101
|
Xiong Q, Tian X, Xu C, Ma B, Liu W, Sun B, Ru Q, Shu X. PM 2 .5 exposure-induced ferroptosis in neuronal cells via inhibiting ERK/CREB pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2201-2213. [PMID: 35608139 DOI: 10.1002/tox.23586] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 03/25/2022] [Accepted: 05/06/2022] [Indexed: 05/16/2023]
Abstract
PM2.5 exposure has been demonstrated to correlate with neurological disorders recently. Ferroptosis is recognized as a newly found programmed form of cell death associated with neurodegenerative diseases, while glutathione peroxidase 4 (GPX4) is a key regulator of ferroptosis. However, the relationship between PM2.5 -induced neurotoxicity and ferroptosis is still unclear. The current study aims to investigate if ferroptosis is involved in neurotoxicity post PM2.5 exposure and its underlying mechanism. The PM2.5 -treated neuronal Neuro-2a (N2A) and SH-SY5Y cells were applied to the current study. The results showed that PM2.5 significantly increased the neuronal cell death, yet the ferroptosis antagonist Ferrostain-1 (Fer-1) markedly decreased the cell death induced by PM2.5 . Western blot further confirmed that ferroptosis was triggered post PM2.5 treatment in N2A cells by decreasing expressions of GPX4 and ferritin heavy chain (FTH), as well as enhancing expressions of ferritin light chain (FTL) and transferrin receptor protein (TFRC). Meanwhile, PM2.5 treatment augmented neuronal oxidative damage and mitochondrial dysfunction. The bioinformatic analysis indicated that CREB could be the regulator of GPX4, and our results showed that ERK/CREB pathway was down-regulated in N2A cells post PM2.5 treatment. The addition of ERK1/2 agonist post PM2.5 treatment significantly inhibit ferroptosis via increasing the expression of GPX4. Taken together, the present study demonstrated that PM2.5 -induced ferroptosis via inhibiting ERK/CREB pathway, and these findings will advance our knowledge of PM2.5 -induced cytotoxicity in the nervous system.
Collapse
Affiliation(s)
- Qi Xiong
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Xiang Tian
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Congyue Xu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Baomiao Ma
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Wei Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Binlian Sun
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| | - Qin Ru
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
- Wuhan Economic and Technological Development Zone, Jianghan University, Wuhan City, China
| | - Xiji Shu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan, People's Republic of China
| |
Collapse
|
102
|
Li K, Zhang Q, Wang T, Rong R, Hu X, Zhang Y. Laboratory investigation of pollutant emissions and PM 2.5 toxicity of underground coal fires. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155537. [PMID: 35489495 DOI: 10.1016/j.scitotenv.2022.155537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 06/14/2023]
Abstract
Widespread underground coal fires (UCFs) release large amounts of pollutants, thus leading to air pollution and health impacts. However, this topic has not been widely investigated, especially regarding the potential health hazards. We quantified the pollutant emissions and analyzed the physicochemical properties of UCF PM2.5 in a laboratory study of coal smoldering under a simulated UCF background. The emission factors of CO2, CO, and PM2.5 were 2489 ± 35, 122 ± 9, 12.90 ± 1.79 g/kg, respectively. UCF PM2.5 are carbonaceous particles with varied morphology and complex composition, including heavy metals, silica and polycyclic aromatic hydrocarbons (PAHs). The main PAHs components were those with 2-4 rings. Benzoapyrene (BaP) and indeno[1,2, 3-cd]pyrene (IcdP) were important contributors to the carcinogenesis of these PAHs. We quantitatively evaluate the toxicity of inhaled UCF PM2.5 using a nasal inhalation exposure system. The target organs of UCF PM2.5 are lungs, liver, and kidneys. UCF PM2.5 presented an enriched chemical composition and induced inflammation and oxidative stress, which together mediated multiple organ injury. Long-term PM2.5 metabolism is the main cause of persistent toxicity, which might lead to long-term chronic diseases. Therefore, local authorities should recognize the importance and effects of UCF emissions, especially PM2.5, to establish control and mitigation measures.
Collapse
Affiliation(s)
- Kaili Li
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China
| | - Qixing Zhang
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China.
| | - Tong Wang
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory (HFIPS), Chinese Academy of Science, Hefei 230031, China; University of Science and Technology of China, Hefei 230026, China
| | - Rui Rong
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Xiaowen Hu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yongming Zhang
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
103
|
He X, Zhang L, Liu S, Wang J, Liu Y, Xiong A, Jiang M, Luo L, Ying X, Li G. Methyltransferase-like 3 leads to lung injury by up-regulation of interleukin 24 through N6-methyladenosine-dependent mRNA stability and translation efficiency in mice exposed to fine particulate matter 2.5. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 308:119607. [PMID: 35718042 DOI: 10.1016/j.envpol.2022.119607] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter 2.5 (PM2.5) exposure leads to the progress of pulmonary disease. It has been reported that N6-methyladenosine (m6A) modification was involved in various biological processes and diseases. However, the critical role of m6A modification in pulmonary disease during PM2.5 exposure remains elusive. Here, we revealed that lung inflammation and mucus production caused by PM2.5 were associated with m6A modification. Both in vivo and in vitro assays demonstrated that PM2.5 exposure elevated the total level of m6A modification as well as the methyltransferase like 3 (METTL3) expression. Integration analysis of m6A RNA immunoprecipitation-seq (meRIP-seq) and RNA-seq discovered that METTL3 up-regulated the expression level and the m6A modification of Interleukin 24 (IL24). Importantly, we explored that the stability of IL24 mRNA was enhanced due to the increased m6A modification. Moreover, the data from qRT-PCR showed that PM2.5 also increased YTH N6-Methyladenosine RNA Binding Protein 1 (YTHDF1) expression, and the up-regulated YTHDF1 augmented IL24 mRNA translation efficiency. Down-regulation of Mettl3 reduced Il24 expression and ameliorated the pulmonary inflammation and mucus secretion in mice exposed to PM2.5. Taken together, our finding provided a comprehensive insight for revealing the significant role of m6A regulators in the lung injury via METTL3/YTHDF1-coupled epitranscriptomal regulation of IL24.
Collapse
Affiliation(s)
- Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Shengbin Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Yao Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Manling Jiang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China
| | - Li Luo
- 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
| | - Xiong Ying
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu, 610000, China
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China; Department of Pulmonary and Critical Care Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, China.
| |
Collapse
|
104
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
105
|
Lin Z, Yang X, Guan L, Qin L, Ding J, Zhou L. The link between ferroptosis and airway inflammatory diseases: A novel target for treatment. Front Mol Biosci 2022; 9:985571. [PMID: 36060261 PMCID: PMC9428508 DOI: 10.3389/fmolb.2022.985571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is an iron-dependent mode of cell death characterized by intracellular lipid peroxide accumulation and a redox reaction imbalance. Compared with other modes of cell death, ferroptosis has specific biological and morphological features. The iron-dependent lipid peroxidation accumulation is manifested explicitly in the abnormal metabolism of intracellular lipid oxides catalyzed by excessive iron ions with the production of many reactive oxygen species and over-oxidization of polyunsaturated fatty acids. Recent studies have shown that various diseases, which include intestinal diseases and cancer, are associated with ferroptosis, but few studies are related to airway inflammatory diseases. This review provides a comprehensive analysis of the primary damage mechanisms of ferroptosis and summarizes the relationship between ferroptosis and airway inflammatory diseases. In addition to common acute and chronic airway inflammatory diseases, we also focus on the progress of research on COVID-19 in relation to ferroptosis. New therapeutic approaches and current issues to be addressed in the treatment of inflammatory airway diseases using ferroptosis are further proposed.
Collapse
|
106
|
Gene Expression Changes Induced by Exposure of RAW 264.7 Macrophages to Particulate Matter of Air Pollution: The Role of Endotoxins. Biomolecules 2022; 12:biom12081100. [PMID: 36008994 PMCID: PMC9405577 DOI: 10.3390/biom12081100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the variable chemical and physical characteristics of particulate air pollutants, inflammation and oxidative stress have been identified as common mechanisms for cell damage and negative health influences. These effects are produced by organic components, especially by endotoxins. This study analyzed the gene expression profile after exposure of RAW 264.7 cells to the standard particulate matter (PM) material, NIST1648a, and PM with a reduced organic matter content, LAp120, in comparison to the effects of lipopolysaccharide (LPS). The selected parameters of cell viability, cell cycle progression, and metabolic and inflammatory activity were also investigated. Both forms of PM negatively influenced the parameters of cell activity. These results were generally reflected in the gene expression profile. Only NIST1648a, excluding LAp120, contained endotoxins and showed small but statistically significant pro-inflammatory activity. However, the gene expression profiling revealed strong pro-inflammatory cell activation induced by NIST1648a that was close to the effects of LPS. Changes in gene expression triggered by LAp120 were relatively small. The observed differences in the effects of NIST1648a and LAp120 were related to the content of organic matter in which bacterial endotoxins play an important role. However, other organic compounds and their interactions with other PM components also appear to be of significant importance.
Collapse
|
107
|
From Iron Metabolism to Ferroptosis: Pathologic Changes in Coronary Heart Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6291889. [PMID: 35993022 PMCID: PMC9385341 DOI: 10.1155/2022/6291889] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022]
Abstract
Coronary heart disease (CHD) is closely related to oxidative stress and inflammatory response and is the most common cardiovascular disease (CVD). Iron is an essential mineral that participates in many physiological and biochemical reactions in the human body. Meanwhile, on the negative side, iron has an active redox capacity, which leads to the accumulation of reactive oxygen species (ROS) and lipid peroxidation. There is growing evidence that disordered iron metabolism is involved in CHD's pathological progression. And the result of disordered iron metabolism is associated with iron overload-induced programmed cell death, often called ferroptosis. That features iron-dependent lipid peroxidation. Ferroptosis may play a crucial role in the development of CHD, and targeting ferroptosis may be a promising option for treating CHD. Here, we review the mechanisms of iron metabolism in cardiomyocytes (CMs) and explain the correlation between iron metabolism and ferroptosis. Meanwhile, we highlight the specific roles of iron metabolism and ferroptosis in the main pathological progression of CHD.
Collapse
|
108
|
Zou L, Li B, Xiong L, Wang Y, Xie W, Huang X, Liang Y, Wei T, Liu N, Chang X, Bai C, Wu T, Xue Y, Zhang T, Tang M. Urban fine particulate matter causes cardiac hypertrophy through calcium-mediated mitochondrial bioenergetics dysfunction in mice hearts and human cardiomyocytes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119236. [PMID: 35367502 DOI: 10.1016/j.envpol.2022.119236] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
In recent years, the cardiovascular toxicity of urban fine particulate matter (PM2.5) has sparked significant alarm. Mitochondria produce 90% of ATP and make up 30% of the volume of cardiomyocytes. Thus knowledge of myocardial mitochondrial dysfunction due to PM2.5 exposure is essential for further cardiotoxic effects. Here, the mechanism of PM2.5-induced cardiac hypertrophy through calcium overload and mitochondrial dysfunction was investigated in vivo and in vitro. Male and female BALB/c mice were given 1.28, 5.5, and 11 mg PM2.5/kg bodyweight weekly through oropharyngeal inhalation for four weeks and were assigned to low, medium, and high dose groups, respectively. PM2.5-induced myocardial edema and cardiac hypertrophy were detected in the high-dose group. Mitochondria were scattered and ruptured with abnormal ultrastructural morphology. In vitro experiments on human cardiomyocyte AC16 showed that exposure to PM2.5 for 24 h caused opened mitochondrial permeability transition pore --leading to excessive calcium production, decreased mitochondrial membrane potential, weakened mitochondrial respiratory metabolism capacity, and decreased ATP production. Nevertheless, the administration of calcium chelator ameliorated the mitochondrial damage in the PM2.5-treated group. Our in vivo and in vitro results confirmed that calcium overload under PM2.5 exposure triggered mTOR/AKT/GSK-3β activation, leading to mitochondrial bioenergetics dysfunction and cardiac hypertrophy.
