1
|
Jo YS, Song JW. Air Pollution and Interstitial Lung Disease. Tuberc Respir Dis (Seoul) 2025; 88:45-55. [PMID: 39542009 PMCID: PMC11704737 DOI: 10.4046/trd.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/11/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
This review article explores the multifaceted relationship between air pollution and interstitial lung diseases (ILDs), particularly focusing on idiopathic pulmonary fibrosis, the most severe form of fibrotic ILD. Air pollutants are mainly composed of particulate matter, ozone (O3), nitrogen dioxide (NO2), carbon monoxide (CO), and sulfur dioxide (SO2). They are recognized as risk factors for several respiratory diseases. However, their specific effects on ILDs and related mechanisms have not been thoroughly studied yet. Emerging evidence suggests that air pollutants may contribute to the development and acute exacerbation of ILDs. Longitudinal studies have indicated that air pollution can adversely affect the prognosis of disease by decreasing lung function and increasing mortality. Lots of in vitro, in vivo , and epidemiologic studies have proposed possible mechanisms linking ILDs to air pollution, including inflammation and oxidative stress induced by exposure to air pollutants, which may induce mitochondrial dysfunction, promote cellular senescence, and disrupt normal epithelial repair processes. Despite these findings, effective interventions to mitigate effects of air pollution on ILD are not well established yet. This review emphasizes the urgent need to address air pollution as a key environmental risk factor for ILDs and calls for further studies to clarify its effects and develop preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Yong Suk Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Li Y, Zhong Y, Li C, Han Z, Cui Y, He R, Liu Y, Cui Q, He D, Hu Z, Zhang Q, Bai J. Interleukin-9 promotes EMT-mediated PM 2.5-induced pulmonary fibrosis by activating the STAT3 pathway. Arch Toxicol 2024; 98:4047-4058. [PMID: 39259283 DOI: 10.1007/s00204-024-03864-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
This study investigated the impact of PM2.5 on promoting EMT in PM2.5-induced pulmonary fibrosis (PF) development and explored molecular mechanisms of the IL-9/STAT3/Snail/TWIST1 signaling pathway in PF owing to PM2.5. Four groups of male SD rats were formed: control (0 mg/kg.bw), low (1 mg/kg.bw), medium (5 mg/kg.bw), and high-dose (25 mg/kg.bw) PM2.5 groups. Experimental rats were subjected to PM2.5 exposure via intratracheal instillation, given once weekly for 16 weeks. 24 h after the final exposure, blood, BALF, and lung tissues were collected. Pulmonary epithelial cells underwent cultivation and exposure to varying PM2.5 concentrations with/without inhibitors for 24 h, after which total protein was extracted for relevant protein assays. The findings demonstrated that PM2.5 damaged lung tissue to different degrees and led to PF in rats. Rats subjected to PM2.5 exposure exhibited elevated concentrations of IL-9 protein in both serum and BALF, and elevated levels of IL-9 and its receptor, IL-9R, in lung tissues, compared to control counterparts. Furthermore, PM2.5-exposed groups demonstrated significantly augmented protein levels of p-STAT3, Snail, TWIST1, Vimentin, COL-I, and α-SMA, while displaying notably diminished levels of E-Cadherin compared to control group. The same findings were observed in PM2.5-treated cells. In BEAS-2B cells co-treated with Stattic (STAT3 inhibitor) and PM2.5, the opposite results occurred. Similar results were obtained for cells co-treated with IL-9-neutralizing antibody and PM2.5. Our findings suggest PM2.5 mediates PF development by promoting IL-9 expression, leading to STAT3 phosphorylation and upregulation of Snail and TWIST1 expression, triggering EMT occurrence and progression in lung epithelial cells.
Collapse
Affiliation(s)
- Yuxuan Li
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Yi Zhong
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Chenwen Li
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
- Yongchuan District Center for Disease Control and Prevention, Chongqing, 402160, China
| | - Zhixia Han
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Yan Cui
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Renjiang He
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Yingyi Liu
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Qinlin Cui
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Daping He
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Zhengquan Hu
- Luzhou Ecological Environment Monitoring Center of Sichuan Province, Luzhou, 646000, China.
| | - Qingbi Zhang
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China.
| | - Jun Bai
- Environmental Health Effects and Risk Assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Meng T, He J, Huo Q, Wang Y, Ren Q, Kang Y. Association of Stress Defense System With Fine Particulate Matter Exposure: Mechanism Analysis and Application Prospects. J Appl Toxicol 2024. [PMID: 39538419 DOI: 10.1002/jat.4724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The association between the stress defense system and exposure to fine particulate matter (PM2.5) is a hot topic in the field of environmental health. PM2.5 pollution is an increasingly serious issue, and its impact on health cannot be ignored. The stress defense system is an important biological mechanism for maintaining cell and internal environment homeostasis, playing a crucial role in PM2.5-induced damage and diseases. The association between PM2.5 exposure and activation of the stress defense system has been reported. Moderate PM2.5 exposure rapidly mobilizes the stress defense system, while excessive PM2.5 exposure may exceed its compensatory and coping abilities, resulting in system imbalance and dysfunction that triggers pathological changes in cells and tissues, thereby increasing the risk of chronic diseases, such as respiratory diseases, cardiovascular diseases, and cancer. This detailed review focuses on the composition, function, and regulatory mechanisms of the antioxidant defense system, autophagy system, ubiquitin-proteasome system, and inflammatory response system, which are all components of the stress defiance system. In particular, the influence of PM2.5 exposure on each of these defense systems and their roles in responding to PM2.5-induced damage was investigated to provide an in-depth understanding of the pathogenesis of PM2.5 exposure, accurately assess potential hazards, and formulate prevention and intervention strategies for health damage caused by PM2.5 exposure.
Collapse
Affiliation(s)
- Tao Meng
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
- Doctoral Innovation Station of Shanxi Province, Key Laboratory of TCM Prevention and Treatment of Dementia Disease, The Fifth People's Hospital of Datong, Datong, China
| | - Jing He
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qianru Huo
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yajie Wang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qingchun Ren
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yihui Kang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| |
Collapse
|
4
|
Drake C, Zobl W, Escher SE. Assessment of pulmonary fibrosis using weighted gene co-expression network analysis. FRONTIERS IN TOXICOLOGY 2024; 6:1465704. [PMID: 39512679 PMCID: PMC11540828 DOI: 10.3389/ftox.2024.1465704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
For many industrial chemicals toxicological data is sparse regarding several regulatory endpoints, so there is a high and often unmet demand for NAMs that allow for screening and prioritization of these chemicals. In this proof of concept case study we propose multi-gene biomarkers of compounds' ability to induce lung fibrosis and demonstrate their application in vitro. For deriving these biomarkers we used weighted gene co-expression network analysis to reanalyze a study where the time-dependent pulmonary gene-expression in mice treated with bleomycin had been documented. We identified eight modules of 58 to 273 genes each which were particularly activated during the different phases (inflammatory; acute and late fibrotic) of the developing fibrosis. The modules' relation to lung fibrosis was substantiated by comparison to known markers of lung fibrosis from DisGenet. Finally, we show the modules' application as biomarkers of chemical inducers of lung fibrosis based on an in vitro study of four diketones. Clear differences could be found between the lung fibrosis inducing diketones and other compounds with regard to their tendency to induce dose-dependent increases of module activation as determined using a previously proposed differential activation score and the fraction of differentially expressed genes in the modules. Accordingly, this study highlights the potential use of composite biomarkers mechanistic screening for compound-induced lung fibrosis.
Collapse
|
5
|
Zeng Y, Zhang R, Jiang Y, Li D, Chen L, Dong G, Zhang R, Niu Y, Chen W, Chen S. Interactions between fibroblasts and monocyte-derived cells in chronic lung injuries induced by real-ambient particulate matter exposure. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 899:503807. [PMID: 39326935 DOI: 10.1016/j.mrgentox.2024.503807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Long-term exposure to fine particulate matter (PM2.5) can lead to chronic lung injury, including inflammation, idiopathic pulmonary fibrosis, and cancer. Mesenchymal cells, such as fibroblasts, myeloid-derived suppressor cells (MDSCs), and interstitial macrophages (IMs), contribute to immune regulation in lung, yet their diversity and functions upon long-term exposure to particulate matter (PM) remain inadequately characterized. In this study, we conducted a 16-week real-ambient PM exposure experiment on C57BL/6 J male mice in Shijiazhuang, China. We used single-cell RNA sequencing to analyze the cellular and molecular changes in lung tissues. Notably, we revealed a significant increase in specific fibroblast (ATX+, Col5a1+Meg3+, universal fibroblasts) and monocyte-derived cell subpopulations (monocytic-MDSCs (M-MDSCs), Lyve1loMHC-Ⅱhi IMs, Lyve1hiMHC-Ⅱlo IMs) that exhibited pro-inflammatory and pro-fibrotic functions. These cell subpopulations engaged in immunosuppressive signaling pathways and interactions with various cytokines, shaping a pulmonary microenvironment similar to those associated with cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). This altered immune environment may promote the development of pulmonary fibrosis caused by PM exposure, underscoring the intricate roles of mesenchymal cells in chronic lung injury and highlighting the cancer-causing potential of PM2.5 exposure.
Collapse
Affiliation(s)
- Youjin Zeng
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Zhang
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Jiang
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Guanghui Dong
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujie Niu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Wen Chen
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen Chen
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
6
|
Afthab M, Hambo S, Kim H, Alhamad A, Harb H. Particulate matter-induced epigenetic modifications and lung complications. Eur Respir Rev 2024; 33:240129. [PMID: 39537244 PMCID: PMC11558539 DOI: 10.1183/16000617.0129-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Air pollution is one of the leading causes of early deaths worldwide, with particulate matter (PM) as an emerging factor contributing to this trend. PM is classified based on its physical size, which ranges from PM10 (diameter ≤10 μm) to PM2.5 (≤2.5 μm) and PM0.5 (≤0.5 μm). Smaller-sized PM can move freely through the air and readily infiltrate deep into the lungs, intensifying existing health issues and exacerbating complications. Lung complications are the most common issues arising from PM exposure due to the primary site of deposition in the respiratory system. Conditions such as asthma, COPD, idiopathic pulmonary fibrosis, lung cancer and various lung infections are all susceptible to worsening due to PM exposure. PM can epigenetically modify specific target sites, further complicating its impact on these conditions. Understanding these epigenetic mechanisms holds promise for addressing these complications in cases of PM exposure. This involves studying the effect of PM on different gene expressions and regulation through epigenetic modifications, including DNA methylation, histone modifications and microRNAs. Targeting and manipulating these epigenetic modifications and their mechanisms could be promising strategies for future treatments of lung complications. This review mainly focuses on different epigenetic modifications due to PM2.5 exposure in the various lung complications mentioned above.
Collapse
Affiliation(s)
- Muhammed Afthab
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Shadi Hambo
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Hyunji Kim
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Ali Alhamad
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| | - Hani Harb
- Institute for Medical Microbiology and Virology, University Hospital Dresden, TU Dresden, Dresden, Germany
| |
Collapse
|
7
|
Yang H, Shi P, Li M, Liu S, Mou B, Xia Y, Sun J. Plasma proteome mediate the impact of PM 2.5 on stroke: A 2-step Mendelian randomization study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116624. [PMID: 38908058 DOI: 10.1016/j.ecoenv.2024.116624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The objectives of this study were to measure the mediation effect of plasma proteins and to clarify their mediating role in the relationship between stroke risk and particulate matter 2.5 (PM2.5) exposure. The possible mediating role of plasma proteins on the causative link between PM2.5 exposure and stroke incidence were examined using a two-step Mendelian randomization (MR) approach based on two-sample Mendelian randomization (TSMR). The findings revealed a significant positive causal relationship between PM2.5 exposure and stroke, with an inverse variance weighted odds ratio of 1.219 (95 % CI: 1.002 - 1.482, P < 0.05). Additionally, a positive causal association was identified between PM2.5 exposure and several plasma proteins, including FAM134B, SAP, ITGB7, Elafin, and DCLK3. Among these, FAM134B, ITGB7, Elafin, and DCLK3 also demonstrated a positive causal association with stroke, whereas only SAP was found to be negatively causally associated with stroke. Remarkably, four plasma proteins, namely DCLK3, FAM134B, Elafin, and ITGB7, were identified as mediators, accounting for substantial proportions (14.5 %, 13.6 %, 11.1 %, and 9.9 %) of the causal association between PM2.5 and stroke. These results remained robust across various sensitivity analyses. Consequently, the study highlights the significant and independent impact of PM2.5 on stroke risk and identifies specific plasma proteins as potential targets for preventive interventions against PM2.5-induced stroke.
