1
|
Li YL, Chang PY, Chuang TW, Hsieh YC, Wang BS, Chen SY, Chiou HY. Association of long-term ozone exposure with the incidence and progression of hypertension, diabetes, and chronic kidney disease: A national retrospective cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 975:179209. [PMID: 40187338 DOI: 10.1016/j.scitotenv.2025.179209] [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: 09/06/2024] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
Evidence suggests that ozone is associated with an increased risk of hypertension, diabetes, or chronic kidney disease (CKD). However, the associations of ozone exposure with the dynamic progression of these diseases among Asian population remain unknown. This study included 9,256,945 participants from Taiwan's National Health Insurance Research Database between 2006 and 2021. Multimorbidity was defined as the coexistence of CKD and either hypertension or diabetes. The ordinary kriging method was used to estimate daily concentrations of ozone, sulfur dioxide, carbon monoxide, nitrogen dioxide, suspended fine particles, and suspended particles. Then, five-year average concentrations of pollutants were calculated. We performed multi-state survival models to analyze the association between ozone and dynamic progression of these diseases. During follow-up, 3,555,498 participants experienced hypertension, diabetes, or CKD; 656,515 experienced multimorbidity; and 792,555 died. Ozone exposure was significantly associated with incidence of the results in all transitions. The hazard ratios of each IQR (3.57 ppb) increment in ozone for the transition to incident disease were 1.016 [95 % confidence interval (CI): 1.014, 1.017], for the transition to death were 1.04 [95 % CI: 1.036, 1.043], for the transition to multimorbidity were 1.015 [95 % CI: 1.012, 1.017]. Furthermore, with each IQR increase of ozone, the hazard ratios for transition from the disease incidence to death and from multimorbidity to death were 1.03 [95 % CI: 1.026, 1.033] and 1.007 [95 % CI: 1.002, 1.013], respectively. Our results suggest long-term exposure to ozone might be an important determinant for the incidence and dynamic progression of hypertension, diabetes, and CKD in Taiwan.
Collapse
Affiliation(s)
- Yu-Ling Li
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Po-Ya Chang
- Department of Leisure Industry and Health Promotion, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Ting-Wu Chuang
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chen Hsieh
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Bo-Sian Wang
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Szu-Ying Chen
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Hung-Yi Chiou
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; Master Program in Applied Epidemiology, College of Public Health, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Hussain S, Majumder N, Mazumder MHH, Lewis SE, Olapeju O, Velayutham M, Amin MS, Brundage K, Kelley EE, Vanoirbeek J. Intermittent ozone inhalation during house dust mite-induced sensitization primes for adverse asthma phenotype. Redox Biol 2024; 76:103330. [PMID: 39244793 PMCID: PMC11407077 DOI: 10.1016/j.redox.2024.103330] [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: 06/11/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
The ability of air pollution to induce acute exacerbation of asthma is well documented. However, the ability of ozone (O3), the most reactive gaseous component of air pollution, to function as a modulator during sensitization is not well established. C57BL/6 J male mice were intranasally sensitized to house dust mite (HDM) (40 μg/kg) for 3 weeks on alternate days in parallel with once-a-week O3 exposure (1 ppm). Mice were euthanized 24 h following the last HDM challenge. Lung lavage, histology, lung function (both forced oscillation and forced expiration-based), immune cell profiling, inflammation (pulmonary and systemic), and immunoglobulin production were assessed. Compared to HDM alone, HDM + O3 leads to a significant increase in peribronchial inflammation (p < 0.01), perivascular inflammation (p < 0.001) and methacholine-provoked large airway hyperreactivity (p < 0.05). Serum total IgG and IgE and HDM-specific IgG1 were 3-5 times greater in HDM + O3 co-exposure compared to PBS and O3-exposed groups. An increase in activated/mature lung total and monocyte-derived dendritic cells (p < 0.05) as well as T-activated, and T memory lymphocyte subset numbers (p < 0.05) were noted in the HDM + O3 group compared to HDM alone group. Concurrent O3 inhalation and HDM sensitization also caused significantly greater (p < 0.05) lung tissue interleukin-17 pathway gene expression and mediator levels in the serum. Redox imbalance was manifested by impaired lung antioxidant defense and increased oxidants. O3 inhalation during allergic sensitization coalesces in generating a significantly worse TH17 asthmatic phenotype.
Collapse
Affiliation(s)
- Salik Hussain
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA; Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, USA.
| | - Nairrita Majumder
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Md Habibul Hasan Mazumder
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Sara E Lewis
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Olanrewaju Olapeju
- Pathology, Anatomy and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Murugesan Velayutham
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA; Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Md Shahrier Amin
- Pathology, Anatomy and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Kathleen Brundage
- Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Eric E Kelley
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Jeroen Vanoirbeek
- KU Leuven, Department of Public Health and Primary Care, Centre for Environment and Health, Leuven, Belgium
| |
Collapse
|
3
|
Peng S, Chen B, Li Z, Sun J, Liu F, Yin X, Zhou Y, Shen H, Xiang H. Ambient ozone pollution impairs glucose homeostasis and contributes to renal function decline: Population-based evidence. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115803. [PMID: 38091674 PMCID: PMC10790241 DOI: 10.1016/j.ecoenv.2023.115803] [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: 08/26/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024]
Abstract
Particulate matter pollution could increase the risk of kidney disease, while evidence for ozone exposure is less well-established. Here, we aimed to evaluate the effect of ozone pollution on renal function and explore mechanisms. We first conducted a cross-sectional study based on Wuhan Chronic Disease Cohort Study baseline information. We recruited 2699 eligible participants, estimated their residential ozone concentrations, collected fasting peripheral blood samples for biochemical analysis and calculated the estimated glomerular filtration rate (eGFR). The linear regression model was applied to evaluate the long-term association between ozone pollution and eGFR. Then, we recruited another 70 volunteers as a panel with 8 rounds follow-up visits. We calculated the eGFR and measured fasting blood glucose and lipid levels. The linear mixed-effect model along with mediation analysis were performed to confirm the short-term association and explore potential mechanisms, respectively. For the long-term association, a 10.95 μg/m3 increment of 3-year ozone exposure was associated with 2.96 mL/min/1.73 m2 decrease in eGFR (95%CI: -4.85, -1.06). Furthermore, the drinkers exhibited a pronounced declination of eGFR (-7.46 mL/min/1.73 m2, 95%CI: -11.84, -3.08) compared to non-drinkers in relation to ozone exposure. Additionally, a 19.02 μg/m3 increase in 3-day ozone concentrations was related to 2.51 mL/min/1.73 m2 decrease in eGFR (95%CI: -3.78, -1.26). Hyperglycemia and insulin resistance mediated 12.2% and 16.5% of the aforementioned association, respectively. Our findings indicated that higher ozone pollution could affect renal function, and the hyperglycemia and insulin resistance linked to ozone might be the underlying mechanisms.
