1
|
Cao H, Li B, Mu M, Li S, Chen H, Tao H, Wang W, Zou Y, Zhao Y, Liu Y, Tao X. Nicotine suppresses crystalline silica-induced astrocyte activation and neuronal death by inhibiting NF-κB in the mouse hippocampus. CNS Neurosci Ther 2024; 30:e14508. [PMID: 37864452 PMCID: PMC11017465 DOI: 10.1111/cns.14508] [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: 06/22/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/22/2023] Open
Abstract
AIMS Exposure to crystalline silica (CS) in occupational settings induces chronic inflammation in the respiratory system and, potentially, the brain. Some workers are frequently concurrently exposed to both CS and nicotine. Here, we explored the impact of nicotine on CS-induced neuroinflammation in the mouse hippocampus. METHODS In this study, we established double-exposed models of CS and nicotine in C57BL/6 mice. To assess depression-like behavior, experiments were conducted at 3, 6, and 9 weeks. Serum inflammatory factors were analyzed by ELISA. Hippocampus was collected for RNA sequencing analysis and examining the gene expression patterns linked to inflammation and cell death. Microglia and astrocyte activation and hippocampal neuronal death were assessed using immunohistochemistry and immunofluorescence staining. Western blotting was used to analyze the NF-κB expression level. RESULTS Mice exposed to CS for 3 weeks showed signs of depression. This was accompanied by elevated IL-6 in blood, destruction of the blood-brain barrier, and activation of astrocytes caused by an increased NF-κB expression in the CA1 area of the hippocampus. The elevated levels of astrocyte-derived Lcn2 and upregulated genes related to inflammation led to higher neuronal mortality. Moreover, nicotine mitigated the NF-κB expression, astrocyte activation, and neuronal death, thereby ameliorating the associated symptoms. CONCLUSION Silica exposure induces neuroinflammation and neuronal death in the mouse hippocampal CA1 region and depressive behavior. However, nicotine inhibits CS-induced neuroinflammation and neuronal apoptosis, alleviating depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Hangbing Cao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Bing Li
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Shanshan Li
- School of PharmacyBengbu Medical CollegeBengbuChina
| | - Haoming Chen
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Huihui Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Wenyang Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yuanjie Zou
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yehong Zhao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yang Liu
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| |
Collapse
|
2
|
Gui J, Liu J, Wang L, Yang X, Tian B, Luo H, Huang D, Han Z, Yang J, Ding R, Fang Z, Li X, Cheng L, Jiang L. Autophagy alleviates hippocampal neuroinflammation by inhibiting the NLRP3 inflammasome in a juvenile rat model exposed particulate matter. Toxicology 2024; 502:153730. [PMID: 38237716 DOI: 10.1016/j.tox.2024.153730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/01/2024] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Ambient fine particulate matter (PM) is a global public and environmental problem. PM is closely associated with several neurological diseases, which typically involve neuroinflammation. We investigated the impact of PM exposure on neuroinflammation using both in vivo (in a juvenile rat model with PM exposure concentrations of 1, 2, and 10 mg/kg for 28 days) and in vitro (in BV-2 and HT-22 cell models with PM concentrations of 50-200 μg/ml for 24 h). We observed that PM exposure induced the activation of the NLRP3 inflammasome, leading to the production of IL-1β and IL-18 in the rat hippocampus and BV-2 cells. Furthermore, inhibition of the NLRP3 inflammasome with MCC950 effectively reduced neuroinflammation and ameliorated hippocampal damage. In addition, autophagy activation was observed in the hippocampus of PM-exposed rats, and the promotion of autophagy by rapamycin (Rapa) effectively attenuated the NLRP3-mediated neuroinflammation induced by PM exposure. However, autophagic flow was blocked in BV-2 cells exposed to PM, and Rapa failed to ameliorate NLRP3 inflammasome activation. We found that autophagy was activated in HT-22 cells exposed to PM and that treatment with Rapa reduced the release of reactive oxygen species (ROS) and malondialdehyde (MDA), as well as cell apoptosis. In a subsequent coculture model of BV-2 and HT-22 cells, we observed the activation of the NLRP3 inflammasome in BV-2 cells when the HT-22 cells were exposed to PM, and this activation was alleviated when PM-exposed HT-22 cells were pretreated with Rapa. Overall, our study revealed that PM exposure triggered hippocampal neuroinflammation by activating the NLRP3 inflammasome. Notably, autophagy mitigated NLRP3 inflammasome activation, potentially by reducing neuronal ROS and apoptosis. This research emphasized the importance of reducing PM exposure and provided valuable insight into its neurotoxicity.
