1
|
Ran Z, Yang J, Liu L, Wu S, An Y, Hou W, Cheng T, Zhang Y, Zhang Y, Huang Y, Zhang Q, Wan J, Li X, Xing B, Ye Y, Xu P, Chen Z, Zhao J, Li R. Chronic PM 2.5 exposure disrupts intestinal barrier integrity via microbial dysbiosis-triggered TLR2/5-MyD88-NLRP3 inflammasome activation. ENVIRONMENTAL RESEARCH 2024; 258:119415. [PMID: 38906446 DOI: 10.1016/j.envres.2024.119415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND PM2.5, a known public health risk, is increasingly linked to intestinal disorders, however, the mechanisms of its impact are not fully understood. PURPOSE This study aimed to explore the impact of chronic PM2.5 exposure on intestinal barrier integrity and to uncover the underlying molecular mechanisms. METHODS C57BL/6 J mice were exposed to either concentrated ambient PM2.5 (CPM) or filtered air (FA) for six months to simulate urban pollution conditions. We evaluated intestinal barrier damage, microbial shifts, and metabolic changes through histopathology, metagenomics, and metabolomics. Analysis of the TLR signaling pathway was also conducted. RESULTS The mean concentration of PM2.5 in the CPM exposure chamber was consistently measured at 70.9 ± 26.8 μg/m³ throughout the study period. Our findings show that chronic CPM exposure significantly compromises intestinal barrier integrity, as indicated by reduced expression of the key tight junction proteins Occludin and Tjp1/Zo-1. Metagenomic sequencing revealed significant shifts in the microbial landscape, identifying 35 differentially abundant species. Notably, there was an increase in pro-inflammatory nongastric Helicobacter species and a decrease in beneficial bacteria, such as Lactobacillus intestinalis, Lactobacillus sp. ASF360, and Eubacterium rectale. Metabolomic analysis further identified 26 significantly altered metabolites commonly associated with intestinal diseases. A strong correlation between altered bacterial species and metabolites was also observed. For example, 4 Helicobacter species all showed positive correlations with 13 metabolites, including Lactate, Bile acids, Pyruvate and Glutamate. Additionally, increased expression levels of TLR2, TLR5, Myd88, and NLRP3 proteins were noted, and their expression patterns showed a strong correlation, suggesting a possible involvement of the TLR2/5-MyD88-NLRP3 signaling pathway. CONCLUSIONS Chronic CPM exposure induces intestinal barrier dysfunction, microbial dysbiosis, metabolic imbalance, and activation of the TLR2/5-MyD88-NLRP3 inflammasome. These findings highlight the urgent need for intervention strategies to mitigate the detrimental effects of air pollution on intestinal health and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zihan Ran
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Jingcheng Yang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China; Greater Bay Area Institute of Precision Medicine, 115 Jiaoxi Road, Guangzhou 511458, China
| | - Liang Liu
- Clinical Research Unit, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaobo Wu
- Department of Laboratory Medicine, Tinglin Hospital of Jinshan District, No. 80 Siping North Road, Shanghai 201505, China
| | - YanPeng An
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Wanwan Hou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Tianyuan Cheng
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Youyi Zhang
- School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yiqing Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yechao Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Qianyue Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China
| | - Jiaping Wan
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China
| | - Xuemei Li
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Baoling Xing
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Yuchen Ye
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Penghao Xu
- School of Biological Sciences, Georgia Insitute of Technology, Atlanta, GA, USA
| | - Zhenghu Chen
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Department of Pathology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China.
| | - Jinzhuo Zhao
- School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China.
| | - Rui Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai 200011, China.
| |
Collapse
|
2
|
Ding R, Huang L, Yan K, Sun Z, Duan J. New insight into air pollution-related cardiovascular disease: an adverse outcome pathway framework of PM2.5-associated vascular calcification. Cardiovasc Res 2024; 120:699-707. [PMID: 38636937 DOI: 10.1093/cvr/cvae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| |
Collapse
|
3
|
Tu M, Liu R, Xue J, Xiao B, Li J, Liang L. Urban Particulate Matter Triggers Meibomian Gland Dysfunction. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 38315493 PMCID: PMC10851789 DOI: 10.1167/iovs.65.2.8] [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: 08/28/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Purpose The meibomian gland (MG), as the largest modified sebaceous gland, is potentially damaged by urban particulate matter (UPM) based on epidemiological evidence, but the specific experimental mechanisms remain unknown. This study investigated the effects of UPM on MG dysfunction (MGD) in rodent models. Methods Female C57BL/6J mice received eye drops containing UPM suspension or PBS for 14 days. The proliferative capacity and progenitor of MG were evaluated by immunofluorescence. Cell apoptosis was confirmed by TUNEL assay, along with the analysis of caspase family expression. Lipid accumulation was visualized by Oil Red O staining and LipidTox staining. Ductal hyperkeratinization, neutrophil infiltration, and pyroptosis activation were detected through immunostaining. The relative gene expression and signaling pathway activation were determined by Western blot analysis. Results Administration of UPM caused MGD-like clinical signs, manifested as distinct corneal epithelial erosion, increased MG orifice occlusion, and glandular dropout. UPM exposure significantly induced progenitor loss, cellular apoptosis, and lipogenic disorder in MG, by reducing P63/Lrig1 expression and increasing cleaved caspase-8, -9, and -3 and meibum lipogenic protein (HMGCR/SREBP-1) expression. UPM-treated mice exhibited ductal hyperkeratinization and neutrophil recruitment. Simultaneously, pyroptosis was motivated, as indicated by the heightened expression of NLRP3 and the cleavage of caspase-1 and -4 and gasdermin D, as well as the increase in IL-1β and IL-18 downstream. The underlying pathological mechanisms of UPM involve the phosphorylation of mitogen-activated protein kinase and nuclear factor-κB. Conclusions These results provided direct evidence for the toxicity of UPM in MG. UPM-induced activation of pyroptosis and mitogen-activated protein kinase/nuclear factor-κB signaling pathway might account for the inflammatory MGD.
Collapse
Affiliation(s)
- Mengqian Tu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ren Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jianwen Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Bing Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lingyi Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
4
|
Caceres L, Abogunloko T, Malchow S, Ehret F, Merz J, Li X, Sol Mitre L, Magnani N, Tasat D, Mwinyella T, Spiga L, Suchanek D, Fischer L, Gorka O, Colin Gissler M, Hilgendorf I, Stachon P, Rog-Zielinska E, Groß O, Westermann D, Evelson P, Wolf D, Marchini T. Molecular mechanisms underlying NLRP3 inflammasome activation and IL-1β production in air pollution fine particulate matter (PM 2.5)-primed macrophages. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122997. [PMID: 38000727 PMCID: PMC10804998 DOI: 10.1016/j.envpol.2023.122997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Exposure to air pollution fine particulate matter (PM2.5) aggravates respiratory and cardiovascular diseases. It has been proposed that PM2.5 uptake by alveolar macrophages promotes local inflammation that ignites a systemic response, but precise underlying mechanisms remain unclear. Here, we demonstrate that PM2.5 phagocytosis leads to NLRP3 inflammasome activation and subsequent release of the pro-inflammatory master cytokine IL-1β. Inflammasome priming and assembly was time- and dose-dependent in inflammasome-reporter THP-1-ASC-GFP cells, and consistent across PM2.5 samples of variable chemical composition. While inflammasome activation was promoted by different PM2.5 surrogates, significant IL-1β release could only be observed after stimulation with transition-metal rich Residual Oil Fly Ash (ROFA) particles. This effect was confirmed in primary human monocyte-derived macrophages and murine bone marrow-derived macrophages (BMDMs), and by confocal imaging of inflammasome-reporter ASC-Citrine BMDMs. IL-1β release by ROFA was dependent on the NLRP3 inflammasome, as indicated by lack of IL-1β production in ROFA-exposed NLRP3-deficient (Nlrp3-/-) BMDMs, and by specific NLRP3 inhibition with the pharmacological compound MCC950. In addition, while ROFA promoted the upregulation of pro-inflammatory gene expression and cytokines release, MCC950 reduced TNF-α, IL-6, and CCL2 production. Furthermore, inhibition of TNF-α with a neutralizing antibody decreased IL-1β release in ROFA-exposed BMDMs. Using electron tomography, ROFA particles were observed inside intracellular vesicles and mitochondria, which showed signs of ultrastructural damage. Mechanistically, we identified lysosomal rupture, K+ efflux, and impaired mitochondrial function as important prerequisites for ROFA-mediated IL-1β release. Interestingly, specific inhibition of superoxide anion production (O2•-) from mitochondrial respiratory Complex I, but not III, blunted IL-1β release in ROFA-exposed BMDMs. Our findings unravel the mechanism by which PM2.5 promotes IL-1β release in macrophages and provide a novel link between innate immune response and exposure to air pollution PM2.5.
