1
|
Park BJ, Dhong KR, Park HJ. Cordyceps militaris Grown on Germinated Rhynchosia nulubilis (GRC) Encapsulated in Chitosan Nanoparticle (GCN) Suppresses Particulate Matter (PM)-Induced Lung Inflammation in Mice. Int J Mol Sci 2024; 25:10642. [PMID: 39408971 PMCID: PMC11477187 DOI: 10.3390/ijms251910642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Cordyceps militaris grown on germinated Rhynchosia nulubilis (GRC) exerts various biological effects, including anti-allergic, anti-inflammatory, and immune-regulatory effects. In this study, we investigated the anti-inflammatory effects of GRC encapsulated in chitosan nanoparticles (CN) against particulate matter (PM)-induced lung inflammation. Optimal CN (CN6) (CHI: TPP w/w ratio of 4:1; TPP pH 2) exhibited a zeta potential of +22.77 mV, suitable for GRC encapsulation. At different GRC concentrations, higher levels (60 and 120 mg/mL) led to increased negative zeta potential, enhancing stability. The optimal GRC concentration for maximum entrapment (31.4 ± 1.35%) and loading efficiency (7.6 ± 0.33%) of GRC encapsulated in CN (GCN) was 8 mg/mL with a diameter of 146.1 ± 54 nm and zeta potential of +30.68. In vivo studies revealed that administering 300 mg/kg of GCN significantly decreased the infiltration of macrophages and T cells in the lung tissues of PM-treated mice, as shown by immunohistochemical analysis of CD4 and F4/80 markers. Additionally, GCN ameliorated PM-induced lung tissue damage, inflammatory cell infiltration, and alveolar septal hypertrophy. GCN also decreased total cells and neutrophils, showing notable anti-inflammatory effects in the bronchoalveolar lavage fluid (BALF) from PM-exposed mice, compared to GRC. Next the anti-inflammatory properties of GCN were further explored in PM- and LPS-exposed RAW264.7 cells; it significantly reduced PM- and LPS-induced cell death, NO production, and levels of inflammatory cytokine mRNAs (IL-1β, IL-6, and COX-2). GCN also suppressed NF-κB/MAPK signaling pathways by reducing levels of p-NF-κB, p-ERK, and p-c-Jun proteins, indicating its potential in managing PM-related inflammatory lung disease. Furthermore, GCN significantly reduced PM- and LPS-induced ROS production. The enhanced bioavailability of GRC components was demonstrated by an increase in fluorescence intensity in the intestinal absorption study using FITC-GCN. Our data indicated that GCN exhibited enhanced bioavailability and potent anti-inflammatory and antioxidant effects in cells and in vivo, making it a promising candidate for mitigating PM-induced lung inflammation and oxidative stress.
Collapse
Affiliation(s)
- Byung-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| | - Kyu-Ree Dhong
- Magicbullettherapeutics Inc., 150 Yeongdeungpo-ro, Yeongdeungpo-gu, Seoul 07292, Republic of Korea;
| | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| |
Collapse
|
2
|
Zeng F, Pang G, Hu L, Sun Y, Peng W, Chen Y, Xu D, Xia Q, Zhao L, Li Y, He M. Subway Fine Particles (PM 2.5)-Induced Pro-Inflammatory Response Triggers Airway Epithelial Barrier Damage Through the TLRs/NF-κB-Dependent Pathway In Vitro. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 39189708 DOI: 10.1002/tox.24403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024]
Abstract
Subways are widely used in major cities around the world, and subway fine particulate matter (PM2.5) is the main source of daily PM2.5 exposure for urban residents. Exposure to subway PM2.5 leads to acute inflammatory damage in humans, which has been confirmed in mouse in vivo studies. However, the concrete mechanism by which subway PM2.5 causes airway damage remains obscure. In this study, we found that subway PM2.5 triggered release of pro-inflammatory cytokines such as interleukin 17E, tumor necrosis factor α, transforming growth factor β, and thymic stromal lymphopoietin from human bronchial epithelial cells (BEAS-2B) in a dose-effect relationship. Subsequently, supernatant recovered from the subway PM2.5 group significantly increased expression of the aforementioned cytokines in BEAS-2B cells compared with the subway PM2.5 group. Additionally, tight junctions (TJs) of BEAS-2B cells including zonula occludens-1, E-cadherin, and occludin were decreased by subway PM2.5 in a dose-dependent manner. Moreover, supernatant recovered from the subway PM2.5 group markedly decreased the expression of these TJs compared with the control group. Furthermore, inhibitors of toll-like receptors (TLRs) and nuclear factor-kappa B (NF-κB), as well as chelate resins (e.g., chelex) and deferoxamine, remarkably ameliorated the observed changes of cytokines and TJs caused by subway PM2.5 in BEAS-2B cells. Therefore, these results suggest that subway PM2.5 induced a decline of TJs after an initial ascent of cytokine expression, and subway PM2.5 altered expression of both cytokines and TJs by activating TLRs/NF-κB-dependent pathway in BEAS-2B cells. The metal components of subway PM2.5 may contribute to the airway epithelial injury.
Collapse
Affiliation(s)
- Fanmei Zeng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Guanhua Pang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Liwen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Yuan Sun
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Wen Peng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yuwei Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Dan Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Qing Xia
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Luwei Zhao
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yifei Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Miao He
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Shenyang, China
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, China
| |
Collapse
|
3
|
Zhang T, Lui KH, Ho SSH, Chen J, Chuang HC, Ho KF. Characterization of airborne endotoxin in personal exposure to fine particulate matter (PM 2.5) and bioreactivity for elderly residents in Hong Kong. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116530. [PMID: 38833976 DOI: 10.1016/j.ecoenv.2024.116530] [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: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
The heavy metals and bioreactivity properties of endotoxin in personal exposure to fine particulate matter (PM2.5) were characterized in the analysis. The average personal exposure concentrations to PM2.5 were ranged from 6.8 to 96.6 μg/m3. The mean personal PM2.5 concentrations in spring, summer, autumn, and winter were 32.1±15.8, 22.4±11.8, 35.3±11.9, and 50.2±19.9 μg/m3, respectively. There were 85 % of study targets exceeded the World Health Organization (WHO) PM2.5 threshold (24 hours). The mean endotoxin concentrations ranged from 1.086 ± 0.384-1.912 ± 0.419 EU/m3, with a geometric mean (GM) varied from 1.034 to 1.869. The concentration of iron (Fe) (0.008-1.16 μg/m3) was one of the most abundant transition metals in the samples that could affect endotoxin toxicity under Toll-Like Receptor 4 (TLR4) stimulation. In summer, the interleukin 6 (IL-6) levels showed statistically significant differences compared to other seasons. Spearman correlation analysis showed endotoxin concentrations were positively correlated with chromium (Cr) and nickel (Ni), implying possible roles as nutrients and further transport via adhering to the surface of fine inorganic particles. Mixed-effects model analysis demonstrated that Tumor necrosis factor-α (TNF-α) production was positively associated with endotoxin concentration and Cr as a combined exposure factor. The Cr contained the highest combined effect (0.205-0.262), suggesting that Cr can potentially exacerbate the effect of endotoxin on inflammation and oxidative stress. The findings will be useful for practical policies for mitigating air pollution to protect the public health of the citizens.
Collapse
Affiliation(s)
- Tianhang Zhang
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Hei Lui
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Steven Sai Hang Ho
- Division of Atmosphere Sciences, Desert Research Institute, Reno, NV 89512, United States; Hong Kong Premium Services and Research Laboratory, Cheung Sha Wan, Kowloon, Hong Kong, China
| | - Jiayao Chen
- School of Architecture, Planning and Environmental Policy, University College Dublin, Dublin, Ireland
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kin Fai Ho
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Zhao H, Zhan C, Li B, Fang Z, Zhong M, He Y, Chen F, Chen Z, Zhang G, Zhong N, Lai K, Chen R. Non-allergic eosinophilic inflammation and airway hyperresponsiveness induced by diesel engine exhaust through activating ILCs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116403. [PMID: 38710145 DOI: 10.1016/j.ecoenv.2024.116403] [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: 11/30/2023] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
RATIONALE Diesel engine exhaust (DEE) is associated with the development and exacerbation of asthma. Studies have shown that DEE can aggravate allergen-induced eosinophilic inflammation in lung. However, it remains not clear that whether DEE alone could initiate non-allergic eosinophilic inflammation and airway hyperresponsiveness (AHR) through innate lymphoid cells (ILCs) pathway. OBJECTIVE This study aims to investigate the airway inflammation and hyperresponsiveness and its relationship with ILC after DEE exposure. METHOD Non-sensitized BALB/c mice were exposed in the chamber of diesel exhaust or filtered air for 2, 4, and 6 weeks (4 h/day, 6 days/week). Anti-CD4 mAb or anti-Thy1.2 mAb was administered by intraperitoneal injection to inhibit CD4+T or ILCs respectively. AHR、airway inflammation and ILCs were assessed. RESULT DEE exposure induced significantly elevated level of neutrophils, eosinophils, collagen content at 4, 6 weeks. Importantly, the airway AHR was only significant in the 4weeks-DEE exposure group. No difference of the functional proportions of Th2 cells was found between exposure group and control group. The proportions of IL-5+ILC2, IL-17+ILC significantly increased in 2, 4weeks-DEE exposure group. After depletion of CD4+T cells, both the proportion of IL-5+ILC2 and IL-17A ILCs was higher in the 4weeks-DEE exposure group which induced AHR, neutrophilic and eosinophilic inflammation accompanied by the IL-5, IL-17A levels. CONCLUSION Diesel engine exhaust alone can imitate asthmatic characteristics in mice model. Lung-resident ILCs are one of the major effectors cells responsible for a mixed Th2/Th17 response and AHR.
Collapse
Affiliation(s)
- Huasi Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R.China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China
| | - Bizhou Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; Department of Respiratory Medicine, Guangzhou Panyu Central Hospital, Guangzhou, P.R.China
| | - Zhangfu Fang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, P.R.China
| | - Mingyu Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China
| | - Yaowei He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China
| | - Fagui Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhe Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China
| | - Guojun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R.China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; Guangzhou National Lab, Guangzhou, P.R.China.
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critial Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; Guangzhou National Lab, Guangzhou, P.R.China.
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R.China; Guangzhou National Lab, Guangzhou, P.R.China.
| |
Collapse
|
5
|
Bhujel B, Oh S, Hur W, Lee S, Chung HS, Lee H, Park JH, Kim JY. Effect of Exposure to Particulate Matter on the Ocular Surface in an Experimental Allergic Eye Disease Mouse Model. Bioengineering (Basel) 2024; 11:498. [PMID: 38790364 PMCID: PMC11118833 DOI: 10.3390/bioengineering11050498] [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: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
In response to the escalating concern over the effect of environmental factors on ocular health, this study aimed to investigate the impact of air pollution-associated particulate matter (PM) on ocular allergy and inflammation. C57BL/6 mice were sensitized with ovalbumin (OVA) topically and aluminum hydroxide via intraperitoneal injection. Two weeks later, the mice were challenged with OVA and exposed to PM. Three groups-naive, OVA, and OVA-sensitized with PM exposure (OVA + PM) groups-were induced to an Allergic Eye disease (AED) model. Parameters including clinical signs, histological changes, inflammatory cell infiltration, serum OVA-specific immunoglobulins E (IgE) levels, mast cells degranulation, cellular apoptosis and T-cell cytokines were studied. The results demonstrate that exposure with PM significantly exacerbates ocular allergy, evidenced by increased eye-lid edema, mast cell degranulation, inflammatory cytokines (IL-4, IL-5 and TNF-α), cell proliferation (Ki67), and serum IgE, polymorphonuclear leukocytes (PMN), and apoptosis and reduced goblet cells. These findings elucidate the detrimental impact of PM exposure on exacerbating the severity of AED. Noticeably, diminished goblet cells highlight disruptions in ocular surface integrity, while increased PMN infiltration with an elevated production of IgE signifies a systemic allergic response with inflammation. In conclusion, this study not only scientifically substantiates the association between air pollution, specifically PM, and ocular health, but also underscores the urgency for further exploration and targeted interventions to mitigate the detrimental effects of environmental pollutants on ocular surfaces.
Collapse
Affiliation(s)
- Basanta Bhujel
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Seheon Oh
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Woojune Hur
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Seorin Lee
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
- Department of Medical Science, University of Ulsan Graduate School, Seoul 05505, Republic of Korea
| | - Ho Seok Chung
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
| | - Hun Lee
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
| | | | - Jae Yong Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea; (B.B.); (S.O.); (W.H.); (S.L.); (H.S.C.); (H.L.)
| |
Collapse
|
6
|
Skevaki C, Nadeau KC, Rothenberg ME, Alahmad B, Mmbaga BT, Masenga GG, Sampath V, Christiani DC, Haahtela T, Renz H. Impact of climate change on immune responses and barrier defense. J Allergy Clin Immunol 2024; 153:1194-1205. [PMID: 38309598 DOI: 10.1016/j.jaci.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/05/2024]
Abstract
Climate change is not just jeopardizing the health of our planet but is also increasingly affecting our immune health. There is an expanding body of evidence that climate-related exposures such as air pollution, heat, wildfires, extreme weather events, and biodiversity loss significantly disrupt the functioning of the human immune system. These exposures manifest in a broad range of stimuli, including antigens, allergens, heat stress, pollutants, microbiota changes, and other toxic substances. Such exposures pose a direct and indirect threat to our body's primary line of defense, the epithelial barrier, affecting its physical integrity and functional efficacy. Furthermore, these climate-related environmental stressors can hyperstimulate the innate immune system and influence adaptive immunity-notably, in terms of developing and preserving immune tolerance. The loss or failure of immune tolerance can instigate a wide spectrum of noncommunicable diseases such as autoimmune conditions, allergy, respiratory illnesses, metabolic diseases, obesity, and others. As new evidence unfolds, there is a need for additional research in climate change and immunology that covers diverse environments in different global settings and uses modern biologic and epidemiologic tools.