Collapse
Affiliation(s)
- Jianxiong Gui
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Jie Liu
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Lingman Wang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Xiaoyue Yang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Bing Tian
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Hanyu Luo
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Dishu Huang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Ziyao Han
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Jiaxin Yang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Ran Ding
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Zhixu Fang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Xue Li
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Li Cheng
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China.
| |
Collapse
|
3
|
Cao Z, Yang A, White AJ, Purdy F, Li C, Luo Z, D’Aloisio AA, Suarez L, Deming-Halverson S, Pinto JM, Chen JC, Werder EJ, Kaufman JD, Sandler DP, Chen H. Ambient Air Pollutants and Olfaction among Women 50-79 Years of Age from the Sister Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:87012. [PMID: 37594315 PMCID: PMC10436839 DOI: 10.1289/ehp12066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Poor olfaction is common in older adults and may have profound adverse implications on their health. However, little is known about the potential environmental contributors to poor olfaction. OBJECTIVE We investigated ambient fine particulate matter [PM ≤ 2.5 μ m in aerodynamic diameter (PM 2.5 )] and nitrogen dioxide (NO 2 ) in relation to poor olfaction in middle-aged to older women. METHODS The Sister Study is a nationwide cohort of 50,884 women in the United States with annual average air pollutant exposures estimated based on participants' residences from enrollment (2003-2009) through 2017. This analysis was limited to 3,345 women, 50-79 years of age as of January 2018, who completed the Brief Smell Identification Test (B-SIT) in 2018-2019. Poor olfaction was defined as a B-SIT score of ≤ 9 in the primary analysis. We conducted multivariable logistic regressions, accounting for covariates and study sampling design. RESULTS Overall, we found little evidence for associations of air pollutants with poor olfaction. The odds ratio (OR) and 95% confidence interval (CI) of poor olfaction for each interquartile range (IQR) increment of air pollutants in 2006 were 1.03 (95% CI: 0.91, 1.17) for PM 2.5 (per 3.3 μ g / m 3 ) and 1.08 (95% CI: 0.96, 1.22) for NO 2 (per 5.7 ppb ). Results were similar in the analyses using the most recent (2017) or the cumulative average (2006-2017) air pollutant exposure data. Secondary analyses suggested potential association in certain subgroups. The OR per IQR was 1.35 (95% CI: 1.11, 1.65) for PM 2.5 among younger participants (< 54.2 years of age) and 1.87 (95% CI: 1.29, 2.71) for NO 2 among current smokers. DISCUSSION This study did not find convincing evidence that air pollutants have lasting detrimental effects on the sense of smell of women 50-79 years of age. The subgroup analyses are exploratory, and the findings need independent confirmation. https://doi.org/10.1289/EHP12066.
Collapse
Affiliation(s)
- Zichun Cao
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Aiwen Yang
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Alexandra J. White
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Frank Purdy
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Chenxi Li
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Zhehui Luo
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Aimee A. D’Aloisio
- Social & Scientific Systems, DLH Holdings Corporation, Durham, North Carolina, USA
| | - Lourdes Suarez
- Social & Scientific Systems, DLH Holdings Corporation, Durham, North Carolina, USA
| | | | - Jayant M. Pinto
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Jiu-Chiuan Chen
- Department of Population and Public Health Sciences, University of Southern California (USC), Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Emily J. Werder
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Joel D. Kaufman
- Department of Environmental and Occupational Health Sciences, University of Washington School of Medicine (UW Medicine), Seattle, Washington, USA
- Department of Medicine, UW Medicine, Seattle, Washington, USA
- Department of Epidemiology, UW Medicine, Seattle, Washington, USA
| | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| |
Collapse
|
4
|
Liu F, Liu C, Liu Y, Wang J, Wang Y, Yan B. Neurotoxicity of the air-borne particles: From molecular events to human diseases. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131827. [PMID: 37315411 DOI: 10.1016/j.jhazmat.2023.131827] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Exposure to PM2.5 is associated with an increased incidence of CNS diseases in humans, as confirmed by numerous epidemiological studies. Animal models have demonstrated that PM2.5 exposure can damage brain tissue, neurodevelopmental issues and neurodegenerative diseases. Both animal and human cell models have identified oxidative stress and inflammation as the primary toxic effects of PM2.5 exposure. However, understanding how PM2.5 modulates neurotoxicity has proven challenging due to its complex and variable composition. This review aims to summarize the detrimental effects of inhaled PM2.5 on the CNS and the limited understanding of its underlying mechanism. It also highlights new frontiers in addressing these issues, such as modern laboratory and computational techniques and chemical reductionism tactics. By utilizing these approaches, we aim to fully elucidate the mechanism of PM2.5-induced neurotoxicity, treat associated diseases, and ultimately eliminate pollution.
Collapse
Affiliation(s)
- Fang Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong 250014, China
| | - Chunyan Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong 250014, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Jiahui Wang
- College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yibing Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong 250014, China.
| | - Bing Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| |
Collapse
|
5
|
Serafin P, Zaremba M, Sulejczak D, Kleczkowska P. Air Pollution: A Silent Key Driver of Dementia. Biomedicines 2023; 11:biomedicines11051477. [PMID: 37239148 DOI: 10.3390/biomedicines11051477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
In 2017, the Lancet Commission on Dementia Prevention, Intervention, and Care included air pollution in its list of potential risk factors for dementia; in 2018, the Lancet Commission on Pollution concluded that the evidence for a causal relationship between fine particulate matter (PM) and dementia is encouraging. However, few interventions exist to delay or prevent the onset of dementia. Air quality data are becoming increasingly available, and the science underlying the associated health effects is also evolving rapidly. Recent interest in this area has led to the publication of population-based cohort studies, but these studies have used different approaches to identify cases of dementia. The purpose of this article is to review recent evidence describing the association between exposure to air pollution and dementia with special emphasis on fine particulate matter of 2.5 microns or less. We also summarize here the proposed detailed mechanisms by which air pollutants reach the brain and activate the innate immune response. In addition, the article also provides a short overview of existing limitations in the treatment of dementia.