Collapse
Affiliation(s)
- Lourdes Caceres
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Tijani Abogunloko
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Sara Malchow
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Fabienne Ehret
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany
| | - Julian Merz
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Deborah Tasat
- Universidad Nacional de General San Martín, Escuela de Ciencia y Tecnología, B1650, General San Martín, Argentina
| | - Timothy Mwinyella
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Lisa Spiga
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Dymphie Suchanek
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Larissa Fischer
- Faculty of Biology, University of Freiburg, 79104, Freiburg im Breisgau, Germany; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Mark Colin Gissler
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Eva Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany.
| | - Timoteo Marchini
- Department of Cardiology and Angiology, University Heart Center, University of Freiburg, 79106, Freiburg im Breisgau, Germany; Faculty of Medicine, University of Freiburg, 79110, Freiburg im Breisgau, Germany; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, C1113AAD, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), C1113AAD, Buenos Aires, Argentina
| |
Collapse
|
5
|
Marchini T. Redox and inflammatory mechanisms linking air pollution particulate matter with cardiometabolic derangements. Free Radic Biol Med 2023; 209:320-341. [PMID: 37852544 DOI: 10.1016/j.freeradbiomed.2023.10.396] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Air pollution is the largest environmental risk factor for disease and premature death. Among the different components that are present in polluted air, fine particulate matter below 2.5 μm in diameter (PM2.5) has been identified as the main hazardous constituent. PM2.5 mainly arises from fossil fuel combustion during power generation, industrial processes, and transportation. Exposure to PM2.5 correlates with enhanced mortality risk from cardiovascular diseases (CVD), such as myocardial infarction and stroke. Over the last decade, it has been increasingly suggested that PM2.5 affects CVD already at the stage of risk factor development. Among the multiple biological mechanisms that have been described, the interplay between oxidative stress and inflammation has been consistently highlighted as one of the main drivers of pulmonary, systemic, and cardiovascular effects of PM2.5 exposure. In this context, PM2.5 uptake by tissue-resident immune cells in the lung promotes oxidative and inflammatory mediators release that alter tissue homeostasis at remote locations. This pathway is central for PM2.5 pathogenesis and might account for the accelerated development of risk factors for CVD, including obesity and diabetes. However, transmission and end-organ mechanisms that explain PM2.5-induced impaired function in metabolic active organs are not completely understood. In this review, the main features of PM2.5 physicochemical characteristics related to PM2.5 ability to induce oxidative stress and inflammation will be presented. Hallmark and recent epidemiological and interventional studies will be summarized and discussed in the context of current air quality guidelines and legislation, knowledge gaps, and inequities. Lastly, mechanistic studies at the intersection between redox metabolism, inflammation, and function will be discussed, with focus on heart and adipose tissue alterations. By offering an integrated analysis of PM2.5-induced effects on cardiometabolic derangements, this review aims to contribute to a better understanding of the pathogenesis and potential interventions of air pollution-related CVD.
Collapse
Affiliation(s)
- Timoteo Marchini
- Vascular Immunology Laboratory, Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany; Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris (IBIMOL), Facultad de Farmacia y Bioquímica, C1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Guan L, Shi H, Tian J, Wang X, Liu N, Wang C, Zhang Z. PM 2.5 induces the inflammatory response in rat spleen lymphocytes through autophagy activation of NLRP3 inflammasome. Mol Immunol 2023; 161:74-81. [PMID: 37506549 DOI: 10.1016/j.molimm.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023]
Abstract
Recent evidence has suggested that fine particulate matter (PM2.5) can induce inflammatory injury in spleen. However, the underlying mechanisms of injury remain enigmatic. In this study, we aim to clarify the inflammatory injury mechanisms of PM2.5 through investigating the crosstalk between autophagy and nod-like receptor protein 3 (NLRP3) inflammasome. The spleen lymphocytes were extracted from SD rats and subjected to PM2.5 and its water-soluble components. The CCK-8 assay was utilized to explore the effects of PM2.5 and its water-soluble components on lymphocytes. Then, the effects of PM2.5 and its water-soluble components exposure on autophagy and NLRP3 inflammasome were detected by qRT-PCR, western blotting, and immunofluorescence staining. The autophagosome production was observed under the transmission electron microscope. The autophagy inhibitor 3-methyladenine (3MA) following PM2.5 water-soluble components was used to investigate the regulation of NLRP3 inflammasome by autophagy. We found that PM2.5 and its water-soluble components decreased the viability of spleen lymphocytes in a dose-dependent manner. PM2.5 exposure and its water-soluble components exposure activated the autophagy and NlRP3 inflammasome, as indicated by an increased expression of LC3, P62, NLRP3, Caspase-1 p10, and increased release of IL-1β. Furthermore, the treatment with autophagy inhibitor 3MA attenuated the production of autophagosome and NLRP3 inflammasome induced by PM2.5 water-soluble components with decreased expression of NLRP3, Caspase-1 p10, and diminished production of IL-1β. These results suggested that PM2.5 and its water-soluble components could induce autophagy and inflammatory response through NLRP3 inflammasome in spleen lymphocytes, while the NLRP3 inflammasome induced by PM2.5 could be significantly alleviated by inhibition of autophagy, further providing new insights for the understanding of spleen injury caused by PM2.5.
Collapse
Affiliation(s)
- Linlin Guan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of Clinical Laboratory, Shanxi Provincial People's Hospital,Taiyuan, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of public health, Linyi Cancer Hospital, Linyi, China
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China.
| |
Collapse
|
7
|
Brugge D, Li J, Zamore W. On the Need for Human Studies of PM Exposure Activation of the NLRP3 Inflammasome. TOXICS 2023; 11:202. [PMID: 36976967 PMCID: PMC10059209 DOI: 10.3390/toxics11030202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
Particulate matter air pollution is associated with blood inflammatory biomarkers, however, the biological pathways from exposure to periferal inflammation are not well understood. We propose that the NLRP3 inflammasome is likely stimulated by ambient particulate matter, as it is by some other particles and call for more research into this pathway.
