1
|
Sivakumar B, Kurian GA. PM 2.5 Induced Vascular and Myocardial Calcification Impairs Ischemia-reperfusion Tolerance via Mitochondrial Dysregulation. Cell Biochem Biophys 2025:10.1007/s12013-025-01758-7. [PMID: 40249523 DOI: 10.1007/s12013-025-01758-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2025] [Indexed: 04/19/2025]
Abstract
Cardiovascular diseases (CVD) are intricately linked to vascular dysfunction, with growing evidence implicating particulate matter (PM2.5) as a major factor. This study addresses the urgent need to understand how PM2.5 exposure influences cardiac vulnerability to ischemia-reperfusion (IR) injury by investigating the underlying mechanisms of vascular and myocardial alterations. The aim was to assess the progressive impact of PM2.5 exposure on vascular and myocardial function, mainly focusing on mitochondrial integrity and calcification processes. Adult Wistar female rats were subjected to PM2.5 at a concentration of 250 µg/m3 for 3 h daily over 1, 7, 14, and 21 days. Cardiac endurance to IR injury was assessed using the Langendorff perfusion method. Findings revealed that exposure for 7 days or more induced vascular calcification, upregulating calcification-related genes and causing calcium accumulation, while endothelial dysfunction and impaired vascular contractility manifested earlier. Myocardial calcification and hemodynamic impairments became evident after 14 days, correlating with progressive mitochondrial dysfunction in both vascular and cardiac tissues. By day 21, severe mitochondrial damage and elevated cardiac sensitivity to IR injury were observed, accompanied by increased metal deposition in the vasculature and myocardium. The study concludes that PM2.5 exposure drives a cascade of vascular and myocardial alterations, with vascular dysfunction preceding myocardial calcification. These findings emphasize the need for strategies to mitigate PM2.5 induced cardiovascular risks, particularly by targeting mitochondrial health and vascular integrity.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Cardiovascular Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Gino A Kurian
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur, Tamil Nadu, India.
| |
Collapse
|
2
|
Wang L, Wang B, Liao J, Zhang J, Su X, Yan J, Xu W, Lin J, Sun G, Wang L, Tang L. Cardiovascular Emergency Hospitalization Risks of PM 2.5 Transition Metals: A Time-Stratified Case-Crossover Study. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:402-413. [PMID: 40270527 PMCID: PMC12012663 DOI: 10.1021/envhealth.4c00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 04/25/2025]
Abstract
PM2.5 pollution poses significant health risks in urban areas, yet the specific cardiovascular impacts of its hazardous components, especially transition metals, remain insufficiently understood. This study evaluated the associations of PM2.5 components on acute myocardial infarction (AMI) and acute aortic dissections (AAD) emergency hospitalizations (n = 9985) using a time-stratified case-crossover between 2017 and 2023 in Xiamen, China. We collected comprehensive data on daily air pollutants, PM2.5 components (water-soluble ions, carbon components, metals, and other elements), and meteorological variables. Conditional logistic regressions were used to estimate odds ratios (OR) per the interquartile range (IQR) of exposures. Our finding reveals significant short-term associations of exposures to air pollutants and PM2.5 components with increased cardiovascular emergency hospitalizations. The strongest associations were observed between cumulative 3-day lagged (lag 0-3) PM2.5 transition metals including Mn [odds ratio, OR = 1.106 (95% CI: 1.032-1.186)], Fe [OR = 1.078, (95% CI: 1.015-1.145)], V [OR = 1.117 (95% CI: 1.024-1.219)], and Zn [OR = 1.08, (95% CI: 1.005-1.161)] exposure with AMI. These associations were stronger among older (age >65 years), male patients, and during colder seasons. Our study highlights the underexplored subacute cardiovascular risks of PM2.5 transition metals, underscoring the need to integrate them into urban air quality management to promote environmental sustainability.
Collapse
Affiliation(s)
- Lin Wang
- Key
Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Bin Wang
- Department
of Emergency, Xiamen Cardiovascular Hospital of Xiamen University,
School of Medicine, Xiamen University, Xiamen 361008, China
| | - Jiawen Liao
- Department
of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
of Americca
| | - Jieru Zhang
- Xiamen
Environmental Monitoring Station, Xiamen 361021, China
| | - Xin Su
- School
of Future Technology (SFT), China University
of Geosciences, Wuhan 430074, China
| | - Jinshan Yan
- Key
Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Xu
- Key
Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Jiyi Lin
- Department
of Emergency, Xiamen Cardiovascular Hospital of Xiamen University,
School of Medicine, Xiamen University, Xiamen 361008, China
| | - Guangfeng Sun
- Department
of Emergency, Xiamen Cardiovascular Hospital of Xiamen University,
School of Medicine, Xiamen University, Xiamen 361008, China
| | - Lunche Wang
- School
of Future Technology (SFT), China University
of Geosciences, Wuhan 430074, China
| | - Lina Tang
- Key
Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
3
|
Wāng Y, Wang C, Jiang Y, Wang T, Wu T, Tang M. Carbonaceous cores serve as surrogates for environmental particulate matter inducing vascular endothelial inflammation via inflammasome activation. JOURNAL OF HAZARDOUS MATERIALS 2025; 486:137011. [PMID: 39736255 DOI: 10.1016/j.jhazmat.2024.137011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Ambient particulate matter (PM) exposure is a known risk factor for cardiovascular diseases. Epidemiological studies have shown the association between PM exposure and vascular complications, including vasculitis, embolism, hypertension, stroke, and atherosclerosis. However, the exact mechanisms underlying its vascular toxicity, especially in relation to short-term exposures, remain incompletely understood. This study investigates the role of PM and its carbonaceous cores in driving vascular endothelial inflammation via inflammasome activation. We hypothesized that PM SRM1648a exposure induces vascular endothelial inflammation through oxidative stress and inflammasome activation. Short-term exposure to PM SRM1648a was assessed in BALB/c mice for systemic inflammation and oxidative stress biomarkers, alongside in vitro studies in HUVECs and EA.hy926 endothelial cells to elucidate inflammasome activation pathways. PM SRM1648a exposure significantly altered redox balance and cytokine profiles in mice and upregulated NLRP3/NLRC4 inflammasomes in vascular endothelial cells, leading to caspase-1/IL-1β activation. Intriguingly, pyroptosis was not the primary mode of cell death. In vitro studies demonstrated that antioxidants glutathione monoethyl ester effectively mitigated oxidative stress and inflammasome activation in endothelial cells. This study highlights the critical role of ROS-mediated inflammasome activation in vascular inflammation induced by PM SRM1648a, with carbon-based cores as key contributors.
Collapse
Affiliation(s)
- Yán Wāng
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Chunzhi Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yang Jiang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Wang
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui 231100, China
| | - Tianshu Wu
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Meng Tang
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
4
|
Wang AYL, Aviña AE, Liu YY, Chang YC, Kao HK. Transcription Factor Blimp-1: A Central Regulator of Oxidative Stress and Metabolic Reprogramming in Chronic Inflammatory Diseases. Antioxidants (Basel) 2025; 14:183. [PMID: 40002370 PMCID: PMC11851694 DOI: 10.3390/antiox14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
B-lymphocyte-induced maturation protein 1 (Blimp-1) is a transcription factor that, among other functions, modulates metabolism and helps to regulate antioxidant pathways, which is important in the context of chronic inflammatory diseases like diabetes, cardiovascular disease, and autoimmune disease. In immune cell function, Blimp-1 has a modulatory role in the orchestration of metabolic reprogramming and as a promoter of anti-inflammatory cytokines, including IL-10, responsible for modulating oxidative stress and immune homeostasis. Moreover, Blimp-1 also modulates key metabolic aspects, such as glycolysis and fatty acid oxidation, which regulate reactive oxygen species levels, as well as tissue protection. This review depicts Blimp-1 as an important regulator of antioxidant defenses and anti-inflammation and suggests that the protein could serve as a therapeutic target in chronic inflammatory and metabolic dysregulation conditions. The modulation of Blimp-1 in diseases such as diabetic coronary heart disease and atherosclerosis could alleviate oxidative stress, augment the protection of tissues, and improve disease outcomes. The therapeutic potential for the development of new treatments for these chronic conditions lies in the synergy between the regulation of Blimp-1 and antioxidant therapies, which are future directions that may be pursued. This review emphasizes Blimp-1's emerging importance as a novel regulator in the pathogenesis of inflammatory diseases, providing new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Ana Elena Aviña
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Yu Liu
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Huang-Kai Kao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
5
|
Wilczyński T, Zalejska-Fiolka J, Sapeta-Wieckowska S, Sarnat-Kucharczyk M, Rokicki W. In situ oxidative stress in patients with epiretinal membrane. Acta Biochim Pol 2024; 71:13581. [PMID: 39494364 PMCID: PMC11528693 DOI: 10.3389/abp.2024.13581] [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: 07/25/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
Context Oxidative stress is an important factor for vitreomacular interface disease development in a theoretical model. Purpose The aim of the study was to evaluate the correlation between oxidative stress in the human epiretinal membrane (ERM) and retinal morphological changes. Material and methods The study included patients scheduled for vitrectomy with epiretinal membrane removal. LogMAR best corrected visual acuity was assessed and optical coherence tomography was performed. Patients were divided into three groups: Type 1 - epiretinal membrane with premacular fibrosis; type 2 - epiretinal membrane with co-existing layer hole; and type 3 - ERM with co-existing full-thickness macular hole. During vitrectomy, epiretinal membranes were collected. Total oxidant status was determined by an automated colorimetric method in homogenates of epiretinal membrane. Statistical analysis The Mann-Whitney U test, Kruskal-Wallis test and Spearman linear correlation analysis were used. Statistical significance was set with a level of α = 0.05. Results Twenty-one Caucasian women (60%) and 14 men (40%) were included in the study. The average age of participants was 74.7 years (95% CI: 71.13-75.45). The mean best corrected visual acuity LogMAR value in the group was 0.8 (95% CI: 0.9-0.7). The mean ratio of total oxidant status to protein level in the collected samples was 0.161 (95% CI: 0.08-0.23) µmol/mg of protein. No correlation was found between total oxidant status and the degree of morphological retinal changes. Conclusion The study found no significant correlation between the level of oxidative stress in epiretinal membrane and retinal morphological changes.
Collapse
Affiliation(s)
- Tomasz Wilczyński
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
| | - Jolanta Zalejska-Fiolka
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Sabina Sapeta-Wieckowska
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
| | - Monika Sarnat-Kucharczyk
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Wojciech Rokicki
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
6
|
Li J, Yao Y, Shang B, Xie Y, Yin H, Song Y, Ma J. Ambient air pollution, low-grade inflammation, and lung function: Evidences from the UK Biobank. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116998. [PMID: 39244880 DOI: 10.1016/j.ecoenv.2024.116998] [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: 05/04/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/10/2024]
Abstract
The associations of ambient air pollution exposure and low-grade inflammation with lung function remain uncertain. In this study, 276,289 subjects were enrolled in the UK Biobank. Individual exposure to ambient air pollution (including nitrogen dioxide [NO2], nitrogen oxides [NOx]), and particulate matter [PM2.5, PM10, PMcoarse]) were estimated by using the land-use regression model. Forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1) were tested, and low-grade inflammation score (INFLA score) was calculated for each subject. In this cross-sectional study, the median concentrations of air pollution were 9.89 µg/m3 for PM2.5, 15.98 µg/m3 for PM10, 6.09 µg/m3 for PMcoarse, 25.60 µg/m3 for NO2, and 41.46 µg/m3 for NOx, respectively. We observed that PM2.5, PM10, PMcoarse, NO2, NOx was negatively associated with lung function. Besides, significant positive associations between PM exposure and low-grade inflammation were noted. Per interquartile range (IQR) increase in PM2.5, PM10, and PMcoarse was related to higher INFLA score, and the β (95 % CI) was 0.06 (0.03, 0.08), 0.03 (0.02, 0.05), and 0.03 (0.01, 0.04), respectively. Additionally, we found significant negative associations between INFLA scores and lung function. One-unit increase in INFLA score was linked with 12.41- and 11.31-ml decreases in FVC and FEV1, respectively. Compared with individuals with low air pollution exposure and low INFLA scores, participants with high air pollution and high INFLA scores had the lowest FVC and FEV1. Additionally, we observed that INFLA scores could modify the relationships of PM2.5, NO2, and NOx with FVC and FEV1 (Pinteraction <0.05). The negative impact of air pollutants on lung function was more pronounced in subjects with high INFLA scores in comparison to those with low INFLA scores. In conclusion, we demonstrated negative associations between ambient air pollution and lung function, and the observed associations were strengthened and modified by low-grade inflammation.