Collapse
Affiliation(s)
- Pawel Serafin
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
| | - Malgorzata Zaremba
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research (CBP), Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego Str., 02-106 Warsaw, Poland
| | - Patrycja Kleczkowska
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Maria Sklodowska-Curie, Medical Academy in Warsaw, Solidarnosci 12 Str., 03-411 Warsaw, Poland
| |
Collapse
|
6
|
Yi W, Ji Y, Gao H, Luo S, Pan R, Song J, He Y, Li Y, Wu Y, Yan S, Liang Y, Sun X, Jin X, Mei L, Cheng J, Su H. Effects of urban particulate matter on gut microbiome and partial schizophrenia-like symptoms in mice: Evidence from shotgun metagenomic and metabolomic profiling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 857:159305. [PMID: 36216056 DOI: 10.1016/j.scitotenv.2022.159305] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Epidemiological evidence reported that particulate matter (PM) was associated with increased schizophrenia (SCZ) risk. Disturbance of gut microbiome was involved in SCZ. However, it remains unclear whether PM induces SCZ-like symptoms and how gut microbiome regulates them. Therefore, a multi-omics animal experiment was conducted to verify how urban PM induces SCZ-like behavior and altered gut microbiota and metabolic pathways. METHODS Using a completely random design, mice were divided into three groups: PM group, control group and MK801 group, which received daily tracheal instillation of PM solution, sterile PBS solution and intraperitoneal injection of MK801 (establish SCZ model), respectively. After a 14-day intervention, feces were collected for multi-omics testing (shotgun metagenomic sequencing and untargeted metabolomic profiling), followed by open field test, tail suspension test, and passive avoidance test. Besides, fecal microbiome of PM group and control group were transplanted into "pseudo-sterile" mice, then behavioral tests were conducted. RESULTS Similar to MK801 group, mice in PM group showed SCZ-like symptoms, including increased spontaneous activity, excitability, anxiety and decreased learning and spatial memory. PM exposure significantly increased the relative abundance of Verrucomicrobia and decreased that of Fibrobacteres et al. The metabolism pathways of estrogen signaling (estriol, 16-glucuronide-estriol and 21-desoxycortisol) and choline metabolism (phosphocholine) were significantly altered by PM exposure. Verrucomicrobia was negatively correlated with the level of estriol, which was correlated with decreased learning and spatial memory. Fibrobacteres and Deinococcus-Thermus were positively correlated with the level of phosphocholine, which was correlated with increased spontaneous activity, excitability and anxiety. Fecal microbiome transplantation from PM group mice reproduced excitability and anxiety symptoms. CONCLUSIONS Exposure to PM may affect composition of gut microbiome and alterations of estrogen signaling pathway and choline metabolism pathway, which were associated with partial SCZ-like behaviors. But whether gut microbiome regulates these metabolic pathways and behaviors remains to be determined.
Collapse
Affiliation(s)
- Weizhuo Yi
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yifu Ji
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Hua Gao
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Shengyong Luo
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Rubing Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Jian Song
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yangyang He
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yuxuan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yudong Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Shuangshuang Yan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Yunfeng Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Xiaoni Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Xiaoyu Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Lu Mei
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Jian Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China
| | - Hong Su
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, China.
| |
Collapse
|
7
|
Zhang J, Yang Y, Al-Ahmady ZS, Du W, Duan J, Liao Z, Sun Q, Wei Z, Hua J. Maternal exposure to PM 2.5 induces cognitive impairment in offspring via cerebellar neuroinflammation and oxidative stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114425. [PMID: 38321695 DOI: 10.1016/j.ecoenv.2022.114425] [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: 09/14/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 09/02/2023]
Abstract
Available evidence suggest that exposure to PM2.5 during pregnancy is associated with reduced cognitive function in offspring. This study aimed to investigate the effects of maternal exposure to PM2.5 on offspring cognitive function and to elucidate the underlying mechanisms. In this work, pregnant C57BL/6 female mice were exposed to concentrated ambient PM2.5 or filtered air from day 0.5 (=vaginal plug) to day 15.5 in the Shanghai Meteorological and Environmental Animal Exposure System, and offspring cerebellar tissues were collected on embryonic day 15.5, as well as postnatal days 0, 10 and 42. The mean PM2.5 concentrations exposed to the pregnant mice were 73.06 ± 4.90 μg/m3 and 11.15 ± 2.71 μg/m3 in the concentrated ambient PM2.5 and filtered air chambers, respectively. Maternal concentrated PM2.5 exposure was negatively correlated with offspring spatial memory significantly as assessed by the Morris water maze. Compared with the filtered air group, PM2.5-exposed offspring mice had reduced cerebellar microglia. Both RNA and protein levels of IL-8 and TNF-α were elevated in the concentrated ambient PM2.5 group. PM2.5 exposure increased the level of 8-OHG in miRNA of microglia and Purkinje cells in 6-week-old offspring. The level of prostaglandin F2α (8-iso-PGF2Aα) in the cerebellum was increased at different growing stages of offspring after gestational exposure of PM2.5. These results suggested that maternal air pollution exposure might cause inflammatory damage and oxidative stress to the cerebellum, contributing to reduced cognitive performance in mice offspring.
