Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 16353, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004997

ABSTRACT

The influence of maternal high-fat diet (HFD) on metabolic response to ozone was examined in Long-Evans rat offspring. F0 females were fed control diet (CD; 10%kcal from fat) or HFD (60%kcal from fat) starting at post-natal day (PND) 30. Rats were bred on PND 72. Dietary regimen was maintained until PND 30 when all offspring were switched to CD. On PND 40, F1 offspring (n = 10/group/sex) were exposed to air or 0.8 ppm ozone for 5 h. Serum samples were collected for global metabolomic analysis (n = 8/group/sex). Offspring from HFD dams had increased body fat and weight relative to CD. Metabolomic analysis revealed significant sex-, diet-, and exposure-related changes. Maternal HFD increased free fatty acids and decreased phospholipids (male > female) in air-exposed rats. Microbiome-associated histidine and tyrosine metabolites were increased in both sexes, while 1,5-anhydroglucitol levels decreased in males indicating susceptibility to insulin resistance. Ozone decreased monohydroxy fatty acids and acyl carnitines and increased pyruvate along with TCA cycle intermediates in females (HFD > CD). Ozone increased various amino acids, polyamines, and metabolites of gut microbiota in HFD female offspring indicating gut microbiome alterations. Collectively, these data suggest that maternal HFD increases offspring susceptibility to metabolic alterations in a sex-specific manner when challenged with environmental stressors.


Subject(s)
Diet, High-Fat , Maternal Nutritional Physiological Phenomena/physiology , Ozone/administration & dosage , Prenatal Exposure Delayed Effects/metabolism , Stress, Physiological/physiology , Animals , Fatty Acids, Nonesterified/blood , Female , Gastrointestinal Microbiome , Male , Metabolomics , Phospholipids/blood , Pregnancy , Rats , Rats, Long-Evans , Sex Factors
2.
Sci Rep ; 9(1): 17925, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784596

ABSTRACT

Agonists of ß2 adrenergic receptors (ß2AR) and glucocorticoid receptors (GR) are prescribed to treat pulmonary diseases. Since ozone effects are mediated through the activation of AR and GR, we hypothesized that the treatment of rats with relevant therapeutic doses of long acting ß2AR agonist (LABA; clenbuterol; CLEN) and/or GR agonist (dexamethasone; DEX) would exacerbate ozone-induced pulmonary and systemic changes. In the first study, male 12-week-old Wistar-Kyoto rats were injected intraperitoneally with vehicle (saline), CLEN (0.004 or 0.02 mg/kg), or DEX (0.02 or 0.1 mg/kg). Since dual therapy is commonly used, in the second study, rats received either saline or combined CLEN + DEX (each at 0.005 or 0.02 mg/kg) one day prior to and on both days of exposure (air or 0.8ppm ozone, 4 hr/day x 2-days). In air-exposed rats CLEN, DEX or CLEN + DEX did not induce lung injury or inflammation, however DEX and CLEN + DEX decreased circulating lymphocytes, spleen and thymus weights, increased free fatty acids (FFA) and produced hyperglycemia and glucose intolerance. Ozone exposure of vehicle-treated rats increased bronchoalveolar lavage fluid protein, albumin, neutrophils, IL-6 and TNF-α. Ozone decreased circulating lymphocytes, increased FFA, and induced hypeerglycemia  and glucose intolerance. Drug treatment did not reverse ozone-induced ventillatory changes, however, lung effects (protein and albumin leakage, inflammation, and IL-6 increase) were exacerbated by CLEN and CLEN + DEX pre-treatment in a dose-dependent manner (CLEN > CLEN + DEX). Systemic effects induced by DEX and CLEN + DEX but not CLEN in air-exposed rats were analogous to and more pronounced than those induced by ozone. These data suggest that adverse air pollution effects might be exacerbated in people receiving LABA or LABA plus glucocorticoids.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Clenbuterol/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Lung/drug effects , Ozone/pharmacology , Adrenergic beta-2 Receptor Agonists/adverse effects , Animals , Clenbuterol/adverse effects , Dexamethasone/adverse effects , Drug Interactions , Fatty Acids/metabolism , Glucocorticoids/adverse effects , Glucose/metabolism , Interleukin-6/metabolism , Lung/metabolism , Lymphocytes/drug effects , Male , Ozone/adverse effects , Rats , Rats, Wistar , Spleen/drug effects , Spleen/metabolism , Thymus Gland/drug effects , Thymus Gland/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
J Toxicol Environ Health A ; 82(2): 86-98, 2019.
Article in English | MEDLINE | ID: mdl-30755101

ABSTRACT

There is growing interest in understanding how maternal diet might affect the sensitivity of offspring to environmental exposures. Previous studies demonstrated that adult rat offspring (approximately 6-months-old) from dams given a high-fat diet (HFD) prior to, during, and after pregnancy displayed elevated pulmonary responses to an acute ozone (O3) exposure. The aim of this study was to examine the influence of maternal and perinatal HFD on pulmonary and metabolic responses to O3 in male and female young-adult offspring (approximately 3-month old). One-month-old F0 female Long-Evans rats commenced HFD (60% kcal from fat) or control diet (CD; 10.5% kcal from fat) and were bred on PND 72. Offspring were maintained on respective HFD or CD until PND 29 when all groups were switched to CD. The 3-months-old female and male offspring (n = 10/group) were exposed to air or 0.8 ppm O3 for 5hr/day for 2 consecutive days. Maternal and perinatal HFD significantly increased body weight and body fat % in offspring regardless of gender. Ozone exposure, but not maternal and perinatal diet, induced hyperglycemia and glucose intolerance in the offspring. Ozone-induced alterations in pulmonary function were exacerbated by maternal and perinatal HFD in both offspring genders. Pulmonary injury/inflammation markers in response to O3 exposure such as bronchoalveolar lavage fluid total protein, lactate dehydrogenase, total cells, and neutrophils were further augmented in offspring (males>females) from dams fed the HFD. Data suggest that maternal and perinatal HFD may enhance the susceptibility of offspring to O3-induced pulmonary injury and that these effects may be sex-specific.