Collapse
Affiliation(s)
- Huajie Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Peng Shi
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Mingzheng Li
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Shuailing Liu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Baohua Mou
- First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Yinglan Xia
- Zhejiang Greentown Cardiovascular Hospital, Hangzhou 310000, China
| | - Jiaxing Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
8
|
Yu SL, Koo H, Kang Y, Jeon HJ, Kang M, Choi DH, Lee SY, Son JW, Lee DC. Exosomal miR-196b secreted from bronchial epithelial cells chronically exposed to low-dose PM 2.5 promotes invasiveness of adjacent and lung cancer cells. Toxicol Lett 2024; 399:9-18. [PMID: 38971455 DOI: 10.1016/j.toxlet.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Fine particulate matter (PM2.5) is a risk factor for pulmonary diseases and lung cancer, and inhaled PM2.5 is mainly deposited in the bronchial epithelium. In this study, we investigated the effect of long-term exposure to low-dose PM2.5 on BEAS-2B cells derived from the normal bronchial epithelium. BEAS-2B cells chronically exposed to a concentration of 5 µg/ml PM2.5 for 30 passages displayed the phenotype promoting epithelial-mesenchymal transition (EMT) and cell invasion. Cellular internalization of exosomes (designated PM2.5 Exo) extracted from BEAS-2B cells chronically exposed to low-dose PM2.5 promoted cell invasion in vitro and metastatic potential in vivo. Hence, to identify the key players driving phenotypic alterations, we analyzed microRNA (miRNA) expression profiles in PM2.5 Exo. Five miRNAs with altered expression were selected: miRNA-196b-5p, miR-135a-2-5p, miR-3117-3p, miR-218-5p, and miR-497-5p. miR-196b-5p was the most upregulated in both BEAS-2B cells and isolated exosomes after PM2.5 exposure. In a functional validation study, genetically modified exosomes overexpressing a miR-196b-5p mimic induced an enhanced invasive phenotype in BEAS-2B cells. Conversely, miR-196b-5p inhibition diminished the PM2.5-enhanced EMT and cell invasion. These findings indicate that exosomal miR-196b-5p may be a candidate biomarker for predicting the malignant behavior of the bronchial epithelium and a therapeutic target for inhibiting PM2.5-triggered pathogenesis.
Collapse
Affiliation(s)
- Seong-Lan Yu
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea.
| | - Han Koo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Yujin Kang
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Hye Jin Jeon
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Minho Kang
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Dong Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Su Yel Lee
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Ji Woong Son
- Division Pulmonology, Department of Internal Medicine, Konyang, University Hospital, Daejeon 35365, Republic of Korea
| | - Dong Chul Lee
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
9
|
Kim C, Jeong SH, Lee H, Nam YJ, Lee H, Choi JY, Lee YS, Kim J, Park YH, Lee JH. Subchronic particulate matter exposure underlying polyhexamethylene guanidine phosphate-induced lung injury: Quantitative and qualitative evaluation with chest computed tomography. Heliyon 2024; 10:e34562. [PMID: 39113974 PMCID: PMC11305277 DOI: 10.1016/j.heliyon.2024.e34562] [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: 08/26/2023] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Our study was to explore the effects of subchronic particulate matter (PM) exposure on lung injury induced by polyhexamethylene guanidine phosphate (PHMG-p) in a rat model. Specifically, we investigated pulmonary inflammation, fibrosis, and tumor formation using chest computed tomography (CT), and histopathologic examination. PHMG-p was administered intratracheally to 20 male rats. After an initial week of PHMG-p treatment, the experimental group (PM group) received intratracheal administration of PM suspension, while the control group received normal saline. This regimen was continued for 10 weeks to induce subchronic PM exposure. Chest CT scans were conducted on all rats, followed by the extraction of both lungs for histopathological analysis. All CT images underwent comprehensive quantitative and qualitative analyses. Pulmonary inflammation was markedly intensified in rats subjected to subchronic PM exposure in the PM group compared to those in the control. Similarly, lung fibrosis was more severe in the PM group as observed on both chest CT and histopathologic examination. Quantitative chest CT analysis revealed that the mean lesion volume was significantly greater in the PM group than in the control group. Although the incidence of bronchiolo-alveolar hyperplasia was higher in the PM group compared to the control group, this difference was not statistically significant. In summary, subchronic PM exposure exacerbated pulmonary inflammation and fibrosis underlying lung injury induced by PHMG-p.
Collapse
Affiliation(s)
- Cherry Kim
- Department of Radiology, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Sang Hoon Jeong
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Hong Lee
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Yoon Jeong Nam
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Hyejin Lee
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Jin Young Choi
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Yu-Seon Lee
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Jaeyoung Kim
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Yoon Hee Park
- Medical Science Research Center, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| | - Ju-Han Lee
- Department of Pathology, Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi, 15355, South Korea
| |
Collapse
|
10
|
Zheng X, Wang Q, Xu X, Huang X, Chen J, Huo X. Associations of insulin sensitivity and immune inflammatory responses with child blood lead (Pb) and PM 2.5 exposure at an e-waste recycling area during the COVID-19 lockdown. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:296. [PMID: 38980420 DOI: 10.1007/s10653-024-02066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024]
Abstract
Fine particular matter (PM2.5) and lead (Pb) exposure can induce insulin resistance, elevating the likelihood of diabetes onset. Nonetheless, the underlying mechanism remains ambiguous. Consequently, we assessed the association of PM2.5 and Pb exposure with insulin resistance and inflammation biomarkers in children. A total of 235 children aged 3-7 years in a kindergarten in e-waste recycling areas were enrolled before and during the Corona Virus Disease 2019 (COVID-19) lockdown. Daily PM2.5 data was collected and used to calculate the individual PM2.5 daily exposure dose (DED-PM2.5). Concentrations of whole blood Pb, fasting blood glucose, serum insulin, and high mobility group box 1 (HMGB1) in serum were measured. Compared with that before COVID-19, the COVID-19 lockdown group had lower DED-PM2.5 and blood Pb, higher serum HMGB1, and lower blood glucose and homeostasis model assessment of insulin resistance (HOMA-IR) index. Decreased DED-PM2.5 and blood Pb levels were linked to decreased levels of fasting blood glucose and increased serum HMGB1 in all children. Increased serum HMGB1 levels were linked to reduced levels of blood glucose and HOMA-IR. Due to the implementation of COVID-19 prevention and control measures, e-waste dismantling activities and exposure levels of PM2.5 and Pb declined, which probably reduced the association of PM2.5 and Pb on insulin sensitivity and diabetes risk, but a high level of risk of chronic low-grade inflammation remained. Our findings add new evidence for the associations among PM2.5 and Pb exposure, systemic inflammation and insulin resistance, which could be a possible explanation for diabetes related to environmental exposure.
Collapse
Affiliation(s)
- Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China
- Center for Reproductive Medicine, Clinical Research Center, Shantou Central Hospital, Shantou, 515041, Guangdong, China
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xiaofan Huang
- Center for Reproductive Medicine, Clinical Research Center, Shantou Central Hospital, Shantou, 515041, Guangdong, China
| | - Jiaxue Chen
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
11
|
Chen PC, Yen MH, Hsiao SY, Kao WC, Wang MT, Chiou PC, Chao CC. Melatonin prevents pulmonary fibrosis caused by PM 2.5 exposure by targeting epithelial-mesenchymal transition. Toxicol Appl Pharmacol 2024; 487:116949. [PMID: 38688425 DOI: 10.1016/j.taap.2024.116949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Pulmonary fibrosis is a lung disorder characterized by the accumulation of abnormal extracellular matrix, scar tissue formation, and tissue stiffness. Type II alveolar epithelial cells (AEII) play a critical role in repairing lung tissue after injury, and repeated injury to these cells is a key factor in the development of pulmonary fibrosis. Chronic exposure to PM2.5, a type of air pollution, has been shown to increase the incidence and severity of pulmonary fibrosis by enhancing the activation of EMT in lung epithelial cells. Melatonin, a hormone with antioxidant properties, has been shown to prevent EMT and reduce fibrosis in previous studies. However, the mechanism through which melatonin targets EMT to prevent pulmonary fibrosis caused by PM2.5 exposure has not been extensively discussed before. In this current study, we found that melatonin effectively prevented pulmonary fibrosis caused by prolonged exposure to PM2.5 by targeting EMT. The study demonstrated changes in cellular morphology and expression of EMT markers. Furthermore, the cell migratory potential induced by prolonged exposure to PM2.5 was greatly reduced by melatonin treatment. Finally, in vivo animal studies showed reduced EMT markers and improved pulmonary function. These findings suggest that melatonin has potential clinical use for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Po-Chun Chen
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan; Translational medicine center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ming-Hong Yen
- Department of Chest Surgery, Cathay General Hospital, New Taipei City, Taiwan
| | - Sheng-Yen Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Chen Kao
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Mei-Ting Wang
- Division of Physical Medicine and Rehabilitation, Fu Jen Catholic University Hospital, Taipei, Taiwan, ROC
| | - Pei-Chen Chiou
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
12
|
Liu B, Han Y, Ye Y, Wei X, Li G, Jiang W. Atmospheric fine particulate matter (PM 2.5) induces pulmonary fibrosis by regulating different cell fates via autophagy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 923:171396. [PMID: 38438032 DOI: 10.1016/j.scitotenv.2024.171396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
The presence of respiratory diseases demonstrates a positive correlation with atmospheric fine particulate matter (PM2.5) exposure. The respiratory system is the main target organ affected by PM2.5, and exposure to PM2.5 elevates the likelihood of developing pulmonary fibrosis (PF). In this study, lung epithelial cell (BEAS-2B) and fibroblast (NIH-3T3) were used as in vitro exposure models to explore the mechanisms of PF. PM2.5 exposure caused mitochondrial damage in BEAS-2B cells and increased a fibrotic phenotype in NIH-3T3 cells. Epithelial cells and fibroblasts have different fates after PM2.5 exposure due to their different sensitivities to trigger autophagy. Exposure to PM2.5 inhibits mitophagy in BEAS-2B cells, which hinders the removal of damaged mitochondria and triggers cell death. In this process, the nuclear retention of the mitophagy-related protein Parkin prevents it from being recruited to mitochondria, resulting in mitophagy inhibition. In contrast, fibroblasts exhibit increased levels of autophagy, which may isolate PM2.5 and cause abnormal fibroblast proliferation and migration. Fibrotic phenotypes such as collagen deposition and increased α-actin also appear in fibroblasts. Our results identify PM2.5 as a trigger of PF and delineate the molecular mechanism of autophagy in PM2.5 induced PF, which provides new insights into the pulmonary injury.
Collapse
Affiliation(s)
- Bingyan Liu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Yangchen Han
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Yiyuan Ye
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Xiaoran Wei
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Gang Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Wei Jiang
- Environment Research Institute, Shandong University, Qingdao 266237, China.
| |
Collapse
|
13
|
Li S, Qu L, Zhou L, Zhan N, Liu L, Ling Y, Chen Q, Lai W, Lin N, Li J. Biomass fuels related-PM 2.5 promotes lung fibroblast-myofibroblast transition through PI3K/AKT/TRPC1 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116309. [PMID: 38599156 DOI: 10.1016/j.ecoenv.2024.116309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Emerging evidence has suggested that exposure to PM2.5 is a significant contributing factor to the development of chronic obstructive pulmonary disease (COPD). However, the underlying biological effects and mechanisms of PM2.5 in COPD pathology remain elusive. In this study, we aimed to investigate the implication and regulatory effect of biomass fuels related-PM2.5 (BRPM2.5) concerning the pathological process of fibroblast-to-myofibroblast transition (FMT) in the context of COPD. In vivo experimentation revealed that exposure to biofuel smoke was associated with airway inflammation in rats. After 4 weeks of exposure, there was inflammation in the small airways, but no significant structural changes in the airway walls. However, after 24 weeks, airway remodeling occurred due to increased collagen deposition, myofibroblast proliferation, and tracheal wall thickness. In vitro, cellular immunofluorescence results showed that with stimulation of BRPM2.5 for 72 h, the cell morphology of fibroblasts changed significantly, most of the cells changed from spindle-shaped to star-shaped irregular, α-SMA stress fibers appeared in the cytoplasm and the synthesis of type I collagen increased. The collagen gel contraction experiment showed that the contractility of fibroblasts was enhanced. The expression level of TRPC1 in fibroblasts was increased. Specific siRNA-TRPC1 blocked BRPM2.5-induced FMT and reduced cell contractility. Additionally, specific siRNA-TRPC1 resulted in a decrease in the augment of intracellular Ca2+ concentration ([Ca2+]i) induced by BRPM2.5. Notably, it was found that the PI3K inhibitor, LY294002, inhibited enhancement of AKT phosphorylation level, FMT occurrence, and elevation of TRPC1 protein expression induced by BRPM2.5. The findings indicated that BRPM2.5 is capable of inducing the FMT, with the possibility of mediation by PI3K/AKT/TRPC1. These results hold potential implications for the understanding of the molecular mechanisms involved in BRPM2.5-induced COPD and may aid in the development of novel therapeutic strategies for pathological conditions characterized by fibrosis.