Collapse
Affiliation(s)
- Jia Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuxin Yao
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Binxing Shang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yujia Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Haoyu Yin
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuanchao Song
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
7
|
Mu C, Li Q, Niu Y, Hu T, Li Y, Wang T, Yu X, Lv Y, Tang H, Jiang J, Xu H, Zheng Y, Han W. Chronic diesel exhaust exposure induced pulmonary vascular remodeling a potential trajectory for traffic related pulmonary hypertension. Respir Res 2024; 25:348. [PMID: 39342206 PMCID: PMC11439202 DOI: 10.1186/s12931-024-02976-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND As one of the most common traffic-related pollutants, diesel exhaust (DE) confers high risk for cardiovascular and respiratory diseases. However, its impact on pulmonary vessels is still unclear. METHODS To explore the effects of DE exposure on pulmonary vascular remodeling, our study analyzed the number and volume of small pulmonary vessels in the diesel engine testers (the DET group) from Luoyang Diesel Engine Factory and the controls (the non-DET group) from the local water company, using spirometry and carbon content in airway macrophage (CCAM) in sputum. And then we constructed a rat model of chronic DE exposure, in which 12 rats were divided into the DE group (6 rats with 16-week DE exposure) and the control group (6 rats with 16-week clean air exposure). During right heart catheterization, right ventricular systolic pressure (RVSP) was assessed by manometry. Macrophage migration inhibitory factor (MIF) in lung tissues and bronchoalveolar lavage fluid (BALF) were measured by qRT-PCR and ELISA, respectively. Histopathological analysis for cardiovascular remodeling was also performed. RESULTS In DET cohort, the number and volume of small pulmonary vessels in CT were positively correlated with CCAM in sputum (P<0.05). Rat model revealed that chronic DE-exposed rats had elevated RVSP, along with increased wall thickness of pulmonary small vessels and right the ventricle. What's more, the MIF levels in BALF and lung tissues were higher in DE-exposed rats than the controls. CONCLUSION Apart from airway remodeling, DE also induces pulmonary vascular remodeling, which will lead to cardiopulmonary dysfunction.
Collapse
Affiliation(s)
- Chaohui Mu
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266071, China
| | - Qinghai Li
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266071, China
- Qingdao Key Lab for Common Diseases, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Yong Niu
- National Institute of Occupational Health and Posing Control, China CDC, Beijing, 100050, China
| | - Ting Hu
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266071, China
| | - Yanting Li
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Tao Wang
- Qingdao Key Lab for Common Diseases, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China
| | - Xinjuan Yu
- Qingdao Key Lab for Common Diseases, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China
| | - Yiqiao Lv
- Department of Pulmonary and Critical Care Medicine, Qingdao Hospital, Dalian Medical University, Dalian, 116000, China
| | - Huiling Tang
- Department of Pulmonary and Critical Care Medicine, Qingdao Hospital, Dalian Medical University, Dalian, 116000, China
| | - Jing Jiang
- Department of Ultrasound, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China
| | - Haibin Xu
- Department of Radiology, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, 266071, China.
| | - Wei Han
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266071, China.
- Qingdao Key Lab for Common Diseases, Qingdao Hospital, University of Rehabilitation and Health Sciences, Qingdao, 266071, China.
- School of Public Health, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
8
|
Offer S, Di Bucchianico S, Czech H, Pardo M, Pantzke J, Bisig C, Schneider E, Bauer S, Zimmermann EJ, Oeder S, Hartner E, Gröger T, Alsaleh R, Kersch C, Ziehm T, Hohaus T, Rüger CP, Schmitz-Spanke S, Schnelle-Kreis J, Sklorz M, Kiendler-Scharr A, Rudich Y, Zimmermann R. The chemical composition of secondary organic aerosols regulates transcriptomic and metabolomic signaling in an epithelial-endothelial in vitro coculture. Part Fibre Toxicol 2024; 21:38. [PMID: 39300536 DOI: 10.1186/s12989-024-00600-x] [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: 02/02/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The formation of secondary organic aerosols (SOA) by atmospheric oxidation reactions substantially contributes to the burden of fine particulate matter (PM2.5), which has been associated with adverse health effects (e.g., cardiovascular diseases). However, the molecular and cellular effects of atmospheric aging on aerosol toxicity have not been fully elucidated, especially in model systems that enable cell-to-cell signaling. METHODS In this study, we aimed to elucidate the complexity of atmospheric aerosol toxicology by exposing a coculture model system consisting of an alveolar (A549) and an endothelial (EA.hy926) cell line seeded in a 3D orientation at the air‒liquid interface for 4 h to model aerosols. Simulation of atmospheric aging was performed on volatile biogenic (β-pinene) or anthropogenic (naphthalene) precursors of SOA condensing on soot particles. The similar physical properties for both SOA, but distinct differences in chemical composition (e.g., aromatic compounds, oxidation state, unsaturated carbonyls) enabled to determine specifically induced toxic effects of SOA. RESULTS In A549 cells, exposure to naphthalene-derived SOA induced stress-related airway remodeling and an early type I immune response to a greater extent. Transcriptomic analysis of EA.hy926 cells not directly exposed to aerosol and integration with metabolome data indicated generalized systemic effects resulting from the activation of early response genes and the involvement of cardiovascular disease (CVD) -related pathways, such as the intracellular signal transduction pathway (PI3K/AKT) and pathways associated with endothelial dysfunction (iNOS; PDGF). Greater induction following anthropogenic SOA exposure might be causative for the observed secondary genotoxicity. CONCLUSION Our findings revealed that the specific effects of SOA on directly exposed epithelial cells are highly dependent on the chemical identity, whereas non directly exposed endothelial cells exhibit more generalized systemic effects with the activation of early stress response genes and the involvement of CVD-related pathways. However, a greater correlation was made between the exposure to the anthropogenic SOA compared to the biogenic SOA. In summary, our study highlights the importance of chemical aerosol composition and the use of cell systems with cell-to-cell interplay on toxicological outcomes.
Collapse
Affiliation(s)
- Svenja Offer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany.
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany.
| | - Hendryk Czech
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Jana Pantzke
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Christoph Bisig
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Eric Schneider
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Stefanie Bauer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elias J Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elena Hartner
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Thomas Gröger
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Rasha Alsaleh
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Christian Kersch
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Till Ziehm
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Thorsten Hohaus
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Christopher P Rüger
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Jürgen Schnelle-Kreis
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Martin Sklorz
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Astrid Kiendler-Scharr
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| |
Collapse
|
9
|
Zhang T, Lui KH, Ho SSH, Chen J, Chuang HC, Ho KF. Characterization of airborne endotoxin in personal exposure to fine particulate matter (PM 2.5) and bioreactivity for elderly residents in Hong Kong. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116530. [PMID: 38833976 DOI: 10.1016/j.ecoenv.2024.116530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
The heavy metals and bioreactivity properties of endotoxin in personal exposure to fine particulate matter (PM2.5) were characterized in the analysis. The average personal exposure concentrations to PM2.5 were ranged from 6.8 to 96.6 μg/m3. The mean personal PM2.5 concentrations in spring, summer, autumn, and winter were 32.1±15.8, 22.4±11.8, 35.3±11.9, and 50.2±19.9 μg/m3, respectively. There were 85 % of study targets exceeded the World Health Organization (WHO) PM2.5 threshold (24 hours). The mean endotoxin concentrations ranged from 1.086 ± 0.384-1.912 ± 0.419 EU/m3, with a geometric mean (GM) varied from 1.034 to 1.869. The concentration of iron (Fe) (0.008-1.16 μg/m3) was one of the most abundant transition metals in the samples that could affect endotoxin toxicity under Toll-Like Receptor 4 (TLR4) stimulation. In summer, the interleukin 6 (IL-6) levels showed statistically significant differences compared to other seasons. Spearman correlation analysis showed endotoxin concentrations were positively correlated with chromium (Cr) and nickel (Ni), implying possible roles as nutrients and further transport via adhering to the surface of fine inorganic particles. Mixed-effects model analysis demonstrated that Tumor necrosis factor-α (TNF-α) production was positively associated with endotoxin concentration and Cr as a combined exposure factor. The Cr contained the highest combined effect (0.205-0.262), suggesting that Cr can potentially exacerbate the effect of endotoxin on inflammation and oxidative stress. The findings will be useful for practical policies for mitigating air pollution to protect the public health of the citizens.
Collapse
Affiliation(s)
- Tianhang Zhang
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Hei Lui
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Steven Sai Hang Ho
- Division of Atmosphere Sciences, Desert Research Institute, Reno, NV 89512, United States; Hong Kong Premium Services and Research Laboratory, Cheung Sha Wan, Kowloon, Hong Kong, China
| | - Jiayao Chen
- School of Architecture, Planning and Environmental Policy, University College Dublin, Dublin, Ireland
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kin Fai Ho
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
10
|
Zhu A, Cao Y, Li C, Yu J, Liu M, Xu K, Ruan Y. Effects of major air pollutants on angina hospitalizations: a correlation study. BMC Public Health 2024; 24:1877. [PMID: 39004712 PMCID: PMC11247793 DOI: 10.1186/s12889-024-19380-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Angina is a crucial risk signal for cardiovascular disease. However, few studies have evaluated the effects of ambient air pollution exposure on angina. OBJECTIVE We aimed to explore the short-term effects of air pollution on hospitalization for angina and its lag effects. METHODS We collected data on air pollutant concentrations and angina hospitalizations from 2013 to 2020. Distributed lag nonlinear model (DLNM) was used to evaluate the short-term effects of air pollutants on angina hospitalization under different lag structures. Stratified analysis by sex, age and season was obtained. RESULTS A total of 39,110 cases of angina hospitalization were included in the study. The results showed a significant positive correlation between PM2.5, SO2, NO2, and CO and angina hospitalization. Their maximum harmful effects were observed at lag0-7 (RR = 1.042; 95% CI: 1.017, 1.068), lag0-3 (RR = 1.067; 95% CI: 1.005, 1.133), lag0-6 (RR = 1.078; 95% CI: 1.041, 1.117), and lag0-6 (RR = 1.244; 95% CI: 1.109, 1.397), respectively. PM10 did not have an overall risk effect on angina hospitalization, but it did have a risk effect on women and the elderly. O3 was significantly negatively correlated with angina hospitalization, with the most pronounced effect observed at lag0-6 (RR = 0.960; 95% CI: 0.940, 0.982). Stratified analysis results showed that women and the elderly were more susceptible to pollutants, and the adverse effects of pollutants were stronger in the cold season. CONCLUSION Short-term exposure to PM2.5, SO2, NO2, and CO increases the risk of hospitalization for angina.
Collapse
Affiliation(s)
- Anning Zhu
- School of Public Health, Lanzhou University, Lanzhou, 730000, PR China
| | - Yongqin Cao
- Gansu Provincial Center for Disease Control and Prevention, Lanzhou, 730000, PR China
| | - Chunlan Li
- Third People's Hospital of Gansu Province, Lanzhou, 730000, PR China
| | - Jingze Yu
- School of Public Health, Lanzhou University, Lanzhou, 730000, PR China
| | - Miaoxin Liu
- School of Public Health, Lanzhou University, Lanzhou, 730000, PR China
| | - Ke Xu
- School of Public Health, Lanzhou University, Lanzhou, 730000, PR China
| | - Ye Ruan
- School of Public Health, Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
11
|
Tiekwe JE, Ongbayokolak N, Dabou S, Natheu CK, Goka MS, Nya Biapa PC, Annesi-Maesano I, Telefo PB. Respiratory Symptoms and Changes of Oxidative Stress Markers among Motorbike Drivers Chronically Exposed to Fine and Ultrafine Air Particles: A Case Study of Douala and Dschang, Cameroon. J Clin Med 2024; 13:3816. [PMID: 38999382 PMCID: PMC11242172 DOI: 10.3390/jcm13133816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Recent studies revealed that the high production of reactive oxidative species due to exposure to fine or ultrafine particles are involved in many chronic respiratory disorders. However, the poor standard of clinical data in sub-Saharan countries makes the assessment of our knowledge on the health impacts of air pollution in urban cities very difficult. Objective: The aim of this study was to evaluate the distribution of respiratory disorders associated with exposure to fine and ultrafine air particles through the changes of some oxidative stress biomarkers among motorbike drivers from two cities of Cameroon. Methods: A cross-sectional survey using a standardized questionnaire was conducted in 2019 on 191 motorcycle drivers (MDs) working in Douala and Dschang. Then, the activities of superoxide dismutase (SOD) and the level of malondialdehyde (MDA) were measured using colorimetric methods. The data of participants, after being clustered in Microsoft Excel, were analyzed and statistically compared using SPSS 20 software. Results: The motorbike drivers recruited from both cities were from 21 to 40 years old, with a mean age of 29.93 (±0.82). The distribution of respiratory disorders, such as a runny nose, cold, dry cough, chest discomfort, and breathlessness, was significantly increased among MDs in Douala. According to the results of biological assays, SOD and MDA were significantly greater among the MDs recruited in Douala compared to those of Dschang. The change in these oxidative stress markers was significantly positively correlated with the mobilization of monocytes and negatively correlated with neutrophils, showing the onset and progression of subjacent inflammatory reactions, and it seemed to be significantly influenced by the location MDs lived in. Conclusions: Through this study, we have confirmed the evidence supporting that the onset and progression of oxidative stress is caused by the long-term exposure to fine or ultrafine air particles among working people living in urban cities. Further studies should be conducted to provide evidence for the cellular damage and dysfunction related to the chronic exposure to fine particulate matter (PM) in the air among working people in the metropolitan sub-Saharan Africa context.