Subject(s)
Diet, High-Fat/adverse effects , Lung/drug effects , Ozone/adverse effects , Age Factors , Animals , Female , Lactation , Lung/metabolism , Male , Ozone/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Long-Evans
4.
Toxicol Sci ; 167(2): 559-572, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30351402

ABSTRACT

Air pollution is a complex mixture of particulate matter and gases linked to adverse clinical outcomes. As such, studying responses to individual pollutants does not account for the potential biological responses resulting from the interaction of various constituents within an ambient air shed. We previously reported that exposure to high levels of the gaseous pollutant acrolein perturbs myocardial synchrony. Here, we examined the effects of repeated, intermittent co-exposure to low levels of concentrated ambient particulates (CAPs) and acrolein on myocardial synchrony and the role of transient receptor potential cation channel A1 (TRPA1), which we previously linked to air pollution-induced sensitization to triggered cardiac arrhythmia. Female B6129 and Trpa1-/- mice (n = 6/group) were exposed to filtered air (FA), CAPs (46 µg/m3 of PM2.5), Acrolein (0.42 ppm), or CAPs+Acrolein for 3 h/day, 2 days/week for 4 weeks. Cardiac ultrasound was conducted to assess cardiac synchronicity and function before and after the first exposure and after the final exposure. Heart rate variability (HRV), an indicator of autonomic tone, was assessed after the final exposure. Strain delay (time between peak strain in adjacent cardiac wall segments), an index of myocardial dyssynchrony, increased by 5-fold after the final CAPs+Acrolein exposure in B6129 mice compared with FA, CAPs, or Acrolein-exposed B6129 mice, and CAPs+Acrolein-exposed Trpa1-/- mice. Only exposure to acrolein alone increased the HRV high frequency domain (5-fold) in B6129 mice, but not in Trpa1-/- mice. Thus, repeated inhalation of pollutant mixtures may increase risk for cardiac responses compared with single or multiple exposures to individual pollutants through TRPA1 activation.


Subject(s)
Acrolein/toxicity , Air Pollutants/toxicity , Arrhythmias, Cardiac/chemically induced , Inhalation Exposure/adverse effects , Myocardium/metabolism , Particulate Matter/toxicity , TRPA1 Cation Channel/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Drug Synergism , Female , Heart Rate/drug effects , Mice , Mice, Knockout , TRPA1 Cation Channel/genetics
5.
Reprod Toxicol ; 83: 63-72, 2019 01.
Article in English | MEDLINE | ID: mdl-30528429

ABSTRACT

Prenatal exposure to ozone has been linked to low birth weight in people and fetal growth restriction in rats. Clinical recommendations suggest use of low dose aspirin to lower risk of preeclampsia and intrauterine growth restriction in high-risk pregnancies, yet its utility in mitigating the postnatal effects of gestational ozone exposure is unknown. The present study investigated the possibility of low dose aspirin to mitigate the effects of ozone exposure during pregnancy. Exposure to ozone impaired uterine arterial flow and induced growth restriction in fetuses of both sexes. Aspirin treatment induced marginal improvements in ozone-induced uterine blood flow impairment. However, this resulted in a protection of fetal weight in dams given aspirin only in early pregnancy. Aspirin administration for the entirety of gestation increased placental weight and reduced antioxidant status, suggesting that prolonged exposure to low dose aspirin may induce placental inefficiency in our model of growth restriction.


Subject(s)
Air Pollutants/toxicity , Aspirin/administration & dosage , Fetal Growth Retardation/prevention & control , Oxidants/toxicity , Ozone/toxicity , Protective Agents/administration & dosage , Uterus/drug effects , Animals , Drug Administration Schedule , Female , Fetal Growth Retardation/chemically induced , Rats, Long-Evans , Regional Blood Flow/drug effects , Ultrasonography, Doppler , Uterine Artery/drug effects , Uterine Artery/physiology , Uterus/blood supply , Uterus/diagnostic imaging
6.
Toxicol Sci ; 166(2): 288-305, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30379318