Collapse
Affiliation(s)
- Shuni Li
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Lihui Qu
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Lifen Zhou
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Na Zhan
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Linmei Liu
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Yuquan Ling
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Qingzi Chen
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Wuping Lai
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Nan Lin
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| | - Jianhua Li
- State Key Laboratory of Respiratory disease, Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, RP China
| |
Collapse
|
14
|
Xu S, Ma L, Wu T, Tian Y, Wu L. Assessment of cellular senescence potential of PM2.5 using 3D human lung fibroblast spheroids in vitro model. Toxicol Res (Camb) 2024; 13:tfae037. [PMID: 38500513 PMCID: PMC10944558 DOI: 10.1093/toxres/tfae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Background Epidemiological studies demonstrate that particulate matter 2.5 (PM2.5) exposure closely related to chronic respiratory diseases. Cellular senescence plays an important role in many diseases. However, it is not fully clear whether PM2.5 exposure could induce cellular senescence in the human lung. In this study, we generated a three-dimensional (3D) spheroid model using isolated primary human lung fibroblasts (HLFs) to investigate the effects of PM2.5 on cellular senescence at the 3D level. Methods 3D spheroids were exposed to 25-100 μg/ml of PM2.5 in order to evaluate the impact on cellular senescence. SA-β-galactosidase activity, cell proliferation, and the expression of key genes and proteins were detected. Results Exposure of the HLF spheroids to PM2.5 yielded a more sensitive cytotoxicity than 2D HLF cell culture. Importantly, PM2.5 exposure induced the rapid progression of cellular senescence in 3D HLF spheroids, with a dramatically increased SA-β-Gal activity. In exploiting the mechanism underlying the effect of PM2.5 on senescence, we found a significant increase of DNA damage, upregulation of p21 protein levels, and suppression of cell proliferation in PM2.5-treated HLF spheroids. Moreover, PM2.5 exposure created a significant inflammatory response, which may be at least partially associated with the activation of TGF-β1/Smad3 axis and HMGB1 pathway. Conclusions Our results indicate that PM2.5 could induce DNA damage, inflammation, and cellular senescence in 3D HLF spheroids, which may provide a new evidence for PM2.5 toxicity based on a 3D model which has been shown to be more in vivo-like in their phenotype and physiology than 2D cultures.
Collapse
Affiliation(s)
- Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
| | - Lin Ma
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
| | - Tao Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Shushan District, Hefei, Anhui 230031, China
| | - Yushan Tian
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision and Test Center, 6 Cuizhu Street, New & High-tech Industry Development District, Zhengzhou, Henan 450001, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Shushan District, Hefei, Anhui 230031, China
| |
Collapse
|
15
|
Chao C, Hsiao S, Kao W, Chiou P, Huang C, Wang M, Chen P. Pyrroloquinoline quinone ameliorates PM2.5-induced pulmonary fibrosis through targeting epithelial-mesenchymal transition. J Cell Mol Med 2024; 28:e18299. [PMID: 38613355 PMCID: PMC11015390 DOI: 10.1111/jcmm.18299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Pulmonary fibrosis is a lung disorder affecting the lungs that involves the overexpressed extracellular matrix, scarring and stiffening of tissue. The repair of lung tissue after injury relies heavily on Type II alveolar epithelial cells (AEII), and repeated damage to these cells is a crucial factor in the development of pulmonary fibrosis. Studies have demonstrated that chronic exposure to PM2.5, a form of air pollution, leads to an increase in the incidence and severity of pulmonary fibrosis by stimulation of epithelial-mesenchymal transition (EMT) in lung epithelial cells. Pyrroloquinoline quinone (PQQ) is a bioactive compound found naturally that exhibits potent anti-inflammatory and anti-oxidative properties. The mechanism by which PQQ prevents pulmonary fibrosis caused by exposure to PM2.5 through EMT has not been thoroughly discussed until now. In the current study, we discovered that PQQ successfully prevented PM2.5-induced pulmonary fibrosis by targeting EMT. The results indicated that PQQ was able to inhibit the expression of type I collagen, a well-known fibrosis marker, in AEII cells subjected to long-term PM2.5 exposure. We also found the alterations of cellular structure and EMT marker expression in AEII cells with PM2.5 incubation, which were reduced by PQQ treatment. Furthermore, prolonged exposure to PM2.5 considerably reduced cell migratory ability, but PQQ treatment helped in reducing it. In vivo animal experiments indicated that PQQ could reduce EMT markers and enhance pulmonary function. Overall, these results imply that PQQ might be useful in clinical settings to prevent pulmonary fibrosis.
Collapse
Affiliation(s)
- Chia‐Chia Chao
- Department of Respiratory TherapyFu Jen Catholic UniversityNew TaipeiTaiwan
| | - Sheng‐Yen Hsiao
- Division of Hematology‐Oncology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of NursingChung Hwa University of Medical TechnologyTainanTaiwan
| | - Wan‐Chen Kao
- Division of Hematology‐Oncology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
| | - Pei‐Chen Chiou
- Department of Respiratory TherapyFu Jen Catholic UniversityNew TaipeiTaiwan
| | - Chieh‐Chen Huang
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
| | - Mei‐Ting Wang
- Division of Physical Medicine and RehabilitationFu Jen Catholic University HospitalTaipeiTaiwan, ROC
| | - Po‐Chun Chen
- School of Life ScienceNational Taiwan Normal UniversityTaipeiTaiwan
- Translational medicine centerShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Department of Medical ResearchChina Medical University Hospital, China Medical UniversityTaichungTaiwan
| |
Collapse
|
16
|
Moonwiriyakit A, Dinsuwannakol S, Sontikun J, Timpratueang K, Muanprasat C, Khemawoot P. Fine particulate matter PM2.5 and its constituent, hexavalent chromium induce acute cytotoxicity in human airway epithelial cells via inflammasome-mediated pyroptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104416. [PMID: 38492761 DOI: 10.1016/j.etap.2024.104416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
PM2.5-induced airway injury contributes to an increased rate of respiratory morbidity. However, the relationship between PM2.5 toxicants and acute cytotoxic effects remains poorly understood. This study aimed to investigate the mechanisms of PM2.5- and its constituent-induced cytotoxicity in human airway epithelial cells. Exposure to PM2.5 resulted in dose-dependent cytotoxicity within 24 h. Among the PM2.5 constituents examined, Cr(VI) at the dose found in PM2.5 exhibited cytotoxic effects. Both PM2.5 and Cr(VI) cause necrosis while also upregulating the expression of proinflammatory cytokine transcripts. Interestingly, exposure to the conditioned PM, obtained from adsorption in the Cr(VI)-reducing agents, FeSO4 and EDTA, showed a decrease in cytotoxicity. Furthermore, PM2.5 mechanistically enhances programmed pyroptosis through the activation of NLRP3/caspase-1/Gasdermin D pathway and increase of IL-1β. These pyroptosis markers were reduced when exposure to conditioned PM. These findings provide a deeper understanding of mechanisms underlying PM2.5 and Cr(VI) in acute airway toxicity.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand.
| | - Sasiwimol Dinsuwannakol
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Kanokphorn Timpratueang
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| |
Collapse
|
17
|
Yoon HY, Kim SY, Song JW. Effects of indoor air pollution on clinical outcomes in patients with interstitial lung disease: protocol of a multicentre prospective observational study. BMJ Open Respir Res 2024; 11:e002053. [PMID: 38262669 PMCID: PMC10806566 DOI: 10.1136/bmjresp-2023-002053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrosing interstitial lung disease with a poor prognosis. While there is evidence suggesting that outdoor air pollution affects the clinical course of IPF, the impact of indoor air pollution on patients with IPF has not been extensively studied. Therefore, this prospective multicentre observational study aims to investigate the association between indoor air pollution and clinical outcomes in patients with IPF. METHODS AND ANALYSIS This study enrolled 140 patients with IPF from 12 medical institutes in the Seoul and Metropolitan areas of the Republic of Korea. Over the course of 1 year, participants visited the institutes every 3 months, during which their clinical data and blood samples were collected. Additionally, indoor exposure to particulate matter ≤2.5 µm (PM2.5) was measured using MicroPEM (RTI International, Research Triangle Park, North Carolina, USA) in each participant's house for 5 days every 3 months. Lung function was assessed using both site spirometry at each institution and portable spirometry at each participant's house every 3 months. The study will analyse the impact of indoor PM2.5 on clinical outcomes, including mortality, acute exacerbation, changes in lung function and health-related quality of life, in the participants. This study represents the first attempt to evaluate the influence of indoor air pollution on the prognosis of patients with IPF. ETHICS AND DISSEMINATION This study has received approval from the institutional review board of all participating institutions, including Asan Medical Center, Seoul, Republic of Korea (2021-0072). TRIAL REGISTRATION NUMBER KCT0006217.
Collapse
Affiliation(s)
- Hee-Young Yoon
- Division of Allergy and Respiratory Diseases, Soonchunhyang University Seoul Hospital, Seoul, Korea (the Republic of)
| | - Sun-Young Kim
- Department of Cancer AI & Digital Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea (the Republic of)
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Korea (the Republic of)
| |
Collapse
|
18
|
Gavito-Covarrubias D, Ramírez-Díaz I, Guzmán-Linares J, Limón ID, Manuel-Sánchez DM, Molina-Herrera A, Coral-García MÁ, Anastasio E, Anaya-Hernández A, López-Salazar P, Juárez-Díaz G, Martínez-Juárez J, Torres-Jácome J, Albarado-Ibáñez A, Martínez-Laguna Y, Morán C, Rubio K. Epigenetic mechanisms of particulate matter exposure: air pollution and hazards on human health. Front Genet 2024; 14:1306600. [PMID: 38299096 PMCID: PMC10829887 DOI: 10.3389/fgene.2023.1306600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024] Open
Abstract
Environmental pollution nowadays has not only a direct correlation with human health changes but a direct social impact. Epidemiological studies have evidenced the increased damage to human health on a daily basis because of damage to the ecological niche. Rapid urban growth and industrialized societies importantly compromise air quality, which can be assessed by a notable accumulation of air pollutants in both the gas and the particle phases. Of them, particulate matter (PM) represents a highly complex mixture of organic and inorganic compounds of the most variable size, composition, and origin. PM being one of the most complex environmental pollutants, its accumulation also varies in a temporal and spatial manner, which challenges current analytical techniques used to investigate PM interactions. Nevertheless, the characterization of the chemical composition of PM is a reliable indicator of the composition of the atmosphere, the quality of breathed air in urbanized societies, industrial zones and consequently gives support for pertinent measures to avoid serious health damage. Epigenomic damage is one of the most promising biological mechanisms of air pollution-derived carcinogenesis. Therefore, this review aims to highlight the implication of PM exposure in diverse molecular mechanisms driving human diseases by altered epigenetic regulation. The presented findings in the context of pan-organic cancer, fibrosis, neurodegeneration and metabolic diseases may provide valuable insights into the toxicity effects of PM components at the epigenomic level and may serve as biomarkers of early detection for novel targeted therapies.
Collapse
Affiliation(s)
- Dulcemaría Gavito-Covarrubias
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
- Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Josué Guzmán-Linares
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Dulce María Manuel-Sánchez
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alejandro Molina-Herrera
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Miguel Ángel Coral-García
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Estela Anastasio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Arely Anaya-Hernández
- Centro de Investigación en Genética y Ambiente, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Primavera López-Salazar
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gabriel Juárez-Díaz
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Javier Martínez-Juárez
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julián Torres-Jácome
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alondra Albarado-Ibáñez
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Ygnacio Martínez-Laguna
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| |
Collapse
|
19
|
Wang X, Zhang D, Zhu Y, Li D, Shen L, Wang Q, Gao Y, Li X, Yu M. Protein lysine acetylation played an important role in NH 3-induced AEC2 damage and pulmonary fibrosis in piglets. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168303. [PMID: 37939958 DOI: 10.1016/j.scitotenv.2023.168303] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/10/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
Gaseous ammonia (NH3), as a main air pollutant in pig farms and surrounding areas, directly affects animal and human health. The lung, as an important organ for gas exchange in the respiratory system, is damaged after NH3 exposure, but the underlying mechanism needs to be further explored. In this study, seven weeks old piglets were exposed to 50 ppm NH3 for 30 days, and displayed pulmonary fibrosis. Then, the toxicological mechanism of NH3-induced pulmonary fibrosis was explored from the aspects of whole genome wide protein expression and post-translational modification. Totally, 404 differentially expressed proteins (DEPs) and 136 differentially lysine acetylated proteins (DAPs) were identified. The expression or lysine acetylation levels of proteins involved in mitochondrial energy metabolism including fatty acid oxidation (CPT1A, ACADVL, ACADS, HADHA, and HADHB), TCA cycle (IDH2 and MDH2), and oxidative phosphorylation (NDUFB7, NDUFV1, ATP5PB, ATP5F1A, COX5A, and COX5B) were significantly changed after NH3 exposure, which suggested that NH3 disrupted mitochondrial energy metabolism in the lung of piglets. Next, we found that type 2 alveolar epithelial cells (AEC2) damaged after NH3 exposure in vivo and in vitro. Integrin-linked kinase (ILK) was enriched in focal adhesion pathway, and showed significantly up-regulated acetylation levels at K191 (FC = 2.99) and K209 sites (FC = 1.52) after NH3 exposure. We illustrated that ILK-K191 hyper-acetylation inhibited AEC2 proliferation and induced AEC2 apoptosis by down-regulating pAKT-S473 in vitro. In conclusion, for the first time, our study revealed that protein acetylation played an important role in the process of NH3-induced pulmonary fibrosis in piglets. Our findings provided valuable insights into toxicological harm of NH3 to human health.