Collapse
Affiliation(s)
- Joseph Eloge Tiekwe
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
- Division of Pharmacology and Toxicology, Faculty of Medicine, University of Leipzig, 04109 Leipzig, Germany
| | - Nadine Ongbayokolak
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Solange Dabou
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Cerge Kamhoua Natheu
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Marie Stéphanie Goka
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
| | | | - Isabella Annesi-Maesano
- Desbrest Institute of Epidemiology and Public Health, University Montpellier, INSERM, 34090 Montpellier, France
- Division of Respiratory Medicine, Allergology, and Thoracic Oncology, University Hospital of Montpellier, 34295 Montpellier, France
| | - Phélix Bruno Telefo
- Department of Biochemistry, University of Dschang, Dschang P.O. Box 67, Cameroon
| |
Collapse
|
12
|
Gu C, Sun Y, Mao M, Liu J, Li X, Zhang X. Mechanism of simulated lunar dust-induced lung injury in rats based on transcriptomics. Toxicol Res (Camb) 2024; 13:tfad108. [PMID: 38179001 PMCID: PMC10762671 DOI: 10.1093/toxres/tfad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 01/06/2024] Open
Abstract
Lunar dust particles are an environmental threat to lunar astronauts, and inhalation of lunar dust can cause lung damage. The current study explored the mechanism of lunar dust simulant (CLDS-i) inducing inflammatory pulmonary injury. Wistar rats were exposed to CLDS-i for 4 h/d and 7d/week for 4 weeks. Pathological results showed that a large number of inflammatory cells gathered and infiltrated in the lung tissues of the simulated lunar dust group, and the alveolar structures were destroyed. Transcriptome analysis confirmed that CLDS-i was mainly involved in the regulation of activation and differentiation of immune inflammatory cells, activated signaling pathways related to inflammatory diseases, and promoted the occurrence and development of inflammatory injury in the lung. Combined with metabolomics analysis, the results of joint analysis of omics were found that the genes Kmo, Kynu, Nos3, Arg1 and Adh7 were involved in the regulation of amino acid metabolism in rat lung tissues, and these genes might be the key targets for the treatment of amino acid metabolic diseases. In addition, the imbalance of amino acid metabolism might be related to the activation of nuclear factor kappaB (NF-κB) signaling pathway. The results of quantitative real-time polymerase chain reaction and Western blot further confirmed that CLDS-i may promote the occurrence and development of lung inflammation and lead to abnormal amino acid metabolism by activating the B cell activation factor (BAFF)/ B cell activation factor receptor (BAFFR)-mediated NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chen Gu
- College of Basic Medical Sciences, Shenyang Medical College, Huanghe North Street 146, Shenyang 110034, China
| | - Yan Sun
- School of Pharmacy, Shenyang Medical College, Huanghe North Street 146, Shenyang 110034, China
| | - Meiqi Mao
- College of Basic Medical Sciences, Shenyang Medical College, Huanghe North Street 146, Shenyang 110034, China
| | - Jinguo Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Nanta Street 114, Shenyang 110016, China
| | - Xiongyao Li
- Center for Lunar and Planetary Sciences, Institute of Geochemistry, Chinese Academy of Sciences, Lincheng West Road 99, Guiyang 550081, China
| | - Xiaoping Zhang
- State Key Laboratory of Lunar and Planetary Sciences, Macau University of Science and Technology, Weilong Road, Taipa, Macau 999078, China
| |
Collapse
|
13
|
Zhang Y, Jiang M, Xiong Y, Zhang L, Xiong A, Wang J, He X, Li G. Integrated analysis of ATAC-seq and RNA-seq unveils the role of ferroptosis in PM2.5-induced asthma exacerbation. Int Immunopharmacol 2023; 125:111209. [PMID: 37976599 DOI: 10.1016/j.intimp.2023.111209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/19/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND PM2.5 exposure increases asthma exacerbation risk and worsens airway inflammation and mucus secretion, but the underlying mechanisms, especially the epigenetic modification changes, are not fully understood. METHODS ATAC-seq was conducted in Beas-2B cells to explore the differential chromatin accessibilities before and after exposure to PM2.5. RNA-seq was applied to screen the differentially expressed genes (DEGs) as well. The integrated analysis of ATAC-seq and RNA-seq was performed. The key up-regulated genes in the ferroptosis signaling pathway were identified by combined analysis with the FerrDb database and then verified. Meanwhile, to access the role of PM2.5-induced ferroptosis in asthma mice, house dust mites (HDM) were employed to conduct an allergic asthma mice model, and the ferroptosis-specific inhibitor (Ferrostatin-1, Fer-1) was used. The H&E staining, PAS staining, airway hyperresponsiveness, and bronchoalveolar lavage fluid (BALF) cell counting were used to investigate the impact of PM2.5-induced ferroptosis in asthma mice. RESULTS A total of 4,921 regions with differential accessibility were identified, encompassing 4,031 unique genes. Among these, 250 regions exhibited increased accessibility while 4,671 regions displayed reduced accessibility. Through the integrated analysis of ATAC-seq and RNA-seq, ferroptosis was determined as the key enriched pathway based on up-regulated DEGs and increased chromatin accessibilities. Furthermore, the decreased cell viability, accelerated lipid peroxide and morphological changes in mitochondria observed upon PM2.5 exposure were rescued by Fer-1, which are indicative of ferroptosis. By overlapping with ferroptosis-related genes from the FerrDb database, FTH1 and FTL were identified as the prominent up-regulated genes with increased chromatin accessibility in ferroptosis pathway. In addition, ChIP-qPCR analysis indicated that histone modification like H3K4me3 and H3K27ac positively regulated FTH1 and FTL expression. Subsequently, in PM2.5-exposed asthmatic mice, inhibition of ferroptosis effectively attenuated airway inflammation and mucus secretion. CONCLUSION These findings shed light on the molecular mechanisms underlying PM2.5-induced asthma exacerbation, with epigenetic modifications playing a pivotal role. Furthermore, it suggests the therapeutic potential of targeting ferroptosis as an intervention strategy.
Collapse
Affiliation(s)
- Yi Zhang
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Manling Jiang
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu 610000, China
| | - Lei Zhang
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Anying Xiong
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Junyi Wang
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China
| | - Xiang He
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China.
| | - Guoping Li
- School of Medicine, Southwest Jiaotong University, Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu 610031, China.
| |
Collapse
|
14
|
Kim HJ, Yang J, Herath KHINM, Jeon YJ, Son YO, Kwon D, Kim HJ, Jee Y. Oral Administration of Sargassum horneri Suppresses Particulate Matter-Induced Oxidative DNA Damage in Alveolar Macrophages of Allergic Airway Inflammation: Relevance to PM-Mediated M1/M2 AM Polarization. Mol Nutr Food Res 2023; 67:e2300462. [PMID: 37986167 DOI: 10.1002/mnfr.202300462] [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: 07/04/2023] [Indexed: 11/22/2023]
Abstract
SCOPE Particulate matter (PM) can cause cellular oxidative damage and promote respiratory diseases. It has recently shown that Sargassum horneri ethanol extract (SHE) containing sterols and gallic acid reduces PM-induced oxidative stress in mice lung cells through ROS scavenging and metal chelating. In this study, the role of alveolar macrophages (AMs) is identified that are particularly susceptible to DNA damage due to PM-triggered oxidative stress in lungs of OVA-sensitized mice exposed to PM. METHODS AND RESULTS The study scrutinizes if PM exposure causes oxidative DNA damage to AMs differentially depending on their type of polarization. Further, SHE's potential is investigated in reducing oxidative DNA damage in polarized AMs and restoring AM polarization in PM-induced allergic airway inflammation. The study discovers that PM triggers prolonged oxidative stress to AMs, leading to lipid peroxidation in them and alveolar epithelial cells. Particularly, AMs are polarized to M2 phenotype (F4/80+ CD206+ ) with enhanced oxidative DNA damage when subject to PM-induced oxidative stress. However, SHE repairs oxidative DNA damage in M1- and M2-polarized AMs and reduces AMs polarization imbalance due to PM exposure. CONCLUSION These results suggest the possibility of SHE as beneficial foods against PM-induced allergic airway inflammation via suppression of AM dysfunction.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Food Bioengineering, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jiwon Yang
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | | | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Doyoung Kwon
- College of Pharmacy, Jeju National University, Jeju, 63243, Republic of Korea
- Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Hyun Jung Kim
- Department of Food Bioengineering, Jeju National University, Jeju, 63243, Republic of Korea
| | - Youngheun Jee
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea
- Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea
| |
Collapse
|
15
|
Hu L, Xu C, Tang X, Yu S, Wang L, Li Q, Zhou X. Fine particulate matter promotes airway inflammation and mucin production by activating endoplasmic reticulum stress and the IRE1α/NOD1/NF‑κB pathway. Int J Mol Med 2023; 52:96. [PMID: 37654182 PMCID: PMC10555484 DOI: 10.3892/ijmm.2023.5299] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
Fine particulate matter (PM2.5) is a type of small particle that is <2.5 µm in diameter that may cause airway inflammation. Thus, the present study aimed to explore the effects of PM2.5 on endoplasmic reticulum (ER) stress and airway inflammation in human airway epithelial cells. For this purpose, HBE135‑E6E7 airway epithelial cells were cultured and exposed to specific concentrations of PM2.5 for various periods of time, and cell viability was determined using a Cell Counting Kit‑8 assay. The results of the present study demonstrated that exposure to PM2.5 increased the mRNA and protein expression levels of interleukin (IL)‑6, tumor necrosis factor (TNF)‑α and mucin 5AC (MUC5AC). Moreover, the expression levels of ER stress‑related proteins, such as glucose‑regulated protein 78, CCAAT‑enhancer binding protein homologous protein, activating transcription factor 6, protein kinase R‑like ER kinase (PERK), phosphorylated (p‑)PERK, inositol‑requiring enzyme 1α (IRE1α) and p‑IRE1α, and nucleotide‑binding oligomerization domain 1 (NOD1) expression levels were increased following exposure to PM2.5. Transfection with IRE1α small interfering RNA (siRNA) led to the increased production of IL‑6, TNF‑α and MUC5AC. Moreover, the expression of NOD1 and the translocation of NF‑κB p65 were inhibited following transfection with IRE1α siRNA. In addition, the results of the present study demonstrated that transfection with NOD1 siRNA decreased the production of IL‑6, TNF‑α and MUC5AC, and decreased the translocation of NF‑κB p65. The expression levels of IL‑6, TNF‑α and MUC5AC were increased in the HBE135‑E6E7 cells following treatment with C12‑iE‑DAP, a NOD1 agonist. Moreover, treatment with C12‑iE‑DAP led to the activation of NF‑κB p65. Collectively, the results of the present study suggest that PM2.5 promotes airway inflammation and mucin production by activating ER stress in HBE135‑E6E7 airway epithelial cells, and that the IRE1α/NOD1/NF‑κB pathway may be involved in this process.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Chaoqun Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Emergency and Trauma College, Hainan Medical University, Haikou, Hainan 579199, P.R. China
| | - Xiang Tang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Shanjun Yu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Lijun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| |
Collapse
|
16
|
Lazaro CM, Victorio JA, Davel AP, Oliveira HCF. CETP expression ameliorates endothelial function in female mice through estrogen receptor-α and endothelial nitric oxide synthase pathway. Am J Physiol Heart Circ Physiol 2023; 325:H592-H600. [PMID: 37539470 DOI: 10.1152/ajpheart.00365.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Endothelial dysfunction is an early manifestation of atherosclerosis. The cholesteryl ester transfer protein (CETP) has been considered proatherogenic by reducing plasma HDL levels. However, CETP may exhibit cell- or tissue-specific effects. We have previously reported that male mice expressing the human CETP gene show impaired endothelium-mediated vascular relaxation associated with oxidative stress. Although sexual dimorphisms on the metabolic role of CETP have been proposed, possible sex differences in the vascular effects of CETP were not previously studied. Thus, here we investigated the endothelial function of female CETP transgenic mice as compared with nontransgenic controls (NTg). Aortas from CETP females presented preserved endothelium-dependent relaxation to acetylcholine and an endothelium-dependent reduction of phenylephrine-induced contraction. eNOS phosphorylation (Ser1177) and calcium-induced NO levels were enhanced, whereas reactive oxygen species (ROS) production and NOX2 and SOD2 expression were reduced in the CETP female aortas. Furthermore, CETP females exhibited increased aortic relaxation to 17β-estradiol (E2) and upregulation of heat shock protein 90 (HSP90) and caveolin-1, proteins that stabilize estrogen receptor (ER) in the caveolae. Indeed, CETP females showed an increased E2-induced relaxation in a manner sensitive to estrogen receptor-α (ERα) and HSP90 inhibitors methylpiperidinopyrazole (MPP) and geldanamycin, respectively. MPP also impaired the relaxation response to acetylcholine in CETP but not in NTg females. Altogether, the study indicates that CETP expression ameliorates the anticontractile endothelial effect and relaxation to E2 in females. This was associated with less ROS production, and increased eNOS-NO and E2-ERα pathways. These results highlight the need for considering the sex-specific effects of CETP on cardiovascular risk.NEW & NOTEWORTHY Here we demonstrated that CETP expression has a sex-specific impact on the endothelium function. Contrary to what was described for males, CETP-expressing females present preserved endothelium-dependent relaxation to acetylcholine and improved relaxation response to 17β-estradiol. This was associated with less ROS production, increased eNOS-derived NO, and increased expression of proteins that stabilize estrogen receptor-α (ERα), thus increasing E2-ERα signaling sensitivity. These results highlight the need for considering the sex-specific effects of CETP on cardiovascular risk.