Collapse
Affiliation(s)
- Chrysanthi Skevaki
- Institute of Laboratory Medicine, member of the German Center for Lung Research and the Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Barrak Alahmad
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass; Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Blandina T Mmbaga
- Kilimanjaro Christian Medical University College, Moshi, Tanzania; Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | - Gileard G Masenga
- Kilimanjaro Christian Medical University College, Moshi, Tanzania; Department of Obstetrics and Gynecology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Vanitha Sampath
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Mass; Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Harald Renz
- Institute of Laboratory Medicine, member of the German Center for Lung Research and the Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany; Kilimanjaro Christian Medical University College, Moshi, Tanzania; Department of Clinical Immunology and Allergology, Laboratory of Immunopathology, Sechenov University, Moscow, Russia.
| |
Collapse
|
7
|
Cafora M, Rovelli S, Cattaneo A, Pistocchi A, Ferrari L. Short-term exposure to fine particulate matter exposure impairs innate immune and inflammatory responses to a pathogen stimulus: A functional study in the zebrafish model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 348:123841. [PMID: 38521398 DOI: 10.1016/j.envpol.2024.123841] [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: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Short-term exposure to fine particulate matter (PM2.5) is associated with the activation of adverse inflammatory responses, increasing the risk of developing acute respiratory diseases, such as those caused by pathogen infections. However, the functional mechanisms underlying this evidence remain unclear. In the present study, we generated a zebrafish model of short-term exposure to a specific PM2.5, collected in the northern metropolitan area of Milan, Italy. First, we assessed the immunomodulatory effects of short-term PM2.5 exposure and observed that it elicited pro-inflammatory effects by inducing the expression of cytokines and triggering hyper-activation of both neutrophil and macrophage cell populations. Moreover, we examined the impact of a secondary infectious pro-inflammatory stimulus induced through the injection of Pseudomonas aeruginosa lipopolysaccharide (Pa-LPS) molecules after exposure to short-term PM2.5. In this model, we demonstrated that the innate immune response was less responsive to a second pro-inflammatory infectious stimulus. Indeed, larvae exhibited dampened leukocyte activation and impaired production of reactive oxygen species. The obtained results indicate that short-term PM2.5 exposure alters the immune microenvironment and affects the inflammatory processes, thus potentially weakening the resistance to pathogen infections.
Collapse
Affiliation(s)
- Marco Cafora
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sabrina Rovelli
- RAHH LAB, Department of Science and High Technology, University of Insubria, Como, Italy
| | - Andrea Cattaneo
- RAHH LAB, Department of Science and High Technology, University of Insubria, Como, Italy
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Unit of Occupational Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy.
| |
Collapse
|
8
|
Lira GVDAG, da Silva GAP, Bezerra PGDM, Sarinho ESC. Avoidance of Inhaled Pollutants and Irritants in Asthma from a Salutogenic Perspective. J Asthma Allergy 2024; 17:237-250. [PMID: 38524100 PMCID: PMC10960548 DOI: 10.2147/jaa.s445864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/19/2023] [Indexed: 03/26/2024] Open
Abstract
Much is known about the role of aeroallergens in asthma, but little is described about the damage caused by inhaled pollutants and irritants to the respiratory epithelium. In this context, the most frequent pollutants and irritants inhaled in the home environment were identified, describing the possible repercussions that may occur in the respiratory tract of the pediatric population with asthma and highlighting the role of the caregiver in environmental control through a salutogenic perspective. Searches were carried out in the MEDLINE/PubMed, Web of Science, Lilacs and Scopus databases for articles considered relevant for the theoretical foundation of this integrative review, in which interactions between exposure to pollutants and inhaled irritants and lung involvement. Articles published in the last 10 years that used the following descriptors were considered: air pollution; tobacco; particulate matter; disinfectants; hydrocarbons, fluorinated; odorants; chloramines; pesticide; asthma; and beyond Antonovsky's sense of coherence. Exposure to smoke and some substances found in cleaning products, such as benzalkonium chloride, ethylenediaminetetraacetic acid and monoethanolamine, offer potential risks for sensitization and exacerbation of asthma. The vast majority of the seven main inhaled products investigated provoke irritative inflammatory reactions and oxidative imbalance in the respiratory epithelium. In turn, the caregiver's role is essential in health promotion and the clinical control of paediatric asthma. From a salutogenic point of view, pollutants and irritants inhaled at home should be carefully investigated in the clinical history so that strategies to remove or reduce exposures can be used by caregivers of children and adolescents with asthma.
Collapse
Affiliation(s)
- Georgia Véras de Araújo Gueiros Lira
- Allergy and Immunology Research Centre, Federal University of Pernambuco, Recife, PE, Brazil
- Department of Paediatrics, Federal University of Pernambuco, Recife, PE, Brazil
| | | | | | - Emanuel S C Sarinho
- Allergy and Immunology Research Centre, Federal University of Pernambuco, Recife, PE, Brazil
- Department of Paediatrics, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
9
|
Park E, Kim BY, Lee S, Son KH, Bang J, Hong SH, Lee JW, Uhm KO, Kwak HJ, Lim HJ. Diesel exhaust particle exposure exacerbates ciliary and epithelial barrier dysfunction in the multiciliated bronchial epithelium models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116090. [PMID: 38364346 DOI: 10.1016/j.ecoenv.2024.116090] [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: 09/25/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
Airway epithelium, the first defense barrier of the respiratory system, facilitates mucociliary clearance against inflammatory stimuli, such as pathogens and particulates inhaled into the airway and lung. Inhaled particulate matter 2.5 (PM2.5) can penetrate the alveolar region of the lung, and it can develop and exacerbate respiratory diseases. Although the pathophysiological effects of PM2.5 in the respiratory system are well known, its impact on mucociliary clearance of airway epithelium has yet to be clearly defined. In this study, we used two different 3D in vitro airway models, namely the EpiAirway-full-thickness (FT) model and a normal human bronchial epithelial cell (NHBE)-based air-liquid interface (ALI) system, to investigate the effect of diesel exhaust particles (DEPs) belonging to PM2.5 on mucociliary clearance. RNA-sequencing (RNA-Seq) analyses of EpiAirway-FT exposed to DEPs indicated that DEP-induced differentially expressed genes (DEGs) are related to ciliary and microtubule function and inflammatory-related pathways. The exposure to DEPs significantly decreased the number of ciliated cells and shortened ciliary length. It reduced the expression of cilium-related genes such as acetylated α-tubulin, ARL13B, DNAH5, and DNAL1 in the NHBEs cultured in the ALI system. Furthermore, DEPs significantly increased the expression of MUC5AC, whereas they decreased the expression of epithelial junction proteins, namely, ZO1, Occludin, and E-cadherin. Impairment of mucociliary clearance by DEPs significantly improved the release of epithelial-derived inflammatory and fibrotic mediators such as IL-1β, IL-6, IL-8, GM-CSF, MMP-1, VEGF, and S100A9. Taken together, it can be speculated that DEPs can cause ciliary dysfunction, hyperplasia of goblet cells, and the disruption of the epithelial barrier, resulting in the hyperproduction of lung injury mediators. Our data strongly suggest that PM2.5 exposure is directly associated with ciliary and epithelial barrier dysfunction and may exacerbate lung injury.
Collapse
Affiliation(s)
- Eunsook Park
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea
| | - Bu-Yeo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, South Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, South Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 215565, South Korea
| | - Jihye Bang
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea
| | - Se Hyang Hong
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea
| | - Joong Won Lee
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea
| | - Kyung-Ok Uhm
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea
| | - Hyun-Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin Univerisity, Seonbuk-Gu, Seoul 02707, South Korea
| | - Hyun Joung Lim
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Chungju, Chungcheongbuk-do 28159, South Korea.
| |
Collapse
|
10
|
Lin TY, Makrufardi F, Tung NT, Manullang A, Chang PJ, Lo CY, Chiu TH, Tung PH, Lin CH, Lin HC, Wang CH, Lin SM. Different Impacts of Traffic-Related Air Pollution on Early-Onset and Late-Onset Asthma. J Asthma Allergy 2024; 17:195-208. [PMID: 38505396 PMCID: PMC10949997 DOI: 10.2147/jaa.s451725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Abstract
Background Early-onset asthma (EOA) and late-onset asthma (LOA) are two distinct phenotypes. Air pollution has been associated with an increase in poorer asthma outcomes. The objective of this study was to examine the effects of traffic-related air pollution (TRAP) on asthma outcomes in EOA and LOA patients. Methods A cross-sectional study was conducted on 675 asthma patients (LOA: 415) recruited from a major medical center in Taiwan. The land-use regression (LUR) model was used to estimate the level of exposure to PM10, PM2.5, NO2, and O3 on an individual level. We investigated the association between TRAP and asthma outcomes in EOA and LOA patients, stratified by allergic sensitization status, using a regression approach. Results An increase in PM10 was associated with younger age of onset, increased asthma duration, and decreased lung function in EOA patients (p<0.05). An increase in PM10 was associated with older age of onset, and decreased asthma duration, eosinophil count, and Asthma Control Test (ACT) score in LOA patients. An increase in PM2.5 was associated with younger age of onset, increased asthma duration, decreased eosinophil count, and lung function in EOA patients (p<0.05). An increase in PM2.5 was associated with decreased lung function and ACT score in LOA patients. An increase in NO2 was associated with increased eosinophil count and decreased lung function in EOA patients (p<0.05). An increase in O3 was associated with decreased lung function in LOA patients (p<0.05). In addition, associations of TRAP with age of onset and eosinophil counts were mainly observed in both EOA and LOA patients with allergic sensitization, and an association with ACT was mainly observed in LOA patients without allergic sensitization. Conclusion The impact of TRAP on age of onset, eosinophil count, and lung function in EOA patients, and ACT in LOA patients, was affected by the status of allergic sensitization.
Collapse
Affiliation(s)
- Ting-Yu Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Firdian Makrufardi
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Nguyen Thanh Tung
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Otorhinolaryngology Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Amja Manullang
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Jui Chang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Hsuan Chiu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Pi-Hung Tung
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chiung-Hung Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
11
|
Kayalar Ö, Rajabi H, Konyalilar N, Mortazavi D, Aksoy GT, Wang J, Bayram H. Impact of particulate air pollution on airway injury and epithelial plasticity; underlying mechanisms. Front Immunol 2024; 15:1324552. [PMID: 38524119 PMCID: PMC10957538 DOI: 10.3389/fimmu.2024.1324552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
Air pollution plays an important role in the mortality and morbidity of chronic airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Particulate matter (PM) is a significant fraction of air pollutants, and studies have demonstrated that it can cause airway inflammation and injury. The airway epithelium forms the first barrier of defense against inhaled toxicants, such as PM. Airway epithelial cells clear airways from inhaled irritants and orchestrate the inflammatory response of airways to these irritants by secreting various lipid mediators, growth factors, chemokines, and cytokines. Studies suggest that PM plays an important role in the pathogenesis of chronic airway diseases by impairing mucociliary function, deteriorating epithelial barrier integrity, and inducing the production of inflammatory mediators while modulating the proliferation and death of airway epithelial cells. Furthermore, PM can modulate epithelial plasticity and airway remodeling, which play central roles in asthma and COPD. This review focuses on the effects of PM on airway injury and epithelial plasticity, and the underlying mechanisms involving mucociliary activity, epithelial barrier function, airway inflammation, epithelial-mesenchymal transition, mesenchymal-epithelial transition, and airway remodeling.
Collapse
Affiliation(s)
- Özgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Hadi Rajabi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Deniz Mortazavi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Gizem Tuşe Aksoy
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Jun Wang
- Department of Biomedicine and Biopharmacology, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Türkiye
| |
Collapse
|
12
|
Xiong A, He X, Liu S, Ran Q, Zhang L, Wang J, Jiang M, Niu B, Xiong Y, Li G. Oxidative stress-mediated activation of FTO exacerbates impairment of the epithelial barrier by up-regulating IKBKB via N6-methyladenosine-dependent mRNA stability in asthmatic mice exposed to PM2.5. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116067. [PMID: 38325270 DOI: 10.1016/j.ecoenv.2024.116067] [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: 10/24/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
In order to comprehend the underlying mechanisms contributing to the development and exacerbation of asthma resulting from exposure to fine particulate matter (PM2.5), we established an asthmatic model in fat mass and obesity-associated gene knockdown mice subjected to PM2.5 exposure. Histological analyses using hematoxylin-eosin (HE) and Periodic Acid-Schiff (PAS) staining revealed that the down-regulation of the fat mass and obesity-associated gene (Fto) expression significantly ameliorated the pathophysiological alterations observed in asthmatic mice exposed to PM2.5. Furthermore, the down-regulation of Fto gene expression effectively attenuated damage to the airway epithelial barrier. Additionally, employing in vivo and in vitro models, we elucidated that PM2.5 modulated FTO expression by inducing oxidative stress. Asthmatic mice exposed to PM2.5 exhibited elevated Fto expression, which correlated with increased levels of reactive oxygen species. Similarly, when cells were exposed to PM2.5, FTO expression was up-regulated in a ROS-dependent manner. Notably, the administration of N-acetyl cysteine successfully reversed the PM2.5-induced elevation in FTO expression. Concurrently, we performed transcriptome-wide Methylated RNA immunoprecipitation Sequencing (MeRIP-seq) analysis subsequent to PM2.5 exposure. Through the implementation of Gene Set Enrichment Analysis and m6A-IP-qPCR, we successfully identified inhibitor of nuclear factor kappa B kinase subunit beta (IKBKB) as a target gene regulated by FTO. Interestingly, exposure to PM2.5 led to increased expression of IKBKB, while m6A modification on IKBKB mRNA was reduced. Furthermore, our investigation revealed that PM2.5 also regulated IKBKB through oxidative stress. Significantly, the down-regulation of IKBKB effectively mitigated epithelial barrier damage in cells exposed to PM2.5 by modulating nuclear factor-kappa B (NF-κB) signaling. Importantly, we discovered that decreased m6A modification on IKBKB mRNA facilitated by FTO enhanced its stability, consequently resulting in up-regulation of IKBKB expression. Collectively, our findings propose a novel role for FTO in the regulation of IKBKB through m6A-dependent mRNA stability in the context of PM2.5-induced oxidative stress. Therefore, it is conceivable that the utilization of antioxidants or inhibition of FTO could represent potential therapeutic strategies for the management of asthma exacerbated by PM2.5 exposure.
