1
|
Poniedziałek B, Rzymski P, Zarębska-Michaluk D, Flisiak R. Viral respiratory infections and air pollution: A review focused on research in Poland. CHEMOSPHERE 2024; 359:142256. [PMID: 38723686 DOI: 10.1016/j.chemosphere.2024.142256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
The COVID-19 pandemic has reinforced an interest in the relationship between air pollution and respiratory viral infections, indicating that their burden can be increased under poor air quality. This paper reviews the pathways through which air pollutants can enhance susceptibility to such infections and aggravate their clinical course and outcome. It also summarizes the research exploring the links between various viral infections and exposure to solid and gaseous pollution in Poland, a region characterized by poor air quality, especially during a heating season. The majority of studies focused on concentrations of particulate matter (PM; 86.7%); the other pollutants, i.e., BaP, benzene, CO, NOx, O3, and SO2, were studied less often and sometimes only in the context of a particular infection type. Most research concerned COVID-19, showing that elevated levels of PM and NO2 correlated with higher morbidity and mortality, while increased PM2.5 and benzo[a]pyrene levels were related to worse clinical course and outcome in hospitalized, regardless of age and dominant SARS-CoV-2 variant. PM10 and PM2.5 levels were also associated with the incidence of influenza-like illness and, along with NO2 concentrations, with a higher rate of children's hospitalizations due to lower respiratory tract RSV infections. Higher levels of air pollutants also increased hospitalization due to bronchitis (PM, NOx, and O3) and emergency department admission due to viral croup (PM10, PM2.5, NOx, CO, and benzene). Although the conducted studies imply only correlations and have other limitations, as discussed in the present paper, it appears that improving air quality through reducing combustion processes in energy production in Poland should be perceived as a part of multilayered protection measures against respiratory viral infections, decreasing the healthcare costs of COVID-19, lower tract RSV infections, influenza, and other respiratory viral diseases prevalent between autumn and early spring, in addition to other health and climate benefits.
Collapse
Affiliation(s)
- Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | | | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
2
|
Ruggles A, Benakis C. Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut- and Lung-Brain Axis. Cells 2024; 13:803. [PMID: 38786027 PMCID: PMC11119296 DOI: 10.3390/cells13100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Recent evidence indicates that exposure to environmental toxins, both short-term and long-term, can increase the risk of developing neurological disorders, including neurodegenerative diseases (i.e., Alzheimer's disease and other dementias) and acute brain injury (i.e., stroke). For stroke, the latest systematic analysis revealed that exposure to ambient particulate matter is the second most frequent stroke risk after high blood pressure. However, preclinical and clinical stroke investigations on the deleterious consequences of environmental pollutants are scarce. This review examines recent evidence of how environmental toxins, absorbed along the digestive tract or inhaled through the lungs, affect the host cellular response. We particularly address the consequences of environmental toxins on the immune response and the microbiome at the gut and lung barrier sites. Additionally, this review highlights findings showing the potential contribution of environmental toxins to an increased risk of stroke. A better understanding of the biological mechanisms underlying exposure to environmental toxins has the potential to mitigate stroke risk and other neurological disorders.
Collapse
Affiliation(s)
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81337 Munich, Germany;
| |
Collapse
|
3
|
Baltazar-García EA, Vargas-Guerrero B, Lima A, Boavida Ferreira R, Mendoza-Magaña ML, Ramírez-Herrera MA, Baltazar-Díaz TA, Domínguez-Rosales JA, Salazar-Montes AM, Gurrola-Díaz CM. Deflamin Attenuated Lung Tissue Damage in an Ozone-Induced COPD Murine Model by Regulating MMP-9 Catalytic Activity. Int J Mol Sci 2024; 25:5063. [PMID: 38791100 PMCID: PMC11121448 DOI: 10.3390/ijms25105063] [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: 03/28/2024] [Revised: 04/26/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is comprised of histopathological alterations such as pulmonary emphysema and peribronchial fibrosis. Matrix metalloproteinase 9 (MMP-9) is one of the key enzymes involved in both types of tissue remodeling during the development of lung damage. In recent studies, it was demonstrated that deflamin, a protein component extracted from Lupinus albus, markedly inhibits the catalytic activity of MMP-9 in experimental models of colon adenocarcinoma and ulcerative colitis. Therefore, in the present study, we investigated for the first time the biological effect of deflamin in a murine COPD model induced by chronic exposure to ozone. Ozone exposure was carried out in C57BL/6 mice twice a week for six weeks for 3 h each time, and the treated group was orally administered deflamin (20 mg/kg body weight) after each ozone exposure. The histological results showed that deflamin attenuated pulmonary emphysema and peribronchial fibrosis, as evidenced by H&E and Masson's trichrome staining. Furthermore, deflamin administration significantly decreased MMP-9 activity, as assessed by fluorogenic substrate assay and gelatin zymography. Interestingly, bioinformatic analysis reveals a plausible interaction between deflamin and MMP-9. Collectively, our findings demonstrate the therapeutic potential of deflamin in a COPD murine model, and suggest that the attenuation of the development of lung tissue damage occurs by deflamin-regulated MMP-9 catalytic activity.
Collapse
Affiliation(s)
- Elia Ana Baltazar-García
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Belinda Vargas-Guerrero
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Ana Lima
- CECAV—Centro de Ciência Animal e Veterinária, Faculty of Veterinary Medicine, Lusófona University, Campo Grande, 376, 1749-024 Lisbon, Portugal;
| | - Ricardo Boavida Ferreira
- LEAF—Landscape Environment Agriculture and Food, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal;
| | - María Luisa Mendoza-Magaña
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (M.L.M.-M.); (M.A.R.-H.)
| | - Mario Alberto Ramírez-Herrera
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (M.L.M.-M.); (M.A.R.-H.)
| | - Tonatiuh Abimael Baltazar-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - José Alfredo Domínguez-Rosales
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Adriana María Salazar-Montes
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Carmen Magdalena Gurrola-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| |
Collapse
|
4
|
Gutor SS, Salinas RI, Nichols DS, Bazzano JMR, Han W, Gokey JJ, Vasiukov G, West JD, Newcomb DC, Dikalova AE, Richmond BW, Dikalov SI, Blackwell TS, Polosukhin VV. Repetitive sulfur dioxide exposure in mice models post-deployment respiratory syndrome. Am J Physiol Lung Cell Mol Physiol 2024; 326:L539-L550. [PMID: 38410870 PMCID: PMC11380962 DOI: 10.1152/ajplung.00239.2023] [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: 07/28/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Soldiers deployed to Iraq and Afghanistan have a higher prevalence of respiratory symptoms than nondeployed military personnel and some have been shown to have a constellation of findings on lung biopsy termed post-deployment respiratory syndrome (PDRS). Since many of the subjects in this cohort reported exposure to sulfur dioxide (SO2), we developed a model of repetitive exposure to SO2 in mice that phenocopies many aspects of PDRS, including adaptive immune activation, airway wall remodeling, and pulmonary vascular (PV) disease. Although abnormalities in small airways were not sufficient to alter lung mechanics, PV remodeling resulted in the development of pulmonary hypertension and reduced exercise tolerance in SO2-exposed mice. SO2 exposure led to increased formation of isolevuglandins (isoLGs) adducts and superoxide dismutase 2 (SOD2) acetylation in endothelial cells, which were attenuated by treatment with the isoLG scavenger 2-hydroxybenzylamine acetate (2-HOBA). In addition, 2-HOBA treatment or Siruin-3 overexpression in a transgenic mouse model prevented vascular remodeling following SO2 exposure. In summary, our results indicate that repetitive SO2 exposure recapitulates many aspects of PDRS and that oxidative stress appears to mediate PV remodeling in this model. Together, these findings provide new insights regarding the critical mechanisms underlying PDRS.NEW & NOTEWORTHY We developed a mice model of "post-deployment respiratory syndrome" (PDRS), a condition in Veterans with unexplained exertional dyspnea. Our model successfully recapitulates many of the pathological and physiological features of the syndrome, revealing involvement of the ROS-isoLGs-Sirt3-SOD2 pathway in pulmonary vasculature pathology. Our study provides additional knowledge about effects and long-term consequences of sulfur dioxide exposure on the respiratory system, serving as a valuable tool for future PDRS research.
Collapse
Affiliation(s)
- Sergey S Gutor
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Rodrigo I Salinas
- Department of Chemistry, Emory University, Atlanta, Georgia, United States
| | - David S Nichols
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Julia M R Bazzano
- Department of Surgery, Emory University, Atlanta, Georgia, United States
| | - Wei Han
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jason J Gokey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Georgii Vasiukov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - James D West
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Dawn C Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Anna E Dikalova
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Bradley W Richmond
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Sergey I Dikalov
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Timothy S Blackwell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Vasiliy V Polosukhin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
5
|
Skevaki C, Nadeau KC, Rothenberg ME, Alahmad B, Mmbaga BT, Masenga GG, Sampath V, Christiani DC, Haahtela T, Renz H. Impact of climate change on immune responses and barrier defense. J Allergy Clin Immunol 2024; 153:1194-1205. [PMID: 38309598 DOI: 10.1016/j.jaci.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/05/2024]
Abstract
Climate change is not just jeopardizing the health of our planet but is also increasingly affecting our immune health. There is an expanding body of evidence that climate-related exposures such as air pollution, heat, wildfires, extreme weather events, and biodiversity loss significantly disrupt the functioning of the human immune system. These exposures manifest in a broad range of stimuli, including antigens, allergens, heat stress, pollutants, microbiota changes, and other toxic substances. Such exposures pose a direct and indirect threat to our body's primary line of defense, the epithelial barrier, affecting its physical integrity and functional efficacy. Furthermore, these climate-related environmental stressors can hyperstimulate the innate immune system and influence adaptive immunity-notably, in terms of developing and preserving immune tolerance. The loss or failure of immune tolerance can instigate a wide spectrum of noncommunicable diseases such as autoimmune conditions, allergy, respiratory illnesses, metabolic diseases, obesity, and others. As new evidence unfolds, there is a need for additional research in climate change and immunology that covers diverse environments in different global settings and uses modern biologic and epidemiologic tools.
