Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Int J Mol Sci ; 25(7)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38612589

ABSTRACT

Lung cancer is the leading cause of cancer death worldwide. Polycyclic aromatic hydrocarbons (PAHs) are metabolized by the cytochrome P450 (CYP)1A and 1B1 to DNA-reactive metabolites, which could lead to mutations in critical genes, eventually resulting in cancer. Omega-3 fatty acids, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are beneficial against cancers. In this investigation, we elucidated the mechanisms by which omega-3 fatty acids EPA and DHA will attenuate PAH-DNA adducts and lung carcinogenesis and tumorigenesis mediated by the PAHs BP and MC. Adult wild-type (WT) (A/J) mice, Cyp1a1-null, Cyp1a2-null, or Cyp1b1-null mice were exposed to PAHs benzo[a]pyrene (BP) or 3-methylcholanthrene (MC), and the effects of omega-3 fatty acid on PAH-mediated lung carcinogenesis and tumorigenesis were studied. The major findings were as follows: (i) omega-3 fatty acids significantly decreased PAH-DNA adducts in the lungs of each of the genotypes studied; (ii) decreases in PAH-DNA adduct levels by EPA/DHA was in part due to inhibition of CYP1B1; (iii) inhibition of soluble epoxide hydrolase (sEH) enhanced the EPA/DHA-mediated prevention of pulmonary carcinogenesis; and (iv) EPA/DHA attenuated PAH-mediated carcinogenesis in part by epigenetic mechanisms. Taken together, our results suggest that omega-3 fatty acids have the potential to be developed as cancer chemo-preventive agents in people.


Subject(s)
Fatty Acids, Omega-3 , Polycyclic Aromatic Hydrocarbons , Humans , Adult , Mice , Animals , Fatty Acids, Omega-3/pharmacology , DNA Adducts , Carcinogenesis , Cell Transformation, Neoplastic , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology
2.
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.
Environ Sci Technol ; 57(38): 14373-14383, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37683087

ABSTRACT

Transition metal catalysts can significantly enhance the pyrolytic remediation of soils contaminated with polycyclic aromatic hydrocarbons (PAHs). Significantly higher pyrene removal efficiency was observed after the pyrolytic treatment of Fe-enriched bentonite (1.8% wt ion-exchanged content) relative to natural bentonite or soil (i.e., 93% vs 48% and 4%) at the unprecedentedly low temperature of 150 °C with only 15 min treatment time. DFT calculations showed that bentonite surfaces with Fe3+ or Cu2+ adsorb pyrene stronger than surfaces with Zn2+ or Na+. Enhanced pyrene adsorption results from increased charge transfer from its aromatic π-bonds to the cation site, which destabilizes pyrene allowing for faster degradation at lower temperatures. UV-Vis and GC-MS analyses revealed pyrene decomposition products in extracts of samples treated at 150 °C, including small aromatic compounds. As the pyrolysis temperature increased above 200 °C, product distribution shifted from extractable compounds to char coating the residue particles. No extractable byproducts were detected after treatment at 400 °C, indicating that char was the final product of pyrene decomposition. Tests with human lung cells showed that extracts of samples pyrolyzed at 150 °C were toxic; thus, high removal efficiency by pyrolytic treatment does not guarantee detoxification. No cytotoxicity was observed for extracts from Fe-bentonite samples treated at 300 °C, inferring that char is an appropriate treatment end point. Overall, we demonstrate that transition metals in clay can catalyze pyrolytic reactions at relatively low temperatures to decrease the energy and contact times required to meet cleanup standards. However, mitigating residual toxicity may require higher pyrolysis temperatures.