ABSTRACT

We have shown that acute ozone inhalation activates sympathetic-adrenal-medullary and hypothalamus-pituitary-adrenal stress axes, and adrenalectomy (AD) inhibits ozone-induced lung injury and inflammation. Therefore, we hypothesized that stress hormone receptor agonists (ß2 adrenergic-ß2AR and glucocorticoid-GR) will restore the ozone injury phenotype in AD, while exacerbating effects in sham-surgery (SH) rats. Male Wistar Kyoto rats that underwent SH or AD were treated with vehicles (saline + corn oil) or ß2AR agonist clenbuterol (CLEN, 0.2 mg/kg, i.p.) + GR agonist dexamethasone (DEX, 2 mg/kg, s.c.) for 1 day and immediately prior to each day of exposure to filtered air or ozone (0.8 ppm, 4 h/day for 1 or 2 days). Ozone-induced increases in PenH and peak-expiratory flow were exacerbated in CLEN+DEX-treated SH and AD rats. CLEN+DEX affected breath waveform in all rats. Ozone exposure in vehicle-treated SH rats increased bronchoalveolar lavage fluid (BALF) protein, N-acetyl glucosaminidase activity (macrophage activation), neutrophils, and lung cytokine expression while reducing circulating lymphocyte subpopulations. AD reduced these ozone effects in vehicle-treated rats. At the doses used herein, CLEN+DEX treatment reversed the protection offered by AD and exacerbated most ozone-induced lung effects while diminishing circulating lymphocytes. CLEN+DEX in air-exposed SH rats also induced marked protein leakage and reduced circulating lymphocytes but did not increase BALF neutrophils. In conclusion, circulating stress hormones and their receptors mediate ozone-induced vascular leakage and inflammatory cell trafficking to the lung. Those receiving ß2AR and GR agonists for chronic pulmonary diseases, or with increased circulating stress hormones due to psychosocial stresses, might have altered sensitivity to air pollution.


Subject(s)
Adrenalectomy , Adrenergic beta-2 Receptor Agonists/pharmacology , Lung Injury/drug therapy , Ozone/toxicity , Pneumonia/drug therapy , Receptors, Glucocorticoid/agonists , Animals , Bronchoalveolar Lavage Fluid/chemistry , Clenbuterol/pharmacokinetics , Corticosterone/blood , Cytokines/metabolism , Dexamethasone/pharmacology , Epinephrine/blood , G-Protein-Coupled Receptor Kinase 3/metabolism , Leukocytes/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Lymphocytes/metabolism , Male , Pneumonia/chemically induced , Pneumonia/metabolism , Random Allocation , Rats , Rats, Inbred WKY , Respiratory Function Tests
7.
Inhal Toxicol ; 30(4-5): 169-177, 2018.
Article in English | MEDLINE | ID: mdl-30086657

ABSTRACT

A cell culture exposure system (CCES) was developed to expose cells established at an air-liquid interface (ALI) to volatile chemicals. We characterized the CCES by exposing indigo dye-impregnated filter inserts inside culture wells to 125 ppb ozone (O3) for 1 h at flow rates of 5 and 25 mL/min/well; the reaction of O3 with an indigo dye produces a fluorescent product. A 5-fold increase in fluorescence at 25 mL/min/well versus 5 mL/min/well was observed, suggesting higher flows were more effective. We then exposed primary human bronchial epithelial cells (HBECs) to 0.3 ppm acrolein for 2 h at 3, 5, and 25 mL/min/well and compared our results against well-established in vitro exposure chambers at the U.S. EPA's Human Studies Facility (HSF Chambers). We measured transcript changes of heme oxygenase-1 (HMOX1) and interleukin-8 (IL-8), as well as lactate dehydrogenase (LDH) release, at 0, 1, and 24 h post-exposure. Comparing responses from HSF Chambers to the CCES, differences were only observed at 1 h post-exposure for HMOX1. Here, the HSF Chamber produced a ∼6-fold increase while the CCES at 3 and 5 mL/min/well produced a ∼1.7-fold increase. Operating the CCES at 25 mL/min/well produced a ∼4.5-fold increase; slightly lower than the HSF Chamber. Our biological results, supported by our comparison against the HSF Chambers, agree with our fluorescence results, suggesting that higher flows through the CCES are more effective at delivering volatile chemicals to cells. This new CCES will be deployed to screen the toxicity of volatile chemicals in EPA's chemical inventories.


Subject(s)
Acrolein/toxicity , Bronchi/drug effects , Epithelial Cells/drug effects , Toxicity Tests/methods , Volatile Organic Compounds/toxicity , Biomarkers/metabolism , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Inhalation Exposure , Interleukin-8/genetics , Interleukin-8/metabolism , L-Lactate Dehydrogenase/metabolism , Risk Assessment , Spectrometry, Fluorescence , Volatilization
8.
Inhal Toxicol ; 30(4-5): 178-186, 2018.
Article in English | MEDLINE | ID: mdl-29947284

ABSTRACT

Apelin has cardiopulmonary protective properties that promote vasodilation and maintenance of the endothelial barrier. While reductions in apelin have been identified as a contributor to various lung diseases, including pulmonary edema, its role in the effect of air pollutants has not been examined. Thus, in the current study, we sought to investigate if apelin is a downstream target of inhaled ozone and if such change in expression is related to altered DNA methylation in the lung. Male, Long-Evans rats were exposed to filtered air or 1.0 ppm ozone for 4 h. Ventilation changes were assessed using whole-body plethysmography immediately following exposure, and markers of pulmonary edema and inflammation were assessed in the bronchoaveolar lavage (BAL) fluid. The enzymatic regulators of DNA methylation were measured in the lung, along with methylation and hydroxymethylation of the apelin promoter. Data showed that ozone exposure was associated with increased enhanced pause and protein leakage in the BAL fluid. Ozone exposure reduced DNA cytosine-5-methyltransferase (DNMT) activity and Dnmt3a/b gene expression. Exposure-induced upregulation of proliferating cell nuclear antigen, indicative of DNA damage, repair, and maintenance methylation. Increased methylation and reduced hydroxymethylation were measured on the apelin promoter. These epigenetic modifications accompanied ozone-induced reduction of apelin expression and development of pulmonary edema. In conclusion, epigenetic regulation, specifically increased methylation of the apelin promoter downstream of DNA damage, may lead to reductions in protective signaling of the apelinergic system, contributing to the pulmonary edema observed following the exposure to oxidant air pollution.