Collapse
Affiliation(s)
- Chrysanthi Skevaki
- Institute of Laboratory Medicine, member of the German Center for Lung Research and the Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Barrak Alahmad
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass; Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Blandina T Mmbaga
- Kilimanjaro Christian Medical University College, Moshi, Tanzania; Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | - Gileard G Masenga
- Kilimanjaro Christian Medical University College, Moshi, Tanzania; Department of Obstetrics and Gynecology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Vanitha Sampath
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass; Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Harald Renz
- Institute of Laboratory Medicine, member of the German Center for Lung Research and the Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany; Kilimanjaro Christian Medical University College, Moshi, Tanzania; Department of Clinical Immunology and Allergology, Laboratory of Immunopathology, Sechenov University, Moscow, Russia.
| |
Collapse
|
6
|
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
|
7
|
Hazlehurst MF, Dearborn LC, Sherris AR, Loftus CT, Adgent MA, Szpiro AA, Ni Y, Day DB, Kaufman JD, Thakur N, Wright RJ, Sathyanarayana S, Carroll KN, Moore PE, Karr CJ. Long-term ozone exposure and lung function in middle childhood. ENVIRONMENTAL RESEARCH 2024; 241:117632. [PMID: 37967704 PMCID: PMC11067856 DOI: 10.1016/j.envres.2023.117632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Ozone (O3) exposure interrupts normal lung development in animal models. Epidemiologic evidence further suggests impairment with higher long-term O3 exposure across early and middle childhood, although study findings to date are mixed and few have investigated vulnerable subgroups. METHODS Participants from the CANDLE study, a pregnancy cohort in Shelby County, TN, in the ECHO-PATHWAYS Consortium, were included if children were born at gestational age >32 weeks, completed a spirometry exam at age 8-9, and had a valid residential history from birth to age 8. We estimated lifetime average ambient O3 exposure based on each child's residential history from birth to age 8, using a validated fine-resolution spatiotemporal model. Spirometry was performed at the age 8-9 year study visit to assess Forced Expiratory Volume in the first second (FEV1) and Forced Vital Capacity (FVC) as primary outcomes; z-scores were calculated using sex-and-age-specific reference equations. Linear regression with robust variance estimators was used to examine associations between O3 exposure and continuous lung function z-scores, adjusted for child, sociodemographic, and home environmental factors. Potential susceptible subgroups were explored using a product term in the regression model to assess effect modification by child sex, history of bronchiolitis in infancy, and allergic sensitization. RESULTS In our sample (n = 648), O3 exposure averaged from birth to age 8 was modest (mean 26.6 [SD 1.1] ppb). No adverse associations between long-term postnatal O3 exposure were observed with either FEV1 (β = 0.12, 95% CI: -0.04, 0.29) or FVC (β = 0.03, 95% CI: -0.13, 0.19). No effect modification by child sex, history of bronchiolitis in infancy, or allergic sensitization was detected for associations with 8-year average O3. CONCLUSIONS In this sample with low O3 concentrations, we did not observe adverse associations between O3 exposures averaged from birth to age 8 and lung function in middle childhood.
Collapse
Affiliation(s)
- Marnie F Hazlehurst
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA.
| | - Logan C Dearborn
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Allison R Sherris
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Margaret A Adgent
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam A Szpiro
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Yu Ni
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA; School of Public Health, College of Health and Human Services, San Diego State University, San Diego, CA, USA
| | - Drew B Day
- Center for Child Health, Behavior, and Development of Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA, USA
| | - Joel D Kaufman
- Departments of Epidemiology and of Environmental and Occupational Health Sciences, School of Public Health, and Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Neeta Thakur
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rosalind J Wright
- Departments of Pediatrics and of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, School of Medicine and Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, and Seattle Children's Research Institute, Seattle, WA, USA
| | - Kecia N Carroll
- Departments of Pediatrics and of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul E Moore
- Division of Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Catherine J Karr
- Department of Pediatrics, School of Medicine and Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
8
|
Pat Y, Ogulur I, Yazici D, Mitamura Y, Cevhertas L, Küçükkase OC, Mesisser SS, Akdis M, Nadeau K, Akdis CA. Effect of altered human exposome on the skin and mucosal epithelial barrier integrity. Tissue Barriers 2023; 11:2133877. [PMID: 36262078 PMCID: PMC10606824 DOI: 10.1080/21688370.2022.2133877] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 10/24/2022] Open
Abstract
Pollution in the world and exposure of humans and nature to toxic substances is continuously worsening at a rapid pace. In the last 60 years, human and domestic animal health has been challenged by continuous exposure to toxic substances and pollutants because of uncontrolled growth, modernization, and industrialization. More than 350,000 new chemicals have been introduced to our lives, mostly without any reasonable control of their health effects and toxicity. A plethora of studies show exposure to these harmful substances during this period with their implications on the skin and mucosal epithelial barrier and increasing prevalence of allergic and autoimmune diseases in the context of the "epithelial barrier hypothesis". Exposure to these substances causes an epithelial injury with peri-epithelial inflammation, microbial dysbiosis and bacterial translocation to sub-epithelial areas, and immune response to dysbiotic bacteria. Here, we provide scientific evidence on the altered human exposome and its impact on epithelial barriers.
Collapse
Affiliation(s)
- Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Medical Microbiology, Faculty of Medicine, Aydin Menderes University, Turkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Turkey
| | - Ozan C Küçükkase
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sanne S Mesisser
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
9
|
De Silva NS, Siewiera J, Alkhoury C, Nader GPF, Nadalin F, de Azevedo K, Couty M, Izquierdo HM, Bhargava A, Conrad C, Maurin M, Antoniadou K, Fouillade C, Londono-Vallejo A, Behrendt R, Bertotti K, Serdjebi C, Lanthiez F, Gallwitz L, Saftig P, Herrero-Fernández B, Saez A, González-Granado JM, van Niel G, Boissonnas A, Piel M, Manel N. Nuclear envelope disruption triggers hallmarks of aging in lung alveolar macrophages. NATURE AGING 2023; 3:1251-1268. [PMID: 37723209 DOI: 10.1038/s43587-023-00488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/16/2023] [Indexed: 09/20/2023]
Abstract
Aging is characterized by gradual immune dysfunction and increased disease risk. Genomic instability is considered central to the aging process, but the underlying mechanisms of DNA damage are insufficiently defined. Cells in confined environments experience forces applied to their nucleus, leading to transient nuclear envelope rupture (NER) and DNA damage. Here, we show that Lamin A/C protects lung alveolar macrophages (AMs) from NER and hallmarks of aging. AMs move within constricted spaces in the lung. Immune-specific ablation of lamin A/C results in selective depletion of AMs and heightened susceptibility to influenza virus-induced pathogenesis and lung cancer growth. Lamin A/C-deficient AMs that persist display constitutive NER marks, DNA damage and p53-dependent senescence. AMs from aged wild-type and from lamin A/C-deficient mice share a lysosomal signature comprising CD63. CD63 is required to limit damaged DNA in macrophages. We propose that NER-induced genomic instability represents a mechanism of aging in AMs.
Collapse
Affiliation(s)
| | - Johan Siewiera
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Chantal Alkhoury
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | | | - Kevin de Azevedo
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Mickaël Couty
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team van Niel, Paris, France
| | | | - Anvita Bhargava
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Cécile Conrad
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | | | - Charles Fouillade
- Institut Curie, PSL Research University, Université Paris-Saclay, CNRS, INSERM, UMR3347, U1021, Orsay, France
| | | | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | | | | | - François Lanthiez
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Lisa Gallwitz
- Biochemical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Paul Saftig
- Biochemical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Beatriz Herrero-Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Angela Saez
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, Spain
| | - José María González-Granado
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12). Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid. CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Guillaume van Niel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team van Niel, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Nicolas Manel
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| |
Collapse
|
10
|
Blaskovic S, Anagnostopoulou P, Borisova E, Schittny D, Donati Y, Haberthür D, Zhou-Suckow Z, Mall MA, Schlepütz CM, Stampanoni M, Barazzone-Argiroffo C, Schittny JC. Airspace Diameter Map-A Quantitative Measurement of All Pulmonary Airspaces to Characterize Structural Lung Diseases. Cells 2023; 12:2375. [PMID: 37830589 PMCID: PMC10571657 DOI: 10.3390/cells12192375] [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: 07/03/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
(1) Background: Stereological estimations significantly contributed to our understanding of lung anatomy and physiology. Taking stereology fully 3-dimensional facilitates the estimation of novel parameters. (2) Methods: We developed a protocol for the analysis of all airspaces of an entire lung. It includes (i) high-resolution synchrotron radiation-based X-ray tomographic microscopy, (ii) image segmentation using the free machine-learning tool Ilastik and ImageJ, and (iii) calculation of the airspace diameter distribution using a diameter map function. To evaluate the new pipeline, lungs from adult mice with cystic fibrosis (CF)-like lung disease (βENaC-transgenic mice) or mice with elastase-induced emphysema were compared to healthy controls. (3) Results: We were able to show the distribution of airspace diameters throughout the entire lung, as well as separately for the conducting airways and the gas exchange area. In the pathobiological context, we observed an irregular widening of parenchymal airspaces in mice with CF-like lung disease and elastase-induced emphysema. Comparable results were obtained when analyzing lungs imaged with μCT, sugges-ting that our pipeline is applicable to different kinds of imaging modalities. (4) Conclusions: We conclude that the airspace diameter map is well suited for a detailed analysis of unevenly distri-buted structural alterations in chronic muco-obstructive lung diseases such as cystic fibrosis and COPD.
Collapse
Affiliation(s)
- Sanja Blaskovic
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (S.B.); (E.B.); (D.S.); (D.H.)
| | | | - Elena Borisova
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (S.B.); (E.B.); (D.S.); (D.H.)
| | - Dominik Schittny
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (S.B.); (E.B.); (D.S.); (D.H.)
| | - Yves Donati
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211 Genève, Switzerland; (Y.D.); (C.B.-A.)