Subject(s)
Bentonite , Polycyclic Aromatic Hydrocarbons , Humans , Temperature , Bentonite/chemistry , Pyrolysis , Pyrenes/chemistry , Soil
4.
Biol Sex Differ ; 14(1): 50, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37553579

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) is characterized by an arrest in lung development and is a leading cause of morbidity in premature neonates. It has been well documented that BPD disproportionally affects males compared to females, but the molecular mechanisms behind this sex-dependent bias remain unclear. Female mice show greater preservation of alveolarization and angiogenesis when exposed to hyperoxia, accompanied by increased miR-30a expression. In this investigation, we tested the hypothesis that loss of miR-30a would result in male and female mice experiencing similar impairments in alveolarization and angiogenesis under hyperoxic conditions. METHODS: Wild-type and miR-30a-/- neonatal mice were exposed to hyperoxia [95% FiO2, postnatal day [PND1-5] or room air before being euthanized on PND21. Alveolarization, pulmonary microvascular development, differences in lung transcriptome, and miR-30a expression were assessed in lungs from WT and miR-30a-/- mice of either sex. Blood transcriptomic signatures from preterm newborns (with and without BPD) were correlated with WT and miR-30a-/- male and female lung transcriptome data. RESULTS: Significantly, the sex-specific differences observed in WT mice were abrogated in the miR-30a-/- mice upon exposure to hyperoxia. The loss of miR-30a expression eliminated the protective effect in females, suggesting that miR-30a plays an essential role in regulating alveolarization and angiogenesis. Transcriptome analysis by whole lung RNA-Seq revealed a significant response in the miR-30a-/- female hyperoxia-exposed lung, with enrichment of pathways related to cell cycle and neuroactive ligand-receptor interaction. Gene expression signature in the miR-30a-/- female lung associated with human BPD blood transcriptomes. Finally, we showed the spatial localization of miR-30a transcripts in the bronchiolar epithelium. CONCLUSIONS: miR-30a could be one of the biological factors mediating the resilience of the female preterm lung to neonatal hyperoxic lung injury. A better understanding of the effects of miR-30a on pulmonary angiogenesis and alveolarization may lead to novel therapeutics for treating BPD.


Bronchopulmonary dysplasia (BPD) is a lung condition that affects babies born prematurely, causing problems with their lung development. Interestingly, BPD tends to affect boys more than girls, but we do not fully understand why. To investigate this, we conducted a study using mice. Female mice had better lung development and blood vessel formation when exposed to high oxygen levels. We found higher expression of a molecule called miR-30a in the female mice and seemed to be protective. So, we wanted to see if removing miR-30a would have the same effect on both male and female mice. To test this, we exposed newborn mice without miR-30a and normal mice to high oxygen levels or regular room air. Interestingly, the differences between normal males and females were no longer present in the mice without miR-30a. This suggested that miR-30a plays an important role in lung development. We also identified that the female mice without miR-30a, when exposed to high oxygen, had the greatest number of genes affected, and these gene changes were like those seen in blood samples from premature babies with BPD. Finally, we report that miR-30a was in a specific part of the lung called the bronchiolar epithelium. Overall, this study suggests that miR-30a is crucial in protecting premature lungs from damage caused by high oxygen levels. By understanding how miR-30a affects lung development, we may be able to develop new treatments for BPD in the future.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , MicroRNAs , Animals , Female , Male , Mice , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Hyperoxia/complications , Hyperoxia/metabolism , Lung/metabolism , Lung Injury/genetics , Lung Injury/complications , Lung Injury/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Sex Factors
5.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L314-L326, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37368978