Subject(s)
Apelin/genetics , DNA Damage , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Inhalation Exposure , Lung/drug effects , Ozone/toxicity , Pulmonary Edema/chemically induced , Animals , Apelin/metabolism , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Lung/metabolism , Lung/physiopathology , Male , Proliferating Cell Nuclear Antigen/metabolism , Promoter Regions, Genetic , Pulmonary Edema/genetics , Pulmonary Edema/metabolism , Pulmonary Edema/physiopathology , Pulmonary Ventilation/drug effects , Rats, Long-Evans , DNA Methyltransferase 3B
9.
Toxicol Appl Pharmacol ; 347: 104-114, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29627347

ABSTRACT

Numerous studies have demonstrated that short-term air pollution exposure causes cardiac autonomic imbalance as measured by heart rate variability (HRV). We previously showed that a single exposure to acrolein, a ubiquitous gaseous component of air pollution, not only causes autonomic imbalance, but also increases arrhythmia through transient receptor potential A1 (TRPA1) cation channels. Thus, the goal of this study was to characterize acrolein-induced autonomic changes in both normal and TRPA1-knockout mice (KO). Conscious, unrestrained C57BL/6 (WT) and KO mice were exposed to 3 ppm acrolein for 3 h. Separate groups were treated with either atenolol (sympathetic blocker), atropine (parasympathetic blocker) or hexamethonium (autonomic neurotransmission blocker), immediately before exposure. Electrocardiogram (ECG) and heart rate (HR) were recorded continuously before, during and after exposure. Exposure to acrolein produced significant increases in standard deviation of normal-to-normal R-R intervals (SDNN), Root Mean Square of the Successive Differences (RMSSD) and Low-Frequency (LF), as well as an increase in arrhythmia in WT mice. Treatment with atenolol reduced this response while atropine enhanced it, and both drugs blocked the acrolein-induced increase in arrhythmia; hexamethonium had no effect. On the other hand, neither acrolein nor any drug had an effect in the KO mice. Thus, acrolein-induced HRV responses appear to be mediated by a combined parasympathetic and sympathetic modulation. KO mice did not demonstrate any increases in HRV with exposure to acrolein. These data demonstrate that the cardiac effects of irritant air pollutants likely involve disruption of homeostatic balance and altered regulation even in healthy animals.


Subject(s)
Acrolein/toxicity , Air Pollutants/toxicity , Arrhythmias, Cardiac/chemically induced , Heart Rate/drug effects , Heart/drug effects , Heart/innervation , Parasympathetic Nervous System/drug effects , Sympathetic Nervous System/drug effects , TRPA1 Cation Channel/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Electrocardiography , Female , Mice, Inbred C57BL , Mice, Knockout , Parasympathetic Nervous System/metabolism , Parasympathetic Nervous System/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , TRPA1 Cation Channel/deficiency , TRPA1 Cation Channel/genetics , Time Factors
10.
Toxicol Sci ; 163(1): 57-69, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29329427

ABSTRACT

Fish, olive, and coconut oil dietary supplementation have several cardioprotective benefits, but it is not established if they protect against air pollution-induced adverse effects. We hypothesized that these dietary supplements would attenuate ozone-induced systemic and pulmonary effects. Male Wistar Kyoto rats were fed either a normal diet, or a diet supplemented with fish, olive, or coconut oil for 8 weeks. Animals were then exposed to air or ozone (0.8 ppm), 4 h/day for 2 days. Ozone exposure increased phenylephrine-induced aortic vasocontraction, which was completely abolished in rats fed the fish oil diet. Despite this cardioprotective effect, the fish oil diet increased baseline levels of bronchoalveolar lavage fluid (BALF) markers of lung injury and inflammation. Ozone-induced pulmonary injury/inflammation were comparable in rats on normal, coconut oil, and olive oil diets with altered expression of markers in animals fed the fish oil diet. Fish oil, regardless of exposure, led to enlarged, foamy macrophages in the BALF that coincided with decreased pulmonary mRNA expression of cholesterol transporters, cholesterol receptors, and nuclear receptors. Serum microRNA profile was assessed and demonstrated marked depletion of a variety of microRNAs in animals fed the fish oil diet, several of which were of splenic origin. No ozone-specific changes were noted. Collectively, these data indicate that although fish oil offered vascular protection from ozone exposure, it increased pulmonary injury/inflammation and impaired lipid transport mechanisms resulting in foamy macrophage accumulation, demonstrating the need to be cognizant of potential off-target pulmonary effects that might offset the overall benefit of this vasoprotective supplement.


Subject(s)
Aorta/drug effects , Dietary Fats/administration & dosage , Lung Injury/chemically induced , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Ozone/toxicity , Animals , Aorta/physiopathology , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Coconut Oil/administration & dosage , Fish Oils/administration & dosage , Foam Cells/cytology , Inflammation , Lung Injury/immunology , Lung Injury/physiopathology , Male , Muscle, Smooth, Vascular/physiopathology , Olive Oil/administration & dosage , Rats, Inbred WKY
11.
Toxicol Appl Pharmacol ; 339: 161-171, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29247675