Collapse
Affiliation(s)
- Carolina M Lazaro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Jamaira A Victorio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Ana Paula Davel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| |
Collapse
|
17
|
Xu G, Zhao Y, Tao Y, Xiong C, Lv M, Gao Q, Zhang F, An Z, Wu W. Lias overexpression alleviates pulmonary injury induced by fine particulate matter in mice. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:6585-6603. [PMID: 37341891 DOI: 10.1007/s10653-023-01651-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/07/2023] [Indexed: 06/22/2023]
Abstract
Oxidative stress and inflammation are mechanisms underlying toxicity induced by fine particulate matter (PM2.5). The antioxidant baseline of the human body modulates the intensity of oxidative stress in vivo. This present study aimed to evaluate the role of endogenous antioxidants in alleviating PM2.5-induced pulmonary injury using a novel mouse model (LiasH/H) with an endogenous antioxidant capacity of approximately 150% of its wild-type counterpart (Lias+/+). LiasH/H and wild-type (Lias+/+) mice were randomly divided into control and PM2.5 exposure groups (n = 10), respectively. Mice in the PM2.5 group and the control group were intratracheally instilled with PM2.5 suspension and saline, respectively, once a day for 7 consecutive days. The metal content, major pathological changes in the lung, and levels of oxidative stress and inflammation biomarkers were examined. The results showed that PM2.5 exposure induced oxidative stress in mice. Overexpression of the Lias gene significantly increased the antioxidant levels and decreased inflammatory responses induced by PM2.5. Further study found that LiasH/H mice exerted their antioxidant function by activating the ROS-p38MAPK-Nrf2 pathway. Therefore, the novel mouse model is useful for the elucidation of the mechanisms of pulmonary injury induced by PM2.5.
Collapse
Affiliation(s)
- Guangcui Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Yingzheng Zhao
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Yingjun Tao
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Cheng Xiong
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Mengdi Lv
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Qiyu Gao
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Fengquan Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China.
| |
Collapse
|
18
|
Bavarsad K, Farbood Y, Mard SA, Khoshnam SE, Dianat M, Jahangiri HM, Khorsandi LS, Goudarzi G, Sarkaki A. Effects of Gallic Acid on Memory Deficits and Electrophysiological Impairments Induced by Cerebral Ischemia/Reperfusion in Rats Following Exposure to Ambient Dust Storm. Neurochem Res 2023:10.1007/s11064-023-03953-5. [PMID: 37222948 DOI: 10.1007/s11064-023-03953-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/25/2023]
Abstract
We aimed to investigate the probable protective effects of gallic acid (GA) on cognitive deficits, hippocampal long term potentiation (LTP) impairments, and molecular changes induced by cerebral ischemia/reperfusion (I/R) in rats following exposure to ambient dust storm. After pretreatment with GA (100 mg/kg), or vehicle (Veh) (normal saline, 2 ml/kg) for ten days, and 60 minutes' exposure to dust storm including PM (PM, 2000-8000 g/m3) every day, 4-vessel occlusion (4VO) type of I/R was induced. Three days after I/R induction, we evaluated behavioral, electrophysiological, histopathological, molecular and brain tissue inflammatory cytokine changes. Our findings indicated that pretreatment with GA significantly reduced cognitive impairments caused by I/R (P < 0.05) and hippocampal LTP impairments caused by I/R after PM exposure (P < 0.001). Additionally, after exposure to PM, I/R significantly elevated the tumor necrosis factor α content (P < 0.01) and miR-124 level (P < 0.001) while pre-treatment with GA reduced the level of miR-124 (P < 0.001). Histopathological results also revealed that I/R and PM caused cell death in the hippocampus CA1 area (P < 0.001) and that GA decreased the rate of cell death (P < 0.001). Our findings show that GA can prevent brain inflammation, and thus cognitive and LTP deficits caused by I/R, PM exposure, or both.
Collapse
Affiliation(s)
- Kowsar Bavarsad
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
| | - Seyed Ali Mard
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
| | - Seyed Esmaeil Khoshnam
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
| | - Mahin Dianat
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
| | - Hamzeh Mirshekari Jahangiri
- Department of Physiology, Faculty of Medicine, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Laya Sadat Khorsandi
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
- Department of Anatomical Sciences, Cellular & Molecular Research Center, Medical Basic Sciences Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholamreza Goudarzi
- National Institute for Medical Research Development "NIMAD", Tehran, Iran
- Department of Environmental Health Engineering, Environmental Technology Research Center, Jundishapur University of Medical Science, Ahvaz, Iran
| | - Alireza Sarkaki
- National Institute for Medical Research Development "NIMAD", Tehran, Iran.
- Medicinal Plants Research Center, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medica Sciences, Ahvaz, Iran.
| |
Collapse
|
19
|
Ji SM, Choi JS, Lee JY, Kim S, Bae WY, Jang YW, Kim JE, Lee SH, Nam S, Jeong JW. Mild exposure to fine particulate matter promotes angiogenesis in non-small cell lung carcinoma. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121715. [PMID: 37120000 DOI: 10.1016/j.envpol.2023.121715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
Fine particulate matter (PM2.5) is associated with public health problems worldwide. Especially, PM2.5 induces epigenetic and microenvironmental changes in lung cancer. Angiogenesis is important for the development and growth of cancer and is mediated by angiogenic factors, including vascular endothelial growth factor. However, the effects of mild PM2.5 exposure on angiogenesis in lung cancer remain unclear. In this study, we examined angiogenic effects using relatively lower concentrations of PM2.5 than in other studies and found that PM2.5 increased angiogenic activities in both endothelial cells and non-small cell lung carcinoma cells. PM2.5 also promoted the growth and angiogenesis of lung cancer via the induction of hypoxia-inducible factor-1α (HIF-1α) in a xenograft mouse tumor model. Angiogenic factors, including vascular endothelial growth factor (VEGF), were highly expressed in lung cancer patients in countries with high PM2.5 levels in the atmosphere, and high expression of VEGF in lung cancer patients lowered the survival rate. Collectively, these results provide new insight into the mechanisms by which mild exposure to PM2.5 is involved in HIF-1α-mediated angiogenesis in lung cancer patients.
Collapse
Affiliation(s)
- Su Min Ji
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae-Sun Choi
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Clinical Research Institute, Kyung Hee University Medical Center, Seoul, 02447, Republic of Korea
| | - Ji Young Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sungyeon Kim
- Department of Genome Medicine and Science, AI Convergence Center for Medical Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, Republic of Korea
| | - Woom-Yee Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ye Won Jang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee Univeristy, Seoul, 02447, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seungyoon Nam
- Department of Genome Medicine and Science, AI Convergence Center for Medical Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
20
|
Sousa AS, Passos MP, Ruberti OM, Jarrete AP, Delbin MA. Evaluation of coronary function in female rats with severe type 1 diabetes: Effects of combined treatment with insulin and pyridoxamine. Microvasc Res 2023; 146:104474. [PMID: 36592817 DOI: 10.1016/j.mvr.2022.104474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/06/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND This study aimed to evaluate the coronary function, myocardium, and epicardial adipose tissue (EAT) in female rats with severe type 1 diabetes and the effects of combined treatment with insulin and pyridoxamine (AGEs inhibitor). METHODS Female Wistar rats were divided into groups: control (CTR, n = 13), type 1 diabetes (DM1, n = 12), type 1 diabetes treated with insulin (DM1 + INS, n = 11), and type 1 diabetes treated with insulin and pyridoxamine (DM1 + INS + PDX, n = 14). The vascular responsiveness was performed in the septal coronary artery and the protein expressions of AGE, RAGE, GPER, NF-kB was evaluated in the left ventricle (LV), as well as the reactive oxygen species (ROS) was measured in LV and in EAT. We analyzed plasma levels of glucose, estradiol, Nε-carboxymethylisine (CML), thiobarbituric acid reactive substances (TBARS), catalase (CAT), and superoxide dismutase (SOD). RESULTS The maximal responses to ACh were reduced in the DM1 compared with the CTR group, accompanied by an increase in circulating glucose, CML, and TBARS. Additionally, the expression of NF-kB in LV and generation of ROS in the presence of MnTMPyP (SOD mimetic) were increased in the DM1 group compared with CTR. Only the combined treatment was effective for fully re-establish ACh relaxation response, NF-kB protein expression, ROS generation, and increased SOD activity in the DM1 + INS + PDX group. CONCLUSION The reduction of the endothelium-dependent relaxation response in the septal coronary artery of female rats with severe type 1 diabetes was normalized with the combined treatment with insulin and pyridoxamine, associated with reduced inflammation and oxidative stress in the myocardium and increased circulating antioxidant activity.
Collapse
Affiliation(s)
- Andressa S Sousa
- Department of Structural and Functional Biology, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Matheus P Passos
- Department of Structural and Functional Biology, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Olivia M Ruberti
- Department of Structural and Functional Biology, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Aline P Jarrete
- Department of Structural and Functional Biology, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Maria A Delbin
- Department of Structural and Functional Biology, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
21
|
Feng S, Huang F, Zhang Y, Feng Y, Zhang Y, Cao Y, Wang X. The pathophysiological and molecular mechanisms of atmospheric PM 2.5 affecting cardiovascular health: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114444. [PMID: 38321663 DOI: 10.1016/j.ecoenv.2022.114444] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5, with aerodynamic diameter less than 2.5 µm) is a leading environmental risk factor for global cardiovascular health concern. OBJECTIVE To provide a roadmap for those new to this field, we reviewed the new insights into the pathophysiological and cellular/molecular mechanisms of PM2.5 responsible for cardiovascular health. MAIN FINDINGS PM2.5 is able to disrupt multiple physiological barriers integrity and translocate into the systemic circulation and get access to a range of secondary target organs. An ever-growing body of epidemiological and controlled exposure studies has evidenced a causal relationship between PM2.5 exposure and cardiovascular morbidity and mortality. A variety of cellular and molecular biology mechanisms responsible for the detrimental cardiovascular outcomes attributable to PM2.5 exposure have been described, including metabolic activation, oxidative stress, genotoxicity, inflammation, dysregulation of Ca2+ signaling, disturbance of autophagy, and induction of apoptosis, by which PM2.5 exposure impacts the functions and fates of multiple target cells in cardiovascular system or related organs and further alters a series of pathophysiological processes, such as cardiac autonomic nervous system imbalance, increasing blood pressure, metabolic disorder, accelerated atherosclerosis and plaque vulnerability, platelet aggregation and thrombosis, and disruption in cardiac structure and function, ultimately leading to cardiovascular events and death. Therein, oxidative stress and inflammation were suggested to play pivotal roles in those pathophysiological processes. CONCLUSION Those biology mechanisms have deepen insights into the etiology, course, prevention and treatment of this public health concern, although the underlying mechanisms have not yet been entirely clarified.
Collapse
Affiliation(s)
- Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China; Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China.
| | - Fangfang Huang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yuqi Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yashi Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Ying Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yunchang Cao
- The Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Xinming Wang
- Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| |
Collapse
|
22
|
Bae HR, Chandy M, Aguilera J, Smith EM, Nadeau KC, Wu JC, Paik DT. Adverse effects of air pollution-derived fine particulate matter on cardiovascular homeostasis and disease. Trends Cardiovasc Med 2022; 32:487-498. [PMID: 34619335 PMCID: PMC9063923 DOI: 10.1016/j.tcm.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022]
Abstract
Air pollution is a rapidly growing major health concern around the world. Atmospheric particulate matter that has a diameter of less than 2.5 µm (PM2.5) refers to an air pollutant composed of particles and chemical compounds that originate from various sources. While epidemiological studies have established the association between PM2.5 exposure and cardiovascular diseases, the precise cellular and molecular mechanisms by which PM2.5 promotes cardiovascular complications are yet to be fully elucidated. In this review, we summarize the various sources of PM2.5, its components, and the concentrations of ambient PM2.5 in various settings. We discuss the experimental findings to date that evaluate the potential adverse effects of PM2.5 on cardiovascular homeostasis and function, and the possible therapeutic options that may alleviate PM2.5-driven cardiovascular damage.
