1
|
Zimmermann EJ, Das A, Huber A, Gawlitta N, Kuhn E, Schlager C, Gutmann B, Krebs T, Schnelle-Kreis J, Delaval MN, Zimmermann R. Toxicological effects of long-term continuous exposure to ambient air on human bronchial epithelial Calu-3 cells exposed at the air-liquid interface. ENVIRONMENTAL RESEARCH 2025; 269:120759. [PMID: 39755196 DOI: 10.1016/j.envres.2025.120759] [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: 09/11/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/06/2025]
Abstract
Air pollution significantly contributes to the global burden of respiratory and cardiovascular diseases. While single source/compound studies dominate current research, long-term, multi-pollutant studies are crucial to understanding the health impacts of environmental aerosols. Our study aimed to use the first air-liquid interface (ALI) aerosol exposure system adapted for long-term in vitro exposures for ambient air in vitro exposure. The automated exposure system was adapted to enable long-term cell exposure. ALI human bronchial epithelial cells (Calu-3) were continuously exposed for 72 h to the ambient air from a European urban area (3 independent exposures). Experimental evaluation included comprehensive toxicological assessments coupled to physical and chemical characterization of the aerosol. Exposure to ambient air resulted in increased significant cytotoxicity and a non-significant decrease in cell viability. Differential gene expressions were indicated for genes related to inflammation (IL1B, IL6) and to xenobiotic metabolism (CYP1A1, CYP1B1) with possible correlations to the PM2.5 content. Common air pollutants were identified such as the carcinogenic benz[a]pyrene (≤3.4 ng m-3/24h) and PM2.5 (≤11.6 μg m-3/24h) with a maximum particle number mean of 4.4 × 10-3 m3/24h. For the first time, ALI human lung epithelial cells were exposed for 72 h to continuous airflow of ambient air. Despite direct exposure to ambient aerosols, only small decrease in cell viability and gene expression changes was observed. We propose this experimental set-up combining comprehensive aerosol characterization and long-term continuous ALI cell exposure for the identification of hazardous compounds or compound mixtures in ambient air.
Collapse
Affiliation(s)
- E J Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, 18051, Germany
| | - A Das
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, 18051, Germany
| | - A Huber
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, 18051, Germany
| | - N Gawlitta
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany
| | - E Kuhn
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany
| | - C Schlager
- Vitrocell Systems GmbH, 79183, Waldkirch, Germany
| | - B Gutmann
- Vitrocell Systems GmbH, 79183, Waldkirch, Germany
| | - T Krebs
- Vitrocell Systems GmbH, 79183, Waldkirch, Germany
| | - J Schnelle-Kreis
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany
| | - M N Delaval
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany.
| | - R Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Munich, 85764, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, 18051, Germany
| |
Collapse
|
2
|
Hamlington KL, Liu AH. Airway inflammation, asthma, and soot. Pediatr Res 2025; 97:13-14. [PMID: 39181983 DOI: 10.1038/s41390-024-03447-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 07/13/2024] [Indexed: 08/27/2024]
Affiliation(s)
- Katharine L Hamlington
- Section of Pulmonary and Sleep Medicine, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andrew H Liu
- Section of Pulmonary and Sleep Medicine, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
3
|
Le Souëf PN, Adachi Y, Anastasiou E, Ansotegui IJ, Badellino HA, Banzon T, Beltrán CP, D'Amato G, El-Sayed ZA, Gómez RM, Hossny E, Kalayci Ö, Morais-Almeida M, Nieto-Garcia A, Peden DB, Phipatanakul W, Wang JY, Wan IJ, Wong G, Xepapadaki P, Papadopoulos NG. Global change, climate change, and asthma in children: Direct and indirect effects - A WAO Pediatric Asthma Committee Report. World Allergy Organ J 2024; 17:100988. [PMID: 39582513 PMCID: PMC11584610 DOI: 10.1016/j.waojou.2024.100988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/21/2024] [Accepted: 10/05/2024] [Indexed: 11/26/2024] Open
Abstract
The twenty-first century has seen a fundamental shift in disease epidemiology with anthropogenic environmental change emerging as the likely dominant factor affecting the distribution and severity of current and future human disease. This is especially true of allergic diseases and asthma with their intimate relationship with the natural environment. Climate change-related variables including increased ambient temperature, heat waves, extreme weather events, air pollution, and rainfall distribution, all can directly affect asthma in children, but each of these variables also indirectly affects asthma via alterations in pollen production and release, outdoor allergen exposure or the microbiome. Air pollution, with its many and varied respiratory consequences, is likely to have the greatest effect, as it has increased globally due to rapid increases in fossil fuel combustion, global population, crowding, and megacities, as well as forest burning and trees succumbing to an increasingly hostile environment. Human activities have also caused substantial deterioration of the global microbiome with reductions in biodiversity for molds, bacteria, and viruses. Reduced microbiome diversity has, in turn, been associated with increases in Th2 allergic responses and allergic disease. The collective effect of these changes has already shifted allergy and asthma disease patterns. Given that changes in climate have been relatively small to date, the unavoidable, much greater shifts in climate in the future are concerning. Determining the relative scale of the direct versus indirect effects of climate change variables is needed if effective avoidance and adaptive measures are to be implemented. This would also require much more basic, epidemiological, and clinical research to understand the causal mechanisms, the most relevant climate factors involved, the regions most affected and, most importantly, effective and actionable adaptation measures. We suggest that allergy and respiratory health workers should follow current guidance to reduce present risks related to climate change and watch for new recommendations to reduce future risks. Since the respiratory system is the one most affected by climate change, they also need to call for more research in this area and show strong leadership in advocating for urgent action to protect children by reducing or reversing factors that have led to our deteriorating climate.
Collapse
Affiliation(s)
- Peter N. Le Souëf
- School of Medicine, University of Western Australia and Telethon Kids Institute, Perth, Australia
| | - Yuichi Adachi
- Pediatric Allergy Center, Toyama Red Cross Hospital, Toyama, Japan
| | - Eleni Anastasiou
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Tina Banzon
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cesar Pozo Beltrán
- Pediatric Allergy and Immunology, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Gennaro D'Amato
- Division of Respiratory Diseases and Allergy, High Specialty Hospital A.Cardarelli, Naples and School of Specialization in Respiratory Disease, University of Naples Federico II, Italy
| | - Zeinab A. El-Sayed
- Pediatric Allergy, Immunology, And Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | | | - Elham Hossny
- Pediatric Allergy, Immunology, And Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Ömer Kalayci
- Hacettepe University School of Medicine, Ankara, Turkey
| | | | - Antonio Nieto-Garcia
- Pediatric Pulmonology and Allergy Unit, Hospital Universitari i Politècnic La Fe, Health Research Institute La Fe, Valencia, Spain
| | - David B. Peden
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Environmental Medicine, Asthma and Lung Biology Division of Pediatric Allergy & Immunology, The School of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA
| | - Wanda Phipatanakul
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiu-Yao Wang
- Allergy, Immunology and Microbiome Research Center, China Medical University Children's Hospital, Taichung, Taiwan
| | - I-Jen Wan
- Department of Pediatrics, Taipei Hospital Ministry of Health and Welfare, School of Medicine, National Yang Ming Chiao Tung University, China Medical University, Taiwan
| | - Gary Wong
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Paraskevi Xepapadaki
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G. Papadopoulos
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Santoro M, Costabile F, Gualtieri M, Rinaldi M, Paglione M, Busetto M, Di Iulio G, Di Liberto L, Gherardi M, Pelliccioni A, Monti P, Barbara B, Grollino MG. Associations between fine particulate matter, gene expression, and promoter methylation in human bronchial epithelial cells exposed within a classroom under air-liquid interface. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 358:124471. [PMID: 38950846 DOI: 10.1016/j.envpol.2024.124471] [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: 02/28/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Associations between indoor air pollution from fine particulate matter (PM with aerodynamic diameter dp < 2.5 μm) and human health are poorly understood. Here, we analyse the concentration-response curves for fine and ultrafine PM, the gene expression, and the methylation patterns in human bronchial epithelial cells (BEAS-2B) exposed at the air-liquid interface (ALI) within a classroom in downtown Rome. Our results document the upregulation of aryl hydrocarbon receptor (AhR) and genes associated with xenobiotic metabolism (CYP1A1 and CYP1B1) in response to single exposure of cells to fresh urban aerosols at low fine PM mass concentrations within the classroom. This is evidenced by concentrations of ultrafine particles (UFPs, dp < 0.1 μm), polycyclic aromatic hydrocarbons (PAH), and ratios of black carbon (BC) to organic aerosol (OA). Additionally, an interleukin 18 (IL-18) down-regulation was found during periods of high human occupancy. Despite the observed gene expression dysregulation, no changes were detected in the methylation levels of the promoter regions of these genes, indicating that the altered gene expression is not linked to changes in DNA methylation and suggesting the involvement of another epigenetic mechanism in the gene regulation. Gene expression changes at low exposure doses have been previously reported. Here, we add the possibility that lung epithelial cells, when singly exposed to real environmental concentrations of fine PM that translate into ultra-low doses of treatment, may undergo epigenetic alteration in the expression of genes related to xenobiotic metabolism. Our findings provide a perspective for future indoor air quality regulations. We underscore the potential role of indoor UFPs as carriers of toxic molecules with low-pressure weather conditions, when rainfall and strong winds may favour low levels of fine PM.
Collapse
Affiliation(s)
- Massimo Santoro
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Development (ENEA), 00123, Rome, Italy
| | - Francesca Costabile
- Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Fosso del Cavaliere, 00133, Rome, Italy; NBFC - National Biodiversity Future Center, NBFC, 90133, Palermo, Italy.
| | - Maurizio Gualtieri
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126, Milan, Italy
| | - Matteo Rinaldi
- NBFC - National Biodiversity Future Center, NBFC, 90133, Palermo, Italy; Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Gobetti, 40129, Bologna, Italy
| | - Marco Paglione
- NBFC - National Biodiversity Future Center, NBFC, 90133, Palermo, Italy; Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Gobetti, 40129, Bologna, Italy
| | - Maurizio Busetto
- Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Gobetti, 40129, Bologna, Italy
| | - Gianluca Di Iulio
- Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Fosso del Cavaliere, 00133, Rome, Italy; Department of Public Health and Infectious Disease - University of Rome "La Sapienza", via Eudossiana 18, 00184, Rome, Italy
| | - Luca Di Liberto
- Institute of Atmospheric Sciences and Climate - Italian National Research Council (ISAC - CNR), Via Fosso del Cavaliere, 00133, Rome, Italy
| | - Monica Gherardi
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, 00078, Rome, Italy
| | - Armando Pelliccioni
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, 00078, Rome, Italy
| | - Paolo Monti
- Department of Civil, Building and Environmental Engineering - University of Rome "La Sapienza", via Eudossiana 18, 00184, Rome, Italy
| | - Benassi Barbara
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Development (ENEA), 00123, Rome, Italy
| | - Maria Giuseppa Grollino
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Development (ENEA), 00123, Rome, Italy
| |
Collapse
|
5
|
Li S, Huff RD, Rider CF, Yuen ACY, Carlsten C. Controlled human exposures to diesel exhaust or particle-depleted diesel exhaust with allergen modulates transcriptomic responses in the lung. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 945:173688. [PMID: 38851342 DOI: 10.1016/j.scitotenv.2024.173688] [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: 02/26/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
The evidence associating traffic-related air pollution (TRAP) with allergic asthma is growing, but the underlying mechanisms for this association remain unclear. The airway epithelium is the primary tissue exposed to TRAP, hence understanding its interactions with TRAP and allergen is important. Diesel exhaust (DE), a paradigm of TRAP, consists of particulate matter (PM) and gases. Modern diesel engines often have catalytic diesel particulate filters to reduce PM output, but these may increase gaseous concentrations, and their benefits on human health cannot be assumed. We conducted a randomized, double-blinded, crossover study using our unique in vivo human exposure system to investigate the effects of DE and allergen co-exposure, with or without particle depletion as a proxy for catalytic diesel particulate filters, on the airway epithelial transcriptome. Participants were exposed for 2 h before an allergen inhalation challenge, with each receiving filtered air and saline (FA-S), filtered air and allergen (FA-A), DE and allergen (DE-A), or particle-depleted DE and allergen (PDDE-A), over four different occasions, each separated by a 4-week washout period. Endobronchial brushings were collected 48 h after each exposure, and total RNA was sequenced. Differentially expressed genes (DEGs) were identified using DESeq2, followed by GO enrichment and pathway analysis. FA-A, DE-A, and PDDE-A exposures significantly modulated genes relative to FA-S, with 462 unique DEGs identified. FA-A uniquely modulated the highest number (↑178, ↓155), followed by DE-A (↑44, ↓23), and then PDDE-A exposure (↑15, ↓2); 6 DEGs (↑4, ↓2) were modulated by all three conditions. Exposure to PDDE-A resulted in modulation of 285 DEGs compared to DE-A exposure, further revealing 26 biological process GO terms, including "cellular response to chemokine" and "inflammatory response". The transcriptional epithelial response to diesel exhaust and allergen co-exposure is enriched in inflammatory mediators, the pattern of which is altered upon particle depletion.
