1
|
Foley HB, Eckel SP, Yang T, Vigil M, Chen X, Marsit C, Farzan SF, Bastain TM, Habre R, Breton CV. EV-miRNA associated with environmental air pollution exposures in the MADRES cohort. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae019. [PMID: 39529802 PMCID: PMC11552520 DOI: 10.1093/eep/dvae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/02/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Air pollution is a hazardous contaminant, exposure to which has substantial consequences for health during critical periods, such as pregnancy. MicroRNA (miRNA) is an epigenetic mechanism that modulates transcriptome responses to the environment and has been found to change in reaction to air pollution exposure. The data are limited regarding extracellular-vesicle (EV) miRNA variation associated with air pollution exposure during pregnancy and in susceptible populations who may be disproportionately exposed. This study aimed to identify EV-miRNA expression associated with ambient, residential exposure to PM2.5, PM10, NO2, O3 and with traffic-related NOx in 461 participants of the MADRES cohort, a low income, predominantly Hispanic pregnancy cohort based in Los Angeles, CA. This study used residence-based modeled air pollution data as well as Nanostring panels for EVmiRNA extracted with Qiagen exoRNeasy kits to evaluate 483 miRNA in plasma in early and late pregnancy. Average air pollution exposures were considered separately for 1-day, 1-week, and 8-week windows before blood collection in both early and late pregnancy. This study identified 63 and 66 EV-miRNA significantly associated with PM2.5 and PM10, respectively, and 2 miRNA associated with traffic-related NOX (False Discovery Rate-adjusted P-value < .05). Of 103 unique EV-miRNA associated with PM, 92% were associated with lung conditions according to HMDD (Human miRNA Disease Database) evidence. In particular, EV-miRNA previously identified with air pollution exposure also associated with PM2.5 and PM10 in this study were: miR-126, miR-16-5p, miR-187-3p, miR200b-3p, miR486-3p, and miR-582-3p. There were no significant differences in average exposures in early vs late pregnancy. Significant EV-miRNAs were only identified in late pregnancy with an 8-week exposure window, suggesting a vulnerable timeframe of exposure, rather than an acute response. These results describe a wide array of EV-miRNA for which expression is affected by PM exposure and may be in part mediating the biological response to ambient air pollution, with potential for health implications in pregnant women and their children.
Collapse
Affiliation(s)
- Helen Bermudez Foley
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Sandrah P Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Tingyu Yang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Mario Vigil
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Xinci Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, United States
| | - Shohreh F Farzan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Theresa M Bastain
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Rima Habre
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
- Spatial Sciences Institute, Dornsife College of Arts and Sciences, University of Southern California, Los Angeles, CA 90089, United States
| | - Carrie V Breton
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| |
Collapse
|
2
|
Ehrhardt B, Roeder T, Krauss-Etschmann S. Drosophila melanogaster as an Alternative Model to Higher Organisms for In Vivo Lung Research. Int J Mol Sci 2024; 25:10324. [PMID: 39408654 PMCID: PMC11476989 DOI: 10.3390/ijms251910324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
COPD and asthma are lung diseases that cause considerable burden to more than 800 million people worldwide. As both lung diseases are so far incurable, it is mandatory to understand the mechanisms underlying disease development and progression for developing novel therapeutic approaches. Exposures to environmental cues such as cigarette smoke in earliest life are known to increase disease risks in the individual's own future. To explore the pathomechanisms leading to later airway disease, mammalian models are instrumental. However, such in vivo experiments are time-consuming and burdensome for the animals, which applies in particular to transgenerational studies. Along this line, the fruit fly Drosophila melanogaster comes with several advantages for research in this field. The short lifespan facilitates transgenerational studies. A high number of evolutionary conserved signaling pathways, together with a large toolbox for tissue-specific gene modification, has the potential to identify novel target genes involved in disease development. A well-defined airway microbiome could help to untangle interactions between disease development and microbiome composition. In the following article, Drosophila melanogaster is therefore presented and discussed as an alternative in vivo model to investigate airway diseases that can complement and/or replace models in higher organisms.
Collapse
Affiliation(s)
- Birte Ehrhardt
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), 24118 Kiel, Germany
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
- DZL Laboratory for Experimental Microbiome Research, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts-University Kiel, 24105 Kiel, Germany
| |
Collapse
|
3
|
Ge Y, Liu Y, Ji B, Fang Y, Xie Y, Sakurai R, Wang J, Zhang Z, Wang Y, Wang X, Rehan VK. Evidence for Wnt signaling's central involvement in perinatal nicotine exposure-induced offspring lung pathology and its modulation by electroacupuncture. Biomed Pharmacother 2023; 168:115824. [PMID: 37925937 DOI: 10.1016/j.biopha.2023.115824] [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: 08/08/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023] Open
Abstract
OBJECTIVE Many factors during pregnancy can induce intrauterine growth restriction (IUGR), resulting in various adverse perinatal outcomes such as low birth weight and multiple organ disorders. Among these factors, prenatal smoke/nicotine exposure is a common cause of IUGR, often associated with altered fetal lung development. The classical Wnt signaling pathway plays a vital role in lung development, and its alterations are commonly associated with developmental lung pathologies. The purpose of this study was to determine whether electroacupuncture (EA) at "Zusanli" (ST 36) points protects perinatal nicotine exposure (PNE)-induced offspring lung dysplasia through Wnt/β-catenin signaling pathway and to identify specific Wnt signaling pathway targets of EA. METHODS Following a well-established protocol, nicotine (1 mg/kg/ body weight) was administered subcutaneously to pregnant Sprague Dawley rat dams from gestational day 6 to postnatal day 21. In the EA group, dams were treated with EA at both ST 36 acupoints, while in another experimental group, Wnt/β-catenin signaling pathway agonist was injected subcutaneously (2 mg/kg/ body weight). Offspring body weight (PND 1, 7, 14, and 21), lung weight, Wnt signaling markers, pulmonary function, and lung morphology were determined at sacrifice on PND 21. Specifically, Western blotting and Real-time PCR were used to detect the protein and mRNA levels of critical Wnt signaling markers Wnt2, Wnt7b, FZD4, FZD7, LRP5, and LRP6 in the offspring lung. The protein levels of β-catenin in lung tissue of offspring rats were detected by ELISA that of LEF-1 by Western blotting. RESULTS Compared to the control group, the body and lung weights of the offspring rats were significantly decreased in the nicotine-only exposed group. The pulmonary function determined as FVC, PEF, TV, and Cdyn was also significantly decreased, while PIF was significantly increased. The protein levels and mRNA expression of Wnt2, Wnt7b, FZD4, FZD7, LRP5, and LRP6 in the lung tissue of the PNE offspring rats were significantly increased. With EA administration at ST 36 acupoints concomitant with nicotine administration, the body and lung weights, pulmonary function (FVC, PEF, PIF, TV, and Cdyn), protein and mRNA levels Wnt signaling pathway markers (Wnt2, Wnt7b, FZD4, FZD7, LRP5, LRP6, β-catenin, and LEF-1) normalized and were not different from the control group. Notably, Wnt agonists agonist administration blocked the protective effects of EA against PNE-induced lung morphological, molecular, and function changes, highlighting the central significance of Wnt pathway signaling in PNE-induced offspring pulmonary pathology and its modulation by EA at ST 36 acupoints. CONCLUSION Concomitant maternal EA at ST 36 acupoints from gestational day 6 to PND 21 protects against offspring PNE-induced lung phenotype. The protective effect is achieved by regulating the expression of Wnt ligand proteins (Wnt2 and Wnt7b) and receptor proteins (FZD4, FZD7, LRP5, and LRP6) upstream of the Wnt/β-catenin signaling pathway intermediates β-catenin, and LEF-1.