ABSTRACT

Growth differentiation factor 15 (GDF15) is a divergent member of the transforming growth factor-ß (TGF-ß) superfamily, and its expression increases under various stress conditions, including inflammation, hyperoxia, and senescence. GDF15 expression is increased in neonatal murine bronchopulmonary dysplasia (BPD) models, and GDF15 loss exacerbates oxidative stress and decreases cellular viability in vitro. Our overall hypothesis is that the loss of GDF15 will exacerbate hyperoxic lung injury in the neonatal lung in vivo. We exposed neonatal Gdf15-/- mice and wild-type (WT) controls on a similar background to room air or hyperoxia (95% [Formula: see text]) for 5 days after birth. The mice were euthanized on postnatal day 21 (PND 21). Gdf15-/- mice had higher mortality and lower body weight than WT mice after exposure to hyperoxia. Hyperoxia exposure adversely impacted alveolarization and lung vascular development, with a greater impact in Gdf15-/- mice. Interestingly, Gdf15-/- mice showed lower macrophage count in the lungs compared with WT mice both under room air and after exposure to hyperoxia. Analysis of the lung transcriptome revealed marked divergence in gene expression and enriched biological pathways in WT and Gdf15-/- mice and differed markedly by biological sex. Notably, pathways related to macrophage activation and myeloid cell homeostasis were negatively enriched in Gdf15-/- mice. Loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice.NEW & NOTEWORTHY We show for the first time that loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice. We also highlight the distinct pulmonary transcriptomic response in the Gdf15-/- lung including pathways related to macrophage recruitment and activation.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , Animals , Female , Mice , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Hyperoxia/metabolism , Lung/metabolism , Lung Injury/genetics , Lung Injury/metabolism , Mice, Inbred C57BL
6.
Redox Biol ; 64: 102790, 2023 08.
Article in English | MEDLINE | ID: mdl-37348155

ABSTRACT

Oxygen supplementation is life saving for premature infants and for COVID-19 patients but can induce long-term pulmonary injury by triggering inflammation, with xenobiotic-metabolizing CYP enzymes playing a critical role. Murine studies showed that CYP1B1 enhances, while CYP1A1 and CYP1A2 protect from, hyperoxic lung injury. In this study we tested the hypothesis that Cyp1b1-null mice would revert hyperoxia-induced transcriptomic changes observed in WT mice at the transcript and pathway level. Wild type (WT) C57BL/6J and Cyp1b1-null mice aged 8-10 weeks were maintained in room air (21% O2) or exposed to hyperoxia (>95% O2) for 48h. Transcriptomic profiling was conducted using the Illumina microarray platform. Hyperoxia exposure led to robust changes in gene expression and in the same direction in WT, Cyp1a1-, Cyp1a2-, and Cyp1b1-null mice, but to different extents for each mouse genotype. At the transcriptome level, all Cyp1-null murine models reversed hyperoxia effects. Gene Set Enrichment Analysis identified 118 hyperoxia-affected pathways mitigated only in Cyp1b1-null mice, including lipid, glutamate, and amino acid metabolism. Cell cycle genes Cdkn1a and Ccnd1 were induced by hyperoxia in both WT and Cyp1b1-null mice but mitigated in Cyp1b1-null O2 compared to WT O2 mice. Hyperoxia gene signatures associated positively with bronchopulmonary dysplasia (BPD), which occurs in premature infants (with supplemental oxygen being one of the risk factors), but only in the Cyp1b1-null mice did the gene profile after hyperoxia exposure show a partial rescue of BPD-associated transcriptome. Our study suggests that CYP1B1 plays a pro-oxidant role in hyperoxia-induced lung injury.


Subject(s)
Bronchopulmonary Dysplasia , COVID-19 , Hyperoxia , Lung Injury , Humans , Infant, Newborn , Animals , Mice , Hyperoxia/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Lung Injury/genetics , Lung Injury/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Mice, Inbred C57BL , COVID-19/metabolism , Oxygen/metabolism , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/complications , Mice, Knockout , Lung/metabolism , Animals, Newborn
7.
Mol Metab ; 72: 101717, 2023 06.
Article in English | MEDLINE | ID: mdl-37004989