Collapse
Affiliation(s)
- Lingyue Zou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Binjing Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Lilin Xiong
- Department of Environmental Health, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, 210003, China
| | - Yan Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Wenjing Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xiaoquan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ying Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tingting Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Na Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xiaoru Chang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Changcun Bai
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tianshu Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
109
|
Pryor JT, Cowley LO, Simonds SE. The Physiological Effects of Air Pollution: Particulate Matter, Physiology and Disease. Front Public Health 2022; 10:882569. [PMID: 35910891 PMCID: PMC9329703 DOI: 10.3389/fpubh.2022.882569] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/15/2022] [Indexed: 01/19/2023] Open
Abstract
Nine out of 10 people breathe air that does not meet World Health Organization pollution limits. Air pollutants include gasses and particulate matter and collectively are responsible for ~8 million annual deaths. Particulate matter is the most dangerous form of air pollution, causing inflammatory and oxidative tissue damage. A deeper understanding of the physiological effects of particulate matter is needed for effective disease prevention and treatment. This review will summarize the impact of particulate matter on physiological systems, and where possible will refer to apposite epidemiological and toxicological studies. By discussing a broad cross-section of available data, we hope this review appeals to a wide readership and provides some insight on the impacts of particulate matter on human health.
Collapse
Affiliation(s)
- Jack T. Pryor
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Woodrudge LTD, London, United Kingdom
| | - Lachlan O. Cowley
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stephanie E. Simonds
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- *Correspondence: Stephanie E. Simonds
| |
Collapse
|
110
|
Tian J, Shi H, Wang X, Wu H, Wang C, Liu N, Wang D, Shi D, Zhang H, Zhang Z. The cardiotoxicity of asthmatic rats after traffic-related PM 2.5 and water-soluble components exposure mediated by endoplasmic reticulum stress and autophagy. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:50704-50716. [PMID: 35235115 DOI: 10.1007/s11356-022-19496-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) is closely related to cardiopulmonary diseases; it is known that the respiratory system is related to the cardiovascular system. This study aimed to investigate the toxic effects of traffic-related PM2.5 (TRPM2.5) and water-soluble components (WSC) on hearts of asthmatic rats and explore potential molecular mechanisms. Here, ovalbumin (OVA)-sensitized asthmatic rats were intratracheally instilled with TRPM2.5 and WSC every 3 days in total of eight times. Significant myocardial pathological changes were observed in the TRPM2.5 and WSC group by hematoxylin-eosin (HE) staining. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) results demonstrated TRPM2.5 and WSC aggravated apoptosis of myocardial cells, which may be triggered by endoplasmic reticulum stress (ERS), as manifested by elevated GRP78, CHOP, and caspase-12. Likewise, TRPM2.5 and WSC activated autophagy via upregulation of LC3 and p62 gene and protein expression. In conclusion, TRPM2.5 and WSC may aggravate heart injury in asthmatic rats, possibly through the activation of ERS and autophagy signaling pathway.
Collapse
Affiliation(s)
- Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Hongyan Wu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Dan Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Dongxing Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
111
|
Wang Y, Shen Z, Zhao S, Huang D, Wang X, Wu Y, Pei C, Shi S, Jia N, He Y, Wang Z. Sipeimine ameliorates PM2.5-induced lung injury by inhibiting ferroptosis via the PI3K/Akt/Nrf2 pathway: A network pharmacology approach. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113615. [PMID: 35567927 DOI: 10.1016/j.ecoenv.2022.113615] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) exposure can cause lung injury and a large number of respiratory diseases. Sipeimine is a steroidal alkaloid isolated from Fritillaria roylei which has been associated with anti-inflammatory, antitussive and antiasthmatic properties. In this study, we explored the potential effects of sipeimine against PM2.5-induced lung injury in Sprague Dawley rats. Sipeimine alleviated lung injury caused by PM2.5 and decreased pulmonary edema, inflammation and the levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the bronchoalveolar lavage fluid. In addition, sipeimine upregulated the glutathione (GSH) expression and downregulated the expression of 4-hydroxynonenal (4-HNE), tissue iron and malondialdehyde (MDA). The downregulation of proteins involved in ferroptosis, including nuclear factor E2-related factor 2 (Nrf2), glutathione peroxidase 4 (GPX4), heme oxygenase-1 (HO-1) and solute carrier family 7 member 11 (SLC7A11) was reversed by sipeimine. The administration of RSL3, a potent ferroptosis-triggering agent, blocked the effects of sipeimine. Using network pharmacology, we found that the effects of sipeimine were presumably mediated through the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. A PI3K inhibitor (LY294002) blocked the PI3K/Akt signaling pathway and reversed the effects of sipeimine. Overall, this study suggested that the protective effect of sipeimine against PM2.5-induced lung injury was mainly mediated through the PI3K/Akt pathway, ultimately leading to a reduction in ferroptosis.
Collapse
Affiliation(s)
- Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Yongcan Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue, Chengdu, Sichuan 611137, China.
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China.
| |
Collapse
|
112
|
Li Y, Yang Y, Yang Y. Multifaceted Roles of Ferroptosis in Lung Diseases. Front Mol Biosci 2022; 9:919187. [PMID: 35813823 PMCID: PMC9263225 DOI: 10.3389/fmolb.2022.919187] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/20/2022] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a distinct type of programmed cell death (PCD) that depends on iron and is characterized by the accumulation of intracellular iron, exhaustion of glutathione, deactivation of glutathione peroxidase, and promotion of lipid peroxidation. Recently, accumulated investigations have demonstrated that ferroptosis is strongly correlated with the initiation and development of many lung diseases. In this review, we summarized the contribution of ferroptosis to the pathologic process of lung diseases, namely, obstructive lung diseases (chronic obstructive pulmonary disease, asthma, and cystic fibrosis), interstitial lung diseases (pulmonary fibrosis of different causes), pulmonary diseases of vascular origin (ischemia-reperfusion injury and pulmonary hypertension), pulmonary infections (bacteria, viruses, and fungi), acute lung injury, acute respiratory distress syndrome, obstructive sleep apnea, pulmonary alveolar proteinosis, and lung cancer. We also discussed the therapeutic potential of targeting ferroptosis for these lung diseases.
Collapse
Affiliation(s)
- Yi Li
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Yang
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Yongfeng Yang
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yongfeng Yang,
| |
Collapse
|
113
|
Shi F, Zhang Z, Cui H, Wang J, Wang Y, Tang Y, Yang W, Zou P, Ling X, Han F, Liu J, Chen Q, Liu C, Cao J, Ao L. Analysis by transcriptomics and metabolomics for the proliferation inhibition and dysfunction through redox imbalance-mediated DNA damage response and ferroptosis in male reproduction of mice and TM4 Sertoli cells exposed to PM 2.5. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113569. [PMID: 35512470 DOI: 10.1016/j.ecoenv.2022.113569] [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: 01/30/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Sertoli cells play a pivotal role in the complex spermatogenesis process. This study aimed to investigate the effects of PM2.5 on Sertoli cells using the TM4 cell line and a real time whole-body PM2.5 exposure mouse model, and further explore the underlying mechanisms through the application of metabolomics and transcriptomics. The results in vivo and in vitro showed that PM2.5 reduced Sertoli cells number in seminiferous tubules and inhibited cell proliferation. PM2.5 exposure also induced Sertoli cell dysfunction by increasing androgen binding protein (ABP) concentration, reducing the blood-testis barrier (BTB)-related protein expression, and decreasing glycolysis capacity and lactate production. The results of transcriptomics, metabolomics, and integrative analysis of multi-omics in the TM4 Sertoli cells revealed the activation of xenobiotic metabolism, and the disturbance of glutathione and purine metabolism after PM2.5 exposure. Further tests verified the reduced GSH/GSSG ratio and the elevation of xanthine oxidase (XO) activity in the PM2.5-exposed TM4 cells, indicating that excessive reactive oxygen species (ROS) was generated via metabolic disorder caused by PM2.5. Moreover, the redox imbalance was proved by the increase in the mitochondrial ROS level, superoxide dismutase (SOD) and catalase (CAT) activity, as well as the activation of the Nrf2 antioxidative pathway. Further study found that the redox imbalance caused by PM2.5 induced DNA damage response and cell cycle arrest. Additionally, PM2.5 induced ferroptosis through iron overload and lipid peroxidation. Taken all together, our study provided new insights for understanding proliferation inhibition and dysfunction of TM4 Sertoli cells exposed to PM2.5 via metabolic disorder and redox imbalance-mediated DNA damage response and ferroptosis.
Collapse
Affiliation(s)
- Fuquan Shi
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhonghao Zhang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Haonan Cui
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jiankang Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yimeng Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Tang
- Institution of Health and Family Planning Supervision of Wei'yang District of Xi'an City, Xi'an 710016, China
| | - Wang Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Peng Zou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xi Ling
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Fei Han
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jinyi Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Cuiqing Liu
- School of Basic Medical Sciences and Public Health, Joint China-US Research Center for Environment and Pulmonary Diseases, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
114
|
Guo X, Lin Y, Lin Y, Zhong Y, Yu H, Huang Y, Yang J, Cai Y, Liu F, Li Y, Zhang QQ, Dai J. PM2.5 induces pulmonary microvascular injury in COPD via METTL16-mediated m6A modification. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 303:119115. [PMID: 35259473 DOI: 10.1016/j.envpol.2022.119115] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) exposure is a significant cause of chronic obstructive pulmonary disease (COPD), but the detailed mechanisms involved in COPD remain unclear. In this study, we established PM2.5-induced COPD rat models and showed that PM2.5 induced pulmonary microvascular injury via accelerating vascular endothelial apoptosis, increasing vascular permeability, and reducing angiogenesis, thereby contributing to COPD development. Moreover, microvascular injury in COPD was validated by measurements of plasma endothelial microparticles (EMPs) and serum VEGF in COPD patients. We then performed m6A sequencing, which confirmed that altered N6-methyladenosine (m6A) modification was induced by PM2.5 exposure. The results of a series of experiments demonstrated that the expression of methyltransferase-like protein 16 (METTL16), an m6A regulator, was upregulated in PM2.5-induced COPD rats, while the expression of other regulators did not differ upon PM2.5-induction. To clarify the regulatory effect of METTL16-mediated m6A modification induced by PM2.5 on pulmonary microvascular injury, cell apoptosis, permeability, and tube formation, the m6A level in METTL16-knockdown pulmonary microvascular endothelial cells (PMVECs) was evaluated, and the target genes of METTL16 were identified from a set of the differentially expressed and m6A-methylated genes associated with vascular injury and containing predicted sites of METTL16 methylation. The results showed that Sulfatase 2 (Sulf2) and Cytohesin-1 (Cyth1) containing the predicted METTL16 methylation sites, exhibited higher m6A methylation and were downregulated after PM2.5 exposure. Further studies demonstrated that METTL16 may regulate Sulf2 expression via m6A modification and thereby contribute to PM2.5-induced microvascular injury. These findings not only provide a better understanding of the role played by m6A modification in PM2.5-induced microvascular injury, but also identify a new therapeutic target for COPD.