Collapse
Affiliation(s)
- Shouxin Peng
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China; Global Health Institute, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Bingbing Chen
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Zhaoyuan Li
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China; Global Health Institute, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Jinhui Sun
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China; Global Health Institute, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Feifei Liu
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China; Global Health Institute, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Xiaoyi Yin
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Yi Zhou
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China
| | - Huanfeng Shen
- School of Resource and Environmental Sciences, Wuhan University, Wuhan 430079, Hubei, PR China.
| | - Hao Xiang
- Department of Global Health, School of Public Health, Wuhan University, Wuhan 430071, Hubei, PR China; Global Health Institute, Wuhan University, Wuhan 430071, Hubei, PR China.
| |
Collapse
|
4
|
Struß N, Dieter S, Schwarz K, Badorrek P, Hohlfeld JM. Sodium Chloride versus Lactose as a Carrier for House Dust Mite Allergen in Allergen Chamber Studies: A Clinical Study to Assess Noninferiority. Int Arch Allergy Immunol 2023; 184:1143-1152. [PMID: 37586347 DOI: 10.1159/000531878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/29/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION In the Fraunhofer allergen challenge chamber (ACC), a standardized, universal, good manufacturing practice-conforming technology using a spray dried solution of lactose (L) and allergen extract has been established. In this study, we investigated the noninferiority of hypertonic sodium chloride (S) versus L as a carrier for house dust mite (HDM) allergen to simplify manufacturing, reduce costs, and allow for wider use. METHODS Using a participant-blinded, sham exposure-controlled, single-arm, sequential intervention study, we challenged adults with HDM allergic rhinitis five times in the ACC. Participants were first exposed to S, L, and clean air (block 1), followed by S + HDM and L + HDM (block 2). Primary endpoints were mean total nasal symptom score (TNSS) and mean nasal secretion weight. RESULTS 19 participants were enrolled in the study (10 females; mean age 32 years [22-49], 4 with mild allergic asthma). The safety profile of S + HDM and L + HDM was similar; eight participants experienced mild procedure-related adverse events including tiredness, cough, and dyspnea. Due to dropouts, 13 participants completed the study and were evaluated. Mean TNSS and nasal secretion were as follows: S 0.98, 0.28 g; L 1.1, 0.20 g; clean air 1.1, 0.23 g; S + HDM 5.7, 4.8 g; L + HDM 5.1, 5.1 g. Separate block 1/block 2 MANOVAs with TNSS and nasal secretion as dependent variables revealed no significant differences between the carriers, neither alone and compared with clean air (p = 0.2059, Wilk's λ = 0.78) nor combined with HDM (p = 0.3474, Wilk's λ = 0.89). Noninferiority of S was established using a meta-analysis-based minimal clinical important difference of -0.55: mean TNSS difference between S + HDM and L + HDM was 0.62 (90% CI: -0.51 to 1.74). CONCLUSION S as an HDM carrier was safe and well tolerated. It was noninferior to L which makes it an adequate and easy-to-use carrier substitute.
