1
|
Das S, Rundblad A, Marques IF, Soares AG, Jaddoe VW, Vrijheid M, Nieuwenhuijsen M, Verlouw J, Matthews J, Holven KB, Thoresen M, Timpson NJ, Felix JF, Ulven SM. Air pollution exposure is associated with gene expression in children. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae025. [PMID: 39723337 PMCID: PMC11668970 DOI: 10.1093/eep/dvae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Environmental exposures, including air pollutants and lack of natural spaces, are associated with suboptimal health outcomes in children. We aimed to study the associations between environmental exposures and gene expression in children. Associations of exposure to particulate matter (PM) with diameter <2.5 (PM2.5) and < 10 (PM10) micrometers, nitrogen dioxide, green spaces, and blue space, with whole-blood gene expression were explored in children from the Dutch Generation R Study (n = 172). Analyses were adjusted for age, sex, batch, maternal education, and area socioeconomic status. Follow-up analysis was carried out using lymphoblastoid cell line gene expression in children from the ALSPAC Study (n = 946). Gene set enrichment analysis (GSEA) using hallmark and immune gene sets from the molecular signature database was carried out to identify significantly over-represented gene sets for insights into biological mechanisms Exposure to PM2.5 was associated with expression of 86 genes in discovery analyses in the Generation R Study [false discovery rate (FDR)-adjusted P-value < .25]. Of these, PM2.5 was also associated with GNG11 expression in the same direction in follow-up analysis (FDR-adjusted P-value < .05). The remaining exposures showed much fewer associations in the discovery analyses. GSEA using PM2.5 association results for both cohorts indicated suppression of gene sets related to interferon response and response to bacterial and viral exposure. In conclusion, gene expression analysis performed in two independent cohorts suggests that PM2.5 exposure in children may be involved in interferon and microbial infection responses.
Collapse
Affiliation(s)
- Siddhartha Das
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Amanda Rundblad
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Irene Fontes Marques
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Ana Goncalves Soares
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - Vincent W Jaddoe
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Martine Vrijheid
- Institute for Global Health Barcelona, Dr. Aiguader 88, Barcelona 08003, Spain
- Institute for Global Health Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona 08002, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain
| | - Mark Nieuwenhuijsen
- Institute for Global Health Barcelona, Dr. Aiguader 88, Barcelona 08003, Spain
- Institute for Global Health Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona 08002, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain
| | - Joost Verlouw
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Kirsten B Holven
- National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo 0424, Norway
| | - Magne Thoresen
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - Janine F Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Stine M Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| |
Collapse
|
2
|
Smith LC, Abramova E, Vayas K, Rodriguez J, Gelfand-Titiyevksiy B, Roepke TA, Laskin JD, Gow AJ, Laskin DL. Transcriptional profiling of lung macrophages following ozone exposure in mice identifies signaling pathways regulating immunometabolic activation. Toxicol Sci 2024; 201:103-117. [PMID: 38897669 PMCID: PMC11347782 DOI: 10.1093/toxsci/kfae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
Macrophages play a key role in ozone-induced lung injury by regulating both the initiation and resolution of inflammation. These distinct activities are mediated by pro-inflammatory and anti-inflammatory/proresolution macrophages which sequentially accumulate in injured tissues. Macrophage activation is dependent, in part, on intracellular metabolism. Herein, we used RNA-sequencing (seq) to identify signaling pathways regulating macrophage immunometabolic activity following exposure of mice to ozone (0.8 ppm, 3 h) or air control. Analysis of lung macrophages using an Agilent Seahorse showed that inhalation of ozone increased macrophage glycolytic activity and oxidative phosphorylation at 24 and 72 h post-exposure. An increase in the percentage of macrophages in S phase of the cell cycle was observed 24 h post ozone. RNA-seq revealed significant enrichment of pathways involved in innate immune signaling and cytokine production among differentially expressed genes at both 24 and 72 h after ozone, whereas pathways involved in cell cycle regulation were upregulated at 24 h and intracellular metabolism at 72 h. An interaction network analysis identified tumor suppressor 53 (TP53), E2F family of transcription factors (E2Fs), cyclin-dependent kinase inhibitor 1A (CDKN1a/p21), and cyclin D1 (CCND1) as upstream regulators of cell cycle pathways at 24 h and TP53, nuclear receptor subfamily 4 group a member 1 (NR4A1/Nur77), and estrogen receptor alpha (ESR1/ERα) as central upstream regulators of mitochondrial respiration pathways at 72 h. To assess whether ERα regulates metabolic activity, we used ERα-/- mice. In both air and ozone-exposed mice, loss of ERα resulted in increases in glycolytic capacity and glycolytic reserve in lung macrophages with no effect on mitochondrial oxidative phosphorylation. Taken together, these results highlight the complex interaction between cell cycle, intracellular metabolism, and macrophage activation which may be important in the initiation and resolution of inflammation following ozone exposure.
Collapse
Affiliation(s)
- Ley Cody Smith
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT 06269, United States
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Elena Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Kinal Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Jessica Rodriguez
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Benjamin Gelfand-Titiyevksiy
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, United States
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| |
Collapse
|
3
|
Moos P, Cheminant J, Adhikari U, Venosa A. Transcriptomic-based roadmap to the healthy and ozone-exposed lung. CURRENT OPINION IN TOXICOLOGY 2024; 37:100445. [PMID: 38187954 PMCID: PMC10769160 DOI: 10.1016/j.cotox.2023.100445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The lung is constantly exposed to a myriad of exogenous stressors. Ground-level ozone represents a ubiquitous and extremely reactive anthropogenic toxicant, impacting the health of millions across the globe. While abundant, epidemiological, in vivo, and in vitro data focuses the ozone toxicity in individual cell types (e.g. epithelial type II, alveolar macrophages) or signaling pathways involved in the injury (e.g., akt, glutathione). When appropriately used, bulk and single cell RNA sequencing techniques have the potential to provide complete, and in certain cases unbiased, information of the molecular events taking place in the steady state and injured lung, and even capture the phenotypic diversity of neighboring cells. To this end, this review compiles information pertaining to the latest understanding of lung cell identity and activation in the steady state and ozone exposed lung. In addition, it discusses the value and benefits of multi-omics approaches and other tools developed to predict cell-cell communication and dissect spatial heterogeneity.