Collapse
Affiliation(s)
- Xiaolan Guo
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yuyin Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yingnan Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yue Zhong
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Hongjiao Yu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yibin Huang
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Jingwen Yang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Qingyuan, 511500, China
| | - Ying Cai
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Qingyuan, 511500, China
| | - FengDong Liu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yuanyuan Li
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China
| | - Qian-Qian Zhang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianwei Dai
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Center for Reproductive Medicine, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510000, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Qingyuan, 511500, China; State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
115
|
Hu P, Xu Y, Jiang Y, Huang J, Liu Y, Wang D, Tao T, Sun Z, Liu Y. The mechanism of the imbalance between proliferation and ferroptosis in pulmonary artery smooth muscle cells based on the activation of SLC7A11. Eur J Pharmacol 2022; 928:175093. [PMID: 35700835 DOI: 10.1016/j.ejphar.2022.175093] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic, progressive pulmonary vascular disease. Pulmonary vascular remodelling (PVR) is one of the main pathological features of PAH. The main cause of PVR is cell death inhibition and excessive proliferation in pulmonary artery smooth muscle cells (PASMCs), which are also affected by oxidative stress. Ferroptosis is a newly identified form of cell death, which is associated with oxidative damage. It depends on the excessive accumulation of lipid peroxides and reactive oxygen species (ROS) in cells. Solute carrier family 7 member 11 (SLC7A11) is a subunit of the cystine/glutamate antiporter system Xc-, which inhibits ferroptosis by eliminating ROS through the promotion of GSH synthesis in cancer cells. However, very few studies exist on the relationship between ferroptosis and SLC7A11 in PAH. In this study, SLC7A11 was up-regulated in Sugen5416/hypoxia-induced PAH rats and patients with PAH. Moreover, SLC7A11 inhibited ferroptosis and promoted proliferation by overexpressing SLC7A11 in PASMCs. Additionally, ubiquitin aldehyde binding 1 (OTUB1), the main regulator of SLC7A11 stability, was involved in the ferroptosis and proliferation of PASMCs. Furthermore, erastin induced ferroptosis by inhibiting SLC7A11 and glutathione peroxidase 4 (GPX4) expressions in vivo and in vitro, suggesting that the continuous proliferation in hypoxic PASMCs could be reversed by erastin. Therefore, this study identifies novel targets and new research directions regarding PAH pathogenesis and treatment.
Collapse
Affiliation(s)
- Panpan Hu
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China
| | - Yi Xu
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China; Department of Pharmacy, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222061, China
| | - Yanjiao Jiang
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China
| | - Jie Huang
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China
| | - Yi Liu
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China
| | - Dapeng Wang
- Department of Intensive Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ting Tao
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China
| | - Zengxian Sun
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China; Department of Pharmacy, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222061, China
| | - Yun Liu
- Department of Pharmacy, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222061, China; Department of Pharmacy, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222061, China.
| |
Collapse
|
116
|
Assessing the neurotoxicity of airborne nano-scale particulate matter in human iPSC-derived neurons using a transcriptomics benchmark dose model. Toxicol Appl Pharmacol 2022; 449:116109. [DOI: 10.1016/j.taap.2022.116109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022]
|
117
|
Ferroptosis is involved in the benzene-induced hematotoxicity in mice via iron metabolism, oxidative stress and NRF2 signaling pathway. Chem Biol Interact 2022; 362:110004. [DOI: 10.1016/j.cbi.2022.110004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 12/27/2022]
|
118
|
Yalamanchili J, Hennigan CJ, Reed BE. Precipitation of aqueous transition metals in particulate matter during the dithiothreitol (DTT) oxidative potential assay. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2022; 24:762-772. [PMID: 35388859 DOI: 10.1039/d2em00005a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transition metals in particulate matter (PM) are hypothesized to have enhanced toxicity based on their oxidative potential (OP). The acellular dithiothreitol (DTT) assay is widely used to measure the OP of PM and its chemical components. In our prior study, we showed that the DTT assay (pH 7.4, 0.1 M phosphate buffer, 37 °C) provides favorable thermodynamic conditions for precipitation of multiple metals present in PM. This study utilizes multiple techniques to characterize the precipitation of aqueous metals present at low concentrations in the DTT assay. Metal precipitation was identified using laser particle light scattering analysis, direct chemical measurement of aqueous metal removal, and microscopic imaging. Experiments were run with aqueous metals from individual metal salts and a well-characterized urban PM standard (NIST SRM-1648a, Urban Particulate Matter). Our results demonstrated rapid precipitation of metals in the DTT assay. Metal precipitation was independent of DTT but dependent on metal concentration. Metal removal in the chemically complex urban PM samples exceeded the thermodynamic predictions and removal seen in single metal salt experiments, suggesting co-precipitation and/or adsorption may have occurred. These results have broad implications for other acellular assays that study PM metals using phosphate buffer, and subsequently, the PM toxicity inferred from these assays.
Collapse
Affiliation(s)
- Jayashree Yalamanchili
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, USA.
| | - Christopher J Hennigan
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, USA.
| | - Brian E Reed
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, USA.
| |
Collapse
|
119
|
Yuan W, Xia H, Xu Y, Xu C, Chen N, Shao C, Dai Z, Chen R, Tao A. The role of ferroptosis in endothelial cell dysfunction. Cell Cycle 2022; 21:1897-1914. [PMID: 35579940 DOI: 10.1080/15384101.2022.2079054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a form of iron-dependent cell death caused by an excessive accumulation of reactive oxygen species and lipid peroxidation. The importance of ferroptosis in the occurrence and progression of various diseases is gradually being recognized; however, the exact biological effects and potential mechanisms of endothelial cell ferroptosis remain unclear. The endothelium forms the innermost layer of the blood vessels and lymphatic vessels. It acts as an important functional interface, responds to various pathological stimuli and causes endothelial dysfunction. Here, we review recent findings to elucidate the role of ferroptosis in endothelial cells under different pathophysiologic settings.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hao Xia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yao Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chong Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Nan Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aibin Tao
- Department of Cardiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
120
|
Lin H, Chen M, Gao Y, Wang Z, Jin F. Tussilagone protects acute lung injury from PM2.5 via alleviating Hif-1α/NF-κB-mediated inflammatory response. ENVIRONMENTAL TOXICOLOGY 2022; 37:1198-1210. [PMID: 35112795 PMCID: PMC9303425 DOI: 10.1002/tox.23476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/07/2021] [Accepted: 01/16/2022] [Indexed: 05/06/2023]
Abstract
Environmental pollution, especially particulate matter in the air, is a serious threat to human health. Long-term inhalation of particulate matter with a diameter < 2.5 μm (PM2.5) induced irreversible respiratory and lung injury. However, it is not clear whether temporary exposure to massive PM2.5 would result in epithelial damage and lung injury. More importantly, it is urgent to clarify the mechanisms of PM2.5 cytotoxicity and develop a defensive and therapeutic approach. In this study, we demonstrated that temporary exposure with PM2.5 induced lung epithelial cell apoptosis via promoting cytokines expression and inflammatory factors secretion. The cytotoxicity of PM2.5 could be alleviated by tussilagone (TSL), which is a natural compound isolated from the flower buds of Tussilago farfara. The mechanism study indicated that PM2.5 promoted the protein level of Hif-1α by reducing its degradation mediated by PHD2 binding, which furtherly activated NF-κB signaling and inflammatory response. Meanwhile, TSL administration facilitated the interaction of the Hif-1α/PHD2 complex and restored the Hif-1α protein level increased by PM2.5. When PHD2 was inhibited in epithelial cells, the protective function of TSL on PM2.5 cytotoxicity was attenuated and the expression of cytokines was retrieved. Expectedly, the in vivo study also suggested that temporary PM2.5 exposure led to acute lung injury. TSL treatment could effectively relieve the damage and decrease the expression of inflammatory cytokines by repressing Hif-1α level and NF-κB activation. Our findings provide a new therapeutic strategy for air pollution-related respiratory diseases, and TSL would be a potential preventive medicine for PM2.5 cytotoxicity.
Collapse
Affiliation(s)
- Hongwei Lin
- Respiration Department of Tangdu Hospital, Air force Military Medical UniversityXi'anChina
| | - Min Chen
- Respiration Department of Tangdu Hospital, Air force Military Medical UniversityXi'anChina
| | - Yanjun Gao
- Respiration Department of Tangdu Hospital, Air force Military Medical UniversityXi'anChina
| | - Zaiqiang Wang
- Respiration Department of Tangdu Hospital, Air force Military Medical UniversityXi'anChina
| | - Faguang Jin
- Respiration Department of Tangdu Hospital, Air force Military Medical UniversityXi'anChina
| |
Collapse
|
121
|
Han D, Yao Y, Chen L, Miao Z, Xu S. Apigenin ameliorates di(2-ethylhexyl) phthalate-induced ferroptosis: The activation of glutathione peroxidase 4 and suppression of iron intake. Food Chem Toxicol 2022; 164:113089. [PMID: 35500696 DOI: 10.1016/j.fct.2022.113089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/28/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely artificial persistent organic pollutant, the contamination of which infiltrates daily human life from many aspects, imperceptibly causing damage to multiple organs in the body, including the liver. Apigenin (APG) is widely distributed in vegetables and fruits and can relieve or prevent the injuries caused by exogenous chemicals through various pharmacological effects, such as antioxidant effects. To investigate the mechanism of DEHP-induced liver injury and the antagonistic effects of APG, we treated AML12 cells with 1 mM DEHP and/or APG. Ultrastructural morphology analysis indicated that DEHP induced typical ferroptosis-like damage. In addition, we found that DEHP exposure induced ferroptosis by enhancing reactive oxygen species (ROS) levels, disrupting iron homeostasis and lipid peroxidation, and regulating the expression of ferroptosis-related genes. Notably, supplementation with APG significantly inhibited these abnormal changes, and molecular docking further showed evidence of the activating effects of APG ligand on glutathione peroxidase 4 (GPX4). These results demonstrated that the protective effects of APG on DEHP-induced ferroptosis were achieved by activating GPX4 and suppressing intracellular iron accumulation. This information not only adds to DEHP toxicological data but also provides a basis for the practical application of APG.