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - David Haberthür
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (S.B.); (E.B.); (D.S.); (D.H.)
| | - Zhe Zhou-Suckow
- Department of Translational Pulmonology, University Hospital Heidelberg, Translational Lung Research Center (TLRC), A Member of German Center for Lung Research (DZL), 69120 Heidelberg, Germany;
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, 10115 Berlin, Germany;
- Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, 10115 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, 10115 Berlin, Germany
| | - Christian M. Schlepütz
- Swiss Light Source, Paul Scherrer Institute, 5232 Villigen, Switzerland; (C.M.S.); (M.S.)
| | - Marco Stampanoni
- Swiss Light Source, Paul Scherrer Institute, 5232 Villigen, Switzerland; (C.M.S.); (M.S.)
- Institute for Biomedical Engineering, University and ETH Zürich, 8093 Zurich, Switzerland
| | - Constance Barazzone-Argiroffo
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211 Genève, Switzerland; (Y.D.); (C.B.-A.)
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Johannes C. Schittny
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (S.B.); (E.B.); (D.S.); (D.H.)
| |
Collapse
|
11
|
Wei Y, Fei L, Wang Y, Zhang M, Chen Z, Guo H, Ge S, Zhu S, Dong P, Yang K, Xie N, Zhao G. A time-series analysis of short-term ambient ozone exposure and hospitalizations from acute myocardial infarction in Henan, China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:93242-93254. [PMID: 37507564 PMCID: PMC10447277 DOI: 10.1007/s11356-023-28456-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023]
Abstract
Epidemiological studies in recent years have identified an association between exposure to air pollutants and acute myocardial infarction (AMI); however, the association between short-term ozone (O3) exposure and AMI hospitalization remains unclear, particularly in developing countries. Therefore, this study collected information on 24,489 AMI patients, including daily air pollutant and meteorological data in Henan, China, between 2016 and 2021. A distributed lagged nonlinear model combined with a Poisson regression model was used to estimate the nonlinear lagged effect of O3 on AMI hospitalizations. We also quantified the effects of O3 on the number of AMI hospitalizations, hospitalization days, and hospitalization costs. The results showed that single- and dual-pollution models of O3 at lag0, lag1, and lag (01-07) were risk factors for AMI hospitalizations, with the most significant effect at lag03 (RR = 1.132, 95% CI:1.083-1.182). Further studies showed that males, younger people (15-64 years), warm seasons, and long sunshine duration were more susceptible to O3. Hospitalizations attributable to O3 during the study period accounted for 11.66% of the total hospitalizations, corresponding to 2856 patients, 33,492 hospital days, and 90 million RMB. Maintaining O3 at 10-130 µg/m3 can prevent hundreds of AMI hospitalizations and save millions of RMB per year in Henan, China. In conclusion, we found that short-term exposure to O3 was significantly associated with an increased risk of hospitalization for AMI in Henan, China, and that further reductions in ambient O3 levels may have substantial health and economic benefits for patients and local healthcare facilities.
Collapse
Affiliation(s)
- Yulong Wei
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Lin Fei
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Yongbin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Min Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, SE5 9NU, UK
| | - Zhigang Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Huige Guo
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Shiqi Ge
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Sen Zhu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Pingshuan Dong
- Department of Cardiology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Kan Yang
- Department of Cardiovascular Surgery, Nanyang Affiliated Hospital of Zhengzhou University, Nanyang Central Hospital, Nanyang, 473009, China
| | - Na Xie
- The Cardiology Department of the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China.
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China.
| |
Collapse
|
12
|
Dearborn LC, Hazlehurst MF, Loftus CT, Szpiro AA, Carroll KN, Moore PE, Adgent MA, Barrett ES, Nguyen RHN, Sathyanarayana S, LeWinn KZ, Bush NR, Kaufman JD, Karr CJ. Role of Air Pollution in the Development of Asthma Among Children with a History of Bronchiolitis in Infancy. Epidemiology 2023; 34:554-564. [PMID: 37042935 PMCID: PMC10563986 DOI: 10.1097/ede.0000000000001613] [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: 01/13/2022] [Accepted: 03/12/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND Infants experiencing bronchiolitis are at increased risk for asthma, but few studies have identified modifiable risk factors. We assessed whether early life air pollution influenced child asthma and wheeze at age 4-6 years among children with a history of bronchiolitis in the first postnatal year. METHODS Children with caregiver-reported physician-diagnosed bronchiolitis were drawn from ECHO-PATHWAYS, a pooled longitudinal cohort from six US cities. We estimated their air pollution exposure from age 1 to 3 years from validated spatiotemporal models of fine particulate matter (PM 2.5 ), nitrogen dioxide (NO 2 ), and ozone (O 3 ). Caregivers reported children's current wheeze and asthma at age 4-6 years. We used modified Poisson regression to estimate relative risks (RR) and 95% confidence intervals (CI), adjusting for child, maternal, and home environmental factors. We assessed effect modification by child sex and maternal history of asthma with interaction models. RESULTS A total of 224 children had caregiver-reported bronchiolitis. Median (interquartile range) 2-year pollutant concentrations were 9.3 (7.8-9.9) µg/m 3 PM 2.5 , 8.5 (6.4-9.9) ppb NO 2 , and 26.6 (25.6-27.7) ppb O 3 . RRs (CI) for current wheeze per 2-ppb higher O 3 were 1.3 (1.0-1.7) and 1.4 (1.1-1.8) for asthma. NO 2 was inversely associated with wheeze and asthma whereas associations with PM 2.5 were null. We observed interactions between NO 2 and PM 2.5 and maternal history of asthma, with lower risks observed among children with a maternal history of asthma. CONCLUSION Our results are consistent with the hypothesis that exposure to modest postnatal O 3 concentrations increases the risk of asthma and wheeze among the vulnerable subpopulation of infants experiencing bronchiolitis.
Collapse
Affiliation(s)
- Logan C Dearborn
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
| | - Marnie F Hazlehurst
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
| | - Christine T Loftus
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
| | - Adam A Szpiro
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA
| | - Kecia N Carroll
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, NY
| | - Paul E Moore
- Division of Allergy, Immunology, and Pulmonology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Margaret A Adgent
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Environmental and Occupational Health Sciences Institute, Rutgers School of Public Health, Piscataway, NJ
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Ruby HN Nguyen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sheela Sathyanarayana
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA
- Seattle Children’s Research Institute, Seattle, WA
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA
| | - Nicole R Bush
- Department of Psychiatry and Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA
| | - Joel D Kaufman
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
- Department of Medicine, School of Medicine, University of Washington; Seattle, WA
| | - Catherine J Karr
- From the Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| |
Collapse
|
13
|
Gutor SS, Salinas RI, Nichols DS, Bazzano JMR, Han W, Gokey JJ, Vasiukov G, West JD, Newcomb DC, Dikalova AE, Richmond BW, Dikalov SI, Blackwell TS, Polosukhin VV. Repetitive Sulfur Dioxide Exposure in Mice Models Post-Deployment Respiratory Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540867. [PMID: 37292948 PMCID: PMC10245576 DOI: 10.1101/2023.05.15.540867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Soldiers deployed to Iraq and Afghanistan have a higher prevalence of respiratory symptoms than non-deployed military personnel and some have been shown to have a constellation of findings on lung biopsy termed post-deployment respiratory syndrome (PDRS). Since many of the deployers in this cohort reported exposure to sulfur dioxide (SO 2 ), we developed a model of repetitive exposure to SO 2 in mice that phenocopies many aspects of PDRS, including adaptive immune activation, airway wall remodeling, and pulmonary vascular disease (PVD). Although abnormalities in small airways were not sufficient to alter lung mechanics, PVD was associated with the development of pulmonary hypertension and reduced exercise tolerance in SO 2 exposed mice. Further, we used pharmacologic and genetic approaches to demonstrate a critical role for oxidative stress and isolevuglandins in mediating PVD in this model. In summary, our results indicate that repetitive SO 2 exposure recapitulates many aspects of PDRS and that oxidative stress may mediate PVD in this model, which may be helpful for future mechanistic studies examining the relationship between inhaled irritants, PVD, and PDRS.
Collapse
|
14
|
Singh P, Aulakh GK. Modulation of low-dose ozone and LPS exposed acute mouse lung inflammation by IF1 mediated ATP hydrolysis inhibitor, BTB06584. Front Immunol 2023; 14:1126574. [PMID: 36993977 PMCID: PMC10040673 DOI: 10.3389/fimmu.2023.1126574] [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: 12/18/2022] [Accepted: 02/16/2023] [Indexed: 03/16/2023] Open
Abstract
Ozone and bacterial lipopolysaccharide (LPS) are common air pollutants that are related to high hospital admissions due to airway hyperreactivity and increased susceptibility to infections, especially in children, older population and individuals with underlying conditions. We modeled acute lung inflammation (ALI) by exposing 6-8 week old male mice to 0.005 ppm ozone for 2 h followed by 50 μg of intranasal LPS. We compared the immunomodulatory effects of single dose pre-treatment with CD61 blocking antibody (clone 2C9.G2), ATPase inhibitor BTB06584 against propranolol as the immune-stimulant and dexamethasone as the immune-suppressant in the ALI model. Ozone and LPS exposure induced lung neutrophil and eosinophil recruitment as measured by respective peroxidase (MPO and EPX) assays, systemic leukopenia, increased levels of lung vascular neutrophil regulatory chemokines such as CXCL5, SDF-1, CXCL13 and a decrease in immune-regulatory chemokines such as BAL IL-10 and CCL27. While CD61 blocking antibody and BTB06584 produced maximum increase in BAL leukocyte counts, protein content and BAL chemokines, these treatments induced moderate increase in lung MPO and EPX content. CD61 blocking antibody induced maximal BAL cell death, a markedly punctate distribution of NK1.1, CX3CR1, CD61. BTB06584 preserved BAL cell viability with cytosolic and membrane distribution of Gr1 and CX3CR1. Propranolol attenuated BAL protein, protected against BAL cell death, induced polarized distribution of NK1.1, CX3CR1 and CD61 but presented with high lung EPX. Dexamethasone induced sparse cell membrane distribution of CX3CR1 and CD61 on BAL cells and displayed very low lung MPO and EPX levels despite highest levels of BAL chemokines. Our study unravels ATPase inhibitor IF1 as a novel drug target for lung injury.