Collapse
Affiliation(s)
- Shijia Li
- Air Pollution Exposure Laboratory (APEL), Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Ryan D Huff
- Air Pollution Exposure Laboratory (APEL), Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Christopher F Rider
- Air Pollution Exposure Laboratory (APEL), Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Agnes C Y Yuen
- Air Pollution Exposure Laboratory (APEL), Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Chris Carlsten
- Air Pollution Exposure Laboratory (APEL), Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
6
|
Lukyanchuk A, Muraki N, Kawai T, Sato T, Hata K, Ito T, Tajima A. Long-term exposure to diesel exhaust particles induces concordant changes in DNA methylation and transcriptome in human adenocarcinoma alveolar basal epithelial cells. Epigenetics Chromatin 2024; 17:24. [PMID: 39103936 DOI: 10.1186/s13072-024-00549-3] [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: 03/08/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Diesel exhaust particles (DEP), which contain hazardous compounds, are emitted during the combustion of diesel. As approximately one-third of the vehicles worldwide use diesel, there are growing concerns about the risks posed by DEP to human health. Long-term exposure to DEP is associated with airway hyperresponsiveness, pulmonary fibrosis, and inflammation; however, the molecular mechanisms behind the effects of DEP on the respiratory tract are poorly understood. Such mechanisms can be addressed by examining transcriptional and DNA methylation changes. Although several studies have focused on the effects of short-term DEP exposure on gene expression, research on the transcriptional effects and genome-wide DNA methylation changes caused by long-term DEP exposure is lacking. Hence, in this study, we investigated transcriptional and DNA methylation changes in human adenocarcinoma alveolar basal epithelial A549 cells caused by prolonged exposure to DEP and determined whether these changes are concordant. RESULTS DNA methylation analysis using the Illumina Infinium MethylationEPIC BeadChips showed that the methylation levels of DEP-affected CpG sites in A549 cells changed in a dose-dependent manner; the extent of change increased with increasing dose reaching the statistical significance only in samples exposed to 30 µg/ml DEP. Four-week exposure to 30 µg/ml of DEP significantly induced DNA hypomethylation at 24,464 CpG sites, which were significantly enriched for DNase hypersensitive sites, genomic regions marked by H3K4me1 and H3K27ac, and several transcription factor binding sites. In contrast, 9,436 CpG sites with increased DNA methylation levels were significantly overrepresented in genomic regions marked by H3K27me3 as well as H3K4me1 and H3K27ac. In parallel, gene expression profiling by RNA sequencing demonstrated that long-term exposure to DEP altered the expression levels of 2,410 genes, enriching 16 gene sets including Xenobiotic metabolism, Inflammatory response, and Senescence. In silico analysis revealed that the expression levels of 854 genes correlated with the methylation levels of the DEP-affected cis-CpG sites. CONCLUSIONS To our knowledge, this is the first report of genome-wide transcriptional and DNA methylation changes and their associations in A549 cells following long-term exposure to DEP.
Collapse
Affiliation(s)
- Alexandra Lukyanchuk
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Krasnoyarsk State Medical University Named After Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Naomi Muraki
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Department of Human Biology and Anatomy, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tsuyoshi Ito
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
7
|
Han R, Zhu D, Sha J, Zhao B, Jin P, Meng C. Decoding the role of DNA methylation in allergic diseases: from pathogenesis to therapy. Cell Biosci 2024; 14:89. [PMID: 38965641 PMCID: PMC11225420 DOI: 10.1186/s13578-024-01270-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024] Open
Abstract
Allergic diseases, characterized by a broad spectrum of clinical manifestations and symptoms, encompass a significant category of IgE-mediated atopic disorders, including asthma, allergic rhinitis, atopic dermatitis, and food allergies. These complex conditions arise from the intricate interplay between genetic and environmental factors and are known to contribute to socioeconomic burdens globally. Recent advancements in the study of allergic diseases have illuminated the crucial role of DNA methylation (DNAm) in their pathogenesis. This review explores the factors influencing DNAm in allergic diseases and delves into their mechanisms, offering valuable perspectives for clinicians. Understanding these epigenetic modifications aims to lay the groundwork for improved early prevention strategies. Moreover, our analysis of DNAm mechanisms in these conditions seeks to enhance diagnostic and therapeutic approaches, paving the way for more effective management of allergic diseases in the future.
Collapse
Affiliation(s)
- Ruiming Han
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dongdong Zhu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, Changchun, China
| | - Jichao Sha
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, Changchun, China
| | - Boning Zhao
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, 615 Michael ST NE, Atlanta, GA, 30322, USA.
| | - Cuida Meng
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, Changchun, China.
| |
Collapse
|
8
|
Berni Canani R, Caminati M, Carucci L, Eguiluz-Gracia I. Skin, gut, and lung barrier: Physiological interface and target of intervention for preventing and treating allergic diseases. Allergy 2024; 79:1485-1500. [PMID: 38439599 DOI: 10.1111/all.16092] [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: 09/05/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024]
Abstract
The epithelial barriers of the skin, gut, and respiratory tract are critical interfaces between the environment and the host, and they orchestrate both homeostatic and pathogenic immune responses. The mechanisms underlying epithelial barrier dysfunction in allergic and inflammatory conditions, such as atopic dermatitis, food allergy, eosinophilic oesophagitis, allergic rhinitis, chronic rhinosinusitis, and asthma, are complex and influenced by the exposome, microbiome, individual genetics, and epigenetics. Here, we review the role of the epithelial barriers of the skin, digestive tract, and airways in maintaining homeostasis, how they influence the occurrence and progression of allergic and inflammatory conditions, how current treatments target the epithelium to improve symptoms of these disorders, and what the unmet needs are in the identification and treatment of epithelial disorders.
Collapse
Affiliation(s)
- Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Marco Caminati
- Allergy Unit and Asthma Centre, Verona Integrated University Hospital and Department of Medicine, University of Verona, Verona, Italy
| | - Laura Carucci
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Ibon Eguiluz-Gracia
- Allergy Unit, Hospital Regional Universitario de Malága, Malaga, Spain
- Allergy Group, Biomedical Research Institute of Malaga (IBIMA)-BIONAND Platform, RICORS Inflammatory Diseases, Malaga, Spain
| |
Collapse
|
9
|
Papadopoulos NG, Akdis CA, Akdis M, Damialis A, Esposito G, Fergadiotou I, Goroncy C, Guitton P, Gotua M, Erotokritou K, Jartti T, Murray C, Nenes A, Nikoletseas S, Finotto S, Pandis SN, Ramiconi V, Simpson A, Soudunsaari A, Stårbröst A, Staiano M, Varriale A, Xepapadaki P, Zuberbier T, Annesi-Maesano I. Addressing adverse synergies between chemical and biological pollutants at schools-The 'SynAir-G' hypothesis. Allergy 2024; 79:294-301. [PMID: 37654007 DOI: 10.1111/all.15857] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023]
Abstract
While the number and types of indoor air pollutants is rising, much is suspected but little is known about the impact of their potentially synergistic interactions, upon human health. Gases, particulate matter, organic compounds but also allergens and viruses, fall within the 'pollutant' definition. Distinct populations, such as children and allergy and asthma sufferers are highly susceptible, while a low socioeconomic background is a further susceptibility factor; however, no specific guidance is available. We spend most of our time indoors; for children, the school environment is of paramount importance and potentially amenable to intervention. The interactions between some pollutant classes have been studied. However, a lot is missing with respect to understanding interactions between specific pollutants of different classes in terms of concentrations, timing and sequence, to improve targeting and upgrade standards. SynAir-G is a European Commission-funded project aiming to reveal and quantify synergistic interactions between different pollutants affecting health, from mechanisms to real life, focusing on the school setting. It will develop a comprehensive and responsive multipollutant monitoring system, advance environmentally friendly interventions, and disseminate the generated knowledge to relevant stakeholders in accessible and actionable formats. The aim of this article it to put forward the SynAir-G hypothesis, and describe its background and objectives.
Collapse
Affiliation(s)
- Nikolaos G Papadopoulos
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - 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
| | - Mubeccel 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
| | - Athanasios Damialis
- Department of Ecology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | | | | | - Maia Gotua
- Center for Allergy and Immunology Research (CAIR), Tbilisi, Georgia
| | | | - Tuomas Jartti
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Department of Pediatrics, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Clare Murray
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Athanasios Nenes
- Laboratory of Atmospheric Processes and their Impacts, School of Architecture, Civil and Environmental Engineering, Swiss Institute of Technology, Lausanne, Switzerland
| | - Sotirios Nikoletseas
- Computer Engineering and Informatics Department, University of Patras, Patras, Greece
| | - Susetta Finotto
- Molecular Pneumology Department, University Hospital of Erlangen, Erlangen, Germany
| | - Spyros N Pandis
- Institute of Chemical Engineering Sciences (ICEHT), Foundation for Research and Technology Hellas (FORTH), Patras, Greece
| | - Valeria Ramiconi
- The European Federation of Allergy and Airways Diseases Patients' Association (EFA), Brussels, Belgium
| | - Angela Simpson
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK
| | | | | | - Maria Staiano
- Institute of Food Science, CNR Italy, Avellino, Italy
| | - Antonio Varriale
- Institute of Food Science, CNR Italy, Avellino, Italy
- URT-ISA, CNR at Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paraskevi Xepapadaki
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Torsten Zuberbier
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
- Global Allergy & Asthma European Network of Excellence-GA2LEN, Berlin, Germany
| | - Isabella Annesi-Maesano
- Department of Allergic and Respiratory Disease, Institut Desbrest of Epidemiology and Public Health, University of Montpellier and INSERM, Montpellier University Hospital, Montpellier, France
| |
Collapse
|
10
|
Cochrane KM, Elango R, Devlin AM, Mayer C, Hutcheon JA, Karakochuk CD. Supplementation with (6 S)-5-methyltetrahydrofolic acid appears as effective as folic acid in maintaining maternal folate status while reducing unmetabolised folic acid in maternal plasma: a randomised trial of pregnant women in Canada. Br J Nutr 2024; 131:92-102. [PMID: 37649241 DOI: 10.1017/s0007114523001733] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Folic acid supplementation is recommended during pregnancy to support healthy fetal development; (6S)-5-methyltetrahydrofolic acid ((6S)-5-MTHF) is available in some commercial prenatal vitamins as an alternative to folic acid, but its effect on blood folate status during pregnancy is unknown. To address this, we randomised sixty pregnant individuals at 8-21 weeks' gestation to 0·6 mg/d folic acid or (6S)-5-MTHF × 16 weeks. Fasting blood specimens were collected at baseline and after 16 weeks (endline). Erythrocyte and serum folate were quantified via microbiological assay (as globally recommended) and plasma unmetabolised folic acid (UMFA) via LC-MS/MS. Differences in biochemical folate markers between groups were explored using multivariable linear/quantile regression, adjusting for baseline concentrations, dietary folate intake and gestational weeks. At endline (n 54), the mean values and standard deviations (or median, inter-quartile range) of erythrocyte folate, serum folate and plasma UMFA (nmol/l) in those supplemented with (6S)-5-MTHF v. folic acid, respectively, were 1826 (sd 471) and 1998 (sd 421); 70 (sd 13) and 78 (sd 17); 0·5 (0·4, 0·8) and 1·3 (0·9, 2·1). In regression analyses, erythrocyte and serum folate did not differ by treatment group; however, concentrations of plasma UMFA in pregnancy were 0·6 nmol/l higher (95 % CI 0·2, 1·1) in those supplementing with folic acid as compared with (6S)-5-MTHF. In conclusion, supplementation with (6S)-5-MTHF may reduce plasma UMFA by ∼50 % as compared with supplementation with folic acid, the biological relevance of which is unclear. As folate is currently available for purchase in both forms, the impact of circulating maternal UMFA on perinatal outcomes needs to be determined.