Collapse
Affiliation(s)
- Nadja Struß
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Susanne Dieter
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Katharina Schwarz
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Philipp Badorrek
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Jens M Hohlfeld
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover BREATH, Hannover, Germany
| |
Collapse
|
5
|
Laursen KR, Christensen NV, Mulder FA, Schullehner J, Hoffmann HJ, Jensen A, Møller P, Loft S, Olin AC, Rasmussen BB, Rosati B, Strandberg B, Glasius M, Bilde M, Sigsgaard T. Airway and systemic biomarkers of health effects after short-term exposure to indoor ultrafine particles from cooking and candles - A randomized controlled double-blind crossover study among mild asthmatic subjects. Part Fibre Toxicol 2023; 20:26. [PMID: 37430267 DOI: 10.1186/s12989-023-00537-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/28/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND There is insufficient knowledge about the systemic health effects of exposure to fine (PM2.5) and ultrafine particles emitted from typical indoor sources, including cooking and candlelight burning. We examined whether short-term exposure to emissions from cooking and burning candles cause inflammatory changes in young individuals with mild asthma. Thirty-six non-smoking asthmatics participated in a randomized controlled double-blind crossover study attending three exposure sessions (mean PM2.5 µg/m3; polycyclic aromatic hydrocarbons ng/m3): (a) air mixed with emissions from cooking (96.1; 1.1), (b) air mixed with emissions from candles (89.8; 10), and (c) clean filtered air (5.8; 1.0). Emissions were generated in an adjacent chamber and let into a full-scale exposure chamber where participants were exposed for five hours. Several biomarkers were assessed in relation to airway and systemic inflammatory changes; the primary outcomes of interest were surfactant Protein-A (SP-A) and albumin in droplets in exhaled air - novel biomarkers for changes in the surfactant composition of small airways. Secondary outcomes included cytokines in nasal lavage, cytokines, C-reactive protein (CRP), epithelial progenitor cells (EPCs), genotoxicity, gene expression related to DNA-repair, oxidative stress, and inflammation, as well as metabolites in blood. Samples were collected before exposure start, right after exposure and the next morning. RESULTS SP-A in droplets in exhaled air showed stable concentrations following candle exposure, while concentrations decreased following cooking and clean air exposure. Albumin in droplets in exhaled air increased following exposure to cooking and candles compared to clean air exposure, although not significant. Oxidatively damaged DNA and concentrations of some lipids and lipoproteins in the blood increased significantly following exposure to cooking. We found no or weak associations between cooking and candle exposure and systemic inflammation biomarkers including cytokines, CRP, and EPCs. CONCLUSIONS Cooking and candle emissions induced effects on some of the examined health-related biomarkers, while no effect was observed in others; Oxidatively damaged DNA and concentrations of lipids and lipoproteins were increased in blood after exposure to cooking, while both cooking and candle emissions slightly affected the small airways including the primary outcomes SP-A and albumin. We found only weak associations between the exposures and systemic inflammatory biomarkers. Together, the results show the existence of mild inflammation following cooking and candle exposure.
Collapse
Affiliation(s)
- Karin Rosenkilde Laursen
- Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Nichlas Vous Christensen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus, Denmark
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Frans Aa Mulder
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus, Denmark
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Jörg Schullehner
- Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark
- Geological Survey of Denmark and Greenland, Aarhus, Denmark
| | - Hans Jürgen Hoffmann
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Annie Jensen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Aarhus, Denmark
| | - Peter Møller
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Aarhus, Denmark
| | - Steffen Loft
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Aarhus, Denmark
| | - Anna-Carin Olin
- Department of Public Health and Community Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Bernadette Rosati
- Department of Chemistry, Aarhus University, Aarhus, Denmark
- Faculty of Physics, University of Vienna, Vienna, Austria
| | - Bo Strandberg
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | | | - Merete Bilde
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Torben Sigsgaard
- Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Travagli V, Iorio EL. The Biological and Molecular Action of Ozone and Its Derivatives: State-of-the-Art, Enhanced Scenarios, and Quality Insights. Int J Mol Sci 2023; 24:ijms24108465. [PMID: 37239818 DOI: 10.3390/ijms24108465] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The ultimate objective of this review is to encourage a multi-disciplinary and integrated methodological approach that, starting from the recognition of some current uncertainties, helps to deepen the molecular bases of ozone treatment effects on human and animal well-being and to optimize their performance in terms of reproducibility of results, quality, and safety. In fact, the common therapeutic treatments are normally documented by healthcare professionals' prescriptions. The same applies to medicinal gases (whose uses are based on their pharmacological effects) that are intended for patients for treatment, diagnostic, or preventive purposes and that have been produced and inspected in accordance with good manufacturing practices and pharmacopoeia monographs. On the contrary, it is the responsibility of healthcare professionals, who thoughtfully choose to use ozone as a medicinal product, to achieve the following objectives: (i) to understand the molecular basis of the mechanism of action; (ii) to adjust the treatment according to the clinical responses obtained in accordance with the principles of precision medicine and personalized therapy; (iii) to ensure all quality standards.
Collapse
Affiliation(s)
- Valter Travagli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Viale Aldo Moro 2, 53100 Siena, Italy
| | - Eugenio Luigi Iorio
- International Observatory of Oxidative Stress, 84127 Salerno, Italy
- Campus Uberlândia, Universidade de Uberaba (UNIUBE), Uberlândia 38055-500, Brazil
| |
Collapse
|
7
|
Ogrotis I, Koufakis T, Kotsa K. Changes in the Global Epidemiology of Type 1 Diabetes in an Evolving Landscape of Environmental Factors: Causes, Challenges, and Opportunities. Medicina (B Aires) 2023; 59:medicina59040668. [PMID: 37109626 PMCID: PMC10141720 DOI: 10.3390/medicina59040668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
The worldwide incidence of type 1 diabetes mellitus (T1DM) has increased in recent decades. The reasons behind this phenomenon are not yet fully understood. Early life infections, prenatal and perinatal factors, and diet composition have been associated with the triggering of autoimmunity and the risk of presentation of T1DM. However, the rapid increase in new cases of the disease raises the hypothesis that lifestyle factors, which have traditionally been associated with type 2 diabetes, such as obesity and unhealthy eating patterns could also play a role in the genesis of autoimmune diabetes. This article aims to highlight the changing epidemiology of T1DM and the importance of properly recognizing the environmental factors behind it, as well as the connections with the pathogenesis of the disorder and the need to prevent or delay T1DM and its long-term complications.