ABSTRACT

Recent studies showed that the circulating stress hormones, epinephrine and corticosterone/cortisol, are involved in mediating ozone-induced pulmonary effects through the activation of the sympathetic-adrenal-medullary (SAM) and hypothalamus-pituitary-adrenal (HPA) axes. Hence, we examined the role of adrenergic and glucocorticoid receptor inhibition in ozone-induced pulmonary injury and inflammation. Male 12-week old Wistar-Kyoto rats were pretreated daily for 7days with propranolol (PROP; a non-selective ß adrenergic receptor [AR] antagonist, 10mg/kg, i.p.), mifepristone (MIFE; a glucocorticoid receptor [GR] antagonist, 30mg/kg, s.c.), both drugs (PROP+MIFE), or respective vehicles, and then exposed to air or ozone (0.8ppm), 4h/d for 1 or 2 consecutive days while continuing drug treatment. Ozone exposure alone led to increased peak expiratory flow rates and enhanced pause (Penh); with greater increases by day 2. Receptors blockade minimally affected ventilation in either air- or ozone-exposed rats. Ozone exposure alone was also associated with marked increases in pulmonary vascular leakage, macrophage activation, neutrophilic inflammation and lymphopenia. Notably, PROP, MIFE and PROP+MIFE pretreatments significantly reduced ozone-induced pulmonary vascular leakage; whereas PROP or PROP+MIFE reduced neutrophilic inflammation. PROP also reduced ozone-induced increases in bronchoalveolar lavage fluid (BALF) IL-6 and TNF-α proteins and/or lung Il6 and Tnfα mRNA. MIFE and PROP+MIFE pretreatments reduced ozone-induced increases in BALF N-acetyl glucosaminidase activity, and lymphopenia. We conclude that stress hormones released after ozone exposure modulate pulmonary injury and inflammatory effects through AR and GR in a receptor-specific manner. Individuals with pulmonary diseases receiving AR and GR-related therapy might experience changed sensitivity to air pollution.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Hormone Antagonists/pharmacology , Lung Injury/metabolism , Ozone/toxicity , Receptors, Adrenergic/metabolism , Receptors, Glucocorticoid/metabolism , Adrenergic beta-Antagonists/therapeutic use , Animals , Bronchoalveolar Lavage Fluid , Hormone Antagonists/therapeutic use , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lung Injury/chemically induced , Lung Injury/drug therapy , Male , Mifepristone/pharmacology , Mifepristone/therapeutic use , Rats , Rats, Inbred WKY , Receptors, Glucocorticoid/antagonists & inhibitors
12.
Inhal Toxicol ; 30(11-12): 439-447, 2018.
Article in English | MEDLINE | ID: mdl-30642191

ABSTRACT

Exposure to wildland fire-related particulate matter (PM) causes adverse health outcomes. However, the impacts of specific biomass sources remain unclear. The purpose of this study was to investigate cardiopulmonary responses in rats following exposure to PM extracts collected from peat fire smoke. We hypothesized that peat smoke PM would dose-dependently alter cardiopulmonary function. Male Sprague-Dawley rats (n = 8/group) were exposed to 35 µg (Lo PM) or 350 µg (Hi PM) of peat smoke PM extracts suspended in saline, or saline alone (Vehicle) via oropharyngeal aspiration (OA). Ventilatory expiration times, measured in whole-body plethysmographs immediately after OA, were the lowest in Hi PM exposed subjects at 6 min into recovery (p = .01 vs. Lo PM, p = .08 vs. Vehicle) and resolved shortly afterwards. The next day, we evaluated cardiovascular function in the same subjects via cardiac ultrasound under isoflurane anesthesia. Compared to Vehicle, Hi PM had 45% higher end systolic volume (p = .03) and 17% higher pulmonary artery blood flow acceleration/ejection time ratios, and both endpoints expressed significant increasing linear trends by dose (p = .01 and .02, respectively). In addition, linear trend analyses across doses detected an increase for end diastolic volume and decreases for ejection fraction and fractional shortening. These data suggest that exposure to peat smoke constituents modulates regulation of ventricular ejection and filling volumes, which could be related to altered blood flow in the pulmonary circulation. Moreover, early pulmonary responses to peat smoke PM point to irritant/autonomic mechanisms as potential drivers of later cardiovascular responses.


Subject(s)
Air Pollutants/adverse effects , Heart/drug effects , Lung/drug effects , Smoke/adverse effects , Soil , Animals , Heart/diagnostic imaging , Heart/physiology , Heart Function Tests , Lung/physiology , Male , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Pulmonary Circulation/drug effects , Pulmonary Ventilation/drug effects , Rats, Sprague-Dawley , Ultrasonography , Ventricular Function, Left/drug effects
13.
Environ Health Perspect ; 125(12): 127005, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269335

ABSTRACT

BACKGROUND: Epidemiological studies suggest that increased ozone exposure during gestation may compromise fetal growth. In particular, the implantation stage of pregnancy is considered a key window of susceptibility for this outcome. OBJECTIVES: The main goals of this study were to investigate the effects of short-term ozone inhalation during implantation on fetal growth outcomes and to explore the potential for alterations in uterine arterial flow as a contributing mechanism. METHODS: Pregnant Long-Evans rats were exposed to filtered air, 0.4 ppm ozone, or 0.8 ppm ozone for 4 h/d during implantation, on gestation days (GD) 5 and 6. Tail cuff blood pressure and uterine artery Doppler ultrasound were measured on GD 15, 19, and 21. To assess whether peri-implantation ozone exposure resulted in sustained pulmonary or systemic health effects, bronchoalveolar lavage fluid, serum metabolic and inflammatory end points, and kidney histopathology were evaluated in dams at GD 21. Growth parameters assessed in GD 21 offspring included fetal weight, length, and body composition. RESULTS: Measures of maternal uterine arterial flow, including resistance index and mean velocity, indicated that resistance increased between GD 15 and GD 21 in 0.8 ppm dams but decreased in controls, although absolute values were similar in both groups on GD 21. Ozone-exposed dams also had lower serum glucose and higher free fatty acid concentrations than controls on GD 21. On GD 21, both male and female offspring had lower body weight than controls, and pooled subsets of 3 male and 3 female fetuses from litters exposed to 0.8 ppm ozone had lower lean mass and fat mass than pooled control offspring. CONCLUSIONS: Findings from our experimental model suggest that the offspring of dams exposed to ozone during implantation had reduced growth compared with controls, possibly as a consequence of ozone-induced vascular dysfunction. https://doi.org/10.1289/EHP2019.