Collapse
Affiliation(s)
- Jiajia Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yingying Yang
- Clinical Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zahraa S Al-Ahmady
- Pharmacology Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom; Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Wenchong Du
- NTU Psychology, School of Social Sciences, Nottingham Trent University, Nottingham NG1 1BU, United Kingdom
| | - Jinjin Duan
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medical, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore; Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Zhejiang 310053, China
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Jing Hua
- Shanghai Key Laboratory of Maternal Fetal Medicine, Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
8
|
Lee SH, Lin CY, Chen TF, Chou CCK, Chiu MJ, Tee BL, Liang HJ, Cheng TJ. Distinct brain lipid signatures in response to low-level PM 2.5 exposure in a 3xTg-Alzheimer's disease mouse inhalation model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156456. [PMID: 35660587 DOI: 10.1016/j.scitotenv.2022.156456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/11/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) poses a significant risk to human health. The molecular mechanisms underlying low-level PM2.5-induced neurotoxicity in the central nervous system remain unclear. In addition, changes in lipids in response to PM2.5 exposure have not yet been fully elucidated. In this study, 3xTg-Alzheimer's disease (AD) mice experienced continuous whole-body exposure to non-concentrated PM2.5 for three consecutive months, while control mice inhaled particulate matter-filtered air over the same time span. A liquid chromatography-mass spectrometry-based lipidomic platform was used to determine the distinct lipid profiles of various brain regions. The average PM2.5 concentration during the exposure was 11.38 μg/m3, which was close to the regulation limits of USA and Taiwan. The partial least squares discriminant analysis model showed distinct lipid profiles in the cortex, hippocampus, and olfactory bulb, but not the cerebellum, of mice in the exposure group. Increased levels of fatty acyls, glycerolipids, and sterol lipids, as well as the decreased levels of glycerophospholipids and sphingolipids in PM2.5-exposed mouse brains may be responsible for the increased energy demand, membrane conformation, neuronal loss, antioxidation, myelin function, and cellular signaling pathways associated with AD development. Our research suggests that subchronic exposure to low levels of PM2.5 may cause neurotoxicity by changing the lipid profiles in a susceptible model. Lipidomics is a powerful tool to study the early effects of PM2.5-induced AD toxicity.
Collapse
Affiliation(s)
- Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ching-Yu Lin
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Charles C-K Chou
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA, USA
| | - Hao-Jan Liang
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
9
|
Chen TF, Lee SH, Zheng WR, Hsu CC, Cho KH, Kuo LW, Chou CCK, Chiu MJ, Tee BL, Cheng TJ. White matter pathology in alzheimer's transgenic mice with chronic exposure to low-level ambient fine particulate matter. Part Fibre Toxicol 2022; 19:44. [PMID: 35768852 PMCID: PMC9245233 DOI: 10.1186/s12989-022-00485-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/29/2022] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Air pollution, especially fine particulate matter (PM), can cause brain damage, cognitive decline, and an increased risk of neurodegenerative disease, especially alzheimer's disease (AD). Typical pathological findings of amyloid and tau protein accumulation have been detected in the brain after exposure in animal studies. However, these observations were based on high levels of PM exposure, which were far from the WHO guidelines and those present in our environment. In addition, white matter involvement by air pollution has been less reported. Thus, this experiment was designed to simulate the true human world and to discuss the possible white matter pathology caused by air pollution. RESULTS 6 month-old female 3xTg-AD mice were divided into exposure and control groups and housed in the Taipei Air Pollutant Exposure System (TAPES) for 5 months. The mice were subjected to the Morris water maze test after exposure and were then sacrificed with brain dissection for further analyses. The mean mass concentration of PM2.5 during the exposure period was 13.85 μg/m3. After exposure, there was no difference in spatial learning function between the two groups, but there was significant decay of memory in the exposure group. Significantly decreased total brain volume and more neuronal death in the cerebral and entorhinal cortex and demyelination of the corpus callosum were noted by histopathological staining after exposure. However, there was no difference in the accumulation of amyloid or tau on immunohistochemistry staining. For the protein analysis, amyloid was detected at significantly higher levels in the cerebral cortex, with lower expression of myelin basic protein in the white matter. A diffuse tensor image study also revealed insults in multiple white matter tracts, including the optic tract. CONCLUSIONS In conclusion, this pilot study showed that even chronic exposure to low PM2.5 concentrations still caused brain damage, such as gross brain atrophy, cortical neuron damage, and multiple white matter tract damage. Typical amyloid cascade pathology did not appear prominently in the vulnerable brain region after exposure. These findings imply that multiple pathogenic pathways induce brain injury by air pollution, and the optic nerve may be another direct invasion route in addition to olfactory nerve.