Collapse
Affiliation(s)
- Yunpeng Ge
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yitian Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Bo Ji
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yang Fang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yana Xie
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Reiko Sakurai
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90502, USA
| | - Jiajia Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ziyue Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yifei Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xu Wang
- School of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing 102488, China
| | - Virender K Rehan
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90502, USA
| |
Collapse
|
4
|
Kurihara C, Kuniyoshi KM, Rehan VK. Preterm Birth, Developmental Smoke/Nicotine Exposure, and Life-Long Pulmonary Sequelae. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040608. [PMID: 37189857 DOI: 10.3390/children10040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
This review delineates the main pulmonary issues related to preterm birth, perinatal tobacco/nicotine exposure, and its effects on offspring, focusing on respiratory health and its possible transmission to subsequent generations. We review the extent of the problem of preterm birth, prematurity-related pulmonary effects, and the associated increased risk of asthma later in life. We then review the impact of developmental tobacco/nicotine exposure on offspring asthma and the significance of transgenerational pulmonary effects following perinatal tobacco/nicotine exposure, possibly via its effects on germline epigenetics.
Collapse
Affiliation(s)
- Chie Kurihara
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Katherine M Kuniyoshi
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Virender K Rehan
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Rumph JT, Stephens VR, Ameli S, Brown LK, Rayford KJ, Nde PN, Osteen KG, Bruner-Tran KL. A Paternal Fish Oil Diet Preconception Reduces Lung Inflammation in a Toxicant-Driven Murine Model of New Bronchopulmonary Dysplasia. Mar Drugs 2023; 21:161. [PMID: 36976210 PMCID: PMC10052688 DOI: 10.3390/md21030161] [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: 12/22/2022] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
New bronchopulmonary dysplasia (BPD) is a neonatal disease that is theorized to begin in utero and manifests as reduced alveolarization due to inflammation of the lung. Risk factors for new BPD in human infants include intrauterine growth restriction (IUGR), premature birth (PTB) and formula feeding. Using a mouse model, our group recently reported that a paternal history of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increased his offspring's risk of IUGR, PTB, and new BPD. Additionally, formula supplementation of these neonates worsened the severity of pulmonary disease. In a separate study, we reported that a paternal preconception fish oil diet prevented TCDD-driven IUGR and PTB. Not surprisingly, eliminating these two major risk factors for new BPD also significantly reduced development of neonatal lung disease. However, this prior study did not examine the potential mechanism for fish oil's protective effect. Herein, we sought to determine whether a paternal preconception fish oil diet attenuated toxicant-associated lung inflammation, which is an important contributor to the pathogenesis of new BPD. Compared to offspring of standard diet TCDD-exposed males, offspring of TCDD-exposed males provided a fish oil diet prior to conception exhibited a significant reduction in pulmonary expression of multiple pro-inflammatory mediators (Tlr4, Cxcr2, Il-1 alpha). Additionally, neonatal lungs of pups born to fish oil treated fathers exhibited minimal hemorrhaging or edema. Currently, prevention of BPD is largely focused on maternal strategies to improve health (e.g., smoking cessation) or reduce risk of PTB (e.g., progesterone supplementation). Our studies in mice support a role for also targeting paternal factors to improve pregnancy outcomes and child health.
Collapse
Affiliation(s)
- Jelonia T. Rumph
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Victoria R. Stephens
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - LaKendria K. Brown
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kevin G. Osteen
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| | - Kaylon L. Bruner-Tran
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Yim G, Roberts A, Ascherio A, Wypij D, Kioumourtzoglou MA, Weisskopf AMG. Smoking During Pregnancy and Risk of Attention-deficit/Hyperactivity Disorder in the Third Generation. Epidemiology 2022; 33:431-440. [PMID: 35213510 PMCID: PMC9010055 DOI: 10.1097/ede.0000000000001467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Animal experiments indicate that environmental factors, such as cigarette smoke, can have multigenerational effects through the germline. However, there are little data on multigenerational effects of smoking in humans. We examined the associations between grandmothers' smoking while pregnant and risk of attention-deficit/hyperactivity disorder (ADHD) in her grandchildren. METHODS Our study population included 53,653 Nurses' Health Study II (NHS-II) participants (generation 1 [G1]), their mothers (generation 0 [G0]), and their 120,467 live-born children (generation 2 [G2]). In secondary analyses, we used data from 23,844 mothers of the nurses who were participants in the Nurses' Mothers' Cohort Study (NMCS), a substudy of NHS-II. RESULTS The prevalence of G0 smoking during the pregnancy with the G1 nurse was 25%. ADHD was diagnosed in 9,049 (7.5%) of the grandchildren (G2). Grand-maternal smoking during pregnancy was associated with increased odds of ADHD among the grandchildren (adjusted odds ratio [aOR] = 1.2; 95% confidence interval [CI] = 1.1, 1.2), independent of G1 smoking during pregnancy. In the Nurses' Mothers' Cohort Study, odds of ADHD increased with increasing cigarettes smoked per day by the grandmother (1-14 cigarettes: aOR = 1.1; 95% CI = 1.0, 1.2; 15+: aOR = 1.2; 95% CI = 1.0, 1.3), compared with nonsmoking grandmothers. CONCLUSIONS Grandmother smoking during pregnancy is associated with an increased risk of ADHD among the grandchildren.