Collapse
Affiliation(s)
- Xiaotong Wang
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Di Zhang
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Yaxue Zhu
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Daojie Li
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Long Shen
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiankun Wang
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| | - Yun Gao
- College of Engineering, the Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoping Li
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Smart Animal Farming Technology, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Mei Yu
- College of Animal Science and Technology, Center for Advanced Science in Animal Breeding and Health Breeding, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
20
|
Ha JH, Lee BW, Yi DH, Lee SJ, Kim WI, Pak SW, Kim HY, Kim SH, Shin IS, Kim JC, Lee IC. Particulate matter-mediated oxidative stress induces airway inflammation and pulmonary dysfunction through TXNIP/NF-κB and modulation of the SIRT1-mediated p53 and TGF-β/Smad3 pathways in mice. Food Chem Toxicol 2024; 183:114201. [PMID: 38013002 DOI: 10.1016/j.fct.2023.114201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023]
Abstract
Exposure to particulate matter is currently recognized as a serious aggravating factor of respiratory diseases. In this study, we investigated the effects of particulate matter (PM) on the respiratory system in BALB/c mice and NCI-H292 cells. PM (0, 2.5, 5 and 20 mg/kg) was administered to mice by intra-tracheal instillation for 7 days. After a 7 day-repeated treatment of PM, we evaluated inflammatory cytokines/cell counts in bronchoalveolar lavage fluid (BALF) and conducted pulmonary histology and functional test. We also investigated the role of TXNIP/NF-κB and SIRT1-mediated p53 and TGF-β/Smad3 pathways in PM-induced airway inflammation and pulmonary dysfunction. PM caused a significant increase in pro-inflammatory cytokines, inflammatory cell counts in bronchoalveolar lavage fluid. PM-mediated oxidative stress down-regulated thioredoxin-1 and up-regulated thioredoxin-interacting protein and activation of nuclear factor-kappa B in the lung tissue and PM-treated NCI-H292 cells. PM suppressed sirtuin1 protein levels and increased p53 acetylation in PM-exposed mice and PM-treated NCI-H292 cells. In addition, PM caused inflammatory cell infiltration and the thickening of alveolar walls by exacerbating the inflammatory response in the lung tissue. PM increased levels of transforming growth factor-β, phosphorylation of Smad3 and activation of α-smooth muscle actin, and collagen type1A2 in PM-exposed mice and PM-treated NCI-H292 cells. In pulmonary function tests, PM exposure impaired pulmonary function resembling pulmonary fibrosis, characterized by increased resistance and elastance of the respiratory system, and resistance, elastance, and damping of lung tissues, whereas decreased compliance of the respiratory system, forced expired volume and forced vital capacity. Overall, PM-mediated oxidative stress caused airway inflammation and pulmonary dysfunction with pulmonary fibrosis via TXNIP pathway/NF-κB activation and modulation of the SIRT1-mediated TGF-β/Smad3 pathways. The results of this study can provide fundamental data on the potential adverse effects and underlying mechanism of pulmonary fibrosis caused by PM exposure as a public health concern. Due to the potential toxicity of PM, people with respiratory disease must be careful with PM exposure.
Collapse
Affiliation(s)
- Ji-Hye Ha
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chungnam National University, Daejeon, Republic of Korea
| | - Ba-Wool Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Da-Hye Yi
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Hyeon-Young Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea.
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea.
| |
Collapse
|
21
|
Craig NA, Scruggs AM, Berens JP, Deng F, Chen Y, Dvonch JT, Huang SK. Promotion of myofibroblast differentiation through repeated treatment of fibroblasts to low concentrations of PM 2.5. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 105:104329. [PMID: 38036232 PMCID: PMC11010492 DOI: 10.1016/j.etap.2023.104329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 11/25/2023] [Indexed: 12/02/2023]
Abstract
Exposure to particulate matter ≤ 2.5 µm (PM2.5) is a risk factor for many lung diseases. Although the toxicologic effects of PM2.5 on airway epithelium are well-described, the effects of PM2.5 on fibroblasts in the lung are less studied. Here, we sought to examine the effects of PM2.5 on the differentiation of fibroblasts into myofibroblasts. Although a single treatment of fibroblasts did not result in a change in collagen or the myofibroblast marker α-SMA, exposing fibroblasts to sequential treatments with PM2.5 at low concentrations caused a robust increase in these proteins. Treatment of fibroblasts with IMD0354, an inhibitor to nuclear factor κB, but not with an antagonist to aryl hydrocarbon receptor, abolished the ability of PM2.5 to induce myofibroblast differentiation. These data demonstrate that potential impact of PM2.5 to fibroblast activation and fibrosis and support the importance of utilizing low concentrations and varying exposure protocols to toxicologic studies.
Collapse
Affiliation(s)
- Nathan A Craig
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anne M Scruggs
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jack P Berens
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | - Yahong Chen
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - J Timothy Dvonch
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Wu T, Liu B, Wei Y, Li Z. TGF-β Regulates m 6A RNA Methylation after PM 2.5 Exposure. TOXICS 2023; 11:1026. [PMID: 38133427 PMCID: PMC10747615 DOI: 10.3390/toxics11121026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
PM2.5 exposure leads to a variety of respiratory diseases, including pulmonary fibrosis, metastatic lung cancer, etc. Exposure to PM2.5 results in the alteration of epigenetic modification. M6A RNA methylation is an essential epigenetic modification that regulates gene expression at the post-transcriptional level. Our previous study found that PM2.5 exposure up-regulated m6A RNA methylation and TGF-β expression level in the lung, but the mechanisms and pathways of PM2.5 regulation of m6A RNA methylation are still unclear. Moreover, a previous study reported that the TGF-β signal pathway could regulate m6A RNA methylation. Based on this evidence, we investigate the role of the TGF-β signaling pathway in PM2.5-induced m6A RNA methylation with the A549 cell line. Our results showed that PM2.5 could induce upregulation of m6A RNA methylation, accompanied by increased expression of TGF-β, Smad3, methyltransferase-like 3 (METTL3), methyltransferase-like 14 (METTL14). Furthermore, these alterations induced by PM2.5 exposure could be reversed by treatment with TGF-β inhibitor. Therefore, we speculated that the TGF-β signal pathway plays an indispensable role in regulating m6A RNA methylation after PM2.5 exposure. Our study demonstrates that PM2.5 exposure influences m6A RNA methylation by inducing the alteration of the TGF-β signal pathway, which could be an essential mechanism for lung-related diseases induced by PM2.5 exposure.
Collapse
Affiliation(s)
| | | | | | - Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; (T.W.); (B.L.); (Y.W.)
| |
Collapse
|
23
|
Ghimire A, Hasan F, Guan X, Potter P, Guo C, Lomnicki S. Oxidation 1-methyl naphthalene based on the synergy of environmentally persistent free radicals (EPFRs) and PAHs in particulate matter (PM) surface. CHEMOSPHERE 2023; 341:140002. [PMID: 37648160 PMCID: PMC10548478 DOI: 10.1016/j.chemosphere.2023.140002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/15/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
Studies of the environmental fate through the interactions of particle-associated polycyclic aromatic hydrocarbons (PAHs) with environmentally persistent free radicals (EPFRs) are presented. The formation of PAHs and EPFRs typically occurs side by side during combustion-processes. The laboratory simulation studies of the model PAH molecule 1-Methylnaphthalene (1-MN) interaction with model EPFRs indicate a transformational synergy between these two pollutants due to mutual and matrix interactions. EPFRs, thorough its redox cycle result in the oxidation of PAHs into oxy-/hydroxy-PAHs. EPFRs have been shown before to produce OH radical during its redox cycle in aqueous media and this study has shown that produced OH radical can transform other PM constituents resulting in alteration of PM chemistry. In model PM, EPFRs driven oxidation process of 1-MN produced 1,4-naphthoquinone, 1-naphthaldehyde, 4-hydroxy-4-methylnaphthalen-1-one, and various isomers of (hydroxymethyl) naphthalene. Differences were observed in oxidation product yields, depending on whether EPFRs and PAHs were cohabiting the same PM or present on separate PM. This effect is attributed to the OH radical concentration gradient as a factor in the oxidation process, further strengthening the hypothesis of EPFRs' role in the PAH oxidation process. This finding is revealing new environmental role of EPFRs in a natural degradation process of PAHs. Additionally, it points to implications of such PM surface chemistry in the changing mobility of PAHs into an aqueous medium, thus increasing their bioavailability.
Collapse
Affiliation(s)
- Ajit Ghimire
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States
| | - Farhana Hasan
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States
| | - Xia Guan
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States
| | - Phillip Potter
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States
| | - Chuqi Guo
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States
| | - Slawo Lomnicki
- Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA, 70803, United States.
| |
Collapse
|
24
|
Zhang C, Ma T, Liu C, Ma D, Wang J, Liu M, Ran J, Wang X, Deng X. PM 2.5 induced liver lipid metabolic disorders in C57BL/6J mice. Front Endocrinol (Lausanne) 2023; 14:1212291. [PMID: 37780625 PMCID: PMC10539470 DOI: 10.3389/fendo.2023.1212291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/16/2023] [Indexed: 10/03/2023] Open
Abstract
PM2.5 can cause adverse health effects via several pathways, such as inducing pulmonary and systemic inflammation, penetration into circulation, and activation of the autonomic nervous system. In particular, the impact of PM2.5 exposure on the liver, which plays an important role in metabolism and detoxification to maintain internal environment homeostasis, is getting more attention in recent years. In the present study, C57BL/6J mice were randomly assigned and treated with PM2.5 suspension and PBS solution for 8 weeks. Then, hepatic tissue was prepared and identified by metabolomics analysis and transcriptomics analysis. PM2.5 exposure can cause extensive metabolic disturbances, particularly in lipid and amino acids metabolic dysregulation.128 differential expression metabolites (DEMs) and 502 differently expressed genes (DEGs) between the PM2.5 exposure group and control group were detected. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that DEGs were significantly enriched in two disease pathways, non-alcoholic fatty liver disease (NAFLD) and type II diabetes mellitus (T2DM), and three signaling pathways, which are TGF-beta signaling, AMPK signaling, and mTOR signaling. Besides, further detection of acylcarnitine levels revealed accumulation in liver tissue, which caused restricted lipid consumption. Furthermore, lipid droplet accumulation in the liver was confirmed by Oil Red O staining, suggesting hepatic steatosis. Moreover, the aberrant expression of three key transcription factors revealed the potential regulatory effects in lipid metabolic disorders, the peroxisomal proliferative agent-activated receptors (PPARs) including PPARα and PPARγ is inhibited, and the activated sterol regulator-binding protein 1 (SREBP1) is overexpressed. Our results provide a novel molecular and genetic basis for a better understanding of the mechanisms of PM2.5 exposure-induced hepatic metabolic diseases, especially in lipid metabolism.
Collapse
Affiliation(s)
- Chenxiao Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tengfei Ma
- College of Basic Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ding Ma
- College of Basic Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- College of Basic Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Liu
- College of Basic Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinjun Ran
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueting Wang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobei Deng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Garcia A, Santa-Helena E, De Falco A, de Paula Ribeiro J, Gioda A, Gioda CR. Toxicological Effects of Fine Particulate Matter (PM 2.5): Health Risks and Associated Systemic Injuries-Systematic Review. WATER, AIR, AND SOIL POLLUTION 2023; 234:346. [PMID: 37250231 PMCID: PMC10208206 DOI: 10.1007/s11270-023-06278-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 03/29/2023] [Indexed: 05/31/2023]
Abstract
Previous studies focused on investigating particulate matter with aerodynamic diameter ≤ 2.5 µm (PM2.5) have shown the risk of disease development, and association with increased morbidity and mortality rates. The current review investigate epidemiological and experimental findings from 2016 to 2021, which enabled the systemic overview of PM2.5's toxic impacts on human health. The Web of Science database search used descriptive terms to investigate the interaction among PM2.5 exposure, systemic effects, and COVID-19 disease. Analyzed studies have indicated that cardiovascular and respiratory systems have been extensively investigated and indicated as the main air pollution targets. Nevertheless, PM2.5 reaches other organic systems and harms the renal, neurological, gastrointestinal, and reproductive systems. Pathologies onset and/or get worse due to toxicological effects associated with the exposure to this particle type, since it can trigger several reactions, such as inflammatory responses, oxidative stress generation and genotoxicity. These cellular dysfunctions lead to organ malfunctions, as shown in the current review. In addition, the correlation between COVID-19/Sars-CoV-2 and PM2.5 exposure was also assessed to help better understand the role of atmospheric pollution in the pathophysiology of this disease. Despite the significant number of studies about PM2.5's effects on organic functions, available in the literature, there are still gaps in knowledge about how this particulate matter can hinder human health. The current review aimed to approach the main findings about the effect of PM2.5 exposure on different systems, and demonstrate the likely interaction of COVID-19/Sars-CoV-2 and PM2.5.