Subject(s)
Air Pollutants/adverse effects , Fetal Development/drug effects , Inhalation Exposure , Maternal Exposure , Ozone/adverse effects , Uterine Artery/physiology , Animals , Dose-Response Relationship, Drug , Embryo Implantation , Female , Male , Random Allocation , Rats/growth & development , Rats/physiology , Rats, Long-Evans , Regional Blood Flow , Ultrasonography, Doppler , Uterine Artery/diagnostic imaging
14.
Toxicol Appl Pharmacol ; 329: 249-258, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28623178

ABSTRACT

Ozone-induced systemic effects are modulated through activation of the neuro-hormonal stress response pathway. Adrenal demedullation (DEMED) or bilateral total adrenalectomy (ADREX) inhibits systemic and pulmonary effects of acute ozone exposure. To understand the influence of adrenal-derived stress hormones in mediating ozone-induced lung injury/inflammation, we assessed global gene expression (mRNA sequencing) and selected proteins in lung tissues from male Wistar-Kyoto rats that underwent DEMED, ADREX, or sham surgery (SHAM) prior to their exposure to air or ozone (1ppm), 4h/day for 1 or 2days. Ozone exposure significantly changed the expression of over 2300 genes in lungs of SHAM rats, and these changes were markedly reduced in DEMED and ADREX rats. SHAM surgery but not DEMED or ADREX resulted in activation of multiple ozone-responsive pathways, including glucocorticoid, acute phase response, NRF2, and PI3K-AKT. Predicted targets from sequencing data showed a similarity between transcriptional changes induced by ozone and adrenergic and steroidal modulation of effects in SHAM but not ADREX rats. Ozone-induced increases in lung Il6 in SHAM rats coincided with neutrophilic inflammation, but were diminished in DEMED and ADREX rats. Although ozone exposure in SHAM rats did not significantly alter mRNA expression of Ifnγ and Il-4, the IL-4 protein and ratio of IL-4 to IFNγ (IL-4/IFNγ) proteins increased suggesting a tendency for a Th2 response. This did not occur in ADREX and DEMED rats. We demonstrate that ozone-induced lung injury and neutrophilic inflammation require the presence of circulating epinephrine and corticosterone, which transcriptionally regulates signaling mechanisms involved in this response.


Subject(s)
Adrenal Cortex/metabolism , Adrenal Medulla/metabolism , Corticosterone/blood , Epinephrine/blood , Lung Injury/chemically induced , Lung/metabolism , Ozone , Pneumonia/chemically induced , Stress, Physiological , Adrenal Cortex/surgery , Adrenal Medulla/surgery , Adrenalectomy , Animals , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Inflammation Mediators/metabolism , Lung/pathology , Lung Injury/blood , Lung Injury/genetics , Lung Injury/prevention & control , Male , Neutrophils/metabolism , Oxidative Stress , Pneumonia/blood , Pneumonia/genetics , Pneumonia/pathology , Pneumonia/prevention & control , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred WKY , Signal Transduction , Stress, Physiological/genetics , Transcription, Genetic
15.
Toxicol Sci ; 158(2): 454-464, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28541489

ABSTRACT

Previous studies have demonstrated that exposure to the pulmonary irritant ozone causes myriad systemic metabolic and pulmonary effects attributed to sympathetic and hypothalamus-pituitary-adrenal (HPA) axis activation, which are exacerbated in metabolically impaired models. We examined respiratory and systemic effects following exposure to a sensory irritant acrolein to elucidate the systemic and pulmonary consequences in healthy and diabetic rat models. Male Wistar and Goto Kakizaki (GK) rats, a nonobese type II diabetic Wistar-derived model, were exposed by inhalation to 0, 2, or 4 ppm acrolein, 4 h/d for 1 or 2 days. Exposure at 4 ppm significantly increased pulmonary and nasal inflammation in both strains with vascular protein leakage occurring only in the nose. Acrolein exposure (4 ppm) also caused metabolic impairment by inducing hyperglycemia and glucose intolerance (GK > Wistar). Serum total cholesterol (GKs only), low-density lipoprotein (LDL) cholesterol (both strains), and free fatty acids (GK > Wistar) levels increased; however, no acrolein-induced changes were noted in branched-chain amino acid or insulin levels. These responses corresponded with a significant increase in corticosterone and modest but insignificant increases in adrenaline in both strains, suggesting activation of the HPA axis. Collectively, these data demonstrate that acrolein exposure has a profound effect on nasal and pulmonary inflammation, as well as glucose and lipid metabolism, with the systemic effects exacerbated in the metabolically impaired GKs. These results are similar to ozone-induced responses with the exception of lung protein leakage and ability to alter branched-chain amino acid and insulin levels, suggesting some differences in neuroendocrine regulation of these two air pollutants.