Collapse
|
15
|
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: 3] [Impact Index Per Article: 3.0] [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
|
16
|
Siegel J, Gill N, Ramanathan M, Patadia M. Unified Airway Disease. Otolaryngol Clin North Am 2023; 56:39-53. [DOI: 10.1016/j.otc.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Abstract
Lung epithelium, the lining that covers the inner surface of the respiratory tract, is directly exposed to the environment and thus susceptible to airborne toxins, irritants, and pathogen-induced damages. In adult mammalian lungs, epithelial cells are generally quiescent but can respond rapidly to repair of damaged tissues. Evidence from experimental injury models in rodents and human clinical samples has led to the identification of these regenerative cells, as well as pathological metaplastic states specifically associated with different forms of damages. Here, we provide a compendium of cells and cell states that exist during homeostasis in normal lungs and the lineage relationships between them. Additionally, we discuss various experimental injury models currently being used to probe the cellular sources-both resident and recruited-that contribute to repair, regeneration, and remodeling following acute and chronic injuries. Finally, we discuss certain maladaptive regeneration-associated cell states and their role in disease pathogenesis.
Collapse
Affiliation(s)
- Arvind Konkimalla
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina 27710, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
18
|
Huot-Marchand S, Nascimento M, Culerier E, Bourenane M, Savigny F, Panek C, Serdjebi C, Le Bert M, Quesniaux VFJ, Ryffel B, Broz P, Riteau N, Gombault A, Couillin I. Cigarette smoke-induced gasdermin D activation in bronchoalveolar macrophages and bronchial epithelial cells dependently on NLRP3. Front Immunol 2022; 13:918507. [PMID: 36045672 PMCID: PMC9421433 DOI: 10.3389/fimmu.2022.918507] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic pulmonary inflammation and chronic obstructive pulmonary disease (COPD) are major health issues largely due to air pollution and cigarette smoke (CS) exposure. The role of the innate receptor NLRP3 (nucleotide-binding domain and leucine-rich repeat containing protein 3) orchestrating inflammation through formation of an inflammasome complex in CS-induced inflammation or COPD remains controversial. Using acute and subchronic CS exposure models, we found that Nlrp3-deficient mice or wild-type mice treated with the NLRP3 inhibitor MCC950 presented an important reduction of inflammatory cells recruited into the bronchoalveolar space and of pulmonary inflammation with decreased chemokines and cytokines production, in particular IL-1β demonstrating the key role of NLRP3. Furthermore, mice deficient for Caspase-1/Caspase-11 presented also decreased inflammation parameters, suggesting a role for the NLRP3 inflammasome. Importantly we showed that acute CS-exposure promotes NLRP3-dependent cleavage of gasdermin D in macrophages present in the bronchoalveolar space and in bronchial airway epithelial cells. Finally, Gsdmd-deficiency reduced acute CS-induced lung and bronchoalveolar space inflammation and IL-1β secretion. Thus, we demonstrated in our model that NLRP3 and gasdermin D are key players in CS-induced pulmonary inflammation and IL-1β release potentially through gasdermin D forming-pore and/or pyroptoctic cell death.
Collapse
Affiliation(s)
| | | | - Elodie Culerier
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
| | | | | | - Corinne Panek
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
| | | | - Marc Le Bert
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
| | | | - Bernhard Ryffel
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Riteau
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
- *Correspondence: Isabelle Couillin, ; Nicolas Riteau,
| | | | - Isabelle Couillin
- University of Orleans and CNRS, INEM-UMR7355, Orleans, France
- *Correspondence: Isabelle Couillin, ; Nicolas Riteau,
| |
Collapse
|
19
|
Marzec JM, Nadadur SS. Inflammation resolution in environmental pulmonary health and morbidity. Toxicol Appl Pharmacol 2022; 449:116070. [PMID: 35618031 PMCID: PMC9872158 DOI: 10.1016/j.taap.2022.116070] [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: 02/18/2022] [Revised: 05/04/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023]
Abstract
Inflammation and resolution are dynamic processes comprised of inflammatory activation and neutrophil influx, followed by mediator catabolism and efferocytosis. These critical pathways ensure a return to homeostasis and promote repair. Over the past decade research has shown that diverse mediators play a role in the active process of resolution. Specialized pro-resolving mediators (SPMs), biosynthesized from fatty acids, are released during inflammation to facilitate resolution and are deficient in a variety of lung disorders. Failed resolution results in remodeling and cellular deposition through pro-fibrotic myofibroblast expansion that irreversibly narrows the airways and worsens lung function. Recent studies indicate environmental exposures may perturb and deregulate critical resolution pathways. Environmental xenobiotics induce lung inflammation and generate reactive metabolites that promote oxidative stress, injuring the respiratory mucosa and impairing gas-exchange. This warrants recognition of xenobiotic associated molecular patterns (XAMPs) as new signals in the field of inflammation biology, as many environmental chemicals generate free radicals capable of initiating the inflammatory response. Recent studies suggest that unresolved, persistent inflammation impacts both resolution pathways and endogenous regulatory mediators, compromising lung function, which over time can progress to chronic lung disease. Chronic ozone (O3) exposure overwhelms successful resolution, and in susceptible individuals promotes asthma onset. The industrial contaminant cadmium (Cd) bioaccumulates in the lung to impair resolution, and recurrent inflammation can result in chronic obstructive pulmonary disease (COPD). Persistent particulate matter (PM) exposure increases systemic cardiopulmonary inflammation, which reduces lung function and can exacerbate asthma, COPD, and idiopathic pulmonary fibrosis (IPF). While recurrent inflammation underlies environmentally induced pulmonary morbidity and may drive the disease process, our understanding of inflammation resolution in this context is limited. This review aims to explore inflammation resolution biology and its role in chronic environmental lung disease(s).
Collapse
Affiliation(s)
- Jacqui M Marzec
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Srikanth S Nadadur
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
20
|
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
|
21
|
Celebi Sozener Z, Özbey Yücel Ü, Altiner S, Ozdel Oztürk B, Cerci P, Türk M, Gorgülü Akin B, Akdis M, Yilmaz I, Ozdemir C, Mungan D, Akdis CA. The External Exposome and Allergies: From the Perspective of the Epithelial Barrier Hypothesis. FRONTIERS IN ALLERGY 2022; 3:887672. [PMID: 35873598 PMCID: PMC9304993 DOI: 10.3389/falgy.2022.887672] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/29/2022] [Indexed: 01/04/2023] Open
Abstract
Introduction In the last decades, we have seen a rapid increase in the prevalence of allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, and food allergies. The environmental changes caused by industrialization, urbanization and modernization, including dramatic increases in air pollutants such as particulate matter (PM), diesel exhaust, nitrogen dioxide (NO2), ozone (O3), alarming effects of global warming, change and loss of biodiversity, affect both human health and the entire ecosystem. Objective In this review, we aimed to discuss the effects of the external exposome on epithelial barriers and its relationship with the development of allergic diseases by considering the changes in all stakeholders of the outer exposome together, in the light of the recently proposed epithelial barrier hypothesis. Method To reach current, prominent, and comprehensive studies on the subject, PubMed databases were searched. We included the more resounding articles with reliable and strong results. Results Exposure to altered environmental factors such as increased pollution, microplastics, nanoparticles, tobacco smoke, food emulsifiers, detergents, and household cleaners, and climate change, loss and change in microbial biodiversity, modifications in the consumption of dietary fatty acids, the use of emulsifiers, preservatives and the decrease in the antioxidant content of the widely consumed western diet may disrupt the epithelial barriers of the skin, respiratory and gastrointestinal tracts, making us more vulnerable to exogeneous allergens and microbes. Epithelial cell activation, microbial dysbiosis and bacterial translocation disrupt the immune balance and a chronic Th2 inflammation ensues. Conclusion Dramatic increases in air pollution, worrisome effects of global warming, dysbiosis, changing dietary habits and the complex interactions of all these factors affect the epithelial barriers and local and systemic inflammation. We want to draw attention to the emerging health effects of environmental changes and to motivate the public to influence government policies for the well-being of humans and the nature of the earth and the well-being of future generations.