Collapse
Affiliation(s)
- Gyeyoon Yim
- From the Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Andrea Roberts
- From the Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Alberto Ascherio
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - David Wypij
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Cardiology, Children's Hospital Boston, Boston, MA
| | | | - And Marc G Weisskopf
- From the Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
7
|
Accordini S, Calciano L, Johannessen A, Benediktsdóttir B, Bertelsen RJ, Bråbäck L, Dharmage SC, Forsberg B, Gómez Real F, Holloway JW, Holm M, Janson C, Jõgi NO, Jõgi R, Malinovschi A, Marcon A, Martínez-Moratalla Rovira J, Sánchez-Ramos JL, Schlünssen V, Torén K, Jarvis D, Svanes C. Prenatal and prepubertal exposures to tobacco smoke in men may cause lower lung function in future offspring: a three-generation study using a causal modelling approach. Eur Respir J 2021; 58:2002791. [PMID: 33795316 PMCID: PMC8529197 DOI: 10.1183/13993003.02791-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 03/11/2021] [Indexed: 11/24/2022]
Abstract
Mechanistic research suggests that lifestyle and environmental factors impact respiratory health across generations by epigenetic changes transmitted through male germ cells. Evidence from studies on humans is very limited.We investigated multigeneration causal associations to estimate the causal effects of tobacco smoking on lung function within the paternal line. We analysed data from 383 adult offspring (age 18-47 years; 52.0% female) and their 274 fathers, who had participated in the European Community Respiratory Health Survey (ECRHS)/Respiratory Health in Northern Europe, Spain and Australia (RHINESSA) generation study and had provided valid measures of pre-bronchodilator lung function. Two counterfactual-based, multilevel mediation models were developed with: paternal grandmothers' smoking in pregnancy and fathers' smoking initiation in prepuberty as exposures; fathers' forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC), or FEV1/FVC z-scores as potential mediators (proxies of unobserved biological mechanisms that are true mediators); and offspring's FEV1 and FVC, or FEV1/FVC z-scores as outcomes. All effects were summarised as differences (Δ) in expected z-scores related to fathers' and grandmothers' smoking history.Fathers' smoking initiation in prepuberty had a negative direct effect on both offspring's FEV1 (Δz-score -0.36, 95% CI -0.63- -0.10) and FVC (-0.50, 95% CI -0.80- -0.20) compared with fathers' never smoking. Paternal grandmothers' smoking in pregnancy had a negative direct effect on fathers' FEV1/FVC (-0.57, 95% CI -1.09- -0.05) and a negative indirect effect on offspring's FEV1/FVC (-0.12, 95% CI -0.21- -0.03) compared with grandmothers' not smoking before fathers' birth nor during fathers' childhood.Fathers' smoking in prepuberty and paternal grandmothers' smoking in pregnancy may cause lower lung function in offspring. Our results support the concept that lifestyle-related exposures during these susceptibility periods influence the health of future generations.
Collapse
Affiliation(s)
- Simone Accordini
- Unit of Epidemiology and Medical Statistics, Dept of Diagnostics and Public Health, University of Verona, Verona, Italy
- Equal contribution as first authors
| | - Lucia Calciano
- Unit of Epidemiology and Medical Statistics, Dept of Diagnostics and Public Health, University of Verona, Verona, Italy
- Equal contribution as first authors
| | - Ane Johannessen
- Centre for International Health, Dept of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | | | - Randi Jacobsen Bertelsen
- Dept of Clinical Science, University of Bergen, Bergen, Norway
- Oral Health Centre of Expertise in Western Norway/Vestland, Bergen, Norway
| | - Lennart Bråbäck
- Section of Sustainable Health, Dept of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Shyamali C Dharmage
- Allergy and Lung Health Unit, School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Bertil Forsberg
- Section of Sustainable Health, Dept of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Francisco Gómez Real
- Dept of Clinical Science, University of Bergen, Bergen, Norway
- Dept of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christer Janson
- Dept of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Nils O Jõgi
- Dept of Clinical Science, University of Bergen, Bergen, Norway
- Lung Clinic, Tartu University Hospital, Tartu, Estonia
| | - Rain Jõgi
- Lung Clinic, Tartu University Hospital, Tartu, Estonia
| | - Andrei Malinovschi
- Dept of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Alessandro Marcon
- Unit of Epidemiology and Medical Statistics, Dept of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Jesús Martínez-Moratalla Rovira
- Servicio de Neumología, Complejo Hospitalario Universitario de Albacete (CHUA), Servicio de Salud de Castilla-La Mancha (SESCAM), Albacete, Spain
| | | | | | - Kjell Torén
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Deborah Jarvis
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Equal contribution as last authors
| | - Cecilie Svanes
- Centre for International Health, Dept of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Equal contribution as last authors
| |
Collapse
|
8
|
Collaco JM, Aoyama BC, Rice JL, McGrath-Morrow SA. Influences of environmental exposures on preterm lung disease. Expert Rev Respir Med 2021; 15:1271-1279. [PMID: 34114906 PMCID: PMC8453051 DOI: 10.1080/17476348.2021.1941886] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/09/2021] [Indexed: 01/09/2023]
Abstract
Introduction: Environmental factors play a critical role in the progression or resolution of chronic respiratory diseases. However, studies are limited on the impact of environmental risk factors on individuals born prematurely with lung disease after they leave the neonatal intensive care unit and are discharged into the home environment.Areas covered: In this review, we cover current knowledge of environmental exposures that impact outcomes of preterm respiratory disease, including air pollution, infections, and disparities. The limited data do suggest that certain exposures should be avoided and there are potential preventative strategies for other exposures. There is a need for additional research outside the neonatal intensive care unit that focuses on individual and community-level factors that affect long-term outcomes.Expert opinion: Preterm respiratory disease can impose a significant burden on infants, children, and young adults born prematurely, but may improve for many individuals over time. In this review, we outline the exposures that may potentially hasten, delay, or prevent resolution of lung injury in preterm children.
Collapse
Affiliation(s)
- Joseph M. Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brianna C. Aoyama
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jessica L. Rice
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sharon A. McGrath-Morrow
- Division of Pulmonary and Sleep, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Role of Epigenetics in the Pathogenesis, Treatment, Prediction, and Cellular Transformation of Asthma. Mediators Inflamm 2021; 2021:9412929. [PMID: 34566492 PMCID: PMC8457970 DOI: 10.1155/2021/9412929] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Asthma is a mysterious disease with heterogeneity in etiology, pathogenesis, and clinical phenotypes. Although ongoing studies have provided a better understanding of asthma, its natural history, progression, pathogenesis, diversified phenotypes, and even the exact epigenetic linkage between childhood asthma and adult-onset/old age asthma remain elusive in many aspects. Asthma heritability has been established through genetic studies, but genetics is not the only influencing factor in asthma. The increasing incidence and some unsolved queries suggest that there may be other elements related to asthma heredity. Epigenetic mechanisms link genetic and environmental factors with developmental trajectories in asthma. This review provides an overview of asthma epigenetics and its components, including several epigenetic studies on asthma, and discusses the epigenetic linkage between childhood asthma and adult-onset/old age asthma. Studies involving asthma epigenetics present valuable novel approaches to solve issues related to asthma. Asthma epigenetic research guides us towards gene therapy and personalized T cell therapy, directs the discovery of new therapeutic agents, predicts long-term outcomes in severe cases, and is also involved in the cellular transformation of childhood asthma to adult-onset/old age asthma.