ABSTRACT

OBJECTIVE: Mitophagy removes damaged mitochondria to maintain cellular homeostasis. Aryl hydrocarbon receptor (AhR) expression in the liver plays a crucial role in supporting normal liver functions, but its impact on mitochondrial function is unclear. Here, we identified a new role of AhR in the regulation of mitophagy to control hepatic energy homeostasis. METHODS: In this study, we utilized primary hepatocytes from AhR knockout (KO) mice and AhR knockdown AML12 hepatocytes. An endogenous AhR ligand, kynurenine (Kyn), was used to activate AhR in AML12 hepatocytes. Mitochondrial function and mitophagy process were comprehensively assessed by MitoSOX and mt-Keima fluorescence imaging, Seahorse XF-based oxygen consumption rate measurement, and Mitoplate S-1 mitochondrial substrate utilization analysis. RESULTS: Transcriptomic analysis indicated that mitochondria-related gene sets were dysregulated in AhR KO liver. In both primary mouse hepatocytes and AML12 hepatocyte cell lines, AhR inhibition strongly suppressed mitochondrial respiration rate and substrate utilization. AhR inhibition also blunted the fasting response of several essential autophagy genes and the mitophagy process. We further identified BCL2 interacting protein 3 (BNIP3), a mitophagy receptor that senses nutrient stress, as an AhR target gene. AhR is directly recruited to the Bnip3 genomic locus, and Bnip3 transcription was enhanced by AhR endogenous ligand treatment in wild-type liver and abolished entirely in AhR KO liver. Mechanistically, overexpression of Bnip3 in AhR knockdown cells mitigated the production of mitochondrial reactive oxygen species (ROS) and restored functional mitophagy. CONCLUSIONS: AhR regulation of the mitophagy receptor BNIP3 coordinates hepatic mitochondrial function. Loss of AhR induces mitochondrial ROS production and impairs mitochondrial respiration. These findings provide new insight into how endogenous AhR governs hepatic mitochondrial homeostasis.


Subject(s)
Mitochondria , Receptors, Aryl Hydrocarbon , Mice , Animals , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Ligands , Mitochondria/metabolism , Liver/metabolism , Mice, Knockout , Homeostasis
8.
Pediatr Pulmonol ; 58(1): 271-278, 2023 01.
Article in English | MEDLINE | ID: mdl-36205439

ABSTRACT

Growth and differentiation factor 15 (GDF15) is a stress-responsive cytokine, and its expression increases during inflammation, hyperoxia, and senescence. Significantly, GDF15 is secreted by the placenta, and maternal levels increase throughout pregnancy. Serum GDF15 level is a promising biomarker for many lung diseases like pulmonary hypertension and pulmonary fibrosis. However, circulating GDF15 levels in preterm infants and their role as a predictor of respiratory outcomes have not been studied. We hypothesized that GDF15 levels would increase with gestational age at birth, and that postnatal GDF15 will be correlated with adverse respiratory outcomes in preterm infants. Scavenged blood samples were retrieved from 57 preterm infants at five time points, from birth until 36-weeks postmenstrual age (PMA). GDF15 levels were measured using ELISA in 114 samples. We performed two-sample t-test, correlation and linear regression, logistic regression, and mixed-effects linear models for statistical analysis, and significance was identified when p < 0.05. Contrary to our hypothesis, for every 1-week increase in gestational age at birth, the predicted GDF15 level decreased by 475.0 pg/ml (p < 0.001). Greater PMA was significantly associated with lower serum GDF15 levels (p < 0.001). Interestingly, higher GDF15 levels were associated with a longer need for mechanical ventilation (p = 0.034), prolonged respiratory support need (p < 0.001), and length of hospital stay (p = 0.006). In conclusion, in preterm infants, GDF15 levels show an inverse correlation with gestational age at birth, with higher levels in more preterm babies, and levels trend down postnatally. Furthermore, longitudinal GDF15 levels through 36 weeks PMA predict adverse respiratory outcomes in preterm infants.