Collapse
Affiliation(s)
- Kelsey M Cochrane
- Food, Nutrition, and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Healthy Starts, Vancouver, Canada
| | - Rajavel Elango
- BC Children's Hospital Research Institute, Healthy Starts, Vancouver, Canada
- Population and Public Health, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Angela M Devlin
- BC Children's Hospital Research Institute, Healthy Starts, Vancouver, Canada
- Paediatrics, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Chantal Mayer
- Obstetrics and Gynaecology, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Jennifer A Hutcheon
- BC Children's Hospital Research Institute, Healthy Starts, Vancouver, Canada
- Obstetrics and Gynaecology, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Crystal D Karakochuk
- Food, Nutrition, and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Healthy Starts, Vancouver, Canada
| |
Collapse
|
11
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, Proteomic, and Metabolomic Correlates of Traffic-Related Air Pollution in the Context of Cardiorespiratory Health: A Systematic Review, Pathway Analysis, and Network Analysis. TOXICS 2023; 11:1014. [PMID: 38133415 PMCID: PMC10748071 DOI: 10.3390/toxics11121014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead to cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
12
|
Singh N, Nagar E, Gautam A, Kapoor H, Arora N. Resveratrol mitigates miR-212-3p mediated progression of diesel exhaust-induced pulmonary fibrosis by regulating SIRT1/FoxO3. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:166063. [PMID: 37544448 DOI: 10.1016/j.scitotenv.2023.166063] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/19/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Diesel exhaust (DE) exposure contributes to the progression of chronic respiratory diseases and is associated with dysregulation of microRNA expression. The present study aims to investigate the involvement of miRNAs and target genes in DE-induced lung fibrosis. METHODS C57BL/6 mice were divided into three groups. Group 1 mice were exposed to filtered air (Control). Group 2 mice were exposed to DE for 30 min per day, 5 days per week, for 8 weeks (DE). Group 3 mice received DE exposure along with resveratrol on alternate days for the last 2 weeks (DE + RES). Mice were sacrificed to isolate RNA from lung tissue for miRNA microarray profiling. Bronchoalveolar lavage fluid and lung tissues were collected for cell count and biochemical analysis. RESULTS DE exposure resulted in differential expression of 28 miRNAs with fold change >2 (p < 0.05). The upregulated miR-212-3p was selected for further analysis. Consensus analysis revealed enrichment of SIRT1 in the FoxO pathway, along with a co-annotation of reduced body weight (p < 0.05). A549 cells transfected with a miR-212-3p inhibitor showed a dose-dependent increase in SIRT1 expression, indicating SIRT1 as a direct target. Treatment with resveratrol restored SIRT1 and miR-212-3p expression and led to a reduction in inflammatory cytokines (p < 0.05). The modulation of SIRT1 correlated negatively with macrophage infiltration, confirming its role in regulating cellular infiltration and lung inflammation. Fibronectin, alpha-SMA, and collagen levels were significantly decreased in DE + RES compared to DE group suggesting modulation of cellular functions and resolution of lung fibrosis. Furthermore, a significant decrease in FoxO3a and TGF-β gene expressions was observed upon resveratrol administration thereby downregulating pro-fibrotic pathway. CONCLUSIONS The present study demonstrates resveratrol treatment stabilizes SIRT1 gene expression by attenuating miR-212-3p in DE-exposed mice, leading to downregulation of TGF-β and FoxO3a expressions. The study highlights the therapeutic role of resveratrol in the treatment of DE-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Naresh Singh
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ekta Nagar
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anshu Gautam
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Himanshi Kapoor
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Naveen Arora
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, proteomic, and metabolomic correlates of traffic-related air pollution: A systematic review, pathway analysis, and network analysis relating traffic-related air pollution to subclinical and clinical cardiorespiratory outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.30.23296386. [PMID: 37873294 PMCID: PMC10592990 DOI: 10.1101/2023.09.30.23296386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease, and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
14
|
Messingschlager M, Bartel-Steinbach M, Mackowiak SD, Denkena J, Bieg M, Klös M, Seegebarth A, Straff W, Süring K, Ishaque N, Eils R, Lehmann I, Lermen D, Trump S. Genome-wide DNA methylation sequencing identifies epigenetic perturbations in the upper airways under long-term exposure to moderate levels of ambient air pollution. ENVIRONMENTAL RESEARCH 2023; 233:116413. [PMID: 37343754 DOI: 10.1016/j.envres.2023.116413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/30/2023] [Accepted: 06/11/2023] [Indexed: 06/23/2023]
Abstract
While the link between exposure to high levels of ambient particulate matter (PM) and increased incidences of respiratory and cardiovascular diseases is widely recognized, recent epidemiological studies have shown that low PM concentrations are equally associated with adverse health effects. As DNA methylation is one of the main mechanisms by which cells regulate and stabilize gene expression, changes in the methylome could constitute early indicators of dysregulated signaling pathways. So far, little is known about PM-associated DNA methylation changes in the upper airways, the first point of contact between airborne pollutants and the human body. Here, we focused on cells of the upper respiratory tract and assessed their genome-wide DNA methylation pattern to explore exposure-associated early regulatory changes. Using a mobile epidemiological laboratory, nasal lavage samples were collected from a cohort of 60 adults that lived in districts with records of low (Simmerath) or moderate (Stuttgart) PM10 levels in Germany. PM10 concentrations were verified by particle measurements on the days of the sample collection and genome-wide DNA methylation was determined by enzymatic methyl sequencing at single-base resolution. We identified 231 differentially methylated regions (DMRs) between moderately and lowly PM10 exposed individuals. A high proportion of DMRs overlapped with regulatory elements, and DMR target genes were involved in pathways regulating cellular redox homeostasis and immune response. In addition, we found distinct changes in DNA methylation of the HOXA gene cluster whose methylation levels have previously been linked to air pollution exposure but also to carcinogenesis in several instances. The findings of this study suggest that regulatory changes in upper airway cells occur at PM10 levels below current European thresholds, some of which may be involved in the development of air pollution-related diseases.
Collapse
Affiliation(s)
- Marey Messingschlager
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany; Freie Universität Berlin, Institute for Biology, Königin-Luise-Strasse 12-16, 14195, Berlin, Germany
| | - Martina Bartel-Steinbach
- Fraunhofer Institute for Biomedical Engineering IBMT, Josef-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Sebastian D Mackowiak
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Johanna Denkena
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Bieg
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Klös
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| | - Anke Seegebarth
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| | - Wolfgang Straff
- Environmental Medicine and Health Effects Assessment, German Environment Agency, Corrensplatz 1, 14195, Berlin, Germany
| | - Katrin Süring
- Environmental Medicine and Health Effects Assessment, German Environment Agency, Corrensplatz 1, 14195, Berlin, Germany
| | - Naveed Ishaque
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Roland Eils
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany; German Center for Lung Research (DZL), Germany; Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Germany; Freie Universität Berlin, Department of Mathematics and Computer Science, Arnimallee 14, 14195, Berlin, Germany
| | - Irina Lehmann
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany; German Center for Lung Research (DZL), Germany.
| | - Dominik Lermen
- Fraunhofer Institute for Biomedical Engineering IBMT, Josef-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Saskia Trump
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
15
|
Cardenas A, Fadadu RP, Koppelman GH. Epigenome-wide association studies of allergic disease and the environment. J Allergy Clin Immunol 2023; 152:582-590. [PMID: 37295475 PMCID: PMC10564109 DOI: 10.1016/j.jaci.2023.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
The epigenome is at the intersection of the environment, genotype, and cellular response. DNA methylation of cytosine nucleotides, the most studied epigenetic modification, has been systematically evaluated in human studies by using untargeted epigenome-wide association studies (EWASs) and shown to be both sensitive to environmental exposures and associated with allergic diseases. In this narrative review, we summarize findings from key EWASs previously conducted on this topic; interpret results from recent studies; and discuss the strengths, challenges, and opportunities regarding epigenetics research on the environment-allergy relationship. The majority of these EWASs have systematically investigated select environmental exposures during the prenatal and early childhood periods and allergy-associated epigenetic changes in leukocyte-isolated DNA and more recently in nasal cells. Overall, many studies have found consistent DNA methylation associations across cohorts for certain exposures, such as smoking (eg, aryl hydrocarbon receptor repressor gene [AHRR] gene), and allergic diseases (eg, EPX gene). We recommend the integration of both environmental exposures and allergy or asthma within long-term prospective designs to strengthen causality as well as biomarker development. Future studies should collect paired target tissues to examine compartment-specific epigenetic responses, incorporate genetic influences in DNA methylation (methylation quantitative trait locus), replicate findings across diverse populations, and carefully interpret epigenetic signatures from bulk, target tissue or isolated cells.
Collapse
Affiliation(s)
- Andres Cardenas
- Department of Epidemiology and Population Health, Stanford School of Medicine, Stanford University, Stanford, Calif
| | - Raj P Fadadu
- School of Medicine, University of California, San Francisco, Calif
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, Groningen, The Netherlands; Groningen Research Institute of Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
16
|
Gonzalez N, Rao N, Dean M, Lee D, Hurson AN, Baris D, Schwenn M, Johnson A, Prokunina-Olsson L, Friesen MC, Zhu B, Rothman N, Silverman DT, Koutros S. Nitrated Polycyclic Aromatic Hydrocarbon (Nitro-PAH) Signatures and Somatic Mutations in Diesel Exhaust-Exposed Bladder Tumors. Cancer Epidemiol Biomarkers Prev 2023; 32:840-847. [PMID: 36996403 PMCID: PMC10239365 DOI: 10.1158/1055-9965.epi-22-1208] [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: 11/16/2022] [Revised: 02/01/2023] [Accepted: 03/24/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Diesel exhaust is a complex mixture, including polycyclic aromatic hydrocarbons (PAH) and nitrated PAHs (nitro-PAH), many of which are potent mutagens and possible bladder carcinogens. To explore the association between diesel exposure and bladder carcinogenesis, we examined the relationship between exposure and somatic mutations and mutational signatures in bladder tumors. METHODS Targeted sequencing was conducted in bladder tumors from the New England Bladder Cancer Study. Using data on 797 cases and 1,418 controls, two-stage polytomous logistic regression was used to evaluate etiologic heterogeneity between bladder cancer subtypes and quantitative, lifetime estimates of respirable elemental carbon (REC), a surrogate for diesel exposure. Poisson regression was used to evaluate associations between REC and mutational signatures. RESULTS We observed significant heterogeneity in the diesel-bladder cancer risk relationship, with a strong positive association among cases with high-grade, nonmuscle invasive TP53-mutated tumors compared with controls [ORTop Tertile vs.Unexposed, 4.8; 95% confidence interval (CI), 2.2-10.5; Ptrend < 0.001; Pheterogeneity = 0.002]. In muscle-invasive tumors, we observed a positive association between diesel exposure and the nitro-PAH signatures of 1,6-dintropyrene (RR, 1.93; 95% CI, 1.28-2.92) and 3-nitrobenzoic acid (RR, 1.97; 95% CI, 1.33-2.92). CONCLUSIONS The relationship between diesel exhaust and bladder cancer was heterogeneous based on the presence of TP53 mutations in tumors, further supporting the link between PAH exposure and TP53 mutations in carcinogenesis. Future studies that can identify nitro-PAH signatures in exposed tumors are warranted to add human data supporting the link between diesel and bladder cancer. IMPACT This study provides additional insight into the etiology and possible mechanisms related to diesel exhaust-induced bladder cancer.
Collapse
Affiliation(s)
- Nicole Gonzalez
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Nina Rao
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Michael Dean
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Donghyuk Lee
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
- Department of Statistics, Pusan National University, Busan, Korea
| | - Amber N. Hurson
- Trans-Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Dalsu Baris
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | | | | | - Ludmila Prokunina-Olsson
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Melissa C. Friesen
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Bin Zhu
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Debra T. Silverman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Stella Koutros
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
18
|
Mechanisms of DNA methylation and histone modifications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:51-92. [PMID: 37019597 DOI: 10.1016/bs.pmbts.2023.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The field of genetics has expanded a lot in the past few decades due to the accessibility of human genome sequences, but still, the regulation of transcription cannot be explicated exclusively by the sequence of DNA of an individual. The coordination and crosstalk between chromatin factors which are conserved is indispensable for all living creatures. The regulation of gene expression has been dependent on the methylation of DNA, post-translational modifications of histones, effector proteins, chromatin remodeler enzymes that affect the chromatin structure and function, and other cellular activities such as DNA replication, DNA repair, proliferation and growth. The mutation and deletion of these factors can lead to human diseases. Various studies are being performed to identify and understand the gene regulatory mechanisms in the diseased state. The information from these high throughput screening studies is able to aid the treatment developments based on the epigenetics regulatory mechanisms. This book chapter will discourse on various modifications and their mechanisms that take place on histones and DNA that regulate the transcription of genes.
Collapse
|
19
|
Serafini MM, Maddalon A, Iulini M, Galbiati V. Air Pollution: Possible Interaction between the Immune and Nervous System? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192316037. [PMID: 36498110 PMCID: PMC9738575 DOI: 10.3390/ijerph192316037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/14/2022] [Accepted: 11/26/2022] [Indexed: 06/01/2023]
Abstract
Exposure to environmental pollutants is a serious and common public health concern associated with growing morbidity and mortality worldwide, as well as economic burden. In recent years, the toxic effects associated with air pollution have been intensively studied, with a particular focus on the lung and cardiovascular system, mainly associated with particulate matter exposure. However, epidemiological and mechanistic studies suggest that air pollution can also influence skin integrity and may have a significant adverse impact on the immune and nervous system. Air pollution exposure already starts in utero before birth, potentially causing delayed chronic diseases arising later in life. There are, indeed, time windows during the life of individuals who are more susceptible to air pollution exposure, which may result in more severe outcomes. In this review paper, we provide an overview of findings that have established the effects of air pollutants on the immune and nervous system, and speculate on the possible interaction between them, based on mechanistic data.