Collapse
Affiliation(s)
- Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Wan-Ru Zheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Ching-Chou Hsu
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Kuan-Hung Cho
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Charles C-K Chou
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA, USA
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan.
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Neuroinflammation in Low-Level PM2.5-Exposed Rats Illustrated by PET via an Improved Automated Produced [18F]FEPPA: A Feasibility Study. Mol Imaging 2022; 2022:1076444. [PMID: 35903248 PMCID: PMC9328187 DOI: 10.1155/2022/1076444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 05/18/2022] [Indexed: 11/26/2022] Open
Abstract
Background [18F]FEPPA is a potent TSPO imaging agent that has been found to be a potential tracer for imaging neuroinflammation. In order to fulfill the demand of this tracer for preclinical and clinical studies, we have developed a one-pot automated synthesis with simplified HPLC purification of this tracer, which was then used for PET imaging of neuroinflammation in fine particulate matter- (PM2.5-) exposed rats. Results Using this automated synthesis method, the RCY of the [18F]FEPPA was 38 ± 4% (n = 17, EOB) in a synthesis time of 83 ± 8 min from EOB. The radiochemical purity and molar activities were greater than 99% and 209 ± 138 GBq/μmol (EOS, n = 15), respectively. The quality of the [18F]FEPPA synthesized by this method met the U.S. Pharmacopoeia (USP) criteria. The stability test showed that the [18F]FEPPA was stable at 21 ± 2°C for up to 4 hr after the end of synthesis (EOS). Moreover, microPET imaging showed that increased tracer activity of [18F]FEPPA in the brain of PM2.5-exposed rats (n = 6) were higher than that of normal controls (n = 6) and regional-specific. Conclusions Using the improved semipreparative HPLC purification, [18F]FEPPA has been produced in high quantity, high quality, and high reproducibility and, for the first time, used for PET imaging the effects of PM2.5 in the rat brain. It is ready to be used for imaging inflammation in various clinical or preclinical studies, especially for nearby PET centers without cyclotrons.
Collapse
|
11
|
Ma YH, Chen HS, Liu C, Feng QS, Feng L, Zhang YR, Hu H, Dong Q, Tan L, Kan HD, Zhang C, Suckling J, Zeng Y, Chen RJ, Yu JT. Association of Long-term Exposure to Ambient Air Pollution With Cognitive Decline and Alzheimer's Disease-Related Amyloidosis. Biol Psychiatry 2022; 93:780-789. [PMID: 35953319 DOI: 10.1016/j.biopsych.2022.05.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Air pollution induces neurotoxic reactions and may exert adverse effects on cognitive health. We aimed to investigate whether air pollutants accelerate cognitive decline and affect neurobiological signatures of Alzheimer's disease (AD). METHODS We used a population-based cohort from the Chinese Longitudinal Healthy Longevity Survey with 31,573 participants and a 10-year follow-up (5878 cognitively unimpaired individuals in Chinese Longitudinal Healthy Longevity Survey followed for 5.95 ± 2.87 years), and biomarker-based data from the Chinese Alzheimer's Biomarker and Lifestyle study including 1131 participants who underwent cerebrospinal fluid measurements of AD-related amyloid-β (Aβ) and tau proteins. Cognitive impairment was determined by education-corrected performance on the China-Modified Mini-Mental State Examination. Annual exposures to fine particulate matter (PM2.5), ground-level ozone (O3), and nitrogen dioxide (NO2) were estimated at areas of residence. Exposures were aggregated as 2-year averages preceding enrollments using Cox proportional hazards or linear models. RESULTS Long-term exposure to PM2.5 (per 20 μg/m3) increased the risk of cognitive impairment (hazard ratio, 1.100; 95% CI: 1.026-1.180), and similar associations were observed from separate cross-sectional analyses. Exposures to O3 and NO2 yielded elevated risk but with nonsignificant estimates. Individuals exposed to high PM2.5 manifested increased amyloid burdens as reflected by cerebrospinal fluid-AD biomarkers. Moreover, PM2.5 exposure-associated decline in global cognition was partly explained by amyloid pathology as measured by cerebrospinal fluid-Aβ42/Aβ40, P-tau/Aβ42, and T-tau/Aβ42, with mediation proportions ranging from 16.95% to 21.64%. CONCLUSIONS Long-term exposure to PM2.5 contributed to the development of cognitive decline, which may be partly explained by brain amyloid accumulation indicative of increased AD risk.