Collapse
Affiliation(s)
- Dongxu Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yujie Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Lu Chen
- College of Animal Science, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Zhiying Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
122
|
Lai TC, Chen YC, Cheng HH, Lee TL, Tsai JS, Lee IT, Peng KT, Lee CW, Hsu LF, Chen YL. Combined exposure to fine particulate matter and high glucose aggravates endothelial damage by increasing inflammation and mitophagy: the involvement of vitamin D. Part Fibre Toxicol 2022; 19:25. [PMID: 35351169 PMCID: PMC8966234 DOI: 10.1186/s12989-022-00462-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Background Cardiovascular diseases (CVDs) are related to particulate matter (PM2.5) exposure. Researchers have not clearly determined whether hyperglycemia, a hallmark of diabetes, exacerbates PM2.5-induced endothelial damage. Thus, this study aimed to investigate the combined effects of PM2.5 and high glucose on endothelial damage. Results Here, we treated human umbilical vein endothelial cells (HUVECs) with 30 mM high glucose and 50 μg/mL PM (HG + PM) to simulate endothelial cells exposed to hyperglycemia and air pollution. First, we showed that HUVECs exposed to PM under high glucose conditions exhibited significant increases in cell damage and apoptosis compared with HUVECs exposed to PM or HG alone. In addition, PM significantly increased the production of reactive oxygen species (ROS) in HUVECs and mitochondria treated with HG and decreased the expression of superoxide dismutase 1 (SOD1), a free radical scavenging enzyme. The coexposure group exhibited significantly increased ROS production in cells and mitochondria, a lower mitochondrial membrane potential, and increased levels of the autophagy-related proteins p62, microtubule-associated protein 1 light chain 3β (LC3B), and mitophagy-related protein BCL2 interacting protein 3 (Bnip3). Moreover, autophagosome-like structures were observed in the HG + PM group using transmission electron microscopy. The expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) were also increased through the JNK/p38 signaling pathway in the HG + PM group. As a ROS scavenger, vitamin D treatment effectively protected cells under HG and PM conditions by increasing cell viability, reducing mitochondrial ROS production, and suppressing the formation of mitophagy and inflammation. Furthermore, diabetes was induced in mice by administering streptozotocin (STZ). Mice were treated with PM by intratracheal injection. Vitamin D effectively alleviated oxidative stress, mitophagy, and inflammation in the aortas of mice treated with STZ and PM. Conclusion Taken together, simultaneous exposure to PM and high glucose exerts significant harmful effects on endothelial cells by inducing ROS production, mitophagy, and inflammation, while vitamin D reverses these effects. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00462-1.
Collapse
Affiliation(s)
- Tsai-Chun Lai
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yu-Chen Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Hui-Hua Cheng
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Tzu-Lin Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Jaw-Shiun Tsai
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan.,Center for Complementary and Integrated Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Kuo-Ti Peng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, 613016, Taiwan.,College of Medicine, Chang Gung University, Guishan District, Taoyuan City, 333323, Taiwan
| | - Chiang-Wen Lee
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, 613016, Taiwan. .,Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, 613016, Taiwan. .,Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan.
| | - Lee-Fen Hsu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County, 613016, Taiwan. .,Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, 613016, Taiwan.
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan.
| |
Collapse
|
123
|
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
|
124
|
Shang Y, Xue W, Kong J, Chen Y, Qiu X, An X, Li Y, Wang H, An J. Ultrafine black carbon caused mitochondrial oxidative stress, mitochondrial dysfunction and mitophagy in SH-SY5Y cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 813:151899. [PMID: 34838543 DOI: 10.1016/j.scitotenv.2021.151899] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/02/2021] [Accepted: 11/19/2021] [Indexed: 06/13/2023]
Abstract
Exposure to ambient ultrafine black carbon (uBC, with aerodynamic diameter less than 100 nm) is associated with many neurodegenerative diseases. Oxidative stress is the predominantly reported neurotoxic effects caused by uBC exposure. Mitochondrion is responsible for production of majority of ROS in cells and mitochondrial dysfunction is closely related to adverse nervous outcomes. Mitophagy is an important cellular process to eliminate dysfunctional or damaged mitochondria. However, the mechanisms that modulate mitophagy and mitochondrial dysfunction initiated by uBC remain to be elucidated. The purpose of this study was to investigate how mitochondrial oxidative stress regulated mitochondrial dysfunction and mitophagy in human neuroblastoma cell line (SH-SY5Y) after uBC treatment. RNA interference was further applied to explore the roles of mitophagy in mitochondrial dysfunction. We found uBC triggered cell apoptosis via ROS-mitochondrial apoptotic pathway. The uBC also caused serious mitochondrial damage and respiratory dysfunction, indicated by the abnormalities in mitochondrial division and fusion related proteins, decreased mitochondria number and ATP level. Increased PTEN induced putative kinase 1 (PINK1) and Parkin protein levels and the autolysosome numbers suggested uBC could promote Pink1/Parkin-dependent mitophagy process in SH-SY5Y cells. Mitophagy inhibition could reserve mitochondria number and ATP activity, but not fusion and division related protein levels in SH-SY5Y cells exposed to uBC. Administration of a mitochondria-targeted antioxidant (mitoquinone) significantly eliminated uBC caused apoptosis, mitochondrial dysfunction and mitophagy. Our data suggested mitochondrial oxidative stress regulated uBC induced mitochondrial dysfunction and PINK1/Parkin-dependent mitophagy. PINK1/Parkin-dependent mitophagy probably participated in regulating uBC caused mitochondrial dysfunction but not by controlling mitochondrial fusion and division related proteins. Our results may provide some new insights and evidences to understand the mechanisms of neurotoxicity induced by uBC.
Collapse
Affiliation(s)
- Yu Shang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China; State Environmental Protection Key Laboratory of Formation and Prevention of Urban Air Pollution Complex, Shanghai Academy of Environment Sciences, Shanghai 200233, China
| | - Wanlei Xue
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Jiexing Kong
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yingjun Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Xinghua Qiu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Xingqin An
- State Key Laboratory of Severe Weather & Key Laboratory of Atmospheric Chemistry of CMA, Chinese Academy of Meteorological Sciences, Beijing 100081, China
| | - Yi Li
- State Key Laboratory of Severe Weather & Key Laboratory of Atmospheric Chemistry of CMA, Chinese Academy of Meteorological Sciences, Beijing 100081, China
| | - Hongli Wang
- State Environmental Protection Key Laboratory of Formation and Prevention of Urban Air Pollution Complex, Shanghai Academy of Environment Sciences, Shanghai 200233, China
| | - Jing An
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
125
|
Guo Y, Lu C, Hu K, Cai C, Wang W. Ferroptosis in Cardiovascular Diseases: Current Status, Challenges, and Future Perspectives. Biomolecules 2022; 12:biom12030390. [PMID: 35327582 PMCID: PMC8945958 DOI: 10.3390/biom12030390] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases (CVDs) are still a major cause of global mortality and disability, seriously affecting people’s lives. Due to the severity and complexity of these diseases, it is important to find new regulatory mechanisms to treat CVDs. Ferroptosis is a new kind of regulatory cell death currently being investigated. Increasing evidence showed that ferroptosis plays an important role in CVDs, such as in ischemia/reperfusion injury, heart failure, cardiomyopathy, and atherosclerosis. Protecting against CVDs by targeting ferroptosis is a promising approach; therefore, in this review, we summarized the latest regulatory mechanism of ferroptosis and the current studies related to each CVD, followed by critical perspectives on the ferroptotic treatment of CVDs and the future direction of this intriguing biology.
Collapse
Affiliation(s)
- Yi Guo
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Chanjun Lu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Ke Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
| | - Weici Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.L.); (K.H.); (C.C.)
- Correspondence: ; Tel.: +86-180-7170-5166
| |
Collapse
|
126
|
Chen GH, Song CC, Pantopoulos K, Wei XL, Zheng H, Luo Z. Mitochondrial oxidative stress mediated Fe-induced ferroptosis via the NRF2-ARE pathway. Free Radic Biol Med 2022; 180:95-107. [PMID: 35045311 DOI: 10.1016/j.freeradbiomed.2022.01.012] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a regulated form of cell death induced by iron (Fe)-dependent lipid peroxidation. At present, the underlying molecular mechanisms remain elusive. Herein, we hypothesized that mitochondria and the NRF2 (transcription factor nuclear factor E2-related factor 2) are potential mediators of ferroptosis, considering their well-established involvement in the oxidative stress pathway. We found that a high iron diet increased hepatic iron content and promoted glutathione (GSH) depletion, lipid peroxidation and oxidative stress. Dietary iron overload also decreased mRNA and protein expression levels of glutathione peroxidase 4 (GPX4) and cystine-glutamate antiporter (SLC7A11), and increased mRNA and protein expression of acyl-CoA synthetase long-chain family member 4 (ACSL4), which are all markers of ferroptosis. Consistent with ferroptosis, iron overload promoted lipid peroxidation and the generation of mitochondrial reactive oxygen species (ROS), and decreased the mitochondrial membrane potential (MMP). Pre-treatment with deferoxamine mesylate (DFO, an iron chelator) alleviated ROS generation and lipid peroxidation, indicating a causative link between iron overload and lipid peroxidation. Suppression of mitochondrial oxidative stress attenuated ferroptosis. Experiments with HEK293T cells revealed that Fe-induced ferroptosis involved direct inhibition of NRF2 binding to antioxidant response elements (AREs) within the promoters of the gpx4 and slc7a11 genes, which in turn induced transcriptional silencing. In conclusion, our study provided a direct link between mitochondrial oxidative stress and ferroptosis via the NRF2-ARE pathway.
Collapse
Affiliation(s)
- Guang-Hui Chen
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan, 430070, China
| | - Chang-Chun Song
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan, 430070, China
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, H3T 1E2, Quebec, Canada
| | - Xiao-Lei Wei
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan, 430070, China
| | - Hua Zheng
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan, 430070, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan, 430070, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
127
|
Yang Z, Shi J, Chen L, Fu C, Shi D, Qu H. Role of Pyroptosis and Ferroptosis in the Progression of Atherosclerotic Plaques. Front Cell Dev Biol 2022; 10:811196. [PMID: 35186925 PMCID: PMC8850398 DOI: 10.3389/fcell.2022.811196] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Pyroptosis is a special way of programmed cell death which is dependent on the activation of cysteinyl aspartate specific proteinase 1 (Caspase-1) and Caspase-4/5/11. Ferroptosis is an iron-dependent cell death that characterized by the intra-cellular lipid peroxidation-mediated membrane damage. Pyroptosis or ferroptosis in macrophages, smooth muscle cells, and vascular endothelial cells are believed to be closely related to the progression of atherosclerotic plaques. Therefore, we discuss the role of pyroptosis and ferroptosis in the development of atherosclerotic plaques and may provide new strategies for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Zhen Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Junhe Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
| | - Li Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| |
Collapse
|
128
|
Chen Y, He Y, Wei X, Jiang DS. Targeting regulated cell death in aortic aneurysm and dissection therapy. Pharmacol Res 2022; 176:106048. [PMID: 34968685 DOI: 10.1016/j.phrs.2021.106048] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/11/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023]
Abstract
Regulated cell death (RCD) is a basic biological phenomenon associated with cell and tissue homeostasis. Recent studies have enriched our understanding of RCD, and many novel cell death types, such as ferroptosis and pyroptosis, have been discovered and defined. Aortic aneurysm and dissection (AAD) is a life-threatening condition, but the pathogenesis remains largely unclear. A series of studies have indicated that the death of smooth muscle cells, endothelial cells and inflammatory cells participates in the development of AAD and that corresponding interventions could alleviate disease progression. Many treatments against cell death have been used to impede the process of AAD in vitro and in vivo, which provides strategies to protect against this condition. In this review, we focus on various types of regulated cell death and provide a framework of their roles in AAD, and the information contributes to further exploration of the molecular mechanisms of AAD.