Collapse
Affiliation(s)
- Doug Brugge
- Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Jianghong Li
- Institute for Community Research, Hartford, CT 06106, USA
| | - Wig Zamore
- Somerville Transportation Equity Partnership, Somerville, MA 02145, USA
| |
Collapse
|
8
|
Feng S, Huang F, Zhang Y, Feng Y, Zhang Y, Cao Y, Wang X. The pathophysiological and molecular mechanisms of atmospheric PM 2.5 affecting cardiovascular health: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114444. [PMID: 38321663 DOI: 10.1016/j.ecoenv.2022.114444] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5, with aerodynamic diameter less than 2.5 µm) is a leading environmental risk factor for global cardiovascular health concern. OBJECTIVE To provide a roadmap for those new to this field, we reviewed the new insights into the pathophysiological and cellular/molecular mechanisms of PM2.5 responsible for cardiovascular health. MAIN FINDINGS PM2.5 is able to disrupt multiple physiological barriers integrity and translocate into the systemic circulation and get access to a range of secondary target organs. An ever-growing body of epidemiological and controlled exposure studies has evidenced a causal relationship between PM2.5 exposure and cardiovascular morbidity and mortality. A variety of cellular and molecular biology mechanisms responsible for the detrimental cardiovascular outcomes attributable to PM2.5 exposure have been described, including metabolic activation, oxidative stress, genotoxicity, inflammation, dysregulation of Ca2+ signaling, disturbance of autophagy, and induction of apoptosis, by which PM2.5 exposure impacts the functions and fates of multiple target cells in cardiovascular system or related organs and further alters a series of pathophysiological processes, such as cardiac autonomic nervous system imbalance, increasing blood pressure, metabolic disorder, accelerated atherosclerosis and plaque vulnerability, platelet aggregation and thrombosis, and disruption in cardiac structure and function, ultimately leading to cardiovascular events and death. Therein, oxidative stress and inflammation were suggested to play pivotal roles in those pathophysiological processes. CONCLUSION Those biology mechanisms have deepen insights into the etiology, course, prevention and treatment of this public health concern, although the underlying mechanisms have not yet been entirely clarified.
Collapse
Affiliation(s)
- Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China; Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China.
| | - Fangfang Huang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yuqi Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yashi Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Ying Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yunchang Cao
- The Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Xinming Wang
- Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| |
Collapse
|
9
|
Chiu YH, Chiu HP, Lin MY. Synergistic effect of probiotic and postbiotic on attenuation of PM2.5-induced lung damage and allergic response. J Food Sci 2023; 88:513-522. [PMID: 36463413 DOI: 10.1111/1750-3841.16398] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022]
Abstract
To date, few studies have been conducted on the relationship between postbiotics and air pollution, and there is limited knowledge if postbiotic and probiotic have synergistic effects. Therefore, we created a PM-induced lung inflammation mice model and demonstrated the effect of probiotic, postbiotic, and their combination treatment on attenuation of PM2.5-induced lung damage and allergic response. The mice were intratracheally given PM2.5 triggering conditions of acute lung damage and allergic response. Our results showed that individual treatment of probiotic and postbiotic reduced body weight loss by 47.1% and 48.9%, but the results did not show any effect on polarizing IFN-γ/IL-4 ratio. In addition, PM2.5-induced overactive expression of IgE treated by probiotic and postbiotic was reduced by 33.2% and 30.4%, respectively. While combination treatment of probiotic and postbiotic exerted a synergistic effect, especially considerably on improving IgE reduction by 57.1%, body weight loss by 78.3%, and IFN-γ/IL-4 ratio boost by 87.5%. To sum up the above functionality, these research findings may help establish a novel platform for postbiotic application, formulation, and mechanistic selection with regard to PM2.5-induced lung injury. PRACTICAL APPLICATION: Allergic inflammation caused by PM2.5 is not like common allergens (ex. Pollens, ovalbumin, dust mites), which simply skewing Th1/Th2 polarization to Th2. Thus using probiotics screened by Th1-skewing criteria might not be the best choice to treat on PM2.5-induced symptoms. This research proposed a combination of probiotics and postbiotics on modulating immunity homeostasis, and consequently attenuating complications of PM2.5-induced lung damage. These research findings may help establish a novel platform for postbiotic application, formulation and mechanistic selection with regard to PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Yi-Heng Chiu
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, ROC, Taiwan.,Chambio Co., Ltd., Taichung, ROC, Taiwan
| | | | - Meei-Yn Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, ROC, Taiwan
| |
Collapse
|
10
|
Li L, Song M, Zhou J, Sun X, Lei Y. Ambient particulate matter exposure causes visual dysfunction and retinal neuronal degeneration. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114231. [PMID: 36327781 DOI: 10.1016/j.ecoenv.2022.114231] [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: 07/06/2022] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 06/16/2023]
Abstract
PM2.5 pollution is related to neurotoxic and vascular effects in eye diseases such as glaucoma. This study investigates the adverse effects of PM2.5 exposure on visual function and retinal neurons. A versatile aerosol concentration enrichment system was used to expose mice to either control air or PM2.5 polluted air. Six months after PM2.5 exposure, visual function was measured by electroretinography (ERG). Hematoxylin and eosin staining and immunofluorescence staining were used for histopathological analysis. Protein markers of apoptosis, astrocytic reactivity, inflammatory cytokines, lipid peroxidation, protein nitration and DNA damage response were quantified with ELISA, western blot or detected using immunofluorescence and immunohistochemistry. After six months of exposure, PM2.5-exposed mice responded poorly to light stimuli compared with those exposed to the control air. PM2.5 exposure caused retinal thinning and reduction in the expression of retinal ganglion cell-selective marker RNA-binding protein with multiple splicing (RBPMS). Further, positive TUNEL staining was observed in the inner nucleus and outer nuclear layers of the retinae after exposure to PM2.5, which was accompanied by the activation of apoptosis signaling molecules p53, caspase-3 and Bax. PM2.5 induced the release of inflammatory cytokines including tumor necrosis factor-α and cleaved interleukin-1β. Furthermore, increased levels of 8-OHdG and γ-H2AX in the mouse retinea were indicative of DNA single and double strand breaks by PM2.5 exposure, which activated PARP-1 mediated DNA damage and repair. In conclusion, this study demonstrates sub-chronic systemic exposure to concentrated PM2.5 causes visual dysfunction and retinal neuronal degeneration. DATA AVAILABILITY: The datasets during and/or analyzed during the current study available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Liping Li
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Maomao Song
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Ji Zhou
- Shanghai Key Laboratory of Meteorology and Health, Shanghai Meteorological Bureau, Shanghai 200030, China; Shanghai Typhoon Institute, CMA, Shanghai 200030, China; Department of Atmospheric and Oceanic Sciences & Institute of Atmospheric Sciences, Fudan University, Shanghai 200031, China.
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200031, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Yuan Lei
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200031, China.
| |
Collapse
|
11
|
Sidwell A, Smith SC, Roper C. A comparison of fine particulate matter (PM 2.5) in vivo exposure studies incorporating chemical analysis. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2022; 25:422-444. [PMID: 36351256 DOI: 10.1080/10937404.2022.2142345] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The complex, variable mixtures present in fine particulate matter (PM2.5) have been well established, and associations between chemical constituents and human health are expanding. In the past decade, there has been an increase in PM2.5 toxicology studies that include chemical analysis of samples. This investigation is a crucial component for identifying the causal constituents for observed adverse health effects following exposure to PM2.5. In this review, investigations of PM2.5 that used both in vivo models were explored and chemical analysis with a focus on respiratory, cardiovascular, central nervous system, reproductive, and developmental toxicity was examined to determine if chemical constituents were considered in the interpretation of the toxicity findings. Comparisons between model systems, PM2.5 characteristics, endpoints, and results were made. A vast majority of studies observed adverse effects in vivo following exposure to PM2.5. While limited, investigations that explored connections between chemical components and measured endpoints noted significant associations between biological measurements and a variety of PM2.5 constituents including elements, ions, and organic/elemental carbon, indicating the need for such analysis. Current limitations in available data, including relatively scarce statistical comparisons between collected toxicity and chemical datasets, are provided. Future progress in this field in combination with epidemiologic research examining chemical composition may support regulatory standards of PM2.5 to protect human health.