Collapse
Affiliation(s)
- Philip Moos
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Jenna Cheminant
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Ujjwal Adhikari
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| |
Collapse
|
4
|
Çelebi Sözener Z, Treffeisen ER, Özdel Öztürk B, Schneider LC. Global warming and implications for epithelial barrier disruption and respiratory and dermatologic allergic diseases. J Allergy Clin Immunol 2023; 152:1033-1046. [PMID: 37689250 PMCID: PMC10864040 DOI: 10.1016/j.jaci.2023.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Global warming has direct and indirect effects, as well as short- and long-term impacts on the respiratory and skin barriers. Extreme temperature directly affects the airway epithelial barrier by disrupting the structural proteins and by triggering airway inflammation and hyperreactivity. It enhances tidal volume and respiratory rate by affecting the thermoregulatory system, causing specific airway resistance and reflex bronchoconstriction via activation of bronchopulmonary vagal C fibers and upregulation of transient receptor potential vanilloid (TRPV) 1 and TRPV4. Heat shock proteins are activated under heat stress and contribute to both epithelial barrier dysfunction and airway inflammation. Accordingly, the frequency and severity of allergic rhinitis and asthma have been increasing. Heat activates TRPV3 in keratinocytes, causing the secretion of inflammatory mediators and eventually pruritus. Exposure to air pollutants alters the expression of genes that control skin barrier integrity and triggers an immune response, increasing the incidence and prevalence of atopic dermatitis. There is evidence that extreme temperature, heavy rains and floods, air pollution, and wildfires increase atopic dermatitis flares. In this narrative review, focused on the last 3 years of literature, we explore the effects of global warming on respiratory and skin barrier and their clinical consequences.
Collapse
Affiliation(s)
- Zeynep Çelebi Sözener
- Division of Immunology and Allergic Diseases, Ankara Bilkent City Hospital, Ankara, Turkey.
| | - Elsa R Treffeisen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Betül Özdel Öztürk
- Division of Immunology and Allergic Diseases, Bolu Izzet Baysal Training and Research Hospital, Bolu, Turkey
| | - Lynda C Schneider
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| |
Collapse
|
5
|
Sundar IK, Duraisamy SK, Choudhary I, Saini Y, Silveyra P. Acute and Repeated Ozone Exposures Differentially Affect Circadian Clock Gene Expression in Mice. Adv Biol (Weinh) 2023; 7:e2300045. [PMID: 37204107 PMCID: PMC10657336 DOI: 10.1002/adbi.202300045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Circadian rhythms have an established role in regulating physiological processes, such as inflammation, immunity, and metabolism. Ozone, a common environmental pollutant with strong oxidative potential, is implicated in lung inflammation/injury in asthmatics. However, whether O3 exposure affects the expression of circadian clock genes in the lungs is not known. In this study, changes in the expression of core clock genes are analyzed in the lungs of adult female and male mice exposed to filtered air (FA) or O3 using qRT-PCR. The findings are confirmed using an existing RNA-sequencing dataset from repeated FA- and O3 -exposed mouse lungs and validated by qRT-PCR. Acute O3 exposure significantly alters the expression of clock genes in the lungs of females (Per1, Cry1, and Rora) and males (Per1). RNA-seq data revealing sex-based differences in clock gene expression in the airway of males (decreased Nr1d1/Rev-erbα) and females (increased Skp1), parenchyma of females and males (decreased Nr1d1 and Fbxl3 and increased Bhlhe40 and Skp1), and alveolar macrophages of males (decreased Arntl/Bmal1, Per1, Per2, Prkab1, and Prkab2) and females (increased Cry2, Per1, Per2, Csnk1d, Csnk1e, Prkab2, and Fbxl3). These findings suggest that lung inflammation caused by O3 exposure affects clock genes which may regulate key signaling pathways.
Collapse
Affiliation(s)
- Isaac Kirubakaran Sundar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Santhosh Kumar Duraisamy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University, School of Public Health, Bloomington, IN, USA
| |
Collapse
|
6
|
Perryman AN, Kim HYH, Payton A, Rager JE, McNell EE, Rebuli ME, Wells H, Almond M, Antinori J, Alexis NE, Porter NA, Jaspers I. Plasma sterols and vitamin D are correlates and predictors of ozone-induced inflammation in the lung: A pilot study. PLoS One 2023; 18:e0285721. [PMID: 37186612 PMCID: PMC10184915 DOI: 10.1371/journal.pone.0285721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Ozone (O3) exposure causes respiratory effects including lung function decrements, increased lung permeability, and airway inflammation. Additionally, baseline metabolic state can predispose individuals to adverse health effects from O3. For this reason, we conducted an exploratory study to examine the effect of O3 exposure on derivatives of cholesterol biosynthesis: sterols, oxysterols, and secosteroid (25-hydroxyvitamin D) not only in the lung, but also in circulation. METHODS We obtained plasma and induced sputum samples from non-asthmatic (n = 12) and asthmatic (n = 12) adult volunteers 6 hours following exposure to 0.4ppm O3 for 2 hours. We quantified the concentrations of 24 cholesterol precursors and derivatives by UPLC-MS and 30 cytokines by ELISA. We use computational analyses including machine learning to determine whether baseline plasma sterols are predictive of O3 responsiveness. RESULTS We observed an overall decrease in the concentration of cholesterol precursors and derivatives (e.g. 27-hydroxycholesterol) and an increase in concentration of autooxidation products (e.g. secosterol-B) in sputum samples. In plasma, we saw a significant increase in the concentration of secosterol-B after O3 exposure. Machine learning algorithms showed that plasma cholesterol was a top predictor of O3 responder status based on decrease in FEV1 (>5%). Further, 25-hydroxyvitamin D was positively associated with lung function in non-asthmatic subjects and with sputum uteroglobin, whereas it was inversely associated with sputum myeloperoxidase and neutrophil counts. CONCLUSION This study highlights alterations in sterol metabolites in the airway and circulation as potential contributors to systemic health outcomes and predictors of pulmonary and inflammatory responsiveness following O3 exposure.