Collapse
Affiliation(s)
- Hye Ryeong Bae
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Juan Aguilera
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Eric M Smith
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
The macrophage senescence hypothesis: the role of poor heat shock response in pulmonary inflammation and endothelial dysfunction following chronic exposure to air pollution. Inflamm Res 2022; 71:1433-1448. [PMID: 36264363 DOI: 10.1007/s00011-022-01647-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/18/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Cardiovascular diseases (CVD) have been associated with high exposure to fine particulate air pollutants (PM2.5). Alveolar macrophages are the first defense against inhaled particles. As soon as they phagocytize the particles, they reach an inflammatory phenotype, which affects the surrounding cells and associates with CVD. Not coincidentally, CVD are marked by a depleted heat shock response (HSR), defined by a deficit in inducing 70-kDa heat shock protein (HSP70) expression during stressful conditions. HSP70 is a powerful anti-inflammatory chaperone, whose reduced levels trigger a pro-inflammatory milieu, cellular senescence, and a senescence-associated secretory phenotype (SASP). However, whether macrophage senescence is the main mechanism by which PM2.5 propagates low-grade inflammation remains unclear. OBJECTIVE AND DESIGN In this article, we review evidence supporting that chronic exposure to PM2.5 depletes HSR and determines the ability to solve the initial stress. RESULTS AND DISCUSSION When exposed to PM2.5, macrophages increase the production of reactive oxygen species, which activate nuclear factor-kappa B (NF-κB). NF-κB is naturally a pro-inflammatory factor that drives prostaglandin E2 (PGE2) synthesis and causes fever. PGE2 can be converted into prostaglandin A2, a powerful inducer of HSR. Therefore, when transiently activated, NF-κB can trigger the anti-inflammatory response through negative feedback, by inducing HSP70 expression. However, when chronically activated, NF-κB heads a set of pathways involved in mitochondrial dysfunction, endoplasmic reticulum stress, unfolded protein response, inflammasome activation, and apoptosis. During chronic exposure to PM2.5, cells cannot properly express sirtuin-1 or activate heat shock factor-1 (HSF-1), which delays the resolution phase of inflammation. Since alveolar macrophages are the first immune defense against PM2.5, we suppose that the pollutant impairs HSR and, consequently, induces cellular senescence. Accordingly, senescent macrophages change its secretory phenotype to a more inflammatory one, known as SASP. Finally, macrophages' SASP would propagate the systemic inflammation, leading to endothelial dysfunction and atherosclerosis.
Collapse
|
24
|
Peres EC, Victorio JA, Nunes-Souza V, Breithaupt-Faloppa AC, Rabelo LA, Tavares-de-Lima W, Davel AP, Rossoni LV. Simvastatin protects against intestinal ischemia/reperfusion-induced pulmonary artery dysfunction. Life Sci 2022; 306:120851. [PMID: 35926590 DOI: 10.1016/j.lfs.2022.120851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 10/16/2022]
Abstract
AIMS The lung is an important target organ damage in intestinal ischemia/reperfusion (II/R), but mechanisms involved in II/R-induced pulmonary artery (PA) dysfunction, as well as its treatment, are not clear. The present study aimed to investigate the mechanisms involved in the II/R-induced PA dysfunction and a possible protective role of acute simvastatin pretreatment. MAIN METHODS Male Wistar rats were subjected to occlusion of the superior mesenteric artery for 45 min followed by 2 h reperfusion (II/R) or sham-operated surgery (sham). In some rats, simvastatin (20 mg/kg, oral gavage) was administrated 1 h before II/R. KEY FINDINGS II/R reduced acetylcholine-induced relaxation and phenylephrine-induced contraction of PA segments, which were prevented by acute simvastatin pretreatment in vivo or restored by inducible nitric oxide synthase (iNOS) inhibition in situ with 1400 W. Elevated reactive oxygen species (ROS) levels and higher nuclear translocation of nuclear factor kappa B (NFκB) subunit p65 were observed in PA of II/R rats and prevented by simvastatin. Moreover, simvastatin increased superoxide dismutase (SOD) activity and endothelial nitric oxide synthase (eNOS) expression in PA of the II/R group as well as prevented the increased levels of interleukin (IL)-1β and IL-6 in lung explants following II/R. SIGNIFICANCE The study suggests that pretreatment with a single dose of simvastatin prevents the II/R-induced increase of inflammatory factors and oxidative stress, as well as PA endothelial dysfunction and adrenergic hyporreactivity. Therefore, acute simvastatin administration could be therapeutic for pulmonary vascular disease in patients suffering from intestinal ischemic events.
Collapse
Affiliation(s)
- Emília C Peres
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jamaira A Victorio
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Valéria Nunes-Souza
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiza A Rabelo
- Laboratory of Cardiovascular Reactivity, Department of Physiology and Pharmacology, Institute of Biological Sciences, Federal University of Alagoas, Brazil
| | - Wothan Tavares-de-Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Paula Davel
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Luciana V Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
25
|
Chen H, Oliver BG, Pant A, Olivera A, Poronnik P, Pollock CA, Saad S. Effects of air pollution on human health - Mechanistic evidence suggested by in vitro and in vivo modelling. ENVIRONMENTAL RESEARCH 2022; 212:113378. [PMID: 35525290 DOI: 10.1016/j.envres.2022.113378] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Airborne particulate matter (PM) comprises both solid and liquid particles, including carbon, sulphates, nitrate, and toxic heavy metals, which can induce oxidative stress and inflammation after inhalation. These changes occur both in the lung and systemically, due to the ability of the small-sized PM (i.e. diameters ≤2.5 μm, PM2.5) to enter and circulate in the bloodstream. As such, in 2016, airborne PM caused ∼4.2 million premature deaths worldwide. Acute exposure to high levels of airborne PM (eg. during wildfires) can exacerbate pre-existing illnesses leading to hospitalisation, such as in those with asthma and coronary heart disease. Prolonged exposure to PM can increase the risk of non-communicable chronic diseases affecting the brain, lung, heart, liver, and kidney, although the latter is less well studied. Given the breadth of potential disease, it is critical to understand the mechanisms underlying airborne PM exposure-induced disorders. Establishing aetiology in humans is difficult, therefore, in-vitro and in-vivo studies can provide mechanistic insights. We describe acute health effects (e.g. exacerbations of asthma) and long term health effects such as the induction of chronic inflammatory lung disease, and effects outside the lung (e.g. liver and renal change). We will focus on oxidative stress and inflammation as this is the common mechanism of PM-induced disease, which may be used to develop effective treatments to mitigate the adverse health effect of PM exposure.
Collapse
Affiliation(s)
- Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia; Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Sydney, NSW, 2037, Australia
| | - Anushriya Pant
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Annabel Olivera
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Philip Poronnik
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Carol A Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia.
| |
Collapse
|
26
|
Wang H, Wang T, Rui W, Xie J, Xie Y, Zhang X, Guan L, Li G, Lei Z, Schiffelers RM, Sluijter JPG, Xiao J. Extracellular vesicles enclosed-miR-421 suppresses air pollution (PM 2.5 )-induced cardiac dysfunction via ACE2 signalling. J Extracell Vesicles 2022; 11:e12222. [PMID: 35536587 PMCID: PMC9089227 DOI: 10.1002/jev2.12222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/03/2022] [Accepted: 04/15/2022] [Indexed: 01/05/2023] Open
Abstract
Air pollution, via ambient PM2.5, is a big threat to public health since it associates with increased hospitalisation, incidence rate and mortality of cardiopulmonary injury. However, the potential mediators of pulmonary injury in PM2.5‐induced cardiovascular disorder are not fully understood. To investigate a potential cross talk between lung and heart upon PM2.5 exposure, intratracheal instillation in vivo, organ culture ex vivo and human bronchial epithelial cells (Beas‐2B) culture in vitro experiments were performed respectively. The exposed supernatants of Beas‐2B were collected to treat primary neonatal rat cardiomyocytes (NRCMs). Upon intratracheal instillation, subacute PM2.5 exposure caused cardiac dysfunction, which was time‐dependent secondary to lung injury in mice, thereby demonstrating a cross‐talk between lungs and heart potentially mediated via small extracellular vesicles (sEV). We isolated sEV from PM2.5‐exposed mice serum and Beas‐2B supernatants to analyse the change of sEV subpopulations in response to PM2.5. Single particle interferometric reflectance imaging sensing analysis (SP‐IRIS) demonstrated that PM2.5 increased CD63/CD81/CD9 positive particles. Our results indicated that respiratory system‐derived sEV containing miR‐421 contributed to cardiac dysfunction post‐PM2.5 exposure. Inhibition of miR‐421 by AAV9‐miR421‐sponge could significantly reverse PM2.5‐induced cardiac dysfunction in mice. We identified that cardiac angiotensin converting enzyme 2 (ACE2) was a downstream target of sEV‐miR421, and induced myocardial cell apoptosis and cardiac dysfunction. In addition, we observed that GW4869 (an inhibitor of sEV release) or diminazene aceturate (DIZE, an activator of ACE2) treatment could attenuate PM2.5‐induced cardiac dysfunction in vivo. Taken together, our results suggest that PM2.5 exposure promotes sEV‐linked miR421 release after lung injury and hereby contributes to PM2.5‐induced cardiac dysfunction via suppressing ACE2.
Collapse
Affiliation(s)
- Hongyun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Tianhui Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Wei Rui
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Jinxin Xie
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Yuling Xie
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xiao Zhang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Longfei Guan
- China-America Institute Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhiyong Lei
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | | | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
27
|
PM2.5 Concentration Measurement Based on Image Perception. ELECTRONICS 2022. [DOI: 10.3390/electronics11091298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
PM2.5 in the atmosphere causes severe air pollution and dramatically affects the normal production and lives of residents. The real-time monitoring of PM2.5 concentrations has important practical significance for the construction of ecological civilization. The mainstream PM2.5 concentration prediction algorithms based on electrochemical sensors have some disadvantages, such as high economic cost, high labor cost, time delay, and more. To this end, we propose a simple and effective PM2.5 concentration prediction algorithm based on image perception. Specifically, the proposed method develops a natural scene statistical prior to estimating the saturation loss caused by the ’haze’ formed by PM2.5. After extracting the prior features, this paper uses the feedforward neural network to achieve the mapping function from the proposed prior features to the PM2.5 concentration values. Experiments constructed on the public Air Quality Image Dataset (AQID) show the superiority of our proposed PM2.5 concentration measurement method compared to state-of-the-art related PM2.5 concentration monitoring methods.
Collapse
|
28
|
PM2.5 Exposure and Asthma Development: The Key Role of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3618806. [PMID: 35419163 PMCID: PMC9001082 DOI: 10.1155/2022/3618806] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/24/2022] [Indexed: 12/21/2022]
Abstract
Oxidative stress is defined as the imbalance between reactive oxygen species (ROS) production and the endogenous antioxidant defense system, leading to cellular damage. Asthma is a common chronic inflammatory airway disease. The presence of asthma tends to increase the production of reactive oxygen species (ROS), and the antioxidant system in the lungs is insufficient to mitigate it. Therefore, asthma can lead to an exacerbation of airway hyperresponsiveness and airway inflammation. PM2.5 exposure increases ROS levels. Meanwhile, the accumulation of ROS will further enhance the oxidative stress response, resulting in DNA, protein, lipid, and other cellular and molecular damage, leading to respiratory diseases. An in-depth study on the relationship between oxidative stress and PM2.5-related asthma is helpful to understand the pathogenesis and progression of the disease and provides a new direction for the treatment of the disease. This paper reviews the research progress of oxidative stress in PM2.5-induced asthma as well as highlights the therapeutic potentials of antioxidant approaches in treatment of asthma.
Collapse
|
29
|
Qin Y, Zhang H, Jiang B, Chen J, Zhang T. Food bioactives lowering risks of chronic diseases induced by fine particulate air pollution: a comprehensive review. Crit Rev Food Sci Nutr 2022; 63:7811-7836. [PMID: 35317688 DOI: 10.1080/10408398.2022.2051162] [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] [Indexed: 11/03/2022]
Abstract
Airborne particulate matter (PM) exerts huge negative impacts on human health worldwide, not only targeting the respiratory system but more importantly inducing and aggravating associated chronic diseases like asthma, lung cancer, atherosclerosis, diabetes mellitus and Alzheimer diseases. Food-derived bioactive compounds like vitamins, dietary polyphenols, omega-3 polyunsaturated fatty acids and sulforaphane are feasible alternative therapeutic approaches against PM-mediated potential health damages, drawing great attention in recent years. In this review, the association between PM exposure and risks of developing chronic diseases, and the detailed mechanisms underlying the detrimental effects of PM will be discussed. Subsequently, principal food-derived bioactive compounds, with emphasize on the preventative or protective effects against PM, along with potential mechanisms will be elucidated. This comprehensive review will discuss and present current research findings to reveal the nutritional intervention as a preventative or therapeutic strategy against ambient air pollution, thereby lowering the risk of developing chronic diseases.