Collapse
Affiliation(s)
- Yue Chen
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi He
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
129
|
Liu T, Bao R, Wang Q, Hao W, Liu Y, Chang S, Wang M, Li Y, Liu Z, Sun Y. SiO 2-induced ferroptosis in macrophages promotes the development of pulmonary fibrosis in silicosis models. Toxicol Res (Camb) 2022; 11:42-51. [PMID: 35237410 PMCID: PMC8882780 DOI: 10.1093/toxres/tfab105] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 09/06/2023] Open
Abstract
Silicosis is a devastating disease that, without effective treatment, endangers the health of miners. Therefore, studies exploring the pathogenesis of SiO2-induced pulmonary fibrosis are necessary to develop treatments for silicosis. Although macrophages are known to play a pivotal role in SiO2-induced pulmonary fibrosis, the underlying mechanism remains unknown. Here, we explored whether ferroptosis was involved in SiO2-induced pulmonary fibrosis. To this end, C57BL/6 mice and mouse macrophage (RAW264.7) cells and mouse lung fibroblast (MLF) cells were subjected to iron content, cell viability, enzyme-linked immunosorbent assay, immunofluorescence staining, histological, western blotting, quantitative reverse transcription-PCR, reactive oxygen species, and lipid peroxidation analysis. In vivo, SiO2 was found to damage the lung alveolar structure, cause infiltration of inflammatory cells, and facilitate fibrosis. Additionally, it increased the iron concentration and lipid peroxidation as well as altered the expression of ferroptosis-related genes and the mitochondrial morphology in macrophages. In vitro, ferroptosis occurred in SiO2-treated RAW264.7 cells, which showed iron overload, lipid peroxidation, and gene alterations. Furthermore, ferrostatin-1 (Fer-1) attenuated ferroptosis in SiO2-treated RAW264.7 cells by inhibiting lipid peroxidation and cell death and regulating ferroptosis-related genes expression, in addition to attenuating the secretion of pro-fibrotic cytokines and fibrosis. Collectively, SiO2 induces ferroptosis in macrophages, which leads to the secretion of pro-fibrotic cytokines and fibrosis.
Collapse
Affiliation(s)
- Taiyang Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Rui Bao
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Qiushi Wang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Wei Hao
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yaoyang Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Sirong Chang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Meng Wang
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yuanyuan Li
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Zhihong Liu
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| | - Yue Sun
- School of Public Health and Management, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
- NHC KEY Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 75000, Ningxia, China
| |
Collapse
|
130
|
Shan H, Li X, Ouyang C, Ke H, Yu X, Tan J, Chen J, Wang C, Zhang L, Tang Y, Yu L, Li W. Salidroside prevents PM2.5-induced BEAS-2B cell apoptosis via SIRT1-dependent regulation of ROS and mitochondrial function. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113170. [PMID: 35026589 DOI: 10.1016/j.ecoenv.2022.113170] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/28/2021] [Accepted: 01/03/2022] [Indexed: 06/14/2023]
Abstract
PM2.5 is a harmful air pollutant currently threatening public health. It has been closely linked to increased morbidity of bronchial asthma and lung cancer worldwide. Salidroside (Sal), an active component extracted from Rhodiola rosea, has been reported to ameliorate the progression of asthma. However, there are few studies on the protective effect of salidroside on PM2.5-induced bronchial epithelial cell injury, and the related molecular mechanism is not clear. Here, we aimed to explore the protective effect and related mechanism of Sal on PM2.5 bronchial injury. We chose 50 μg/mL PM2.5 for 24 h as a PM2.5-induced cell damage model. After that BEAS-2B cells were pretreated with 40, 80, 160 µM Sal for 24 h and then exposed to 50 μg/mL PM2.5 for 24 h. We found that Sal pretreatment significantly inhibited the decrease of cell viability induced by PM2.5. Sal was effective in preventing PM2.5-induced apoptotic features, including Ca2+ overload, the cleavages of caspase 3, and the increases in levels of caspase 9 and Bcl-2-associated X protein (Bax), ultimately, Sal significantly inhibited PM2.5-induced apoptosis. Sal improved mitochondrial membrane potential, inhibited the release of cytochrome c from the mitochondria to cytoplasm. Sal alleviated ROS production, decreased the level of MDA, prevented the reduction of CAT, SOD and GSH-Px and increased the expression of NF-E2-related factor 2 (Nrf2), HO-1 and superoxide dismutase 1 (SOD1) in cells exposed to PM2.5. Furthermore, Sal improved the decrease of SIRT1 and PGC-1 α expression levels caused by PM2.5. In addition, inhibition of SIRT1 by EX527 (SIRT1 inhibitor) reversed the protective effects of Sal, including the decrease of ROS level, the increase of membrane potential level and the decrease of apoptosis level. Thus, Sal may be regarded as a potential drug to prevent PM2.5-induced apoptosis of bronchial epithelial cells and other diseases with similar pathological mechanisms.
Collapse
Affiliation(s)
- Hui Shan
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China
| | - Xiaohong Li
- Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China; Department of Nutrition and Food Hygiene, School of Public Health, Weifang Medical College, Weifang, China
| | - Chuan Ouyang
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China
| | - Hongyang Ke
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China
| | - Xiaoli Yu
- Key Laboratory of health inspection and quarantine, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China
| | - Jinfeng Tan
- Weifang environmental monitoring station, Weifang, China
| | - Junhao Chen
- Key Laboratory of health inspection and quarantine, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China
| | - Chunping Wang
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China
| | - Liping Zhang
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China
| | - Yunfeng Tang
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China
| | - Li Yu
- School of basic medicine, Weifang Medical University, Weifang, China
| | - Wanwei Li
- Department of Environmental Hygiene, School of Public Health, Weifang Medical College, Weifang, China; Public Health Demonstration Center, School of Public Health, Weifang Medical College, Weifang, China.
| |
Collapse
|
131
|
Yang N, Wang Q, Ding B, Gong Y, Wu Y, Sun J, Wang X, Liu L, Zhang F, Du D, Li X. Expression profiles and functions of ferroptosis-related genes in the placental tissue samples of early- and late-onset preeclampsia patients. BMC Pregnancy Childbirth 2022; 22:87. [PMID: 35100981 PMCID: PMC8805258 DOI: 10.1186/s12884-022-04423-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/24/2022] [Indexed: 01/21/2023] Open
Abstract
Background The accumulation of reactive oxygen species (ROS) resulting from upregulated levels of oxidative stress is commonly implicated in preeclampsia (PE). Ferroptosis is a novel form of iron-dependent cell death instigated by lipid peroxidation that likely plays an important role in PE pathogenesis. This study aimed to investigate the expression profiles and functions of ferroptosis-related genes (FRGs) in early-onset preeclampsia (EOPE) and late-onset preeclampsia (LOPE). Methods Gene expression data and clinical information were downloaded from the Gene Expression Omnibus (GEO) database. The “limma” R package was used to screen differentially expressed genes. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein–protein interaction (PPI) network analyses were conducted to investigate the bioinformatics functions and molecular interactions of significantly different FRGs. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to verify the expression of hub FRGs in PE. Results A total of 4215 differentially expressed genes (DEGs) were identified between EOPE and preterm cases while 556 DEGs were found between LOPE and term controls. Twenty significantly different FRGs were identified in EOPE subtypes, while only 3 FRGs were identified in LOPE subtypes. Functional enrichment analysis revealed that the differentially expressed FRGs were mainly involved in EOPE and enriched in hypoxia- and iron-related pathways, such as the response to hypoxia, iron homeostasis and iron ion binding process. PPI network analysis and verification by RT-qPCR resulted in the identification of the following five FRGs of interest: FTH1, HIF1A, FTL, MAPK8 and PLIN2. Conclusions EOPE and LOPE have distinct underlying molecular mechanisms, and ferroptosis may be mainly implicated in the pathogenesis of EOPE. Further studies are necessary for deeper inquiry into placental ferroptosis and its role in the pathogenesis of EOPE. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04423-6.
Collapse
Affiliation(s)
- Nana Yang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Qianghua Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Biao Ding
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Yingying Gong
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Yue Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Junpei Sun
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Xuegu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Lei Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Feng Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Danli Du
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China.
| | - Xiang Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China.
| |
Collapse
|
132
|
Leng Y, Luo X, Yu J, Jia H, Yu B. Ferroptosis: A Potential Target in Cardiovascular Disease. Front Cell Dev Biol 2022; 9:813668. [PMID: 35127725 PMCID: PMC8811289 DOI: 10.3389/fcell.2021.813668] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/31/2021] [Indexed: 12/22/2022] Open
Abstract
Ferroptosis is a new form of regulatory cell death characterized by iron-dependent and intracellular lipid peroxidation. Ferroptosis can be divided into two stages. The first stage is iron overload in the cell, which generates a large amount of reactive oxygen species through the Fenton reaction, and the second stage results from an imbalance of the intracellular antioxidant system. Excessive phospholipid hydroperoxides cannot be removed by reduction reactions, as this could destroy the cell membrane structure and interfere with mitochondrial function, eventually leading to ferroptosis of the cell. Cardiovascular diseases have gradually become the leading cause of death in modern society. The relationship between ferroptosis and the occurrence and progression of cardiovascular disease has become a research hotspot in recent years. In this review, we summarize the mechanism of ferroptosis and its specific role in cardiovascular disease.
Collapse
Affiliation(s)
- Yanlong Leng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
| | - Xing Luo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
| | - Jiaying Yu
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haibo Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
- *Correspondence: Haibo Jia,
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
| |
Collapse
|
133
|
Zhai X, Wang J, Sun J, Xin L. PM 2.5 induces inflammatory responses via oxidative stress-mediated mitophagy in human bronchial epithelial cells. Toxicol Res (Camb) 2022; 11:195-205. [PMID: 35237424 PMCID: PMC8882786 DOI: 10.1093/toxres/tfac001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) is a ubiquitous air pollutant, and it has been reported to be closely associated with lung inflammatory injury. In this study, the potential molecular mechanisms underlying PM2.5-induced cellular inflammation in human bronchial epithelial (BEAS-2B) cells were investigated. MATERIALS AND METHODS Ambient PM2.5 particulates from Suzhou, China, were collected and re-suspended in ultrapure water. Cellular damages, characterized by oxidative stress, mitochondrial injury, and inflammatory cytokine production, were determined in 24 h PM2.5-treated BEAS-2B cells with or without 3-methyladenine (3-MA; autophagy inhibitor) pretreatment. Biomarkers related to oxidative damage, inflammatory injury and autophagy signaling pathways were also measured. RESULTS Uptake of PM2.5 in BEAS-2B cells induced cellular oxidative damage, mitochondrial injury, and inflammatory responses as indicated by a significant decrease in GSH/GSSG ratio, increased MDA content, dilated mitochondria with loss and rupture of crista, and production of inflammatory cytokines. Activation of Nrf-2/TXNIP-mediated NF-κB and Bnip3L/NIX-dependent mitophagy signaling pathways, as well as accumulation of autophagosomes and autolysosomes, were also observed. A 6 h pretreatment of 3-MA increased PM2.5-induced oxidative damage and cellular inflammation as indicated by increasing protein levels of HO-1, TXNIP, Bnip3L/NIX and IL-8 gene expression. CONCLUSIONS PM2.5 induced cellular inflammatory injury by oxidative stress, mitochondrial dysfunction, and mitophagy initiation. Although induction of Bnip3L/NIX-mediated mitophagy in BEAS-2B cells appeared to confer protection in response to PM2.5, dysfunction of autophagic flux may be a critical contributor to defective mitophagy and cellular inflammatory response.