Collapse
Affiliation(s)
- Allie Sidwell
- Department of Biology, University of Mississippi, Mississippi, MS, USA
| | - Samuel Cole Smith
- Department of Bio-Molecular Sciences, University of Mississippi, Mississippi, MS, USA
| | - Courtney Roper
- Department of Bio-Molecular Sciences, University of Mississippi, Mississippi, MS, USA
| |
Collapse
|
12
|
Ferrara F, Cordone V, Pecorelli A, Benedusi M, Pambianchi E, Guiotto A, Vallese A, Cervellati F, Valacchi G. Ubiquitination as a key regulatory mechanism for O 3-induced cutaneous redox inflammasome activation. Redox Biol 2022; 56:102440. [PMID: 36027676 PMCID: PMC9425076 DOI: 10.1016/j.redox.2022.102440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 10/26/2022] Open
Abstract
NLRP1 is one of the major inflammasomes modulating the cutaneous inflammatory responses and therefore linked to a variety of cutaneous conditions. Although NLRP1 has been the first inflammasome to be discovered, only in the past years a significant progress was achieved in understanding the molecular mechanism and the stimuli behind its activation. In the past decades a crescent number of studies have highlighted the role of air pollutants as Particulate Matter (PM), Cigarette Smoke (CS) and Ozone (O3) as trigger stimuli for inflammasomes activation, especially via Reactive Oxygen Species (ROS) mediators. However, whether NLRP1 can be modulated by air pollutants via oxidative stress and the mechanism behind its activation is still poorly understood. Here we report for the first time that O3, one of the most toxic pollutants, activates the NLRP1 inflammasome in human keratinocytes via oxidative stress mediators as hydrogen peroxide (H2O2) and 4-hydroxy-nonenal (4HNE). Our data suggest that NLRP1 represents a target protein for 4HNE adduction that possibly leads to its proteasomal degradation and activation via the possible involvement of E3 ubiquitin ligase UBR2. Of note, Catalase (Cat) treatment prevented inflammasome assemble and inflammatory cytokines release as well as NLRP1 ubiquitination in human keratinocytes upon O3 exposure. The present work is a mechanistic study that follows our previous work where we have showed the ability of O3 to induce cutaneous inflammasome activation in humans exposed to this pollutant. In conclusion, our results suggest that O3 triggers the cutaneous NLRP1 inflammasome activation by ubiquitination and redox mechanism.
Collapse
Affiliation(s)
- Francesca Ferrara
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Valeria Cordone
- Dept. of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA
| | - Mascia Benedusi
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Erika Pambianchi
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA.
| | - Anna Guiotto
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA
| | - Andrea Vallese
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA; Dept. of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy; Dept. of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Label-free detection and quantification of ultrafine particulate matter in lung and heart of mouse and evaluation of tissue injury. Part Fibre Toxicol 2022; 19:51. [PMID: 35883088 PMCID: PMC9316794 DOI: 10.1186/s12989-022-00493-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022] Open
Abstract
While it is known that air borne ultrafine particulate matter (PM) may pass through the pulmonary circulation of blood at the alveolar level between lung and heart and cross the air-blood barrier, the mechanism and effects are not completely clear. In this study the imaging method fluorescence lifetime imaging microscopy is adopted for visualization with high spatial resolution and quantification of ultrafine PM particles in mouse lung and heart tissues. The results showed that the median numbers of particles in lung of mice exposed to ultrafine particulate matter of diameter less than 2.5 µm was about 2.0 times more than that in the filtered air (FA)-treated mice, and about 1.3 times more in heart of ultrafine PM-treated mice than in FA-treated mice. Interestingly, ultrafine PM particles were more abundant in heart than lung, likely due to how ultrafine PM particles are cleared by phagocytosis and transport via circulation from lungs. Moreover, heart tissues showed inflammation and amyloid deposition. The component analysis of concentrated airborne ultrafine PM particles suggested traffic exhausts and industrial emissions as predominant sources. Our results suggest association of ultrafine PM exposure to chronic lung and heart tissue injuries. The current study supports the contention that industrial air pollution is one of the causative factors for rising levels of chronic pulmonary and cardiac diseases.
Collapse
|
14
|
Prieux R, Ferrara F, Cervellati F, Guiotto A, Benedusi M, Valacchi G. Inflammasome involvement in CS-induced damage in HaCaT keratinocytes. In Vitro Cell Dev Biol Anim 2022; 58:335-348. [PMID: 35428946 PMCID: PMC9076721 DOI: 10.1007/s11626-022-00658-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 12/14/2022]
Abstract
Cigarette smoke (CS) alters cutaneous biological processes such as redox homeostasis and inflammation response that might be involved in promoting skin inflammatory conditions. Exposure to CS has also been linked to a destabilization of the NLRP3 inflammasome in pollution target tissues such as the lung epithelium, resulting in a more vulnerable immunological response to several exogenous and endogenous stimuli related to oxidative stress. Thus, CS has an adverse effect on host defense, increasing the susceptibility to develop lung infections and pathologies. In the skin, another direct target of pollution, inflammasome disorders have been linked to an increasing number of diseases such as melanoma, psoriasis, vitiligo, atopic dermatitis, and acne, all conditions that have been connected directly or indirectly to pollution exposure. The inflammasome machinery is an important innate immune sensor in human keratinocytes. However, the role of CS in the NLRP1 and NLRP3 inflammasome in the cutaneous barrier has still not been investigated. In the present study, we were able to determine in keratinocytes exposed to CS an increased oxidative damage evaluated by 4-HNE protein adduct and carbonyl formation. Of note is that, while CS inhibited NLRP3 activation, it was able to activate NLRP1, leading to an increased secretion of the proinflammatory cytokines IL-1β and IL-18. This study highlights the importance of the inflammasome machinery in CS that more in general, in pollution, affects cutaneous tissues and the important cross-talk between different members of the NLRP inflammasome family.
Collapse
Affiliation(s)
- Roxane Prieux
- Department of Neurosciences and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Department of Neurosciences and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Department of Neurosciences and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Anna Guiotto
- Department of Neurosciences and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Mascia Benedusi
- Department of Neurosciences and Rehabilitation, University of Ferrara, Ferrara, Italy.
| | - Giuseppe Valacchi
- Department of Environment and Prevention, University of Ferrara, Ferrara, Italy.
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA.