Collapse
|
10
|
Breton CV, Landon R, Kahn LG, Enlow MB, Peterson AK, Bastain T, Braun J, Comstock SS, Duarte CS, Hipwell A, Ji H, LaSalle JM, Miller RL, Musci R, Posner J, Schmidt R, Suglia SF, Tung I, Weisenberger D, Zhu Y, Fry R. Exploring the evidence for epigenetic regulation of environmental influences on child health across generations. Commun Biol 2021; 4:769. [PMID: 34158610 PMCID: PMC8219763 DOI: 10.1038/s42003-021-02316-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Environmental exposures, psychosocial stressors and nutrition are all potentially important influences that may impact health outcomes directly or via interactions with the genome or epigenome over generations. While there have been clear successes in large-scale human genetic studies in recent decades, there is still a substantial amount of missing heritability to be elucidated for complex childhood disorders. Mounting evidence, primarily in animals, suggests environmental exposures may generate or perpetuate altered health outcomes across one or more generations. One putative mechanism for these environmental health effects is via altered epigenetic regulation. This review highlights the current epidemiologic literature and supporting animal studies that describe intergenerational and transgenerational health effects of environmental exposures. Both maternal and paternal exposures and transmission patterns are considered, with attention paid to the attendant ethical, legal and social implications.
Collapse
Affiliation(s)
- Carrie V Breton
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Remy Landon
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Linda G Kahn
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alicia K Peterson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Theresa Bastain
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joseph Braun
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Cristiane S Duarte
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Alison Hipwell
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Ji
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, Davis, CA, USA
| | | | - Rashelle Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jonathan Posner
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Rebecca Schmidt
- Department of Public Health Sciences, UC Davis School of Medicine, Davis, CA, USA
| | | | - Irene Tung
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Weisenberger
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California and Department of Epidemiology and Biostatistics, University of California, San Francisco, Oakland, CA, USA
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
11
|
Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, Schmidt‐Weber CB, Maes T, Rebane A, Krauss‐Etschmann S, Rådinger M. Spotlight on microRNAs in allergy and asthma. Allergy 2021; 76:1661-1678. [PMID: 33128813 PMCID: PMC8246745 DOI: 10.1111/all.14646] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/16/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
In past 10 years, microRNAs (miRNAs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases and their potential as biomarkers in liquid biopsies. They act as master post‐transcriptional regulators that control most cellular processes. As one miRNA can target several mRNAs, often within the same pathway, dysregulated expression of miRNAs may alter particular cellular responses and contribute, or lead, to the development of various diseases. In this review, we give an overview of the current research on miRNAs in allergic diseases, including atopic dermatitis, allergic rhinitis, and asthma. Specifically, we discuss how individual miRNAs function in the regulation of immune responses in epithelial cells and specialized immune cells in response to different environmental factors and respiratory viruses. In addition, we review insights obtained from experiments with murine models of allergic airway and skin inflammation and offer an overview of studies focusing on miRNA discovery using profiling techniques and bioinformatic modeling of the network effect of multiple miRNAs. In conclusion, we highlight the importance of research into miRNA function in allergy and asthma to improve our knowledge of the molecular mechanisms involved in the pathogenesis of this heterogeneous group of diseases.
Collapse
Affiliation(s)
- Julie Weidner
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Sabine Bartel
- Department of Pathology and Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Ayse Kılıç
- Channing Division of Network Medicine Brigham and Women's Hospital Boston MA USA
| | - Ulrich M. Zissler
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Harald Renz
- Institut für Laboratoriumsmedizin und Pathobiochemie Philipps University of Marburg Marburg Germany
| | - Jürgen Schwarze
- Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Carsten B. Schmidt‐Weber
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Tania Maes
- Department of Respiratory Medicine Ghent University Ghent Belgium
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Susanne Krauss‐Etschmann
- Research Center Borstel Borstel Germany
- Institute of Experimental Medicine Christian‐Albrechts University Kiel Kiel Germany
| | - Madeleine Rådinger
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
12
|
Sheikhpour M, Maleki M, Ebrahimi Vargoorani M, Amiri V. A review of epigenetic changes in asthma: methylation and acetylation. Clin Epigenetics 2021; 13:65. [PMID: 33781317 PMCID: PMC8008616 DOI: 10.1186/s13148-021-01049-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/04/2021] [Indexed: 12/30/2022] Open
Abstract
Several studies show that childhood and adulthood asthma and its symptoms can be modulated through epigenetic modifications. Epigenetic changes are inheritable modifications that can modify the gene expression without changing the DNA sequence. The most common epigenetic alternations consist of DNA methylation and histone modifications. How these changes lead to asthmatic phenotype or promote the asthma features, in particular by immune pathways regulation, is an understudied topic. Since external effects, like exposure to tobacco smoke, air pollution, and drugs, influence both asthma development and the epigenome, elucidating the role of epigenetic changes in asthma is of great importance. This review presents available evidence on the epigenetic process that drives asthma genes and pathways, with a particular focus on DNA methylation, histone methylation, and acetylation. We gathered and assessed studies conducted in this field over the past two decades. Our study examined asthma in different aspects and also shed light on the limitations and the important factors involved in the outcomes of the studies. To date, most of the studies in this area have been carried out on DNA methylation. Therefore, the need for diagnostic and therapeutic applications through this molecular process calls for more research on the histone modifications in this disease.