Subject(s)
Acrolein/toxicity , Respiratory System/drug effects , Stress, Physiological/drug effects , Animals , Cholesterol/blood , Corticosterone/blood , Diabetes Mellitus, Experimental/metabolism , Epinephrine/blood , Fatty Acids, Nonesterified/blood , Glucose Intolerance , Hyperglycemia/chemically induced , Inhalation Exposure , Male , Rats , Rats, Wistar
16.
Cardiovasc Toxicol ; 17(2): 97-108, 2017 04.
Article in English | MEDLINE | ID: mdl-26894885

ABSTRACT

Acrolein is an irritating aldehyde generated during combustion of organic compounds. Altered autonomic activity has been documented following acrolein inhalation, possibly impacting myocardial synchrony and function. Given the ubiquitous nature of acrolein in the environment, we sought to better define the immediate and delayed functional cardiac effects of acrolein inhalation in vivo. We hypothesized that acrolein inhalation would increase markers of cardiac mechanical dysfunction, i.e., myocardial dyssynchrony and performance index in mice. Male C57Bl/6J mice were exposed to filtered air (FA) or acrolein (0.3 or 3.0 ppm) for 3 h in whole-body plethysmography chambers (n = 6). Echocardiographic analyses were performed 1 day before exposure and at 1 and 24 h post-exposure. Speckle tracking echocardiography revealed that circumferential strain delay (i.e., dyssynchrony) was increased at 1 and 24 h following exposure to 3.0 ppm, but not 0.3 ppm, when compared to pre-exposure and/or FA exposure. Pulsed wave Doppler of transmitral blood flow revealed that acrolein exposure at 0.3 ppm, but not 3.0 ppm, increased the Tei index of myocardial performance (i.e., decreased global heart performance) at 1 and 24 h post-exposure compared to pre-exposure and/or FA exposure. We conclude that short-term inhalation of acrolein can acutely modify cardiac function in vivo and that echocardiographic evaluation of myocardial synchrony and performance following exposure to other inhaled pollutants could provide broader insight into the health effects of air pollution.


Subject(s)
Acrolein/toxicity , Air Pollutants/toxicity , Inhalation Exposure/adverse effects , Myocardial Contraction/drug effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Function, Left/drug effects , Animals , Cardiotoxicity , Dose-Response Relationship, Drug , Echocardiography, Doppler, Color , Echocardiography, Doppler, Pulsed , Lung/drug effects , Lung/physiopathology , Male , Mice, Inbred C57BL , Plethysmography, Whole Body , Pneumonia/chemically induced , Pneumonia/physiopathology , Pulmonary Ventilation/drug effects , Risk Assessment , Time Factors , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology
17.
Toxicol Appl Pharmacol ; 324: 51-60, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27746315

ABSTRACT

Short-term exposure to ambient air pollution is linked with adverse cardiovascular effects. While previous research focused primarily on particulate matter-induced responses, gaseous air pollutants also contribute to cause short-term cardiovascular effects. Mechanisms underlying such effects have not been adequately described, however the immediate nature of the response suggests involvement of irritant neural activation and downstream autonomic dysfunction. Thus, this study examines the role of TRPA1, an irritant sensory receptor found in the airways, in the cardiac response of mice to acrolein and ozone. Conscious unrestrained wild-type C57BL/6 (WT) and TRPA1 knockout (KO) mice implanted with radiotelemeters were exposed once to 3ppm acrolein, 0.3ppm ozone, or filtered air. Heart rate (HR) and electrocardiogram (ECG) were recorded continuously before, during and after exposure. Analysis of ECG morphology, incidence of arrhythmia and heart rate variability (HRV) were performed. Cardiac mechanical function was assessed using a Langendorff perfusion preparation 24h post-exposure. Acrolein exposure increased HRV independent of HR, as well as incidence of arrhythmia. Acrolein also increased left ventricular developed pressure in WT mice at 24h post-exposure. Ozone did not produce any changes in cardiac function. Neither gas produced ECG effects, changes in HRV, arrhythmogenesis, or mechanical function in KO mice. These data demonstrate that a single exposure to acrolein causes cardiac dysfunction through TRPA1 activation and autonomic imbalance characterized by a shift toward parasympathetic modulation. Furthermore, it is clear from the lack of ozone effects that although gaseous irritants are capable of eliciting immediate cardiac changes, gas concentration and properties play important roles.


Subject(s)
Acrolein/toxicity , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Heart Rate/drug effects , Heart Rate/physiology , Transient Receptor Potential Channels/physiology , Acrolein/administration & dosage , Animals , Arrhythmias, Cardiac/physiopathology , Electrocardiography/methods , Female , Inhalation Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , TRPA1 Cation Channel , Telemetry/methods
18.
Toxicol Appl Pharmacol ; 306: 47-57, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27368153