- Department of Food and Nutrition, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|
15
|
Zeng X, Liu D, Wu W, Huo X. PM 2.5 exposure inducing ATP alteration links with NLRP3 inflammasome activation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:24445-24456. [PMID: 35064883 PMCID: PMC8783591 DOI: 10.1007/s11356-021-16405-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/04/2021] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) has been the primary air pollutant and the fourth leading risk factor for disease and death in the world. Exposure to PM2.5 is related to activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, but the mechanism of PM2.5 affecting the NLRP3 inflammasome is still unclear. Previous studies have shown that PM2.5 can cause alterations in adenosine triphosphate (ATP), and an increase in extracellular ATP and a decrease in intracellular ATP can trigger the activation process of the NLRP3 inflammasome. Therefore, we emphasize that ATP changes may be the central link and key mechanism of PM2.5 exposure that activates the NLRP3 inflammasome. This review briefly elucidates and summarizes how PM2.5 acts on ATP and subsequently further impacts the NLRP3 inflammasome. Investigation of ATP changes due to exposure to PM2.5 may be essential to regulate NLRP3 inflammasome activation and treat inflammation-related diseases such as coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| | - Dongling Liu
- Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Weidong Wu
- Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
16
|
Wang Y, Xiong L, Huang X, Ma Y, Zou L, Liang Y, Xie W, Wu Y, Chang X, Wang Z, Tang M. Intermittent exposure to airborne particulate matter induces subcellular dysfunction and aortic cell damage in BALB/c mice through multi-endpoint assessment at environmentally relevant concentrations. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127169. [PMID: 34592597 DOI: 10.1016/j.jhazmat.2021.127169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
Airborne particulate matter (PM) has been linked to cardiovascular diseases, but the underlying mechanisms remain unclear, especially at realistic exposure levels. In this study, both male and female BALB/c mice were employed to assess vascular homeostasis following a standard urban particulate matter, PM SRM1648a, via oropharyngeal aspiration at three environmentally relevant concentrations. The tested indicators included histopathological observation and lipid deposition, as well as redox biology and inflammatory responses. Furthermore, endothelial monolayer, vascular cell apoptosis and subcellular function were assessed to decipher whether episodic PM SRM1648a exposure leads to vascular damage after multiple periods of treatment, including subacute (4 weeks) and subchronic (8 weeks) durations. As a result, PM aspiration caused thickening of airways, leukocytes infiltration and adhesion to alveoli, with the spot of particles engulfed by pulmonary macrophages. Meanwhile, it induced local and systemic oxidative stress and inflammation, but limited pathological changes were captured throughout aortic tissues after either subacute or subchronic treatment. Furthermore, even in the absence of aortic impairment, vascular cell equilibrium has been disturbed by the characteristics of endothelial monolayer disintegration and cell apoptosis. Mechanistically, PM SRM1648a activated molecular markers of ER stress (BIP) and mitochondrial dynamics (DRP1) at both transcriptional and translational levels, which were strongly correlated to ox-inflammation and could serve as early checkpoints of hazardous events. In summary, our data basically indicate that episodic exposure of BALB/c mice to PM SRM1648a exerts limited effects on vascular histopathological alterations, but induces vascular cell apoptosis and subcellular dysfunction, to which local and systemic redox biology and inflammation are probably correlated.
Collapse
Affiliation(s)
- Yan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Lilin Xiong
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China; Department of Environmental Health, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu 210003, China
| | - Xiaoquan Huang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lingyue Zou
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ying Liang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Wenjing Xie
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yongya Wu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Xiaoru Chang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhihui Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW With cardiovascular disease (CVD) being the top cause of deaths worldwide, it is important to ensure healthy cardiovascular aging through enhanced understanding and prevention of adverse health effects exerted by external factors. This review aims to provide an updated understanding of environmental influences on cardiovascular aging, by summarizing epidemiological and mechanistic evidence for the cardiovascular health impact of major environmental stressors, including air pollution, endocrine-disrupting chemicals (EDCs), metals, and climate change. RECENT FINDINGS Recent studies generally support positive associations of exposure to multiple chemical environmental stressors (air pollution, EDCs, toxic metals) and extreme temperatures with increased risks of cardiovascular mortality and morbidity in the population. Environmental stressors have also been associated with a number of cardiovascular aging-related subclinical changes including biomarkers in the population, which are supported by evidence from relevant experimental studies. The elderly and patients are the most vulnerable demographic groups to majority environmental stressors. Future studies should account for the totality of individuals' exposome in addition to single chemical pollutants or environmental factors. Specific factors most responsible for the observed health effects related to cardiovascular aging remain to be elucidated.
Collapse
Affiliation(s)
- Yang Lan
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Yanta District, Xi'an City, Shaanxi Province, 710061, People's Republic of China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China
- Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Yanta District, Xi'an City, Shaanxi Province, 710061, People's Republic of China.
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China.
- Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Stachyra K, Wiśniewska A, Kiepura A, Kuś K, Rolski F, Czepiel K, Chmura Ł, Majka G, Surmiak M, Polaczek J, van Eldik R, Suski M, Olszanecki R. Inhaled silica nanoparticles exacerbate atherosclerosis through skewing macrophage polarization towards M1 phenotype. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 230:113112. [PMID: 34953274 DOI: 10.1016/j.ecoenv.2021.113112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 05/15/2023]
Abstract
BACKGROUND AND AIMS Exposure to environmental nanoparticles is related to the adverse impact on health, including cardiovascular system. Various forms of nanoparticles have been reported to interact with endothelium and induce inflammation. However, the potential role of nanoparticles in the pathogenesis of atherosclerosis and their mechanisms of action are still unclear. The aim of this study was to investigate the effect of two broadly used nanomaterials, which also occur in natural environment - silicon oxide (SiO2) and ferric oxide (Fe2O3) in the form of nanoparticles (NPs) - on the development of atherosclerosis. METHODS We used apolipoprotein E-knockout mice exposed to silica and ferric oxide nanoparticles in a whole body inhalation chamber. RESULTS Inhaled silica nanoparticles augmented the atherosclerotic lesions and increased the percentage of pro-inflammatory M1 macrophages in both the plaque and the peritoneum in apoE-/- mice. Exposure to ferric oxide nanoparticles did not enhance atherogenesis process, however, it caused significant changes in the atherosclerotic plaque composition (elevated content of CD68-positive macrophages and enlarged necrotic core accompanied by the decreased level of M1 macrophages). Both silica and ferric oxide NPs altered the phenotype of T lymphocytes in the spleen by promoting polarization towards Th17 cells. CONCLUSIONS Exposure to silica and ferric oxide nanoparticles exerts impact on atherosclerosis development and plaque composition. Pro-atherogenic abilities of silica nanoparticles are associated with activation of pro-inflammatory macrophages.
Collapse
Affiliation(s)
- Kamila Stachyra
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Anna Kiepura
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Katarzyna Kuś
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Filip Rolski
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 265 Wielicka Street, 30-663 Krakow, Poland
| | - Klaudia Czepiel
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Łukasz Chmura
- Chair of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Grzegorz Majka
- Chair of Immunology, Faculty of Medicine, Jagiellonian University Medical College, 18 Czysta Street, 31-121 Krakow, Poland
| | - Marcin Surmiak
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Street, 31-066 Krakow, Poland
| | - Justyna Polaczek
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland
| | - Rudi van Eldik
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland; Department of Chemistry and Pharmacy, University of Erlangen-Nuremberg, Egerlandstrasse 1, 91058 Erlangen, Germany
| | - Maciej Suski
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland
| | - Rafał Olszanecki
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Krakow, Poland.
| |
Collapse
|
19
|
Jiang J, Zhang G, Yu M, Gu J, Zheng Y, Sun J, Ding S. Quercetin improves the adipose inflammatory response and insulin signaling to reduce "real-world" particulate matter-induced insulin resistance. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:2146-2157. [PMID: 34365603 DOI: 10.1007/s11356-021-15829-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/31/2021] [Indexed: 06/13/2023]
Abstract
Numerous epidemiological data and experimental studies support a strong link between fine particulate matter (less than 2.5 mm in aerodynamic diameter, PM2.5) exposure and the development of insulin resistance/type 2 diabetes mellitus (T2DM). Quercetin (Que), a flavonoid compound with anti-inflammatory effects, has been confirmed to improve glucose metabolic disorders in rodents and humans. In this study, we investigated the underlying mechanisms of particulate matter (PM)-induced glucose metabolic disorder and subsequently examined the protective effect and mechanism of quercetin supplementation. Male C57BL/6 mice in the control group and PM group were exposed to ambient filtered air (FA) or PM (6 h/day, 7 days/week) for 18 weeks. Mice in the Que group were exposed to PM for 18 weeks and administered Que (50 or 100 mg/kg bw). Glucose tolerance, insulin sensitivity, and systemic and visceral white adipose tissue (vWAT) inflammatory responses were measured. The expression of proteins involved in insulin signal transduction in vWAT was assessed. Chronic PM exposure caused systemic and vWAT inflammation characterized by an increase in serum IL-6 and TNF-α levels and increased vWAT macrophage filtration, triggering NLRP3 inflammasome activation, impairing the classic glucose metabolism signal in vWAT, and inducing whole-body insulin resistance. Moreover, Que administration significantly alleviated systemic and vWAT inflammation, abolished NLRP3 inflammasome activation, and improved signaling abnormalities characteristic of insulin resistance in vWAT and adipocytes. Based on these findings, chronic PM exposure activated the NLRP3 inflammasome and subsequently caused systemic and WAT inflammation and impaired insulin signaling in vWAT and adipocytes. Most importantly, Que administration inhibited NLRP3 inflammasome-mediated inflammation and insulin signaling in vWAT to improve these adverse effects.