Collapse
Affiliation(s)
- Alexia N. Perryman
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Hye-Young H. Kim
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America
| | - Alexis Payton
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Julia E. Rager
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Erin E. McNell
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Meghan E. Rebuli
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Heather Wells
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Martha Almond
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jamie Antinori
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Neil E. Alexis
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Ned A. Porter
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States of America
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
7
|
Zhao J, Yang Q, Liu Z, Xu P, Tian L, Yan J, Li K, Lin B, Bian L, Xi Z, Liu X. The impact of subchronic ozone exposure on serum metabolome and the mechanisms of abnormal bile acid and arachidonic acid metabolisms in the liver. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114573. [PMID: 36701875 DOI: 10.1016/j.ecoenv.2023.114573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 06/17/2023]
Abstract
Ambient ozone (O3) pollution can induce respiratory and cardiovascular toxicity. However, its impact on the metabolome and the underlying mechanisms remain unclear. This study first investigated the serum metabolite changes in rats exposed to 0.5 ppm O3 for 3 months using untargeted metabolomic approach. Results showed chronic ozone exposure significantly altered the serum levels of 34 metabolites with potential increased risk of digestive, respiratory and cardiovascular disease. Moreover, bile acid synthesis and secretion, and arachidonic acid (AA) metabolism became the most prominent affected metabolic pathways after O3 exposure. Further studies on the mechanisms found that the elevated serum toxic bile acid was not due to the increased biosynthesis in the liver, but the reduced reuptake from the portal vein to hepatocytes owing to repressed Ntcp and Oatp1a1, and the decreased bile acid efflux in hepatocytes as a results of inhibited Bsep, Ostalpha and Ostbeta. Meanwhile, decreased expressions of detoxification enzyme of SULT2A1 and the important regulators of FXR, PXR and HNF4α also contributed to the abnormal bile acids. In addition, O3 promoted the conversion of AA into thromboxane A2 (TXA2) and 20-hydroxyarachidonic acid (20-HETE) in the liver by up-regulation of Fads2, Cyp4a and Tbxas1 which resulting in decreased AA and linoleic acid (LA), and increased thromboxane B2 (TXB2) and 20-HETE in the serum. Furthermore, apparent hepatic chronic inflammation, fibrosis and abnormal function were found in ozone-exposed rats. These results indicated chronic ozone exposure could alter serum metabolites by interfering their metabolism in the liver, and inducing liver injury to aggravate metabolic disorders.
Collapse
Affiliation(s)
- Jiao Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Qingcheng Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Zhiyuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Pengfei Xu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| |
Collapse
|
8
|
Beentjes D, Shears RK, French N, Neill DR, Kadioglu A. Mechanistic Insights into the Impact of Air Pollution on Pneumococcal Pathogenesis and Transmission. Am J Respir Crit Care Med 2022; 206:1070-1080. [PMID: 35649181 PMCID: PMC9704843 DOI: 10.1164/rccm.202112-2668tr] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is the leading cause of pneumonia and bacterial meningitis. A number of recent studies indicate an association between the incidence of pneumococcal disease and exposure to air pollution. Although the epidemiological evidence is substantial, the underlying mechanisms by which the various components of air pollution (particulate matter and gases such as NO2 and SO2) can increase susceptibility to pneumococcal infection are less well understood. In this review, we summarize the various effects air pollution components have on pneumococcal pathogenesis and transmission; exposure to air pollution can enhance host susceptibility to pneumococcal colonization by impairing the mucociliary activity of the airway mucosa, reducing the function and production of key antimicrobial peptides, and upregulating an important pneumococcal adherence factor on respiratory epithelial cells. Air pollutant exposure can also impair the phagocytic killing ability of macrophages, permitting increased replication of S. pneumoniae. In addition, particulate matter has been shown to activate various extra- and intracellular receptors of airway epithelial cells, which may lead to increased proinflammatory cytokine production. This increases recruitment of innate immune cells, including macrophages and neutrophils. The inflammatory response that ensues may result in significant tissue damage, thereby increasing susceptibility to invasive disease, because it allows S. pneumoniae access to the underlying tissues and blood. This review provides an in-depth understanding of the interaction between air pollution and the pneumococcus, which has the potential to aid the development of novel treatments or alternative strategies to prevent disease, especially in areas with high concentrations of air pollution.