Collapse
Affiliation(s)
- Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Mobina Maleki
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Ebrahimi Vargoorani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Microbiology, College of Basic Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Vahid Amiri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Mazzoccoli G, Kvetnoy I, Mironova E, Yablonskiy P, Sokolovich E, Krylova J, Carbone A, Anderson G, Polyakova V. The melatonergic pathway and its interactions in modulating respiratory system disorders. Biomed Pharmacother 2021; 137:111397. [PMID: 33761613 DOI: 10.1016/j.biopha.2021.111397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
Melatonin is a key intracellular neuroimmune-endocrine regulator and coordinator of multiple complex and interrelated biological processes. The main functions of melatonin include the regulation of neuroendocrine and antioxidant system activity, blood pressure, rhythms of the sleep-wake cycle, the retardation of ageing processes, as well as reseting and optimizing mitochondria and thereby the cells of the immune system. Melatonin and its agonists have therefore been mooted as a treatment option across a wide array of medical disorders. This article reviews the role of melatonin in the regulation of respiratory system functions under normal and pathological conditions. Melatonin can normalize the structural and functional organization of damaged lung tissues, by a number of mechanisms, including the regulation of signaling molecules, oxidant status, lipid raft function, optimized mitochondrial function and reseting of the immune response over the circadian rhythm. Consequently, melatonin has potential clinical utility for bronchial asthma, chronic obstructive pulmonary disease, lung cancer, lung vascular diseases, as well as pulmonary and viral infections. The integration of melatonin's effects with the alpha 7 nicotinic receptor and the aryl hydrocarbon receptor in the regulation of mitochondrial function are proposed as a wider framework for understanding the role of melatonin across a wide array of diverse pulmonary disorders.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy.
| | - Igor Kvetnoy
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; Department of Pathology, Saint Petersburg State University, University Embankment, 7/9, Saint Petersburg 199034, Russian Federation
| | - Ekaterina Mironova
- Saint Petersburg Institute of Bioregulation and Gerontology, Dynamo Ave., 3, Saint Petersburg 197110, Russian Federation
| | - Petr Yablonskiy
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation
| | - Evgenii Sokolovich
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation
| | - Julia Krylova
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; Pavlov First Saint Petersburg State Medical University, Lev Tolstoy str. 6-8, Saint Petersburg 197022, Russian Federation
| | - Annalucia Carbone
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy
| | | | - Victoria Polyakova
- Saint Petersburg Institute of Phthisiopulmonology, Lygovsky Ave. 2-4, Saint Petersburg 191036, Russian Federation; St. Petersburg State Pediatric Medical University, Litovskaia str. 2, Saint-Petersburg 194100, Russian Federation
| |
Collapse
|
14
|
Mahon GM, Koppelman GH, Vonk JM. Grandmaternal smoking, asthma and lung function in the offspring: the Lifelines cohort study. Thorax 2021; 76:441-447. [PMID: 33542091 PMCID: PMC8070652 DOI: 10.1136/thoraxjnl-2020-215232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 12/01/2022]
Abstract
Background/objective Limited research exists regarding the association between grandmaternal smoking during pregnancy and the risk for asthma and altered lung function in grandchildren. This study aimed to investigate this association in a three-generation design. Methods 37 291 participants (25 747 adults and 11 544 children) were included from the Lifelines study, a prospective longitudinal three generation cohort study in The Netherlands. Spirometry was available in 69.5% and 61.1% of the included adults and children. Logistic and linear regression were used to analyse the association between grandmaternal smoking during pregnancy and (1) asthma, (2) early childhood asthma (ie, onset before 6 years) and (3) lung function level. Maternal and paternal grandmaternal smoking were studied separately and the analyses were stratified by adult/child and by gender. The analyses were adjusted for gender, current smoking, birth variables and socioeconomic status. Results In the adult population, maternal grandmaternal smoking during pregnancy was associated with a higher risk for asthma (OR (95% CI): 1.38 (1.06 to 1.79)), early childhood asthma (1.49 (95% CI 1.06 to 2.11)) and a lower FEV1/FVC% predicted (B (95% CI): −1.04 (−1.91 to −0.16) in men. These findings were not observed in a separate analysis of children that participated in this study. There was also no significant association between paternal grandmaternal smoking and asthma/lung function. Conclusion Maternal grandmaternal smoking during pregnancy is associated with higher asthma risk and lower lung function in male grandchildren and a reverse effect in male grandchildren of subsequent generations. Our study highlights the deep-rooted effects of tobacco smoking across generations.
Collapse
Affiliation(s)
- Gillian M Mahon
- Department of Pediatric Pulmonology and Pediatric Allergology, GRIAC Research Institute, University Medical Center Groningen Beatrix Children's Hospital, Groningen, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, GRIAC Research Institute, University Medical Center Groningen Beatrix Children's Hospital, Groningen, The Netherlands
| | - Judith M Vonk
- Department of Epidemiology, GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Tissue-Resident Type 2 Innate Lymphoid Cells Arrest Alveolarization in Bronchopulmonary Dysplasia. J Immunol Res 2020; 2020:8050186. [PMID: 33178840 PMCID: PMC7648679 DOI: 10.1155/2020/8050186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/09/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe complication of the respiratory system associated with preterm birth. Type 2 innate lymphoid cells (ILC2s) play a major role in tissue homeostasis, inflammation, and wound healing. However, the role in BPD remains unclear. The present study showed that ILC2s, interleukin-4 (IL-4), IL-13, and anti-inflammatory (M2) macrophages increased significantly in BPD mice as compared to the control mice. Administration with recombinant mouse IL-33 amplified the above phenomena and aggravated the alveolar structural disorder and functional injury in mice subjected to BPD, and the opposite was true with anti-ST2 antibody. In addition, the depletion of ILC2s in BPD mice with anti-CD90.2 antibody substantially abolished the destructive effect on BPD. In the treatment of BPD with dexamethasone, the number of ILC2s and M2 macrophages and levels of IL-4 and IL-13 decreased with remission as compared to the control group. This study identified a major destructive role of the ILC2s in BPD that could be attenuated as a therapeutic strategy.