Collapse
Affiliation(s)
- Yang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Hua Zhang
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Bo Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Jingjing Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
30
|
Li X, Zhang W, Laden F, Curhan GC, Rimm EB, Guo X, Hart JE, Wu S. Dietary nitrate intake and vegetable consumption, ambient particulate matter, and risk of hypertension in the Nurses' Health study. ENVIRONMENT INTERNATIONAL 2022; 161:107100. [PMID: 35066305 PMCID: PMC11968144 DOI: 10.1016/j.envint.2022.107100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Studies have suggested that dietary nitrate could lower blood pressure levels whereas ambient particulate matter (PM) may increase risk of hypertension. However, it is unknown if these exposures may modify each other. OBJECTIVES We collected information on dietary nitrate intake and vegetables consumption and estimated long-term exposures to ambient PM for women in the Nurses' Health Study. METHODS Hazard ratios (HRs) and 95% confidence intervals (95% CIs) for risk of hypertension were calculated using Cox proportional hazards models with adjustment for potential demographic, lifestyle and dietary confounders. Interactions were assessed with multiplicative interaction terms and stratified models. RESULTS Increases in dietary nitrate intake (per 150 mg/d) and green leafy vegetables consumption (per serving/day) were both significantly associated with decreases in hypertension risk (both multivariable-adjusted HRs were 0.97, 95% CI: 0.94, 0.99). Long-term exposure to ambient PM with an aerodynamic diameter ≤ 2.5 µm (PM2.5) was associated with an increased risk of hypertension, with a multivariable-adjusted HR of hypertension of 1.06 (95% CI: 1.02, 1.11) per 10 µg/m3 increase in PM2.5. Ambient PM2.5 significantly modified the associations of dietary nitrate intake (Pinteraction = 0.02) and green leafy vegetables consumption (Pinteraction = 0.004). The associations with dietary factors were gradually weakened with increasing PM2.5: the fully-adjusted HRs for risk of hypertension were 0.94 (95% CI: 0.89, 0.99) and 0.94 (95% CI: 0.90, 0.99) for per 150 mg/d increase in nitrate intake and per serving/d increase in green leafy vegetables consumption, respectively, in the lowest PM2.5 quartile, and 1.00 (95% CI: 0.94, 1.06) and 1.02 (95% CI: 0.97, 1.08), respectively, in the highest PM2.5 quartile. CONCLUSION Our research highlights a potentially protective effect of dietary nitrate intake in the prevention of hypertension and suggests that these benefits are attenuated by increasing exposure to ambient PM2.5.
Collapse
Affiliation(s)
- Xianggui Li
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Weidong Zhang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Francine Laden
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Exposure, Epidemiology and Risk Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gary C Curhan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Renal (Kidney) Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric B Rimm
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Jaime E Hart
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Exposure, Epidemiology and Risk Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| |
Collapse
|
31
|
Hahad O, Kuntic M, Frenis K, Chowdhury S, Lelieveld J, Lieb K, Daiber A, Münzel T. Physical Activity in Polluted Air-Net Benefit or Harm to Cardiovascular Health? A Comprehensive Review. Antioxidants (Basel) 2021; 10:1787. [PMID: 34829658 PMCID: PMC8614825 DOI: 10.3390/antiox10111787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 12/15/2022] Open
Abstract
Both exposure to higher levels of polluted air and physical inactivity are crucial risk factors for the development and progression of major noncommunicable diseases and, in particular, of cardiovascular disease. In this context, the World Health Organization estimated 4.2 and 3.2 million global deaths per year in response to ambient air pollution and insufficient physical activity, respectively. While regular physical activity is well known to improve general health, it may also increase the uptake and deposit of air pollutants in the lungs/airways and circulation, due to increased breathing frequency and minute ventilation, thus increasing the risk of cardiovascular disease. Thus, determining the tradeoff between the health benefits of physical activity and the potential harmful effects of increased exposure to air pollution during physical activity has important public health consequences. In the present comprehensive review, we analyzed evidence from human and animal studies on the combined effects of physical activity and air pollution on cardiovascular and other health outcomes. We further report on pathophysiological mechanisms underlying air pollution exposure, as well as the protective effects of physical activity with a focus on oxidative stress and inflammation. Lastly, we provide mitigation strategies and practical recommendations for physical activity in areas with polluted air.
Collapse
Affiliation(s)
- Omar Hahad
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany;
| | - Marin Kuntic
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
| | - Katie Frenis
- Department of Hematology/Oncology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Sourangsu Chowdhury
- Atmospheric Chemistry Department, Max Planck Institute for Chemistry, 55122 Mainz, Germany; (S.C.); (J.L.)
| | - Jos Lelieveld
- Atmospheric Chemistry Department, Max Planck Institute for Chemistry, 55122 Mainz, Germany; (S.C.); (J.L.)
- Climate and Atmosphere Research Center, The Cyprus Institute, Nicosia 2121, Cyprus
| | - Klaus Lieb
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany;
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| |
Collapse
|
32
|
Park J, Lee KH, Kim H, Woo J, Heo J, Lee CH, Yi SM, Yoo CG. The impact of organic extracts of seasonal PM 2.5 on primary human lung epithelial cells and their chemical characterization. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:59868-59880. [PMID: 34148195 PMCID: PMC8541986 DOI: 10.1007/s11356-021-14850-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/08/2021] [Indexed: 05/13/2023]
Abstract
Lung epithelial cells serve as the first line of defense against various inhaled pollutant particles. To investigate the adverse health effects of organic components of fine particulate matter (PM2.5) collected in Seoul, South Korea, we selected 12 PM2.5 samples from May 2016 to January 2017 and evaluated the effects of organic compounds of PM2.5 on inflammation, cellular aging, and macroautophagy in human lung epithelial cells isolated directly from healthy donors. Organic extracts of PM2.5 specifically induced neutrophilic chemokine and interleukin-8 expression via extracellular signal-regulated kinase activation. Moreover, PM2.5 significantly increased the expression of aging markers (p16, p21, and p27) and activated macroautophagy. Average mass concentrations of organic and elemental carbon had no significant correlations with PM2.5 effects. However, polycyclic aromatic hydrocarbons and n-alkanes were the most relevant components of PM2.5 that correlated with neutrophilic inflammation. Vegetative detritus and residential bituminous coal combustion sources strongly correlated with neutrophilic inflammation, aging, and macroautophagy activation. These data suggest that the chemical composition of PM2.5 is important for determining the adverse health effects of PM2.5. Our study provides encouraging evidence to regulate the harmful components of PM2.5 in Seoul.
Collapse
Affiliation(s)
- Jieun Park
- Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehakno, Jongno-gu, Seoul, 03080, Korea
| | - Hyewon Kim
- Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Jisu Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehakno, Jongno-gu, Seoul, 03080, Korea
| | - Jongbae Heo
- Busan Development Institute, 955 Jungangdae-ro, Busanjin-gu, Busan, 47210, Korea.
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehakno, Jongno-gu, Seoul, 03080, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Muk Yi
- Graduate School of Public Health, Seoul National University, Seoul, Korea
- Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 101 Daehakno, Jongno-gu, Seoul, 03080, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
33
|
Harmon AC, Noël A, Subramanian B, Perveen Z, Jennings MH, Chen YF, Penn AL, Legendre K, Paulsen DB, Varner KJ, Dugas TR. Inhalation of particulate matter containing free radicals leads to decreased vascular responsiveness associated with an altered pulmonary function. Am J Physiol Heart Circ Physiol 2021; 321:H667-H683. [PMID: 34415187 PMCID: PMC8794232 DOI: 10.1152/ajpheart.00725.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022]
Abstract
Airborne particulate matter (PM) is associated with an increased risk for cardiovascular diseases. Although the goal of thermal remediation is to eliminate organic wastes through combustion, when incomplete combustion occurs, organics chemisorb to transition metals to generate PM-containing environmentally persistent free radicals (EPFRs). Similar EPFR species have been detected in PM found in diesel and gasoline exhaust, woodsmoke, and urban air. Prior in vivo studies demonstrated that EPFRs reduce cardiac function secondary to elevations in pulmonary arterial pressures. In vitro studies showed that EPFRs increase ROS and cytokines in pulmonary epithelial cells. We thus hypothesized that EPFR inhalation would promote lung inflammation and oxidative stress, leading to systemic inflammation, vascular endothelial injury, and a decline in vascular function. Mice were exposed to EPFRs for either 4 h or for 4 h/day for 10 days and lung and vascular function were assessed. After a 4-h exposure, plasma nitric oxide (NO) was reduced while endothelin-1 (ET-1) was increased, however lung function was not altered. After 10 day, plasma NO and ET-1 levels were again altered and lung tidal volume was reduced. These time course studies suggested the vasculature may be an early target of injury. To test this hypothesis, an intermediate time point of 3 days was selected. Though the mice exhibited no marked inflammation in either the lung or the blood, we did note significantly reduced endothelial function concurrent with a reduction in lung tidal volume and an elevation in annexin V protein levels in the lung. Although vascular dysfunction was not dependent upon inflammation, it may be associated with an injury at the air-blood interface. Gene expression analysis suggested roles for oxidative stress and aryl hydrocarbon receptor (Ahr) signaling. Studies probing the relationship between pulmonary oxidative stress and AhR signaling at the air-blood interface with vascular dysfunction seem warranted.NEW & NOTEWORTHY Particulate matter (PM) resulting from the combustion of organic matter is known to contribute to cardiopulmonary disease. Despite hypotheses that cardiovascular dysfunction occurring after PM exposures is secondary to lung or systemic inflammation, these studies investigating exposures to PM-containing environmentally persistent free radicals (EPFRs) demonstrate that cardiovascular dysfunction precedes pulmonary inflammation. The cardiopulmonary health consequences of EPFRs have yet to be thoroughly evaluated, especially in healthy, adult mice. Our data suggest the vasculature as a direct target of PM exposure, and our studies aimed to elucidate the mechanisms contributing to EPFR-induced vascular dysfunction.
Collapse
Affiliation(s)
- Ashlyn C Harmon
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | | | - Zakia Perveen
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Merilyn H Jennings
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Yi-Fan Chen
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Kelsey Legendre
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Daniel B Paulsen
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Kurt J Varner
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Tammy R Dugas
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| |
Collapse
|
34
|
Ning R, Li Y, Du Z, Li T, Sun Q, Lin L, Xu Q, Duan J, Sun Z. The mitochondria-targeted antioxidant MitoQ attenuated PM 2.5-induced vascular fibrosis via regulating mitophagy. Redox Biol 2021; 46:102113. [PMID: 34425389 PMCID: PMC8379696 DOI: 10.1016/j.redox.2021.102113] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Short-term PM2.5 exposure is related to vascular remodeling and stiffness. Mitochondria-targeted antioxidant MitoQ is reported to improve the occurrence and development of mitochondrial redox-related diseases. At present, there is limited data on whether MitoQ can alleviate the vascular damage caused by PM2.5. Therefore, the current study was aimed to evaluate the protective role of MitoQ on aortic fibrosis induced by PM2.5 exposure. Vascular Doppler ultrasound manifested PM2.5 damaged both vascular function and structure in C57BL/6J mice. Histopathological analysis found that PM2.5 induced aortic fibrosis and disordered elastic fibers, accompanied by collagen I/III deposition and synthetic phenotype remodeling of vascular smooth muscle cells; while these alterations were partially alleviated following MitoQ treatment. We further demonstrated that mitochondrial dysfunction, including mitochondrial reactive oxygen species (ROS) overproduction and activated superoxide dismutase 2 (SOD2) expression, decreased mitochondrial membrane potential (MMP), oxygen consumption rate (OCR), ATP and increased intracellular Ca2+, as well as mitochondrial fragmentation caused by increased Drp1 expression and decreased Mfn2 expression, occurred in PM2.5-exposed aorta or human aortic vascular smooth muscle cells (HAVSMCs), which were reversed by MitoQ. Moreover, the enhanced expressions of LC3II/I, p62, PINK1 and Parkin regulated mitophagy in PM2.5-exposed aorta and HAVSMCs were weakened by MitoQ. Transfection with PINK1 siRNA in PM2.5-exposed HAVSMCs further improved the effects of MitoQ on HAVSMCs synthetic phenotype remodeling, mitochondrial fragmentation and mitophagy. In summary, our data demonstrated that MitoQ treatment had a protective role in aortic fibrosis after PM2.5 exposure through mitochondrial quality control, which regulated by mitochondrial ROS/PINK1/Parkin-mediated mitophagy. Our study provides a possible targeted therapy for PM2.5-induced arterial stiffness.
Collapse
Affiliation(s)
- Ruihong Ning
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Zhou Du
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lisen Lin
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qing Xu
- Core Facilities Center, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
35
|
The cardiovascular effects of air pollution: Prevention and reversal by pharmacological agents. Pharmacol Ther 2021; 232:107996. [PMID: 34571110 PMCID: PMC8941724 DOI: 10.1016/j.pharmthera.2021.107996] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
Air pollution is associated with staggering levels of cardiovascular morbidity and mortality. Airborne particulate matter (PM), in particular, has been associated with a wide range of detrimental cardiovascular effects, including impaired vascular function, raised blood pressure, alterations in cardiac rhythm, blood clotting disorders, coronary artery disease, and stroke. Considerable headway has been made in elucidating the biological processes underlying these associations, revealing a labyrinth of multiple interacting mechanistic pathways. Several studies have used pharmacological agents to prevent or reverse the cardiovascular effects of PM; an approach that not only has the advantages of elucidating mechanisms, but also potentially revealing therapeutic agents that could benefit individuals that are especially susceptible to the effects of air pollution. This review gathers investigations with pharmacological agents, offering insight into the biology of how PM, and other air pollutants, may cause cardiovascular morbidity.