Collapse
Affiliation(s)
- Daan Beentjes
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Rebecca K Shears
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Neil French
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Daniel R Neill
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Aras Kadioglu
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
9
|
Tovar A, Smith GJ, Nalesnik MB, Thomas JM, McFadden KM, Harkema JR, Kelada SNP. A Locus on Chromosome 15 Contributes to Acute Ozone-induced Lung Injury in Collaborative Cross Mice. Am J Respir Cell Mol Biol 2022; 67:528-538. [PMID: 35816602 PMCID: PMC9651200 DOI: 10.1165/rcmb.2021-0326oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Ozone (O3)-induced respiratory toxicity varies considerably within the human population and across inbred mouse strains, indicative of gene-environment interactions (GxE). Though previous studies have identified several quantitative trait loci (QTL) and candidate genes underlying responses to O3 exposure, precise mechanisms of susceptibility remain incompletely described. We sought to update our understanding of the genetic architecture of O3 responsiveness using the Collaborative Cross (CC) recombinant inbred mouse panel. We evaluated hallmark O3-induced inflammation and injury phenotypes in 56 CC strains after exposure to filtered air or 2 ppm O3, and performed focused genetic analysis of variation in lung injury, as reflected by protein in lung lavage fluid. Strain-dependent responses to O3 were clear, and QTL mapping revealed two novel loci on Chr (Chromosomes) 10 (peak, 26.2 Mb; 80% confidence interval [CI], 24.6-43.6 Mb) and 15 (peak, 47.1 Mb; 80% CI, 40.2-54.9 Mb), the latter surpassing the 95% significance threshold. At the Chr 15 locus, C57BL/6J and CAST/EiJ founder haplotypes were associated with higher lung injury responses compared with all other CC founder haplotypes. With further statistical analysis and a weight of evidence approach, we delimited the Chr 15 QTL to an ∼2 Mb region containing 21 genes (10 protein coding) and nominated three candidate genes, namely Oxr1, Rspo2, and Angpt1. Gene and protein expression data further supported Oxr1 and Angpt1 as priority candidate genes. In summary, we have shown that O3-induced lung injury is modulated by genetic variation, identified two high priority candidate genes, and demonstrated the value of the CC for detecting GxE.
Collapse
Affiliation(s)
- Adelaide Tovar
- Department of Genetics
- Curriculum in Genetics and Molecular Biology, and
| | - Gregory J. Smith
- Department of Genetics
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Morgan B. Nalesnik
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | | | | | - Jack R. Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Samir N. P. Kelada
- Department of Genetics
- Curriculum in Genetics and Molecular Biology, and
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
10
|
Nguyen J, Deering-Rice CE, Armstrong BS, Massa C, Reilly CA, Venosa A. Parenchymal and Inflammatory Cell Responses to Single and Repeated Ozone Exposure in Healthy and Surfactant Protein-C Mutant Lung. Toxicol Sci 2022; 189:107-123. [PMID: 35866636 DOI: 10.1093/toxsci/kfac074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Mutations in the alveolar epithelial-specific gene encoding for surfactant protein C (SP-C) are linked to pulmonary disease. Ozone (O3) is a ubiquitous pollutant known to exacerbate stress through oxidative injury and inflammation. To comprehend the structural, functional, and immunological impact of single and repeated O3 exposure, SP-CWT and SP-CI73T mice were exposed to air or O3 (0.8 ppm, 3 h, up to x4 consecutive days). O3 was associated with mitochondrial and autophagic activation (PINK1, LC3B, and p62), focal remodeling, and inflammation localized at the terminal bronchiole-to-alveolar junctions. Histological damage was exacerbated by repeated exposure. Single O3 challenge resulted in transient elastin fiber loss, while repeated exposure resulted in marked increases in elastance in SP-CI73T mice. Flow cytometric analysis revealed increases in classical monocyte and monocyte-derived macrophages recruitment in conditions of repeated exposure, which peaked earlier (24 h) in SP-CI73T mice. Immunohistochemical analysis also showed clustering of Arg-1+ and CD206+ activated cells within regions of remodeled lung. Lymphoid cell analysis identified CX3CR1-B220+ B cells accumulating after single (24/72 h). Repeated exposure produces a switch in the phenotype of these B cells CX3CR1+ (72 h) only in SP-CWT mice. SP-CI73T mutants also displayed depletion in NK1.1+NKp46+ NK cells in lung, as well as bone marrow, blood, and spleen. These results illustrate the cumulative impact of O3 on lung structure and function in healthy lung, and aberrant myeloid and lymphoid recruitment in SP-C mutants responding to challenge. Together, this work highlights the significance of modeling environmental exposure across the spectrum of genetic susceptibility, consistent with human disease.
Collapse
Affiliation(s)
- Jacklyn Nguyen
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Brittnie S Armstrong
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Christopher Massa
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah
| |
Collapse
|
11
|
Abstract
Ozone (O3), a criterion air pollutant produced as a product of internal combustion, generates increased inflammation, lung permeability, and airway hyperreactivity when exposed to rodents in laboratory settings. Airway hyperreactivity is defined as an exaggerated acute obstructive response of the airways to one or more nonspecific stimuli. Lung permeability is a measure of barrier functions that separate internal and external environments to limit access of pathogens and other noxious material. By modeling in vivo O3 exposure in rodents, this allows investigators to explore pulmonary and nonpulmonary O3 effects as a means of understanding its impact on human health and lung function. Furthermore, direct effects of O3 on epithelial permeability can be defined using in vitro exposures to airway epithelial cells. This chapter will focus on methods of generating O3 and then exposing rodents and cultured epithelial cells in laboratory settings.