Collapse
Affiliation(s)
- Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China; National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China.
| | - Shengbin Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Qin Ran
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Manling Jiang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Bin Niu
- Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan friendship hospital, Chengdu 610000, China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu third people's hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China.
| |
Collapse
|
13
|
Immormino RM, Smeekens JM, Mathai PI, Clough KM, Nguyen JT, Ghio AJ, Cook DN, Kulis MD, Moran TP. Different airborne particulates trigger distinct immune pathways leading to peanut allergy in a mouse model. Allergy 2024; 79:432-444. [PMID: 37804001 PMCID: PMC11017991 DOI: 10.1111/all.15908] [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: 04/10/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Environmental exposure to peanut through non-oral routes is a risk factor for peanut allergy. Early-life exposure to air pollutants, including particulate matter (PM), is associated with sensitization to foods through unknown mechanisms. We investigated whether PM promotes sensitization to environmental peanut and the development of peanut allergy in a mouse model. METHODS C57BL/6J mice were co-exposed to peanut and either urban particulate matter (UPM) or diesel exhaust particles (DEP) via the airways and assessed for peanut sensitization and development of anaphylaxis following peanut challenge. Peanut-specific CD4+ T helper (Th) cell responses were characterized by flow cytometry and Th cytokine production. Mice lacking select innate immune signaling genes were used to study mechanisms of PM-induced peanut allergy. RESULTS Airway co-exposure to peanut and either UPM- or DEP-induced systemic sensitization to peanut and anaphylaxis following peanut challenge. Exposure to UPM or DEP triggered activation and migration of lung dendritic cells to draining lymph nodes and induction of peanut-specific CD4+ Th cells. UPM- and DEP-induced distinct Th responses, but both stimulated expansion of T follicular helper (Tfh) cells essential for peanut allergy development. MyD88 signaling was critical for UPM- and DEP-induced peanut allergy, whereas TLR4 signaling was dispensable. DEP-induced peanut allergy and Tfh-cell differentiation depended on IL-1 but not IL-33 signaling, whereas neither cytokine alone was necessary for UPM-mediated sensitization. CONCLUSION Environmental co-exposure to peanut and PM induces peanut-specific Tfh cells and peanut allergy in mice.
Collapse
Affiliation(s)
- Robert M. Immormino
- Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Johanna M. Smeekens
- Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC Food Allergy Initiative, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Priscilla I. Mathai
- Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Katelyn M. Clough
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | | - Andrew J. Ghio
- Human Studies Facility, United States Environmental Protection Agency, Chapel Hill, North Carolina, USA
| | - Donald N. Cook
- Division of Intramural Research, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, North Carolina, USA
| | - Michael D. Kulis
- Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC Food Allergy Initiative, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Timothy P. Moran
- Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
14
|
Park HW, Lee HS. IL-23 contributes to Particulate Matter induced allergic asthma in the early life of mice and promotes asthma susceptibility. J Mol Med (Berl) 2024; 102:129-142. [PMID: 37994911 DOI: 10.1007/s00109-023-02393-6] [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: 05/16/2023] [Revised: 10/10/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Air pollutant exposure leads to and exacerbates respiratory diseases. Particulate Matter (PM) is a major deleterious factor in the pathophysiology of asthma. Nonetheless, studies on the effects and mechanisms of exposure in the early life of mice remain unresolved. This study aimed to investigate changes in allergic phenotypes and effects on allergen-specific memory T cells resulting from co-exposure of mice in the early life to PM and house dust mites (HDM) and to explore the role of interleukin-23 (IL-23) in this process. PM and low-dose HDM were administered intranasally in 4-day-old C57BL/6 mice. After confirming an increase in IL-23 expression in mouse lung tissues, changes in the asthma phenotype and lung effector/memory Th2 or Th17 cells were evaluated after intranasal administration of anti-IL-23 antibody (Ab) during co-exposure to PM and HDM. Evaluation was performed up to 7 weeks after the last administration. Co-exposure to PM and low-dose HDM resulted in increases in airway hyperresponsiveness (AHR), eosinophils, neutrophils, and persistent Th2/Th17 effector/memory cells, which were all inhibited by anti-IL-23 Ab administration. When low-dose HDM was administered twice after a 7-week rest, mice exposed to PM and HDM during the previous early life period exhibited re-increases AHR, eosinophil count, HDM-specific IgG1, and effector/memory Th2 and Th17 cell populations. However, anti-IL-23 Ab administration during the early life period resulted in inhibition. Co-exposure to PM and low-dose HDM reinforced the allergic phenotypes and allergen-specific memory responses in early life of mice. During this process, IL-23 contributes to the enhancement of effector/memory Th2/Th17 cells and allergic phenotypes. KEY MESSAGES: PM-induced IL-23 expression, allergic responses in HDMinstilled mice of early life period. PM-induced effector/memory Th2/Th17 cells in HDMinstilled mice of early life period. Inhibition of IL-23 reduced the increase in allergic responses. Inhibition of IL-23 reduced the increase in allergic responses. After the resting period, HDM administration showed re-increase in allergic responses. Inhibition of IL-23 reduced the HDM-recall allergic responses.
Collapse
Affiliation(s)
- Heung-Woo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Seung Lee
- Institute of Allergy and Clinical Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
15
|
Cao J, Hou S, Chen Z, Yan J, Chao L, Qian Y, Li J, Yan X. Interleukin-37 relieves PM2.5-triggered lung injury by inhibiting autophagy through the AKT/mTOR signaling pathway in vivo and in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115816. [PMID: 38091678 DOI: 10.1016/j.ecoenv.2023.115816] [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: 08/27/2023] [Revised: 11/27/2023] [Accepted: 12/09/2023] [Indexed: 01/12/2024]
Abstract
Autophagy mediates PM2.5-related lung injury (LI) and is tightly linked to inflammation and apoptosis processes. IL-37 has been demonstrated to regulate autophagy. This research aimed to examine the involvement of IL-37 in the progression of PM2.5-related LI and assess whether autophagy serves as a mediator for its effects.To create a model of PM2.5-related LI, this research employed a nose-only PM2.5 exposure system and utilized both human IL-37 transgenic mice and wild-type mice. The hIL-37tg mice demonstrated remarkable reductions in pulmonary inflammation and pathological LI compared to the WT mice. Additionally, they exhibited activation of the AKT/mTOR signaling pathway, which served to regulate the levels of autophagy and apoptosis.Furthermore, in vitro experiments revealed a dose-dependent upregulation of autophagy and apoptotic proteins following exposure to PM2.5 DMSO extraction. Simultaneously, p-AKT and p-mTOR expression was found to decrease. However, pretreatment with IL-37 demonstrated a remarkable reduction in the levels of autophagy and apoptotic proteins, along with an elevation of p-AKT and p-mTOR. Interestingly, pretreatment with rapamycin, an autophagy inducer, weakened the therapeutic impact of IL-37. Conversely, the therapeutic impact of IL-37 was enhanced when treated with 3-MA, a potent autophagy inhibitor. Moreover, the inhibitory effect of IL-37 on autophagy was successfully reversed by administering AKT inhibitor MK2206. The findings suggest that IL-37 can inhibit both the inflammatory response and autophagy, leading to the alleviation of PM2.5-related LI. At the molecular level, IL-37 may exert its anti autophagy and anti apoptosis effects by activating the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jing Cao
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Shujie Hou
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Zixiao Chen
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine,The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Lingshan Chao
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Yuxing Qian
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Jingwen Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China
| | - Xixin Yan
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Respiratory Critical Care Medicine, Hebei Institute of Respiratory Diseases, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
16
|
Le Gall-Lanotto C, Verdin A, Cazier F, Bataille-Savattier A, Guéré C, Dorr MM, Fluhr JW, Courcot D, Vié K, Misery L. Road-traffic-related air pollution contributes to skin barrier alteration and growth defect of sensory neurons. Exp Dermatol 2024; 33:e15009. [PMID: 38284185 DOI: 10.1111/exd.15009] [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: 06/14/2023] [Revised: 12/12/2023] [Accepted: 12/23/2023] [Indexed: 01/30/2024]
Abstract
The effects of air pollution on health are gaining increasing research interest with limited data on skin alterations available. It was suggested that air pollution is a trigger factor for sensitive skin (SS). However, this data was based on surveys with a lack of experimental data. SS is related to altered skin nerve endings and cutaneous neurogenic inflammation. TTe present study was to assess the in vitro effect of particulate matter (PM) on epidermis and nerve ending homeostasis. PM samples were collected according to a validated protocol. Reconstructed human epidermis (RHE, Episkin®) was exposed to PM and subsequently the supernatants were transferred to a culture of PC12 cells differentiated into sensory neurons (SN). Cell viability, axonal growth and neuropeptide-release were measured. The modulation of the expression of different inflammatory, keratinocytes differentiation and neurites growth markers was assessed. PM samples contained a high proportion of particles with a size below 1 μm and a complex chemical composition. Transcriptomic and immunohistochemical analyses revealed that PM altered keratinocytes terminal differentiation and induced an inflammatory response. While viability and functionality of the SN were not modified, their outgrowth was significantly decreased after incubation with PM-exposed Episkin® supernatants. This was closely related to the modification of nerve growth factor/semaphorin 3A balance. This study showed that air pollutants have negative effects on keratinocytes and sensory nerve endings including inflammatory responses. These effects are probably involved in the SS pathophysiology and might be involved in inflammatory skin disorders.
Collapse
Affiliation(s)
| | - Anthony Verdin
- EA4492-Unit of Environmental Chemistry And Interactions With Living Organisms (UCEIV), SFR Condorcet FR CNRS 3417, Université du Littoral Côte d'Opale, Dunkerque, France
| | - Fabrice Cazier
- Common Center of Measurements (CCM), Université du Littoral Côte d'Opale, Dunkerque, France
| | | | | | | | - Joachim W Fluhr
- Univ Brest, LIEN, Brest, France
- Department of Dermatology, University Hospital, Brest, France
- Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin, Germany
- Institute of Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany
| | - Dominique Courcot
- EA4492-Unit of Environmental Chemistry And Interactions With Living Organisms (UCEIV), SFR Condorcet FR CNRS 3417, Université du Littoral Côte d'Opale, Dunkerque, France
| | | | - Laurent Misery
- Univ Brest, LIEN, Brest, France
- Department of Dermatology, University Hospital, Brest, France
| |
Collapse
|
17
|
Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases. Eur J Immunol 2024; 54:e2249982. [PMID: 37804068 DOI: 10.1002/eji.202249982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Epithelial/immune interactions are characterized by the different properties of the various epithelial tissues, the mediators involved, and the varying immune cells that initiate, sustain, or abrogate allergic diseases on the surface. The intestinal mucosa, respiratory mucosa, and regular skin feature structural differences according to their primary function and surroundings. In the context of these specialized functions, the active role of the epithelium in shaping immune responses is increasingly recognizable. Crosstalk between epithelial and immune cells plays an important role in maintaining homeostatic conditions. While cells of the myeloid cell lineage, mainly macrophages, are the dominating immune cell population in the skin and the respiratory tract, lymphocytes comprise most intraepithelial immune cells in the intestine under healthy conditions. Common to all surface epithelia is the fact that innate immune cells represent the first line of immunosurveillance that either directly defeats invading pathogens or initiates and coordinates more effective successive immune responses involving adaptive immune cells and effector cells. Pharmacological approaches for the treatment of allergic and chronic inflammatory diseases involving epithelial barriers target immunological mediators downstream of the epithelium (such as IL-4, IL-5, IL-13, and IgE). The next generation of therapeutics involves upstream events of the inflammatory cascade, such as epithelial-derived alarmins and related mediators.
Collapse
Affiliation(s)
- Melanie Albrecht
- Molecular Allergology, Vice President´s Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University of Marburg, Marburg, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
18
|
Weng CM, Lee MJ, Chao W, Lin YR, Chou CJ, Chen MC, Chou CL, Tsai IL, Lin CH, Fan Chung K, Kuo HP. Airway epithelium IgE-FcεRI cross-link induces epithelial barrier disruption in severe T2-high asthma. Mucosal Immunol 2023; 16:685-698. [PMID: 37536562 DOI: 10.1016/j.mucimm.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Although high-affinity immunoglobulin (Ig)E receptor (FcεRI) expression is upregulated in type 2 (T2)-high asthmatic airway epithelium, its functional role in airway epithelial dysfunction has not been elucidated. Here we report the upregulated expression of FcεRI and p-EGFR (Epidermal Growth Factor Receptor), associated with decreased expression of E-cadherin and claudin-18 in bronchial biopsies of severe T2-high asthmatics compared to mild allergic asthmatics and non-T2 asthmatics. Monomeric IgE (mIgE) decreased the expression of junction proteins, E-cadherin, claudin-18, and ZO-1, and increased alarmin messenger RNA and protein expression in cultured primary bronchial epithelial cells from T2-high asthmatics. Epithelial FcεRI ligation with mIgE decreased transepithelial electric resistance in air-liquid interface cultured epithelial cells. FcεRI ligation with mIgE or IgE- Dinitrophenyl or serum of high-level allergen-specific IgE activated EGFR and Akt via activation of Src family kinases, mediating alarmin expression, junctional protein loss, and increased epithelial permeability. Furthermore, tracheal instillation of mIgE in house dust mite-sensitized mice induced airway hyper-responsiveness, junction protein loss, epithelial cell shedding, and increased epithelial permeability. Thus, our results suggest that IgE-FcεRI cross-linking in the airway epithelium is a potential and unnoticed mechanism for impaired barrier function, increased mucosal permeability, and EGFR-mediated alarmin production in T2-high asthma.