Collapse
Affiliation(s)
- Zeynep Celebi Sozener
- Clinic of Immunology and Allergic Diseases, Ankara City Hospital, Ankara, Turkey
- *Correspondence: Zeynep Celebi Sozener ; orcid.org/0000-0003-4188-0959
| | - Ümüs Özbey Yücel
- Department of Nutrition and Diet, Ankara University, Ankara, Turkey
| | - Seda Altiner
- Division of Immunology and Allergic Diseases, Department of Internal Medicine, School of Medicine, Ankara University, Ankara, Turkey
| | - Betül Ozdel Oztürk
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, School of Medicine, Ankara University, Ankara, Turkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic Diseases, Eskisehir City Hospital, Eskisehir, Turkey
| | - Murat Türk
- Clinic of Immunology and Allergic Diseases, Kayseri City Hospital, Kayseri, Turkey
| | - Begüm Gorgülü Akin
- Clinic of Immunology and Allergic Diseases, Ankara City Hospital, Ankara, Turkey
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Insu Yilmaz
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, Erciyes University, Kayseri, Turkey
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Dilsad Mungan
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, School of Medicine, Ankara University, Ankara, Turkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Cezmi A. Akdis ; orcid.org/0000-0001-8020-019X
| |
Collapse
|
22
|
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
|
23
|
Wang Y, Zhang Y, Zhang L, Li M, Zhu P, Ji W, Liang R, Qiin L, Wu W, Feng F, Jin Y. [Angiotensin-converting enzyme 2 particapates in ozone-induced lung inflammation and airway remodeling in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:860-867. [PMID: 35790436 DOI: 10.12122/j.issn.1673-4254.2022.06.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the roles of angiotensin-converting enzyme 2 (ACE2) in ozone-induced pulmonary inflammation and airway remodeling in mice. METHODS Sixteen wild-type (WT) C57BL/6J mice and 16 ACE2 knock-out (KO) mice were exposed to either filtered air or ozone (0.8 ppm) for 3 h per day for 5 consecutive days. Masson's staining and HE staining were used to observe lung pathologies. Bronchoalveolar lavage fluid (BALF) was collected and the total cell count was determined. The total proteins and cytokines in BALF were determined by BCA and ELISA method. The transcription levels of airway remodeling-related indicators in the lung tissues were detected using real-time quantitative PCR. The airway resistance of the mice was measured using a small animal ventilator with methacholine stimulation. RESULTS Following ozoneexposure ACE2 KO mice had significantly higher lung pathological scores than WT mice (P < 0.05). Masson staining results showed that compared with ozone-exposed WT mice, ozone-exposed ACE2 KO mice presented with significantly larger area of collagen deposition in the bronchi [(19.62±3.16)% vs (6.49±1.34)%, P < 0.05] and alveoli [(21.63±3.78)% vs (4.44±0.99)%, P < 0.05]. The total cell count and total protein contents in the BALF were both higher in ozone-exposed ACE2 KO mice than in WT mice, but these differences were not statistically significant (P > 0.05). The concentrations of IL-6, IL-1β, TNF-α, CXCL1/KC and MCP-1 in the BALF were all higher in ozone-exposed ACE2 KO mice than in ozone-exposed WT mice, but only the difference in IL-1β was statistically significant (P < 0.05). The transcription levels of MMP-9, MMP-13, TIMP 4, COL1A1, and TGF-β in the lung tissues were all significantly higher in ozone-exposed ACE2 KO mice (P < 0.01). No significant difference was found in airway resistance between ozone-exposed ACE KO mice and WT mice after challenge with 0, 10, 25, or 100 mg/mL of methacholine. CONCLUSION ACE2 participates in ozone-induced lung inflammation and airway remodeling in mice.
Collapse
Affiliation(s)
- Y Wang
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Y Zhang
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - L Zhang
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - M Li
- Department of Toxicology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - P Zhu
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - W Ji
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - R Liang
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - L Qiin
- Institute of Chronic and Non-communicable Disease Prevention and Control, Henan Provincial Center for Disease Control and Prevention, Zhengzhou 450001, China
| | - W Wu
- Department of Occupational and Environmental Health, School of Public Health, Xinxiang Medical University, Xinxiang 453000, China
| | - F Feng
- Department of Toxicology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Y Jin
- Department of epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
24
|
Celebi Sozener Z, Ozdel Ozturk B, Cerci P, Turk M, Gorgulu Akin B, Akdis M, Altiner S, Ozbey U, Ogulur I, Mitamura Y, Yilmaz I, Nadeau K, Ozdemir C, Mungan D, Akdis CA. Epithelial barrier hypothesis: Effect of the external exposome on the microbiome and epithelial barriers in allergic disease. Allergy 2022; 77:1418-1449. [PMID: 35108405 PMCID: PMC9306534 DOI: 10.1111/all.15240] [Citation(s) in RCA: 158] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/11/2022]
Abstract
Environmental exposure plays a major role in the development of allergic diseases. The exposome can be classified into internal (e.g., aging, hormones, and metabolic processes), specific external (e.g., chemical pollutants or lifestyle factors), and general external (e.g., broader socioeconomic and psychological contexts) domains, all of which are interrelated. All the factors we are exposed to, from the moment of conception to death, are part of the external exposome. Several hundreds of thousands of new chemicals have been introduced in modern life without our having a full understanding of their toxic health effects and ways to mitigate these effects. Climate change, air pollution, microplastics, tobacco smoke, changes and loss of biodiversity, alterations in dietary habits, and the microbiome due to modernization, urbanization, and globalization constitute our surrounding environment and external exposome. Some of these factors disrupt the epithelial barriers of the skin and mucosal surfaces, and these disruptions have been linked in the last few decades to the increasing prevalence and severity of allergic and inflammatory diseases such as atopic dermatitis, food allergy, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, and asthma. The epithelial barrier hypothesis provides a mechanistic explanation of how these factors can explain the rapid increase in allergic and autoimmune diseases. In this review, we discuss factors affecting the planet's health in the context of the 'epithelial barrier hypothesis,' including climate change, pollution, changes and loss of biodiversity, and emphasize the changes in the external exposome in the last few decades and their effects on allergic diseases. In addition, the roles of increased dietary fatty acid consumption and environmental substances (detergents, airborne pollen, ozone, microplastics, nanoparticles, and tobacco) affecting epithelial barriers are discussed. Considering the emerging data from recent studies, we suggest stringent governmental regulations, global policy adjustments, patient education, and the establishment of individualized control measures to mitigate environmental threats and decrease allergic disease.
Collapse
Affiliation(s)
| | - Betul Ozdel Ozturk
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic DiseasesEskisehir City HospitalEskisehirTurkey
| | - Murat Turk
- Clinic of Immunology and Allergic DiseasesKayseri City HospitalKayseriTurkey
| | - Begum Gorgulu Akin
- Clinic of Immunology and Allergic DiseasesAnkara City HospitalAnkaraTurkey
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Seda Altiner
- Clinic of Internal Medicine Division of Immunology and Allergic DiseasesKahramanmaras Necip Fazil City HospitalKahramanmarasTurkey
| | - Umus Ozbey
- Department of Nutrition and DietAnkara UniversityAnkaraTurkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Insu Yilmaz
- Department of Chest DiseasesDivision of Immunology and Allergic DiseasesErciyes UniversityKayseriTurkey
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University School of MedicineDivision of Pulmonary and Critical Care MedicineDepartment of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Cevdet Ozdemir
- Institute of Child HealthDepartment of Pediatric Basic SciencesIstanbul UniversityIstanbulTurkey
- Istanbul Faculty of MedicineDepartment of PediatricsDivision of Pediatric Allergy and ImmunologyIstanbul UniversityIstanbulTurkey
| | - Dilsad Mungan
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| |
Collapse
|
25
|
Das S, Saha A, Patowary P, Niri P, Goyary D, Karmakar S, Chattopadhyay P. Assessment of toxicological consequences upon acute inhalation exposure to chemically improvised nonlethal riot control combinational formulation (NCF) containing oleoresin capsicum and skatole. Toxicol Res (Camb) 2021; 10:1129-1143. [PMID: 34956616 DOI: 10.1093/toxres/tfab095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 09/20/2021] [Accepted: 09/28/2021] [Indexed: 11/12/2022] Open
Abstract
Sensory irritation is an acute adverse effect leading to temporary disability posed by riot control agents in various deployable forms are utilized by defense personal in violent mob attacks but their irreversible toxic effects and risk assessment have been a matter of concern. These intimidating risks of available riot control agents have led to exploring the pulmonary toxicity profile of the oil in water emulsion formulation developed for vicious crowd controls containing an irritant oleoresin capsicum, a malodorant (skatole), and a commercial dye, followed by characterization using standard methods. Nonlethal riot control combinational formulation (NCF) has been aimed to be the best possible low-lethal alternative for riot control measures. In this study, 30 min of acute inhalation exposure of NCF was given to Wistar rats and various respiratory parameters like lung dynamics, bronchoalveolar lavage fluid (BALF) cytological assays, pro-inflammatory cytokines estimation, antioxidant activity, collagen accumulation, cytotoxicity, in vivo lung imaging, western blot, histology of lung tissue, etc. were investigated to validate its potentiality and rate of irritation reversibility as nonlethal agents. An exaggerated physiological change like sensory irritation, changes in lung functional variables, increased pro-inflammatory cytokines, etc. were noticed initially without airway obstruction as the expression of nociceptive TRPV1 protein did not alter the physiological regulation of protective proteins like Nrf2 and HO-1 and also no abnormality was found in lung tissue architecture. In conclusion, it can be stated that this formulation can be explored as a nonlethal riot control agent intending to generate discomfort but with early reversibility of sensory irritation and no recurrence of toxicity.
Collapse
Affiliation(s)
- Sanghita Das
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| | - Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| | - Danswrang Goyary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| | - Sanjeev Karmakar
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory, Solmara, Tezpur, Assam 784001, India
| |
Collapse
|
26
|
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: 2.7] [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
|
27
|
Environmental Exposures and Lung Aging: Molecular Mechanisms and Implications for Improving Respiratory Health. Curr Environ Health Rep 2021; 8:281-293. [PMID: 34735706 PMCID: PMC8567983 DOI: 10.1007/s40572-021-00328-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Inhaled environmental exposures cause over 12 million deaths per year worldwide. Despite localized efforts to reduce environmental exposures, tobacco smoking and air pollution remain the urgent public health challenges that are contributing to the growing prevalence of respiratory diseases. The purpose of this review is to describe the mechanisms through which inhaled environmental exposures accelerate lung aging and cause overt lung disease. RECENT FINDINGS Environmental exposures related to fossil fuel and tobacco combustion and occupational exposures related to silica and coal mining generate oxidative stress and inflammation in the lungs. Sustained oxidative stress causes DNA damage, epigenetic instability, mitochondrial dysfunction, and cell cycle arrest in key progenitor cells in the lung. As a result, critical repair mechanisms are impaired, leading to premature destruction of the lung parenchyma. Inhaled environmental exposures accelerate lung aging by injuring the lungs and damaging the cells responsible for wound healing. Interventions that minimize exposure to noxious antigens are critical to improve lung health, and novel research is required to expand our knowledge of therapies that may slow or prevent premature lung aging.