Collapse
Affiliation(s)
| | | | - Jiaojiao Sun
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Lili Xin
- Corresponding author: School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
134
|
Hao R, Ge J, Song X, Li F, Sun-Waterhouse D, Li D. Cadmium induces ferroptosis and apoptosis by modulating miR-34a-5p/Sirt1axis in PC12 cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:41-51. [PMID: 34558789 DOI: 10.1002/tox.23376] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/06/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
Cadmium (Cd) is a potent neurotoxic metal present in the environment and food. In this study, CdCl2 (2 or 4 μM) induced cytotoxicity and neurotoxicity in PC12 cells, causing decreases in cell viability and NEP protein expression and increase in p-tau protein expression. For the first time, CdCl2 -initiated injury was found to result from the induction of not only apoptosis but also ferroptosis, as evidenced by the increased iron content, ROS production, and mitochondrial membrane potential along with changes in the expressions of iron death-related genes (FTH1, GPX4, ASCL4, PTGS2, and NOX1) and levels of caspase9, Bax, and Bcl-2 proteins. The molecular mechanisms leading to apoptosis and ferroptosis at least included the participation of the miR-34a-5p/Sirt1 axis, in which miR-34a-5p promoted CdCl2 -induced neurotoxicity through targeting Sirt1. Knocking out miR-34a-5p attenuated CdCl2 -induced damage of PC12 cells, cytotoxicity and neurotoxicity. This research provides the underlying molecular mechanisms of CdCl2 -induced damage and asserts the role of miRNAs as critical regulators.
Collapse
Affiliation(s)
- Rili Hao
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
| | - Junlin Ge
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
| | - Xinyu Song
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
| | - Feng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Taian, China
| |
Collapse
|
135
|
Cao W, Wang X, Li J, Yan M, Chang CH, Kim J, Jiang J, Liao YP, Tseng S, Kusumoputro S, Lau C, Huang M, Han P, Lu P, Xia T. NLRP3 inflammasome activation determines the fibrogenic potential of PM 2.5 air pollution particles in the lung. J Environ Sci (China) 2022; 111:429-441. [PMID: 34949371 DOI: 10.1016/j.jes.2021.04.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 06/14/2023]
Abstract
Airborne fine particulate matter (PM2.5) is known to cause respiratory inflammation such as chronic obstructive pulmonary disease and lung fibrosis. NLRP3 inflammasome activation has been implicated in these diseases; however, due to the complexity in PM2.5 compositions, it is difficult to differentiate the roles of the components in triggering this pathway. We collected eight real-life PM2.5 samples for a comparative analysis of their effects on NLRP3 inflammasome activation and lung fibrosis. In vitro assays showed that although the PM2.5 particles did not induce significant cytotoxicity at the dose range of 12.5 to 100 µg/mL, they induced potent TNF-α and IL-1β production in PMA differentiated THP-1 human macrophages and TGF-β1 production in BEAS-2B human bronchial epithelial cells. At the dose of 100 µg/mL, PM2.5 induced NLRP3 inflammasome activation by inducing lysosomal damage and cathepsin B release, leading to IL-1β production. This was confirmed by using NLRP3- and ASC-deficient cells as well as a cathepsin B inhibitor, ca-074 ME. Administration of PM2.5 via oropharyngeal aspiration at 2 mg/kg induced significant TGF-β1 production in the bronchoalveolar lavage fluid and collagen deposition in the lung at 21 days post-exposure, suggesting PM2.5 has the potential to induce pulmonary fibrosis. The ranking of in vitro IL-1β production correlates well with the in vivo total cell count, TGF-β1 production, and collagen deposition. In summary, we demonstrate that the PM2.5 is capable of inducing NLRP3 inflammasome activation, which triggers a series of cellular responses in the lung to induce fibrosis.
Collapse
Affiliation(s)
- Wei Cao
- Translational Medical Center, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou 450007, China.
| | - Xiang Wang
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States.
| | - Jiulong Li
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States
| | - Ming Yan
- Basic Medical College, Zhengzhou University, Zhengzhou 450001, China
| | - Chong Hyun Chang
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States
| | - Joshua Kim
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles 90095, CA, United States
| | - Jinhong Jiang
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States
| | - Shannon Tseng
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles 90095, CA, United States
| | - Sydney Kusumoputro
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles 90095, CA, United States
| | - Candice Lau
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles 90095, CA, United States
| | - Marissa Huang
- Department of Integrative Biology and Physiology, University of California, Los Angeles 90095, CA, United States
| | - Pengli Han
- Translational Medical Center, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou 450007, China
| | - Pengju Lu
- Translational Medical Center, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou 450007, China
| | - Tian Xia
- Translational Medical Center, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou 450007, China; Division of NanoMedicine, Department of Medicine, University of California, Los Angeles 90095, CA, United States.
| |
Collapse
|
136
|
Ishihara N, Okuda T, Hagino H, Oguro A, Tani Y, Okochi H, Tokoro C, Fujii-Kuriyama Y, Itoh K, Vogel CF, Ishihara Y. Involvement of polycyclic aromatic hydrocarbons and endotoxin in macrophage expression of interleukin-33 induced by exposure to particulate matter. J Toxicol Sci 2022; 47:201-210. [PMID: 35527008 PMCID: PMC9469799 DOI: 10.2131/jts.47.201] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Air pollutants are important factors that contribute to the development and/or exacerbation of allergic inflammation accompanied by asthma, but experimental evidence still needs to be collected. Interleukin 33 (IL-33) is closely involved in the onset and progression of asthma. In this study, we examined the effects of particulate matter (PM) on IL-33 expression in macrophages. PM2.5 collected in Yokohama, Japan by the cyclone device significantly induced IL-33 expression in human THP-1 macrophages, and the induction was clearly suppressed by pretreatment with the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the Toll-like receptor 4 (TLR4) antagonist TAK-242. PM2.5-induced IL-33 expression was significantly attenuated in AhR-knockout or TLR4-mutated macrophages, suggesting an important role of polycyclic aromatic hydrocarbons (PAHs) and endotoxin in IL-33 stimulation. PM samples derived from tunnel dust slightly but significantly induced IL-33 expression, while road dust PM did not affect IL-33 expression. The PAH concentration in tunnel dust was higher than that in road dust. Tunnel dust or road dust PM contained less endotoxin than PM2.5 collected in Yokohama. These data suggest that the potency of IL-33 induction could depend on the concentration of PAHs as well as endotoxin in PMs. Caution regarding PAHs and endotoxin levels in air pollutants should be taken to prevent IL-33-induced allergic inflammation.
Collapse
Affiliation(s)
- Nami Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan
| | - Tomoaki Okuda
- Faculty of Science and Technology, Keio University, Kanagawa, 223-8522, Japan
| | - Hiroyuki Hagino
- Japan Automobile Research Institute, Ibaraki, 305-0822, Japan
| | - Ami Oguro
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan
| | - Yuto Tani
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Hiroshi Okochi
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Chiharu Tokoro
- School of Creative Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Yoshiaki Fujii-Kuriyama
- Medical Research Institute, Molecular Epidemiology, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, 769-2101, Japan
| | - Christoph F.A. Vogel
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, 95616, USA,Center for Health and the Environment, University of California, Davis, Davis, CA, 95616, USA
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8521, Japan,Center for Health and the Environment, University of California, Davis, Davis, CA, 95616, USA
| |
Collapse
|
137
|
Ren JY, Yin BW, Li X, Zhu SQ, Deng JL, Sun YT, Zhang ZA, Guo ZH, Pei HT, Zhang F, Li RQ, Chen FG, Ma YX. Sesamin attenuates PM 2.5-induced cardiovascular injury by inhibiting ferroptosis in rats. Food Funct 2021; 12:12671-12682. [PMID: 34825691 DOI: 10.1039/d1fo02913d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Objective: This study aimed to elucidate the pharmacological effects of sesamin (Ses) and its mechanism of action towards PM2.5-induced cardiovascular injuries. Method: Forty Sprague Dawley (SD) rats were randomly divided into five groups: a saline control group; a PM2.5 exposure group; and low-, middle-, and high-dose Ses pretreatment groups. The SD rats were pretreated with different concentrations of Ses for 21 days. Afterward, the rats were exposed to ambient PM2.5 by intratracheal instillation every other day for a total of three times. The levels of inflammatory markers, including tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), and interleukin-6 (IL-6), and indicators related to oxidative responses, such as total superoxide dismutase (SOD), reduced glutathione (GSH), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA), were measured in the blood and heart. The expression of ferroptosis-related proteins in heart tissues was determined via western blot and immunohistochemistry. Results: Ses pretreatment substantially ameliorated cardiovascular injuries in rats as evidenced by the decrease in the pathological score and collagen area. The decreased levels of SOD, GSH, and GSH-Px in the heart and serum were inhibited by Ses. In addition, Ses not only notably increased the activity of antioxidant enzymes but also reduced the levels of MDA, CK, LDH, CK-MB, IL-6, TNF-α, IL-1β, and IL-6. Furthermore, Ses pretreatment upregulated the expression levels of GPX4, SLC7A11, TFRC, and FPN1 and inhibited the expression levels of FTH1 and FTL. Conclusion: Ses pretreatment could ameliorate PM2.5-induced cardiovascular injuries perhaps by inhibiting ferroptosis. Therefore, Ses pretreatment may be a novel strategy for the prevention and treatment of PM2.5-induced cardiovascular injury.
Collapse
Affiliation(s)
- Jing-Yi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Bo-Wen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Xiang Li
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Si-Qi Zhu
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jin-Liang Deng
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yi-Ting Sun
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhen-Ao Zhang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zi-Hao Guo
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Huan-Ting Pei
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Fan Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Rui-Qiang Li
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| | - Feng-Ge Chen
- Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, 050017, China
| | - Yu-Xia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
| |
Collapse
|
138
|
Kwag Y, Ye S, Oh J, Lee DW, Yang W, Kim Y, Ha E. Direct and Indirect Effects of Indoor Particulate Matter on Blood Indicators Related to Anemia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182412890. [PMID: 34948498 PMCID: PMC8701383 DOI: 10.3390/ijerph182412890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022]
Abstract
Exposure to indoor particulate matter (PM) is a potential risk factor that increases systemic inflammation and affects erythropoiesis. This study investigated the association between exposure to indoor PM and blood indicators related to anemia (BIRA) in housewives. Indoor PM and blood folate status are important factors in the risk of anemia. This was a housewife cohort study; we recruited 284 housewives in Seoul and Ulsan, Republic of Korea. Indoor exposure to PM2.5 and PM10 was measured by gravimetric analysis and sensors. We investigated the BIRA, such as hemoglobin (Hb), hematocrit, mean corpuscular volume (MCV), mean corpuscular Hb (MCH), and mean corpuscular Hb concentration (MCHC). Statistical analysis was performed by multiple linear regression model and mediation analysis. The association between BIRA and PM was assessed by multiple linear regression models fitted by mediation analyses. The increase in the level of indoor PM2.5 was associated with a decrease in MCV (Beta coefficient (B): −0.069, Standard error (SE): 0.022) and MCH (B: −0.019, SE: 0.009) in gravimetric measurements. The increase in the level of indoor PM2.5 was associated with a decrease in Hb (B: −0.024, SE: 0.011), hematocrit (B: −0.059, SE: 0.033), and MCV (B: −0.081, SE: 0.037) and MCH (B: −0.037, SE: 0.012) in sensor measurements (PM2.5-Lag10). Further, we identified a serum folate-mediated PM effect. The indoor PM exposure was significantly associated with decreased Hb, MCV, and MCH in housewives. Taken together, our data show that exposure to indoor PM is a risk factor for anemia in housewives. Blood folate concentration can be a mediating factor in the effect of indoor PM on BIRA. Therefore, folate intake should be recommended to prevent anemia in housewives. Moreover, indoor PM exposure should be managed.
Collapse
Affiliation(s)
- Youngrin Kwag
- Department of Environmental Medicine, School of Medicine, Ewha Womans University, Seoul KS013, Korea; (Y.K.); (J.O.)