Collapse
|
20
|
Melén E, Koppelman GH, Vicedo-Cabrera AM, Andersen ZJ, Bunyavanich S. Allergies to food and airborne allergens in children and adolescents: role of epigenetics in a changing environment. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:810-819. [PMID: 35985346 DOI: 10.1016/s2352-4642(22)00215-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Allergic diseases affect millions of children and adolescents worldwide. In this Review, we focus on allergies to food and airborne allergens and provide examples of prevalence trends during a time when climate change is of increasing concern. Profound environmental changes have affected natural systems in terms of biodiversity loss, air pollution, and climate. We discuss the potential links between these changes and allergic diseases in children, and the clinical implications. Several exposures of relevance for allergic disease also correlate with epigenetic changes such as DNA methylation. We propose that epigenetics could be a promising tool by which exposures and hazards related to a changing environment can be captured. Epigenetics might also provide promising biomarkers and help to elucidate the mechanisms related to allergic disease initiation and progress.
Collapse
Affiliation(s)
- Erik Melén
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology and Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, Netherlands
| | - Ana Maria Vicedo-Cabrera
- Institute of Social and Preventive Medicine and Oeschger Center for Climate Change Research, University of Bern, Bern, Switzerland
| | | | - Supinda Bunyavanich
- Division of Allergy and Immunology, Department of Pediatrics, and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Leem J, Kim S, Kim JS, Oh JS. ROS-independent cytotoxicity of 9,10-phenanthrenequinone inhibits cell cycle progression and spindle assembly during meiotic maturation in mouse oocytes. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129248. [PMID: 35739767 DOI: 10.1016/j.jhazmat.2022.129248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Diesel exhaust particles (DEPs) are major components of ambient particulate matter and are associated with various adverse health effects. Typically, DEPs contain a vast number of organic compounds, among which 9,10-phenanthrenequinone (9,10-PQ), the quinone derivative of the polycyclic aromatic hydrocarbon phenanthrene, is one of the most abundant and toxic. 9,10-PQ can produce excessive reactive oxygen species (ROS) via redox cycling and exhibit cytotoxicity in various cells. However, the underlying mechanisms involved in cytotoxicity of 9,10-PQ remain elusive. In this study, we investigated the effects of exposure to 9,10-PQ using mouse oocytes as a model system. We found that 9,10-PQ compromised meiotic maturation by impairing acentriolar microtubule organizing center (MTOC) assembly and subsequent spindle formation during meiotic maturation. Moreover, 9,10-PQ exposure prevented cell cycle progression by inhibiting Cdk1 activation via disturbance of cyclin B1 accumulation. Importantly, meiotic defects induced by 9,10-PQ exposure were not rescued by decreasing ROS levels, revealing that 9,10-PQ has ROS-independent activity that regulates cell cycle progression and spindle assembly. Therefore, our findings reveal that 9,10-PQ has novel activity that regulates cell-cycle progression and spindle formation in an ROS-independent manner during meiotic maturation in mouse oocytes.
Collapse
Affiliation(s)
- Jiyeon Leem
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Seul Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
22
|
Kim S, Leem J, Oh JS, Kim JS. Cytotoxicity of 9,10-Phenanthrenequinone Impairs Mitotic Progression and Spindle Assembly Independent of ROS Production in HeLa Cells. TOXICS 2022; 10:toxics10060327. [PMID: 35736935 PMCID: PMC9227850 DOI: 10.3390/toxics10060327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022]
Abstract
The polycyclic aromatic hydrocarbon quinone derivative 9,10-phenanthrenequinone (9,10-PQ) is one of the most abundant and toxic components found in diesel exhaust particles (DEPs). These DEPs are created during diesel fuel combustion and are considered the main source of urban air pollution. As 9,10-PQ can produce excessive reactive oxygen species (ROS) through redox cycling, it has been shown to exert potent cytotoxic effects against various cell types. However, the mechanisms underlying this cytotoxicity remain unclear. In this study, we showed that 9,10-PQ exerts cytotoxicity by impairing mitotic progression and spindle assembly in HeLa cells. Exposure to 9,10-PQ impaired spindle assembly and chromosome alignment, resulting in delayed mitotic entry and progression in HeLa cells. Furthermore, 9,10-PQ exposure decreased the CEP192 and p-Aurora A levels at the spindle poles. Notably, these mitotic defects induced by 9,10-PQ were not rescued by scavenging ROS, implying the ROS-independent activity of 9,10-PQ. Therefore, our results provide the first evidence that 9,10-PQ exerts its cytotoxicity through specific inhibition of mitotic progression and spindle assembly, independent of ROS.
Collapse
Affiliation(s)
- Seul Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Jiyeon Leem
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Jeong Su Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (J.S.O.); (J.-S.K.)
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
- Correspondence: (J.S.O.); (J.-S.K.)
| |
Collapse
|
23
|
Kim S, Hollinger H, Radke EG. 'Omics in environmental epidemiological studies of chemical exposures: A systematic evidence map. ENVIRONMENT INTERNATIONAL 2022; 164:107243. [PMID: 35551006 PMCID: PMC11515950 DOI: 10.1016/j.envint.2022.107243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 03/25/2022] [Accepted: 04/10/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Systematic evidence maps are increasingly used to develop chemical risk assessments. These maps can provide an overview of available studies and relevant study information to be used for various research objectives and applications. Environmental epidemiological studies that examine the impact of chemical exposures on various 'omic profiles in human populations provide relevant mechanistic information and can be used for benchmark dose modeling to derive potential human health reference values. OBJECTIVES To create a systematic evidence map of environmental epidemiological studies examining environmental contaminant exposures with 'omics in order to characterize the extent of available studies for future research needs. METHODS Systematic review methods were used to search and screen the literature and included the use of machine learning methods to facilitate screening studies. The Populations, Exposures, Comparators and Outcomes (PECO) criteria were developed to identify and screen relevant studies. Studies that met the PECO criteria after full-text review were summarized with information such as study population, study design, sample size, exposure measurement, and 'omics analysis. RESULTS Over 10,000 studies were identified from scientific databases. Screening processes were used to identify 84 studies considered PECO-relevant after full-text review. Various contaminants (e.g. phthalate, benzene, arsenic, etc.) were investigated in epidemiological studies that used one or more of the four 'omics of interest: epigenomics, transcriptomics, proteomics, and metabolomics . The epidemiological study designs that were used to explore single or integrated 'omic research questions with contaminant exposures were cohort studies, controlled trials, cross-sectional, and case-control studies. An interactive web-based systematic evidence map was created to display more study-related information. CONCLUSIONS This systematic evidence map is a novel tool to visually characterize the available environmental epidemiological studies investigating contaminants and biological effects using 'omics technology and serves as a resource for investigators and allows for a range of applications in chemical research and risk assessment needs.
Collapse
Affiliation(s)
- Stephanie Kim
- Superfund and Emergency Management Division, Region 2, U.S. Environmental Protection Agency, NY, USA.
| | - Hillary Hollinger
- Office of Pollution Prevention and Toxics, U.S. Environmental Protection Agency, NC, USA.
| | - Elizabeth G Radke
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, D.C, USA.
| |
Collapse
|
24
|
Acute air pollution exposure increases TETs in human PBMCs. J Allergy Clin Immunol 2022; 150:477-488.e9. [DOI: 10.1016/j.jaci.2022.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022]
|
25
|
Long E, Schwartz C, Carlsten C. Controlled human exposure to diesel exhaust: a method for understanding health effects of traffic-related air pollution. Part Fibre Toxicol 2022; 19:15. [PMID: 35216599 PMCID: PMC8876178 DOI: 10.1186/s12989-022-00454-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Diesel exhaust (DE) is a major component of air pollution in urban centers. Controlled human exposure (CHE) experiments are commonly used to investigate the acute effects of DE inhalation specifically and also as a paradigm for investigating responses to traffic-related air pollution (TRAP) more generally. Given the critical role this model plays in our understanding of TRAP's health effects mechanistically and in support of associated policy and regulation, we review the methodology of CHE to DE (CHE-DE) in detail to distill critical elements so that the results of these studies can be understood in context. From 104 eligible publications, we identified 79 CHE-DE studies and extracted information on DE generation, exposure session characteristics, pollutant and particulate composition of exposures, and participant demographics. Virtually all studies had a crossover design, and most studies involved a single DE exposure per participant. Exposure sessions were typically 1 or 2 h in duration, with participants alternating between exercise and rest. Most CHE-DE targeted a PM concentration of 300 μg/m3. There was a wide range in commonly measured co-pollutants including nitrogen oxides, carbon monoxide, and total organic compounds. Reporting of detailed parameters of aerosol composition, including particle diameter, was inconsistent between studies, and older studies from a given lab were often cited in lieu of repeating measurements for new experiments. There was a male predominance in participants, and over half of studies involved healthy participants only. Other populations studied include those with asthma, atopy, or metabolic syndrome. Standardization in reporting exposure conditions, potentially using current versions of engines with modern emissions control technology, will allow for more valid comparisons between studies of CHE-DE, while recognizing that diesel engines in much of the world remain old and heterogeneous. Inclusion of female participants as well as populations more susceptible to TRAP will broaden the applicability of results from CHE-DE studies.
Collapse
Affiliation(s)
- Erin Long
- Faculty of Medicine, University of British Columbia, 317 - 2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Carley Schwartz
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, 2775 Laurel Street 7th Floor, Vancouver, BC, V5Z 1M9, Canada
| | - Christopher Carlsten
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, 2775 Laurel Street 7th Floor, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
26
|
Long E, Carlsten C. Controlled human exposure to diesel exhaust: results illuminate health effects of traffic-related air pollution and inform future directions. Part Fibre Toxicol 2022; 19:11. [PMID: 35139881 PMCID: PMC8827176 DOI: 10.1186/s12989-022-00450-5] [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: 08/12/2021] [Accepted: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
Air pollution is an issue of increasing interest due to its globally relevant impacts on morbidity and mortality. Controlled human exposure (CHE) studies are often employed to investigate the impacts of pollution on human health, with diesel exhaust (DE) commonly used as a surrogate of traffic related air pollution (TRAP). This paper will review the results derived from 104 publications of CHE to DE (CHE-DE) with respect to health outcomes. CHE-DE studies have provided mechanistic evidence supporting TRAP’s detrimental effects on related to the cardiovascular system (e.g., vasomotor dysfunction, inhibition of fibrinolysis, and impaired cardiac function) and respiratory system (e.g., airway inflammation, increased airway responsiveness, and clinical symptoms of asthma). Oxidative stress is thought to be the primary mechanism of TRAP-induced effects and has been supported by several CHE-DE studies. A historical limitation of some air pollution research is consideration of TRAP (or its components) in isolation, limiting insight into the interactions between TRAP and other environmental factors often encountered in tandem. CHE-DE studies can help to shed light on complex conditions, and several have included co-exposure to common elements such as allergens, ozone, and activity level. The ability of filters to mitigate the adverse effects of DE, by limiting exposure to the particulate fraction of polluted aerosols, has also been examined. While various biomarkers of DE exposure have been evaluated in CHE-DE studies, a definitive such endpoint has yet to be identified. In spite of the above advantages, this paradigm for TRAP is constrained to acute exposures and can only be indirectly applied to chronic exposures, despite the critical real-world impact of living long-term with TRAP. Those with significant medical conditions are often excluded from CHE-DE studies and so results derived from healthy individuals may not apply to more susceptible populations whose further study is needed to avoid potentially misleading conclusions. In spite of limitations, the contributions of CHE-DE studies have greatly advanced current understanding of the health impacts associated with TRAP exposure, especially regarding mechanisms therein, with important implications for regulation and policy.
Collapse
Affiliation(s)
- Erin Long
- Faculty of Medicine, University of British Columbia, 317 - 2194 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Christopher Carlsten
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, 2775 Laurel Street 7th Floor, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
27
|
Goodman S, Chappell G, Guyton KZ, Pogribny IP, Rusyn I. Epigenetic alterations induced by genotoxic occupational and environmental human chemical carcinogens: An update of a systematic literature review. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 789:108408. [PMID: 35690411 PMCID: PMC9188653 DOI: 10.1016/j.mrrev.2021.108408] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 01/03/2023]
Abstract
Epigenetic alterations, such as changes in DNA methylation, histones/chromatin structure, nucleosome positioning, and expression of non-coding RNAs, are recognized among key characteristics of carcinogens; they may occur independently or concomitantly with genotoxic effects. While data on genotoxicity are collected through standardized guideline tests, data collected on epigenetic effects is far less uniform. In 2016, we conducted a systematic review of published studies of genotoxic carcinogens that reported epigenetic endpoints to better understand the evidence for epigenetic alterations of human carcinogens, and the potential association with genotoxic endpoints. Since then, the number of studies of epigenetic effects of chemicals has nearly doubled. This review stands as an update on epigenetic alterations induced by occupational and environmental human carcinogens that were previously and recently classified as Group 1 by the International Agency for Research on Cancer. We found that the evidence of epigenetic effects remains uneven across agents. Studies of DNA methylation are most abundant, while reports concerning effects on non-coding RNA have increased over the past 5 years. By contrast, mechanistic toxicology studies of histone modifications and chromatin state alterations remain few. We found that most publications of epigenetic effects of carcinogens were studies in exposed humans or human cells. Studies in rodents represent the second most common species used for epigenetic studies in toxicology, in vivo exposures being the most predominant. Future studies should incorporate dose- and time-dependent study designs and also investigate the persistence of effects following cessation of exposure, considering the dynamic nature of most epigenetic alterations.