Collapse
|
28
|
Michaeloudes C, Abubakar-Waziri H, Lakhdar R, Raby K, Dixey P, Adcock IM, Mumby S, Bhavsar PK, Chung KF. Molecular mechanisms of oxidative stress in asthma. Mol Aspects Med 2021; 85:101026. [PMID: 34625291 DOI: 10.1016/j.mam.2021.101026] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
The lungs are exposed to reactive oxygen species oxygen (ROS) produced as a result of inhalation of oxygen, as well as smoke and other air pollutants. Cell metabolism and the NADPH oxidases (Nox) generate low levels of intracellular ROS that act as signal transduction mediators by inducing oxidative modifications of histones, enzymes and transcription factors. Redox signalling is also regulated by localised production and sensing of ROS in mitochondria, the endoplasmic reticulum (ER) and inside the nucleus. Intracellular ROS are maintained at low levels through the action of a battery of enzymatic and non-enzymatic antioxidants. Asthma is a heterogeneous airway inflammatory disease with different immune endotypes; these include atopic or non-atopic Th2 type immune response associated with eosinophilia, or a non-Th2 response associated with neutrophilia. Airway remodelling and hyperresponsiveness accompany the inflammatory response in asthma. Over-production of ROS resulting from infiltrating immune cells, particularly eosinophils and neutrophils, and a concomitant impairment of antioxidant responses lead to development of oxidative stress in asthma. Oxidative stress is augmented in severe asthma and during exacerbations, as well as by air pollution and obesity, and causes oxidative damage of tissues promoting airway inflammation and hyperresponsiveness. Furthermore, deregulated Nox activity, mitochondrial dysfunction, ER stress and/or oxidative DNA damage, resulting from exposure to irritants, inflammatory mediators or obesity, may lead to redox-dependent changes in cell signalling. ROS play a central role in airway epithelium-mediated sensing, development of innate and adaptive immune responses, and airway remodelling and hyperresponsiveness. Nonetheless, antioxidant compounds have proven clinically ineffective as therapeutic agents for asthma, partly due to issues with stability and in vivo metabolism of these compounds. The compartmentalised nature of ROS production and sensing, and the role of ROS in homeostatic responses and in the action of corticosteroids and β2-adrenergic receptor agonists, adds another layer of complexity to antioxidant therapy development. Nox inhibitors and mitochondrial-targeted antioxidants are in clinical development for a number of diseases but they have not yet been investigated in asthma. A better understanding of the complex role of ROS in the pathogenesis of asthma will highlight new opportunities for more targeted and effective redox therapies.
Collapse
Affiliation(s)
- Charalambos Michaeloudes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom.
| | - Hisham Abubakar-Waziri
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Ramzi Lakhdar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Katie Raby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Piers Dixey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom; Royal Brompton & Harefield NHS Trust, London, UK
| |
Collapse
|
29
|
Yuksel H, Ocalan M, Yilmaz O. E-Cadherin: An Important Functional Molecule at Respiratory Barrier Between Defence and Dysfunction. Front Physiol 2021; 12:720227. [PMID: 34671272 PMCID: PMC8521047 DOI: 10.3389/fphys.2021.720227] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
While breathing, many microorganisms, harmful environmental particles, allergens, and environmental pollutants enter the human airways. The human respiratory tract is lined with epithelial cells that act as a functional barrier to these harmful factors and provide homeostasis between external and internal environment. Intercellular epithelial junctional proteins play a role in the formation of the barrier. E-cadherin is a calcium-dependent adhesion molecule and one of the most important molecules involved in intercellular epithelial barier formation. E-cadherin is not only physical barrier element but also regulates cell proliferation, differentiation and the immune response to environmental noxious agents through various transcription factors. In this study, we aimed to review the role of E-cadherin in the formation of airway epithelial barier, its status as a result of exposure to various environmental triggers, and respiratory diseases associated with its dysfunction. Moreover, the situations in which its abnormal activation can be noxious would be discussed.
Collapse
Affiliation(s)
- Hasan Yuksel
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Merve Ocalan
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Ozge Yilmaz
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| |
Collapse
|
30
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
31
|
Abstract
Since the industrial revolution, air pollution has become a major problem causing several health problems involving the airways as well as the cardiovascular, reproductive, or neurological system. According to the WHO, about 3.6 million deaths every year are related to inhalation of polluted air, specifically due to pulmonary diseases. Polluted air first encounters the airways, which are a major human defense mechanism to reduce the risk of this aggressor. Air pollution consists of a mixture of potentially harmful compounds such as particulate matter, ozone, carbon monoxide, volatile organic compounds, and heavy metals, each having its own effects on the human body. In the last decades, a lot of research investigating the underlying risks and effects of air pollution and/or its specific compounds on the airways, has been performed, involving both in vivo and in vitro experiments. The goal of this review is to give an overview of the recent data on the effects of air pollution on healthy and diseased airways or models of airway disease, such as asthma or chronic obstructive pulmonary disease. Therefore, we focused on studies involving pollution and airway symptoms and/or damage both in mice and humans.
Collapse
|
32
|
Aulakh GK, Brocos Duda JA, Guerrero Soler CM, Snead E, Singh J. Characterization of low-dose ozone-induced murine acute lung injury. Physiol Rep 2021; 8:e14463. [PMID: 32524776 PMCID: PMC7287414 DOI: 10.14814/phy2.14463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022] Open
Abstract
Ozone is a toxic and highly reactive gaseous oxidizing chemical with well‐documented adverse health effects in humans. On the basis of animal and human data, environmental guidelines and air quality standards recommend a threshold for exposure of no more than 0.063 ppm of ozone (daily concentrations). This research describes a standardized sensitive model of sterile murine lung inflammation induced by exposing mice to acute (0, 4 or 24 hr), yet low, levels of ozone (0.005, 0.05 or 0.5 ppm), one that are below the current recommendations for what is considered a safe or “ambient” ozone concentration for humans. Ozone led to concentration and time‐dependent phlogistic cell death in the bronchoalveolar lavage, lung epithelial damage and hemorrhage. Interestingly, we observed distinct large bright CD11b positive cells in the bronchoalveolar lavage, upregulation of lung vascular and alveolar ATP synthase as well as plasminogen and bronchiolar angiostatin expression in ozone‐exposed mice, platelet and neutrophil accumulation in the lung vasculature and an eotaxin‐2, IL‐16, CXCL5, CXCL12, and CXCL13 dominant inflammatory response leading to lung injury. Using a fluorescent intravital microscopy set up, we quantified ozone‐induced extensive alveolar cellular damage. We observed ozone‐induced actin filament disorganization, perturbed respiratory mechanics, acute suppression of the alveolar reactive oxygen species (ROS) production and mitochondrial potential in ventilated lungs. We present evidence of systemic, as well as pulmonary toxicity, at 40‐fold lower ozone concentrations than previously reported in mice. The findings are important in establishing a sensitive means of quantifying structural and functional lung disorganization following exposure to an aerosolized pollutant, even at levels of ozone exposure previously thought to be safe in humans.
Collapse
Affiliation(s)
- Gurpreet Kaur Aulakh
- Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jessica Andrea Brocos Duda
- Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | - Elisabeth Snead
- Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jaswant Singh
- Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
33
|
Pederson WP, Cyphert-Daly JM, Tighe RM, Que LG, Ledford JG. Genetic variation in surfactant protein-A2 alters responses to ozone. PLoS One 2021; 16:e0247504. [PMID: 33617569 PMCID: PMC7899376 DOI: 10.1371/journal.pone.0247504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Increased exposure to Ozone (O3) is associated with adverse health effects in individuals afflicted with respiratory diseases. Surfactant protein-A (SP-A), encoded by SP-A1 and SP-A2, is the largest protein component in pulmonary surfactant and is functionally impaired by O3-oxidation. OBJECTIVE We used humanized SP-A2 transgenic mice with allelic variation corresponding to a glutamine (Q) to lysine (K) amino acid substitution at position 223 in the lectin domain to determine the impact of this genetic variation in regards to O3 exposure. METHODS Mice were exposed to 2ppm O3 or Filtered Air (FA) for 3 hours and 24 hrs post-challenge pulmonary function tests and other parameters associated with inflammation were assessed in the bronchoalveolar lavage (BAL) fluid and lung tissue. Additionally, mouse tracheal epithelial cells were cultured and TEER measurements recorded for each genotype to determine baseline epithelial integrity. RESULTS Compared to FA, O3 exposure led to significantly increased sensitivity to methacholine challenge in all groups of mice. SP-A2 223Q variant mice were significantly protected from O3-induced AHR compared to SP-A-/- and SP-A2 223K mice. Neutrophilia was observed in all genotypes of mice post O3-exposure, however, SP-A2 223Q mice had a significantly lower percentage of neutrophils compared to SP-A-/- mice. Albumin levels in BAL were unchanged in O3-exposed SP-A2 223Q mice compared to their FA controls, while levels were significantly increased in all other genotypes of O3-exposed mice. SP-A 223Q MTECS has significant higher TEER values than all other genotypes, and WT MTECS has significantly higher TEER than the SP-A KO and SP-A 223K MTECS. SIGNIFICANCE Taken together, our study suggests that expression of a glutamine (Q) as position 223 in SP-A2, as opposed to expression of lysine (K), is more protective in acute exposures to ozone and results in attenuated O3-induced AHR, neutrophilia, and vascular permeability.
Collapse
Affiliation(s)
- William P. Pederson
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Jaime M. Cyphert-Daly
- Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Robert M. Tighe
- Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Loretta G. Que
- Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Julie G. Ledford
- Asthma and Airways Disease Research Center, Tucson, Arizona, United States of America
- Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
34
|
Celebi Sözener Z, Cevhertas L, Nadeau K, Akdis M, Akdis CA. Environmental factors in epithelial barrier dysfunction. J Allergy Clin Immunol 2021; 145:1517-1528. [PMID: 32507229 DOI: 10.1016/j.jaci.2020.04.024] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The main interfaces controlling and attempting to homeostatically balance communications between the host and the environment are the epithelial barriers of the skin, gastrointestinal system, and airways. The epithelial barrier constitutes the first line of physical, chemical, and immunologic defenses and provides a protective wall against environmental factors. Following the industrial revolution in the 19th century, urbanization and socioeconomic development have led to an increase in energy consumption, and waste discharge, leading to increased exposure to air pollution and chemical hazards. Particularly after the 1960s, biological and chemical insults from the surrounding environment-the exposome-have been disrupting the physical integrity of the barrier by degrading the intercellular barrier proteins at tight and adherens junctions, triggering epithelial alarmin cytokine responses such as IL-25, IL-33, and thymic stromal lymphopoietin, and increasing the epithelial barrier permeability. A typical type 2 immune response develops in affected organs in asthma, rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, food allergy, and atopic dermatitis. The aim of this article was to discuss the effects of environmental factors such as protease enzymes of allergens, detergents, tobacco, ozone, particulate matter, diesel exhaust, nanoparticles, and microplastic on the integrity of the epithelial barriers in the context of epithelial barrier hypothesis.