Collapse
Affiliation(s)
- Chih-Ming Weng
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Jung Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei Chao
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Rong Lin
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Chun-Ju Chou
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Mei-Chuan Chen
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan; Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Liang Chou
- Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Lin Tsai
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Han-Pin Kuo
- Pulmonary Medicine Research Center, Taipei Medical University, Taipei, Taiwan; Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 0] [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
|
20
|
Chatziparasidis G, Bush A, Chatziparasidi MR, Kantar A. Airway epithelial development and function: A key player in asthma pathogenesis? Paediatr Respir Rev 2023; 47:51-61. [PMID: 37330410 DOI: 10.1016/j.prrv.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 06/19/2023]
Abstract
Though asthma is a common and relatively easy to diagnose disease, attempts at primary or secondary prevention, and cure, have been disappointing. The widespread use of inhaled steroids has dramatically improved asthma control but has offered nothing in terms of altering long-term outcomes or reversing airway remodeling and impairment in lung function. The inability to cure asthma is unsurprising given our limited understanding of the factors that contribute to disease initiation and persistence. New data have focused on the airway epithelium as a potentially key factor orchestrating the different stages of asthma. In this review we summarize for the clinician the current evidence on the central role of the airway epithelium in asthma pathogenesis and the factors that may alter epithelial integrity and functionality.
Collapse
Affiliation(s)
- Grigorios Chatziparasidis
- Paediatric Respiratory Unit, IASO Hospital, Larissa, Thessaly, Greece; Faculty of Nursing, Thessaly University, Greece.
| | - Andrew Bush
- National Heart and Lung Institute, Royal Brompton & Harefield NHS Foundation Trust, London, UK
| | | | - Ahmad Kantar
- Pediatric Asthma and Cough Centre, Instituti Ospedalieri Bergamaschi, University and Research Hospitals, Bergamo, Italy
| |
Collapse
|
21
|
Kumar P, Singh AB, Arora T, Singh S, Singh R. Critical review on emerging health effects associated with the indoor air quality and its sustainable management. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162163. [PMID: 36781134 DOI: 10.1016/j.scitotenv.2023.162163] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Indoor air quality (IAQ) is one of the fundamental elements affecting people's health and well-being. Currently, there is a lack of awareness among people about the quantification, identification, and possible health effects of IAQ. Airborne pollutants such as volatile organic compounds (VOCs), particulate matter (PM), sulfur dioxide (SO2), carbon monoxide (CO), nitrous oxide (NO), polycyclic aromatic hydrocarbons (PAHs) microbial spores, pollen, allergens, etc. primarily contribute to IAQ deterioration. This review discusses the sources of major indoor air pollutants, molecular toxicity mechanisms, and their effects on cardiovascular, ocular, neurological, women, and foetal health. Additionally, contemporary strategies and sustainable methods for regulating and reducing pollutant concentrations are emphasized, and current initiatives to address and enhance IAQ are explored, along with their unique advantages and potentials. Due to their longer exposure times and particular physical characteristics, women and children are more at risk for poor indoor air quality. By triggering many toxicity mechanisms, including oxidative stress, DNA methylation, epigenetic modifications, and gene activation, indoor air pollution can cause a range of health issues. Low birth weight, acute lower respiratory tract infections, Sick building syndromes (SBS), and early death are more prevalent in exposed residents. On the other hand, the main causes of incapacity and early mortality are lung cancer, chronic obstructive pulmonary disease, and cardiovascular disorders. It's crucial to acknowledge anticipated research needs and implemented efficient interventions and policies to lower health hazards.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of Environmental Studies, Satyawati College, University of Delhi, Delhi 52, India
| | - A B Singh
- Institute of Genomics and Integrative Biology (IGIB), Mall Road Campus, Delhi 07, India
| | - Taruna Arora
- Division of Reproductive Biology, Maternal and Child Health, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Sevaram Singh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India; Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| | - Rajeev Singh
- Department of Environmental Studies, Satyawati College, University of Delhi, Delhi 52, India; Department of Environmental Science, Jamia Millia Islamia (A Central University), New Delhi 110025, India.
| |
Collapse
|
22
|
De Volder J, Bontinck A, De Grove K, Dirven I, Haelterman V, Joos G, Brusselle G, Maes T. Trajectory of neutrophilic responses in a mouse model of pollutant-aggravated allergic asthma. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121722. [PMID: 37105460 DOI: 10.1016/j.envpol.2023.121722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
Experimental studies suggest that neutrophils could contribute to allergic asthma pathogenesis, that is mainly driven by type 2 immunity. Inhalation of diesel exhaust particles (DEP) is implicated in both exacerbation and development of asthma. Since exposure to DEP is associated with a neutrophilic component, we aimed to investigate how exposure to the combination of allergens and DEP modulates neutrophilic responses. Human bronchial epithelial cells (HBEC) were exposed to house dust mite (HDM), DEP or HDM + DEP in vitro to determine the expression of neutrophil-recruiting chemokines. Female (C57BL/6 J) mice were intranasally instilled with saline, DEP, HDM or combined HDM + DEP for 3 weeks (subacute) or 6 weeks (chronic). The neutrophilic responses were determined in lung tissue and bronchoalveolar lavage fluid (BALF). Simultaneous exposure to HDM + DEP resulted in increased CXCL1 and CXCL8 mRNA expression by HBEC in vitro. In mice, subacute exposure to HDM + DEP induced a strong mixed eosinophilic/neutrophilic inflammation in BALF and lung and was associated with higher expression of neutrophil-attracting chemokines and NET formation compared to the sole exposures. After chronic HDM + DEP exposure, a similar neutrophilic response was observed, however the NET formation was less pronounced. Interestingly, the increase of BALF eosinophils was also significantly attenuated after chronic HDM + DEP exposure compared to the subacute exposure. Subacute and chronic HDM + DEP exposure induced goblet cell hyperplasia and airway hyperresponsiveness. Our data suggest a role for neutrophils and NETs in pollutant-aggravated eosinophilic allergic asthma. Moreover, subacute exposure to HDM + DEP induces a mixed eosinophilic/neutrophilic response whereas upon chronic HDM + DEP exposure there is a shift in inflammatory response with a more prominent neutrophilic component.
Collapse
Affiliation(s)
- Joyceline De Volder
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Annelies Bontinck
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Katrien De Grove
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Iris Dirven
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Valerie Haelterman
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Guy Joos
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Guy Brusselle
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Tania Maes
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium.
| |
Collapse
|
23
|
Zhu Y, Pan Z, Jing D, Liang H, Cheng J, Li D, Zhou X, Lin F, Liu H, Pan P, Zhang Y. Association of air pollution, genetic risk, and lifestyle with incident adult-onset asthma: A prospective cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114922. [PMID: 37080133 DOI: 10.1016/j.ecoenv.2023.114922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Numerous studies have explored the association of air pollution with asthma but have yielded conflicting results. The exact role of air pollution in the incidence of adult-onset asthma and whether this effect is modified by genetic risk, lifestyle, or their interaction remain uncertain. METHODS We conducted a prospective cohort study on 298,738 participants (aged 37-73 years) registered in the UK Biobank. Cox proportional hazard models were used to evaluate the association of air pollution, including particulate matter (PM2.5, PMcoarse, and PM10), nitrogen dioxide (NO2), and nitrogen oxides (NOx), with asthma incidence. We constructed genetic risk and lifestyle scores, assessed whether the impact of air pollution on adult-onset asthma risk was modified by genetic susceptibility or lifestyle factors, and evaluated the identified interactions. RESULTS We found that each interquartile range increase in annual concentrations of PM2.5, NO2, and NOx was related to 1.04 (95% confidence interval [CI]: 1.01, 1.08), 1.04 (95% CI: 1.00, 1.08), and 1.03 (95% CI: 1.00, 1.06) times the risk of adult-onset asthma, respectively. The size of the effect of air pollution was greater among subpopulations with low genetic risk or unfavorable lifestyles. We also identified an additive interaction effect of air pollution with lifestyle factors, but not with genetic risk, on the risk of adult-onset asthma. CONCLUSION Our analyses show that air pollution increases the risk of adult-onset asthma, but that the size of the effect is modified by lifestyle and genetic risk. These findings emphasize the need for integrated interventions for environmental pollution by the government as well as adherence to healthy lifestyles to prevent adult-onset asthma.
Collapse
Affiliation(s)
- Yiqun Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China
| | - Zhaoyi Pan
- Central South University, Changsha 410008, Hunan, China
| | - Danrong Jing
- Department of Dermatology, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China
| | - Huaying Liang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China
| | - Jun Cheng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dianwu Li
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China
| | - Xin Zhou
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China
| | - Fengyu Lin
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008, Hunan, China.
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008, Hunan, China.
| | - Yan Zhang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Center of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008, Hunan, China.
| |
Collapse
|
24
|
Labohá P, Sychrová E, Brózman O, Sovadinová I, Bláhová L, Prokeš R, Ondráček J, Babica P. Cyanobacteria, cyanotoxins and lipopolysaccharides in aerosols from inland freshwater bodies and their effects on human bronchial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104073. [PMID: 36738853 DOI: 10.1016/j.etap.2023.104073] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Components of cyanobacterial water blooms were quantified in aerosols above agitated water surfaces of five freshwater bodies. The thoracic and respirable aerosol fraction (0.1-10 µm) was sampled using a high-volume sampler. Cyanotoxins microcystins were detected by LC-MS/MS at levels 0.3-13.5 ng/mL (water) and < 35-415 fg/m3 (aerosol). Lipopolysaccharides (endotoxins) were quantified by Pyrogene rFC assay at levels < 10-119 EU/mL (water) and 0.13-0.64 EU/m3 (aerosol). Cyanobacterial DNA was detected by qPCR at concentrations corresponding to 104-105 cells eq./mL (water) and 101-103 cells eq./m3 (aerosol). Lipopolysaccharides isolated from bloom samples induced IL-6 and IL-8 cytokine release in human bronchial epithelial cells Beas-2B, while extracted cyanobacterial metabolites induced both pro-inflammatory and cytotoxic effects. Bloom components detected in aerosols and their bioactivities observed in upper respiratory airway epithelial cells together indicate that aerosols formed during cyanobacterial water blooms could induce respiratory irritation and inflammatory injuries, and thus present an inhalation health risk.
Collapse
Affiliation(s)
- Petra Labohá
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic
| | - Eliška Sychrová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic
| | - Ondřej Brózman
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic
| | - Iva Sovadinová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic
| | - Lucie Bláhová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic
| | - Roman Prokeš
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic; Department of Atmospheric Matter Fluxes and Long-range Transport, Global Change Research Institute of the Czech Academy of Sciences, Bělidla 986/4a, 60300 Brno, Czech Republic
| | - Jakub Ondráček
- Department of Aerosol Chemistry and Physics, Institute of Chemical Process Fundamentals of the Czech Academy of Sciences, Rozvojová 135, 16502 Prague, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, 61137 Brno, Czech Republic; Department of Experimental Phycology and Ecotoxicology, Institute of Botany of the Czech Academy of Sciences, Lidická 25/27, 60200 Brno, Czech Republic.
| |
Collapse
|
25
|
León B. Understanding the development of Th2 cell-driven allergic airway disease in early life. FRONTIERS IN ALLERGY 2023; 3:1080153. [PMID: 36704753 PMCID: PMC9872036 DOI: 10.3389/falgy.2022.1080153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Allergic diseases, including atopic dermatitis, allergic rhinitis, asthma, and food allergy, are caused by abnormal responses to relatively harmless foreign proteins called allergens found in pollen, fungal spores, house dust mites (HDM), animal dander, or certain foods. In particular, the activation of allergen-specific helper T cells towards a type 2 (Th2) phenotype during the first encounters with the allergen, also known as the sensitization phase, is the leading cause of the subsequent development of allergic disease. Infants and children are especially prone to developing Th2 cell responses after initial contact with allergens. But in addition, the rates of allergic sensitization and the development of allergic diseases among children are increasing in the industrialized world and have been associated with living in urban settings. Particularly for respiratory allergies, greater susceptibility to developing allergic Th2 cell responses has been shown in children living in urban environments containing low levels of microbial contaminants, principally bacterial endotoxins [lipopolysaccharide (LPS)], in the causative aeroallergens. This review highlights the current understanding of the factors that balance Th2 cell immunity to environmental allergens, with a particular focus on the determinants that program conventional dendritic cells (cDCs) toward or away from a Th2 stimulatory function. In this context, it discusses transcription factor-guided functional specialization of type-2 cDCs (cDC2s) and how the integration of signals derived from the environment drives this process. In addition, it analyzes observational and mechanistic studies supporting an essential role for innate sensing of microbial-derived products contained in aeroallergens in modulating allergic Th2 cell immune responses. Finally, this review examines whether hyporesponsiveness to microbial stimulation, particularly to LPS, is a risk factor for the induction of Th2 cell responses and allergic sensitization during infancy and early childhood and the potential factors that may affect early-age response to LPS and other environmental microbial components.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Discerning asthma endotypes through comorbidity mapping. Nat Commun 2022; 13:6712. [PMID: 36344522 PMCID: PMC9640644 DOI: 10.1038/s41467-022-33628-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Asthma is a heterogeneous, complex syndrome, and identifying asthma endotypes has been challenging. We hypothesize that distinct endotypes of asthma arise in disparate genetic variation and life-time environmental exposure backgrounds, and that disease comorbidity patterns serve as a surrogate for such genetic and exposure variations. Here, we computationally discover 22 distinct comorbid disease patterns among individuals with asthma (asthma comorbidity subgroups) using diagnosis records for >151 M US residents, and re-identify 11 of the 22 subgroups in the much smaller UK Biobank. GWASs to discern asthma risk loci for individuals within each subgroup and in all subgroups combined reveal 109 independent risk loci, of which 52 are replicated in multi-ancestry meta-analysis across different ethnicity subsamples in UK Biobank, US BioVU, and BioBank Japan. Fourteen loci confer asthma risk in multiple subgroups and in all subgroups combined. Importantly, another six loci confer asthma risk in only one subgroup. The strength of association between asthma and each of 44 health-related phenotypes also varies dramatically across subgroups. This work reveals subpopulations of asthma patients distinguished by comorbidity patterns, asthma risk loci, gene expression, and health-related phenotypes, and so reveals different asthma endotypes.