Subject(s)
Bronchopulmonary Dysplasia , Infant, Premature , Pregnancy , Female , Infant, Newborn , Humans , Gestational Age , Respiration, Artificial , Growth Differentiation Factor 15
9.
Front Immunol ; 13: 1004603, 2022.
Article in English | MEDLINE | ID: mdl-36159798

ABSTRACT

Background: Cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1) pathway, which is regulated by aryl hydrocarbon receptor (AhR) plays an important role in chemical carcinogenesis and xenobiotic metabolism. Recently, we demonstrated that the microbial metabolite Urolithin A (UroA) mitigates colitis through its gut barrier protective and anti-inflammatory activities in an AhR-dependent manner. Here, we explored role of CYP1A1 in UroA-mediated gut barrier and immune functions in regulation of inflammatory bowel disease (IBD). Methods: To determine the role of CYP1A1 in UroA-mediated protectives activities against colitis, we subjected C57BL/6 mice and Cyp1a1 -/- mice to dextran sodium sulphate (DSS)-induced acute colitis model. The phenotypes of the mice were characterized by determining loss of body weight, intestinal permeability, systemic and colonic inflammation. Further, we evaluated the impact of UroA on regulation of immune cell populations by flow cytometry and confocal imaging using both in vivo and ex vivo model systems. Results: UroA treatment mitigated DSS-induced acute colitis in the wildtype mice. However, UroA-failed to protect Cyp1a1 -/- mice against colitis, as evident from non-recovery of body weight loss, shortened colon lengths and colon weight/length ratios. Further, UroA failed to reduce DSS-induced inflammation, intestinal permeability and upregulate tight junction proteins in Cyp1a1 -/- mice. Interestingly, UroA induced the expansion of T-reg cells in a CYP1A1-dependent manner both in vivo and ex vivo models. Conclusion: Our results suggest that CYP1A1 expression is essential for UroA-mediated enhanced gut barrier functions and protective activities against colitis. We postulate that CYP1A1 plays critical and yet unknown functions beyond xenobiotic metabolism in the regulation of gut epithelial integrity and immune systems to maintain gut homeostasis in IBD pathogenesis.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Colitis/pathology , Coumarins , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Inflammation , Mice , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Tight Junction Proteins/metabolism , Xenobiotics/adverse effects
11.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163440

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a cytoplasmic transcription factor that is well-known for regulating xenobiotic metabolism. Studies in knockout and transgenic mice indicate that the AHR plays a vital role in the development of liver and regulation of reproductive, cardiovascular, hematopoietic, and immune homeostasis. In this focused review on lung diseases associated with acute injury and alveolar development, we reviewed and summarized the current literature on the mechanistic role(s) and therapeutic potential of the AHR in acute lung injury, chronic obstructive pulmonary disease, and bronchopulmonary dysplasia (BPD). Pre-clinical studies indicate that endogenous AHR activation is necessary to protect neonatal and adult lungs against hyperoxia- and cigarette smoke-induced injury. Our goal is to provide insight into the high translational potential of the AHR in the meaningful management of infants and adults with these lung disorders that lack curative therapies.


Subject(s)
Acute Lung Injury/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bronchopulmonary Dysplasia/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Acute Lung Injury/drug therapy , Animals , Bronchopulmonary Dysplasia/drug therapy , Gene Expression Regulation/drug effects , Humans , Ligands , Molecular Targeted Therapy , Pulmonary Disease, Chronic Obstructive/drug therapy
12.
Antioxidants (Basel) ; 11(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35052622

ABSTRACT

Numerous human and animal studies have reported positive correlation between carcinogen-DNA adduct levels and cancer occurrence. Therefore, attenuation of DNA adduct levels would be expected to suppress tumorigenesis. In this investigation, we report that the antioxidants omega 3-fatty acids, which are constituents of fish oil (FO), significantly decreased DNA adduct formation by polycyclic aromatic hydrocarbons (PAHs). B6C3F1 male mice were fed an FO or corn oil (CO) diet, or A/J male mice were pre-fed with omega-3 fatty acids eicosapentaenoic acid (EPA) and/or docosahexaenoic acid (DHA). While the B6C3F1 mice were administered two doses of a mixture of seven carcinogenic PAHs including benzo(a)pyrene (BP), the A/J mice were treated i.p. with pure benzo[a]pyrene (BP). Animals were euthanized after 1, 3, or 7 d after PAH treatment. DNA adduct levels were measured by the 32P-postlabeling assay. Our results showed that DNA adduct levels in the lungs of mice 7 d after treatment were significantly decreased in the FO or EPA/DHA groups compared with the CO group. Interestingly, both qPCR and Western blot analyses revealed that FO, DHA and EPA/DHA significantly decreased the expression of cytochrome P450 (CYP) 1B1. CYP1B1 plays a critical role in the metabolic activation of BP to DNA-reactive metabolites. qPCR also showed that the expression of some metabolic and DNA repair genes was induced by BP and inhibited by FO or omega-3 fatty acids in liver, but not lung. Our results suggest that a combination of mechanism entailing CYP1B1 inhibition and the modulation of DNA repair genes contribute to the attenuation of PAH-mediated carcinogenesis by omega 3 fatty acids.