Collapse
Affiliation(s)
- Zeynep Celebi Sözener
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Department of Chest Diseases, Division of Allergy and Immunology, Ankara University School of Medicine, Ankara, Turkey
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Kari Nadeau
- the Naddisy Foundation, Sean Parker Asthma and Allergy Center, Stanford University, Stanford, Calif
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| |
Collapse
|
35
|
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: 7.8] [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
|
36
|
Wagner JG, Barkauskas CE, Vose A, Lewandowski RP, Harkema JR, Tighe RM. Repetitive Ozone Exposures and Evaluation of Pulmonary Inflammation and Remodeling in Diabetic Mouse Strains. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:117009. [PMID: 33253011 PMCID: PMC7703867 DOI: 10.1289/ehp7255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 05/06/2023]
Abstract
BACKGROUND Epidemiological studies support the hypothesis that diabetes alters pulmonary responses to air pollutants like ozone (O 3 ). The mechanism(s) underlying these associations and potential links among diabetes, O 3 , and lung inflammation and remodeling are currently unknown. OBJECTIVES The goal was to determine whether pulmonary responses to repetitive ozone exposures are exacerbated in murine strains that are hyperglycemic and insulin resistant. METHODS Normoglycemic and insulin-sensitive C57BL/6J mice; hyperglycemic, but mildly insulin-resistant, KK mice; and hyperglycemic and markedly insulin-resistant KKAy mice were used for ozone exposure studies. All animals were exposed to filtered air (FA) or repetitive ozone (0.5 ppm O 3 , 4 h/d, for 13 consecutive weekdays). Tissue analysis was performed 24 h following the final exposure. This analysis included bronchoalveolar lavage (BAL) for cell and fluid analysis, and tissue for pathology, immunohistology, mRNA, and hydroxyproline. RESULTS Following repetitive O 3 exposure, higher bronchoalveolar lavage fluid inflammatory cells were observed in all mice (KKAy > KK > C 57 BL / 6 ), with a notable influx of neutrophils and eosinophils in KK and KKAy mice. Although the lungs of O 3 -exposed C57BL/6J and KK mice had minimal centriacinar histological changes without fibrosis, the lungs of O 3 -exposed KKAy mice contained marked epithelial hyperplasia in proximal alveolar ducts and adjacent alveoli with associated centriacinar fibrosis. Fibrosis in O 3 -exposed KKAy lungs was confirmed with immunohistochemistry, tissue hydroxyproline content, and tissue mRNA expression of fibrosis-associated genes (Ccl11, Il13, and Mmp12). Immunofluorescence staining and confocal microscopy revealed alterations in the structure and composition of the airway and alveolar epithelium in regions of fibrosis. DISCUSSION Our results demonstrate that in diabetic animal strains repetitive ambient ozone exposure led to early and exaggerated pulmonary inflammation and remodeling. Changes in distal and interstitial airspaces and the activation of Th2 inflammatory and profibrotic pathways in experimental animals provide a preliminary, mechanistic framework to support the emerging epidemiological associations among air pollution, diabetes, and lung disease. https://doi.org/10.1289/EHP7255.
Collapse
Affiliation(s)
- James G. Wagner
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | | | - Aaron Vose
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Ryan P. Lewandowski
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Jack R. Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Robert M. Tighe
- Department of Medicine, Duke University, Durham, North Carolina, USA
| |
Collapse
|
37
|
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: 112] [Impact Index Per Article: 28.0] [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
|
38
|
Watson JK, Sanders P, Dunmore R, Rosignoli G, Julé Y, Rawlins EL, Mustelin T, May R, Clarke D, Finch DK. Distal lung epithelial progenitor cell function declines with age. Sci Rep 2020; 10:10490. [PMID: 32591591 PMCID: PMC7319976 DOI: 10.1038/s41598-020-66966-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/06/2020] [Indexed: 01/22/2023] Open
Abstract
Tissue stem cell exhaustion is a key hallmark of aging, and in this study, we characterised its manifestation in the distal lung. We compared the lungs of 3- and 22-month old mice. We examined the gross morphological changes in these lungs, the density and function of epithelial progenitor populations and the epithelial gene expression profile. Bronchioles became smaller in their cross-sectional area and diameter. Using long-term EdU incorporation analysis and immunohistochemistry, we found that bronchiolar cell density remained stable with aging, but inferred rates of bronchiolar club progenitor cell self-renewal and differentiation were reduced, indicative of an overall slowdown in cellular turnover. Alveolar Type II progenitor cell density and self-renewal were maintained per unit tissue area with aging, but rates of inferred differentiation into Type I cells, and indeed overall density of Type I cells was reduced. Microarray analysis revealed age-related changes in multiple genes, including some with roles in proliferation and differentiation, and in IGF and TGFβ signalling pathways. By characterising how lung stem cell dynamics change with aging, this study will elucidate how they contribute to age-related loss of pulmonary function, and pathogenesis of common age-related pulmonary diseases.
Collapse
Affiliation(s)
- Julie K Watson
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Philip Sanders
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Rebecca Dunmore
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Guglielmo Rosignoli
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Yvon Julé
- Biocellvia, 10 rue Grignan, Marseille, 13001, France
| | - Emma L Rawlins
- Gurdon Institute, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1QN, UK
| | - Tomas Mustelin
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Richard May
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Deborah Clarke
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K Finch
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
39
|
miRNAs in Lung Development and Diseases. Int J Mol Sci 2020; 21:ijms21082765. [PMID: 32316149 PMCID: PMC7216056 DOI: 10.3390/ijms21082765] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
The development of the lung involves a diverse group of molecules that regulate cellular processes, organ formation, and maturation. The various stages of lung development are marked by accumulation of small RNAs that promote or repress underlying mechanisms, depending on the physiological environment in utero and postnatally. To some extent, the pathogenesis of various lung diseases is regulated by small RNAs. In this review, we discussed miRNAs regulation of lung development and diseases, that is, COPD, asthma, pulmonary fibrosis, and pulmonary arterial hypertension, and also highlighted possible connotations for human lung health.
Collapse
|
40
|
Michaudel C, Bataille F, Maillet I, Fauconnier L, Colas C, Sokol H, Straube M, Couturier-Maillard A, Dumoutier L, van Snick J, Quesniaux VF, Togbe D, Ryffel B. Ozone-Induced Aryl Hydrocarbon Receptor Activation Controls Lung Inflammation via Interleukin-22 Modulation. Front Immunol 2020; 11:144. [PMID: 32161582 PMCID: PMC7053361 DOI: 10.3389/fimmu.2020.00144] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/20/2020] [Indexed: 01/09/2023] Open
Abstract
Airborne ozone exposure causes severe lung injury and inflammation. The aryl hydrocarbon Receptor (AhR) (1), activated in pollutant-induced inflammation, is critical for cytokine production, especially IL-22 and IL-17A. The role of AhR in ozone-induced lung inflammation is unknown. We report here that chronic ozone exposure activates AhR with increased tryptophan and lipoxin A4 production in mice. AhR-/- mice show increased lung inflammation, airway hyperresponsiveness, and tissue remodeling with an increased recruitment of IL-17A and IL-22-expressing cells in comparison to control mice. IL-17A- and IL-22-neutralizing antibodies attenuate lung inflammation in AhR-/- and control mice. Enhanced lung inflammation and recruitment of ILC3, ILC2, and T cells were observed after T cell-specific AhR depletion using the AhRCD4cre-deficient mice. Together, the data demonstrate that ozone exposure activates AhR, which controls lung inflammation, airway hyperresponsiveness, and tissue remodeling via the reduction of IL-22 expression.
Collapse
Affiliation(s)
- Chloé Michaudel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Florent Bataille
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Isabelle Maillet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| | | | - Cyril Colas
- University of Orléans, CNRS ICOA, UMR7311, F-45067, Orléans, France
- CNRS, CBM, UPR4301, University Orléans, Orléans, France
| | - Harry Sokol
- Avenir Team Gut Microbiota and Immunity, Equipe de Recherche Labélisée 1157, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Marjolène Straube
- Avenir Team Gut Microbiota and Immunity, Equipe de Recherche Labélisée 1157, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Aurélie Couturier-Maillard
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Laure Dumoutier
- Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques van Snick
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - Valérie F. Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Dieudonnée Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
- ArtImmune SAS, Orléans, France
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orléans, France
| |
Collapse
|
41
|
Thimmulappa RK, Chattopadhyay I, Rajasekaran S. Oxidative Stress Mechanisms in the Pathogenesis of Environmental Lung Diseases. OXIDATIVE STRESS IN LUNG DISEASES 2019. [PMCID: PMC7120104 DOI: 10.1007/978-981-32-9366-3_5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Globally, respiratory diseases are major cause of disability and mortality, and more alarmingly, it disproportionately affects developing countries, which is largely attributed to poor quality of air. Tobacco smoke and emissions from combustion of fossil fuel and biomass fuel are the major airborne pollutants affecting human lung health. Oxidative stress is the dominant driving force by which the airborne pollutants exert their toxicity in lungs and cause respiratory diseases. Most airborne pollutants are associated with intrinsic oxidative potential and, additionally, stimulate endogenous production of reactive oxygen species (ROS) and reactive nitrogen species (RNS). Elevated ROS and RNS in lungs modulate redox signals and cause irreversible damage to critical biomolecules (lipids, proteins and DNA) and initiate various pathogenic cellular process. This chapter provides an insight into oxidative stress-linked pathogenic cellular process such as lipid peroxidation, inflammation, cell death, mitochondrial dysfunction, endoplasmic reticulum stress, epigenetic changes, profibrotic signals and mucus hypersecretion, which drive the development and progression of lung diseases. Lungs are associated with robust enzymatic and non-enzymatic (GSH, ascorbic acid, uric acid, vitamin E) antioxidant defences. However, sustained production of free radicals due to continuous exposures to airborne pollutants overwhelms lung antioxidant defences and causes oxidative injury. Preclinical studies have demonstrated the critical roles and therapeutic potential of upregulating lung antioxidants for intervention of respiratory diseases; however, so far clinical benefits in antioxidant supplementation trials have been minimal and conflicting. Antioxidants alone may not be effective in treatment of respiratory diseases; however it could be a promising adjunctive therapy.