Collapse
|
27
|
Würzner P, Jörres RA, Karrasch S, Quartucci C, Böse-O'Reilly S, Nowak D, Rakete S. Effect of experimental exposures to 3-D printer emissions on nasal allergen responses and lung diffusing capacity for inhaled carbon monoxide/nitric oxide in subjects with seasonal allergic rhinitis. INDOOR AIR 2022; 32:e13174. [PMID: 36437663 DOI: 10.1111/ina.13174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
3-D printers are widely used. Based on previous findings, we hypothesized that their emissions could enhance allergen responsiveness and reduce lung diffusing capacity. Using a cross-over design, 28 young subjects with seasonal allergic rhinitis were exposed to 3-D printer emissions, either from polylactic acid (PLA) or from acrylonitrile butadiene styrene copolymer (ABS), for 2 h each. Ninety minutes later, nasal allergen challenges were performed, with secretions sampled after 1.5 h. Besides nasal functional and inflammatory responses, assessments included diffusing capacity. There was also an inclusion day without exposure. The exposures elicited slight reductions in lung diffusing capacity for inhaled nitric oxide (DLNO ) that were similar for PLA and ABS. Rhinomanometry showed the same allergen responses after both exposures. In nasal secretions, concentrations of interleukin 6 and tumor necrosis factor were slightly reduced after ABS exposure versus inclusion day, while that of interleukin 5 was slightly increased after PLA exposure versus inclusion.
Collapse
Affiliation(s)
- Philipp Würzner
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Rudolf A Jörres
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stefan Karrasch
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Caroline Quartucci
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Bavarian Health and Food Safety Authority, Institute for Occupational Health and Product Safety, Environmental Health, Munich, Germany
| | - Stephan Böse-O'Reilly
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Department of Public Health, Health Services Research and Health Technology Assessment, Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT - University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Dennis Nowak
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stefan Rakete
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
28
|
Duchesne M, Okoye I, Lacy P. Epithelial cell alarmin cytokines: Frontline mediators of the asthma inflammatory response. Front Immunol 2022; 13:975914. [PMID: 36311787 PMCID: PMC9616080 DOI: 10.3389/fimmu.2022.975914] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
The exposure of the airway epithelium to external stimuli such as allergens, microbes, and air pollution triggers the release of the alarmin cytokines IL-25, IL-33 and thymic stromal lymphopoietin (TSLP). IL-25, IL-33 and TSLP interact with their ligands, IL-17RA, IL1RL1 and TSLPR respectively, expressed by hematopoietic and non-hematopoietic cells including dendritic cells, ILC2 cells, endothelial cells, and fibroblasts. Alarmins play key roles in driving type 2-high, and to a lesser extent type 2-low responses, in asthma. In addition, studies in which each of these three alarmins were targeted in allergen-challenged mice showed decreased chronicity of type-2 driven disease. Consequently, ascertaining the mechanism of activity of these upstream mediators has implications for understanding the outcome of targeted therapies designed to counteract their activity and alleviate downstream type 2-high and low effector responses. Furthermore, identifying the factors which shift the balance between the elicitation of type 2-high, eosinophilic asthma and type-2 low, neutrophilic-positive/negative asthma by alarmins is essential. In support of these efforts, observations from the NAVIGATOR trial imply that targeting TSLP in patients with tezepelumab results in reduced asthma exacerbations, improved lung function and control of the disease. In this review, we will discuss the mechanisms surrounding the secretion of IL-25, IL-33, and TSLP from the airway epithelium and how this influences the allergic airway cascade. We also review in detail how alarmin-receptor/co-receptor interactions modulate downstream allergic inflammation. Current strategies which target alarmins, their efficacy and inflammatory phenotype will be discussed.
Collapse
|
29
|
Milani GP, Cafora M, Favero C, Luganini A, Carugno M, Lenzi E, Pistocchi A, Pinatel E, Pariota L, Ferrari L, Bollati V. PM 2 .5, PM 10 and bronchiolitis severity: A cohort study. Pediatr Allergy Immunol 2022; 33:e13853. [PMID: 36282132 PMCID: PMC9827836 DOI: 10.1111/pai.13853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND A few studies suggest that particulate matter (PM) exposure might play a role in bronchiolitis. However, available data are mostly focused on the risk of hospitalization and come from retrospective studies that provided conflicting results. This prospective study investigated the association between PM (PM2.5 and PM10 ) exposure and the severity of bronchiolitis. METHODS This prospective cohort study was conducted between November 2019 and February 2020 at the pediatric emergency department of the Fondazione IRCCS Ca' Ospedale Maggiore Policlinico, Milan, Italy. Infants <1 year of age with bronchiolitis were eligible. The bronchiolitis severity score was assessed in each infant and a nasal swab was collected to detect respiratory viruses. The daily PM10 and PM2.5 exposure in the 29 preceding days were considered. Adjusted regression models were employed to evaluate the association between the severity score and PM10 and PM2.5 exposure. RESULTS A positive association between the PM2.5 levels and the severity score was found at day-2 (β 0.0214, 95% CI 0.0011-0.0417, p = .0386), day-5 (β 0.0313, 95% CI 0.0054-0.0572, p = .0179), day-14 (β 0.0284, 95% CI 0.0078-0.0490, p = .0069), day-15 (β 0.0496, 95% CI 0.0242-0.0750, p = .0001) and day-16 (β 0.0327, 95% CI 0.0080-0.0574, p = .0093).Similar figures were observed considering the PM10 exposure and limiting the analyses to infants with respiratory syncytial virus. CONCLUSION This study shows for the first time a direct association between PM2.5 and PM10 levels and the severity of bronchiolitis.
Collapse
Affiliation(s)
- Gregorio P Milani
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Cafora
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Chiara Favero
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Anna Luganini
- Department of Life Science and System Biology, Università degli Studi di Torino, Turin, Italy
| | - Michele Carugno
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erica Lenzi
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Eva Pinatel
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Luigi Pariota
- Department of Civil, Architectural and Environmental Engineering, Federico II University of Naples, Naples, Italy
| | - Luca Ferrari
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Bollati
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
30
|
Sundram TKM, Tan ESS, Cheah SC, Lim HS, Seghayat MS, Bustami NA, Tan CK. Impacts of particulate matter (PM 2.5) on the health status of outdoor workers: observational evidence from Malaysia. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:71064-71074. [PMID: 35595900 DOI: 10.1007/s11356-022-20955-y] [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: 12/31/2021] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Ambient air pollution is a significant contributor to disease burden, leading to an estimated 4.2 million premature deaths and 103.1 million disability-adjusted life years (DALYs) annually worldwide. As industrialization and urbanization surge in Asia, air pollution and its corresponding health issues follow suit. Findings on disease burden in developing countries are extremely scanty. This study aimed to determine the concentration of PM2.5 and its impact on respiratory health of outdoor workers in Malaysia. A 2-cycled 3-month cohort study involving 440 participants was conducted. Workers' health status was assessed via (1) Total Ocular Symptom Score (TOSS), (2) Total Nasal Symptom Score (TNSS), (3) St. George's Respiratory Questionnaire (SGPQ), and (4) Asthma Control Test (ACT). The maximum PM2.5 concentration was measured at 122.90 ± 2.07 µg/m3 during third week of August 2016. Meanwhile, the minimum concentration was measured at 57.47 ± 3.80 µg/m3 and 57.47 ± 1.64 µg/m3 during fourth week of July 2016 and first week of August 2017 respectively. Findings revealed that TOSS, TNSS, and SGPQ changes were significantly (p < 0.05) associated with the concentration of PM2.5. Outdoor workers were more significantly (p < 0.05) affected by changes in PM2.5 compared to indoor workers with a moderate correlation (r value ranged from 0.4 to 0.7). Ironically, no significant association was found between ACT assessment and PM2.5. Collectively, our findings suggested that changes in the concentration of PM2.5 threatened the respiratory health of outdoor workers. The existing policy should be strengthened and preventive measures to be enforced safeguarding health status of outdoor workers.
Collapse
Affiliation(s)
| | - Eugenie Sin Sing Tan
- Faculty of Medicine and Health Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Shiau Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Hwee San Lim
- School of Physics, Universiti Sains Malaysia, 11800, Pulau Pinang, Gelugor, Malaysia
| | - Marjan Sadat Seghayat
- Faculty of Medicine, MAHSA University, Bioscience & Nursing, 42610, Jenjarom, Selangor, Malaysia
| | - Normina Ahmad Bustami
- Faculty of Medicine and Health Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Chung Keat Tan
- Faculty of Medicine and Health Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
31
|
Pardo M, Offer S, Hartner E, Di Bucchianico S, Bisig C, Bauer S, Pantzke J, Zimmermann EJ, Cao X, Binder S, Kuhn E, Huber A, Jeong S, Käfer U, Schneider E, Mesceriakovas A, Bendl J, Brejcha R, Buchholz A, Gat D, Hohaus T, Rastak N, Karg E, Jakobi G, Kalberer M, Kanashova T, Hu Y, Ogris C, Marsico A, Theis F, Shalit T, Gröger T, Rüger CP, Oeder S, Orasche J, Paul A, Ziehm T, Zhang ZH, Adam T, Sippula O, Sklorz M, Schnelle-Kreis J, Czech H, Kiendler-Scharr A, Zimmermann R, Rudich Y. Exposure to naphthalene and β-pinene-derived secondary organic aerosol induced divergent changes in transcript levels of BEAS-2B cells. ENVIRONMENT INTERNATIONAL 2022; 166:107366. [PMID: 35763991 DOI: 10.1016/j.envint.2022.107366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
The health effects of exposure to secondary organic aerosols (SOAs) are still limited. Here, we investigated and compared the toxicities of soot particles (SP) coated with β-pinene SOA (SOAβPin-SP) and SP coated with naphthalene SOA (SOANap-SP) in a human bronchial epithelial cell line (BEAS-2B) residing at the air-liquid interface. SOAβPin-SP mostly contained oxygenated aliphatic compounds from β-pinene photooxidation, whereas SOANap-SP contained a significant fraction of oxygenated aromatic products under similar conditions. Following exposure, genome-wide transcriptome responses showed an Nrf2 oxidative stress response, particularly for SOANap-SP. Other signaling pathways, such as redox signaling, inflammatory signaling, and the involvement of matrix metalloproteinase, were identified to have a stronger impact following exposure to SOANap-SP. SOANap-SP also induced a stronger genotoxicity response than that of SOAβPin-SP. This study elucidated the mechanisms that govern SOA toxicity and showed that, compared to SOAs derived from a typical biogenic precursor, SOAs from a typical anthropogenic precursor have higher toxicological potency, which was accompanied with the activation of varied cellular mechanisms, such as aryl hydrocarbon receptor. This can be attributed to the difference in chemical composition; specifically, the aromatic compounds in the naphthalene-derived SOA had higher cytotoxic potential than that of the β-pinene-derived SOA.
Collapse
Affiliation(s)
- Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, ISR-7610001 Rehovot, Israel.
| | - Svenja Offer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Elena Hartner
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Christoph Bisig
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Stefanie Bauer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Jana Pantzke
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Elias J Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Xin Cao
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Stephanie Binder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Evelyn Kuhn
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Anja Huber
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Seongho Jeong
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Uwe Käfer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Eric Schneider
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Arunas Mesceriakovas
- Department of Environmental and Biological Sciences, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, FI-70210 Kuopio, Finland
| | - Jan Bendl
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; University of the Bundeswehr Munich, Institute for Chemistry and Environmental Engineering, Werner- Heisenberg-Weg 39, D-85577 Neubiberg, Germany; Institute for Environmental Studies, Faculty of Science, Charles University, Albertov 6, CZE-12800 Prague, Czech Republic
| | - Ramona Brejcha
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Angela Buchholz
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, FI-70210 Kuopio, Finland
| | - Daniela Gat
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, ISR-7610001 Rehovot, Israel
| | - Thorsten Hohaus
- Institute of Energy and Climate Research, Troposphere (IEK-8), Forschungszentrum Jülich GmbH, Wilhelm-Johen-Str., D-52428 Jülich, Germany
| | - Narges Rastak
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Erwin Karg
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Gert Jakobi
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Markus Kalberer
- Department of Environmental Sciences, University of Basel, Klingelbergstr. 27, CH-4056 Basel, Switzerland
| | - Tamara Kanashova
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany
| | - Yue Hu
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Christoph Ogris
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Annalisa Marsico
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Fabian Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Tali Shalit
- The Mantoux Bioinformatics Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Thomas Gröger
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Christopher P Rüger
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Jürgen Orasche
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Andreas Paul
- Institute of Energy and Climate Research, Troposphere (IEK-8), Forschungszentrum Jülich GmbH, Wilhelm-Johen-Str., D-52428 Jülich, Germany
| | - Till Ziehm
- Institute of Energy and Climate Research, Troposphere (IEK-8), Forschungszentrum Jülich GmbH, Wilhelm-Johen-Str., D-52428 Jülich, Germany
| | - Zhi-Hui Zhang
- Department of Environmental Sciences, University of Basel, Klingelbergstr. 27, CH-4056 Basel, Switzerland
| | - Thomas Adam
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; University of the Bundeswehr Munich, Institute for Chemistry and Environmental Engineering, Werner- Heisenberg-Weg 39, D-85577 Neubiberg, Germany
| | - Olli Sippula
- Department of Environmental and Biological Sciences, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, FI-70210 Kuopio, Finland
| | - Martin Sklorz
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Jürgen Schnelle-Kreis
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Hendryk Czech
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Astrid Kiendler-Scharr
- Institute of Energy and Climate Research, Troposphere (IEK-8), Forschungszentrum Jülich GmbH, Wilhelm-Johen-Str., D-52428 Jülich, Germany
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany; Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Dr.-Lorenz-Weg 2, D-18059 Rostock, Germany
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, ISR-7610001 Rehovot, Israel
| |
Collapse
|
32
|
Wildung M, Herr C, Riedel D, Wiedwald C, Moiseenko A, Ramírez F, Tasena H, Heimerl M, Alevra M, Movsisyan N, Schuldt M, Volceanov-Hahn L, Provoost S, Nöthe-Menchen T, Urrego D, Freytag B, Wallmeier J, Beisswenger C, Bals R, van den Berge M, Timens W, Hiemstra PS, Brandsma CA, Maes T, Andreas S, Heijink IH, Pardo LA, Lizé M. miR449 Protects Airway Regeneration by Controlling AURKA/HDAC6-Mediated Ciliary Disassembly. Int J Mol Sci 2022; 23:ijms23147749. [PMID: 35887096 PMCID: PMC9320302 DOI: 10.3390/ijms23147749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 01/25/2023] Open
Abstract
Airway mucociliary regeneration and function are key players for airway defense and are impaired in chronic obstructive pulmonary disease (COPD). Using transcriptome analysis in COPD-derived bronchial biopsies, we observed a positive correlation between cilia-related genes and microRNA-449 (miR449). In vitro, miR449 was strongly increased during airway epithelial mucociliary differentiation. In vivo, miR449 was upregulated during recovery from chemical or infective insults. miR0449−/− mice (both alleles are deleted) showed impaired ciliated epithelial regeneration after naphthalene and Haemophilus influenzae exposure, accompanied by more intense inflammation and emphysematous manifestations of COPD. The latter occurred spontaneously in aged miR449−/− mice. We identified Aurora kinase A and its effector target HDAC6 as key mediators in miR449-regulated ciliary homeostasis and epithelial regeneration. Aurora kinase A is downregulated upon miR449 overexpression in vitro and upregulated in miR449−/− mouse lungs. Accordingly, imaging studies showed profoundly altered cilia length and morphology accompanied by reduced mucociliary clearance. Pharmacological inhibition of HDAC6 rescued cilia length and coverage in miR449−/− cells, consistent with its tubulin-deacetylating function. Altogether, our study establishes a link between miR449, ciliary dysfunction, and COPD pathogenesis.