Collapse
Affiliation(s)
- Samantha Goodman
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | | | | | - Igor P Pogribny
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
28
|
Konwar C, Asiimwe R, Inkster AM, Merrill SM, Negri GL, Aristizabal MJ, Rider CF, MacIsaac JL, Carlsten C, Kobor MS. Risk-focused differences in molecular processes implicated in SARS-CoV-2 infection: corollaries in DNA methylation and gene expression. Epigenetics Chromatin 2021; 14:54. [PMID: 34895312 PMCID: PMC8665859 DOI: 10.1186/s13072-021-00428-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Understanding the molecular basis of susceptibility factors to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global health imperative. It is well-established that males are more likely to acquire SARS-CoV-2 infection and exhibit more severe outcomes. Similarly, exposure to air pollutants and pre-existing respiratory chronic conditions, such as asthma and chronic obstructive respiratory disease (COPD) confer an increased risk to coronavirus disease 2019 (COVID-19). METHODS We investigated molecular patterns associated with risk factors in 398 candidate genes relevant to COVID-19 biology. To accomplish this, we downloaded DNA methylation and gene expression data sets from publicly available repositories (GEO and GTEx Portal) and utilized data from an empirical controlled human exposure study conducted by our team. RESULTS First, we observed sex-biased DNA methylation patterns in autosomal immune genes, such as NLRP2, TLE1, GPX1, and ARRB2 (FDR < 0.05, magnitude of DNA methylation difference Δβ > 0.05). Second, our analysis on the X-linked genes identified sex associated DNA methylation profiles in genes, such as ACE2, CA5B, and HS6ST2 (FDR < 0.05, Δβ > 0.05). These associations were observed across multiple respiratory tissues (lung, nasal epithelia, airway epithelia, and bronchoalveolar lavage) and in whole blood. Some of these genes, such as NLRP2 and CA5B, also exhibited sex-biased gene expression patterns. In addition, we found differential DNA methylation patterns by COVID-19 status for genes, such as NLRP2 and ACE2 in an exploratory analysis of an empirical data set reporting on human COVID-9 infections. Third, we identified modest DNA methylation changes in CpGs associated with PRIM2 and TATDN1 (FDR < 0.1, Δβ > 0.05) in response to particle-depleted diesel exhaust in bronchoalveolar lavage. Finally, we captured a DNA methylation signature associated with COPD diagnosis in a gene involved in nicotine dependence (COMT) (FDR < 0.1, Δβ > 0.05). CONCLUSION Our findings on sex differences might be of clinical relevance given that they revealed molecular associations of sex-biased differences in COVID-19. Specifically, our results hinted at a potentially exaggerated immune response in males linked to autosomal genes, such as NLRP2. In contrast, our findings at X-linked loci such as ACE2 suggested a potentially distinct DNA methylation pattern in females that may interact with its mRNA expression and inactivation status. We also found tissue-specific DNA methylation differences in response to particulate exposure potentially capturing a nitrogen dioxide (NO2) effect-a contributor to COVID-19 susceptibility. While we identified a molecular signature associated with COPD, all COPD-affected individuals were smokers, which may either reflect an association with the disease, smoking, or may highlight a compounded effect of these two risk factors in COVID-19. Overall, our findings point towards a molecular basis of variation in susceptibility factors that may partly explain disparities in the risk for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Chaini Konwar
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada
| | - Rebecca Asiimwe
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada
| | - Amy M Inkster
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- The Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Sarah M Merrill
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada
| | - Gian L Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Maria J Aristizabal
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada
- The Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, M5S 3B2, Canada
- Department of Biology, Queen' University, Kingston, ON, K7L 3N6, Canada
- Program in Child and Brain Development, CIFAR, MaRS Centre, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Christopher F Rider
- The Department of Respiratory Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Julie L MacIsaac
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada
| | - Christopher Carlsten
- The Department of Respiratory Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Michael S Kobor
- BC Children's Hospital Research Institute (BCCHR), 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V6H 0B3, Canada.
- Program in Child and Brain Development, CIFAR, MaRS Centre, 661 University Ave, Toronto, ON, M5G 1M1, Canada.
- The Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
29
|
Soliai MM, Kato A, Helling BA, Stanhope CT, Norton JE, Naughton KA, Klinger AI, Thompson EE, Clay SM, Kim S, Celedón JC, Gern JE, Jackson DJ, Altman MC, Kern RC, Tan BK, Schleimer RP, Nicolae DL, Pinto JM, Ober C. Multi-omics colocalization with genome-wide association studies reveals a context-specific genetic mechanism at a childhood onset asthma risk locus. Genome Med 2021; 13:157. [PMID: 34629083 PMCID: PMC8504130 DOI: 10.1186/s13073-021-00967-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified thousands of variants associated with asthma and other complex diseases. However, the functional effects of most of these variants are unknown. Moreover, GWASs do not provide context-specific information on cell types or environmental factors that affect specific disease risks and outcomes. To address these limitations, we used an upper airway epithelial cell (AEC) culture model to assess transcriptional and epigenetic responses to rhinovirus (RV), an asthma-promoting pathogen, and provide context-specific functional annotations to variants discovered in GWASs of asthma. METHODS Genome-wide genetic, gene expression, and DNA methylation data in vehicle- and RV-treated upper AECs were collected from 104 individuals who had a diagnosis of airway disease (n=66) or were healthy participants (n=38). We mapped cis expression and methylation quantitative trait loci (cis-eQTLs and cis-meQTLs, respectively) in each treatment condition (RV and vehicle) in AECs from these individuals. A Bayesian test for colocalization between AEC molecular QTLs and adult onset asthma and childhood onset asthma GWAS SNPs, and a multi-ethnic GWAS of asthma, was used to assign the function to variants associated with asthma. We used Mendelian randomization to demonstrate DNA methylation effects on gene expression at asthma colocalized loci. RESULTS Asthma and allergic disease-associated GWAS SNPs were specifically enriched among molecular QTLs in AECs, but not in GWASs from non-immune diseases, and in AEC eQTLs, but not among eQTLs from other tissues. Colocalization analyses of AEC QTLs with asthma GWAS variants revealed potential molecular mechanisms of asthma, including QTLs at the TSLP locus that were common to both the RV and vehicle treatments and to both childhood onset and adult onset asthma, as well as QTLs at the 17q12-21 asthma locus that were specific to RV exposure and childhood onset asthma, consistent with clinical and epidemiological studies of these loci. CONCLUSIONS This study provides evidence of functional effects for asthma risk variants in AECs and insight into RV-mediated transcriptional and epigenetic response mechanisms that modulate genetic effects in the airway and risk for asthma.
Collapse
Affiliation(s)
- Marcus M Soliai
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| | - Atsushi Kato
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Britney A Helling
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | - James E Norton
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Aiko I Klinger
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma E Thompson
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Selene M Clay
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Soyeon Kim
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Matthew C Altman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Robert C Kern
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bruce K Tan
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert P Schleimer
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan L Nicolae
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Jayant M Pinto
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
30
|
Clifford RL, Jones MJ. Greater Than the Sum of Its Parts: Integrating Multiple Data Sets to Decipher Cigarette Smoking Effects on Airway Epithelium. Am J Respir Cell Mol Biol 2021; 65:335-336. [PMID: 34139135 PMCID: PMC8525203 DOI: 10.1165/rcmb.2021-0207ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Rachel L Clifford
- Centre for Respiratory Medicine University of Nottingham and Nottingham NIHR Biomedical Research Centre Nottingham, United Kingdom
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics University of Manitoba Manitoba, Canada and
- Children's Hospital Research Institute of Manitoba Manitoba, Canada
| |
Collapse
|
31
|
Mukherjee S, Dasgupta S, Mishra PK, Chaudhury K. Air pollution-induced epigenetic changes: disease development and a possible link with hypersensitivity pneumonitis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:55981-56002. [PMID: 34498177 PMCID: PMC8425320 DOI: 10.1007/s11356-021-16056-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/16/2021] [Indexed: 05/16/2023]
Abstract
Air pollution is a serious threat to our health and has become one of the major causes of many diseases including cardiovascular disease, respiratory disease, and cancer. The association between air pollution and various diseases has long been a topic of research interest. However, it remains unclear how air pollution actually impacts health by modulating several important cellular functions. Recently, some evidence has emerged about air pollution-induced epigenetic changes, which are linked with the etiology of various human diseases. Among several epigenetic modifications, DNA methylation represents the most prominent epigenetic alteration underlying the air pollution-induced pathogenic mechanism. Several other types of epigenetic changes, such as histone modifications, miRNA, and non-coding RNA expression, have also been found to have been linked with air pollution. Hypersensitivity pneumonitis (HP), one of the most prevalent forms of interstitial lung diseases (ILDs), is triggered by the inhalation of certain organic and inorganic substances. HP is characterized by inflammation in the tissues around the lungs' airways and may lead to irreversible lung scarring over time. This review, in addition to other diseases, attempts to understand whether certain pollutants influence HP development through such epigenetic modifications.
Collapse
Affiliation(s)
- Suranjana Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
| | - Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| |
Collapse
|
32
|
Slob EMA, Richards LB, Vijverberg SJH, Longo C, Koppelman GH, Pijnenburg MWH, Bel EHD, Neerincx AH, Herrera Luis E, Perez-Garcia J, Tim Chew F, Yie Sio Y, Andiappan AK, Turner SW, Mukhopadhyay S, Palmer CNA, Hawcutt D, Jorgensen AL, Burchard EG, Hernandez-Pacheco N, Pino-Yanes M, Maitland-van der Zee AH. Genome-wide association studies of exacerbations in children using long-acting beta2-agonists. Pediatr Allergy Immunol 2021; 32:1197-1207. [PMID: 33706416 PMCID: PMC8328929 DOI: 10.1111/pai.13494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Some children with asthma experience exacerbations despite long-acting beta2-agonist (LABA) treatment. While this variability is partly caused by genetic variation, no genome-wide study until now has investigated which genetic factors associated with risk of exacerbations despite LABA use in children with asthma. We aimed to assess whether genetic variation was associated with exacerbations in children treated with LABA from a global consortium. METHODS A meta-analysis of genome-wide association studies (meta-GWAS) was performed in 1,425 children and young adults with asthma (age 6-21 years) with reported regular use of LABA from six studies within the PiCA consortium using a random effects model. The primary outcome of each study was defined as any exacerbation within the past 6 or 12 months, including at least one of the following: 1) hospital admissions for asthma, 2) a course of oral corticosteroids or 3) emergency room visits because of asthma. RESULTS Genome-wide association results for a total of 82 996 common single nucleotide polymorphisms (SNPs, MAF ≥1%) with high imputation quality were meta-analysed. Eight independent variants were suggestively (P-value threshold ≤5 × 10-6 ) associated with exacerbations despite LABA use. CONCLUSION No strong effects of single nucleotide polymorphisms (SNPs) on exacerbations during LABA use were identified. We identified two loci (TBX3 and EPHA7) that were previously implicated in the response to short-acting beta2-agonists (SABA). These loci merit further investigation in response to LABA and SABA use.
Collapse
Affiliation(s)
- Elise M A Slob
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Pediatric Respiratory Medicine, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - Levi B Richards
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne J H Vijverberg
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Pediatric Respiratory Medicine, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Cristina Longo
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerard H Koppelman
- Department of Paediatric, Pulmonology & Paediatric Allergology, University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, The Netherlands.,University Medical Center Groningen, Groningen Research Institute for Asthma & COPD (GRIAC), University of Groningen, Groningen, The Netherlands
| | - Mariëlle W H Pijnenburg
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Erasmus MC - Sophia, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Elisabeth H D Bel
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne H Neerincx
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Herrera Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Fook Tim Chew
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yang Yie Sio
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anand K Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Steve W Turner
- Department of Child Health, University of Aberdeen, Aberdeen, UK
| | - Somnath Mukhopadhyay
- Academic Department of Paediatrics, Royal Alexandra Children's Hospital, Brighton and Sussex Medical School, Brighton, UK.,Population Pharmacogenetics Group, Biomedical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Colin N A Palmer
- Population Pharmacogenetics Group, Biomedical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Daniel Hawcutt
- Department of Women's and Children's Health, University of Liverpool, Liverpool, UK.,NIHR Alder Hey Clinical Research Facility, Alder Hey Children's Hospital, Liverpool, UK
| | - Andrea L Jorgensen
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Esteban G Burchard
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Natalia Hernandez-Pacheco
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Anke H Maitland-van der Zee
- Department of Respiratory Disease, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Pediatric Respiratory Medicine, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
33
|
Abstract
There has been a substantial increase in the incidence and the prevalence of allergic disorders in the recent decades, which seems to be related to rapid environmental and lifestyle changes, such as higher exposure to factors thought to exert pro-allergic effects but less contact with factors known to be associated with protection against the development of allergies. Pollution is the most remarkable example of the former, while less contact with microorganisms, lower proportion of unprocessed natural products in diet, and others resulting from urbanization and westernization of the lifestyle exemplify the latter. It is strongly believed that the effects of environmental factors on allergy susceptibility and development are mediated by epigenetic mechanisms, i.e. biologically relevant biochemical changes of the chromatin carrying transcriptionally-relevant information but not affecting the nucleotide sequence of the genome. Classical epigenetic mechanisms include DNA methylation and histone modifications, for instance acetylation or methylation. In addition, microRNA controls gene expression at the mRNA level. Such epigenetic mechanisms are involved in crucial regulatory processes in cells playing a pivotal role in allergies. Those include centrally managing cells, such as T lymphocytes, as well as specific structural and effector cells in the affected organs, responsible for the local clinical presentation of allergy, e.g. epithelial or airway smooth muscle cells in asthma. Considering that allergic disorders possess multiple clinical (phenotypes) and mechanistic (endotypes) forms, targeted, stratified treatment strategies based on detailed clinical and molecular diagnostics are required. Since conventional diagnostic or therapeutic approaches do not suffice, this gap could possibly be filled out by epigenetic approaches.