Collapse
|
12
|
Wilson ML, Thysell JA, Baumann KK, Quaranta DV, Liang WS, Erickson MA. Effects of Anesthesia on Ozone-Induced Lung and Systemic Inflammation. Lung 2022; 200:269-275. [PMID: 35199228 PMCID: PMC9038869 DOI: 10.1007/s00408-022-00514-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/18/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE Anesthetics are required for procedures that deliver drugs/biologics, infectious/inflammatory agents, and toxicants directly to the lungs. However, the possible confounding effects of anesthesia on lung inflammation and injury are underreported. Here, we evaluated the effects of two commonly used anesthetic regimens on lung inflammatory responses to ozone in mice. METHODS We tested the effects of brief isoflurane (Iso) or ketamine/xylazine/atipamezole (K/X/A) anesthesia prior to ozone exposure (4 h, 3 ppm) on lung inflammatory responses in mice. Anesthesia regimens modeled those used for non-surgical intratracheal instillations and were administered 1-2 h or 24 h prior to initiating ozone exposure. RESULTS We found that Iso given 1-2 h prior to ozone inhibited inflammatory responses in the lung, and this effect was absent when Iso was given 23-24 h prior to ozone. In contrast, K/X/A given 1-2 h prior to ozone increased lung and systemic inflammation. CONCLUSION Our results highlight the need to comprehensively evaluate anesthesia as an experimental variable in the assessment of lung inflammation in response to ozone and other inflammatory stimuli.
Collapse
Affiliation(s)
- Miranda L Wilson
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jarl A Thysell
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Box 359755, Seattle, WA, 98104, USA
| | - Kristen K Baumann
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA
| | - Danny V Quaranta
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Box 359755, Seattle, WA, 98104, USA
| | - W Sandy Liang
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA
| | - Michelle A Erickson
- Veterans Administration Puget Sound Healthcare System, 1660 S. Columbian Way, S-182, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 325 9th Avenue, Box 359755, Seattle, WA, 98104, USA.
| |
Collapse
|
13
|
Tovar A, Crouse WL, Smith GJ, Thomas JM, Keith BP, McFadden KM, Moran TP, Furey TS, Kelada SNP. Integrative analysis reveals mouse strain-dependent responses to acute ozone exposure associated with airway macrophage transcriptional activity. Am J Physiol Lung Cell Mol Physiol 2022; 322:L33-L49. [PMID: 34755540 PMCID: PMC8721896 DOI: 10.1152/ajplung.00237.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 01/03/2023] Open
Abstract
Acute ozone (O3) exposure is associated with multiple adverse cardiorespiratory outcomes, the severity of which varies across individuals in human populations and inbred mouse strains. However, molecular determinants of response, including susceptibility biomarkers that distinguish who will develop severe injury and inflammation, are not well characterized. We and others have demonstrated that airway macrophages (AMs) are an important resident immune cell type that are functionally and transcriptionally responsive to O3 inhalation. Here, we sought to explore influences of strain, exposure, and strain-by-O3 exposure interactions on AM gene expression and identify transcriptional correlates of O3-induced inflammation and injury across six mouse strains, including five Collaborative Cross (CC) strains. We exposed adult mice of both sexes to filtered air (FA) or 2 ppm O3 for 3 h and measured inflammatory and injury parameters 21 h later. Mice exposed to O3 developed airway neutrophilia and lung injury with strain-dependent severity. In AMs, we identified a common core O3 transcriptional response signature across all strains, as well as a set of genes exhibiting strain-by-O3 exposure interactions. In particular, a prominent gene expression contrast emerged between a low- (CC017/Unc) and high-responding (CC003/Unc) strain, as reflected by cellular inflammation and injury. Further inspection indicated that differences in their baseline gene expression and chromatin accessibility profiles likely contribute to their divergent post-O3 exposure transcriptional responses. Together, these results suggest that aspects of O3-induced respiratory responses are mediated through altered AM transcriptional signatures and further confirm the importance of gene-environment interactions in mediating differential responsiveness to environmental agents.
Collapse
Affiliation(s)
- Adelaide Tovar
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wesley L Crouse
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Gregory J Smith
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph M Thomas
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin P Keith
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathryn M McFadden
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy P Moran
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Terrence S Furey
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Samir N P Kelada
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
14
|
Hathaway QA, Majumder N, Goldsmith WT, Kunovac A, Pinti MV, Harkema JR, Castranova V, Hollander JM, Hussain S. Transcriptomics of single dose and repeated carbon black and ozone inhalation co-exposure highlight progressive pulmonary mitochondrial dysfunction. Part Fibre Toxicol 2021; 18:44. [PMID: 34911549 PMCID: PMC8672524 DOI: 10.1186/s12989-021-00437-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 11/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Air pollution is a complex mixture of particles and gases, yet current regulations are based on single toxicant levels failing to consider potential interactive outcomes of co-exposures. We examined transcriptomic changes after inhalation co-exposure to a particulate and a gaseous component of air pollution and hypothesized that co-exposure would induce significantly greater impairments to mitochondrial bioenergetics. A whole-body inhalation exposure to ultrafine carbon black (CB), and ozone (O3) was performed, and the impact of single and multiple exposures was studied at relevant deposition levels. C57BL/6 mice were exposed to CB (10 mg/m3) and/or O3 (2 ppm) for 3 h (either a single exposure or four independent exposures). RNA was isolated from lungs and mRNA sequencing performed using the Illumina HiSeq. Lung pathology was evaluated by histology and immunohistochemistry. Electron transport chain (ETC) activities, electron flow, hydrogen peroxide production, and ATP content were assessed. RESULTS Compared to individual exposure groups, co-exposure induced significantly greater neutrophils and protein levels in broncho-alveolar lavage fluid as well as a significant increase in mRNA expression of oxidative stress and inflammation related genes. Similarly, a significant increase in hydrogen peroxide production was observed after co-exposure. After single and four exposures, co-exposure revealed a greater number of differentially expressed genes (2251 and 4072, respectively). Of these genes, 1188 (single exposure) and 2061 (four exposures) were uniquely differentially expressed, with 35 mitochondrial ETC mRNA transcripts significantly impacted after four exposures. Both O3 and co-exposure treatment significantly reduced ETC maximal activity for complexes I (- 39.3% and - 36.2%, respectively) and IV (- 55.1% and - 57.1%, respectively). Only co-exposure reduced ATP Synthase activity (- 35.7%) and total ATP content (30%). Further, the ability for ATP Synthase to function is limited by reduced electron flow (- 25%) and translation of subunits, such as ATP5F1, following co-exposure. CONCLUSIONS CB and O3 co-exposure cause unique transcriptomic changes in the lungs that are characterized by functional deficits to mitochondrial bioenergetics. Alterations to ATP Synthase function and mitochondrial electron flow underly a pathological adaptation to lung injury induced by co-exposure.