13.
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
14.
Semin Cancer Biol ; 76: 3-16, 2021 11.
Article in English | MEDLINE | ID: mdl-34242741

ABSTRACT

Lung cancer has the second highest incidence and highest mortality compared to all other cancers. Polycyclic aromatic hydrocarbon (PAH) molecules belong to a class of compounds that are present in tobacco smoke, diesel exhausts, smoked foods, as well as particulate matter (PM). PAH-derived reactive metabolites are significant contributors to lung cancer development. The formation of these reactive metabolites entails metabolism of the parent PAHs by cytochrome P4501A1/1B1 (CYP1A1/1B1) and epoxide hydrolase enzymes. These reactive metabolites then react with DNA to form DNA adducts, which contribute to key gene mutations, such as the tumor suppressor gene, p53 and are linked to pulmonary carcinogenesis. PAH exposure also leads to upregulation of CYP1A1 transcription by binding to the aryl hydrocarbon receptor (AHR) and eliciting transcription of the CYP1A1 promoter, which comprises specific xenobiotic-responsive element (XREs). While hepatic and pulmonary CYP1A1/1B1 metabolize PAHs to DNA-reactive metabolites, the hepatic CYP1A2, however, may protect against lung tumor development by suppressing both liver and lung CYP1A1 enzymes. Further analysis of these enzymes has shown that PAH-exposure also induces sustained transcription of CYP1A1, which is independent of the persistence of the parent PAH. CYP1A2 enzyme plays an important role in the sustained induction of hepatic CYP1A1. PAH exposure may further contribute to pulmonary carcinogenesis by producing epigenetic alterations. DNA methylation, histone modification, long interspersed nuclear element (LINE-1) activation, and non-coding RNA, specifically microRNA (miRNA) alterations may all be induced by PAH exposure. The relationship between PAH-induced enzymatic reactive metabolite formation and epigenetic alterations is a key area of research that warrants further exploration. Investigation into the potential interplay between these two mechanisms may lead to further understanding of the mechanisms of PAH carcinogenesis. These mechanisms will be crucial for the development of effective targeted therapies and early diagnostic tools.


Subject(s)
Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Lung Neoplasms/chemically induced , Polycyclic Aromatic Hydrocarbons/adverse effects , Polycyclic Aromatic Hydrocarbons/metabolism , Animals , Humans
15.
JCI Insight ; 6(13)2021 07 08.
Article in English | MEDLINE | ID: mdl-34061778

ABSTRACT

The main mechanisms underlying sexually dimorphic outcomes in neonatal lung injury are unknown. We tested the hypothesis that hormone- or sex chromosome-mediated mechanisms interact with hyperoxia exposure to impact injury and repair in the neonatal lung. To distinguish sex differences caused by gonadal hormones versus sex chromosome complement (XX versus XY), we used the Four Core Genotypes (FCG) mice and exposed them to hyperoxia (95% FiO2, P1-P4: saccular stage) or room air. This model generates XX and XY mice that each have either testes (with Sry, XXM, or XYM) or ovaries (without Sry, XXF, or XYF). Lung alveolarization and vascular development were more severely impacted in XYM and XYF compared with XXF and XXM mice. Cell cycle-related pathways were enriched in the gonadal or chromosomal females, while muscle-related pathways were enriched in the gonadal males, and immune-response-related pathways were enriched in chromosomal males. Female gene signatures showed a negative correlation with human patients who developed bronchopulmonary dysplasia (BPD) or needed oxygen therapy at 28 days. These results demonstrate that chromosomal sex - and not gonadal sex - impacted the response to neonatal hyperoxia exposure. The female sex chromosomal complement was protective and could mediate sex-specific differences in the neonatal lung injury.