Collapse
|
34
|
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: 112] [Impact Index Per Article: 28.0] [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
|
35
|
de Homdedeu M, Cruz MJ, Sánchez-Díez S, Gómez-Ollés S, Ojanguren I, Ma D, Muñoz X. Role of diesel exhaust particles in the induction of allergic asthma to low doses of soybean. ENVIRONMENTAL RESEARCH 2021; 196:110337. [PMID: 33130171 DOI: 10.1016/j.envres.2020.110337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
INTRODUCTION Exposure to environmental pollutants such as diesel exhaust particles (DEP) increases the risk of asthma and asthma exacerbation. However, the exact mechanisms inducing asthma to low doses of allergens remain poorly understood. The present study aimed to analyse the immunomodulatory effect of the inhalation of DEP in a mouse model exposed to non-asthmagenic doses of soybean hull extract (SHE). MATERIAL AND METHODS BALB/c ByJ mice were randomly divided into four experimental groups. Two groups received nasal instillations of saline and the other two groups received 3 mg ml-1 SHE during 5 days per week for 3 weeks. One group in each pair also received 150 μg of DEP in the same instillations 3 days per week. SHE-specific IgE levels, oxidative stress, leukocyte pattern and optical projection tomography (OPT) imaging studies were assessed. RESULTS Inhalation of SHE and/or DEP increased levels of H2O2 in BAL, while coexposure to SHE and DEP increased SHE-specific IgE levels in serum. Inhalation of SHE alone increased eosinophils, B cells, total and resident monocytes and decreased levels of NK cells, while inhalation of DEP increased neutrophils and decreased total monocytes. Regarding dendritic cells (DC), the inhalation of SHE and/or DEP increased the total population, while the inhalation of SHE alone increased Th2-related DCs (CD11b + Ly6C-) and decreased tolerogenic DCs (CD11b-Ly6C-). However, coexposure to SHE and DEP increased oxidative stress-sensitive DCs (CD11b-Ly6C+) and decreased Th1-related DCs (CD11b + Ly6C+). As regards macrophages, inhalation of SHE and DEP decreased total and alveolar populations. DEP deposition in lung tissue did not differ between groups. CONCLUSION Coexposure to DEP activates the asthmatic response to low doses of soy by triggering the immune response and oxidative stress.
Collapse
Affiliation(s)
- M de Homdedeu
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M J Cruz
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - S Sánchez-Díez
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S Gómez-Ollés
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - I Ojanguren
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - D Ma
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - X Muñoz
- Pulmonology Service, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Enfermedades Respiratorias (CibeRes), Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
36
|
Cardenas A, Fadadu RP, Van Der Laan L, Ward-Caviness C, Granger L, Diaz-Sanchez D, Devlin RB, Bind MA. Controlled human exposures to diesel exhaust: a human epigenome-wide experiment of target bronchial epithelial cells. ENVIRONMENTAL EPIGENETICS 2021; 7:dvab003. [PMID: 33859829 PMCID: PMC8035831 DOI: 10.1093/eep/dvab003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 05/28/2023]
Abstract
Diesel exhaust (DE) is a major contributor to ambient air pollution around the world. It is a known human carcinogen that targets the respiratory system and increases risk for many diseases, but there is limited research on the effects of DE exposure on the epigenome of human bronchial epithelial cells. Understanding the epigenetic impact of this environmental pollutant can elucidate biological mechanisms involved in the pathogenesis of harmful DE-related health effects. To estimate the causal effect of short-term DE exposure on the bronchial epithelial epigenome, we conducted a controlled single-blinded randomized crossover human experiment of exposure to DE and used bronchoscopy and Illumina 450K arrays for data collection and analysis, respectively. Of the 13 participants, 11 (85%) were male and 2 (15%) were female, and 12 (92%) were White and one (8%) was Hispanic; the mean age was 26 years (SD = 3.8 years). Eighty CpGs were differentially methylated, achieving the minimum possible exact P-value of P = 2.44 × 10-4 (i.e. 2/213). In regional analyses, we found two differentially methylated regions (DMRs) annotated to the chromosome 5 open reading frame 63 genes (C5orf63; 7-CpGs) and unc-45 myosin chaperone A gene (UNC45A; 5-CpGs). Both DMRs showed increased DNA methylation after DE exposure. The average causal effects for the DMRs ranged from 1.5% to 6.0% increases in DNA methylation at individual CpGs. In conclusion, we found that short-term DE alters DNA methylation of genes in target bronchial epithelial cells, demonstrating epigenetic level effects of exposure that could be implicated in pulmonary pathologies.
Collapse
Affiliation(s)
- Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley; Berkeley, CA 94704, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94704, USA
| | - Raj P Fadadu
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley; Berkeley, CA 94704, USA
- School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lars Van Der Laan
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley; Berkeley, CA 94704, USA
| | - Cavin Ward-Caviness
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27709, USA
| | - Louis Granger
- Department of Statistics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA
| | - David Diaz-Sanchez
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27709, USA
| | - Robert B Devlin
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27709, USA
| | - Marie-Abèle Bind
- Department of Statistics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
37
|
Ellis AK, Murrieta-Aguttes M, Furey S, Picard P, Carlsten C. Effect of fexofenadine hydrochloride on allergic rhinitis aggravated by air pollutants. ERJ Open Res 2021; 7:00806-2020. [PMID: 33834054 PMCID: PMC8021806 DOI: 10.1183/23120541.00806-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/18/2021] [Indexed: 11/27/2022] Open
Abstract
In recent decades, seasonal allergic rhinitis (SAR) prevalence has increased and recent studies have shown that air pollutants, such as diesel exhaust particles (DEP), can increase inflammatory and allergic biomarkers. The aim of this study was to investigate the effects of DEP on SAR symptoms induced by ragweed and to evaluate the efficacy and safety of fexofenadine HCl 180 mg versus placebo. This phase 3, single-centre, sequential, parallel-group, double-blind, randomised study (NCT03664882) was conducted in an environmental exposure unit (EEU) during sequential exposures: Period 1 (ragweed pollen alone), Period 2 (ragweed pollen+DEP), and Period 3 (ragweed pollen+DEP+single-dose fexofenadine HCl 180 mg or placebo). Efficacy and safety were evaluated in Period 3. Primary endpoints were the area under the curve (AUC) of total nasal symptom score (TNSS) from baseline to hour 12 (AUC0–12) during Period 1 and Period 2; and the AUC of the TNSS from hour 2 to 12 (AUC2–12) during Period 3. 251 out of 257 evaluable subjects were included in the modified intent-to-treat population. Least squares mean difference (95% CI) for TNSS Log AUC0−12 in Period 2 versus Period 1 was 0.13 (0.081–0.182; p<0.0001). Least squares mean difference in TNSS Log AUC2−12 for fexofenadine HCl versus placebo during Period 3 was −0.24 (−0.425–−0.047; p=0.0148). One fexofenadine HCl-related adverse event was observed. SAR symptoms evoked by ragweed were aggravated by DEP. Fexofenadine HCl 180 mg was effective in relieving pollen-induced, air pollution-aggravated allergic rhinitis symptoms. This is the first randomised, double-blind, large study to demonstrate the beneficial effect of a histamine H1-receptor antagonist by reducing ragweed pollen-induced seasonal allergic rhinitis symptoms aggravated by controlled exposure to air pollutantshttps://bit.ly/3oauMFu
Collapse
Affiliation(s)
- Anne K Ellis
- Division of Allergy and Immunology, Dept of Medicine, Queen's University, Kingston, ON, Canada
| | | | - Sandy Furey
- Sanofi Consumer Health Care, Bridgewater, MA, USA
| | | | - Christopher Carlsten
- Air Pollution Exposure Laboratory, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Lambert KA, Markevych I, Yang BY, Bauer CP, Berdel D, von Berg A, Bergmann KC, Lodge C, Koletzko S, Prendergast LA, Schikowski T, Schulz H, Werchan M, Heinrich J, Standl M, Erbas B. Association of early life and acute pollen exposure with lung function and exhaled nitric oxide (FeNO). A prospective study up to adolescence in the GINIplus and LISA cohort. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 763:143006. [PMID: 33131877 DOI: 10.1016/j.scitotenv.2020.143006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/22/2020] [Accepted: 10/08/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Pollen exposure has both acute and chronic detrimental effects on allergic asthma, but little is known about its wider effects on respiratory health. This is increasingly important knowledge as ambient pollen levels are changing with the changing global climate. OBJECTIVE To assess associations of pollen exposure with lung function and fractional exhaled nitric oxide (FeNO) at age 15 in two prospective German birth cohorts, GINIplus and LISA. METHODS Background city-specific pollen exposure was measured in infancy (during the first three months of life), and contemporary (on the day of and 7 days prior to lung function measurement). Greenness levels within circular buffers (100-3000 m) around the birth and 15-year home addresses were calculated using the satellite-derived Normalized Difference Vegetation Index. Regression models were used to assess the associations of grass and birch pollen with lung function and FeNO, and the modifying effects of residential greenness were explored. RESULTS Cumulative early life exposure to grass pollen was associated with reduced lung function in adolescence (FEV1: -4.9 mL 95%CI: -9.2, -0.6 and FVC: -5.2 mL 95%CI: -9.8, -0.5 per doubling of pollen count). Acute grass pollen exposure was associated with increased airway inflammation in all children, with higher FeNO increases in children living in green areas. In contrast acute birch pollen exposure was associated with reduced lung function only in children sensitised to birch allergens. CONCLUSION This study provides suggestive evidence that early pollen exposure has a negative effect on later lung function, which is in turn influenced by acute pollen exposures.
Collapse
Affiliation(s)
- Katrina A Lambert
- Department of Public Health, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Iana Markevych
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, Ludwig Maximilians University of Munich, Munich, Germany; Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Bo-Yi Yang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, PR China
| | - Carl-Peter Bauer
- Department of Pediatrics, Technical University of Munich, Munich, Germany
| | - Dietrich Berdel
- Research Institute, Department of Pediatrics, Marien-Hospital Wesel, Wesel, Germany
| | - Andrea von Berg
- Research Institute, Department of Pediatrics, Marien-Hospital Wesel, Wesel, Germany
| | | | - Caroline Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, The University of Melbourne, Melbourne, Australia
| | - Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany; Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Luke A Prendergast
- Department of Mathematics and Statistics, School of Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Tamara Schikowski
- IUF, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich, Germany
| | | | - Joachim Heinrich
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, Ludwig Maximilians University of Munich, Munich, Germany; Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population & Global Health, The University of Melbourne, Melbourne, Australia
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Bircan Erbas
- Department of Public Health, School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Faculty of Public Health, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
39
|
Lam HCY, Jarvis D, Fuertes E. Interactive effects of allergens and air pollution on respiratory health: A systematic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 757:143924. [PMID: 33310575 PMCID: PMC7812370 DOI: 10.1016/j.scitotenv.2020.143924] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 05/28/2023]
Abstract
BACKGROUND Studies have demonstrated an adverse role of outdoor allergens on respiratory symptoms. It is unknown whether this effect is independent or synergistic of outdoor air pollutants. METHODS We systematically reviewed all epidemiological studies that examined interaction effects between counts of outdoor airborne allergens (pollen, fungal spores) and air pollutants, on any respiratory health outcome in children and adults. We searched the MEDLINE, EMBASE and Scopus databases. Each study was summarized qualitatively and assessed for quality and risk of bias (International Prospective Register for Systematic Reviews, registration number CRD42020162571). RESULTS Thirty-five studies were identified (15 timeseries, eight case-crossovers, 11 panels and one cohort study), of which 12 reported a significant statistical interaction between an allergen and air pollutant. Eight interactions were related to asthma outcomes, including one on lung function measures and wheeze, three to medical consultations for pollinosis and one to allergic symptoms (nasal, ocular or bronchial). There was no consensus as to which allergen or air pollutant is more likely to interact. No study investigated whether interactions are stronger in atopic individuals. CONCLUSION Despite strong evidence from small experimental studies in humans, only a third of studies identified significant allergen-pollutant interactions using common epidemiological study designs. Exposure misclassification, failure to examine subgroups at risk, inadequate statistical power or absence of population-level effects are possible explanations.