Collapse
Affiliation(s)
- Ioannis Ogrotis
- School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-231-099-4706
| |
Collapse
|
8
|
Molot J, Sears M, Marshall LM, Bray RI. Neurological susceptibility to environmental exposures: pathophysiological mechanisms in neurodegeneration and multiple chemical sensitivity. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:509-530. [PMID: 34529912 DOI: 10.1515/reveh-2021-0043] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/13/2021] [Indexed: 05/23/2023]
Abstract
The World Health Organization lists air pollution as one of the top five risks for developing chronic non-communicable disease, joining tobacco use, harmful use of alcohol, unhealthy diets and physical inactivity. This review focuses on how host defense mechanisms against adverse airborne exposures relate to the probable interacting and overlapping pathophysiological features of neurodegeneration and multiple chemical sensitivity. Significant long-term airborne exposures can contribute to oxidative stress, systemic inflammation, transient receptor subfamily vanilloid 1 (TRPV1) and subfamily ankyrin 1 (TRPA1) upregulation and sensitization, with impacts on olfactory and trigeminal nerve function, and eventual loss of brain mass. The potential for neurologic dysfunction, including decreased cognition, chronic pain and central sensitization related to airborne contaminants, can be magnified by genetic polymorphisms that result in less effective detoxification. Onset of neurodegenerative disorders is subtle, with early loss of brain mass and loss of sense of smell. Onset of MCS may be gradual following long-term low dose airborne exposures, or acute following a recognizable exposure. Upregulation of chemosensitive TRPV1 and TRPA1 polymodal receptors has been observed in patients with neurodegeneration, and chemically sensitive individuals with asthma, migraine and MCS. In people with chemical sensitivity, these receptors are also sensitized, which is defined as a reduction in the threshold and an increase in the magnitude of a response to noxious stimulation. There is likely damage to the olfactory system in neurodegeneration and trigeminal nerve hypersensitivity in MCS, with different effects on olfactory processing. The associations of low vitamin D levels and protein kinase activity seen in neurodegeneration have not been studied in MCS. Table 2 presents a summary of neurodegeneration and MCS, comparing 16 distinctive genetic, pathophysiological and clinical features associated with air pollution exposures. There is significant overlap, suggesting potential comorbidity. Canadian Health Measures Survey data indicates an overlap between neurodegeneration and MCS (p < 0.05) that suggests comorbidity, but the extent of increased susceptibility to the other condition is not established. Nevertheless, the pathways to the development of these conditions likely involve TRPV1 and TRPA1 receptors, and so it is hypothesized that manifestation of neurodegeneration and/or MCS and possibly why there is divergence may be influenced by polymorphisms of these receptors, among other factors.
Collapse
Affiliation(s)
- John Molot
- Family Medicine, University of Ottawa Faculty of Medicine, North York, ON, Canada
| | | | | | - Riina I Bray
- Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Yang M, Chang X, Gao Q, Gong X, Zheng J, Liu H, Li K, Zhan H, Wang X, Li S, Sun X, Feng S, Sun Y. LncRNA MEG3 ameliorates NiO nanoparticles-induced pulmonary inflammatory damage via suppressing the p38 mitogen activated protein kinases pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1058-1070. [PMID: 35006638 DOI: 10.1002/tox.23464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/23/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
The lung inflammatory damage could result from the nickel oxide nanoparticles (NiO NPs), in which the underlying mechanism is still unclear. This article explored the roles of long noncoding RNA maternally expressed gene 3 (lncRNA MEG3) and p38 mitogen activated protein kinases (p38 MAPK) pathway in pulmonary inflammatory injury induced by NiO NPs. Wistar rats were treated with NiO NPs suspensions (0.015, 0.06, and 0.24 mg/kg) by intratracheal instillation twice-weekly for 9 weeks. Meanwhile, A549 cells were treated with NiO NPs suspensions (25, 50, and 100 μg/ml) for 24 h. It can be concluded that the NiO NPs did trigger pulmonary inflammatory damage, which was confirmed by the histopathological examination, abnormal changes of inflammatory cells and inflammatory cytokines (IL-1β, IL-6, TGF-β1, TNF-α, IFN-γ, IL-10, CXCL-1 and CXCL-2) in bronchoalveolar lavage fluid (BALF), pulmonary tissue and cell culture supernatant. Furthermore, NiO NPs activated the p38 MAPK pathway and downregulated MEG3 in vivo and in vitro. However, p38 MAPK pathway inhibitor (10 μM SB203580) reversed the alterations in the expression levels of inflammatory cytokines induced by NiO NPs. Meanwhile, over-expressed MEG3 significantly suppressed NiO NPs-induced p38 MAPK pathway activation and inflammatory cytokines changes. Overall, the above results proved that over-expression of lncRNA MEG3 reduced NiO NPs-induced inflammatory damage by preventing the activation of p38 MAPK pathway.
Collapse
Affiliation(s)
- Mengmeng Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Qing Gao
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuefeng Gong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Jinfa Zheng
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Han Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Kun Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haibing Zhan
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Sheng Li
- Department of Public Health, The First People's Hospital of Lanzhou City, Lanzhou, China
| | - Xingchang Sun
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Sanwei Feng
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
10
|
Tempol Preserves Endothelial Progenitor Cells in Male Mice with Ambient Fine Particulate Matter Exposure. Biomedicines 2022; 10:biomedicines10020327. [PMID: 35203535 PMCID: PMC8869086 DOI: 10.3390/biomedicines10020327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/28/2022] Open
Abstract
Ambient fine particulate matter (PM) exposure associates with an increased risk of cardiovascular diseases (CVDs). Major sex differences between males and females exist in epidemiology, pathophysiology, and outcome of CVDs. Endothelial progenitor cells (EPCs) play a vital role in the development and progression of CVDs. PM exposure-induced reduction of EPCs is observed in male, not female, mice with increased reactive oxygen species (ROS) production and oxidative stress. The lung is considered an important source of ROS in mice with PM exposure. The aim of the present study was to investigate the sex differences in pulmonary superoxide dismutase (SOD) expression and ROS production, and to test the effect of SOD mimic Tempol on the populations of EPCs in mice with PM exposure. Both male and female C57BL/6 mice (8–10 weeks) were exposed to intranasal PM or vehicle for 6 weeks. Flow cytometry analysis demonstrated that PM exposure significantly decreased the levels of EPCs (CD34+/CD133+) in both blood and bone marrow with increased ROS production in males, but not in females. ELISA analysis showed higher levels of serum IL-6 and IL-1βin males than in females. Pulmonary expression of the antioxidant enzyme SOD1 was significantly decreased in males after PM exposure, but not in females. Administration of the SOD mimic Tempol in male mice with PM exposure attenuated the production of ROS and inflammatory cytokines, and preserved EPC levels. These data indicated that PM exposure-induced reduction of EPC population in male mice may be due to decreased expression of pulmonary SOD1 in male mice.