Collapse
Affiliation(s)
- Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China; Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hua-Shuai Chen
- School of Business, Xiangtan University, Xiangtan, Hunan, China
| | - Cong Liu
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education and Key Laboratory of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| | - Qiu-Shi Feng
- Department of Sociology, National University of Singapore, Singapore
| | - Lei Feng
- Department of Psychological Medicine and Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hai-Dong Kan
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education and Key Laboratory of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Diseases, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Yi Zeng
- Center for the Study of Aging and Human Development, Medical School of Duke University, Center for Healthy Aging and Development Studies, National School of Development, Raissun Institute for Advanced Studies, Peking University, Beijing, China
| | - Ren-Jie Chen
- School of Public Health, Key Laboratory of Public Health Safety of the Ministry of Education and Key Laboratory of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Ji X, Liu R, Guo J, Li Y, Cheng W, Pang Y, Zheng Y, Zhang R, Tang J. Olfactory bulb microglia activation mediated neuronal death in real-ambient particulate matter exposure mice with depression-like behaviors. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153456. [PMID: 35093369 DOI: 10.1016/j.scitotenv.2022.153456] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/03/2022] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
Growing evidence has indicated that air pollution is associated with depression, and damage of olfactory bulb (OB) is regarded as an early marker for depression. However, the toxicity of fine particulate matter (PM2.5) on OB and underlying mechanisms remains to be elucidated. In our study, a real-ambient PM2.5 exposure system was applied to explore the effects of PM2.5 on OB in C57BL/6 mice for 4 or 8 weeks. After 8 weeks exposure, the mice emerged potential depressive-like responses with reduction and disorder of cells in olfactory bulb tissues. Apoptosis and ultra-microstructure analysis indicated that the real-ambient PM2.5 exposure caused the neuronal death of OB. The immunofluorescence observation and KEGG pathway analysis revealed the real-ambient PM2.5 exposure induced microglia activation along with tumor necrosis factor α (TNFα)-mediated signaling enriched in OB of mice with depression-like behaviors. Moreover, results from ex vivo biosensor assay exhibited that PM2.5 might trigger systemic inflammation with increased levels of various proinflammatory factors to activate microglia. Further in vitro co-culture model identified that the PM2.5 evoked microglia cells activation with TNFα secretion and induced neuronal cells apoptosis via classical caspase3 signaling. Our findings provide new insights that PM2.5 induced microglia activation characterized by the release of TNFα to cause neurotoxicity either by direct action or by circulatory inflammation, resulting in OB damage, which may play a critical role in early diagnosis and pathogenic mechanisms for PM2.5 to cause depression.
Collapse
Affiliation(s)
- Xiaoya Ji
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Rui Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jiajun Guo
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yanting Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wenting Cheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yaxian Pang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jinglong Tang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
13
|
Kim YT, Kim W, Bae MJ, Choi JE, Kim MJ, Oh SS, Park KS, Park S, Lee SK, Koh SB, Kim C. The effect of polycyclic aromatic hydrocarbons on changes in the brain structure of firefighters: An analysis using data from the Firefighters Research on Enhancement of Safety & Health study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 816:151655. [PMID: 34785224 DOI: 10.1016/j.scitotenv.2021.151655] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/17/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are formed during incomplete combustion of organic matter, and firefighters are highly exposed to these toxic compounds at fire sites. Exposure to PAHs can cause cognitive decline and neurodegeneration; however, to date, few studies have examined the potential effects of PAH exposure on structural changes in the brain. We aimed to investigate the association between the four types of PAH metabolites and the corresponding changes in neuroimaging markers based on smoking status and hypertension in male firefighters. For this, we utilized the 2-year follow-up data of 301 Korean male firefighters aged over 40 years. The concentrations of four PAH metabolites in urine were measured. Subcortical volume and cortical thickness were estimated using 3 T magnetic resonance imaging of the brain. A generalized linear model was used to investigate the effects of PAHs on changes in the subcortical volume and cortical thickness. We found an association between 1-hydroxyphenathrene (1-OHPHE) and 2-hydroxyfluorene (2-OHF) and changes in several brain regions in all the study participants. Individuals who had never smoked showed significantly thinner frontal (p < 0.001), parietal (p < 0.001), temporal (p < 0.001), and cingulate lobes (p < 0.001) with 1% increase each in the urinary concentration of 1-OHPHE. Hypertension interacted with the concentration of 1-OHPHE to reduce the volume of gray matter and cause cortical thinning in the frontal, parietal, and temporal lobes. Exposure to PAHs may reduce cortical thickness and subcortical volume, which are definitive markers of neurodegeneration. Notably, hypertension can accelerate the degenerative effects of PAHs.
Collapse
Affiliation(s)
- Yun Tae Kim
- Department of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Woojin Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mun-Joo Bae
- Department of Occupational and Environmental Health, Yonsei University Graduate School of Public Health, Seoul, Republic of Korea
| | - Jee Eun Choi
- Department of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Mi-Ji Kim
- Department of Preventive Medicine and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Sung Soo Oh
- Department of Occupational and Environmental Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Ki Soo Park
- Department of Preventive Medicine and Institute of Health Science, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Sungha Park
- Division of Cardiology, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Koo Lee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Baek Koh
- Department of Preventive Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Changsoo Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Human Complexity and Systems Science, Yonsei University, Incheon, Republic of Korea.