Collapse
Affiliation(s)
- Holly C Y Lam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; MRC Centre for Environment & Health, Imperial College, London, United Kingdom.
| | - Deborah Jarvis
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; MRC Centre for Environment & Health, Imperial College, London, United Kingdom.
| | - Elaine Fuertes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
40
|
Tu Y, Williams GM, Cortés de Waterman AM, Toelle BG, Guo Y, Denison L, Babu GR, Yang BY, Dong GH, Jalaludin B, Marks GB, Knibbs LD. A national cross-sectional study of exposure to outdoor nitrogen dioxide and aeroallergen sensitization in Australian children aged 7-11 years. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 271:116330. [PMID: 33383426 DOI: 10.1016/j.envpol.2020.116330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 06/12/2023]
Abstract
The prevalence of allergic diseases in Australian children is high, but few studies have assessed the potential role of outdoor air pollution in allergic sensitization. We investigated the association between outdoor air pollution and the prevalence of aeroallergen sensitization in a national cross-sectional study of Australian children aged 7-11 years. Children were recruited from 55 participating schools in 12 Australian cities during 2007-2008. Parents completed a detailed (70-item) questionnaire. Outdoor nitrogen dioxide (NO2), as a proxy for exposure to traffic-related emissions, was estimated using measurements from regulatory monitors near each school and a national land-use regression (LUR) model. Three averaging periods were assessed, using information on duration of residence at the address, including lifetime, previous (lifetime, excluding the last year), and recent (the last year only). The LUR model was used as an additional source of recent exposure estimates at school and home addresses. Skin prick tests (SPTs) were performed to measure sensitization to eight common aeroallergens. Multilevel logistic regression estimated the association between NO2 and sensitization (by individual allergens, indoor and outdoor allergens, and all allergens combined), after adjustment for individual- and area-level covariates. In total, 2226 children had a completed questionnaire and SPT. The prevalence of sensitization to any allergen was 44.4%. Sensitization to house dust mites (HDMs) was the most common (36.1%), while sensitization to Aspergillus was the least common (3.4%). Measured mean (±s.d.) NO2 exposure was between 9 (±2.9) ppb and 9.5 (±3.2) ppb, depending on the averaging period. An IQR (4 ppb) increase in measured previous NO2 exposure was associated with greater odds of sensitization to HDMs (OR: 1.21, 95% CI: 1.01-1.43, P = 0.035). We found evidence of an association between relatively low outdoor NO2 concentrations and sensitization to HDMs, but not other aeroallergens, in Australian children aged 7-11 years.
Collapse
Affiliation(s)
- Yanhui Tu
- Faculty of Medicine, School of Public Health, The University of Queensland, Herston, QLD, 4006, Australia
| | - Gail M Williams
- Faculty of Medicine, School of Public Health, The University of Queensland, Herston, QLD, 4006, Australia
| | | | - Brett G Toelle
- Woolcock Institute of Medical Research, The University of Sydney, NSW, 2006, Australia; Sydney Local Health District, Sydney, NSW, 2050, Australia
| | - Yuming Guo
- Centre for Air Pollution, Energy and Health Research, Glebe, NSW, 2037, Australia; Department of Epidemiology and Biostatistics, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Lyn Denison
- ERM Services Australia, Melbourne, VIC, 3000, Australia
| | - Giridhara R Babu
- Indian Institute of Public Health-Bangalore, Public Health Foundation of India, Bangalore, 560023, India
| | - Bo-Yi Yang
- Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guang-Hui Dong
- Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bin Jalaludin
- Centre for Air Pollution, Energy and Health Research, Glebe, NSW, 2037, Australia; Population Health, South Western Sydney Local Health District, Liverpool, NSW, 2170, Australia; Ingham Institute, Liverpool, NSW, 2170, Australia
| | - Guy B Marks
- Centre for Air Pollution, Energy and Health Research, Glebe, NSW, 2037, Australia; Woolcock Institute of Medical Research, The University of Sydney, NSW, 2006, Australia; South Western Sydney Clinical School, The University of New South Wales, Liverpool, NSW, 2170, Australia
| | - Luke D Knibbs
- Faculty of Medicine, School of Public Health, The University of Queensland, Herston, QLD, 4006, Australia; Centre for Air Pollution, Energy and Health Research, Glebe, NSW, 2037, Australia.
| |
Collapse
|
41
|
Sharma J, Parsai K, Raghuwanshi P, Ali SA, Tiwari V, Bhargava A, Mishra PK. Emerging role of mitochondria in airborne particulate matter-induced immunotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116242. [PMID: 33321436 DOI: 10.1016/j.envpol.2020.116242] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 05/05/2023]
Abstract
The immune system is one of the primary targets of airborne particulate matter. Recent evidence suggests that mitochondria lie at the center of particulate matter-induced immunotoxicity. Particulate matter can directly interact with mitochondrial components (proteins, lipids, and nucleic acids) and impairs the vital mitochondrial processes including redox mechanisms, fusion-fission, autophagy, and metabolic pathways. These disturbances impede different mitochondrial functions including ATP production, which acts as an important platform to regulate immunity and inflammatory responses. Moreover, the mitochondrial DNA released into the cytosol or in the extracellular milieu acts as a danger-associated molecular pattern and triggers the signaling pathways, involving cGAS-STING, TLR9, and NLRP3. In the present review, we discuss the emerging role of mitochondria in airborne particulate matter-induced immunotoxicity and its myriad biological consequences in health and disease.
Collapse
Affiliation(s)
- Jahnavi Sharma
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Kamakshi Parsai
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pragati Raghuwanshi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Sophiya Anjum Ali
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Vineeta Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India.
| |
Collapse
|
42
|
Koch S, Welch JF, Tran R, Ramsook AH, Hung A, Carlsten C, Guenette JA, Koehle MS. Ventilatory responses to constant load exercise following the inhalation of a short-acting ß 2-agonist in a laboratory-controlled diesel exhaust exposure study in individuals with exercise-induced bronchoconstriction. ENVIRONMENT INTERNATIONAL 2021; 146:106182. [PMID: 33395924 DOI: 10.1016/j.envint.2020.106182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/30/2020] [Accepted: 10/04/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE Individuals with exercise-induced bronchoconstriction (EIB) use ß2-agonists to reduce respiratory symptoms during acute exercise. The resultingbronchodilation could increase the dose of inhaled pollutants and impair respiratory function when exercise is performedin air pollution. We aimed to assess respiratory responses in individuals with EIB when completing a cycling bout while being exposed to diesel exhaust (DE) or filtered air (FA) with and without the inhalation of salbutamol (SAL), a short-acting ß2-agonist. METHODS In a double-blind, repeated-measures design, 19 participants with EIB (22-33 years of age) completed four visits: FA-placebo (FA-PLA), FA-SAL, DE-PLA, DE-SAL. After the inhalation of either 400 µg of SAL or PLA, participants sat in the exposure chamber for 60 min, breathing either FA or DE (PM2.5 = 300 μg/m3). Participants then cycled for 30 min at 50 % of peak work rate while breathing FA or DE. Respiratory responses were assessed via spirometry, work of breathing (WOB), fractional use of ventilatory capacity (V̇E/V̇E,CAP), area under the maximal expiratory flow-volume curve (MEFVAUC), and dyspnea during and following cycling. RESULTS Bronchodilation in response to SAL and acute cycling was observed, independent of FA/DE exposure. Specifically, FEV1 was increased by 7.7 % (confidence interval (CI): 7.2-8.2 %; p < 0.01) in response to SAL, and MEFVAUC was increased after cycling by 1.1 % (0.9-1.3 %; p = 0.03). Despite a significant decrease in total WOB by 6.2 J/min (4.7-7.5 J/min; p = 0.049) and a reduction in V̇E/V̇E,CAP by 5.8 % (5-6 %, p < 0.01) in the SAL exposures, no changes were observed in dyspnea. The DE exposure significantly increased V̇E/V̇E,CAP by 2.4 % (0.9-3.9 %; p < 0.01), but this did not affect dyspnea. DISCUSSION Our findings suggest that the use of SAL prior to moderate-intensity exercise when breathing high levels of DE, does not reduce respiratory function or exercise ventilatory responses for up to 60 min following exercise.
Collapse
Affiliation(s)
- Sarah Koch
- School of Kinesiology, University of British Columbia, Vancouver, Canada.
| | - Joseph F Welch
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Raymond Tran
- School of Kinesiology, University of British Columbia, Vancouver, Canada; Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Andrew H Ramsook
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, British Columbia, Canada; Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andy Hung
- School of Kinesiology, University of British Columbia, Vancouver, Canada
| | - Christopher Carlsten
- Faculty of Medicine, University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Jordan A Guenette
- School of Kinesiology, University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, British Columbia, Canada; Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S Koehle
- School of Kinesiology, University of British Columbia, Vancouver, Canada; Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
43
|
Huang SK, Tripathi P, Koneva LA, Cavalcante RG, Craig N, Scruggs AM, Sartor MA, Deng F, Chen Y. Effect of concentration and duration of particulate matter exposure on the transcriptome and DNA methylome of bronchial epithelial cells. ENVIRONMENTAL EPIGENETICS 2021; 7:dvaa022. [PMID: 33692908 PMCID: PMC7928203 DOI: 10.1093/eep/dvaa022] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/18/2020] [Accepted: 12/08/2020] [Indexed: 05/04/2023]
Abstract
Exposure to particulate matter (PM) from ambient air pollution is a well-known risk factor for many lung diseases, but the mechanism(s) for this is not completely understood. Bronchial epithelial cells, which line the airway of the respiratory tract, undergo genome-wide level changes in gene expression and DNA methylation particularly when exposed to fine (<2.5 µm) PM (PM2.5). Although some of these changes have been reported in other studies, a comparison of how different concentrations and duration of exposure affect both the gene transcriptome and DNA methylome has not been done. Here, we exposed BEAS-2B, a bronchial epithelial cell line, to different concentrations of PM2.5, and compared how single or repeated doses of PM2.5 affect both the transcriptome and methylome of cells. Widespread changes in gene expression occurred after cells were exposed to a single treatment of high-concentration (30 µg/cm2) PM2.5 for 24 h. These genes were enriched in pathways regulating cytokine-cytokine interactions, Mitogen-Activated Protein Kinase (MAPK) signaling, PI3K-Akt signaling, IL6, and P53. DNA methylomic analysis showed that nearly half of the differentially expressed genes were found to also have DNA methylation changes, with just a slightly greater trend toward overall hypomethylation across the genome. Cells exposed to a lower concentration (1 µg/cm2) of PM2.5 demonstrated a comparable, but more attenuated change in gene expression compared to cells exposed to higher concentrations. There were also many genes affected by lower concentrations of PM2.5, but not higher concentrations. Additionally, repeated exposure to PM2.5 (1 µg/cm2) for seven days resulted in transcriptomic and DNA methylomic changes that were distinct from cells treated with PM2.5 for only one day. Compared to single exposure, repeated exposure to PM2.5 caused a more notable degree of hypomethylation across the genome, though certain genes and regions demonstrated increased DNA methylation. The overall increase in hypomethylation, especially with repeated exposure to PM2.5, was associated with an increase in expression of ten-eleven translocation enzymes. These data demonstrate how variations in concentration and duration of PM2.5 exposure induce distinct differences in the transcriptomic and DNA methylomic profile of bronchial epithelial cells, which may have important implications in the development of both acute and chronic lung disease.