Collapse
Affiliation(s)
- Amanda Garcia
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Eduarda Santa-Helena
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Anna De Falco
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Joaquim de Paula Ribeiro
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Adriana Gioda
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Carolina Rosa Gioda
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| |
Collapse
|
26
|
Chang EM, Chao CC, Wang MT, Hsu CL, Chen PC. PM 2.5 promotes pulmonary fibrosis by mitochondrial dysfunction. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37126650 DOI: 10.1002/tox.23817] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/23/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Pulmonary fibrosis is known as an incurable lung disorder with irreversible progression of chronic injury, myofibroblast proliferation, extracellular matrix (ECM) accumulation, and tissue scarring. Atmospheric particulate matter 2.5 (PM2.5 ) is implicated as a risk factor of several diseases, especially lung diseases such as pulmonary fibrosis. The molecular mechanism which participates PM2.5 -induced pulmonary fibrosis in type II alveolar cells (AEII) has yet to be determined. Our results proved that short- and long-term exposure to PM2.5 significantly stimulated epithelial-mesenchymal transition (EMT) activity in AEII cells, according to, changes in gene signature analyzed by RNA-seq and cell morphology. Furthermore, Gene Ontology (GO) enrichment analysis also suggested that mitochondrial dysfunction was related to progression of pulmonary fibrosis in AEII after PM2.5 exposure. We observed a marked decline in mitochondria membrane potential (MMP), as well as fragmented mitochondria, in AEII cells exposed to PM2.5 , which suggests that energy metabolism is suppressed after PM2.5 exposure. We also confirmed that PM2.5 exposure could influence the expression levels of Mfn1, Mfn2, and Drp1 in AEII. Pretreatment of mitochondrial fusion promoter M1 was able to reverse mitochondrial dysfunction as well as EMT in AEII. These data suggested the key role of mitochondrial fragmentation in AEII, which was induced by PM2.5 exposure, and participated pathogenesis of pulmonary fibrosis. Finally, we investigated the response of lung tissue exposed to PM2.5 in vivo. The data indicated that the lung tissue exposed to PM2.5 obviously induced collagen accumulation. Moreover, IHC results revealed that PM2.5 enhanced Drp1 expression but suppressed Mfn1 and Mfn2 expression in lung tissue. The current study provides novel insight of pulmonary fibrosis caused by PM2.5 exposure.
Collapse
Affiliation(s)
- En-Ming Chang
- Department of Respiratory Care, Shin Kong Wu Ho Su Memorial Hospital, Taipei City, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Mei-Ting Wang
- Division of Physical Medicine and Rehabilitation, Fu Jen Catholic University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei City, Taiwan
| | - Po-Chun Chen
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
27
|
da Silva MC, Fabiano LC, da Costa Salomão KC, de Freitas PLZ, Neves CQ, Borges SC, de Souza Carvalho MDG, Breithaupt-Faloppa AC, de Thomaz AA, Dos Santos AM, Buttow NC. A Rodent Model of Human-Dose-Equivalent 5-Fluorouracil: Toxicity in the Liver, Kidneys, and Lungs. Antioxidants (Basel) 2023; 12:antiox12051005. [PMID: 37237871 DOI: 10.3390/antiox12051005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
5-Fluorouracil (5-FU) is a chemotherapy drug widely used to treat a range of cancer types, despite the recurrence of adverse reactions. Therefore, information on its side effects when administered at a clinically recommended dose is relevant. On this basis, we examined the effects of the 5-FU clinical treatment on the integrity of the liver, kidneys, and lungs of rats. For this purpose, 14 male Wistar rats were divided into treated and control groups and 5-FU was administered at 15 mg/kg (4 consecutive days), 6 mg/kg (4 alternate days), and 15 mg/kg on the 14th day. On the 15th day, blood, liver, kidney, and lung samples were collected for histological, oxidative stress, and inflammatory evaluations. We observed a reduction in the antioxidant markers and an increase in lipid hydroperoxides (LOOH) in the liver of treated animals. We also detected elevated levels of inflammatory markers, histological lesions, apoptotic cells, and aspartate aminotransferase. Clinical treatment with 5-FU did not promote inflammatory or oxidative alterations in the kidney samples; however, histological and biochemical changes were observed, including increased serum urea and uric acid. 5-FU reduces endogenous antioxidant defenses and increases LOOH levels in the lungs, suggesting oxidative stress. Inflammation and histopathological alterations were also detected. The clinical protocol of 5-FU promotes toxicity in the liver, kidneys, and lungs of healthy rats, resulting in different levels of histological and biochemical alterations. These results will be useful in the search for new adjuvants to attenuate the adverse effects of 5-FU in such organs.
Collapse
Affiliation(s)
- Mariana Conceição da Silva
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Lilian Catarim Fabiano
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | | | - Camila Quaglio Neves
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| | | | - Maria das Graças de Souza Carvalho
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-904, SP, Brasil
| | - André Alexandre de Thomaz
- Quantum Electronic Department, Institute of Physics Gleb Wataghin, State University of Campinas, Campinas 13083-872, SP, Brazil
| | - Aline Mara Dos Santos
- Biological Physics and Cell Signaling Laboratory, Institute of Biology, Department of Structural and Functional Biology, State University of Campinas, Campinas 13083-970, SP, Brazil
| | - Nilza Cristina Buttow
- Department of Morphological Science, State University of Maringá, Maringá 87020-900, PR, Brazil
| |
Collapse
|
28
|
Li Y, Du Z, Li T, Ren X, Yu Y, Duan J, Sun Z. MitoQ ameliorates PM 2.5-induced pulmonary fibrosis through regulating the mitochondria DNA homeostasis. CHEMOSPHERE 2023; 330:138745. [PMID: 37088202 DOI: 10.1016/j.chemosphere.2023.138745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Pulmonary fibrosis is a severe pulmonary disease, and may related to PM2.5 exposure. Our study aims to explore the pathogenesis of PM2.5-induced pulmonary fibrosis, and MitoQ protective effect in this process. Our results find that inflammatory cells aggregation and pulmonary fibrosis in mice lung after PM2.5 exposure. Moreover, Collagen I/III overproduction, EMT and TGF-β1/Smad2 pathway activation in mice lung and BEAS-2B after PM2.5 exposure. Fortunately, these changes were partially ameliorated after MitoQ treatment. Meanwhile, severe oxidative stress, mitochondrial homeostasis imbalance, overproduction of 8-oxoG (7,8-dihydro-8-oxoguanine), as well as the inhibition of SIRT3/OGG1 pathway have founded in mice lung or BEAS-2B after PM2.5 exposure, which were alleviated by MitoQ treatment. Collectively, our study found that oxidative stress, especially mitochondrial oxidative stress participates in the PM2.5-induced pulmonary fibrosis, and MitoQ intervention had a protective effect on this progress. Moreover, mitochondrial DNA homeostasis might participate in the pulmonary fibrosis caused by PM2.5 exposure. Our study provides a novel pathogenesis of PM2.5-caused pulmonary fibrosis and a possible targeted therapy for the pulmonary diseases triggered by PM2.5.
Collapse
Affiliation(s)
- Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Zhou Du
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
29
|
Noonin C, Itsaranawet T, Thongboonkerd V. Calcium oxalate crystal-induced secretome derived from proximal tubular cells, not that from distal tubular cells, induces renal fibroblast activation. Eur J Med Res 2023; 28:150. [PMID: 37031165 PMCID: PMC10082508 DOI: 10.1186/s40001-023-01109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/24/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND Kidney stone disease (KSD) is commonly accompanied with renal fibrosis, characterized by accumulation and reorganization of extracellular matrix (ECM). During fibrogenesis, resident renal fibroblasts are activated to become myofibroblasts that actively produce ECM. However, such fibroblast-myofibroblast differentiation in KSD remained unclear. Our present study thus examined effects of secreted products (secretome) derived from proximal (HK-2) vs. distal (MDCK) renal tubular cells exposed to calcium oxalate monohydrate (COM) crystals on activation of renal fibroblasts (BHK-21). METHODS HK-2 and MDCK cells were treated with 100 µg/ml COM crystals under serum-free condition for 16 h. In parallel, the cells maintained in serum-free medium without COM treatment served as the control. Secretome derived from culture supernatant of each sample was mixed (1:1) with fresh serum-free medium and then used for BHK-21 culture for another 24 h. RESULTS Analyses revealed that COM-treated-HK-2 secretome significantly induced proliferation, caused morphological changes, increased spindle index, and upregulated fibroblast-activation markers (F-actin, α-SMA and fibronectin) in BHK-21 cells. However, COM-treated-MDCK secretome had no significant effects on these BHK-21 parameters. Moreover, level of transforming growth factor-β1 (TGF-β1), a profibrotic factor, significantly increased in the COM-treated-HK-2 secretome but not in the COM-treated-MDCK secretome. CONCLUSIONS These data indicate, for the first time, that proximal and distal tubular epithelial cells exposed to COM crystals send different messages to resident renal fibroblasts. Only the secretome derived from proximal tubular cells, not that from the distal cells, induces renal fibroblast activation after their exposure to COM crystals. Such differential effects are partly due to TGF-β1 secretion, which is induced by COM crystals only in proximal tubular cells.
Collapse
Affiliation(s)
- Chadanat Noonin
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 6th Floor - SiMR Building, 2 Wanglang Road, Bangkoknoi, 10700, Bangkok, Thailand
| | - Tanakorn Itsaranawet
- Biological Sciences Program, Mahidol University International College, Nakhon Pathom, 73170, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 6th Floor - SiMR Building, 2 Wanglang Road, Bangkoknoi, 10700, Bangkok, Thailand.
| |
Collapse
|
30
|
Yin L, Han K, Jiang B, Meng Q, Aschner M, Li X, Chen R. NAT10 accelerates pulmonary fibrosis through N4-acetylated TGFB1-initiated epithelial-to-mesenchymal transition upon ambient fine particulate matter exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 322:121149. [PMID: 36731737 DOI: 10.1016/j.envpol.2023.121149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/29/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Exposure to ambient fine particulate matter (PM2.5) has been linked to a higher pulmonary fibrosis risk. Dysregulation of the epitranscriptome results in abnormal expression of mRNAs during fibrosis development. N4-acetylcytidine (ac4C) is one of the most frequent RNA epigenetic alterations, however, its function in PM2.5-triggered fibrosis is yet unknown. In this study, lung epithelial and murine models were established and exposed to PM2.5 to analyze the function of ac4C alteration in pulmonary fibrosis and underlying mechanisms. Meanwhile, the expression levels of only known ac4C "writer" protein, N-acetyltransferase 10 (NAT10), were significantly induced in pulmonary epithelia, relative to the control. Subsequently, NAT10 enhanced the stability of transforming growth factor beta 1 (TGFB1) mRNA as well as protein levels. As an up-stream driver, TGFB1 accelerated EMT and fibrosis process. Inhibition of NAT10 significantly protected against pulmonary EMT and fibrosis driven by PM2.5 exposure, whereas TGFB1 overexpression reversed the protective effects of NAT10 inhibition. Thus, NAT10 accelerated PM2.5-triggered pulmonary fibrosis via increasing TGFB1 mRNA stability in an ac4C-dependent manner. Our results reveal a pivotal role of NAT10-regulated mRNA ac4C acetylation in PM2.5-triggered pulmonary fibrosis and uncover the potential epitranscriptional mechanism.