ABSTRACT

Acute ozone exposure induces a classical stress response with elevated circulating stress hormones along with changes in glucose, protein and lipid metabolism in rats, with similar alterations in ozone-exposed humans. These stress-mediated changes over time have been linked to insulin resistance. We hypothesized that acute ozone-induced stress response and metabolic impairment would persist during subchronic episodic exposure and induce peripheral insulin resistance. Male Wistar Kyoto rats were exposed to air or 0.25ppm or 1.00ppm ozone, 5h/day, 3 consecutive days/week (wk) for 13wks. Pulmonary, metabolic, insulin signaling and stress endpoints were determined immediately after 13wk or following a 1wk recovery period (13wk+1wk recovery). We show that episodic ozone exposure is associated with persistent pulmonary injury and inflammation, fasting hyperglycemia, glucose intolerance, as well as, elevated circulating adrenaline and cholesterol when measured at 13wk, however, these responses were largely reversible following a 1wk recovery. Moreover, the increases noted acutely after ozone exposure in non-esterified fatty acids and branched chain amino acid levels were not apparent following a subchronic exposure. Neither peripheral or tissue specific insulin resistance nor increased hepatic gluconeogenesis were present after subchronic ozone exposure. Instead, long-term ozone exposure lowered circulating insulin and severely impaired glucose-stimulated beta-cell insulin secretion. Thus, our findings in young-adult rats provide potential insights into epidemiological studies that show a positive association between ozone exposures and type 1 diabetes. Ozone-induced beta-cell dysfunction may secondarily contribute to other tissue-specific metabolic alterations following chronic exposure due to impaired regulation of glucose, lipid, and protein metabolism.


Subject(s)
Air Pollutants/toxicity , Ozone/toxicity , Animals , Blood Glucose/analysis , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cholesterol/blood , Epinephrine/blood , Glucose/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/metabolism , Leptin/blood , Liver/drug effects , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats, Inbred WKY
19.
Inhal Toxicol ; 28(7): 313-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27097751

ABSTRACT

Ozone (O3) is known to induce adverse pulmonary and systemic health effects. Importantly, children and older persons are considered at-risk populations for O3-induced dysfunction, yet the mechanisms accounting for the age-related pulmonary responses to O3 are uncertain. In this study, we examined age-related susceptibility to O3 using 1 mo (adolescent), 4 mo (young adult), 12 mo (adult) and 24 mo (senescent) male Brown Norway rats exposed to filtered air or O3 (0.25 and 1.00 ppm), 6 h/day, two days/week for 1 week (acute) or 13 weeks (subchronic). Ventilatory function, assessed by whole-body plethysmography, and bronchoalveolar lavage fluid (BALF) biomarkers of injury and inflammation were used to examine O3-induced pulmonary effects. Relaxation time declined in all ages following the weekly exposures; however, this effect persisted only in the 24 mo rats following a five days recovery, demonstrating an inability to induce adaptation commonly seen with repeated O3 exposures. PenH was increased in all groups with an augmented response in the 4 mo rats following the subchronic O3 exposures. O3 led to increased breathing frequency and minute volume in the 1 and 4 mo animals. Markers of pulmonary permeability were increased in all age groups. Elevations in BALF γ-glutamyl transferase activity and lung inflammation following an acute O3 exposure were noted in only the 1 and 4 mo rats, which likely received an increased effective O3 dose. These data demonstrate that adolescent and young adult animals are more susceptible to changes in ventilation and pulmonary injury/inflammation caused by acute and episodic O3 exposure.


Subject(s)
Air Pollutants/toxicity , Lung/drug effects , Ozone/toxicity , Age Factors , Animals , Bronchoalveolar Lavage Fluid/chemistry , Lung/metabolism , Lung/physiology , Lung Injury , Male , Plethysmography, Whole Body , Rats, Inbred BN , Respiration/drug effects , gamma-Glutamyltransferase/metabolism
20.
Toxicol Sci ; 150(2): 312-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26732886

ABSTRACT

Acute ozone exposure increases circulating stress hormones and induces metabolic alterations in animals. We hypothesized that the increase of adrenal-derived stress hormones is necessary for both ozone-induced metabolic effects and lung injury. Male Wistar-Kyoto rats underwent bilateral adrenal demedullation (DEMED), total bilateral adrenalectomy (ADREX), or sham surgery (SHAM). After a 4 day recovery, rats were exposed to air or ozone (1 ppm), 4 h/day for 1 or 2 days and responses assessed immediately postexposure. Circulating adrenaline levels dropped to nearly zero in DEMED and ADREX rats relative to SHAM. Corticosterone tended to be low in DEMED rats and dropped to nearly zero in ADREX rats. Adrenalectomy in air-exposed rats caused modest changes in metabolites and lung toxicity parameters. Ozone-induced hyperglycemia and glucose intolerance were markedly attenuated in DEMED rats with nearly complete reversal in ADREX rats. Ozone increased circulating epinephrine and corticosterone in SHAM but not in DEMED or ADREX rats. Free fatty acids (P = .15) and branched-chain amino acids increased after ozone exposure in SHAM but not in DEMED or ADREX rats. Lung minute volume was not affected by surgery or ozone but ozone-induced labored breathing was less pronounced in ADREX rats. Ozone-induced increases in lung protein leakage and neutrophilic inflammation were markedly reduced in DEMED and ADREX rats (ADREX > DEMED). Ozone-mediated decreases in circulating white blood cells in SHAM were not observed in DEMED and ADREX rats. We demonstrate that ozone-induced peripheral metabolic effects and lung injury/inflammation are mediated through adrenal-derived stress hormones likely via the activation of stress response pathway.


Subject(s)
Acute Lung Injury/metabolism , Adrenal Glands/metabolism , Hyperglycemia/metabolism , Ozone/toxicity , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Adrenal Glands/surgery , Adrenalectomy , Animals , Corticosterone/blood , Epinephrine/blood , Glucose Intolerance/drug therapy , Hyperglycemia/blood , Hyperglycemia/chemically induced , Inhalation Exposure , Male , Rats, Inbred WKY
SELECTION OF CITATIONS
SEARCH DETAIL
...