Collapse
Affiliation(s)
- Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA
- Correspondence address: Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA. Tel: +1-734-647-6477; Fax: +1-734-764-4556; E-mail:
| | - Priya Tripathi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Lada A Koneva
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Room 2017, Palmer Commons 100 Washtenaw Avenue Ann Arbor, MI 48109-2218, USA
| | - Raymond G Cavalcante
- Epigenomics Core, University of Michigan, Ann Arbor, Medical Science Research Building II Rm C568 1150 W. Medical Center Dr Ann Arbor, MI 48109, USA
| | - Nathan Craig
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Anne M Scruggs
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Room 2017, Palmer Commons 100 Washtenaw Avenue Ann Arbor, MI 48109-2218, USA
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Xueyuan Road 38, Haidian District, Beijing, China
| | - Yahong Chen
- Department of Respiratory Medicine, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, China
| |
Collapse
|
44
|
Chou YH, Tantoh DM, Wu MC, Tyan YS, Chen PH, Nfor ON, Hsu SY, Shen CY, Huang CN, Liaw YP. PM 2.5 exposure and DLEC1 promoter methylation in Taiwan Biobank participants. Environ Health Prev Med 2020; 25:68. [PMID: 33153431 PMCID: PMC7646067 DOI: 10.1186/s12199-020-00909-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Particulate matter (PM) < 2.5 μm (PM2.5) or fine PM is a serious public health concern. It affects DNA methylation and heightens carcinogenesis. Deleted in lung and esophageal cancer 1 (DLEC1) is a tumor suppressor gene. However, aberrant methylation of the gene is associated with several cancers. We evaluated the association between PM2.5 and DLEC1 promoter methylation in Taiwanese adults based on regular outdoor exercise. METHODS We obtained DNA methylation and exercise data of 496 participants (aged between 30 and 70 years) from the Taiwan Biobank (TWB) database. We also extracted PM2.5 data from the Air Quality Monitoring Database (AQMD) and estimated participants' exposure using residential addresses. RESULTS DLEC1 methylation and PM2.5 were positively associated: beta coefficient (β) = 0.114 × 10-3; p value = 0.046. The test for interaction between exercise and PM2.5 on DLEC1 methylation was significant (p value = 0.036). After stratification by exercise habits, PM2.5 and DLEC1 methylation remained significantly associated only among those who exercised regularly (β = 0.237 × 10-3; p value = 0.007). PM2.5 quartile-stratified analyses revealed an inverse association between regular exercise and DLEC1 methylation at PM2.5 < 27.37 μg/m3 (β = - 5.280 × 10-3; p value = 0.009). After combining exercise habits and PM2.5 quartiles, one stratum (i.e., regular exercise and PM2.5 < 27.37 μg/m3) was inversely associated with DLEC1 methylation (β = -5.160 × 10-3, p value = 0.007). CONCLUSIONS We found significant positive associations between PM2.5 and DLEC1 promoter methylation. Regular exercise at PM2.5 < 27.37 μg/m3 seemingly regulated DLEC1 promoter methylation.
Collapse
Affiliation(s)
- Ying-Hsiang Chou
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Disline Manli Tantoh
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Ming-Chi Wu
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- School of Medical Informatics, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Yeu-Sheng Tyan
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Pei-Hsin Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Shu-Yi Hsu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Chao-Yu Shen
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan.
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan.
| | - Chien-Ning Huang
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan.
- Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| | - Yung-Po Liaw
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- Medical Imaging and Big Data Center, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| |
Collapse
|
45
|
Traina G, Barbalace A, Betti F, Bolzacchini E, Bonini M, Contini D, Felice G, Foti T, Mantecca P. What impact of air pollution in pediatric respiratory allergic diseases. Pediatr Allergy Immunol 2020; 31 Suppl 26:26-28. [PMID: 33236436 DOI: 10.1111/pai.13362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
Respiratory allergies are known to affect people all over the world. Environmental factors related to pollution play a significant etiopathogenic role in this regard. Polluting sources are industrial activities and urban traffic, capable of generating various types of pollutants that trigger inflammatory, direct, and indirect damage to tissues, promoting allergic symptoms, even serious ones, and interfering with the pharmacologic response. They are also able to modify pollen, promoting allergic sensitization. Pollution could have played a significant predisposing role in the ongoing morbidity and mortality of SARS-CoV-2.
Collapse
Affiliation(s)
- Giovanni Traina
- Department of Pediatric and Neonatology, Cernusco sul Naviglio and Melzo-ASST Melegnano-Martesana, Milan, Italy
| | | | - Federica Betti
- Department of Pediatric and Neonatology, Cernusco sul Naviglio and Melzo-ASST Melegnano-Martesana, Milan, Italy
| | - Ezio Bolzacchini
- Department of Earth and Enviromental POLARIS, University of Milan Bicocca, Milan, Italy
| | - Maira Bonini
- Agency for health protection of metropolitan area of Milan-Hygiene and public health service, Milan west, Italy
| | - Daniele Contini
- Istituto di Scienze dell'Atmosfera e del Clima, ISAC-CNR, Lecce, Italy
| | - Giuseppe Felice
- Family Pediatrician, Metropolitan area of Milan, Milan, Italy
| | - Tiziana Foti
- Department of Pediatrics, Grande Ospedale Mettopolitano Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Paride Mantecca
- Department of Earth and Enviromental POLARIS, University of Milan Bicocca, Milan, Italy
| |
Collapse
|
46
|
Goobie GC, Nouraie M, Zhang Y, Kass DJ, Ryerson CJ, Carlsten C, Johannson KA. Air Pollution and Interstitial Lung Diseases: Defining Epigenomic Effects. Am J Respir Crit Care Med 2020; 202:1217-1224. [PMID: 32569479 PMCID: PMC7605178 DOI: 10.1164/rccm.202003-0836pp] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Gillian C. Goobie
- Department of Human Genetics, Graduate School of Public Health and
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Yingze Zhang
- Department of Human Genetics, Graduate School of Public Health and
- Department of Medicine and
| | | | - Christopher J. Ryerson
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada; and
| | - Christopher Carlsten
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, British Columbia, Canada; and
| | - Kerri A. Johannson
- Division of Respiratory Medicine, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
47
|
Li H, Ryu MH, Rider CF, Tse W, Clifford RL, Aristizabal MJ, Wen W, Carlsten C. Predominant DNMT and TET mediate effects of allergen on the human bronchial epithelium in a controlled air pollution exposure study. J Allergy Clin Immunol 2020; 147:1671-1682. [PMID: 33069714 DOI: 10.1016/j.jaci.2020.08.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/29/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Epidemiological data show that traffic-related air pollution contributes to the increasing prevalence and severity of asthma. DNA methylation (DNAm) changes may elucidate adverse health effects of environmental exposures. OBJECTIVES We sought to assess the effects of allergen and diesel exhaust (DE) exposures on global DNAm and its regulation enzymes in human airway epithelium. METHODS A total of 11 participants, including 7 with and 4 without airway hyperresponsiveness, were recruited for a randomized, double-blind crossover study. Each participant had 3 exposures: filtered air + saline, filtered air + allergen, and DE + allergen. Forty-eight hours postexposure, endobronchial biopsies and bronchoalveolar lavages were collected. Levels of DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) enzymes, 5-methylcytosine, and 5-hydroxymethylcytosine were determined by immunohistochemistry. Cytokines and chemokines in bronchoalveolar lavages were measured by electrochemiluminescence multiplex assays. RESULTS Predominant DNMT (the most abundant among DNMT1, DNMT3A, and DNMT3B) and predominant TET (the most abundant among TET1, TET2, and TET3) were participant-dependent. 5-Methylcytosine and its regulation enzymes differed between participants with and without airway hyperresponsiveness at baseline (filtered air + saline) and in response to allergen challenge (regardless of DE exposure). Predominant DNMT and predominant TET correlated with lung function. Allergen challenge effect on IL-8 in bronchoalveolar lavages was modified by TET2 baseline levels in the epithelium. CONCLUSIONS Response to allergen challenge is associated with key DNAm regulation enzymes. This relationship is generally unaltered by DE coexposure but is rather dependent on airway hyperresponsiveness status. These enzymes therefore warranted further inquiry regarding their potential in diagnosis, prognosis, and treatment of asthma.
Collapse
Affiliation(s)
- Hang Li
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Air Pollution Exposure Laboratory, Department of Medicine, Division of Respiratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Min Hyung Ryu
- Air Pollution Exposure Laboratory, Department of Medicine, Division of Respiratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher F Rider
- Air Pollution Exposure Laboratory, Department of Medicine, Division of Respiratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Wayne Tse
- Air Pollution Exposure Laboratory, Department of Medicine, Division of Respiratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachel L Clifford
- Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital, Nottingham, United Kingdom
| | - Maria J Aristizabal
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada; Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| | - Weiping Wen
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Chris Carlsten
- Air Pollution Exposure Laboratory, Department of Medicine, Division of Respiratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
48
|
Miller MR, Poland CA. Nanotoxicology: The Need for a Human Touch? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001516. [PMID: 32697439 DOI: 10.1002/smll.202001516] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/28/2020] [Indexed: 06/11/2023]
Abstract
With the ever-expanding number of manufactured nanomaterials (MNMs) under development there is a vital need for nanotoxicology studies that test the potential for MNMs to cause harm to health. An extensive body of work in cell cultures and animal models is vital to understanding the physicochemical characteristics of MNMs and the biological mechanisms that underlie any detrimental actions to cells and organs. In human subjects, exposure monitoring is combined with measurement of selected health parameters in small panel studies, especially in occupational settings. However, the availability of further in vivo human data would greatly assist the risk assessment of MNMs. Here, the potential for controlled inhalation exposures of MNMs in human subjects is discussed. Controlled exposures to carbon, gold, aluminum, and zinc nanoparticles in humans have already set a precedence to demonstrate the feasibility of this approach. These studies have provided considerable insight into the potential (or not) of nanoparticles to induce inflammation, alter lung function, affect the vasculature, reach the systemic circulation, and accumulate in other organs. The need for further controlled exposures of MNMs in human volunteers - to establish no-effect limits, biological mechanisms, and provide vital data for the risk assessment of MNMs - is advocated.
Collapse
Affiliation(s)
- Mark R Miller
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Craig A Poland
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
49
|
Li CH, Sayeau K, Ellis AK. Air Pollution and Allergic Rhinitis: Role in Symptom Exacerbation and Strategies for Management. J Asthma Allergy 2020; 13:285-292. [PMID: 32922045 PMCID: PMC7457822 DOI: 10.2147/jaa.s237758] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/12/2020] [Indexed: 01/24/2023] Open
Abstract
This article reviews the current understanding of the role of air pollution in both the symptom exacerbation and rising prevalence of allergic rhinitis (AR) for the development of future AR therapeutics and management strategies. We discuss the epidemiological evidence for this relationship through birth cohort studies, the economic impact of AR, and the influence of air pollution through the lens of the exposome framework of allergic disease development. This is followed by a discussion on the influence of diesel exhaust and diesel exhaust particles (DEP) from motor vehicle emissions and their implication in the rising prevalence of allergic disease and allergic sensitization through triggering inflammatory signalling pathways that exacerbate AR symptoms. Finally, a summary is provided of clinical trials assessing the influence of air pollution on AR with a depiction of currently available therapies and management strategies. Future directions in the development of AR modalities given the air pollution-mediated symptom exacerbation are challenged with unfolding the complex gene–environment interaction product of heterogenous AR presentation.
Collapse
Affiliation(s)
- Carmen H Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, ON, Canada
| | - Kyle Sayeau
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, ON, Canada
| | - Anne K Ellis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Allergy Research Unit, Kingston Health Sciences Center - KGH Site, Kingston, ON, Canada.,Department of Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
50
|
Alashkar Alhamwe B, Miethe S, Pogge von Strandmann E, Potaczek DP, Garn H. Epigenetic Regulation of Airway Epithelium Immune Functions in Asthma. Front Immunol 2020; 11:1747. [PMID: 32973742 PMCID: PMC7461869 DOI: 10.3389/fimmu.2020.01747] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the respiratory tract characterized by recurrent breathing problems resulting from airway obstruction and hyperresponsiveness. Human airway epithelium plays an important role in the initiation and control of the immune responses to different types of environmental factors contributing to asthma pathogenesis. Using pattern recognition receptors airway epithelium senses external stimuli, such as allergens, microbes, or pollutants, and subsequently secretes endogenous danger signaling molecules alarming and activating dendritic cells. Hence, airway epithelial cells not only mediate innate immune responses but also bridge them with adaptive immune responses involving T and B cells that play a crucial role in the pathogenesis of asthma. The effects of environmental factors on the development of asthma are mediated, at least in part, by epigenetic mechanisms. Those comprise classical epigenetics including DNA methylation and histone modifications affecting transcription, as well as microRNAs influencing translation. The common feature of such mechanisms is that they regulate gene expression without affecting the nucleotide sequence of the genomic DNA. Epigenetic mechanisms play a pivotal role in the regulation of different cell populations involved in asthma pathogenesis, with the remarkable example of T cells. Recently, however, there is increasing evidence that epigenetic mechanisms are also crucial for the regulation of airway epithelial cells, especially in the context of epigenetic transfer of environmental effects contributing to asthma pathogenesis. In this review, we summarize the accumulating evidence for this very important aspect of airway epithelial cell pathobiology.
Collapse
Affiliation(s)
- Bilal Alashkar Alhamwe
- Institute of Laboratory Medicine, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,College of Pharmacy, International University for Science and Technology (IUST), Daraa, Syria.,Center for Tumor Biology and Immunology, Institute of Tumor Immunology, Philipps University Marburg, Marburg, Germany
| | - Sarah Miethe
- Institute of Laboratory Medicine, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Elke Pogge von Strandmann
- Center for Tumor Biology and Immunology, Institute of Tumor Immunology, Philipps University Marburg, Marburg, Germany
| | - Daniel P Potaczek
- Institute of Laboratory Medicine, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,John Paul II Hospital, Kraków, Poland
| | - Holger Garn
- Institute of Laboratory Medicine, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| |
Collapse
|