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul KS013, Korea
| | - Shinhee Ye
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Incheon KS006, Korea;
| | - Jongmin Oh
- Department of Environmental Medicine, School of Medicine, Ewha Womans University, Seoul KS013, Korea; (Y.K.); (J.O.)
| | - Dong-Wook Lee
- Department of Preventive Medicine, College of Medicine, Seoul National University, Seoul KS013, Korea;
| | - Wonho Yang
- Department of Occupational Health, Daegu Catholic University, Gyeongsan-si KS002, Korea;
| | - Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan KS016, Korea
- Correspondence: (Y.K.); (E.H.)
| | - Eunhee Ha
- Department of Environmental Medicine, School of Medicine, Ewha Womans University, Seoul KS013, Korea; (Y.K.); (J.O.)
- Correspondence: (Y.K.); (E.H.)
| |
Collapse
|
139
|
Yu S, Jia J, Zheng J, Zhou Y, Jia D, Wang J. Recent Progress of Ferroptosis in Lung Diseases. Front Cell Dev Biol 2021; 9:789517. [PMID: 34869391 PMCID: PMC8635032 DOI: 10.3389/fcell.2021.789517] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/31/2021] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death due to iron-dependent excess accumulation of lipid peroxides and differs from other programmed cell deaths in morphological and biochemical characteristics. The process of ferroptosis is precisely regulated by iron metabolism, lipid metabolism, amino acid metabolism, and numerous signaling pathways, and plays a complex role in many pathophysiological processes. Recent studies have found that ferroptosis is closely associated with the development and progression of many lung diseases, including acute lung injury, pulmonary ischemia-reperfusion injury, lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. Here, we present a review of the main regulatory mechanisms of ferroptosis and its research progress in the pathogenesis and treatment of lung diseases, with the aim of providing new ideas for basic and clinical research of lung-related diseases.
Collapse
Affiliation(s)
- Shangjiang Yu
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqiu Jia
- Department of Pediatrics, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
140
|
Tang X, Li Z, Yu Z, Li J, Zhang J, Wan N, Zhang J, Cao J. Effect of curcumin on lung epithelial injury and ferroptosis induced by cigarette smoke. Hum Exp Toxicol 2021; 40:S753-S762. [PMID: 34787501 DOI: 10.1177/09603271211059497] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cigarette smoke (CS)-caused ferroptosis was involved in the pathogenesis of COPD, but the role of ferroptosis in lung epithelial injury and inflammation is not clear. Rats were treated with CS or CUR and BEAS-2B cells were exposed to CS extract (CSE), ferrostatin-1 (Fer-1), deferoxamine (DFO), or CUR to detect reactive oxygen species (ROS) accumulation, lipid peroxidation, iron overload, and ferroptosis-related protein, which were the characteristic changes of ferroptosis. Compared with the control group, CSE-treated BEAS-2B cells had more cell death, higher cytotoxicity, and lower cell viability. The infiltration of inflammatory cell around the bronchi in the CS group of rats was more than that in the normal group. Meanwhile, CSE/CS elevated the levels of interleukin-6 and tumor necrosis factor-α in BEAS-2B cells and bronchoalveolar lavage fluid of rats. Besides, accumulative ROS and depleted glutathione was observed in vitro. In BEAS-2B cells and lung tissues of rats, CSE/CS increased malondialdehyde and iron; down-regulated solute carrier family 7, glutathione peroxidase 4, and ferritin heavy chain levels; and up-regulated transferrin receptor level. These changes were rescued by pretreatment of Fer-1 or DFO in vitro, and mitigated by CUR in vitro and in vivo. Collectively, this study reveals that ferroptosis was involved in lung epithelial cell injury and inflammation induced by CS, and CUR may alleviate CS-induced injury, inflammation, and ferroptosis of lung epithelial cell.
Collapse
Affiliation(s)
- Xin Tang
- Department of Oncology, 605425Chongqing University Cancer Hospital, Chongqing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Zhenyu Li
- Department of Pathology, 605425Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Zhi Yu
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jinna Li
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jinbang Zhang
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Nansheng Wan
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jie Cao
- Department of Respiratory and Critical Care Medicine, 117865Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
141
|
Zhao T, Qi W, Yang P, Yang L, Shi Y, Zhou L, Ye L. Mechanisms of cardiovascular toxicity induced by PM 2.5: a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:65033-65051. [PMID: 34617228 DOI: 10.1007/s11356-021-16735-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
An increasing number of studies have shown that exposure to particulate matter with a diameter ≤ 2.5 μm (PM2.5) could affect the onset and development of cardiovascular diseases. To explore the underlying mechanisms, the studies conducted in vitro investigations using different cell lines. In this review, we examined recently published reports cited by PubMed or Web of Science on the topic of cardiovascular toxicity induced by PM2.5 that carried the term in vitro. Here, we summarized the suggested mechanisms of PM2.5 leading to adverse effects and cardiovascular toxicity including oxidative stress; the increase of vascular endothelial permeability; the injury of vasomotor function and vascular reparative capacity in vascular endothelial cell lines; macrophage polarization and apoptosis in macrophage cell lines; and hypermethylation and apoptosis in the AC16 cell line and the related signaling pathways, which provided a new research direction of cardiovascular toxicity of PM2.5.
Collapse
Affiliation(s)
- Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China
| | - Wen Qi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China
| | - Pan Yang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China
- Jilin Provincial Center for Disease Control and Prevention (Jilin Provincial Institute of Public Health), Changchun, China
| | - Liwei Yang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China
| | - Yanbin Shi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China.
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, China.
| |
Collapse
|
142
|
Chen G, Han Y, Zhang H, Tu W, Zhang S. Radiotherapy-Induced Digestive Injury: Diagnosis, Treatment and Mechanisms. Front Oncol 2021; 11:757973. [PMID: 34804953 PMCID: PMC8604098 DOI: 10.3389/fonc.2021.757973] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is one of the main therapeutic methods for treating cancer. The digestive system consists of the gastrointestinal tract and the accessory organs of digestion (the tongue, salivary glands, pancreas, liver and gallbladder). The digestive system is easily impaired during radiotherapy, especially in thoracic and abdominal radiotherapy. In this review, we introduce the physical classification, basic pathogenesis, clinical characteristics, predictive/diagnostic factors, and possible treatment targets of radiotherapy-induced digestive injury. Radiotherapy-induced digestive injury complies with the dose-volume effect and has a radiation-based organ correlation. Computed tomography (CT), MRI (magnetic resonance imaging), ultrasound (US) and endoscopy can help diagnose and evaluate the radiation-induced lesion level. The latest treatment approaches include improvement in radiotherapy (such as shielding, hydrogel spacers and dose distribution), stem cell transplantation and drug administration. Gut microbiota modulation may become a novel approach to relieving radiogenic gastrointestinal syndrome. Finally, we summarized the possible mechanisms involved in treatment, but they remain varied. Radionuclide-labeled targeting molecules (RLTMs) are promising for more precise radiotherapy. These advances contribute to our understanding of the assessment and treatment of radiation-induced digestive injury.
Collapse
Affiliation(s)
- Guangxia Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Yi Han
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Haihan Zhang
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Wenling Tu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Shuyu Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.,West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
143
|
Zou L, Xiong L, Wu T, Wei T, Liu N, Bai C, Huang X, Hu Y, Xue Y, Zhang T, Tang M. NADPH oxidases regulate endothelial inflammatory injury induced by PM 2.5 via AKT/eNOS/NO axis. J Appl Toxicol 2021; 42:738-749. [PMID: 34708887 DOI: 10.1002/jat.4254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 11/11/2022]
Abstract
Fine particulate matter (PM2.5 )-induced detrimental cardiovascular effects have been widely concerned, especially for endothelial cells, which is the first barrier of the cardiovascular system. Among potential mechanisms involved, reactive oxidative species take up a crucial part. However, source of oxidative stress and its relationship with inflammatory response have been rarely studied in PM2.5 -induced endothelial injury. Here, as a key oxidase that catalyzes redox reactions, NADPH oxidase (NOX) was investigated. Human umbilical vein endothelial cells (EA.hy926) were exposed to Standard Reference Material 1648a of urban PM2.5 for 24 h, which resulted in NOX-sourced oxidative stress, endothelial dysfunction, and inflammation induction. These are manifested by the up-regulation of NOX, increase of superoxide anion and hydrogen peroxide, elevated endothelin-1 (ET-1) and asymmetric dimethylarginine (ADMA) level, reduced nitric oxide (NO) production, and down-regulation of phosphorylation of endothelial NO synthase (eNOS) with increased levels of inducible NO synthase, as well as the imbalance between tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor 1 (PAI-1), and changes in the levels of pro-inflammatory and anti-inflammatory factors. However, administration of NOX1/4 inhibitor GKT137831 alleviated PM2.5 -induced elevated endothelial dysfunction biomarkers (NO, ET-1, ADMA, iNOS, and tPA/PAI-1), inflammatory factors (IL-1β, IL-10, and IL-18), and adhesion molecules (ICAM-1, VCAM-1, and P-selectin) and also passivated NOX-dependent AKT and eNOS phosphorylation that involved in endothelial activation. In summary, PM2.5 -induced NOX up-regulation is the source of ROS in EA.hy926, which activated AKT/eNOS/NO signal response leading to endothelial dysfunction and inflammatory damage in EA.hy926 cells.
Collapse
Affiliation(s)
- Lingyue Zou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Lilin Xiong
- Department of Environmental Health, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Tianshu Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Tingting Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Na Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Changcun Bai
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xiaoquan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yuanyuan Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
144
|
Zou HX, Qiu BQ, Lai SQ, Zhou XL, Gong CW, Wang LJ, Yuan MM, He AD, Liu JC, Huang H. Iron Metabolism and Idiopathic Pulmonary Arterial Hypertension: New Insights from Bioinformatic Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5669412. [PMID: 34722766 PMCID: PMC8556088 DOI: 10.1155/2021/5669412] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022]
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a rare vascular disease with a poor prognosis, and the mechanism of its development remains unclear. Further molecular pathology studies may contribute to a comprehensive understanding of IPAH and provide new insights into diagnostic markers and potential therapeutic targets. Iron deficiency has been reported in 43-63% of patients with IPAH and is associated with reduced exercise capacity and higher mortality, suggesting that dysregulated iron metabolism may play an unrecognized role in influencing the development of IPAH. In this study, we explored the regulatory mechanisms of iron metabolism in IPAH by bioinformatic analysis. The molecular function of iron metabolism-related genes (IMRGs) is mainly enriched in active transmembrane transporter activity, and they mainly affect the biological process of response to oxidative stress. Ferroptosis and fluid shear stress and atherosclerosis pathways may be the critical pathways regulating iron metabolism in IPAH. We further identified 7 key genes (BCL2, GCLM, MSMO1, SLC7A11, SRXN1, TSPAN5, and TXNRD1) and 5 of the key genes (BCL2, MSMO1, SLC7A11, TSPAN5, and TXNRD1) as target genes may be regulated by 6 dysregulated miRNAs (miR-483-5p, miR-27a-3p, miR-27b-3p, miR-26b-5p, miR-199a-5p, and miR-23b-3p) in IPAH. In addition, we predicted potential IPAH drugs-celastrol and cinnamaldehyde-that target iron metabolism based on our results. These results provide insights for further definition of the role of dysregulated iron metabolism in IPAH and contribute to a deeper understanding of the molecular mechanisms and potential therapeutic targets of IPAH.