Collapse
|
36
|
Sousa AS, Sponton ACS, Delbin MA. Perivascular adipose tissue and microvascular endothelial dysfunction in obese mice: Beneficial effects of aerobic exercise in adiponectin receptor (AdipoR1) and peNOS Ser1177. Clin Exp Pharmacol Physiol 2021; 48:1430-1440. [PMID: 34260769 DOI: 10.1111/1440-1681.13550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/28/2022]
Abstract
In the present study, we aim to investigate the effects of aerobic physical training on perivascular adipose tissue (PVAT)-induced microvascular dysfunction of the femoral artery in obese mice. Microvascular reactivity was evaluated in control sedentary (c-SD), obese sedentary (o-SD) and obese trained (o-TR) male mice (C57BL6/JUnib), in the absence (PVAT-) or the presence (PVAT+) of femoral artery PVAT. We also analyzed protein expression, vascular nitric oxide (NO) production and reactive oxygen species (ROS) generation in PVAT. The blood glucose, triglycerides and total cholesterol levels were increased in the o-SD group, when compared with the c-SD group. The maximal responses and the potency to acetylcholine (ACh) were decreased in PVAT+ compared with PVAT- rings in the o-SD group, accompanied by a decrease in vascular protein expression of peNOSSer1177 , Cu/Zn-SOD, leptin receptor (Ob-R) and adiponectin receptor (AdipoR1). The protein expression of leptin increased and that of adiponectin decreased in PVAT. Additionally, vascular NO production was reduced and ROS generation was enhanced in PVAT in the o-SD group. Aerobic exercise training was effective for normalizing ACh relaxation response, vascular NO production and ROS generation in the o-TR group. It partially re-established the vascular protein expression of peNOSSer1177 and the PVAT leptin; normalized the vascular Cu/Zn-SOD and AdipoR1 protein expressions. In obese sedentary mice, the presence of PVAT is involved in the process of microvascular dysfunction of the femoral artery in a pathway associated with increased inflammation and ROS generation. The aerobic exercise training normalized the vascular response, the NO production and/or bioavailability and oxidative stress, with improved vascular expressions of Cu/Zn-SOD, peNOSser1177 , and AdipoR1.
Collapse
Affiliation(s)
- Andressa S Sousa
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Amanda C S Sponton
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Maria A Delbin
- Laboratory of Vascular Biology, Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
37
|
Muscat Baron Y. Could the COVID-19 Positive Asymptomatic Tobacco Smoker be a Silent Superspeader? ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021099. [PMID: 33988145 PMCID: PMC8182616 DOI: 10.23750/abm.v92i2.11147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/03/2021] [Indexed: 11/23/2022]
Abstract
There appears to be a connection between COVID-19 infection and an airborne microscopic pollutant called particulate matter which has been suggested to act as vector for viral transmission. The highest human exposure to particulate matter occurs during smoking and to a lesser extent to 2nd hand smoking. This article offers a hypothetical proposition that particulate matter derived from tobacco smoking may act as COVID-19’s vector for infection transmission. With a background smoking Chinese male population of more than 66% and more than 70% of Chinese nonsmokers exposed to 2nd hand smoke, the potential of exhaled smoke acting as a viral vector is significant. If this hypothesis is proven, measures such as face protection to reduce coronavirus-laden particulate matter transmission, measures of social distancing and legislation to protect nonsmokers from contracting the infection through 2nd hand smoking should be implemented. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Yves Muscat Baron
- a:1:{s:5:"en_US";s:55:"Mater Dei Hospital, University of Malta Medical School ";}.
| |
Collapse
|
38
|
Costa-Beber LC, Goettems-Fiorin PB, Dos Santos JB, Friske PT, Heck TG, Hirsch GE, Ludwig MS. Ovariectomy reduces the cardiac cytoprotection in rats exposed to particulate air pollutant. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:23395-23404. [PMID: 33443732 DOI: 10.1007/s11356-021-12350-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/02/2021] [Indexed: 06/12/2023]
Abstract
Fine particulate matter (PM2.5) has been considered a risk factor for cardiovascular diseases by inducing an oxidative and inflammatory phenotype. Besides, the reduction of 17β-estradiol (E2) levels during menopause is a natural risk for cardiovascular outcomes. During the E2 downfall, there is a high requirement of the 70-kDa heat shock proteins (HSP70), which present essential antioxidant, anti-inflammatory, and anti-senescence roles. We investigated if the ovariectomy, an animal model for menopause, could induce additional effects in cardiac health by impairing oxidative and heat shock response parameters of female rats chronically exposed to residual oil fly ash (ROFA; an inorganic fraction of PM2.5). Thus, ROFA was obtained from São Paulo (Brazil) and solubilized it in saline. Further, female Wistar rats were exposed to 50 μL of saline (control group) or ROFA solution (250 μg) (polluted) by intranasal instillation, 5 days/week, 12 weeks. At the 12th week, animals were subdivided into four groups (n = 6 p/group): control, OVX, polluted, and polluted + OVX. Control and polluted were submitted to false surgery, while OVX and polluted + OVX were ovariectomized. ROFA or saline exposure continued for 12 weeks. Ovariectomy reduced the cardiac catalase activity and iHSP70 expression in female rats exposed to ROFA. Neither plasma eHSP72 levels nor H-index (eHSP72 to cardiac iHSP70 ratio) was affected. In conclusion, ovariectomy reduces the cardiac cytoprotection and antioxidant defense, and enhances the susceptibility to premature cellular senescence in rats exposed to ROFA.
Collapse
Affiliation(s)
- Lílian Corrêa Costa-Beber
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil.
- Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil.
| | - Pauline Brendler Goettems-Fiorin
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
- Atmospheric Pollution Laboratory, Postgraduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite 245, Porto Alegre, RS, Brazil
| | - Jaíne Borges Dos Santos
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
| | - Paula Taís Friske
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
| | - Thiago Gomes Heck
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
- Medicine Course, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Gabriela Elisa Hirsch
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Mirna Stela Ludwig
- Research Group in Physiology, Department of Life Sciences, Postgraduate Program in Integral Attention to Health, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000, Bairro Universitário, Ijuí, RS, 98700-000, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
- Medicine Course, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| |
Collapse
|
39
|
Hill BG, Rood B, Ribble A, Haberzettl P. Fine particulate matter (PM 2.5) inhalation-induced alterations in the plasma lipidome as promoters of vascular inflammation and insulin resistance. Am J Physiol Heart Circ Physiol 2021; 320:H1836-H1850. [PMID: 33666505 PMCID: PMC8163652 DOI: 10.1152/ajpheart.00881.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
Fine particulate matter (PM2.5) air pollution exposure increases the risk of developing cardiovascular disease (CVD). Although the precise mechanisms by which air pollution exposure increases CVD risk remain uncertain, research indicates that PM2.5-induced endothelial dysfunction contributes to CVD risk. Previous studies demonstrate that concentrated ambient PM2.5 (CAP) exposure induces vascular inflammation and impairs insulin and vascular endothelial growth factor (VEGF) signaling dependent on pulmonary oxidative stress. To assess whether CAP exposure induces these vascular effects via plasmatic factors, we incubated aortas from naïve mice with plasma isolated from mice exposed to HEPA-filtered air or CAP (9 days) and examined vascular inflammation and insulin and VEGF signaling. We found that treatment of naïve aortas with plasma from CAP-exposed mice activates NF-κBα and induces insulin and VEGF resistance, indicating transmission by plasmatic factor(s). To identify putative factors, we exposed lung-specific ecSOD-transgenic (ecSOD-Tg) mice and wild-type (WT) littermates to CAP at concentrations of either ∼60 µg/m3 (CAP60) or ∼100 µg/m3 (CAP100) and measured the abundance of plasma metabolites by mass spectrometry. In WT mice, both CAP concentrations increased levels of fatty acids such as palmitate, myristate, and palmitoleate and decreased numerous phospholipid species; however, these CAP-induced changes in the plasma lipidome were prevented in ecSOD-Tg mice. Consistent with the literature, we found that fatty acids such as palmitate are sufficient to promote endothelial inflammation. Collectively, our findings suggest that PM2.5 exposure, by inducing pulmonary oxidative stress, promotes unique lipidomic changes characterized by high levels of circulating fatty acids, which are sufficient to trigger vascular pathology.NEW & NOTEWORTHY We found that circulating plasma constituents are responsible for air pollution-induced vascular pathologies. Inhalation of fine particulate matter (≤PM2.5) promotes a unique form of dyslipidemia that manifests in a manner dependent upon pulmonary oxidative stress. The air pollution-engendered dyslipidemic phenotype is characterized by elevated free fatty acid species and diminished phospholipid species, which could contribute to vascular inflammation and loss of insulin sensitivity.
Collapse
Affiliation(s)
- Bradford G Hill
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Benjamin Rood
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Amanda Ribble
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Petra Haberzettl
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
40
|
Aztatzi-Aguilar OG, Pardo-Osorio GA, Uribe-Ramírez M, Narváez-Morales J, De Vizcaya-Ruiz A, Barbier OC. Acute kidney damage by PM 2.5 exposure in a rat model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 83:103587. [PMID: 33460805 DOI: 10.1016/j.etap.2021.103587] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
PM2.5 exposure is associated with a glomerular filtration rate (GFR) reduction, and renal tissue damage. The goal of this study was demonstrate the acute effect of PM2.5 on the kidney. Male rats were acutely exposed to PM2.5 or filtered air. Blood pressure was mesure and early kidney biomarkers were evaluated in serum and urine samples, and also IL-1β, IL-6 and TNFα were determined. Oxidative biomarkers, angiotensin/bradykinin-related proteins, KIM-1, IL-6 and histology were determined. Blood pressure, GFR, and early kidney damage biomarkers increase together with oxidative biomarkers and angiotensin/bradykinin endocrine-related proteins increased after exposure to PM2.5. Urinary IL-6 increased after exposure to PM2.5, whereas in kidney cortex decreased. Histological changes were observed and accompanied by the induction of KIM-1. Acute exposure to PM2.5 not decline kidney function. However, it can induce early kidney damage biomarkers, oxidative stress, inflammation and angiotensin mediators, which perhabs culminates in a lose of renal function.
Collapse
Affiliation(s)
- Octavio Gamaliel Aztatzi-Aguilar
- Departamento de Investigación en Inmunología y Medicina Ambiental, Instituto Nacional de Enfermedades Respiratorias, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, C.P. 14080, Ciudad de México, CDMX, Mexico.
| | - Gabriela Andrea Pardo-Osorio
- Universidad del Valle de México, Av. Observatorio 400. Col. 16 de Septiembre, C.P. 11810, Ciudad de México, CDMX, Mexico.
| | - Marisela Uribe-Ramírez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col San Pedro Zacatenco, C.P. 07360, Ciudad de México, CDMX, Mexico.
| | - Juana Narváez-Morales
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col San Pedro Zacatenco, C.P. 07360, Ciudad de México, CDMX, Mexico.
| | - Andrea De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col San Pedro Zacatenco, C.P. 07360, Ciudad de México, CDMX, Mexico.
| | - Olivier Christophe Barbier
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col San Pedro Zacatenco, C.P. 07360, Ciudad de México, CDMX, Mexico.
| |
Collapse
|
41
|
Xie W, You J, Zhi C, Li L. The toxicity of ambient fine particulate matter (PM2.5) to vascular endothelial cells. J Appl Toxicol 2021; 41:713-723. [DOI: 10.1002/jat.4138] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/17/2020] [Accepted: 12/27/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Jia You
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Chenxi Zhi
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Liang Li
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards University of South China Hengyang China
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study University of South China Hengyang China
| |
Collapse
|
42
|
Circadian Deregulation as Possible New Player in Pollution-Induced Tissue Damage. ATMOSPHERE 2021. [DOI: 10.3390/atmos12010116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Circadian rhythms are 24-h oscillations driven by a hypothalamic master oscillator that entrains peripheral clocks in almost all cells, tissues and organs. Circadian misalignment, triggered by industrialization and modern lifestyles, has been linked to several pathological conditions, with possible impairment of the quality or even the very existence of life. Living organisms are continuously exposed to air pollutants, and among them, ozone or particulate matters (PMs) are considered to be among the most toxic to human health. In particular, exposure to environmental stressors may result not only in pulmonary and cardiovascular diseases, but, as it has been demonstrated in the last two decades, the skin can also be affected by pollution. In this context, we hypothesize that chronodistruption can exacerbate cell vulnerability to exogenous damaging agents, and we suggest a possible common mechanism of action in deregulation of the homeostasis of the pulmonary, cardiovascular and cutaneous tissues and in its involvement in the development of pathological conditions.