Collapse
Affiliation(s)
- Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Nairrita Majumder
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, PO Box 9229, Morgantown, WV, 26506-9229, USA
| | - William T Goldsmith
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, PO Box 9229, Morgantown, WV, 26506-9229, USA
| | - Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Mark V Pinti
- Mitochondria, Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Vince Castranova
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Salik Hussain
- Mitochondria, Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA.
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, PO Box 9229, Morgantown, WV, 26506-9229, USA.
| |
Collapse
|
15
|
Kong X, Bennett WC, Jania CM, Chason KD, German Z, Adouli J, Budney SD, Oby BT, van Heusden C, Lazarowski ER, Jaspers I, Randell SH, Hedgespeth BA, Cruse G, Hua X, Schworer SA, Smith GJ, Kelada SN, Tilley SL. Identification of an ATP/P2X7/mast cell pathway mediating ozone-induced bronchial hyperresponsiveness. JCI Insight 2021; 6:e140207. [PMID: 34546976 PMCID: PMC8663556 DOI: 10.1172/jci.insight.140207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/16/2021] [Indexed: 11/20/2022] Open
Abstract
Ozone is a highly reactive environmental pollutant with well-recognized adverse effects on lung health. Bronchial hyperresponsiveness (BHR) is one consequence of ozone exposure, particularly for individuals with underlying lung disease. Our data demonstrated that ozone induced substantial ATP release from human airway epithelia in vitro and into the airways of mice in vivo and that ATP served as a potent inducer of mast cell degranulation and BHR, acting through P2X7 receptors on mast cells. Both mast cell-deficient and P2X7 receptor-deficient (P2X7-/-) mice demonstrated markedly attenuated BHR to ozone. Reconstitution of mast cell-deficient mice with WT mast cells and P2X7-/- mast cells restored ozone-induced BHR. Despite equal numbers of mast cells in reconstituted mouse lungs, mice reconstituted with P2X7-/- mast cells demonstrated significantly less robust BHR than mice reconstituted with WT mast cells. These results support a model where P2X7 on mast cells and other cell types contribute to ozone-induced BHR.
Collapse
Affiliation(s)
- Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - William C Bennett
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Corey M Jania
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Kelly D Chason
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Zachary German
- Marsico Lung Institute and
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jennifer Adouli
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Samuel D Budney
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Brandon T Oby
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Catharina van Heusden
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Eduardo R Lazarowski
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Ilona Jaspers
- Department of Pediatrics and Center for Environmental Medicine, Asthma, and Lung Biology and
| | - Scott H Randell
- Marsico Lung Institute and
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Barry A Hedgespeth
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Glenn Cruse
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Xiaoyang Hua
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Otolaryngology - Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Stephen A Schworer
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
- Division of Allergy and Immunology, Department of Pediatrics, and
| | - Gregory J Smith
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Samir Np Kelada
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephen L Tilley
- Marsico Lung Institute and
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Transcriptomics Underlying Pulmonary Ozone Pathogenesis Regulated by Inflammatory Mediators in Mice. Antioxidants (Basel) 2021; 10:antiox10091489. [PMID: 34573120 PMCID: PMC8466999 DOI: 10.3390/antiox10091489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Ozone (O3) is the predominant oxidant air pollutant associated with airway inflammation, lung dysfunction, and the worsening of preexisting respiratory diseases. We previously demonstrated the injurious roles of pulmonary immune receptors, tumor necrosis factor receptor (TNFR), and toll-like receptor 4, as well as a transcription factor NF-κB, in response to O3 in mice. In the current study, we profiled time-dependent and TNFR- and NF-κB-regulated lung transcriptome changes by subacute O3 to illuminate the underlying molecular events and downstream targets. Mice lacking Tnfr1/Tnfr2 (Tnfr-/-) or Nfkb1 (Nfkb1-/-) were exposed to air or O3. Lung RNAs were prepared for cDNA microarray analyses, and downstream and upstream mechanisms were predicted by pathway analyses of the enriched genes. O3 significantly altered the genes involved in inflammation and redox (24 h), cholesterol biosynthesis and vaso-occlusion (48 h), and cell cycle and DNA repair (48–72 h). Transforming growth factor-β1 was a predicted upstream regulator. Lack of Tnfr suppressed the immune cell proliferation and lipid-related processes and heightened epithelial cell integrity, and Nfkb1 deficiency markedly suppressed lung cell cycle progress during O3 exposure. Common differentially regulated genes by TNFR and NF-κB1 (e.g., Casp8, Il6, and Edn1) were predicted to protect the lungs from cell death, connective tissue injury, and inflammation. Il6-deficient mice were susceptible to O3-induced protein hyperpermeability, indicating its defensive role, while Tnf-deficient mice were resistant to overall lung injury caused by O3. The results elucidated transcriptome dynamics and provided new insights into the molecular mechanisms regulated by TNFR and NF-κB1 in pulmonary subacute O3 pathogenesis.