Collapse
Affiliation(s)
- Hua-Xi Zou
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Bai-Quan Qiu
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Song-Qing Lai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xue-Liang Zhou
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Cheng-Wu Gong
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Li-Jun Wang
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming-Ming Yuan
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - An-Di He
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ji-Chun Liu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Huang Huang
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
145
|
Wang Y, Xiong L, Yao Y, Ma Y, Liu Q, Pang Y, Tang M. The involvement of DRP1-mediated caspase-1 activation in inflammatory response by urban particulate matter in EA.hy926 human vascular endothelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117369. [PMID: 34182399 DOI: 10.1016/j.envpol.2021.117369] [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: 11/15/2020] [Revised: 04/13/2021] [Accepted: 05/06/2021] [Indexed: 06/13/2023]
Abstract
Atmospheric particulate matter (PM) has been reported to be closely related to cardiovascular adverse events. However, the underlying mode of action remains to be elucidated. Previous studies have documented that PM induces mitochondrial damage and inflammation, the relation between these two biological outcomes is still unclear though. In this study, we used EA.hy926 human vascular endothelial cells and a standard PM, PM SRM1648a to study the potential effects of mitochondrial dysfunction on endothelial inflammatory responses. As a result, PM SRM1648a changes mitochondrial morphology and interrupts mitochondrial dynamics with a persistent tendency of fission in a dose-dependent manner. Additionally, the caspase-1/IL-1β axis is involved in inflammatory responses but not cell pyroptosis in EA.hy926 cells following the exposure to PM SRM1648a. The activation of caspase-1 has implications in inflammation but not pyroptosis, because caspase-1-dependent pyroptosis is not the main modality of cell death in PM SRM1648a-treated EA.hy926 cells. With regard to the association between mitochondrial damage and inflammation in the case of particle stimulation, DRP1-mediated mitochondrial fission is responsible for inflammatory responses as a result of caspase-1 activation. The current study showed that PM SRM1648a has the ability to disturb mitochondrial dynamics, and trigger endothelial inflammation via DRP1/caspase-1/IL-1β regulatory pathway. In a conclusion, mitochondrial fission enables EA.hy926 cells to facilitate caspase-1 activation in response to PM SRM1648a, which is a crucial step for inflammatory reaction in vascular endothelial cells.
Collapse
Affiliation(s)
- Yan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Lilin Xiong
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China; Department of Environmental Health, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, 210003, China
| | - Yongshuai Yao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qing Liu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yanting Pang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
146
|
Liu Z, Xia X, Lv X, Song E, Song Y. Iron-bearing nanoparticles trigger human umbilical vein endothelial cells ferroptotic responses by promoting intracellular iron level. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117345. [PMID: 34004477 DOI: 10.1016/j.envpol.2021.117345] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 06/12/2023]
Abstract
Iron-bearing nanoparticles (IBNPs) were abundant in particulate matter (PM). Due to their high reactivity, IBNPs were considered hazardous to human health, however, their toxic mode-of-action(s) are highly unclear. Ferroptosis is a novel programmed cell death (PCD) that highly associated with intracellular iron. However, the pro-ferroptotic effect of IBNPs has not been characterized. To this end, we ought to investigate whether and how IBNPs (synthetic γ-Fe2O3 and Fe3O4 NPs were selected as the model compounds) are involved in ferroptosis. We found that human umbilical vein endothelial cells (HUVECs) phagocytized large qualities of γ-Fe2O3 and Fe3O4 NPs, resulting in increased intracellular iron level. We further observed the disrupted cystine/glutamate reverse transporter (System Xc-) and glutathione peroxidase 4 (GPX4) signaling in γ-Fe2O3 and Fe3O4 NPs-challenged HUVECs. γ-Fe2O3 and Fe3O4 NPs could also cause mitochondrial fusion and fission dysregulation, activate lipid peroxidation and iron metabolism-related genes in a P53-dependent manner. Together, the ferroptotic activity of IBNPs should be acknowledged for the risk assessment of PM associated health effects.
Collapse
Affiliation(s)
- Zixuan Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaomin Xia
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Xuying Lv
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| |
Collapse
|
147
|
Wang Y, Ma Y, Yao Y, Liu Q, Pang Y, Tang M. Ambient particulate matter triggers defective autophagy and hijacks endothelial cell renewal through oxidative stress-independent lysosomal impairment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 286:117295. [PMID: 34438478 DOI: 10.1016/j.envpol.2021.117295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/20/2021] [Accepted: 04/30/2021] [Indexed: 06/13/2023]
Abstract
Ambient particulate matter (APM) has been authenticated to exert hazards on human vascular endothelial cells, including abnormal autophagy. However, the potential reasons for autophagosome accumulation are still obscure. Since autophagy is a dynamic process, it is imperative to systemically consider the autophagic induction combined with its degradation to reflect realistic scenarios. Therefore, in the current study, different exposure durations were initially employed for the detection of autophagic marker proteins to assess the dynamic autophagic state preliminarily. Additionally, LC3 turn-over and autophagic flux assays were used to determine the specific cause of LC3II upregulation in EA.hy926 human vascular endothelial cells by a type of standard urban particulate matter, PM SRM1648a. As a result, PM SRM1648a stimulates excess autophagic vacuoles in EA. hy926 cells, in which the underlying causes are probably different at varying incubation endpoints. Intriguingly, LC3II upregulation was due to the intensifying autophagic initiation after 6 h of exposure, whereas as exposure period was extended to 24 h, overloaded autophagic vacuoles were attributed to the defective autophagy. Mechanistically, PM SRM1648a damages EA. hy926 cells by inducing lysosomal disequilibrium and resultant autophagic malfunction which are not directly mediated by oxidative stress. These data indicate that appropriate maintenance of lysosomal function and autophagic flux is probably a protective measure against APM-induced endothelial cell damage.
Collapse
Affiliation(s)
- Yan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yongshuai Yao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qing Liu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yanting Pang
- 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
|
148
|
Huang F, Yang R, Xiao Z, Xie Y, Lin X, Zhu P, Zhou P, Lu J, Zheng S. Targeting Ferroptosis to Treat Cardiovascular Diseases: A New Continent to Be Explored. Front Cell Dev Biol 2021; 9:737971. [PMID: 34527678 PMCID: PMC8435746 DOI: 10.3389/fcell.2021.737971] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, including cardiomyopathy, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, vascular injury, stroke, and arrhythmia, are correlated with cardiac and vascular cell death. Ferroptosis is a novel form of non-apoptotic regulated cell death which is characterized by an iron-driven accumulation of lethal lipid hydroperoxides. The initiation and execution of ferroptosis are under the control of several mechanisms, including iron metabolism, glutamine metabolism, and lipid peroxidation. Recently, emerging evidence has demonstrated that ferroptosis can play an essential role in the development of various cardiovascular diseases. Recent researches have shown the ferroptosis inhibitors, iron chelators, genetic manipulations, and antioxidants can alleviate myocardial injury by blocking ferroptosis pathway. In this review, we systematically described the mechanisms of ferroptosis and discussed the role of ferroptosis as a novel therapeutic strategy in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ronghua Yang
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuefeng Lin
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Lu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
149
|
Hu T, Zhu P, Liu Y, Zhu H, Geng J, Wang B, Yuan G, Peng Y, Xu B. PM2.5 induces endothelial dysfunction via activating NLRP3 inflammasome. ENVIRONMENTAL TOXICOLOGY 2021; 36:1886-1893. [PMID: 34173703 DOI: 10.1002/tox.23309] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/02/2021] [Accepted: 06/13/2021] [Indexed: 05/05/2023]
Abstract
PM2.5 (particulate matter <2.5 μm in diameter) is proven to contribute to the development of atherosclerosis. Endothelial cell dysfunction is the initial step of atherosclerosis. The underlying mechanisms of endothelial cell damage exposed to PM2.5 are still obscure. In our study, PM2.5 was administrated to C57BL/6 male mice by intranasal instillation for 2 weeks. Human umbilical vein endothelial cells (HUVECs) were also treated with PM2.5 to evaluate the adverse effect in vitro. The immunohistochemical staining of aortas showed that the expressions of proinflammatory cytokines and endothelial adhesion markers were significantly increased in PM2.5-exposed mice than that in saline-exposed mice. In vitro, PM2.5 could inhibit HUVECs viability and impair cell migration in a concentration-dependent manner. Besides, PM2.5 exposure downregulated eNOS expression while upregulated reactive oxygen species (ROS) levels. Mechanistically, PM2.5 activated the NLRP3 inflammasome in HUVECs while knockdown of NLRP3 could effectively reverse the downregulation of eNOS expression and production of ROS after PM2.5 exposure. In summary, our data showed that PM2.5 could cause endothelial dysfunction, and probably via NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of Cardiology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Ping Zhu
- Department of Endocrinology, Huai'an Hospital of Huai'an City, Huai'an, Jiangsu, China
| | - Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Haoran Zhu
- Department of Cardiology, Huai'an First People's Hospital Clinical College of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Jin Geng
- Department of Cardiology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Bingjian Wang
- Department of Cardiology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Guoliang Yuan
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, Shuyang, Jiangsu, China
| | - Yuzhu Peng
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
150
|
Manivannan J, Sundaresan L. Systems level insights into the impact of airborne exposure on SARS-CoV-2 pathogenesis and COVID-19 outcome - A multi-omics big data study. GENE REPORTS 2021; 25:101312. [PMID: 34401607 PMCID: PMC8358088 DOI: 10.1016/j.genrep.2021.101312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/14/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that led to more than 800,00 deaths and continues to be a major threat worldwide. The scientific community has been studying the risk factors associated with SARS-CoV-2 infection and pathogenesis. Recent studies highlight the possible contribution of atmospheric air pollution, specifically particulate matter (PM) exposure as a co-factor in COVID-19 severity. Hence, meaningful translation of suitable omics datasets of SARS-CoV-2 infection and PM exposure is warranted to understand the possible involvement of airborne exposome on COVID-19 outcome. Publicly available transcriptomic data (microarray and RNA-Seq) related to COVID-19 lung biopsy, SARS-CoV-2 infection in epithelial cells and PM exposure (lung tissue, epithelial and endothelial cells) were obtained in addition with proteome and interactome datasets. System-wide pathway/network analysis was done through appropriate software tools and data resources. The primary findings are; 1. There is no robust difference in the expression of SARS-CoV-2 entry factors upon particulate exposure, 2. The upstream pathways associated with upregulated genes during SARS-CoV-2 infection considerably overlap with that of PM exposure, 3. Similar pathways were differentially expressed during SARS-CoV-2 infection and PM exposure, 4. SARS-CoV-2 interacting host factors were predicted to be associated with the molecular impact of PM exposure and 5. Differentially expressed pathways during PM exposure may increase COVID-19 severity. Based on the observed molecular mechanisms (direct and indirect effects) the current study suggests that airborne PM exposure has to be considered as an additional co-factor in the outcome of COVID-19.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- COVID-19
- COVID19, coronavirus disease 2019
- CTSB, cathepsin B
- CTSL, cathepsin L
- DEG, differentially expressed genes
- GEO, Gene Expression Omnibus
- GSEA, gene set enrichment analysis
- IL-17, interleukin-17
- Microarray
- Omics
- PM, particulate matter
- PPAR, peroxisome proliferator-activated receptors
- PPI, protein-protein interaction
- PTM, post-translational modification
- Particulate matter
- Pathway analysis
- Proteome
- RNA-seq
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- TLR, Toll-like receptor
- TMPRSS2, transmembrane protease, serine 2
- TNF, tumor necrosis factor
- VEGF, vascular endothelial growth factor
- X2K, eXpression2Kinases
Collapse
Affiliation(s)
- Jeganathan Manivannan
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Lakshmikirupa Sundaresan
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|