| |
Collapse
|
14
|
Chang JH, Lee YL, Laiman V, Han CL, Jheng YT, Lee KY, Yeh CT, Kuo HP, Chung KF, Heriyanto DS, Hsiao TC, Wu SM, Ho SC, Chuang KJ, Chuang HC. Air pollution-regulated E-cadherin mediates contact inhibition of proliferation via the hippo signaling pathways in emphysema. Chem Biol Interact 2022; 351:109763. [PMID: 34852269 DOI: 10.1016/j.cbi.2021.109763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 01/07/2023]
Abstract
Air pollution has been linked to emphysema in chronic obstruction pulmonary disease (COPD). However, the underlying mechanisms in the development of emphysema due to air pollution remain unclear. The objective of this study was to investigate the role of components of the Hippo signaling pathway for E-cadherin-mediated contact inhibition of proliferation in the lungs after air pollution exposure. E-Cadherin-mediated contact inhibition of proliferation via the Hippo signaling pathway was investigated in Sprague-Dawley (SD) rats whole-body exposed to air pollution, and in alveolar epithelial A549 cells exposed to diesel exhaust particles (DEPs), E-cadherin-knockdown, and high-mobility group box 1 (HMGB1) treatment. Underlying epithelial differentiation, apoptosis, and senescence were also examined, and the interaction network among these proteins was examined. COPD lung sections were used to confirm the observations in rats. Expressions of HMGB1 and E-cadherin were negatively regulated in the lungs and A549 cells by air pollution, and this was confirmed by knockdown of E-cadherin and by treating A549 cells with HMGB1. Depletion of phosphorylated (p)-Yap occurred after exposure to air pollution and E-cadherin-knockdown, which resulted in decreases of SPC and T1α. Exposure to air pollution and E-cadherin-knockdown respectively downregulated p-Sirt1 and increased p53 levels in the lungs and in A549 cells. Moreover, the protein interaction network suggested that E-cadherin is a key activator in regulating Sirt1 and p53, as well as alveolar epithelial cell differentiation by SPC and T1α. Consistently, downregulation of E-cadherin, p-Yap, SPC, and T1α was observed in COPD alveolar regions with particulate matter (PM) deposition. In conclusion, our results indicated that E-cadherin-mediated cell-cell contact directly regulates the Hippo signaling pathway to control differentiation, cell proliferation, and senescence due to air pollution. Exposure to air pollution may initiate emphysema in COPD patients.
Collapse
Affiliation(s)
- Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Vincent Laiman
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Chia-Li Han
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Teng Jheng
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Han-Pin Kuo
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
15
|
Kang YJ, Tan H, Lee CY, Cho H. An Air Particulate Pollutant Induces Neuroinflammation and Neurodegeneration in Human Brain Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101251. [PMID: 34561961 PMCID: PMC8564420 DOI: 10.1002/advs.202101251] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/10/2021] [Indexed: 05/06/2023]
Abstract
Fine particulate matter (PM2.5), a major component among air pollutants, highlights as a global health concern. Several epidemiological studies show the correlation between chronical PM2.5 exposure and incidents of neurological disorders including Alzheimer's disease. However, the mechanisms have not been well understood, partly due to the lack of model systems that reflect the physiologically relevant innate immunity in human brains. Here, PM2.5-polluted human brain models (PMBs) are created in a 3D microfluidic platform reconstituting key aspects of human brain immunity under the PM2.5 exposure. PM2.5 penetration across a blood-brain barrier (BBB) model and accumulation in the brain tissue side of the model are first validated. Second, the PMB model shows that the BBB-penetrating PM2.5 initiates astrogliosis, resulting in slight neuronal loss and microglial infiltration. Third, it is demonstrated that the infiltrating microglia obtain M1 phenotype induced by interleukin-1β and interferon-γ from neurons and reactive astrocytes under the PM2.5 exposure. Finally, it is observed that additional proinflammatory mediators and nitric oxide released from the M1 microglia exacerbate neuronal damages, such as synaptic impairment, phosphoric tau accumulation, and neuronal death. This study suggests that PM2.5 can be a potential environmental risk factor for dementia mediated by the detrimental neuroinflammation.