Collapse
|
42
|
Lim CC, Hayes RB, Ahn J, Shao Y, Silverman DT, Jones RR, Garcia C, Bell ML, Thurston GD. Long-Term Exposure to Ozone and Cause-Specific Mortality Risk in the United States. Am J Respir Crit Care Med 2019; 200:1022-1031. [PMID: 31051079 PMCID: PMC6794108 DOI: 10.1164/rccm.201806-1161oc] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 05/03/2019] [Indexed: 01/18/2023] Open
Abstract
Rationale: Many studies have linked short-term exposure to ozone (O3) with morbidity and mortality, but epidemiologic evidence of associations between long-term O3 exposure and mortality is more limited.Objectives: To investigate associations of long-term (annual or warm season average of daily 8-h maximum concentrations) O3 exposure with all-cause and cause-specific mortality in the NIH-AARP Diet and Health Study, a large prospective cohort of U.S. adults with 17 years of follow-up from 1995 to 2011.Methods: The cohort (n = 548,780) was linked to census tract-level estimates for O3. Associations between long-term O3 exposure (averaged values from 2002 to 2010) and multiple causes of death were evaluated using multivariate Cox proportional hazards models, adjusted for individual- and census tract-level covariates, and potentially confounding copollutants and temperature.Measurements and Main Results: Long-term annual average exposure to O3 was significantly associated with deaths caused by cardiovascular disease (per 10 ppb; hazard ratio [HR], 1.03; 95% confidence interval [CI], 1.01-1.06), ischemic heart disease (HR, 1.06; 95% CI, 1.02-1.09), respiratory disease (HR, 1.04; 95% CI, 1.00-1.09), and chronic obstructive pulmonary disease (HR, 1.09; 95% CI, 1.03-1.15) in single-pollutant models. The results were robust to alternative models and adjustment for copollutants (fine particulate matter and nitrogen dioxide), although some evidence of confounding by temperature was observed. Significantly elevated respiratory disease mortality risk associated with long-term O3 exposure was found among those living in locations with high temperature (Pinteraction < 0.05).Conclusions: This study found that long-term exposure to O3 is associated with increased risk for multiple causes of mortality, suggesting that establishment of annual and/or seasonal federal O3 standards is needed to more adequately protect public health from ambient O3 exposures.
Collapse
Affiliation(s)
| | - Richard B. Hayes
- Department of Environmental Medicine and
- Department of Population Health, New York University School of Medicine, New York, New York
| | - Jiyoung Ahn
- Department of Environmental Medicine and
- Department of Population Health, New York University School of Medicine, New York, New York
| | - Yongzhao Shao
- Department of Environmental Medicine and
- Department of Population Health, New York University School of Medicine, New York, New York
| | - Debra T. Silverman
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Rena R. Jones
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Cynthia Garcia
- California Air Resources Board, Sacramento, California; and
| | - Michelle L. Bell
- School of Forestry and Environmental Studies, Yale University, New Haven, Connecticut
| | - George D. Thurston
- Department of Environmental Medicine and
- Department of Population Health, New York University School of Medicine, New York, New York
| |
Collapse
|
43
|
Mumby S, Chung KF, Adcock IM. Transcriptional Effects of Ozone and Impact on Airway Inflammation. Front Immunol 2019; 10:1610. [PMID: 31354743 PMCID: PMC6635463 DOI: 10.3389/fimmu.2019.01610] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/27/2019] [Indexed: 12/24/2022] Open
Abstract
Epidemiological and challenge studies in healthy subjects and in individuals with asthma highlight the health impact of environmental ozone even at levels considered safe. Acute ozone exposure in man results in sputum neutrophilia in 30% of subjects particularly young children, females, and those with ongoing cardiopulmonary disease. This may be associated with systemic inflammation although not in all cases. Chronic exposure amplifies these effects and can result in the formation of asthma-like symptoms and immunopathology. Asthmatic patients who respond to ozone (responders) induce a greater number of genes in bronchoalveolar (BAL) macrophages than healthy responders with up-regulation of inflammatory and immune pathways under the control of cytokines and chemokines and the enhanced expression of remodeling and repair programmes including those associated with protease imbalances and cell-cell adhesion. These pathways are under the control of several key transcription regulatory factors including nuclear factor (NF)-κB, anti-oxidant factors such as nuclear factor (erythroid-derived 2)-like 2 NRF2, the p38 mitogen activated protein kinase (MAPK), and priming of the immune system by up-regulating toll-like receptor (TLR) expression. Murine and cellular models of acute and chronic ozone exposure recapitulate the inflammatory effects seen in humans and enable the elucidation of key transcriptional pathways. These studies emphasize the importance of distinct transcriptional networks in driving the detrimental effects of ozone. Studies indicate the critical role of mediators including IL-1, IL-17, and IL-33 in driving ozone effects on airway inflammation, remodeling and hyperresponsiveness. Transcription analysis and proof of mechanisms studies will enable the development of drugs to ameliorate the effects of ozone exposure in susceptible individuals.
Collapse
Affiliation(s)
- Sharon Mumby
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
44
|
Li J, Chen Y, Chen QY, Liu D, Xu L, Cheng G, Yang X, Guo Z, Zeng Y. Role of transient receptor potential cation channel subfamily V member 1 (TRPV1) on ozone-exacerbated allergic asthma in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 247:586-594. [PMID: 30708321 DOI: 10.1016/j.envpol.2019.01.091] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
Around the globe, worsening air pollution is spawning major public health and environmental concerns, especially in the poorest and most populous cities. As a major secondary air pollutant, ozone is a potential risk factor for exacerbated asthma, although the underlying mechanisms remain uncertain. In this study, we aim to investigate the role of ozone on asthma exacerbation using a classic asthmatic model with allergic airway inflammation by treating Balb/c mice with ovalbumin (OVA). Our study shows ozone exposure significantly exacerbated OVA-induced asthmatic phenotypes, including serum immunoglobulin, Th cytokines, inflammatory cell counts, mucus production, airway remodeling, and airway hyper-responsiveness (AHR). Interestingly, expression of transient receptor potential cation channel subfamily V member1 (TRPV1) was also significantly elevated in ozone-exacerbated asthmatic mice and that treatment with TRPV1 antagonist effectively suppressed AHR, airway inflammation and remodeling. The underlying mechanisms of these effects may be associated with suppression of neuropeptide calcitonin gene-related peptide (CGRP) and thymic stromal lymphopoietin (TSLP), an epithelial cell-derived cytokine. Base on the role of TRPV1 in allergic asthma, this study further revealed that inhibition of TRPV1 by TRPV1 antagonist has significant anti-inflammatory effects on ozone-induced asthma exacerbation in this study. Induction of TRPV1 expression may be an important mechanism underlying the increased risks for asthma after exposure to environmental pollutants.
Collapse
Affiliation(s)
- Jinquan Li
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China; Big Data Science and Engineering Research Institute, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Yushan Chen
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, 57 Old Forge, Tuxedo, NY 10987, United States
| | - Dan Liu
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Lang Xu
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Guirong Cheng
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Xu Yang
- Section of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Yan Zeng
- Brain and Cognitive Dysfunction Research Center, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China; Big Data Science and Engineering Research Institute, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
45
|
Smith GJ, Walsh L, Higuchi M, Kelada SNP. Development of a large-scale computer-controlled ozone inhalation exposure system for rodents. Inhal Toxicol 2019; 31:61-72. [PMID: 31021248 PMCID: PMC7055063 DOI: 10.1080/08958378.2019.1597222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/23/2019] [Accepted: 02/04/2019] [Indexed: 01/06/2023]
Abstract
Objective: Complete systems for laboratory-based inhalation toxicology studies are typically not commercially available; therefore, inhalation toxicologists utilize custom-made exposure systems. Here we report on the design, construction, testing, operation and maintenance of a newly developed in vivo rodent ozone inhalation exposure system. Materials and methods: Key design requirements for the system included large-capacity exposure chambers to facilitate studies with large sample sizes, automatic and precise control of chamber ozone concentrations, as well as automated data collection on airflow and environmental conditions. The exposure system contains two Hazelton H-1000 stainless steel and glass exposure chambers, each providing capacity for up to 180 mice or 96 rats. We developed an empirically tuned proportional-integral-derivative control loop that provides stable ozone concentrations throughout the exposure period (typically 3h), after a short ramp time (∼8 min), and across a tested concentration range of 0.2-2 ppm. Specific details on the combination of analog and digital input/output system for environmental data acquisition, control and safety systems are provided, and we outline the steps involved in maintenance and calibration of the system. Results: We show that the exposure system produces consistent ozone exposures both within and across experiments, as evidenced by low coefficients of variation in chamber ozone concentration and consistent biological responses (airway inflammation) in mice, respectively. Conclusion: Thus, we have created a large and robust ozone exposure system, facilitating future studies on the health effects of ozone in rodents.
Collapse
Affiliation(s)
- Gregory J. Smith
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leon Walsh
- United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Mark Higuchi
- United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Samir N. P. Kelada
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|