Collapse
|
11
|
Measurement of oxidatively damaged DNA in mammalian cells using the comet assay: Reflections on validity, reliability and variability. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 873:503423. [PMID: 35094807 DOI: 10.1016/j.mrgentox.2021.503423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022]
Abstract
The comet assay is a simple technique for measurements of low levels of DNA damage and repair in single cells. However, there is variation in background levels of DNA damage in peripheral blood mononuclear cells (PBMCs). This variation has been documented by inter-laboratory ring-trials where identical samples have been analysed in different laboratories using the formamidopyrimidine DNA glycosylase (Fpg)-modified comet assay. The coefficient of variation of background levels of Fpg-sensitive sites was 128 % in the first inter-laboratory validation trial called European Standards Committee on Oxidative DNA Damage. The variation was reduced to 44 % by the end of the project. Subsequent ring-trials by the European Comet Assay Validation Group showed similar inter-laboratory variation in Fpg-sensitive sites in PBMCs (45 %). The lowest inter-laboratory variation in Fpg-sensitive sites in PBMCs was 12 % when using calibration to standardize comet assay descriptors. Introduction of standard comet assay procedures was surprisingly unsuccessful as certain laboratories experienced technical problems using unaccustomed assay conditions. This problem was alleviated by using flexible assay standard conditions rather than a standard protocol in a ring-trial by the hCOMET group. The approach reduced technical problems, but the inter-laboratory variation in Fpg-sensitive sites was not reduced. The ring-trials have not pinpointed specific assay steps as major determinants of the variation in DNA damage levels. It is likely that small differences in several steps cause inter-laboratory variation. Although this variation in reported DNA damage levels causes concern, ring-trials have also shown that the comet assay is a reliable tool in biomonitoring studies.
Collapse
|
12
|
Pfaar O, Bergmann K, Bonini S, Compalati E, Domis N, Blay F, Kam P, Devillier P, Durham SR, Ellis AK, Gherasim A, Haya L, Hohlfeld JM, Horak F, Iinuma T, Jacobs RL, Jacobi HH, Jutel M, Kaul S, Kelly S, Klimek L, Larché M, Lemell P, Mahler V, Nolte H, Okamoto Y, Patel P, Rabin RL, Rather C, Sager A, Salapatek AM, Sigsgaard T, Togias A, Willers C, Yang WH, Zieglmayer R, Zuberbier T, Zieglmayer P. Technical standards in allergen exposure chambers worldwide - an EAACI Task Force Report. Allergy 2021; 76:3589-3612. [PMID: 34028057 DOI: 10.1111/all.14957] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/23/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
Allergen exposure chambers (AECs) can be used for controlled exposure to allergenic and non-allergenic airborne particles in an enclosed environment, in order to (i) characterize the pathological features of respiratory diseases and (ii) contribute to and accelerate the clinical development of pharmacological treatments and allergen immunotherapy for allergic disease of the respiratory tract (such as allergic rhinitis, allergic rhinoconjunctivitis, and allergic asthma). In the guidelines of the European Medicines Agency for the clinical development of products for allergen immunotherapy (AIT), the role of AECs in determining primary endpoints in dose-finding Phase II trials is emphasized. Although methodologically insulated from the variability of natural pollen exposure, chamber models remain confined to supporting secondary, rather than primary, endpoints in Phase III registration trials. The need for further validation in comparison with field exposure is clearly mandated. On this basis, the European Academy of Allergy and Clinical Immunology (EAACI) initiated a Task Force in 2015 charged to gain a better understanding of how AECs can generate knowledge about respiratory allergies and can contribute to the clinical development of treatments. Researchers working with AECs worldwide were asked to provide technical information in eight sections: (i) dimensions and structure of the AEC, (ii) AEC staff, (iii) airflow, air processing, and operating conditions, (iv) particle dispersal, (v) pollen/particle counting, (vi) safety and non-contamination measures, (vii) procedures for symptom assessments, (viii) tested allergens/substances and validation procedures. On this basis, a minimal set of technical requirements for AECs applied to the field of allergology is proposed.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Karl‐Christian Bergmann
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Department of Dermatology and Allergy Allergy Centre Charité Berlin Germany
| | - Sergio Bonini
- Institute of Translational Medicine Italian National Research Council Rome Italy
| | | | - Nathalie Domis
- ALYATEC Environmental Exposure Chamber Strasbourg France
| | - Frédéric Blay
- ALYATEC Environmental Exposure Chamber Strasbourg France
- Chest Diseases Department Strasbourg University Hospital Strasbourg France
| | | | - Philippe Devillier
- Department of Airway Diseases Pharmacology Research Laboratory‐VIM Suresnes, Exhalomics Platform, Hôpital Foch University Paris‐Saclay Suresnes France
| | | | - Anne K. Ellis
- Departments of Medicine and Biomedical & Molecular Sciences Queen's University Kingston ON Canada
- Allergy Research Unit Kingston General Health Research Institute Kingston ON Canada
| | - Alina Gherasim
- ALYATEC Environmental Exposure Chamber Strasbourg France
| | | | - Jens M. Hohlfeld
- Fraunhofer Institute for Toxicology and Experimental Medicine and Department of Respiratory Medicine Hannover Medical School Member of the German Center for Lung Research Hannover Germany
| | | | | | | | | | - Marek Jutel