Subject(s)
Bronchopulmonary Dysplasia , Gonadal Hormones/metabolism , Hyperoxia , Lung Injury , Oxygen Inhalation Therapy , Sex Chromosomes , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/therapy , Female , Humans , Hyperoxia/etiology , Hyperoxia/genetics , Hyperoxia/metabolism , Infant, Newborn , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/metabolism , Male , Mice , Ovary/metabolism , Oxygen Inhalation Therapy/adverse effects , Oxygen Inhalation Therapy/methods , Protective Factors , Risk Factors , Sex Characteristics , Testis/metabolism
16.
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
17.
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).

18.
Redox Biol ; 37: 101718, 2020 10.
Article in English | MEDLINE | ID: mdl-32961439

ABSTRACT

Sex as a biological variable plays a critical role both during lung development and in modulating postnatal hyperoxic lung injury and repair. The molecular mechanisms behind these sex-specific differences need to be elucidated. Our objective was to determine if the neonatal lung epigenomic landscape reconfiguration has profound effects on gene expression and could underlie sex-biased differences in protection from or susceptibility to diseases. Neonatal male and female mice (C57BL/6) were exposed to hyperoxia (95% FiO, PND 1-5: saccular stage) or room air and euthanized on PND 7 and 21. Pulmonary gene expression was studied using RNA-seq on Illumina HiSeq 2500 platform and quantified. Epigenomic landscape was assessed using Chromatin Immunoprecipitation (ChIP-Seq) of the H3K27ac histone modification mark, associated with active genes, enhancers, and super-enhancers. These data were then integrated, pathways identified and validated. Sex-biased epigenetic modulation of gene expression leads to differential regulation of biological processes in the developing lung at baseline and after exposure to hyperoxia. The female lung exhibits a more robust epigenomic response for the H3K27ac mark in response to hyperoxia. Epigenomic changes distribute over genomic and epigenomic domains in a sex-specific manner. The differential epigenomic responses also enrich for key transcription regulators crucial for lung development. In addition, by utilizing H3K27ac as the target epigenomic change we were also able to identify new epigenomic reprogramming at super-enhancers. Finally, we report for the first time that the upregulation of p21 (Cdkn1a) in the injured neonatal lung could be mediated through gain of H3K27ac. These data demonstrate that modulation of transcription via epigenomic landscape alterations may contribute to the sex-specific differences in preterm neonatal hyperoxic lung injury and repair.


Subject(s)
Hyperoxia , Animals , Animals, Newborn , Epigenesis, Genetic , Epigenomics , Female , Hyperoxia/genetics , Lung , Male , Mice , Mice, Inbred C57BL
19.
Free Radic Biol Med ; 160: 208-218, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32791187