Collapse
Affiliation(s)
- You Jung Kang
- Department Mechanical Engineering and Engineering ScienceDepartment of Biological SciencesCenter for Biomedical Engineering and ScienceUniversity of North Carolina at CharlotteCharlotteNC28223USA
- Institute of Quantum BiophysicsDepartment of BiophysicsSungkyunkwan UniversitySuwon‐siGyeonggi‐do16419ROK
| | - Hsih‐Yin Tan
- Institute for Health Innovation & TechnologyNational University of SingaporeSingapore117599Singapore
| | - Charles Y. Lee
- Department Mechanical Engineering and Engineering ScienceDepartment of Biological SciencesCenter for Biomedical Engineering and ScienceUniversity of North Carolina at CharlotteCharlotteNC28223USA
| | - Hansang Cho
- Institute of Quantum BiophysicsDepartment of BiophysicsSungkyunkwan UniversitySuwon‐siGyeonggi‐do16419ROK
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan UniversitySuwon‐siGyeonggi‐do16419ROK
| |
Collapse
|
16
|
Lee SH, Chen YH, Chien CC, Yan YH, Chen HC, Chuang HC, Hsieh HI, Cho KH, Kuo LW, Chou CCK, Chiu MJ, Tee BL, Chen TF, Cheng TJ. Three month inhalation exposure to low-level PM2.5 induced brain toxicity in an Alzheimer's disease mouse model. PLoS One 2021; 16:e0254587. [PMID: 34437570 PMCID: PMC8389369 DOI: 10.1371/journal.pone.0254587] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Although numerous epidemiological studies revealed an association between ambient fine particulate matter (PM2.5) exposure and Alzheimer's disease (AD), the PM2.5-induced neuron toxicity and associated mechanisms were not fully elucidated. The present study assessed brain toxicity in 6-month-old female triple-transgenic AD (3xTg-AD) mice following subchronic exposure to PM2.5 via an inhalation system. The treated mice were whole-bodily and continuously exposed to real-world PM2.5 for 3 months, while the control mice inhaled filtered air. Changes in cognitive and motor functions were evaluated using the Morris Water Maze and rotarod tests. Magnetic resonance imaging analysis was used to record gross brain volume alterations, and tissue staining with hematoxylin and eosin, Nissl, and immunohistochemistry methods were used to monitor pathological changes in microstructures after PM2.5 exposure. The levels of AD-related hallmarks and the oxidative stress biomarker malondialdehyde (MDA) were assessed using Western blot analysis and liquid chromatography-mass spectrometry, respectively. Our results showed that subchronic exposure to environmental levels of PM2.5 induced obvious neuronal loss in the cortex of exposed mice, but without significant impairment of cognitive and motor function. Increased levels of phosphorylated-tau and MDA were also observed in olfactory bulb or hippocampus after PM2.5 exposure, but no amyloid pathology was detected, as reported in previous studies. These results revealed that a relatively lower level of PM2.5 subchronic exposure from the environmental atmosphere still induced certain neurodegenerative changes in the brains of AD mice, especially in the olfactory bulb, entorhinal cortex and hippocampus, which is consistent with the nasal entry and spreading route for PM exposure. Systemic factors may also contribute to the neuronal toxicity. The effects of PM2.5 after a more prolonged exposure period are needed to establish a more comprehensive picture of the PM2.5-mediated development of AD.
Collapse
Affiliation(s)
- Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsuan Chen
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chu-Chun Chien
- Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Horng Yan
- Department of Endocrinology and Metabolism, Kuang Tien General Hospital, Taichung, Taiwan
- Department of Nutrition and Institute of Biomedical Nutrition, Hung Kuang University, Taichung, Taiwan
| | - Hsin-Chang Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-I Hsieh
- Department of Occupational Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Kuan-Hung Cho
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, California, United States of America
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Lai CH, Ho SC, Pan CH, Chen WL, Wang CC, Liang CW, Chien CY, Riediker M, Chuang KJ, Chuang HC. Chronic exposure to metal fume PM 2.5 on inflammation and stress hormone cortisol in shipyard workers: A repeat measurement study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 215:112144. [PMID: 33743405 DOI: 10.1016/j.ecoenv.2021.112144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 06/12/2023]
Abstract
Particulate matter with an aerodynamic diameter of ≤ 2.5 µm (PM2.5) has been linked to adverse health outcomes in welding workers. The objective of this study was to investigate associations of chronic exposure to metal fume PM2.5 in shipyard workers with health outcomes. A longitudinal study was conducted to determine the effects of metal fume PM2.5 on FeNO, urinary metals, urinary oxidative stress, inflammation, and stress hormones in workers. There were 20 office workers and 49 welding workers enrolled in this study who were followed-up for a second year. We observed that Fe, Zn, and Mn were abundant in PM2.5 to which welding workers were personally exposed, whereas PM2.5 to which office workers were personally exposed was dominated by Pb, Cu, and Zn. We observed in the first and/or second visits that urinary 8-iso-prostaglandin F2-α (PGF2α) and 8-hydroxy-2'-deoxy guanosine (8-OHdG) were significantly increased by exposure. An increase in urinary interleukin (IL)-6 and decreases in urinary serotonin and cortisol were observed in the first and/or second visits after exposure. PM2.5 was associated with decreases in urinary 8-OHdG and cortisol among workers. Next, we observed that urinary Ni, Co, and Fe had significantly increased among workers after a year of exposure. Urinary metals were associated with decreases in urinary 8-iso-PGF2α and cortisol among workers. Urinary Ni, Cu, and Fe levels were associated with an increase in urinary IL-6 and a decrease in urinary cortisol among workers. In conclusion, chronic exposure to metal fume PM2.5 was associated with inflammation and a cortisol deficiency in shipyard workers, which could associate with adrenal glands dysfunction.
Collapse
Affiliation(s)
- Ching-Huang Lai
- School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chih-Hong Pan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan; Institute of Labor, Occupational Safety and Health, Ministry of Labor, New Taipei City, Taiwan.
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan; Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan; School of Medicine, National Defense Medical Center, Taipei, Taiwan.
| | - Chung-Ching Wang
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan; Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan; School of Medicine, National Defense Medical Center, Taipei, Taiwan.
| | - Che-Wi Liang
- School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| | - Chi-Yu Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| | - Michael Riediker
- Swiss Centre for Occupational and Environmental Health, Winterthur, Switzerland.
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|