Collapse
|
17
|
Smith GJ, Tovar A, Kanke M, Wang Y, Deshane JS, Sethupathy P, Kelada SNP. Ozone-induced changes in the murine lung extracellular vesicle small RNA landscape. Physiol Rep 2021; 9:e15054. [PMID: 34558223 PMCID: PMC8461034 DOI: 10.14814/phy2.15054] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 01/08/2023] Open
Abstract
Inhalation exposure to ozone (O3 ) causes adverse respiratory health effects that result from airway inflammation, a complex response mediated in part by changes to airway cellular transcriptional programs. These programs may be regulated by microRNAs transferred between cells (e.g., epithelial cells and macrophages) via extracellular vesicles (EV miRNA). To explore this, we exposed female C57BL/6J mice to filtered air (FA), 1, or 2 ppm O3 by inhalation and collected bronchoalveolar lavage fluid (BALF) 21 h later for markers of airway inflammation, EVs, and EV miRNA. Both concentrations of O3 significantly increased markers of inflammation (neutrophils), injury (total protein), and the number of EV-sized particles in the BALF. Imagestream analysis indicated a substantial portion of particles was positive for canonical EV markers (CD81, CD51), and Siglec-F, a marker of alveolar macrophages. Using high-throughput small RNA sequencing, we identified several differentially expressed (DE) BALF EV miRNAs after 1 ppm (16 DE miRNAs) and 2 ppm (99 DE miRNAs) O3 versus FA exposure. O3 concentration-response patterns in EV miRNA expression were apparent, particularly for miR-2137, miR-126-3p, and miR-351-5p. Integrative analysis of EV miRNA expression and airway cellular mRNA expression identified EV miR-22-3p as a candidate regulator of transcriptomic responses to O3 in airway macrophages. In contrast, we did not identify candidate miRNA regulators of mRNA expression data from conducting airways (predominantly composed of epithelial cells). In summary, our data show that O3 exposure alters EV release and EV miRNA expression, suggesting that further investigation of EVs may provide insight into their effects on airway macrophage function and other mechanisms of O3 -induced respiratory inflammation.
Collapse
Affiliation(s)
- Gregory J Smith
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adelaide Tovar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matt Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Yong Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessy S Deshane
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Samir N P Kelada
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
18
|
Sun N, Niu Y, Zhang R, Huang Y, Wang J, Qiu W, Zhang X, Han Z, Bao J, Zhu H, Duan Y, Kan H. Ozone inhalation induces exacerbation of eosinophilic airway inflammation and Th2-skew immune response in a rat model of AR. Biomed Pharmacother 2021; 137:111261. [PMID: 33482509 DOI: 10.1016/j.biopha.2021.111261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Ozone (O3) exposure elicits allergic rhinitis (AR) exacerbations by mechanisms that remain poorly understood. We used a rat model to investigate the effects of O3 on eosinophilic airway inflammation and Th2-related response. METHODS Sprague-Dawley (SD) rats were sensitized and challenged with ovalbumin (OVA) to make AR models. Three groups of AR rats were exposed respectively to 0.5, 1.0, 2.0 ppm of O3 for 2 h daily over 6 weeks consecutively and studied 24 h later. Normal rats exposed to O3 alone were used as controls. Nasal symptoms and OVA-specific immunoglobulin E (OVA-sIg E) in the serum were evaluated. Inflammatory cells in nasal lavage fluid (NLF) were classified and counted. Cytokines protein levels in NLF were assessed by ELISA. The pathological changes in the nasal mucosa were examined by histology. RESULTS The combination of allergen and repeated O3 exposure in rats induced a significant increase of the number of sneezes, nasal rubs, amount of nasal secretion and OVA-sIgE in the serum, accompanied by enhancement of eosinophils in NLF and nasal mucosa. The increase of interleukin-5 (IL-5), IL-13, Eotaxin and decrease of INF-γ protein levels in NLF were detected in AR rats after O3 inhalation. Hematoxylin and eosin staining showed disordered arrangement of the nasal mucosa epithelium and eosinophilic infiltration in a concentration-dependent manner. CONCLUSIONS O3 inhalation deteriorated symptoms in AR rats, and the possible mechanism is that ozone co-exposure could enhance the expression of Th2 cytokines, eosinophilic airway inflammation dose-dependently. The observation is helpful for us to understand the synergistic effect of O3 in the air pollution and allergen on aggravating allergic rhinitis.
Collapse
Affiliation(s)
- Na Sun
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Yue Niu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| | - Ruxin Zhang
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China.
| | - Yu Huang
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Jinchao Wang
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Wenjia Qiu
- Department of Respiratory Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Xueyan Zhang
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhijin Han
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Jing Bao
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai, China
| | - Huili Zhu
- Department of Respiratory Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Yusen Duan
- Shanghai Environmental Monitoring Center, Shanghai, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Venosa A. Senescence in Pulmonary Fibrosis: Between Aging and Exposure. Front Med (Lausanne) 2020; 7:606462. [PMID: 33282895 PMCID: PMC7689159 DOI: 10.3389/fmed.2020.606462] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
To date, chronic pulmonary pathologies represent the third leading cause of death in the elderly population. Evidence-based projections suggest that >65 (years old) individuals will account for approximately a quarter of the world population before the turn of the century. Genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication, are described as the nine “hallmarks” that govern cellular fitness. Any deviation from the normal pattern initiates a complex cascade of events culminating to a disease state. This blueprint, originally employed to describe aberrant changes in cancer cells, can be also used to describe aging and fibrosis. Pulmonary fibrosis (PF) is the result of a progressive decline in injury resolution processes stemming from endogenous (physiological decline or somatic mutations) or exogenous stress. Environmental, dietary or occupational exposure accelerates the pathogenesis of a senescent phenotype based on (1) window of exposure; (2) dose, duration, recurrence; and (3) cells type being targeted. As the lung ages, the threshold to generate an irreversibly senescent phenotype is lowered. However, we do not have sufficient knowledge to make accurate predictions. In this review, we provide an assessment of the literature that interrogates lung epithelial, mesenchymal, and immune senescence at the intersection of aging, environmental exposure and pulmonary fibrosis.