Collapse
Affiliation(s)
- Jinjin Jiang
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China
| | - Guofu Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Min Yu
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China
| | - Juan Gu
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China
| | - Yang Zheng
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China
| | - Jinxia Sun
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China
| | - Shibin Ding
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu Province, People's Republic of China.
| |
Collapse
|
20
|
Song L, Lei L, Jiang S, Pan K, Zeng X, Zhang J, Zhou J, Xie Y, Zhou L, Dong C, Zhao J. NLRP3 inflammasome is involved in ambient PM 2.5-related metabolic disorders in diabetic model mice but not in wild-type mice. Inhal Toxicol 2021; 33:260-267. [PMID: 34641747 DOI: 10.1080/08958378.2021.1980637] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIMS To explore the role of nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome in ambient fine particulate matter (PM2.5)-related metabolic disorders. METHODS In this study, the C57BL/6 and db/db mice were exposed to concentrated PM2.5 or filtered air (FA) using Shanghai Meteorological and Environmental Animal Exposure System (Shanghai-METAS) for 12 weeks. Indices of lipid metabolism, glucose metabolism, insulin sensitivity, and protein expression of NLRP3 inflammasome in visceral adipose tissue (VAT) were measured, respectively. RESULTS The results showed that PM2.5 exposure increased circulatory insulin, triglycerides (TG), and total cholesterol (TC), and decreased high-density lipoprotein (HDL) in both C57BL/6 and db/db mice. The levels of NLRP3-related circulatory inflammatory cytokines including both interleukin (IL)-18 and IL-1β in serum were increased in the PM2.5-exposed mice and accompanied by the elevation in fasting blood glucose and insulin. The results also showed that exposure to PM2.5 promoted the activation of NLRP3, pro-caspase-1, caspase-1, and apoptosis-associated speck-like protein containing CARD (ASC), simultaneously accompanied by the increase of IL-18 and IL-1β expression in VAT, but the statistically significant difference only found in the db/db mice, not in C57BL/6 mice. CONCLUSION The activation of NLRP3 inflammasome might be not the main mechanism of PM2.5-related metabolic disorders in wide type mice but it partly mediated the exacerbation of metabolic disorders in diabetic model mice.
Collapse
Affiliation(s)
- Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Lei Lei
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China.,Shanghai Changning Center for Disease Control and Prevention, Shanghai, China
| | - Kun Pan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Dong
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China.,Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China.,IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Gong K, Chen Y, Liu W, Wang Z. Global research trends of Apolipoprotein E in central nervous system: A scientometric analysis. Int Immunopharmacol 2021; 98:107919. [PMID: 34217139 DOI: 10.1016/j.intimp.2021.107919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 10/21/2022]
Abstract
Apolipoprotein E (apoE, protein; APOE, gene) involves in cholesterol recycling and redistribution by mediating lipoprotein pathways unique to central nervous system (CNS), which is a potential therapeutic target for diseases. We visually analyzed the research hotspots of APOE related to CNS in this work, by scientometric analysis from the Web of Science Core Collection (WOSCC) database over the past two decades. A total of 25,719 references of "APOE" and 836 references of "APOE in CNS" were retrieved from the WOSCC on October 26, 2020, and then VOSviewer 1.6.15, Citespace 5.7.R2 were used for visual analysis. Over the last two decades, the research on the field of APOE in CNS is not faddish. Although many funds, organizations, and scholars were affiliated in this field, organizations and scholars, especially the top teams in this field, still lacked close cooperation with other teams around the world. Few articles with high citations had been published in the last decade, but recent studies still lacked scale and breakthrough, and the keywords associated with APOE appeared more outdated. However, the current researches have not fully elucidated the crosstalk between APOE and neuroinflammation in CNS, some new ideas may rekindle the research enthusiasm of scholars. Although the field of APOE in CNS appeared more outdated. Based on keyword analysis, we hypothesized new ideas for further investigation of neuroinflammation would light the interest of APOE in CNS for the scholars. The crosstalk between ApoE and inflammasome may be the focus of future researches. How APOE modulates the time course or intensity of the inflammasome activation, inflammatory response (proinflammatory or anti-inflammatory), and pathological process of CNS disease deserves future attention in both basic and clinical studies. More apoE/APOE-targeted pharmacological interventions will be available for preclinical experiments and clinical trials and bring hope for patients with CNS diseases.
Collapse
Affiliation(s)
- Kai Gong
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China
| | - Yuhua Chen
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China
| | - Wei Liu
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China.
| | - Zhanxiang Wang
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China.
| |
Collapse
|
22
|
Qu S, Li K, Yang T, Yang Y, Zheng Z, Liu H, Wang X, Zhang Y, Deng S, Zhu X, Chen L, Li Y. Shenlian extract protects against ultrafine particulate matter-aggravated myocardial ischemic injury by inhibiting inflammation response via the activation of NLRP3 inflammasomes. ENVIRONMENTAL TOXICOLOGY 2021; 36:1349-1361. [PMID: 33729688 DOI: 10.1002/tox.23131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
Air pollution is a growing public health burden associated with several negative health effects, especially cardiovascular disease. Shenlian extract (SL), a traditional Chinese medicine, has the effects of clearing heat-toxin and promoting blood circulation for removing blood stasis, and it has long been used to treat cardiovascular diseases and atherosclerosis. This study explored the underlying action mechanism of SL against ultrafine particle-induced myocardial ischemic injury (UFP-MI) through network pharmacology prediction and experimental verification. Male Sprague-Dawley rats with UFP-MI were pre-treated with SL intragastrically for 7 days. All the rats were then euthanized. Inflammatory cytokine detection and histopathological analysis were performed to assess the protective effects of SL. For the mechanism study, differentially expressed genes (DEGs) were identified in UFP-MI rats treated with SL through transcriptomic analysis. Subsequently, in combination with network pharmacology, potential pathways involved in the effects of SL treatment were identified using the Internet-based Computation Platform (www.tcmip.cn) and Cytoscape 3.6.0. Further validation experiments were performed to reveal the mechanism of the therapeutic effects of SL on UFP-MI. The results show that SL significantly suppressed inflammatory cell infiltration into myocardial tissue and exhibited significant anti-inflammatory activity. Transcriptomic analysis revealed that the DEGs after SL treatment had significant anti-inflammatory, immunomodulatory, and anti-viral activities. Network pharmacology analysis illustrated that the targets of SL were mainly involved in regulation of the inflammatory response, apoptotic process, innate immune response, platelet activation, and coagulation process. By combining transcriptomic and network pharmacology data, we found that SL may exert anti-inflammatory effects by acting on the NOD-like signaling pathway to regulate immune response activation and inhibit systemic inflammation. Verification experiments revealed that SL can suppress the secretion of the inflammatory cytokines Interleukin-1 (IL-1), Interleukin-18(IL-18) and Interleukin-33(IL-33) and suppress NLRP3 inflammasome activity. The results suggested that SL can directly inhibit the activation of NLRP3 inflammasomes and reduce the release of cytokines to protect against ultrafine particulate matter-aggravated myocardial ischemic injury.
