation of crucial pathways such as angiogenesis, response to hypoxia, inflammatory response, and p53 pathway. Candidate genes from these pathways were validated at the mRNA level by qPCR. Analysis also revealed sex-specific differences in the modulation of crucial transcription factors. Focusing on the differential modulation of the angiogenesis pathway, we also showed sex-specific differential activation of Hif-1α-regulated genes using ChIP-qPCR and differences in expression of crucial genes (Vegf, VegfR2, and Phd2) modulating angiogenesis. We demonstrate the translational relevance of our findings by showing that our murine sex-specific differences in gene expression correlate with those from a preexisting human BPD data set. In conclusion, we provide novel molecular insights into differential sex-specific modulation of the pulmonary transcriptome in neonatal hyperoxic lung injury and highlight angiogenesis as one of the crucial differentially modulated pathways.


Subject(s)
Gene Expression Regulation , Hyperoxia/metabolism , Lung Injury/metabolism , Neovascularization, Physiologic , Sex Characteristics , Transcriptome , Animals , Female , Hyperoxia/pathology , Lung Injury/pathology , Male , Mice
12.
Sci Rep ; 7(1): 642, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28377578

ABSTRACT

Hyperoxia contributes to lung injury in experimental animals and diseases such as acute respiratory distress syndrome in humans. Cytochrome P450 (CYP)1A enzymes are protective against hyperoxic lung injury (HLI). The molecular pathways and differences in gene expression that modulate these protective effects remain largely unknown. Our objective was to characterize genotype specific differences in the transcriptome and proteome of acute hyperoxic lung injury using the omics platforms: microarray and Reverse Phase Proteomic Array. Wild type (WT), Cyp1a1-/- and Cyp1a2-/- (8-10 wk, C57BL/6J background) mice were exposed to hyperoxia (FiO2 > 0.95) for 48 hours. Comparison of transcriptome changes in hyperoxia-exposed animals (WT versus knock-out) identified 171 genes unique to Cyp1a1-/- and 119 unique to Cyp1a2-/- mice. Gene Set Enrichment Analysis revealed pathways including apoptosis, DNA repair and early estrogen response that were differentially regulated between WT, Cyp1a1-/- and Cyp1a2-/- mice. Candidate genes from these pathways were validated at the mRNA and protein level. Quantification of oxidative DNA adducts with 32P-postlabeling also revealed genotype specific differences. These findings provide novel insights into mechanisms behind the differences in susceptibility of Cyp1a1-/- and Cyp1a2-/- mice to HLI and suggest novel pathways that need to be investigated as possible therapeutic targets for acute lung injury.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Hyperoxia/complications , Lung Injury/etiology , Lung Injury/metabolism , Proteome , Transcriptome , Alleles , Animals , Computational Biology/methods , DNA Repair , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation , Genotype , Lung Injury/pathology , Male , Mice , Mice, Knockout , Molecular Sequence Annotation , Oxidative Stress , Proteomics/methods , Signal Transduction
13.
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
14.
Biochem Biophys Res Commun ; 476(4): 346-351, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27235555

ABSTRACT

Supplemental oxygen, which is routinely administered to preterm infants with pulmonary insufficiency, contributes to bronchopulmonary dysplasia (BPD) in these infants. Hyperoxia also contributes to the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) in adults. The mechanisms of oxygen-mediated pulmonary toxicity are not completely understood. Recent studies have suggested an important role for cytochrome P450 (CYP)1A1/1A2 in the protection against hyperoxic lung injury. The role of CYP1B1 in oxygen-mediated pulmonary toxicity has not been studied. In this investigation, we tested the hypothesis that CYP1B1 plays a mechanistic role in oxygen toxicity in pulmonary cells in vitro. In human bronchial epithelial cell line BEAS-2B, hyperoxic treatment for 1-3 days led to decreased cell viability by about 50-80%. Hyperoxic cytotoxicity was accompanied by an increase in levels of reactive oxygen species (ROS) by up to 110%, and an increase of TUNEL-positive cells by up to 4.8-fold. Western blot analysis showed hyperoxia to significantly down-regulate CYP1B1 protein level. Also, there was a decrease of CYP1B1 mRNA by up to 38% and Cyp1b1 promoter activity by up to 65%. On the other hand, CYP1B1 siRNA appeared to rescue the cell viability under hyperoxia stress, and overexpression of CYP1B1 significantly attenuated hyperoxic cytotoxicity after 48 h of incubation. In immortalized lung endothelial cells derived from Cyp1b1-null and wild-type mice, hyperoxia increased caspase 3/7 activities in a time-dependent manner, but endothelial cells lacking the Cyp1b1 gene showed significantly decreased caspase 3/7 activities after 48 and 72 h of incubation, implying that CYP1B1 might promote apoptosis in wild type lung endothelial cells under hyperoxic stress. In conclusion, our results support the hypothesis that CYP1B1 plays a mechanistic role in pulmonary oxygen toxicity, and CYP1B1-mediated apoptosis could be one of the mechanisms of oxygen toxicity. Thus, CYP1B1 could be a novel target for preventative and/or therapeutic interventions against BPD in infants and ALI/ARDS in adults.


Subject(s)
Acute Lung Injury/prevention & control , Cytochrome P-450 CYP1B1/metabolism , Hyperoxia/metabolism , Lung/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytochrome P-450 CYP1B1/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Enzymologic , Humans , Hyperoxia/genetics , Hyperoxia/pathology , Lung/cytology , Mice, Mutant Strains , RNA, Small Interfering , Receptors, Aryl Hydrocarbon/metabolism
16.
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
17.
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
18.
Biochem Biophys Res Commun ; 450(4): 1345-50, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24997337

ABSTRACT

Caffeine is used to prevent bronchopulmonary dysplasia (BPD) in premature neonates. Hyperoxia contributes to the development of BPD, inhibits cell proliferation and decreases cell survival. The mechanisms responsible for the protective effect of caffeine in pulmonary oxygen toxicity remain largely unknown. A549 and MLE 12 pulmonary epithelial cells were exposed to hyperoxia or maintained in room air, in the presence of different concentrations (0, 0.05, 0.1 and 1mM) of caffeine. Caffeine had a differential concentration-specific effect on cell cycle progression, oxidative stress and viability, with 1mM concentration being deleterious and 0.05 mM being protective. Reactive oxygen species (ROS) generation during hyperoxia was modulated by caffeine in a similar concentration-specific manner. Caffeine at 1mM, but not at the 0.05 mM concentration decreased the G2 arrest in these cells. Taken together this study shows the novel funding that caffeine has a concentration-specific effect on cell cycle regulation, ROS generation, and cell survival in hyperoxic conditions.


Subject(s)
Caffeine/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Lung/drug effects , Oxidative Stress/drug effects , Oxygen/metabolism , Animals , Cell Line , Cyclin-Dependent Kinase 2/metabolism , Dose-Response Relationship, Drug , Histones/metabolism , Lung/cytology , Lung/metabolism , Mice , Reactive Oxygen Species/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...