- Department of Clinical Immunology Wroclaw Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wrocław Poland
| | | | | | - Ludger Klimek
- Center for Rhinology and Allergology Wiesbaden Germany
- Allergy Center Rhineland‐Palatinate Mainz University Medical Center Mainz Germany
| | - Mark Larché
- Divisions of Clinical Immunology & Allergy, and Respirology Department of Medicine and Firestone Institute for Respiratory Health McMaster University Hamilton ON Canada
| | | | | | | | | | - Piyush Patel
- Cliantha Research Limited Mississauga ON Canada
- Providence Therapeutics Toronto ON Canada
| | - Ronald L. Rabin
- Center for Biologics Evaluation and Research US Food and Drug Administration Silver Spring MD USA
| | | | | | | | - Torben Sigsgaard
- Department of Public Health, Section for Environment Occupation and Health Danish Ramazzini Centre Aarhus University Aarhus Denmark
| | - Alkis Togias
- Division of Allergy, Immunology, and Transplantation (DAIT) National Institute of Allergy and Infectious Diseases NIH Bethesda MD USA
| | | | | | | | - Torsten Zuberbier
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Department of Dermatology and Allergy Allergy Centre Charité Berlin Germany
| | - Petra Zieglmayer
- Vienna Challenge Chamber Vienna Austria
- Karl Landsteiner University Krems Austria
| |
Collapse
|
13
|
Malakootian M, Mohammadi A, Nasiri A, Oliveri Conti G, Faraji M. Correlation between heavy metal concentration and oxidative potential of street dust. AIR QUALITY, ATMOSPHERE, & HEALTH 2021; 15:731-738. [PMID: 34868387 PMCID: PMC8627698 DOI: 10.1007/s11869-021-01130-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/10/2021] [Indexed: 05/30/2023]
Abstract
The current study aimed to consider oxidative potential (OP), its spatial distribution, and correlations with heavy metals (HMs) in street dust in Kerman city, Iran. The concentration of HMs in 35 street dust samples was detected by ICP-AES. The OP in samples was measured through dithiothreitol (DTT). The mean concentration of elements followed Zn > Cu > Pb > Cr > As > Cd. The OP value was found to be 7.17 ± 2.98 nmol/min. µg dust in the current study. A strong correlation was observed among the concentrations of As and Cr and OP values in dust samples. More values of OP were observed in the center and west of the Kerman city. According to results of the current study, it could be concluded that OP can be applied as metrics of pollution originated from different sources and human health effects. The amount of OP in the street dust in the Kerman city can be reduced through the use of clean fuels.
Collapse
Affiliation(s)
- Mohammad Malakootian
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Environmental Health Engineering, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Amir Mohammadi
- Department of Public Health, School of Nursing and Midwifery, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Alireza Nasiri
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Gea Oliveri Conti
- Department of Medical Sciences, Surgical and Advanced Technologies “G.F. Ingrassia,”, University of Catania, Catania, Italy
| | - Maryam Faraji
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Environmental Health Engineering, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Scholten RH, Essig YJ, Roursgaard M, Jensen A, Krais AM, Gren L, Dierschke K, Gudmundsson A, Wierzbicka A, Møller P. Inhalation of hydrogenated vegetable oil combustion exhaust and genotoxicity responses in humans. Arch Toxicol 2021; 95:3407-3416. [PMID: 34468814 DOI: 10.1007/s00204-021-03143-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
Biofuels from vegetable oils or animal fats are considered to be more sustainable than petroleum-derived diesel fuel. In this study, we have assessed the effect of hydrogenated vegetable oil (HVO) exhaust on levels of DNA damage in peripheral blood mononuclear cells (PBMCs) as primary outcome, and oxidative stress and inflammation as mediators of genotoxicity. In a randomized cross-over study, healthy humans were exposed to filtered air, inorganic salt particles, exhausts from combustion of HVO in engines with aftertreatment [i.e. emission with nitrogen oxides and low amounts of particulate matter less than 2.5 µm (approximately 1 µg/m3)], or without aftertreatment (i.e. emission with nitrogen oxides and 93 ± 13 µg/m3 of PM2.5). The subjects were exposed for 3 h and blood samples were collected before, within 1 h after the exposure and 24 h after. None of the exposures caused generation of DNA strand breaks and oxidatively damaged DNA, or affected gene expression of factors related to DNA repair (Ogg1), antioxidant defense (Hmox1) or pro-inflammatory cytokines (Ccl2, Il8 and Tnfa) in PBMCs. The results from this study indicate that short-term HVO exhaust exposure is not associated with genotoxic hazard in humans.
Collapse
Affiliation(s)
- Rebecca Harnung Scholten
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Copenhagen K, Denmark
| | - Yona J Essig
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Martin Roursgaard
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Copenhagen K, Denmark
| | - Annie Jensen
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Copenhagen K, Denmark
| | - Annette M Krais
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Louise Gren
- Ergonomics and Aerosol Technology, Department of Design Sciences, Lund University, Box 118, 22100, Lund, Sweden
| | - Katrin Dierschke
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Gudmundsson
- Ergonomics and Aerosol Technology, Department of Design Sciences, Lund University, Box 118, 22100, Lund, Sweden
| | - Aneta Wierzbicka
- Ergonomics and Aerosol Technology, Department of Design Sciences, Lund University, Box 118, 22100, Lund, Sweden
| | - Peter Møller
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Copenhagen K, Denmark.