Collapse
Affiliation(s)
- Lijia Yin
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Ke Han
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Bo Jiang
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Qingtao Meng
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Xiaobo Li
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing, 100069, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China; Beijing laboratory of allergic diseases, Capital Medical University; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
31
|
Coal dust nanoparticles induced pulmonary fibrosis by promoting inflammation and epithelial-mesenchymal transition via the NF-κB/NLRP3 pathway driven by IGF1/ROS-mediated AKT/GSK3β signals. Cell Death Dis 2022; 8:500. [PMID: 36581638 PMCID: PMC9800584 DOI: 10.1038/s41420-022-01291-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Pneumoconiosis is the most common and serious disease among coal miners. In earlier work on this subject, we documented that coal dust (CD) nanoparticles (CD-NPs) induced pulmonary fibrosis (PF) more profoundly than did CD micron particles (CD-MPs), but the mechanism has not been thoroughly studied. Based on the GEO database, jveen, STRING, and Cytoscape tools were used to screen hub genes regulating PF. Particle size distribution of CD were analyzed with Malvern nanoparticle size potentiometer. Combining 8 computational methods, we found that IGF1, POSTN, MMP7, ASPN, and CXCL14 may act as hub genes regulating PF. Based on the high score of IGF1 and its important regulatory role in various tissue fibrosis, we selected it as the target gene in this study. Activation of the IGF1/IGF1R axis promoted CD-NPs-induced PF, and inhibition of the axis activation had the opposite effect in vitro and in vivo. Furthermore, activation of the IGF1/IGF1R axis induced generation of reactive oxygen species (ROS) to promote epithelial-mesenchymal transition (EMT) in alveolar epithelial cells (AECs) to accelerate PF. High-throughput gene sequencing based on lung tissue suggested that cytokine-cytokine receptor interaction and the NF-kB signaling pathway play a key role in PF. Also, ROS induced inflammation and EMT by the activation of the NF-kB/NLRP3 axis to accelerate PF. ROS can induce the activation of AKT/GSK3β signaling, and inhibition of it can inhibit ROS-induced inflammation and EMT by the NF-kB/NLRP3 axis, thereby inhibiting PF. CD-NPs induced PF by promoting inflammation and EMT via the NF-κB/NLRP3 pathway driven by IGF1/ROS-mediated AKT/GSK3β signals. This study provides a valuable experimental basis for the prevention and treatment of coal workers' pneumoconiosis. Illustration of the overall research idea of this study: IGF1 stimulates coal dust nanoparticles induced pulmonary fibrosis by promoting inflammation and EMT via the NF-κB/NLRP3 pathway driven by ROS-mediated AKT/GSK3β signals.
Collapse
|
32
|
Liu H, Nie H, Lai W, Shi Y, Liu X, Li K, Tian L, Xi Z, Lin B. Different exposure modes of PM 2.5 induces bronchial asthma and fibrosis in male rats through macrophage activation and immune imbalance induced by TIPE2 methylation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114200. [PMID: 36274320 DOI: 10.1016/j.ecoenv.2022.114200] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/02/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Exposure to PM2.5 can aggravate the occurrence and development of bronchial asthma and fibrosis. Here, we investigated the differences in bronchial injury caused by different exposure modes of PM2.5 (high concentration intermittent exposure and low concentration continuous exposure), and the mechanism of macrophage activation and respiratory immune imbalance induced by PM2.5, leading to bronchial asthma and airway fibrosis using animal and cell models. A "PM2.5 real-time online concentrated animal whole-body exposure system" was used to conduct PM2.5 respiratory exposure of Wistar rats for 12 weeks, which can enhance oxidative stress in rat bronchus, activate epithelial cells and macrophages, release chemokines, recruit inflammatory cells, release inflammatory factors and extracellular matrix, promote bronchial mucus hypersecretion, inhibit the expression of epithelial cytoskeletal proteins, destroy airway barrier, and induce asthma. Furthermore, PM2.5 induced M2 polarization in lung bronchial macrophages through JAK/STAT and PI3K/Akt signaling pathways, and compared with low concentration continuous exposure, high concentration intermittent exposure of PM2.5 could regulate significantly higher expression of TIPE2 protein through promoter methylation of TIPE2 DNA, thereby activating PI3K/Akt signaling pathway and more effectively inducing M2 polarization of macrophages. Additionally, activated macrophages release IL-23, and activated epithelial cells and macrophages released TGF-β1, which promoted the differentiation of Th17 cells, triggered the Th17 dominant immune response, and activated the TGF-β1/Smad2 signaling pathway, finally causing bronchial fibrosis. Moreover, when the total amount of PM2.5 exposure was equal, high concentration-intermittent exposure was more serious than low concentration-continuous exposure. In vitro experiments, the co-culture models of PM2.5 with BEAS-2B, WL-38 and rat primary alveolar macrophages further confirmed that PM2.5 could induce the macrophage activation through oxidative stress and TIPE2 DNA methylation, and activate the TGF-β1/Smad2 signaling pathway, leading to the occurrence of bronchial fibrosis.
Collapse
Affiliation(s)
- Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Huipeng Nie
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Yue Shi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Xuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China.
| |
Collapse
|
33
|
Zhao C, Pu W, Wazir J, Jin X, Wei L, Song S, Su Z, Li J, Deng Y, Wang H. Long-term exposure to PM2.5 aggravates pulmonary fibrosis and acute lung injury by disrupting Nrf2-mediated antioxidant function. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 313:120017. [PMID: 36007796 DOI: 10.1016/j.envpol.2022.120017] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Epidemiological studies have indicated that exposure to ambient air-borne fine particulate matter (PM2.5) is associated with many cardiopulmonary diseases; however, the underlying pathological mechanisms of PM2.5-induced lung injury remain unknown. In this study, we aimed to assess the impact of acute or prolonged exposure to water-insoluble fractions of PM2.5 (PM2.5 particulate) on lung injury and its molecular mechanisms. Balb/c mice were randomly exposed to PM2.5 once (acute exposure) or once every three days for a total of 6 times (prolonged exposure). Lung, BALF and blood samples were collected, and pulmonary pathophysiological alterations were analyzed. Nrf2 knockout mice were adapted to assess the involvement of Nrf2 in lung injury, and transcriptomic analysis was performed to delineate the mechanisms. Through transcriptomic analysis and validation of Nrf2 knockout mice, we found that acute exposure to PM2.5 insoluble particulates induced neutrophil infiltration-mediated airway inflammation, whereas prolonged exposure to PM2.5 insoluble particulate triggered lung fibrosis by decreasing the transcriptional activity of Nrf2, which resulted in the downregulated expression of antioxidant-related genes. In response to secondary LPS exposure, prolonged PM2.5 exposure induced more severe lung injury, indicating that prolonged PM2.5 exposure induced Nrf2 inhibition weakened its antioxidative defense capacity against oxidative stress injury, leading to the formation of pulmonary fibrosis and increasing its susceptibility to secondary bacterial infection.
Collapse
Affiliation(s)
- Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xiaolu Jin
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Zhonglan Su
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiabin Li
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Yijun Deng
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
34
|
Nie H, Liu H, Shi Y, Lai W, Liu X, Xi Z, Lin B. Effects of Different Concentrations of Oil Mist Particulate Matter on Pulmonary Fibrosis In Vivo and In Vitro. TOXICS 2022; 10:647. [PMID: 36355939 PMCID: PMC9695344 DOI: 10.3390/toxics10110647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Oil-mist particulate matter (OMPM) refers to oily particles with a small aerodynamic equivalent diameter in ambient air. Since the pathogenesis of pulmonary fibrosis (PF) has not been fully elucidated, this study aims to explore the potential molecular mechanisms of the adverse effects of exposure to OMPM at different concentrations in vivo and in vitro on PF. In this study, rats and cell lines were treated with different concentrations of OMPM in vivo and in vitro. Sirius Red staining analysis shows that OMPM exposure could cause pulmonary lesions and fibrosis symptoms. The expression of TGF-β1, α-SMA, and collagen I was increased in the lung tissue of rats. The activities of MMP2 and TIMP1 were unbalanced, and increased N-Cadherin and decreased E-Cadherin upon OMPM exposure in a dose-dependent manner. In addition, OMPM exposure could activate the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways, and the differentiation of human lung fibroblast HFL-1 cells. Therefore, OMPM exposure could induce PF by targeting the lung epithelium and fibroblasts, and activating the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways.
Collapse
|
35
|
Gao J, Lei T, Wang H, Luo K, Wang Y, Cui B, Yu Z, Hu X, Zhang F, Chen Y, Ding W, Lu Z. Dimethylarginine dimethylaminohydrolase 1 protects PM 2.5 exposure-induced lung injury in mice by repressing inflammation and oxidative stress. Part Fibre Toxicol 2022; 19:64. [PMID: 36242005 PMCID: PMC9569114 DOI: 10.1186/s12989-022-00505-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Airborne fine particulate matter with aerodynamic diameter ≤ 2.5 μm (PM2.5) pollution is associated with the prevalence of respiratory diseases, including asthma, bronchitis and chronic obstructive pulmonary disease. In patients with those diseases, circulating asymmetric dimethylarginine (ADMA) levels are increased, which contributes to airway nitric oxide deficiency, oxidative stress and inflammation. Overexpression of dimethylarginine dimethylaminohydrolase 1 (DDAH1), an enzyme degrading ADMA, exerts protective effects in animal models. However, the impact of DDAH1/ADMA on PM2.5-induced lung injury has not been investigated. METHODS Ddah1-/- and DDAH1-transgenic mice, as well as their respective wild-type (WT) littermates, were exposed to either filtered air or airborne PM2.5 (mean daily concentration ~ 50 µg/m3) for 6 months through a whole-body exposure system. Mice were also acutely exposed to 10 mg/kg PM2.5 and/or exogenous ADMA (2 mg/kg) via intratracheal instillation every other day for 2 weeks. Inflammatory response, oxidative stress and related gene expressions in the lungs were examined. In addition, RAW264.7 cells were exposed to PM2.5 and/or ADMA and the changes in intracellular oxidative stress and inflammatory response were determined. RESULTS Ddah1-/- mice developed more severe lung injury than WT mice after long-term PM2.5 exposure, which was associated with greater induction of pulmonary oxidative stress and inflammation. In the lungs of PM2.5-exposed mice, Ddah1 deficiency increased protein expression of p-p65, iNOS and Bax, and decreased protein expression of Bcl-2, SOD1 and peroxiredoxin 4. Conversely, DDAH1 overexpression significantly alleviated lung injury, attenuated pulmonary oxidative stress and inflammation, and exerted opposite effects on those proteins in PM2.5-exposed mice. In addition, exogenous ADMA administration could mimic the effect of Ddah1 deficiency on PM2.5-induced lung injury, oxidative stress and inflammation. In PM2.5-exposed macrophages, ADMA aggravated the inflammatory response and oxidative stress in an iNOS-dependent manner. CONCLUSION Our data revealed that DDAH1 has a marked protective effect on long-term PM2.5 exposure-induced lung injury.
Collapse
Affiliation(s)
- Junling Gao
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Tong Lei
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Hongyun Wang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Kai Luo
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Yuanli Wang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Bingqing Cui
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Zhuoran Yu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Xiaoqi Hu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Fang Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Wenjun Ding
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.
| | - Zhongbing Lu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquanlu, Beijing, 100049, China.
| |
Collapse
|
36
|
Liu D, Liu Y, Wang R, Feng L, Xu L, Jin C. Metabolic profiling disturbance of PM 2.5 revealed by Raman spectroscopy and mass spectrometry-based nontargeted metabolomics. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:74500-74511. [PMID: 35639313 DOI: 10.1007/s11356-022-20506-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/25/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) is an important risk factor affecting human health. Therefore, a quick method for finding metabolic targets in situ in ambient fine particulate matter is crucial. In this study, the impact of PM2.5 on human lung epithelial cells (A549) was investigated by Raman spectroscopy and mass spectrometry (MS)-based nontargeted metabolomics analysis. Raman detection indicated that exposure to PM2.5 reduced the levels of phenylalanine, tyrosine, and nucleotides. Metabolomics results not only demonstrated a significant decrease of the aforementioned metabolites but also added some important metabolite information that could not be detected by Raman spectroscopy. Our study demonstrated that Raman spectroscopy was an in situ, real-time, and rapid detection method for detecting metabolites, especially suitable for the assignment of phenylalanine/tyrosine and nucleotides, which play important roles in cellular growth. Moreover, the metabolic profiling changes observed upon PM2.5 treatment mainly involved phenylalanine, tyrosine metabolism, purine and pyrimidine metabolism, and energy metabolism, clearly demonstrating that PM2.5 can inhibit the synthesis of protein and DNA/RNA and reduce cellular energy supplies, further influencing cellular proliferation and other activities.