Collapse
|
43
|
Lan Y, Ng CT, Ong CN, Yu LE, Bay BH. Transcriptomic analysis identifies dysregulated genes and functional networks in human small airway epithelial cells exposed to ambient PM 2.5. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111702. [PMID: 33396033 DOI: 10.1016/j.ecoenv.2020.111702] [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: 06/26/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Cellular models exhibiting human physiological features of pseudostratified columnar epithelia, provide a more realistic approach for elucidating detailed mechanisms underlying PM2.5-induced pulmonary toxicity. In this study, we characterized the barrier and mucociliary functions of differentiated human small airway epithelial cells (SAECs), cultured at the air-liquid interface (ALI). Due to the presence of mucociliary protection, particle internalization was reduced, with a concomitant decrease in cytotoxicity in differentiated S-ALI cells, as compared to conventional submerged SAEC cultures. After 24-hour exposure to PM2.5 surrogates, 117 up-regulated genes and 156 down-regulated genes were detected in S-ALI cells, through transcriptomic analysis using the Affymetrix Clariom™ S Human Array. Transcription-level changes in >60 signaling pathways, were revealed by functional annotation of the 273 differentially expressed genes, using the PANTHER Gene List Analysis. These pathways are involved in multiple cellular processes, that include inflammation and apoptosis. Exposure to urban PM2.5 led to complex responses in airway epithelia, including a net induction of downstream pro-inflammatory and pro-apoptotic responses. Collectively, this study highlights the importance of using the more advanced ALI model rather than the undifferentiated submerged model, to avoid over-assessment of inhaled particle toxicity in human. The results of our study also suggest that reduction of ambient PM2.5 concentrations would have a protective effect on respiratory health in humans.
Collapse
Affiliation(s)
- Yang Lan
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Cheng Teng Ng
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore
| | - Choon Nam Ong
- NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Liya E Yu
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore 117576, Singapore; NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
44
|
Ficociello G, Inverni A, Massimi L, Buccini G, Canepari S, Uccelletti D. Assessment of the effects of atmospheric pollutants using the animal model Caenorhabditis elegans. ENVIRONMENTAL RESEARCH 2020; 191:110209. [PMID: 32937173 DOI: 10.1016/j.envres.2020.110209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 06/11/2023]
Abstract
Air pollution is recognized as the world's largest environmental health risk. In this work we evaluated in vivo the effects of three relevant components of atmospheric dusts (brake dust, wood pellet ash and Saharan dust) employing the animal model Caenorhabditis elegans. Main endpoints of C. elegans such as life span, brood size and oxidative stress were addressed by exposing the nematodes to different dust concentrations. Brake dust and pellet ash affected the life span and increased significantly the oxidative stress of exposed nematodes, while Saharan dust showed no effects. Water soluble and insoluble fractions of these dusts were used to investigate the impact of the single fraction on C. elegans. The two fractions of brake dust and pellet ash exerted different effects on C. elegans endpoints in terms of life span and oxidative stress response. These fractions acted in different ways on the worm susceptibility to infection of two human pathogens (Staphylococcus aureus and Pseudomonas aeruginosa) affecting the sek-1 gene expression. In conclusion, our study showed that C. elegans is a valuable tool to investigate in vivo possible effects of atmospheric dusts.
Collapse
Affiliation(s)
- Graziella Ficociello
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy
| | - Agnese Inverni
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy; Chemistry Department, Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy
| | - Lorenzo Massimi
- Chemistry Department, Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy
| | - Giulio Buccini
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy
| | - Silvia Canepari
- Chemistry Department, Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy
| | - Daniela Uccelletti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, P.le Aldo Moro, 5, 00185, Rome, Italy.
| |
Collapse
|
45
|
Wyatt LH, Devlin RB, Rappold AG, Case MW, Diaz-Sanchez D. Low levels of fine particulate matter increase vascular damage and reduce pulmonary function in young healthy adults. Part Fibre Toxicol 2020; 17:58. [PMID: 33198760 PMCID: PMC7670817 DOI: 10.1186/s12989-020-00389-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) related mild inflammation, altered autonomic control of cardiovascular function, and changes to cell function have been observed in controlled human exposure studies. METHODS To measure the systemic and cardiopulmonary impacts of low-level PM exposure, we exposed 20 healthy, young volunteers to PM2.5, in the form of concentrated ambient particles (mean: 37.8 μg/m3, SD 6.5), and filtered air (mean: 2.1 μg/m3, SD 2.6). In this double-blind, crossover study the exposure order was randomized. During the 4 h exposure, volunteers (7 females and 13 males) underwent light intensity exercise to regulate ventilation rate. We measured pulmonary, cardiac, and hematologic end points before exposure, 1 h after exposure, and again 20 h after exposure. RESULTS Low-level PM2.5 resulted in both pulmonary and extra-pulmonary changes characterized by alterations in systematic inflammation markers, cardiac repolarization, and decreased pulmonary function. A mean increase in PM2.5 concentration (37.8 μg/m3) significantly increased serum amyloid A (SAA), C-reactive protein (CRP), soluble intercellular adhesion molecule-1 (sICAM-1), and soluble vascular cell adhesion molecule-1 (sVCAM-1), 1 h after exposure by 8.7, 9.1, 10.7, and 6.6%, respectively, relative to the filtered air control. SAA remained significantly elevated (34.6%) 20 h after PM2.5 exposure which was accompanied by a 5.7% decrease in percent neutrophils. Decreased pulmonary function was observed 1 h after exposure through a 0.8 and 1.2% decrease in forced expiratory volume in 1 s (FEV1) and FEV1/ forced vital capacity (FEV1/FVC) respectively. Additionally, sex specific changes were observed in repolarization outcomes following PM2.5 exposure. In males, P-wave and QRS complex were increased by 15.4 and 5.4% 1 h after exposure. CONCLUSIONS This study is the first controlled human exposure study to demonstrate biological effects in response to exposure to concentrated ambient air PM2.5 particles at levels near the PM2.5 US NAAQS standard. CLINICAL TRIAL REGISTRATION INFORMATION clinicaltrials.gov ; Identifier: NCT03232086 . The study was registered retrospectively on July 25, 2017, prior to final data collection on October 25, 2017 and data analysis.
Collapse
Affiliation(s)
- Lauren H Wyatt
- Public Health and Integrated Toxicology Division, Human Studies Facility, United States Environmental Protection Agency (USEPA), Research Triangle Park, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA.
| | - Robert B Devlin
- Public Health and Integrated Toxicology Division, Human Studies Facility, United States Environmental Protection Agency (USEPA), Research Triangle Park, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA
| | - Ana G Rappold
- Public Health and Integrated Toxicology Division, Human Studies Facility, United States Environmental Protection Agency (USEPA), Research Triangle Park, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA
| | - Martin W Case
- Public Health and Integrated Toxicology Division, Human Studies Facility, United States Environmental Protection Agency (USEPA), Research Triangle Park, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA
| | - David Diaz-Sanchez
- Public Health and Integrated Toxicology Division, Human Studies Facility, United States Environmental Protection Agency (USEPA), Research Triangle Park, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA
| |
Collapse
|
46
|
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
|
47
|
Shirazi J, Donzanti MJ, Nelson KM, Zurakowski R, Fromen CA, Gleghorn JP. Significant Unresolved Questions and Opportunities for Bioengineering in Understanding and Treating COVID-19 Disease Progression. Cell Mol Bioeng 2020; 13:259-284. [PMID: 32837585 PMCID: PMC7384395 DOI: 10.1007/s12195-020-00637-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 is a disease that manifests itself in a multitude of ways across a wide range of tissues. Many factors are involved, and though impressive strides have been made in studying this novel disease in a very short time, there is still a great deal that is unknown about how the virus functions. Clinical data has been crucial for providing information on COVID-19 progression and determining risk factors. However, the mechanisms leading to the multi-tissue pathology are yet to be fully established. Although insights from SARS-CoV-1 and MERS-CoV have been valuable, it is clear that SARS-CoV-2 is different and merits its own extensive studies. In this review, we highlight unresolved questions surrounding this virus including the temporal immune dynamics, infection of non-pulmonary tissue, early life exposure, and the role of circadian rhythms. Risk factors such as sex and exposure to pollutants are also explored followed by a discussion of ways in which bioengineering approaches can be employed to help understand COVID-19. The use of sophisticated in vitro models can be employed to interrogate intercellular interactions and also to tease apart effects of the virus itself from the resulting immune response. Additionally, spatiotemporal information can be gleaned from these models to learn more about the dynamics of the virus and COVID-19 progression. Application of advanced tissue and organ system models into COVID-19 research can result in more nuanced insight into the mechanisms underlying this condition and elucidate strategies to combat its effects.
Collapse
Affiliation(s)
- Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Michael J. Donzanti
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Ryan Zurakowski
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| |
Collapse
|
48
|
Gangwar RS, Bevan GH, Palanivel R, Das L, Rajagopalan S. Oxidative stress pathways of air pollution mediated toxicity: Recent insights. Redox Biol 2020; 34:101545. [PMID: 32505541 PMCID: PMC7327965 DOI: 10.1016/j.redox.2020.101545] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/01/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023] Open
Abstract
Ambient air pollution is a leading environmental cause of morbidity and mortality globally with most of the outcomes of cardiovascular origin. While numerous mechanisms are proposed to explain the link between air pollutants and cardiovascular events, the evidence supports a role for oxidative stress as a critical intermediary pathway in the transduction of systemic responses in the cardiovascular system. Indeed, alterations in vascular function are a critical step in the development of cardiometabolic disorders such as hypertension, diabetes, and atherosclerosis. This review will provide an overview of the impact of particulate and gaseous pollutants on oxidative stress from human and animal studies published in the last five years. We discuss current gaps in knowledge and evidence to date implicating the role of oxidative stress with an emphasis on inhalational exposures. We conclude with the identification of gaps, and an exhortation for further studies to elucidate the impact of oxidative stress in air pollution mediated effects. Particulate matter air pollution is the leading risk factor for cardiovascular morbidity and mortality globally. Mechanisms of oxidative stress mediated pathways. How does lung inflammation crucial to inhalational exposure mediate systemic toxicity? Review of recent animal and human exposure studies providing insights into oxidative stress pathways.
Collapse
Affiliation(s)
- Roopesh Singh Gangwar
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Graham H Bevan
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rengasamy Palanivel
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lopa Das
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
49
|
Miller MR. Oxidative stress and the cardiovascular effects of air pollution. Free Radic Biol Med 2020; 151:69-87. [PMID: 31923583 PMCID: PMC7322534 DOI: 10.1016/j.freeradbiomed.2020.01.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
Abstract
Cardiovascular causes have been estimated to be responsible for more than two thirds of the considerable mortality attributed to air pollution. There is now a substantial body of research demonstrating that exposure to air pollution has many detrimental effects throughout the cardiovascular system. Multiple biological mechanisms are responsible, however, oxidative stress is a prominent observation at many levels of the cardiovascular impairment induced by pollutant exposure. This review provides an overview of the evidence that oxidative stress is a key pathway for the different cardiovascular actions of air pollution.
Collapse
Affiliation(s)
- Mark R Miller
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH4 3RL, United Kingdom.
| |
Collapse
|
50
|
Long MH, Zhang C, Xu DQ, Fu WL, Gan XD, Li F, Wang Q, Xia W, Xu DG. PM 2.5 aggravates diabetes via the systemically activated IL-6-mediated STAT3/SOCS3 pathway in rats' liver. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113342. [PMID: 31676093 DOI: 10.1016/j.envpol.2019.113342] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 06/10/2023]
Abstract
PM2.5 exposure aggravates type 2 diabetes, in which inflammatory factors play an important role. In this study, we aimed to explore the mechanisms responsible for aggravating diabetes after PM2.5 exposure, and study the roles of inflammatory factors in insulin-resistant type 2 diabetes. Our study indicated that short-time PM2.5 exposure enhances insulin resistance in type 2 diabetic rats and significantly raises inflammatory factors, including IL-6, TNF-α, and MCP-1, in lungs. However, we found that of these inflammatory factors only IL-6 levels are elevated in blood, liver, adipose tissue, and macrophages, but not in skeletal muscle. IL-6 induced activation of the STAT3/SOCS3 pathway in liver, but not other downstream pathways including STAT1, ERK1/2, and PI3K. Both STAT3 inhibition and IL-6 neutralization effectively alleviated the disorders of glucose metabolism after PM2.5 exposure. Taken together, this suggests that the systemic increase in IL-6 may play an important role in the deterioration of the type 2 diabetes via IL-6/STAT3/SOCS3 pathway in liver after short-time exposure to PM2.5. Besides, we unexpectedly found a stronger resistance to the PM2.5 exposure-induced increase in IL-6 in skeleton muscle than those of many other tissues.
Collapse
Affiliation(s)
- Min-Hui Long
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China; The South China Normal University, Guangzhou, 510631, PR China
| | - Chao Zhang
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Dong-Qun Xu
- Institute of Products Related with Environment and Health, China Center for Diseases Control, Beijing, 100080, PR China
| | - Wen-Liang Fu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Xiang-Dong Gan
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China; The South China Normal University, Guangzhou, 510631, PR China
| | - Fei Li
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Qin Wang
- Institute of Products Related with Environment and Health, China Center for Diseases Control, Beijing, 100080, PR China
| | - Wenrong Xia
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China
| | - Dong-Gang Xu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, PR China.
| |
Collapse
|