Collapse
Affiliation(s)
- Merit Wildung
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
| | - Christian Herr
- Department of Internal Medicine V, Saarland University, 66421 Homburg, Germany; (C.H.); (C.B.); (R.B.)
| | - Dietmar Riedel
- Laboratory for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, 37075 Goettingen, Germany;
| | - Cornelia Wiedwald
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
| | - Alena Moiseenko
- Immunology & Respiratory Department, Boehringer Ingelheim Pharma GmbH, 88400 Biberach an der Riss, Germany;
| | - Fidel Ramírez
- Global Computational Biology and Digital Sciences Department, Boehringer Ingelheim Pharma GmbH, 88400 Biberach an der Riss, Germany;
| | - Hataitip Tasena
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands; (H.T.); (W.T.); (C.-A.B.); (I.H.H.)
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands;
| | - Maren Heimerl
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
| | - Mihai Alevra
- Institute of Neuro- and Sensory Physiology, Goettingen University, 37073 Goettingen, Germany;
| | - Naira Movsisyan
- Oncophysiology Group, Max Planck Institute for Multidisciplinary Sciences, 37075 Goettingen, Germany; (N.M.); (D.U.); (L.A.P.)
| | - Maike Schuldt
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
| | - Larisa Volceanov-Hahn
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
| | - Sharen Provoost
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.P.); (T.M.)
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (T.N.-M.); (J.W.)
| | - Diana Urrego
- Oncophysiology Group, Max Planck Institute for Multidisciplinary Sciences, 37075 Goettingen, Germany; (N.M.); (D.U.); (L.A.P.)
| | - Bernard Freytag
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
| | - Julia Wallmeier
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany; (T.N.-M.); (J.W.)
| | - Christoph Beisswenger
- Department of Internal Medicine V, Saarland University, 66421 Homburg, Germany; (C.H.); (C.B.); (R.B.)
| | - Robert Bals
- Department of Internal Medicine V, Saarland University, 66421 Homburg, Germany; (C.H.); (C.B.); (R.B.)
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands;
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands; (H.T.); (W.T.); (C.-A.B.); (I.H.H.)
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands;
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Centre, 2333 Leiden, The Netherlands;
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands; (H.T.); (W.T.); (C.-A.B.); (I.H.H.)
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands;
| | - Tania Maes
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (S.P.); (T.M.)
| | - Stefan Andreas
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
| | - Irene H. Heijink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands; (H.T.); (W.T.); (C.-A.B.); (I.H.H.)
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands;
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, 9712 Groningen, The Netherlands
| | - Luis A. Pardo
- Oncophysiology Group, Max Planck Institute for Multidisciplinary Sciences, 37075 Goettingen, Germany; (N.M.); (D.U.); (L.A.P.)
| | - Muriel Lizé
- Molecular & Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, 37075 Gottingen, Germany; (M.W.); (C.W.); (M.H.); (L.V.-H.); (S.A.)
- Molecular Oncology, University Medical Center Goettingen, 37077 Goettingen, Germany; (M.S.); (B.F.)
- Immunology & Respiratory Department, Boehringer Ingelheim Pharma GmbH, 88400 Biberach an der Riss, Germany;
- Correspondence:
| |
Collapse
|
33
|
Yang X, Wang Q, Han F, Dong B, Wen B, Li L, Ruan H, Zhang S, Kong J, Zhi H, Wang C, Wang J, Zhang M, Xu D. Pulmonary Benefits of Intervention with Air Cleaner among Schoolchildren in Beijing: A Randomized Double-Blind Crossover Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7185-7193. [PMID: 34491046 DOI: 10.1021/acs.est.1c03146] [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] [Indexed: 06/13/2023]
Abstract
We conducted a crossover study employing air cleaner intervention among 125 schoolchildren aged 9-12 years in a boarding school in Beijing, China. The PM concentrations were monitored, and 27 biomarkers were analyzed. We used the linear mixed-effects model to evaluate the association of intervention/time-weighted PM concentrations with biomarkers. The outcomes showed that air cleaner intervention was associated with FeNO, exhaled breath condensate (EBC) IL-1β, and IL-6, which decreased by 12.57%, 10.83%, and 4.33%, respectively. Similar results were observed in the associations with PMs. Lag 1 day PMs had the strongest relationship with biomarkers, and significant changes were observed in biomarkers such as FEV1, FeNO, EBC 8-iso, and MCP-1. Boys showed higher percentage changes than girls, and the related biomarkers were FeNO, EBC 4-HNE, IL-1β, IL-6, and MCP-1. The results showed that biomarkers such as FeNO, EBC IL-6, MCP-1, and 4-HNE could sensitively reflect the early abnormal response of the respiratory system under short-term PM exposure among healthy schoolchildren and indicated that (1) air cleaners exert a protective effect on children's respiratory system. (2) PM had lag and cumulative effect, lag 1 day had the greatest effect. (3) The boys were more sensitive than the girls.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Qin Wang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Air Quality and Health Monitoring, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Feng Han
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Bin Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Bo Wen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Li Li
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Hongjie Ruan
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Shaoping Zhang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Jian Kong
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Hong Zhi
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Chong Wang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Jun Wang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Chemistry, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Ming Zhang
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Dongqun Xu
- Key Laboratory of Environment and Human Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| |
Collapse
|
34
|
Particulate Matter Exposure and the Changes in Immune Biomarkers: Effects of Biyeom-Go on the Nasal Mucosa of Patients with Allergic Rhinitis and a Particulate Matter-Treated Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4259669. [PMID: 35378908 PMCID: PMC8976652 DOI: 10.1155/2022/4259669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
Abstract
This study was to investigate the effects of Biyeom-go (BYG, an herbal formula) on immune biomarkers present in the nasal mucosa of patients with allergic rhinitis under exposure to particulate matter 2.5 (PM2.5), and on changes in goblet cells and immune biomarkers in mice under exposure to Korea diesel particulate matter (KDP20). Thirty patients showing characteristic allergic rhinitis symptoms were enrolled in Jeonju-si, Korea, and treated with BYG thrice a day for four weeks. Changes in the expression of immune biomarkers (interleukin 4 (IL-4), IL-5, IL-8, IL-13, IL-33, and thymic stromal lymphopoietin (TSLP) mRNA), total nasal symptom scores (TNSS), mini-rhinitis-specific quality of life questionnaire (RQLQ) results, and visual analog scale scores were evaluated after 4 weeks of treatment. Additionally, the difference in PM2.5 concentrations in the air in Jeonju-si, Korea (November, 2019 ∼ March, 2020), was analyzed to determine the change in TNSS. KDP20 (100 μg/mL) was exposed to C57BL/6 mice for 10 days; 0.05% Nasonex (a positive control, mometasone furoate), or BYG was administrated for 5 days twice a day. The expression of inflammatory factors was detected via qRT-PCR using nasopharynx tissue samples of mice. BYG treatment was found to be associated with significant improvement in total nasal symptoms, especially itching and sneezing (p < 0.0001), and mini-RQLQ after 4 weeks. IL-8 (p < 0.01), IL-33 (p < 0.01), and TSLP (p < 0.001) expression levels decreased after BYG treatment. In mice, administration of BYG reduced the number of goblet cells increased through KDP20 treatment. KDP20-induced immune biomarkers (IL-33, TSLP, tumor necrosis factor alpha, and IL-8) were also significantly downregulated in the nasopharynx tissue after BYG treatment. Therefore, BYG may show therapeutic effects against allergic rhinitis in humans, and it was confirmed that the expression of PM-induced inflammatory factors in mice was decreased via BYG treatment.
Collapse
|
35
|
Cano-Granda DV, Ramírez-Ramírez M, M. Gómez D, Hernandez JC. Effects of particulate matter on endothelial, epithelial and immune system cells. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.01.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Particulate Matter (PM) is an air pollutant that is classified according to its aerodynamic diameter into particles with a diameter of less than 10 µm (PM10), a diameter of less than 2.5 µm (PM2.5), and particles ultra-fine with a diameter less than 0.1 µm (PM0.1). PM10 is housed in the respiratory system, while PM2.5 and 0.1 can pass into the circulation to generate systemic alterations. Although several diseases associated with PM exposure, such as respiratory, cardiovascular, and central nervous system, have been documented to cause 4.2 million premature deaths per year worldwide. Few reviews address cellular and molecular mechanisms in the epithelial and endothelial cells of the tissues exposed to PM, which can cause these diseases, this being the objective of the present review. For this, a search was carried out in the NCBI and Google Scholar databases focused on scientific publications that addressed the expression of pro-inflammatory molecules, adhesion molecules, and oxidative radicals, among others, and their relationship with the effects caused by the PM. The main findings include the increase in pro-inflammatory cytokines and dysfunction in the components of the immune response; the formation of reactive oxygen species; changes in epithelial and endothelial function, evidenced by altered expression of adhesion molecules; and the increase in molecules involved in coagulation. Complementary studies are required to understand the molecular effects of harmful health effects and the future approach to strategies to mitigate this response.
Collapse
Affiliation(s)
- Danna V. Cano-Granda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia 2 Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Mariana Ramírez-Ramírez
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia 2 Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Diana M. Gómez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia. Medellín, Colombia
| |
Collapse
|
36
|
Shi Q, Wang Q, Liu L, Chen J, Wang B, Bellusci S, Chen C, Dong N. FGF10 protects against particulate matter (PM)-induced lung injury via regulation of endoplasmic reticulum stress. Int Immunopharmacol 2022; 105:108552. [PMID: 35114441 DOI: 10.1016/j.intimp.2022.108552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/08/2022] [Accepted: 01/16/2022] [Indexed: 11/05/2022]
Abstract
Exposure of the lungs to particulate matter (PM) leads to the development of respiratory disease and involves mechanisms such as oxydative stress, mitochondrial dysfunction and endoplasmic reticulum (ER) stress. However, there are no effective therapies to treat PM-induced lung diseases. Fibroblast growth factor 10 (FGF10) is a multifunctional growth factor mediating mesenchymal-to-epithelial signaling and displaying a significant therapeutic potential following injury. The present research aims to investigate the regulatory mechanism of FGF10 on ER stress in PM-induced lung injury. PM-induced lung injury leads to peribronchial wall thickening and marked infiltration of inflammatory cells which is associated with increased secretion of inflammatory cytokines. The results show that FGF10 treatment attenuates PM-induced lung injury in vivo and reversed ER stress protein GRP78 and CHOP levels. Moreover, comparison of human bronchial epithelial cells cultured with PM and FGF10 vs PM alone shows sustained cell proliferation and restrained secretion of inflammatory cytokines supporting FGF10's protective role. Significantly, both ERK1/2 and PI3K/AKT inhibitors largely abolished the impact of FGF10 on PM-induced ER stress. Taken together, both in vivo and in vitro experiments showed that FGF10, via the activation of ERK1/2 and PI3K/AKT signaling, protects against PM-induced lung injury through the regulation of ER stress. Therefore, FGF10 represents a potential therapy for PM-induced lung injury.
Collapse
Affiliation(s)
- Qiangqiang Shi
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Qiang Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Li Liu
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Junjie Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Beibei Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Saverio Bellusci
- Cardio-Pulmonary Institute (CPI) and Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, 35392, Giessen, Germany.
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China.
| | - Nian Dong
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China.
| |
Collapse
|
37
|
Bai X, Chen H, Oliver BG. The health effects of traffic-related air pollution: A review focused the health effects of going green. CHEMOSPHERE 2022; 289:133082. [PMID: 34843836 DOI: 10.1016/j.chemosphere.2021.133082] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/03/2021] [Accepted: 11/24/2021] [Indexed: 06/13/2023]
Abstract
Traffic-related air pollution (TRAP) is global concern due to both the ecological damage of TRAP and the adverse health effects in Humans. Several strategies to reduce TRAP have been implemented, including the use of sustainable fuels, after-treatment technologies, and new energy vehicles. Such approaches can reduce the exhaust of particulate matter, adsorbed chemicals and a range of gases, but from a health perspective these approaches are not always successful. This review aims to discuss the approaches taken, and to then describe the likely health effects of these changes.