Collapse
Affiliation(s)
- Shuiqing Qu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kai Li
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ting Yang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanmin Yang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyuzn Zheng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Liu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xi Wang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Zhang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuoqiu Deng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxin Zhu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Chen
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujie Li
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Abstract
SUMMARY Exposure to air pollutants has been now associated with detrimental effects on a variety of organs, including the heart, lungs, GI tract, and brain. However, recently it has become clear that pollutant exposure can also promote the development/exacerbation of a variety of skin conditions, including premature aging, psoriasis, acne, and atopic dermatitis. Although the molecular mechanisms by which pollutant exposure results in these cutaneous pathological manifestations, it has been noticed that an inflammatory status is a common denominator of all those skin conditions. For this reason, recently, the activation of a cytosolic multiprotein complex involved in inflammatory responses (the inflammasome) that could promote the maturation of proinflammatory cytokines interleukin-1β and interleukin-18 has been hypothesized to play a key role in pollution-induced skin damage. In this review, we summarize and propose the cutaneous inflammasome as a novel target of pollutant exposure and the eventual usage of inflammasome inhibitor as new technologies to counteract pollution-induced skin damage. Possibly, the ability to inhibit the inflammasome activation could prevent cutaneous inflammaging and ameliorate the health and appearance of the skin.
Collapse
|
24
|
Pan K, Jiang S, Du X, Zeng X, Zhang J, Song L, Lei L, Zhou J, Kan H, Sun Q, Xie Y, Dong C, Zhao J. Parental PM 2 .5 exposure changes Th17/Treg cells in offspring, is associated with the elevation of blood pressure. ENVIRONMENTAL TOXICOLOGY 2021; 36:1152-1161. [PMID: 33605513 DOI: 10.1002/tox.23114] [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: 11/18/2019] [Revised: 12/10/2020] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Epidemiological evidences have indicated that fine particulate matter (PM2.5 ) exposure is associated with the occurrence and development of hypertension. The present study aims to explore the effects of parental PM2.5 exposure on blood pressure in offspring and elucidate the potential mechanism. The parental male and female C57BL/6 mice were exposed to concentrated PM2.5 or filtered air (FA) using Shanghai Meteorological and Environmental Animal Exposure System (Shanghai-METAS) for 16 weeks. At week 12, the mice were assigned to breed offspring. The male offspring mice were further exposed to PM2.5 or FA as above method. During the parental exposure, the average PM2.5 concentration was 133.7 ± 53.32 μg/m3 in PM chamber, whereas the average concentration in FA chamber was 9.4 ± 0.23 μg/m3 . Similarly, during the offspring exposure, the average concentration in PM and FA chamber were 100.76 ± 26.97 μg/m3 and 9.15 ± 0.15 μg/m3 , respectively. The PM2.5 -exposed offspring mice displayed the elevation of blood pressure, the increase of angiotensin II (Ang II), the decrease of angiotensin converting enzyme 2 (ACE2) and Ang (1-7) in serum when compared with the FA-exposed offspring mice. The similar results displayed in the proteins expression of ACE2, AT1R, and Ang (1-7) in vessel and kidney. More importantly, parental PM exposure further induced the increase in serous Ang II and the protein expression of AT1R in vessel, but decrease in ACE2 and Ang (1-7). The serous Ang II was positively associated with splenic T helper type 17 (Th17) cell population and serous IL (interleukin)-17A, but negatively associated with T regular (Treg) cell population and serous IL-10. The results suggested that parental air pollution exposure might induce the elevation of offspring blood pressure via mediate Th17- and Treg-related immune microenvironment.
Collapse
Affiliation(s)
- Kun Pan
- Department of Infectious Disease Control, Center for Disease Control and Prevention of Shangcheng in Hangzhou, Hangzhou, Zhejiang, China
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Lei Lei
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Dong
- Administrative office, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| |
Collapse
|
25
|
Cox LA, Ketelslegers HB, Lewis RJ. The shape of low-concentration dose-response functions for benzene: implications for human health risk assessment. Crit Rev Toxicol 2021; 51:95-116. [PMID: 33853483 DOI: 10.1080/10408444.2020.1860903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Are dose-response relationships for benzene and health effects such as myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) supra-linear, with disproportionately high risks at low concentrations, e.g. below 1 ppm? To investigate this hypothesis, we apply recent mode of action (MoA) and mechanistic information and modern data science techniques to quantify air benzene-urinary metabolite relationships in a previously studied data set for Tianjin, China factory workers. We find that physiologically based pharmacokinetics (PBPK) models and data for Tianjin workers show approximately linear production of benzene metabolites for air benzene (AB) concentrations below about 15 ppm, with modest sublinearity at low concentrations (e.g. below 5 ppm). Analysis of the Tianjin worker data using partial dependence plots reveals that production of metabolites increases disproportionately with increases in air benzene (AB) concentrations above 10 ppm, exhibiting steep sublinearity (J shape) before becoming saturated. As a consequence, estimated cumulative exposure is not an adequate basis for predicting risk. Risk assessments must consider the variability of exposure concentrations around estimated exposure concentrations to avoid over-estimating risks at low concentrations. The same average concentration for a specified duration is disproportionately risky if it has higher variance. Conversely, if chronic inflammation via activation of inflammasomes is a critical event for induction of MDS and other health effects, then sufficiently low concentrations of benzene are predicted not to cause increased risks of inflammasome-mediated diseases, no matter how long the duration of exposure. Thus, we find no evidence that the dose-response relationship is supra-linear at low doses; instead sublinear or zero excess risk at low concentrations is more consistent with the data. A combination of physiologically based pharmacokinetic (PBPK) modeling, Bayesian network (BN) analysis and inference, and partial dependence plots appears a promising and practical approach for applying current data science methods to advance benzene risk assessment.
Collapse
Affiliation(s)
- Louis A Cox
- Cox Associates LLC, Denver, CO, USA.,Department of Business Analytics, University of Colorado, Denver, CO, USA
| | - Hans B Ketelslegers
- Concawe Division, European Petroleum Refiners Association, Brussels, Belgium
| | - R Jeffrey Lewis
- Concawe Division, European Petroleum Refiners Association, Brussels, Belgium.,ExxonMobil Biomedical Sciences, Inc, Clinton, NJ, USA
| |
Collapse
|
26
|
Trushna T, Tripathi AK, Rana S, Tiwari RR. Nutraceuticals with anti-inflammatory and anti-oxidant properties as intervention for reducing the health effects of fine particulate matter: Potential and Prospects. Comb Chem High Throughput Screen 2021; 25:1639-1660. [PMID: 33845731 DOI: 10.2174/1386207324666210412121226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 11/22/2022]
Abstract
Air pollution, especially particulate matter pollution adversely affects human health. A growing pool of evidence has emerged which underscores the potential of individual-level nutritional interventions in attenuating the adverse health impact of exposure to PM2.5. Although controlling emission and reducing the overall levels of air pollution remains the ultimate objective globally, the sustainable achievement of such a target and thus consequent protection of human health will require a substantial amount of time and concerted efforts worldwide. In the meantime, smaller-scale individual-level interventions that can counter the inflammatory or oxidative stress effects triggered by exposure to particulate matter may be utilized to ameliorate the health effects of PM2.5 pollution. One such intervention is incorporation of nutraceuticals in the diet. Here, we present a review of the evidence generated from various in vitro, in vivo and human studies regarding the effects of different anti-inflammatory and antioxidant nutraceuticals in ameliorating the health effects of particulate matter air pollution. The studies discussed in this review suggest that these nutraceuticals when consumed as a part of the diet, or as additional supplementation, can potentially negate the cellular level adverse effects of exposure to particulate pollution. The potential benefits of adopting a non-pharmacological diet-based approach to air pollution-induced disease management have also been discussed. We argue that before a nutraceuticals-based approach can be used for widespread public adoption, further research, especially human clinical trials, is essential to confirm the beneficial action of relevant nutraceuticals and to explore the safe limits of human supplementation and the risk of side effects. Future research should focus on systematically translating bench-based knowledge regarding nutraceuticals gained from in-vitro and in-vivo studies into clinically usable nutritional guidelines.