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) leads to progressive lung injury, which significantly impacts patient morbidity and mortality but may differ clinically between the sexes. Cytochrome P450 (CYP) 1A enzymes are protective against hyperoxic lung injury and may contribute to sex-dependent pathology. NRF2 is a critical transcriptional regulator of antioxidants and loss of NRF2 leads to severe hyperoxic lung injury and mortality in mice. NRF2 deficiencies and polymorphisms have been observed in patients with pulmonary diseases such as chronic obstructive pulmonary disease and severe asthma. No prior studies have evaluated whether there are sex-specific differences in oxygen-mediated lung injury in Nrf2-/- mice and there are few rescue studies. OBJECTIVE: To test the hypothesis that hyperoxia induces greater lung injury and inflammation in Nrf2-/- mice compared to wild type (WT) that differs between sexes, and that this phenotype will be rescued by the administration of the cytochrome P450 (CYP) 1A inducer beta-naphthoflavone (BNF). DESIGN/METHODS: Male and female 8-10-week-old WT or Nrf2-/- C57BL/6 mice were pre-treated with BNF (40 mg/kg) or corn oil control and exposed to hyperoxia (95% O2) for 68 h. Survival, pulmonary edema, neutrophil recruitment, and lung injury scores were evaluated. Gene expression of phase II detoxification enzymes, pulmonary cytokines, and Cyp1a1/2 was quantified. CYP1A1/2 protein expression and catalytic activities were also measured. RESULTS: Hyperoxia exposure greatly reduced survival in Nrf2-/- mice, particularly in females. BNF treatment improved survival by 182.8% in Nrf2-/- females and by 41.4% in Nrf2-/- males as well as in WT females by 85.7%. Females had greater pulmonary edema as measured by lung weight to body weight ratios but was attenuated in all groups except Nrf2-/- females by BNF. Neutrophils doubled in Nrf2-/- lungs compared to WT in hyperoxia but were decreased in BNF-treated females of both genotypes. Pulmonary cytokine gene expression including Il-6 and Tnf-α increased in hyperoxia especially in Nrf2-/- mice and was unaffected by BNF. Pulmonary and hepatic Nqo1 gene expression w-as decreased in Nrf2-/- mice and was largely unaffected by BNF; however pulmonary Ho-1 did not vary significantly between the genotypes and was decreased in WT animals treated with BNF. Activities and protein expression of pulmonary and hepatic CYP1A1/2 were induced via BNF across all groups. Although hepatic Cyp1a2 gene expression was higher in Nrf2-/- males, the catalytic activity was higher in Nrf2-/- females. CONCLUSIONS: Hyperoxia augmented lung injury in Nrf2-/- mice, and pre-treatment with BNF was protective against mortality and injury, eliminating the sex-dependent survival difference in both genotypes. Our results support the hypothesis that NRF2 protects mice against lung injury, and the fact that BNF rescues the lung injury phenotype in Nrf2-/- mice suggests that augmented CYP1A expression by BNF may contribute to the beneficial effects. Further studies could lead to the development of BNF and other flavonoids for the prevention/treatment of hyperoxic lung injury, particularly in vulnerable patients with relative NRF2 deficiency, regardless of sex.


Subject(s)
Hyperoxia , Lung Injury , Animals , Cytochrome P-450 CYP1A1 , Female , Humans , Hyperoxia/genetics , Lung , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Oxygen , beta-Naphthoflavone/toxicity
20.
Expert Opin Drug Metab Toxicol ; 16(11): 1109-1124, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32841068

ABSTRACT

INTRODUCTION: Toxicity of chemotherapy drugs is the leading cause of poor therapeutic outcome in many cancer patients. Gastrointestinal (GI) toxicity and hepatotoxicity are among the most common side effects of current chemotherapies. Emerging studies indicate that many chemotherapy-induced toxicities are driven by drug metabolism, but very few reviews summarize the role of drug metabolism in chemotherapy-induced GI toxicity and hepatotoxicity. In this review, we highlighted the importance of drug metabolizing enzymes (DMEs) in chemotherapy toxicity. AREAS COVERED: Our review demonstrated that altered activity of DMEs play important role in chemotherapy-induced GI toxicity and hepatotoxicity. Besides direct changes in catalytic activities, the transcription of DMEs is also affected by inflammation, cell-signaling pathways, and/or by drugs in cancer patients due to the disease etiology. EXPERT OPINION: More studies should focus on how DMEs are altered during chemotherapy treatment, and how such changes affect the metabolism of chemotherapy drug itself. This mutual interaction between chemotherapies and DMEs can lead to excessive exposure of parent drug or toxic metabolites which ultimately cause GI adverse effect.


Subject(s)
Antineoplastic Agents/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Gastrointestinal Diseases/chemically induced , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Gastrointestinal Diseases/physiopathology , Humans , Neoplasms/drug therapy , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...