Collapse
Affiliation(s)
- Daojie Liu
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yumin Liu
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ruibing Wang
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Feng
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Li Xu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chengyu Jin
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
37
|
Chen H, Oliver BG, Pant A, Olivera A, Poronnik P, Pollock CA, Saad S. Effects of air pollution on human health - Mechanistic evidence suggested by in vitro and in vivo modelling. ENVIRONMENTAL RESEARCH 2022; 212:113378. [PMID: 35525290 DOI: 10.1016/j.envres.2022.113378] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Airborne particulate matter (PM) comprises both solid and liquid particles, including carbon, sulphates, nitrate, and toxic heavy metals, which can induce oxidative stress and inflammation after inhalation. These changes occur both in the lung and systemically, due to the ability of the small-sized PM (i.e. diameters ≤2.5 μm, PM2.5) to enter and circulate in the bloodstream. As such, in 2016, airborne PM caused ∼4.2 million premature deaths worldwide. Acute exposure to high levels of airborne PM (eg. during wildfires) can exacerbate pre-existing illnesses leading to hospitalisation, such as in those with asthma and coronary heart disease. Prolonged exposure to PM can increase the risk of non-communicable chronic diseases affecting the brain, lung, heart, liver, and kidney, although the latter is less well studied. Given the breadth of potential disease, it is critical to understand the mechanisms underlying airborne PM exposure-induced disorders. Establishing aetiology in humans is difficult, therefore, in-vitro and in-vivo studies can provide mechanistic insights. We describe acute health effects (e.g. exacerbations of asthma) and long term health effects such as the induction of chronic inflammatory lung disease, and effects outside the lung (e.g. liver and renal change). We will focus on oxidative stress and inflammation as this is the common mechanism of PM-induced disease, which may be used to develop effective treatments to mitigate the adverse health effect of PM exposure.
Collapse
Affiliation(s)
- Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia; Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Sydney, NSW, 2037, Australia
| | - Anushriya Pant
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Annabel Olivera
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Philip Poronnik
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Carol A Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia.
| |
Collapse
|
38
|
Shamsollahi HR, Yunesian M, Kharrazi S, Jahanbin B, Nazmara S, Rafieian S, Dehghani MH. Characterization of persistent materials of deposited PM 2.5 in the human lung. CHEMOSPHERE 2022; 301:134774. [PMID: 35500624 DOI: 10.1016/j.chemosphere.2022.134774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
Clearance of deposited urban air particulates (PMs) from the lung is vital for the protection of the lung tissue. Several studies have investigated the behavior of immune cells against these particulates in vitro and in vivo. However, the fate of particulates in the lung is yet unclear. Here, we report the results of our investigations on the clearance of particulates from the lung. Twelve normal lung tissue samples were taken from nonsmoking and non-occupationally exposed patients who needed lung lobectomy or segmentectomy. The remaining particulates were isolated from the alveolar area and extracellular matrix (ECM), separately, and their chemical composition was determined using the FE-SEM EDAX and GC-MS. Moreover, urban air PM2.5 was collected in two forms dry and washed. These were characterized too. Our results showed that none of the metals in the deposited particulates structure is fully water-soluble. After contact with mucosal liquid, the alveolar particulates included Fe, Al, Si, Ti, and Ni. These elements were absent in the PMs isolated from ECM. The organics of alveolar and ECM particulates were the same and included tetra-decane, hexadecane, and octa-decane. None of the organics present in the urban air PM2.5, such as PAHs, were available in isolated particulates from the lung tissue. This study shows that the full clearance of inhaled particulates does not happen in the lung. The immune system's primary function is detoxification by removing all components identifiable by immune cells. After that, the remained PMs will be relocated and deposited into the ECM.
Collapse
Affiliation(s)
- Hamid Reza Shamsollahi
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masud Yunesian
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran.
| | - Sharmin Kharrazi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, 14177-55469, Tehran, Iran.
| | - Behnaz Jahanbin
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Shahrokh Nazmara
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Center for Water Quality Research, Institute for Environmental Research, Tehran University of Medical Sciences, Tehran, Iran.
| | - Shahab Rafieian
- General Thoracic Surgery Ward, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hadi Dehghani
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Institute for Environmental Research, Center for Solid Waste Research, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Jia Q, Li Q, Wang Y, Zhao J, Jiang Q, Wang H, Xue W, Zhu Z, Tian L. Lung microbiome and transcriptome reveal mechanisms underlying PM 2.5 induced pulmonary fibrosis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 831:154974. [PMID: 35378184 DOI: 10.1016/j.scitotenv.2022.154974] [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: 01/04/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
Airborne fine particulate matter (PM2.5) is considered to be a risk factor for lung fibrosis, and therefore, it has attracted public attention due to its various physicochemical features and its adverse effects on health. However, little remains to be known regarding the mechanism of PM2.5-induced pulmonary fibrosis. The lung microbiota may be a potential factor involved in the adverse outcomes of pulmonary fibrosis. Meanwhile, miRNAs are thought to be key regulators that participate in the complex interplay between the host and the microbiota. Hence, to investigate the potential mechanisms of pulmonary fibrosis, and to explore the impact of PM2.5-induced alterations in miRNAs and the lung microbiota and possible interaction patterns in mice models, we took advantage of 16S rDNA gene sequencing, miRNAs sequencing (miRNAs-Seq), and mining of public databases profiling. The results of 16S rDNA analysis showed that PM2.5 interfered with the microbial community composition, resulting in Proteobacteria becoming an additional dominant phylum. In addition, differentially expressed miRNAs were enriched in HIF-1 signaling, the IL-17 signaling, as well as Th17 cell differentiation pathways, which are closely related to microbial functional pathways. Significantly, a target miRNA, miR-149-5p, may be a key factor triggering the MAPK signal pathway related to pulmonary fibrosis and disturbing the homeostasis of lung bacterial flora. These results indicate that PM2.5 may lead to interaction between lung microbiota dysbiosis and an imbalance of miRNA levels to form a vicious cycle that promotes lung fibrogenesis. The current study provides new insights into the progression of pulmonary fibrosis.
Collapse
Affiliation(s)
- Qiyue Jia
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qiuyue Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jing Zhao
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qiyue Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Hongwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Wenming Xue
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
40
|
Lakhdar R, Mumby S, Abubakar-Waziri H, Porter A, Adcock IM, Chung KF. Lung toxicity of particulates and gaseous pollutants using ex-vivo airway epithelial cell culture systems. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119323. [PMID: 35447256 DOI: 10.1016/j.envpol.2022.119323] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Air pollution consists of a multi-faceted mix of gases and ambient particulate matter (PM) with diverse organic and non-organic chemical components that contribute to increasing morbidity and mortality worldwide. In particular, epidemiological and clinical studies indicate that respiratory health is adversely affected by exposure to air pollution by both causing and worsening (exacerbating) diseases such as chronic obstructive pulmonary disease (COPD), asthma, interstitial pulmonary fibrosis and lung cancer. The molecular mechanisms of air pollution-induced pulmonary toxicity have been evaluated with regards to different types of PM of various sizes and concentrations with single and multiple exposures over different time periods. These data provide a plausible interrelationship between cellular toxicity and the activation of multiple biological processes including proinflammatory responses, oxidative stress, mitochondrial oxidative damage, autophagy, apoptosis, cell genotoxicity, cellular senescence and epithelial-mesenchymal transition. However, these molecular changes have been studied predominantly in cell lines rather than in primary bronchial or nasal cells from healthy subjects or those isolated from patients with airways disease. In addition, they have been conducted under different cell culture conditions and generally in submerged culture rather than the more relevant air-liquid interface culture and with a variety of air pollutant exposure protocols. Cell types may respond differentially to pollution delivered as an aerosol rather than being bathed in media containing agglomerations of particles. As a result, the actual pathophysiological pathways activated by different PMs in primary cells from the airways of healthy and asthmatic subjects remains unclear. This review summarises the literature on the different methodologies utilised in studying the impact of submicron-sized pollutants on cells derived from the respiratory tract with an emphasis on data obtained from primary human cell. We highlight the critical underlying molecular mechanisms that may be important in driving disease processes in response to air pollution in vivo.
Collapse
Affiliation(s)
- Ramzi Lakhdar
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Sharon Mumby
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Hisham Abubakar-Waziri
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Alexandra Porter
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Ian M Adcock
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Kian Fan Chung
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| |
Collapse
|
41
|
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: 38] [Impact Index Per Article: 12.7] [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
|
42
|
Déméautis T, Delles M, Tomaz S, Monneret G, Glehen O, Devouassoux G, George C, Bentaher A. Pathogenic Mechanisms of Secondary Organic Aerosols. Chem Res Toxicol 2022; 35:1146-1161. [PMID: 35737464 DOI: 10.1021/acs.chemrestox.1c00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Air pollution represents a major health problem and an economic burden. In recent years, advances in air pollution research has allowed particle fractionation and identification of secondary organic aerosol (SOA). SOA is formed from either biogenic or anthropogenic emissions, through a mass transfer from the gaseous mass to the particulate phase in the atmosphere. They can have deleterious impact on health and the mortality of individuals with chronic inflammatory diseases. The pleiotropic effects of SOA could involve different and interconnected pathogenic mechanisms ranging from oxidative stress, inflammation, and immune system dysfunction. The purpose of this review is to present recent findings about SOA pathogenic roles and potential underlying mechanisms focusing on the lungs; the latter being the primary exposed organ to atmospheric pollutants.
Collapse
Affiliation(s)
- Tanguy Déméautis
- Inflammation and Immunity of the Respiratory Epithelium, EA3738 (CICLY), South Medical University Hospital, Lyon 1 Claude Bernard University, 165 Chemin du grand Revoyet, 69395 Pierre-Bénite, France
| | - Marie Delles
- Inflammation and Immunity of the Respiratory Epithelium, EA3738 (CICLY), South Medical University Hospital, Lyon 1 Claude Bernard University, 165 Chemin du grand Revoyet, 69395 Pierre-Bénite, France
| | - Sophie Tomaz
- University of Lyon, Lyon 1 Claude Bernard University, CNRS, IRCELYON, 2 Avenue Albert Einstein, 69626 Villeurbanne, France
| | - Guillaume Monneret
- Pathophysiology of Immunosuppression Associated with Systemic Inflammatory Responses, EA7426 (PI3), Edouard Herriot Hospital, 5 Place d'Arsonval, 69003 Lyon, France
| | - Olivier Glehen
- Inflammation and Immunity of the Respiratory Epithelium, EA3738 (CICLY), South Medical University Hospital, Lyon 1 Claude Bernard University, 165 Chemin du grand Revoyet, 69395 Pierre-Bénite, France.,Digestive and Endocrine Surgery Department, University Hospital of Lyon, Lyon South Hospital,165 Chemin du Grand Revoyet 69495 Pierre-Benite, France
| | - Gilles Devouassoux
- Inflammation and Immunity of the Respiratory Epithelium, EA3738 (CICLY), South Medical University Hospital, Lyon 1 Claude Bernard University, 165 Chemin du grand Revoyet, 69395 Pierre-Bénite, France.,Pulmonology Department, Croix Rousse Hospital, Lyon Civil Hospices, Lyon 1 Claude Bernard University, 103 Grande Rue de la Croix-Rousse, 69004 Lyon, France
| | - Christian George
- University of Lyon, Lyon 1 Claude Bernard University, CNRS, IRCELYON, 2 Avenue Albert Einstein, 69626 Villeurbanne, France
| | - Abderrazzak Bentaher
- Inflammation and Immunity of the Respiratory Epithelium, EA3738 (CICLY), South Medical University Hospital, Lyon 1 Claude Bernard University, 165 Chemin du grand Revoyet, 69395 Pierre-Bénite, France
| |
Collapse
|
43
|
Li Y, Shi T, Li X, Sun H, Xia X, Ji X, Zhang J, Liu M, Lin Y, Zhang R, Zheng Y, Tang J. Inhaled tire-wear microplastic particles induced pulmonary fibrotic injury via epithelial cytoskeleton rearrangement. ENVIRONMENT INTERNATIONAL 2022; 164:107257. [PMID: 35486965 DOI: 10.1016/j.envint.2022.107257] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
Tire wear microplastic particles (TWMPs) are emerging microplastic pollutants that have gained increasing attention lately. However, the health effect of inhaled airborne TWMPs has never been explored before and may already be included in particulate matter morbidity and mortality. Here, we endeavored to address the preliminary study of TWMP inhalation-induced pulmonary toxic effects and its epigenetic mechanisms in C57BL/6 mice. As a result, restricted ventilatory dysfunction and fibrotic pathological changes were observed in TWMP-treaded mice. Further research found that attenuation of miR-1a-3p plays an important role in TWMP-induced lung injury. Results from in vitro study confirmed that cytoskeleton regulatory gene twinfilin-1 was one of the target genes of miR-1a-3p, and involved in cytoskeleton rearrangement caused by TWMP exposure. Mechanistically, miR-1a-3p inhibited the F-actin formation by targeting cytoskeletal regulatory proteins twinfilin-1, leading to TWMP-induced pulmonary fibrotic injury. While we are in the very early stages of explaining the role of epigenetics in TWMP-induced lung injury, the potential for the use of epigenetic marks as biomarkers is high and discoveries made in this field will likely bring us closer to better understanding this crucial mechanism.