Collapse
|
16
|
Liu J, Yu C, Doherty TM, Akbari O, Allard P, Rehan VK. Perinatal nicotine exposure-induced transgenerational asthma: Effects of reexposure in F1 gestation. FASEB J 2020; 34:11444-11459. [PMID: 32654256 PMCID: PMC7839813 DOI: 10.1096/fj.201902386r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 01/31/2023]
Abstract
In a rat model, perinatal nicotine exposure results in an epigenetically driven multi- and trans-generationally transmitted asthmatic phenotype that tends to wane over successive generations. However, the effect of repeat nicotine exposure during the F1 (Filial 1) gestational period on the transmitted phenotype is unknown. Using a well-established rat model, we compared lung function, mesenchymal markers of airway reactivity, and global gonadal DNA methylation changes in F2 offspring in a sex-specific manner following perinatal exposure to nicotine in only the F0 gestation, in both F0 and F1 (F0/F1) gestations, and in neither (control group). Both F0 only and F0/F1 exposure groups showed an asthmatic phenotype, an effect that was more pronounced in the F0/F1 exposure group, especially in males. Testicular global DNA methylation increased, while ovarian global DNA methylation decreased in the F0/F1 exposed group. Since the offspring of smokers are more likely to smoke than the offspring of nonsmokers, this sets the stage for more severe asthma if both mother and grandmother had smoked during their pregnancies. Increased gonadal DNA methylation changes following nicotine reexposure in the F1 generation suggests that epigenetic mechanisms might well underlie the transgenerational inheritance of acquired phenotypic traits in general and nicotine-induced asthma in particular.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pediatrics/Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Celia Yu
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Terence M. Doherty
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Patrick Allard
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Institute for Society and Genetics, UCLA, Los Angeles, CA, USA
| | - Virender K. Rehan
- Department of Pediatrics/Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Institute for Society and Genetics, UCLA, Los Angeles, CA, USA
| |
Collapse
|
17
|
Singh SP, Devadoss D, Manevski M, Sheybani A, Ivanciuc T, Exil V, Agarwal H, Raizada V, Garofalo RP, Chand HS, Sopori ML. Gestational Exposure to Cigarette Smoke Suppresses the Gasotransmitter H 2S Biogenesis and the Effects Are Transmitted Transgenerationally. Front Immunol 2020; 11:1628. [PMID: 32849552 PMCID: PMC7399059 DOI: 10.3389/fimmu.2020.01628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Gestational cigarette smoke (CS) impairs lung angiogenesis and alveolarization, promoting transgenerational development of asthma and bronchopulmonary dysplasia (BPD). Hydrogen sulfide (H2S), a proangiogenic, pro-alveolarization, and anti-asthmatic gasotransmitter is synthesized by cystathionine-γ-lyase (CSE), cystathionine-β-synthase (CBS), and 3-mercaptopyruvate sulfur transferase (3MST). Objective: Determine if gestational CS exposure affected the expression of H2S synthesizing enzymes in the mouse lung and human placenta. Methods: Mice were exposed throughout gestational period to secondhand CS (SS) at approximating the dose of CS received by a pregnant woman sitting in a smoking bar for 3 h/days during pregnancy. Lungs from 7-days old control and SS-exposed pups and human placenta from mothers who were either non-smokers or smokers during pregnancy were analyzed for expression of the enzymes. Measurements: Mouse lungs and human placentas were examined for the expression of CSE, CBS, and 3MST by immunohistochemical staining, qRT-PCR and/or Western blot (WB) analyses. Results: Compared to controls, mouse lung exposed gestationally to SS had significantly lower levels of CSE, CBS, and 3MST. Moreover, the SS-induced suppression of CSE and CBS in F1 lungs was transmitted to the F2 generation without significant change in the magnitude of the suppression. These changes were associated with impaired epithelial-mesenchymal transition (EMT)-a process required for normal lung angiogenesis and alveolarization. Additionally, the placentas from mothers who smoked during pregnancy, expressed significantly lower levels of CSE, CBS, and 3MST, and the effects were partially moderated by quitting smoking during the first trimester. Conclusions: Lung H2S synthesizing enzymes are downregulated by gestational CS and the effects are transmitted to F2 progeny. Smoking during pregnancy decreases H2S synthesizing enzymes is human placentas, which may correlate with the increased risk of asthma/BPD in children.
Collapse
Affiliation(s)
- Shashi P. Singh
- Respiratory Immunology Division, Lovelace Respiratory Research Institute, Albuquerque, NM, United States
| | - Dinesh Devadoss
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Marko Manevski
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Aryaz Sheybani
- Respiratory Immunology Division, Lovelace Respiratory Research Institute, Albuquerque, NM, United States
| | - Teodora Ivanciuc
- Department of Microbiology and Immunology, Galveston, TX, United States
| | - Vernat Exil
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Hemant Agarwal
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Veena Raizada
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | | | - Hitendra S. Chand
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Mohan L. Sopori
- Respiratory Immunology Division, Lovelace Respiratory Research Institute, Albuquerque, NM, United States
| |
Collapse
|
18
|
Yue H, Ji X, Ku T, Li G, Sang N. Sex difference in bronchopulmonary dysplasia of offspring in response to maternal PM 2.5 exposure. JOURNAL OF HAZARDOUS MATERIALS 2020; 389:122033. [PMID: 32004849 DOI: 10.1016/j.jhazmat.2020.122033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/05/2020] [Indexed: 06/10/2023]
Abstract
The adverse effects of fine particulate matters (PM2.5) on respiratory diseases start in utero. In order to investigate whether maternal PM2.5 exposure could lead to bronchopulmonary dysplasia (BPD) in offspring, PM2.5 was collected in Taiyuan, Shanxi, China during the annual heating period. Mice were mated and gestation day 0 (GD0) was considered the day on which a vaginal plug was observed. The plug-positive mice received 3 mg/kg b.w. PM2.5 by oropharyngeal aspiration every other day starting on GD0 and throughout the gestation period. Offspring were sacrificed at postnatal days (PNDs) 1, 7, 14 and 21. We assessed some typical BPD-like symptoms in offspring. The results showed that maternal PM2.5 exposure caused low birth weight, hypoalveolarization, decreased angiogenesis, suppressed production of secretory and surfactant proteins, and increased inflammation in the lungs of male offspring. However, maternal PM2.5 exposure induced only hypoalveolarization and inflammation in the lungs of female offspring. Furthermore, these alterations were reversed during postnatal development. Our results demonstrated that maternal exposure to PM2.5 caused reversible BPD-related consequences in offspring, and male offspring were more sensitive than females. However, these alterations were reversed during postnatal development.
Collapse
Affiliation(s)
- Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
19
|
Legoff L, D’Cruz SC, Tevosian S, Primig M, Smagulova F. Transgenerational Inheritance of Environmentally Induced Epigenetic Alterations during Mammalian Development. Cells 2019; 8:cells8121559. [PMID: 31816913 PMCID: PMC6953051 DOI: 10.3390/cells8121559] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Genetic studies traditionally focus on DNA as the molecule that passes information on from parents to their offspring. Changes in the DNA code alter heritable information and can more or less severely affect the progeny's phenotype. While the idea that information can be inherited between generations independently of the DNA's nucleotide sequence is not new, the outcome of recent studies provides a mechanistic foundation for the concept. In this review, we attempt to summarize our current knowledge about the transgenerational inheritance of environmentally induced epigenetic changes. We focus primarily on studies using mice but refer to other species to illustrate salient points. Some studies support the notion that there is a somatic component within the phenomenon of epigenetic inheritance. However, here, we will mostly focus on gamete-based processes and the primary molecular mechanisms that are thought to contribute to epigenetic inheritance: DNA methylation, histone modifications, and non-coding RNAs. Most of the rodent studies published in the literature suggest that transgenerational epigenetic inheritance through gametes can be modulated by environmental factors. Modification and redistribution of chromatin proteins in gametes is one of the major routes for transmitting epigenetic information from parents to the offspring. Our recent studies provide additional specific cues for this concept and help better understand environmental exposure influences fitness and fidelity in the germline. In summary, environmental cues can induce parental alterations and affect the phenotypes of offspring through gametic epigenetic inheritance. Consequently, epigenetic factors and their heritability should be considered during disease risk assessment.