| |
Collapse
|
15
|
Li X, Haberzettl P, Conklin DJ, Bhatnagar A, Rouchka EC, Zhang M, O’Toole TE. Exposure to Fine Particulate Matter Air Pollution Alters mRNA and miRNA Expression in Bone Marrow-Derived Endothelial Progenitor Cells from Mice. Genes (Basel) 2021; 12:1058. [PMID: 34356074 PMCID: PMC8307414 DOI: 10.3390/genes12071058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Exposure to fine particulate matter (PM2.5) air pollution is associated with quantitative deficits of circulating endothelial progenitor cells (EPCs) in humans. Related exposures of mice to concentrated ambient PM2.5 (CAP) likewise reduces levels of circulating EPCs and induces defects in their proliferation and angiogenic potential as well. These changes in EPC number or function are predictive of larger cardiovascular dysfunction. To identify global, PM2.5-dependent mRNA and miRNA expression changes that may contribute to these defects, we performed a transcriptomic analysis of cells isolated from exposed mice. Compared with control samples, we identified 122 upregulated genes and 44 downregulated genes in EPCs derived from CAP-exposed animals. Functions most impacted by these gene expression changes included regulation of cell movement, cell and tissue development, and cellular assembly and organization. With respect to miRNA changes, we found that 55 were upregulated while 53 were downregulated in EPCs from CAP-exposed mice. The top functions impacted by these miRNA changes included cell movement, cell death and survival, cellular development, and cell growth and proliferation. A subset of these mRNA and miRNA changes were confirmed by qRT-PCR, including some reciprocal relationships. These results suggest that PM2.5-induced changes in gene expression may contribute to EPC dysfunction and that such changes may contribute to the adverse cardiovascular outcomes of air pollution exposure.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA;
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA;
| | - Petra Haberzettl
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Daniel J. Conklin
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Eric C. Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA;
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY 40202, USA
| | - Mei Zhang
- Department of Medicine, University of Louisville Genomics Facility, Louisville, KY 40202, USA;
| | - Timothy E. O’Toole
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
16
|
Singh P, O'Toole TE, Conklin DJ, Hill BG, Haberzettl P. Endothelial progenitor cells as critical mediators of environmental air pollution-induced cardiovascular toxicity. Am J Physiol Heart Circ Physiol 2021; 320:H1440-H1455. [PMID: 33606580 PMCID: PMC8260385 DOI: 10.1152/ajpheart.00804.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 01/15/2023]
Abstract
Environmental air pollution exposure is a leading cause of death worldwide, and with increasing industrialization and urbanization, its disease burden is expected to rise even further. The majority of air pollution exposure-associated deaths are linked to cardiovascular disease (CVD). Although ample research demonstrates a strong correlation between air pollution exposure and CVD risk, the mechanisms by which inhalation of polluted air affects cardiovascular health are not completely understood. Inhalation of environmental air pollution has been associated with endothelial dysfunction, which suggests that air pollution exposure impacts CVD health by inducing endothelial injury. Interestingly, recent studies demonstrate that air pollution exposure affects the number and function of endothelial progenitor cells (EPCs), subpopulations of bone marrow-derived proangiogenic cells that have been shown to play an essential role in maintaining cardiovascular health. In line with their beneficial function, chronically low levels of circulating EPCs and EPC dysfunction (e.g., in diabetic patients) have been associated with vascular dysfunction, poor cardiovascular health, and increases in the severity of cardiovascular outcomes. In contrast, treatments that improve EPC number and function (e.g., exercise) have been found to attenuate cardiovascular dysfunction. Considering the critical, nonredundant role of EPCs in maintaining vascular health, air pollution exposure-induced impairments in EPC number and function could lead to endothelial dysfunction, consequently increasing the risk for CVD. This review article covers novel aspects and new mechanistic insights of the adverse effects of air pollution exposure on cardiovascular health associated with changes in EPC number and function.
Collapse
Affiliation(s)
- Parul Singh
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Timothy E O'Toole
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel J Conklin
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Bradford G Hill
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Petra Haberzettl
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
17
|
Li A, Mei Y, Zhao M, Xu J, Seery S, Li R, Zhao J, Zhou Q, Ge X, Xu Q. The effect of ambient ozone on glucose-homoeostasis: A prospective study of non-diabetic older adults in Beijing. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 761:143308. [PMID: 33223186 DOI: 10.1016/j.scitotenv.2020.143308] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 10/07/2020] [Accepted: 10/19/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To investigate potential effects of short- and medium-term exposure to low levels of ozone (O3) on glucose-homeostasis in non-diabetic older adults. METHODS 166 non-diabetic, older participants in Beijing were deemed eligible to partake in this longitudinal population-based study. Observations were recorded on three separate occasions from November 2016 up until January 2018. Concentrations of outdoor O3 were monitored throughout the study period. Biomarkers indicative of glucose-homeostasis, including fasting blood glucose, insulin, HbAlc, glycated albumin percentage (glycated albumin/albumin), HOMA-IR and HOMA-B were measured at 3 sessions. A linear mixed effects model with random effects was adopted to quantify the effect of O3 across a comprehensive set of glucose-homeostasis markers. RESULTS Short-term O3 exposure positively associated with increased fasting blood glucose, insulin, HOMA-IR and HOMA-B. The effect on glucose occurred at 3-, 5-, 6- and 7-days, although the largest effect manifested on 6-days (5.6%, 95% CI: 1.4, 9.9). Significant associations with both insulin and HOMA-IR were observed on the 3- and 4-days. For HOMA-B, positive associations were identified from 3- to 7-days with estimates ranging from 40.0% (95% CI: 2.3, 91.5) to 83.1% (95% CI: 25.3, 167.5). Stratification suggests that women may be more susceptible to short-term O3 exposure. There does not appear to be a significant association between O3 and glucose-homeostasis in medium-term exposures. CONCLUSIONS In this study, we found that O3 exposure is at least partially associated with type II diabetes in older adults with no prior history of this condition. O3 therefore appears to be a potential risk factor, which is a particular concern when we consider the rise in global concentrations. Evidence also suggests that women may be more susceptible to short-term O3 exposure although we are not quite sure why. Future research may look to investigate this phenomenon further.