Collapse
Affiliation(s)
- Xu Bai
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia; Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Sydney, NSW, 2037, Australia.
| |
Collapse
|
38
|
García-Serna AM, Martín-Orozco E, Jiménez-Guerrero P, Hernández-Caselles T, Pérez-Fernández V, Cantero-Cano E, Muñoz-García M, Molina-Ruano MD, Rojo-Atenza E, García-Marcos L, Morales E, Garcia‐Marcos L, Gimenez‐Banon MJ, Martinez‐Torres A, Morales E, Perez‐Fernandez V, Sanchez‐Solis M, Nieto A, Prieto‐Sanchez MT, Sanchez‐Ferrer M, Fernanez‐Palacios L, Gomez‐Gomez VP, Martinez‐Gracia C, Peso‐Echarri P, Ros‐Berruezo G, Santaella‐Pacual M, Gazquez A, Larque E, Pastor‐Fajardo MT, Sanchez‐Campillo M, Serrano‐Munuera A, Zornoza‐Moreno M, Jimenez‐Guerrero P, Adomnei E, Arense‐Gonzalo JJ, Mendiola J, Navarro‐Lafuente F, Torres‐Cantero AM, Salvador‐Garcia C, Segovia‐Hernández M, Yagüe‐Guirao G, Valero‐Guillén PL, Aviles‐Plaza FV, Cabezas‐Herrera J, Martinez‐Lopez A, Martinez‐Villanueva M, Noguera‐Velasco JA, Franco‐Garcia A, Garcia‐Serna AM, Hernandez‐Caselles T, Martin‐Orozco E, Norte‐Muñoz M, Canovas M, Cantero‐Cano E, de Diego T, Pastor JM, Sola‐Martínez RA, Esteban‐Gil A, Fernández‐Breis JT, Alcántara MV, Hernández S, López‐Soler C. Cytokine profiles in cord blood in relation to prenatal traffic-related air pollution: The NELA cohort. Pediatr Allergy Immunol 2022; 33:e13732. [PMID: 35212052 DOI: 10.1111/pai.13732] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Outdoor air pollution may disturb immune system development. We investigated whether gestational exposure to traffic-related air pollutants (TRAP) is associated with unstimulated cytokine profiles in newborns. METHODS Data come from 235 newborns of the NELA cohort. Innate response-related cytokines (IL-6, IFN-α, IL1-β, and TNF-α), Th1-related (IFN-γ and IL-2), Th2-related (IL-4, IL-5, and IL-13), Th17-related (IL-17 and IL-23), and immunomodulatory cytokine IL-10 were quantified in the supernatant of unstimulated whole umbilical cord blood cells after 7 days of culture using the Luminex technology. Dispersion/chemical transport modeling was used to estimate long-term (whole pregnancy and trimesters) and short-term (15 days before delivery) residential exposures to traffic-related nitrogen dioxide (NO2 ), particulate matter (PM2.5 and PM10 ), and ozone (O3 ). We fitted multivariable logistic regression, Bayesian kernel machine regression (BKMR), and weighted quantile sum (WQS) regression models. RESULTS NO2 during the whole pregnancy increased the odds of detection of IL-1β (OR per 10 µg/m3 increase = 1.37; 95% CI, 1.02, 1.85) and IL-6 (OR per 10 µg/m3 increase = 1.32; 95% CI 1.00, 1.75). Increased odds of detected concentrations of IL-10 was found in newborns exposed during whole pregnancy to higher levels of NO2 (OR per 10 µg/m3 increase = 1.30; 95% CI 0.99, 1.69), PM10 (OR per 10 µg/m3 increase = 1.49; 95% CI 0.95, 2.33), and PM2.5 (OR per 5 µg/m3 increase = 1.56; 95% CI 0.97, 2.51). Exposure to O3 during the whole pregnancy increased the odds of detected IL-13 (OR per 10 µg/m3 increase = 1.22; 95% CI 1.01, 1.49). WQS model revealed first and third trimesters of gestation as windows of higher susceptibility. CONCLUSIONS Gestational exposure to TRAP may increase detection of pro-inflammatory, Th2-related, and T regulatory cytokines in newborns. These changes might influence immune system responses later in life.
Collapse
Affiliation(s)
- Azahara M García-Serna
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Elena Martín-Orozco
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain
| | - Pedro Jiménez-Guerrero
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Regional Atmospheric Modelling Group, Department of Physics, University of Murcia, Murcia, Spain
| | - Trinidad Hernández-Caselles
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain
| | - Virginia Pérez-Fernández
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Public Health Sciences, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | | | - María Dolores Molina-Ruano
- Obstetrics & Gynecology Service, Virgen de la Arrixaca University Clinical Hospital, University of Murcia, Murcia, Spain
| | - Encarna Rojo-Atenza
- Obstetrics & Gynecology Service, Virgen de la Arrixaca University Clinical Hospital, University of Murcia, Murcia, Spain
| | - Luis García-Marcos
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain.,Pediatric Allergy and Pulmonology Units, Virgen de la Arrixaca University Children's Hospital, University of Murcia, Murcia, Spain
| | - Eva Morales
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Public Health Sciences, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
PM10 Alters Trophoblast Cell Function and Modulates miR-125b-5p Expression. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3697944. [PMID: 35036432 PMCID: PMC8759905 DOI: 10.1155/2022/3697944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022]
Abstract
Air pollution is one of the largest global environmental health hazards that threaten premature mortality or morbidity. Particulate matter 10 (PM10) has been demonstrated to contribute to several human diseases via dysregulated miRNA expression. Trophoblast cells play a key role in implantation and placentation for a successful pregnancy. Nonetheless, the PM10 associated trophoblast cell functions during pregnancy and miRNA expression are still unknown. Our study showed that PM10 affected HTR-8/SVneo cell viability and also decreased cell proliferation, migration, and invasion. A high concentration of PM10 caused an increase in HTR-8/SVneo cell apoptosis. Treatment with PM10 induced inflammation through the upregulated IL-1β, IL-6, and TNF-α expression in trophoblast cells. In PM10-treated HTR-8/SVneo cells, miR-125b-5p expression was considerably increased and TXNRD1 was found to be negatively related to miR-125b-5p. Collectively, our findings revealed that PM10 could alter miR-125b-5p expression by targeting TXNRD1 and suppressing trophoblast cell functions. Additional investigations relating to the function of miR-125b-5p and its target on particulate pollution exposure in trophoblast are warranted for future biomarker or effective therapeutic approaches.
Collapse
|
40
|
Huang J, Yang X, Fan F, Hu Y, Wang X, Zhu S, Ren G, Wang G. Outdoor air pollution and the risk of asthma exacerbations in single lag0 and lag1 exposure patterns: a systematic review and meta-analysis. J Asthma 2021; 59:2322-2339. [PMID: 34809505 DOI: 10.1080/02770903.2021.2008429] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Objective: To synthesize evidence regarding the relationship between outdoor air pollution and risk of asthma exacerbations in single lag0 and lag1 exposure patterns.Methods: We performed a systematic literature search using PubMed, Embase, Cochrane Library, Web of Science, ClinicalTrials, China National Knowledge Internet, Chinese BioMedical, and Wanfang databases. Articles published until August 1, 2020 and the reference lists of the relevant articles were reviewed. Two authors independently evaluated the eligible articles and performed structured extraction of the relevant information. Pooled relative risks (RRs) and 95% confidence intervals (CIs) of lag0 and lag1 exposure patterns were estimated using random-effect models.Results: Eighty-four studies met the eligibility criteria and provided sufficient information for meta-analysis. Outdoor air pollutants were associated with increased risk of asthma exacerbations in both single lag0 and lag1 exposure patterns [lag0: RR (95% CI) (pollutants), 1.057(1.011, 1.103) (air quality index, AQI), 1.007 (1.005, 1.010) (particulate matter of diameter ≤ 2.5 μm, PM2.5), 1.009 (1.005, 1.012) (particulate matter of diameter, PM10), 1.010 (1.006, 1.014) (NO2), 1.030 (1.011, 1.048) (CO), 1.005 (1.002, 1.009) (O3); lag1:1.064(1.022, 1.106) (AQI), 1.005 (1.002, 1.008) (PM2.5), 1.007 (1.004, 1.011) (PM10), 1.008 (1.004, 1.012) (NO2), 1.025 (1.007, 1.042) (CO), 1.010 (1.006, 1.013) (O3)], except SO2 [lag0: RR (95% CI), 1.004 (1.000, 1.007); lag1: RR (95% CI), 1.003 (0.999, 1.006)]. Subgroup analyses revealed stronger effects in children and asthma exacerbations associated with other events (including symptoms, lung function changes, and medication use).Conclusion: Outdoor air pollution increases the asthma exacerbation risk in single lag0 and lag1 exposure patterns.Trial registration: PROSPERO, CRD42020204097. https://www.crd.york.ac.uk/.Supplemental data for this article is available online at https://doi.org/10.1080/02770903.2021.2008429 .
Collapse
Affiliation(s)
- Junjun Huang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Xiaoyu Yang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Fangfang Fan
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Hu
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Xi Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Sainan Zhu
- Department of Biostatistics, Peking University First Hospital, Beijing, China
| | - Guanhua Ren
- Department of Library, Peking University First Hospital, Beijing, China
| | - Guangfa Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
41
|
Savouré M, Lequy É, Bousquet J, Chen J, de Hoogh K, Goldberg M, Vienneau D, Zins M, Nadif R, Jacquemin B. Long-term exposures to PM 2.5, black carbon and NO 2 and prevalence of current rhinitis in French adults: The Constances Cohort. ENVIRONMENT INTERNATIONAL 2021; 157:106839. [PMID: 34454361 DOI: 10.1016/j.envint.2021.106839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 08/16/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Rhinitis is one of the most common disease worldwide with a high and increasing prevalence. There is limited knowledge on the link between long-term exposure to air pollution and rhinitis. OBJECTIVES We aim to study the associations between long-term exposure to air pollutants and self-reported current rhinitis among adults from Constances, a large French population-based cohort. METHODS Current rhinitis was defined at inclusion from questionnaire by the presence of sneezing, runny or blocked nose in the last 12 months without a cold or the flu. Annual concentrations of nitrogen dioxide (NO2), particulate matter ≤ 2.5 µm (PM2.5) and black carbon (BC) were estimated at the participants' residential address by European land-use regression models. Cross-sectional associations between annual exposure to each air pollutant and current rhinitis were estimated using logistic models adjusted for age, sex, smoking, education level and French deprivation index. The health prevention centers were taken into account by marginal models with generalized estimating equations. Several supplementary analyses were performed. RESULTS Analyses were performed on 127,108 participants (47 years old on average, 54% women, 19% current smokers). The prevalence of current rhinitis, allergic (AR) and non-allergic (NAR) rhinitis were 36%, 25% and 11% respectively. Adjusted ORs for current rhinitis were 1.13 (1.08, 1.17), 1.12 (1.07, 1.17), and 1.11 (1.06, 1.17) per one interquartile range increase of PM2.5 (4.85 µg/m3), BC (0.88 · 10-5 m-1) and NO2 (17.3 µg/m3) respectively. Significant and positive associations were also found for both AR and NAR. Results were similar in men and women, and in the different smoking strata, and were consistent with meta-analysis or after imputing missing covariates. DISCUSSION An increase of modeled annual average residential exposure to PM2.5, BC, and NO2 was significantly associated with an increase of prevalence of current rhinitis in adults in the French general population. The results suggest that among air pollutants, BC may be of special interest.
Collapse
Affiliation(s)
- Marine Savouré
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Équipe d'Épidémiologie respiratoire intégrative, CESP, 94807 Villejuif, France; Agence de l'environnement et de la Maîtrise de l'Energie, 20, avenue du Grésillé - BP 90406, 49004 Angers Cedex 01, France.
| | - Émeline Lequy
- Université Paris-Saclay, Université de Paris, UVSQ, Inserm, Cohortes Epidémiologiques en population, 94807 Villejuif, France.
| | - Jean Bousquet
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Équipe d'Épidémiologie respiratoire intégrative, CESP, 94807 Villejuif, France.
| | - Jie Chen
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CM Utrecht, the Netherlands.
| | - Kees de Hoogh
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | - Marcel Goldberg
- Université Paris-Saclay, Université de Paris, UVSQ, Inserm, Cohortes Epidémiologiques en population, 94807 Villejuif, France.
| | - Danielle Vienneau
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | - Marie Zins
- Université Paris-Saclay, Université de Paris, UVSQ, Inserm, Cohortes Epidémiologiques en population, 94807 Villejuif, France.
| | - Rachel Nadif
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Équipe d'Épidémiologie respiratoire intégrative, CESP, 94807 Villejuif, France.
| | - Bénédicte Jacquemin
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
42
|
Associations of Air Pollution and Pediatric Asthma in Cleveland, Ohio. ScientificWorldJournal 2021; 2021:8881390. [PMID: 34566522 PMCID: PMC8460381 DOI: 10.1155/2021/8881390] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 11/18/2022] Open
Abstract
Air pollution has been associated with poor health outcomes and continues to be a risk factor for respiratory health in children. While higher particulate matter (PM) levels are associated with increased frequency of symptoms, lower lung function, and increase airway inflammation from asthma, the precise composition of the particles that are more highly associated with poor health outcomes or healthcare utilization are not fully elucidated. PM is measured quantifiably by current air pollution monitoring systems. To better determine sources of PM and speciation of such sources, a particulate matter (PM) source apportionment study, the Cleveland Multiple Air Pollutant Study (CMAPS), was conducted in Cleveland, Ohio, in 2009-2010, which allowed more refined assessment of associations with health outcomes. This article presents an evaluation of short-term (daily) and long-term associations between motor vehicle and industrial air pollution components and pediatric asthma emergency department (ED) visits by evaluating two sets of air quality data with healthcare utilization for pediatric asthma. Exposure estimates were developed using land use regression models for long-term exposures for nitrogen dioxide (NO2) and coarse (i.e., with aerodynamic diameters between 2.5 and 10 μm) particulate matter (PM) and the US EPA Positive Matrix Factorization receptor model for short-term exposures to fine (<2.5 μm) and coarse PM components. Exposure metrics from these two approaches were used in asthma ED visit prevalence and time series analyses to investigate seasonal-averaged short- and long-term impacts of both motor vehicles and industry emissions. Increased pediatric asthma ED visits were found for LUR coarse PM and NO2 estimates, which were primarily contributed by motor vehicles. Consistent, statistically significant associations with pediatric asthma visits were observed, with short-term exposures to components of fine and coarse iron PM associated with steel production. Our study is the first to combine spatial and time series analysis of ED visits for asthma using the same periods and shows that PM related to motor vehicle emissions and iron/steel production are associated with increased pediatric asthma visits.