Collapse
Affiliation(s)
- Louis Legoff
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Shereen Cynthia D’Cruz
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Sergei Tevosian
- University of Florida, Department of Physiological Sciences Box 100144, 1333 Center Drive, Gainesville, FL 32610, USA;
| | - Michael Primig
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Fatima Smagulova
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
- Correspondence:
| |
Collapse
|
20
|
Hammer B, Wagner C, Divac Rankov A, Reuter S, Bartel S, Hylkema MN, Krüger A, Svanes C, Krauss-Etschmann S. In utero exposure to cigarette smoke and effects across generations: A conference of animals on asthma. Clin Exp Allergy 2019; 48:1378-1390. [PMID: 30244507 DOI: 10.1111/cea.13283] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 07/24/2018] [Accepted: 09/01/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND The prevalence of asthma and chronic obstructive pulmonary disease (COPD) has risen markedly over the last decades and is reaching epidemic proportions. However, underlying molecular mechanisms are not fully understood, hampering the urgently needed development of approaches to prevent these diseases. It is well established from epidemiological studies that prenatal exposure to cigarette smoke is one of the main risk factors for aberrant lung function development or reduced fetal growth, but also for the development of asthma and possibly COPD later in life. Of note, recent evidence suggests that the disease risk can be transferred across generations, that is, from grandparents to their grandchildren. While initial studies in mouse models on in utero smoke exposure have provided important mechanistic insights, there are still knowledge gaps that need to be filled. OBJECTIVE Thus, in this review, we summarize current knowledge on this topic derived from mouse models, while also introducing two other relevant animal models: the fruit fly Drosophila melanogaster and the zebrafish Danio rerio. METHODS This review is based on an intensive review of PubMed-listed transgenerational animal studies from 1902 to 2018 and focuses in detail on selected literature due to space limitations. RESULTS This review gives a comprehensive overview of mechanistic insights obtained in studies with the three species, while highlighting the remaining knowledge gaps. We will further discuss potential (dis)advantages of all three animal models. CONCLUSION/CLINICAL RELEVANCE Many studies have already addressed transgenerational inheritance of disease risk in mouse, zebrafish or fly models. We here propose a novel strategy for how these three model organisms can be synergistically combined to achieve a more detailed understanding of in utero cigarette smoke-induced transgenerational inheritance of disease risk.
Collapse
Affiliation(s)
- Barbara Hammer
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany
| | - Christina Wagner
- Invertebrate Models, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Sabine Bartel
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany
| | - Machteld N Hylkema
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Arne Krüger
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany.,Institute for Life Science and Technology, Hanze University of Applied Sciences, Groningen, The Netherlands
| | - Cecilie Svanes
- Centre for International Health, University of Bergen, Bergen, Norway.,Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany.,Institute for Experimental Medicine, Christian-Albrechts-Universitaet zu Kiel, Kiel, Germany
| |
Collapse
|
21
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
22
|
Jeong KS, Kim S, Kim WJ, Kim HC, Bae J, Hong YC, Ha M, Ahn K, Lee JY, Kim Y, Ha E. Cohort profile: Beyond birth cohort study - The Korean CHildren's ENvironmental health Study (Ko-CHENS). ENVIRONMENTAL RESEARCH 2019; 172:358-366. [PMID: 30825686 DOI: 10.1016/j.envres.2018.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/05/2018] [Indexed: 06/09/2023]
Abstract
The Korean CHildren's ENvironmental health Study (Ko-CHENS) is a nationwide prospective birth cohort showing the correlation between the environmental exposures and the health effects to prevent the environmental diseases in children, and it provides the guidelines for the environmental hazardous factors, applying the life-course approach to the environmental-health management system. The Ko-CHENS consists of 5000 Core and 65,000 Main Cohorts. The children in the Core Cohort are followed up at 6 months, every year before their admission into the elementary school, and every 3 years from the first year after this admission. The children in the Cohort will be followed up through the data links (Statistics Korea, National Health Insurance Service [NHIS], and Ministry of Education). The individual biospecimens will be analyzed for 19 substances. The long-term-storage biological samples will be used for the further substance analysis. The Ko-CHENS will investigate whether the environmental variables including the perinatal outdoor and indoor factors and the greenness contribute causally to the health outcomes in the children and adolescents. In addition to the individual surveys, the assessments of the outdoor exposures and health outcomes will use the national air-quality monitoring data and claim data of the NHIS, respectively. The two big-data forms of the Ko-CHENS are as follows: The Ko-CHENS data that can be linked with the nationally registered NHIS health-related database, including the medical utilization and the periodic health screening, and the birth/mortality database in the Statistics; the other is the Big-CHENS dataset that is based on the NHIS mother delivery code, for which the follow-up of almost 97% of the total birth population is expected. The Ko-CHENS is a very cost-effective study that fully exploits the existing national big-data systems with the data linkage.
Collapse
Affiliation(s)
- Kyoung Sook Jeong
- Department of Occupational and Environmental Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Suejin Kim
- Environmental Health Research Division, Department of Environmental Health Research, National Institute of Environmental Research, Ministry of Environment, Incheon, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University, Chuncheon, Republic of Korea
| | - Hwan-Cheol Kim
- Department of Occupational and Environmental Medicine, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jisuk Bae
- Department of Preventive Medicine, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - Yun-Chul Hong
- Institute of Environmental Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Mina Ha
- Department of Preventive Medicine, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Environmental Health Center for Atopic Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji-Young Lee
- Department of Occupational and Environmental Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea.
| | - Eunhee Ha
- Department of Occupational and Environmental Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Zakarya R, Adcock I, Oliver BG. Epigenetic impacts of maternal tobacco and e-vapour exposure on the offspring lung. Clin Epigenetics 2019; 11:32. [PMID: 30782202 PMCID: PMC6381655 DOI: 10.1186/s13148-019-0631-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
In utero exposure to tobacco products, whether maternal or environmental, have harmful effects on first neonatal and later adult respiratory outcomes. These effects have been shown to persist across subsequent generations, regardless of the offsprings' smoking habits. Established epigenetic modifications induced by in utero exposure are postulated as the mechanism underlying the inherited poor respiratory outcomes. As e-cigarette use is on the rise, their potential to induce similar functional respiratory deficits underpinned by an alteration in the foetal epigenome needs to be explored. This review will focus on the functional and epigenetic impact of in utero exposure to maternal cigarette smoke, maternal environmental tobacco smoke, environmental tobacco smoke and e-cigarette vapour on foetal respiratory outcomes.
Collapse
Affiliation(s)
- Razia Zakarya
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Ian Adcock
- Airway Diseases Section, National Heart and Lung Institute, Imperial College London, London, UK
- Biomedical Research Unit, Section of Respiratory Diseases, Royal Brompton and Harefield NHS Trust, London, UK
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia.