Collapse
Affiliation(s)
- Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Yayuan Mei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Samuel Seery
- School of Humanities and Social Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Runkui Li
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Resources and Environmental Information System, Institute of Geographic Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiaxin Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Quan Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Xiaoyu Ge
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
18
|
Abela AG, Fava S. Why is the Incidence of Type 1 Diabetes Increasing? Curr Diabetes Rev 2021; 17:e030521193110. [PMID: 33949935 DOI: 10.2174/1573399817666210503133747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/14/2021] [Accepted: 03/11/2021] [Indexed: 11/22/2022]
Abstract
Type 1 diabetes is a condition that can lead to serious long-term complications and can have significant psychological and quality of life implications. Its incidence is increasing in all parts of the world, but the reasons for this are incompletely understood. Genetic factors alone cannot explain such a rapid increase in incidence; therefore, environmental factors must be implicated. Lifestyle factors have been classically associated with type 2 diabetes. However, there are data implicating obesity and insulin resistance to type 1 diabetes as well (accelerator hypothesis). Cholesterol has also been shown to be correlated with the incidence of type 1 diabetes; this may be mediated by immunomodulatory effects of cholesterol. There is considerable interest in early life factors, including maternal diet, mode of delivery, infant feeding, childhood diet, microbial exposure (hygiene hypothesis), and use of anti-microbials in early childhood. Distance from the sea has recently been shown to be negatively correlated with the incidence of type 1 diabetes. This may contribute to the increasing incidence of type 1 diabetes since people are increasingly living closer to the sea. Postulated mediating mechanisms include hours of sunshine (and possibly vitamin D levels), mean temperature, dietary habits, and pollution. Ozone, polychlorinated biphenyls, phthalates, trichloroethylene, dioxin, heavy metals, bisphenol, nitrates/nitrites, and mercury are amongst the chemicals which may increase the risk of type 1 diabetes. Another area of research concerns the role of the skin and gut microbiome. The microbiome is affected by many of the factors mentioned above, including the mode of delivery, infant feeding, exposure to microbes, antibiotic use, and dietary habits. Research on the reasons why the incidence of type 1 diabetes is increasing not only sheds light on its pathogenesis but also offers insights into ways we can prevent type 1 diabetes.
Collapse
Affiliation(s)
- Alexia G Abela
- Department of Medicine, University of Malta & Mater Dei Hospital, Tal-Qroqq, Msida, Malta
| | - Stephen Fava
- Department of Medicine, University of Malta & Mater Dei Hospital, Tal-Qroqq, Msida, Malta
| |
Collapse
|
19
|
Shkirkova K, Lamorie-Foote K, Connor M, Patel A, Barisano G, Baertsch H, Liu Q, Morgan TE, Sioutas C, Mack WJ. Effects of ambient particulate matter on vascular tissue: a review. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2020; 23:319-350. [PMID: 32972334 PMCID: PMC7758078 DOI: 10.1080/10937404.2020.1822971] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Fine and ultra-fine particulate matter (PM) are major constituents of urban air pollution and recognized risk factors for cardiovascular diseases. This review examined the effects of PM exposure on vascular tissue. Specific mechanisms by which PM affects the vasculature include inflammation, oxidative stress, actions on vascular tone and vasomotor responses, as well as atherosclerotic plaque formation. Further, there appears to be a greater PM exposure effect on susceptible individuals with pre-existing cardiovascular conditions.
Collapse
Affiliation(s)
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California
- Keck School of Medicine, University of Southern California
| | - Michelle Connor
- Zilkha Neurogenetic Institute, University of Southern California
- Keck School of Medicine, University of Southern California
| | - Arati Patel
- Zilkha Neurogenetic Institute, University of Southern California
- Keck School of Medicine, University of Southern California
| | | | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California
- Keck School of Medicine, University of Southern California
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California
- Leonard Davis School of Gerontology, University of Southern California
| |
Collapse
|
20
|
Application of the comet assay in human biomonitoring: An hCOMET perspective. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2019; 783:108288. [PMID: 32192646 DOI: 10.1016/j.mrrev.2019.108288] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022]
Abstract
The comet assay is a well-accepted biomonitoring tool to examine the effect of dietary, lifestyle, environmental and occupational exposure on levels of DNA damage in human cells. With such a wide range of determinants for DNA damage levels, it becomes challenging to deal with confounding and certain factors are inter-related (e.g. poor nutritional intake may correlate with smoking status). This review describes the effect of intrinsic (i.e. sex, age, tobacco smoking, occupational exposure and obesity) and extrinsic (season, environmental exposures, diet, physical activity and alcohol consumption) factors on the level of DNA damage measured by the standard or enzyme-modified comet assay. Although each factor influences at least one comet assay endpoint, the collective evidence does not indicate single factors have a large impact. Thus, controlling for confounding may be necessary in a biomonitoring study, but none of the factors is strong enough to be regarded a priori as a confounder. Controlling for confounding in the comet assay requires a case-by-case approach. Inter-laboratory variation in levels of DNA damage and to some extent also reproducibility in biomonitoring studies are issues that have haunted the users of the comet assay for years. Procedures to collect specimens, and their storage, are not standardized. Likewise, statistical issues related to both sample-size calculation (before sampling of specimens) and statistical analysis of the results vary between studies. This review gives guidance to statistical analysis of the typically complex exposure, co-variate, and effect relationships in human biomonitoring studies.
Collapse
|