Collapse
Affiliation(s)
- Yanting Li
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Teng Shi
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xin Li
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Huimin Sun
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Xia
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaoya Ji
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jianzhong Zhang
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Meike Liu
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Rong Zhang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Jinglong Tang
- School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
44
|
Study on Lung Injury Caused by Fine Particulate Matter and Intervention Effect of Rhodiola wallichiana. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3693231. [PMID: 35432571 PMCID: PMC9007651 DOI: 10.1155/2022/3693231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/10/2022] [Indexed: 11/18/2022]
Abstract
Objective The objective of this study was to observe the protective effect of Rhodiola wallichiana drops in a rat model of fine particulate matter (PM2.5) lung injury. Methods Forty male Wistar rats were randomly divided into blank control (NC), normal saline (NS), PM2.5-infected (PM), and Rhodiola wallichiana (RW) groups. Rats in the NC group were not provided any interventions, whereas those in the NS and PM groups were administered normal saline and PM2.5 suspension by trachea drip once a week for four weeks. Rats in the RW group were intraperitoneally administered Rhodiola wallichiana for 14 days and then administered PM2.5 suspension by trachea drip 7 days after drug delivery. The levels of inflammatory factors such as interleukin-6, interleukin-1β, and tumor necrosis factor-alpha and oxidative stress biomarkers such as 8-hydroxy-2′-deoxyguanosine, 4-hydroxynonenal, and protein carbonyl content were determined in the serum and bronchoalveolar lavage fluid by ELISA. The level of 4-hydroxynonenal in the lung was also determined using Western blotting and immunohistochemical staining. Results Levels of inflammatory factors and oxidative stress biomarkers were all increased in the PM group but decreased in the RW group. Western blotting revealed increased 4-hydroxynonenal levels in the PM group but decreased levels in the RW group. Immunohistochemical staining also provided similar results. Conclusion Rhodiola wallichiana could protect rats from inflammation and oxidative stress injury caused by PM2.5.
Collapse
|
45
|
Jin Y, Qi G, Shou Y, Li D, Liu Y, Guan H, Zhang Q, Chen S, Luo J, Xu L, Li C, Ma W, Chen N, Zheng Y, Yu D. High throughput data-based, toxicity pathway-oriented development of a quantitative adverse outcome pathway network linking AHR activation to lung damages. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:128041. [PMID: 34906874 DOI: 10.1016/j.jhazmat.2021.128041] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
The quantitative adverse outcome pathway (qAOP) is proposed to inform dose-responses at multiple biological levels for the purpose of toxicity prediction. So far, qAOP models concerning human health are scarce. Previously, we proposed 5 key molecular pathways that led aryl hydrogen receptor (AHR) activation to lung damages. The present study assembled an AOP network based on the gene expression signatures of these toxicity pathways, and validated the network using publicly available high throughput data combined with machine learning models. In addition, the AOP network was quantitatively evaluated with omics approaches and bioassays, using 16HBE-CYP1A1 cells exposed to benzo(a)pyrene (BaP), a prototypical AHR activator. Benchmark dose (BMD) analysis of transcriptomics revealed that AHR gene held the lowest BMD value, whereas AHR pathway held the lowest point of departure (PoD) compared to the other 4 pathways. Targeted bioassays were further performed to quantitatively understand the cellular responses, including ROS generation, DNA damage, interleukin-6 production, and extracellular matrix increase marked by collagen expression. Eventually, response-response relationships were plotted using nonlinear model fitting. The present study developed a highly reliable AOP model concerning human health, and validated as well as quantitatively evaluated it, and such a method is likely to be adoptable for risk assessment.
Collapse
Affiliation(s)
- Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Guangshuai Qi
- School of Public Health, Qingdao University, Qingdao, China
| | - Yingqing Shou
- School of Public Health, Qingdao University, Qingdao, China
| | - Daochuan Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuzhen Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - Heyuan Guan
- School of Public Health, Qingdao University, Qingdao, China
| | - Qianqian Zhang
- School of Public Health, Qingdao University, Qingdao, China
| | - Shen Chen
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Ningning Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
46
|
Song X, Liu J, Geng N, Shan Y, Zhang B, Zhao B, Ni Y, Liang Z, Chen J, Zhang L, Zhang Y. Multi-omics analysis to reveal disorders of cell metabolism and integrin signaling pathways induced by PM 2.5. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127573. [PMID: 34753055 DOI: 10.1016/j.jhazmat.2021.127573] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/10/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
Atmospheric fine particle pollution is known to cause many adverse health effects. However, the potential mechanisms of PM2.5-induced cytotoxicity still needs further understanding. Herein, we integrated cytotoxicity, component profiling, metabolomics and proteomics data to deeply explain the biological responses of human bronchial epithelial cells exposed to PM2.5. We observed that PM2.5 caused cell cycle arrest, calcium influx, cell damage and further induced cell apoptosis. The contents of heavy metals and 4-6 rings PAHs in PM2.5 were positively correlated with intracellular ROS, indicating that they might be the important components to induce the above cytotoxicity. Integrated metabolomics and proteomics analysis revealed the significant alterations of many metabolic processes, such as glycolysis, the citric acid cycle, amino acid metabolism and lipid metabolism. Notably, we found that PM2.5 inhibited the integrin signaling pathway, including down-regulating the protein expression of integrins and the phosphorylation of downstream signaling kinases, which might ultimately affect cell cycle progression, cell metabolism and apoptosis. This study provided a comprehensive data resource for the deep understanding of biological toxicity mechanisms caused by atmospheric fine particles in human lung-bronchial epithelium cells.
Collapse
Affiliation(s)
- Xiaoyao Song
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Jianhui Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Ningbo Geng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Yichu Shan
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Baoqin Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Yuwen Ni
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Zhen Liang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Jiping Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Lihua Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Yukui Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
47
|
Singh N, Arora N. Diesel exhaust exposure in mice induces pulmonary fibrosis by TGF-β/Smad3 signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 807:150623. [PMID: 34610407 DOI: 10.1016/j.scitotenv.2021.150623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Epidemiological studies suggest increased risk of lung cancer associated with diesel exhaust (DE) exposure. However, DE-induced lung fibrosis may lead to cancer and needs investigation. OBJECTIVES To study the mechanism involved in the initiation of DE- induced lung fibrosis. METHODS C57BL/6 mice were exposed to DE for 30 min/day for 5 days/weeks for 8 weeks. Pulmonary function test was performed to measure lung function. Mice were euthanized to collect BALF, blood, and lung tissue. BALF was used for cell count and cytokine analysis. Lung tissue slides were stained to examine structural integrity. RNA from lung tissue was used for RT-PCR. Immunoblots were performed to study fibrosis and EMT pathway. RESULTS Mice exposed to DE increase lung resistance and tissue elastance with decrease in inspiratory capacity (p < 0.05) suggesting lung function impairment. BALF showed significantly increased macrophages, neutrophils and monocytes (p < 0.01). Additionally, there was an increase in inflammation and alveolar wall thickening in lungs (p < 0.01) correlates with cellular infiltration. Macrophages had black soot deposition in lung tissue of DE exposed mice. Lung section staining revealed increase in mucus producing goblet cells for clearance of soot in lung. DE exposed lung showed increased collagen deposition and hydroxyproline residue (p < 0.01). Repetitive exposure of DE in mice lead to tissue remodeling in lung, demonstrated by fibrotic foci and smooth muscles. A significant increase in α-SMA and fibronectin (p < 0.05) in lung indicate progression of pulmonary fibrosis. TGF-β/Smad3 signaling was activated with increase in P-smad3 expression in DE exposed mice. Decreased expression of E-cadherin and increased vimentin (p < 0.05) in lungs of DE exposed mice indicate epithelial to mesenchymal transition. CONCLUSION DE exposure to mice induced lung injury and pulmonary fibrosis thereby remodeling tissue. The study demonstrates TGF-β/SMAD3 pathway involvement with an activation of EMT in DE exposed mice.
Collapse
Affiliation(s)
- Naresh Singh
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Naveen Arora
- Allergy and Immunology Section, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
48
|
He Z, Zhang H, Song Y, Yang Z, Cai Z. Exposure to ambient fine particulate matter impedes the function of spleen in the mouse metabolism of high-fat diet. JOURNAL OF HAZARDOUS MATERIALS 2022; 423:127129. [PMID: 34509742 DOI: 10.1016/j.jhazmat.2021.127129] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Epidemiological and experimental evidence has been associating the exposure with ambient fine particulate matter (PM2.5) with metabolic malfunctions such as obesity and cardiovascular disease. As the blood-filter and the important lymphatic organ, spleen participates in the regulation of metabolic balance. In this work, liquid chromatography-mass spectrometry (LC-MS)-based lipidomics, metabolomics and proteomics were performed to study the effects of PM2.5 exposure and high-fat diet (HFD) induced obesity on mice spleen. By comparing the differences in lipids, metabolites, and proteins in the spleens from PM2.5 and HFD treated mice, we discovered the individual and combined effects of the two risk factors. The results showed the PM2.5 exposure altered energy metabolism of the mice, as evidenced by the upregulation of TCA cycle. In addition, the metabolism of branched-chain amino acids was also significantly changed, which might be related to the preventive function of spleen in lipid metabolism. The PM2.5-induced metabolic changes in spleen could further aggravate the adverse impacts of HFD on mice, resulting in impeded splenic metabolism of lipids. This study revealed the effects of PM2.5 and obesity mice spleen, which might be of great significance to public health.
Collapse
Affiliation(s)
- Zhao He
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Hongna Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
49
|
Guo C, Lv S, Liu Y, Li Y. Biomarkers for the adverse effects on respiratory system health associated with atmospheric particulate matter exposure. JOURNAL OF HAZARDOUS MATERIALS 2022; 421:126760. [PMID: 34396970 DOI: 10.1016/j.jhazmat.2021.126760] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/17/2021] [Accepted: 07/25/2021] [Indexed: 06/13/2023]
Abstract
Large amounts of epidemiological evidence have confirmed the atmospheric particulate matter (PM2.5) exposure was positively correlated with the morbidity and mortality of respiratory diseases. Nevertheless, its pathogenesis remains incompletely understood, probably resulting from the activation of oxidative stress, inflammation, altered genetic and epigenetic modifications in the lung upon PM2.5 exposure. Currently, biomarker investigations have been widely used in epidemiological and toxicological studies, which may help in understanding the biologic mechanisms underlying PM2.5-elicited adverse health outcomes. Here, the emerging biomarkers to indicate PM2.5-respiratory system interactions were summarized, primarily related to oxidative stress (ROS, MDA, GSH, etc.), inflammation (Interleukins, FENO, CC16, etc.), DNA damage (8-OHdG, γH2AX, OGG1) and also epigenetic modulation (DNA methylation, histone modification, microRNAs). The identified biomarkers shed light on PM2.5-elicited inflammation, fibrogenesis and carcinogenesis, thus may favor more precise interventions in public health. It is worth noting that some inconsistent findings may possibly relate to the inter-study differentials in the airborne PM2.5 sample, exposure mode and targeted subjects, as well as methodological issues. Further research, particularly by -omics technique to identify novel, specific biomarkers, is warranted to illuminate the causal relationship between PM2.5 pollution and deleterious lung outcomes.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Songqing Lv
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yufan Liu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
50
|
Wang C, Wang J, Zheng X, Zhang J, Zhang J, Qiao G, Liu H, Zhao H, Bai J, Zhang H, Zhang Z. Epigenetic regulation is involved in traffic-related PM 2.5 aggravating allergic airway inflammation in rats. Clin Immunol 2021; 234:108914. [PMID: 34954131 DOI: 10.1016/j.clim.2021.108914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/27/2021] [Accepted: 12/20/2021] [Indexed: 11/29/2022]
Abstract
Increasing fine particulate matter (PM2.5) and epigenetic modifications are closely associated with the pathogenesis of asthma, but the definite mechanism remains unclear. The traffic-related PM2.5 exposure aggravated pulmonary inflammation and changed the methylation level of interferon gamma (Ifng) and interleukin (Il)4 genes, and then altered levels of affiliated cytokines of IFN-γ and IL-4 in rats with allergic airway inflammation. It also increased the level of miR146a and decreased the level of miR31. In addition, transcription factors of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 6 (Stat6) rose; forkhead box P3 (Foxp3) and signal transducer and activator of transcription 4 (Stat4) lowered. The traffic-related PM2.5 altered epigenetic modifications in allergic airway inflammation of rats leading to inflammation exacerbation through impaired regulatory T (Treg) cells function and T-helper type 1 (Th1)/Th2 cells imbalance, which provided a new target for the treatment and control of asthma.
Collapse
Affiliation(s)
- Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jing Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Xin Zheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jiaqi Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jingwei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Guoguo Qiao
- Teaching Experiment Center, School of Public Health, Shanxi Medical University, China
| | - Haifang Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Huichao Zhao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China.
| |
Collapse
|