Collapse
|
43
|
Sun B, Song J, Wang Y, Jiang J, An Z, Li J, Zhang Y, Wang G, Li H, Alexis NE, Jaspers I, Wu W. Associations of short-term PM 2.5 exposures with nasal oxidative stress, inflammation and lung function impairment and modification by GSTT1-null genotype: A panel study of the retired adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 285:117215. [PMID: 33932759 DOI: 10.1016/j.envpol.2021.117215] [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: 12/14/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
PM2.5 (particulate matter ≤2.5 μm in aerodynamic diameter) is a major urban air pollutant worldwide. Its effects on the respiratory system of the susceptible population have been less characterized. This study aimed to estimate the association of short-term PM2.5 exposure with respiratory outcomes of the retired adults, and to examine whether these associations were stronger among the subjects with GSTT-null genotype. 32 healthy subjects (55-77 years) were recruited for five follow-up examinations. Ambient concentrations of PM2.5 were monitored consecutively for 7 days prior to physical examination. Pulmonary outcomes including forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), peak expiratory flow (PEF), and fractional exhaled nitric oxide (FeNO), and nasal fluid concentrations of 8-epi-prostaglandin F2 alpha (8-epi-PGF2α), tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8) and IL-1β were measured. A linear mixed-effect model was introduced to evaluate the associations of PM2.5 concentrations with respiratory outcomes. Additionally, GSTT1 genotype-based stratification was performed to characterize modification on PM2.5-related respiratory outcomes. We found that a 10 μg/m3 increase in PM2.5 was associated with decreases of 0.52 L (95% confidence interval [CI]: -1.04, -0.002), 0.64 L (95% CI: -1.13, -0.16), 0.1 (95% CI: -0.23, 0.04) and 2.87 L/s (95% CI: -5.09, -0.64) in FVC, FEV1, FEV1/FVC ratio and PEF at lag 2, respectively. Meanwhile, marked increases of 80.82% (95% CI: 5.13%, 156.50%) in IL-8, 77.14% (95% CI: 1.88%, 152.40%) in IL-1β and 67.87% (95% CI: 14.85%, 120.88%) in 8-epi-PGF2α were observed as PM2.5 concentration increased by 10 μg/m3 at lag 2. Notably, PM2.5-associated decreases in FVC and PEF and increase in FeNO were stronger among the subjects with GSTT1-null genotype. In summary, short-term exposure to PM2.5 is associated with nasal inflammation, oxidative stress and lung function reduction in the retired subjects. Lung function reduction and inflammation are stronger among the subjects with GSTT1-null genotype.
Collapse
Affiliation(s)
- Beibei Sun
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Ya Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Jing Jiang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Yange Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Gui Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Huijun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China.
| |
Collapse
|
44
|
Ahn K. Intervention of Particulate Matter: What Can We Do for Asthmatic Patients? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:677-680. [PMID: 34486253 PMCID: PMC8419650 DOI: 10.4168/aair.2021.13.5.677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/20/2022]
Affiliation(s)
- Kangmo Ahn
- Environmental Health Center for Atopic Diseases, Samsung Medical Center, Seoul, Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
45
|
Ali MU, Yu Y, Yousaf B, Munir MAM, Ullah S, Zheng C, Kuang X, Wong MH. Health impacts of indoor air pollution from household solid fuel on children and women. JOURNAL OF HAZARDOUS MATERIALS 2021; 416:126127. [PMID: 34492921 DOI: 10.1016/j.jhazmat.2021.126127] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 05/12/2021] [Indexed: 05/11/2023]
Abstract
The inefficient and incomplete combustion of solid fuel (SF) is associated with high levels of indoor air pollutants leading to 3.55 million deaths annually. The risk is higher in women and children, due to their higher exposure duration and unique physical properties. The current article aims to provide a critical overview regarding the use of solid fuel, its associated pollutants, their toxicity mechanisms and, most importantly the associated health impacts, especially in women and children. Pollutants associated with SF mostly include polycyclic aromatic hydrocarbons, particulate matter, nitrous oxide, carbon monoxide and sulfur dioxide, and their concentrations are two- to threefold higher in indoor environments. These pollutants can lead to a variety of health risks by inducing different toxicity mechanisms, such as oxidative stress, DNA methylation, and gene activation. Exposed children have an increased prevalence of low birth weight, acute lower respiratory tract infections, anemia and premature mortality. On the other hand, lung cancer, chronic obstructive pulmonary disease and cardiovascular diseases are the major causes of disability and premature death in women. Indoor air pollution resulting from SF combustion is a major public health threat globally. To reduce the risks, it is important to identify future research gaps and implement effective interventions and policies.
Collapse
Affiliation(s)
- Muhammad Ubaid Ali
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, and State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, China.
| | - Yangmei Yu
- Consortium on Health, Environment, Education and Research (CHEER), Department of Science and Environmental Studies, The Education University of Hong Kong, Hong Kong, China.
| | - Balal Yousaf
- Department of Environment Engineering, Middle East Technical University, Ankara 06800, Turkey; CAS-Key Laboratory of Crust-Mantle Materials and the Environments, School of Earth and Space Sciences, University of Science and Technology of China, Hefei 230026, China.
| | - Mehr Ahmed Mujtaba Munir
- CAS-Key Laboratory of Crust-Mantle Materials and the Environments, School of Earth and Space Sciences, University of Science and Technology of China, Hefei 230026, China.
| | - Sami Ullah
- Department of Forestry, Shaheed Benazir Bhutto University Sheringal, Dir Upper, KPK, Pakistan.
| | - Chunmiao Zheng
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, and State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, China.
| | - Xingxing Kuang
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, and State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, China.
| | - Ming Hung Wong
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, and State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, China; Consortium on Health, Environment, Education and Research (CHEER), Department of Science and Environmental Studies, The Education University of Hong Kong, Hong Kong, China.
| |
Collapse
|
46
|
Abstract
Since the industrial revolution, air pollution has become a major problem causing several health problems involving the airways as well as the cardiovascular, reproductive, or neurological system. According to the WHO, about 3.6 million deaths every year are related to inhalation of polluted air, specifically due to pulmonary diseases. Polluted air first encounters the airways, which are a major human defense mechanism to reduce the risk of this aggressor. Air pollution consists of a mixture of potentially harmful compounds such as particulate matter, ozone, carbon monoxide, volatile organic compounds, and heavy metals, each having its own effects on the human body. In the last decades, a lot of research investigating the underlying risks and effects of air pollution and/or its specific compounds on the airways, has been performed, involving both in vivo and in vitro experiments. The goal of this review is to give an overview of the recent data on the effects of air pollution on healthy and diseased airways or models of airway disease, such as asthma or chronic obstructive pulmonary disease. Therefore, we focused on studies involving pollution and airway symptoms and/or damage both in mice and humans.
Collapse
|
47
|
Losol P, Choi JP, Kim SH, Chang YS. The Role of Upper Airway Microbiome in the Development of Adult Asthma. Immune Netw 2021; 21:e19. [PMID: 34277109 PMCID: PMC8263217 DOI: 10.4110/in.2021.21.e19] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Clinical and molecular phenotypes of asthma are complex. The main phenotypes of adult asthma are characterized by eosinophil and/or neutrophil cell dominant airway inflammation that represent distinct clinical features. Upper and lower airways constitute a unique system and their interaction shows functional complementarity. Although human upper airway contains various indigenous commensals and opportunistic pathogenic microbiome, imbalance of this interactions lead to pathogen overgrowth and increased inflammation and airway remodeling. Competition for epithelial cell attachment, different susceptibilities to host defense molecules and antimicrobial peptides, and the production of proinflammatory cytokine and pattern recognition receptors possibly determine the pattern of this inflammation. Exposure to environmental factors, including infection, air pollution, smoking is commonly associated with asthma comorbidity, severity, exacerbation and resistance to anti-microbial and steroid treatment, and these effects may also be modulated by host and microbial genetics. Administration of probiotic, antibiotic and corticosteroid treatment for asthma may modify the composition of resident microbiota and clinical features. This review summarizes the effect of some environmental factors on the upper respiratory microbiome, the interaction between host-microbiome, and potential impact of asthma treatment on the composition of the upper airway microbiome.
Collapse
Affiliation(s)
- Purevsuren Losol
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| |
Collapse
|
48
|
Misiukiewicz-Stepien P, Paplinska-Goryca M. Biological effect of PM 10 on airway epithelium-focus on obstructive lung diseases. Clin Immunol 2021; 227:108754. [PMID: 33964432 DOI: 10.1016/j.clim.2021.108754] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
Recently, a continuous increase in environmental pollution has been observed. Despite wide-scale efforts to reduce air pollutant emissions, the problem is still relevant. Exposure to elevated levels of airborne particles increased the incidence of respiratory diseases. PM10 constitute the largest fraction of air pollutants, containing particles with a diameter of less than 10 μm, metals, pollens, mineral dust and remnant material from anthropogenic activity. The natural airway defensive mechanisms against inhaled material, such as mucus layer, ciliary clearance and macrophage phagocytic activity, may be insufficient for proper respiratory function. The epithelium layer can be disrupted by ongoing oxidative stress and inflammatory processes induced by exposure to large amounts of inhaled particles as well as promote the development and exacerbation of obstructive lung diseases. This review draws attention to the current state of knowledge about the physical features of PM10 and its impact on airway epithelial cells, and obstructive pulmonary diseases.
Collapse
Affiliation(s)
- Paulina Misiukiewicz-Stepien
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland; Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland.
| | | |
Collapse
|
49
|
Ohlwein S, Hennig F, Lucht S, Schmidt B, Eisele L, Arendt M, Dührsen U, Dürig J, Jöckel KH, Moebus S, Hoffmann B. Air Pollution and Polyclonal Elevation of Serum Free Light Chains: An Assessment of Adaptive Immune Responses in the Prospective Heinz Nixdorf Recall Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:27004. [PMID: 33596105 PMCID: PMC7889003 DOI: 10.1289/ehp7164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Residential exposure to air pollution (AP) has been shown to activate the immune system (IS). Although innate immune responses to AP have been studied extensively, investigations on the adaptive IS are scarce. OBJECTIVES The aim of this study was to investigate the association between short- to long-term AP exposure and polyclonal free light chains (FLC) produced by plasma cells. METHODS We used repeated data from three examinations (t0: 2000-2003; t1: 2006-2008; and t2: 2011-2015) of the population-based German Heinz Nixdorf Recall cohort of initially 4,814 participants (45-75 y old). Residential exposure to total and source-specific particulate matter (PM) with an aerodynamic diameter of 10 or 2.5μm (PM10 and PM2.5 respectively), nitrogen dioxide (NO2), and particle number concentrations (accumulation mode; PNAM) was estimated using a chemistry transport model with different time windows (1- to 365-d mean ± standard deviation) before blood draw. We applied linear mixed models with a random participant intercept to estimate associations between total, traffic- and industry-related AP exposures and log-transformed FLC, controlling for examination time, sociodemographic and lifestyle variables, estimated glomerular filtration rate and season. RESULTS Analyzing 9,933 observations from 4,455 participants, we observed generally positive associations between AP exposures and FLC. We observed strongest associations with middle-term exposures, e.g., 3.0% increase in FLC (95% confidence interval: 1.8%, 4.3%) per interquartile range increase in 91-d mean of NO2 (14.1μg/m³). Across the different pollutants, NO2 showed strongest associations with FLC, followed by PM10 and PNAM. Effect estimates for traffic-related exposures were mostly higher compared with total exposures. Although NO2 and PNAM estimates remained stable upon adjustment for PM, PM estimates decreased considerably upon adjustment for NO2 and PNAM. DISCUSSION Our results suggest that middle-term AP exposures in particular might be positively associated with activation of the adaptive IS. Traffic-related PM, PNAM, and NO2 showed strongest associations. https://doi.org/10.1289/EHP7164.
Collapse
Affiliation(s)
- Simone Ohlwein
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Frauke Hennig
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Sarah Lucht
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Börge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Lewin Eisele
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Marina Arendt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, University Hospital Essen, Germany
| | - Jan Dürig
- Department of Hematology, University Hospital Essen, Germany
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
- Centre for Urban Epidemiology (CUE), Institute of Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Barbara Hoffmann
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
50
|
Asthma and air pollution: recent insights in pathogenesis and clinical implications. Curr Opin Pulm Med 2021; 26:10-19. [PMID: 31724961 DOI: 10.1097/mcp.0000000000000644] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Air pollution has adverse effects on the onset and morbidity of respiratory diseases, including asthma. In this review, we discuss recent insights into the effects of air pollution on the incidence and exacerbation of asthma. We focus on epidemiological studies that describe the association between air pollution exposure and development, mortality, persistence and exacerbations of asthma among different age groups. Moreover, we also provide an update on translational studies describing the mechanisms behind this association. RECENT FINDINGS Mechanisms linking air pollutants such as particulate matter, nitrogen dioxide (NO2) and ozone to the development and exacerbation of asthma include the induction of both eosinophilic and neutrophilic inflammation driven by stimulation of airway epithelium and increase of pro-inflammatory cytokine production, oxidative stress and DNA methylation changes. Although exposure during foetal development is often reported as a crucial timeframe, exposure to air pollution is detrimental in people of all ages, thus influencing asthma onset as well as increase in asthma prevalence, mortality, persistence and exacerbation. SUMMARY In conclusion, this review highlights the importance of reducing air pollution levels to avert the progressive increase in asthma incidence and morbidity.
Collapse
|