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
| |
Collapse
|
24
|
Murrison LB, Brandt EB, Myers JB, Hershey GKK. Environmental exposures and mechanisms in allergy and asthma development. J Clin Invest 2019; 129:1504-1515. [PMID: 30741719 DOI: 10.1172/jci124612] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Environmental exposures interplay with human host factors to promote the development and progression of allergic diseases. The worldwide prevalence of allergic disease is rising as a result of complex gene-environment interactions that shape the immune system and host response. Research shows an association between the rise of allergic diseases and increasingly modern Westernized lifestyles, which are characterized by increased urbanization, time spent indoors, and antibiotic usage. These environmental changes result in increased exposure to air and traffic pollution, fungi, infectious agents, tobacco smoke, and other early-life and lifelong risk factors for the development and exacerbation of asthma and allergic diseases. It is increasingly recognized that the timing, load, and route of allergen exposure affect allergic disease phenotypes and development. Still, our ability to prevent allergic diseases is hindered by gaps in understanding of the underlying mechanisms and interaction of environmental, viral, and allergen exposures with immune pathways that impact disease development. This Review highlights epidemiologic and mechanistic evidence linking environmental exposures to the development and exacerbation of allergic airway responses.
Collapse
Affiliation(s)
- Liza Bronner Murrison
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric B Brandt
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA
| | - Jocelyn Biagini Myers
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
25
|
Arah OA. Commentary: Tobacco smoking and asthma: multigenerational effects, epigenetics and multilevel causal mediation analysis. Int J Epidemiol 2019; 47:1117-1119. [PMID: 30184137 DOI: 10.1093/ije/dyy193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Onyebuchi A Arah
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,Center for Health Policy Research, UCLA Fielding School of Public Health, Los Angeles, CA, USA.,California Center for Population Research, Los Angeles, CA, USA.,Department of Statistics, UCLA College of Letters and Science, Los Angeles, CA, USA
| |
Collapse
|
26
|
Qu W, Zhao WH, Wen X, Yan HY, Liu HX, Hou LF, Ping J. Prenatal nicotine exposure induces thymic hypoplasia in mice offspring from neonatal to adulthood. Toxicol Lett 2018; 304:30-38. [PMID: 30605750 DOI: 10.1016/j.toxlet.2018.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/29/2018] [Accepted: 12/28/2018] [Indexed: 12/24/2022]
Abstract
Clinical study showed that smoking during pregnancy deceased the thymus size in newborns. However, the long-term effect remains unclear. This study was aimed to observe the effects of prenatal nicotine exposure (PNE) on the development of thymus and the T-lymphocyte subpopulation in mice offspring from the neonatal to adulthood. Both the thymus weight and cytometry data indicated that PNE caused persistent thymic hypoplasia in male offspring from neonatal to adult period and transient changes in female offspring from neonatal to prepuberal period. Flow cytometry analysis disclosed a permanent decreased proportion and number of mature CD4 single-positive (SP) T cells in thymus of both sex. In addition, the PNE male offspring showed a more serious thymus atrophy in the ovalbumin (OVA)-sensitized model. Moreover, increased autophagic vacuole and elevated mRNA expression of Beclin 1 were noted in PNE fetal thymus. In conclusion, PNE offspring showed thymus atrophy and CD 4 SP T cell reduction at different life stages. Mechanically, PNE induced excessive autophagy in fetal thymocytes might be involved in these changes. All the results provided evidence for elucidating the PNE-induced programmed immune diseases.
Collapse
Affiliation(s)
- Wen Qu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China.
| | - Wen-Hao Zhao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Xiao Wen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hui-Yi Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Han-Xiao Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Li-Fang Hou
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Jie Ping
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
27
|
Yu Y, Wang L, Gu G. The correlation between Runx3 and bronchial asthma. Clin Chim Acta 2018; 487:75-79. [PMID: 30218658 DOI: 10.1016/j.cca.2018.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
Abstract
Runx3, a member of the Runt-related transcription factor family, has attracted extensive attention due to its important role in the development of immune systems, especially in the differentiation of T cells. Accumulated evidence indicated that altered expression of Runx3 regulates a variety of target genes in different tissues/cells. Studies in animal models suggested that Runx3 may regulate the development of T cell lineage including those of innate lymphoid cells, Treg cells and dendritic cells, which may contribute to the development of hypersensitivity and asthma. Specifically, Runx3 modulates Th1/Th2 balance and hence, the production of interleukins, which induce inflammatory responses. Understanding the roles and mechanisms of Runx3 in the regulation of immune function provides a basis for the design of novel preventive and treatment models for bronchial asthma. This article reviews published data from cell lines, animal models, and patients, concerning the relationship between Runx3 expression alteration and asthma. Epigenetic regulation of Runx3 by DNA hypermethylation and microRNA, and the implication of these pathways in asthma are also discussed.
Collapse
Affiliation(s)
- Yanyan Yu
- The children's hospital affiliated of Suzhou University, Suzhou 215000, Jiangsu Province, China.
| | - Leilei Wang
- Children Asthma Department, Lianyungang Maternal and Child Hospital Jiangsu Province, Lianyungang 222006, Jiangsu Province, China
| | - Guixiong Gu
- The children's hospital affiliated of Suzhou University, Suzhou 215000, Jiangsu Province, China.
| |
Collapse
|
28
|
Fonseca W, Lukacs NW, Ptaschinski C. Factors Affecting the Immunity to Respiratory Syncytial Virus: From Epigenetics to Microbiome. Front Immunol 2018. [PMID: 29515570 PMCID: PMC5825926 DOI: 10.3389/fimmu.2018.00226] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common pathogen that infects virtually all children by 2 years of age and is the leading cause of hospitalization of infants worldwide. While most children experience mild symptoms, some children progress to severe lower respiratory tract infection. Those children with severe disease have a much higher risk of developing childhood wheezing later in life. Many risk factors are known to result in exacerbated disease, including premature birth and early age of RSV infection, when the immune system is relatively immature. The development of the immune system before and after birth may be altered by several extrinsic and intrinsic factors that could lead to severe disease predisposition in children who do not exhibit any currently known risk factors. Recently, the role of the microbiome and the resulting metabolite profile has been an area of intense study in the development of lung disease, including viral infection and asthma. This review explores both known risk factors that can lead to severe RSV-induced disease as well as emerging topics in the development of immunity to RSV and the long-term consequences of severe infection.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States.,University of Michigan, Mary H. Weiser Food Allergy Center, Ann Arbor, MI, United States
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States.,University of Michigan, Mary H. Weiser Food Allergy Center, Ann Arbor, MI, United States
| |
Collapse
|