Collapse
Affiliation(s)
- Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, UT, United States
| |
Collapse
|
20
|
Choudhary I, Vo T, Paudel K, Patial S, Saini Y. Compartment-specific transcriptomics of ozone-exposed murine lungs reveals sex- and cell type-associated perturbations relevant to mucoinflammatory lung diseases. Am J Physiol Lung Cell Mol Physiol 2020; 320:L99-L125. [PMID: 33026818 DOI: 10.1152/ajplung.00381.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ozone is known to cause lung injury, and resident cells of the respiratory tract (i.e., epithelial cells and macrophages) respond to inhaled ozone in a variety of ways that affect their survival, morphology, and functioning. However, a complete understanding of the sex-associated and the cell type-specific gene expression changes in response to ozone exposure is still limited. Through transcriptome profiling, we aimed to analyze gene expression alterations and associated enrichment of biological pathways in three distinct cell type-enriched compartments of ozone-exposed murine lungs. We subchronically exposed adult male and female mice to 0.8 ppm ozone or filtered air. RNA-Seq was performed on airway epithelium-enriched airways, parenchyma, and purified airspace macrophages. Differential gene expression and biological pathway analyses were performed and supported by cellular and immunohistochemical analyses. While a majority of differentially expressed genes (DEGs) in ozone-exposed versus air-exposed groups were common between both sexes, sex-specific DEGs were also identified in all of the three tissue compartments. As compared with ozone-exposed males, ozone-exposed females had significant alterations in gene expression in three compartments. Pathways relevant to cell division and DNA repair were enriched in the ozone-exposed airways, indicating ozone-induced airway injury and repair, which was further supported by immunohistochemical analyses. In addition to cell division and DNA repair pathways, inflammatory pathways were also enriched within the parenchyma, supporting contribution by both epithelial and immune cells. Further, immune response and cytokine-cytokine receptor interactions were enriched in macrophages, indicating ozone-induced macrophage activation. Finally, our analyses also revealed the overall upregulation of mucoinflammation- and mucous cell metaplasia-associated pathways following ozone exposure.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
21
|
Wiegman CH, Li F, Ryffel B, Togbe D, Chung KF. Oxidative Stress in Ozone-Induced Chronic Lung Inflammation and Emphysema: A Facet of Chronic Obstructive Pulmonary Disease. Front Immunol 2020; 11:1957. [PMID: 32983127 PMCID: PMC7492639 DOI: 10.3389/fimmu.2020.01957] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD) caused by cigarette smoke and characterized by chronic inflammation, alveolar destruction (emphysema) and bronchiolar obstruction. Ozone is a gaseous constituent of urban air pollution resulting from photochemical interaction of air pollutants such as nitrogen oxide and organic compounds. While acute exposure to ozone induces airway hyperreactivity and neutrophilic inflammation, chronic ozone exposure in mice causes activation of oxidative pathways resulting in cell death and a chronic bronchial inflammation with emphysema, mimicking cigarette smoke-induced COPD. Therefore, the chronic exposure to ozone has become a model for studying COPD. We review recent data on mechanisms of ozone induced lung disease focusing on pathways causing chronic respiratory epithelial cell injury, cell death, alveolar destruction, and tissue remodeling associated with the development of chronic inflammation and AHR. The initial oxidant insult may result from direct effects on the integrity of membranes and organelles of exposed epithelial cells in the airways causing a stress response with the release of mitochondrial reactive oxygen species (ROS), DNA, and proteases. Mitochondrial ROS and mitochondrial DNA activate NLRP3 inflammasome and the DNA sensors cGAS and STING accelerating cell death pathways including caspases with inflammation enhancing alveolar septa destruction, remodeling, and fibrosis. Inhibitors of mitochondrial ROS, NLRP3 inflammasome, DNA sensor, cell death pathways, and IL-1 represent novel therapeutic targets for chronic airways diseases underlined by oxidative stress.
Collapse
Affiliation(s)
- Coen H. Wiegman
- Section of Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Feng Li
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Dieudonnée Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
- ArtImmune SAS, Orléans, France
| | - Kian Fan Chung
- Section of Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Patial S, Saini Y. Lung macrophages: current understanding of their roles in Ozone-induced lung diseases. Crit Rev Toxicol 2020; 50:310-323. [PMID: 32458707 DOI: 10.1080/10408444.2020.1762537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Through the National Ambient Air Quality Standards (NAAQS), the Clean Air Act of the United States outlines acceptable levels of six different air pollutants considered harmful to humans and the environment. Included in this list is ozone (O3), a highly reactive oxidant gas, respiratory health hazard, and common environmental air pollutant at ground level. The respiratory health effects due to O3 exposure are often associated with molecular and cellular perturbations in the respiratory tract. Periodic review of NAAQS requires comprehensive scientific evaluation of the public health effects of these pollutants, which is formulated through integrated science assessment (ISA) of the most policy-relevant scientific literature. This review focuses on the protective and pathogenic effects of macrophages in the O3-exposed respiratory tract, with emphasis on mouse model-based toxicological studies. Critical findings from 39 studies containing the words O3, macrophage, mice, and lung within the full text were assessed. While some of these studies highlight the presence of disease-relevant pathogenic macrophages in the airspaces, others emphasize a protective role for macrophages in O3-induced lung diseases. Moreover, a comprehensive list of currently known macrophage-specific roles in O3-induced lung diseases is included in this review and the significant knowledge gaps that still exist in the field are outlined. In conclusion, there is a vital need in this field for additional policy-relevant scientific information, including mechanistic studies to further define the role of macrophages in response to O3.
Collapse
Affiliation(s)
- Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|