Collapse
Affiliation(s)
- Tanwi Trushna
- Department of Environmental Health and Epidemiology, ICMR- National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Amit K Tripathi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Sindhuprava Rana
- Department of Bioinformatics, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Rajnarayan R Tiwari
- ICMR- National Institute for Research in Environmental Health (NIREH), Bhopal-462030, Madhya Pradesh. India
| |
Collapse
|
27
|
Ran Z, An Y, Zhou J, Yang J, Zhang Y, Yang J, Wang L, Li X, Lu D, Zhong J, Song H, Qin X, Li R. Subchronic exposure to concentrated ambient PM2.5 perturbs gut and lung microbiota as well as metabolic profiles in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 272:115987. [PMID: 33213950 DOI: 10.1016/j.envpol.2020.115987] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/12/2020] [Accepted: 11/01/2020] [Indexed: 05/21/2023]
Abstract
Exposure to ambient fine particular matter (PM2.5) are linked to an increased risk of metabolic disorders, leading to enhanced rate of many diseases, such as inflammatory bowel disease (IBD), cardiovascular diseases, and pulmonary diseases; nevertheless, the underlying mechanisms remain poorly understood. In this study, BALB/c mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CPM) for 2 months using a versatile aerosol concentration enrichment system(VACES). We found subchronic CPM exposure caused significant lung and intestinal damage, as well as systemic inflammatory reactions. In addition, serum and BALFs (bronchoalveolar lavage fluids) metabolites involved in many metabolic pathways in the CPM exposed mice were markedly disrupted upon PM2.5 exposure. Five metabolites (glutamate, glutamine, formate, pyruvate and lactate) with excellent discriminatory power (AUC = 1, p < 0.001) were identified to predict PM2.5 exposure related toxicities. Furthermore, subchronic exposure to CPM not only significantly decreased the richness and composition of the gut microbiota, but also the lung microbiota. Strong associations were found between several gut and lung bacterial flora changes and systemic metabolic abnormalities. Our study showed exposure to ambient PM2.5 not only caused dysbiosis in the gut and lung, but also significant systemic and local metabolic alterations. Alterations in gut and lung microbiota were strongly correlated with metabolic abnormalities. Our study suggests potential roles of gut and lung microbiota in PM2.5 caused metabolic disorders.
Collapse
Affiliation(s)
- Zihan Ran
- Department of Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhouyuan Road, 201318, Shanghai, China; Inspection and Quarantine Department, The College of Medical Technology, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Road, 201318, Shanghai, China
| | - Yanpeng An
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Fudan University, Shanghai, 200438, China
| | - Ji Zhou
- Department of Atmospheric and Oceanic Sciences & Institute of Atmospheric Sciences, Fudan University, Shanghai, China
| | - Jingmin Yang
- Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute), Chongqing, 400020, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Youyi Zhang
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Jingcheng Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Lei Wang
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Xin Li
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China; Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute), Chongqing, 400020, China
| | - Jiang Zhong
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200011, China
| | - Xingjun Qin
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Rui Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
28
|
Yan Z, Qi Z, Yang X, Ji N, Wang Y, Shi Q, Li M, Zhang J, Zhu Y. The NLRP3 inflammasome: Multiple activation pathways and its role in primary cells during ventricular remodeling. J Cell Physiol 2021; 236:5547-5563. [PMID: 33469931 DOI: 10.1002/jcp.30285] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/18/2022]
Abstract
Inflammasomes are a group of multiprotein signaling complexes located in the cytoplasm. Several inflammasomes have been identified, including NLRP1, NLRP2, NLRP3, AIM2, and NLRC4. Among them, NLRP3 was investigated in most detail, and it was reported that it can be activated by many different stimuli. Increased NLRP3 protein expression and inflammasome assembly lead to caspase-1 mediated maturation and release of IL-1β, which triggers inflammation and pyroptosis. The activation of the NLRP3 inflammasome has been widely reported in studies of tumors and neurological diseases, but relatively few studies on the cardiovascular system. Ventricular remodeling (VR) is an important factor contributing to heart failure (HF) after myocardial infarction (MI). Consequently, delaying VR is of great significance for improving heart function. Studies have shown that the NLRP3 inflammasome plays an essential role in the process of VR. Here, we reviewed the latest studies on the activation pathway of the NLRP3 inflammasome, focusing on the effects of the NLRP3 inflammasome in primary cells during VR, and finally discuss future research directions in this field.
Collapse
Affiliation(s)
- Zhipeng Yan
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhongwen Qi
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoya Yang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nan Ji
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueyao Wang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qi Shi
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meng Li
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junping Zhang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yaping Zhu
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
29
|
Niu L, Li L, Xing C, Luo B, Hu C, Song M, Niu J, Ruan Y, Sun X, Lei Y. Airborne particulate matter (PM 2.5) triggers cornea inflammation and pyroptosis via NLRP3 activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 207:111306. [PMID: 32949934 DOI: 10.1016/j.ecoenv.2020.111306] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 05/05/2023]
Abstract
Although studies have demonstrated that fine particulate matter (PM2.5) induces ocular surface damage, PM2.5 exposure causes cornea toxicity is not entirely clear. The aim of this study is to investigate the role of the nod-like receptor family pyrin domain containing three (NLRP3) inflammasome-mediated pyroptosis in PM2.5-related corneal toxicity. Human corneal epithelial cells (HCECs) were exposed to different concentrations of PM2.5, and the cell viability, expressions of NLRP3 inflammasome mediated pyroptosis axis molecules and intracellular reactive oxygen species (ROS) formation were measured in HCECs. Animal experiments were undertaken to topically apply PM2.5 suspension to mouse eyes for three months and the pyroptosis related molecules in the mouse corneas were measured. RESULTS: Our results showed a dose-dependent decrease of HCEC viability in the PM2.5-treated cells. NLRP3 inflammasome-mediated pyroptosis axis (NLRP3, ASC, GSDMD, caspase-1, IL-1β, and IL-18) were activated in the PM2.5-treated HCECs, accompanied by increased ROS formation. Further in vivo study confirmed the activation of this pathway in the mouse corneas exposed to PM2.5. In conclusion, this study provids novel evidence that PM2.5 induces corneal toxicity by triggering cell pyroptosis.
Collapse
Affiliation(s)
- Liangliang Niu
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - Liping Li
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - Chao Xing
- Animal research center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, China; Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China
| | - Chunchun Hu
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - Maomao Song
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - Jingping Niu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Ye Ruan
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), And Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China; State Key Laboratory of Medical Neurobiology, Institute of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| | - Yuan Lei
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), And Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| |
Collapse
|
30
|
Ambient PM particles reach mouse brain, generate ultrastructural hallmarks of neuroinflammation, and stimulate amyloid deposition, tangles, and plaque formation. TALANTA OPEN 2020. [DOI: 10.1016/j.talo.2020.100013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
31
|
Du X, Zeng X, Zhang J, Pan K, Song L, Zhou J, Zhou L, Xie Y, Sun Q, Ge W, Chen R, Zhao J, Kan H. Ambient fine particulate matter induced the elevation of blood pressure through ACE2/Ang(1-7) pathway: The evidence from urine metabolites. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 203:111044. [PMID: 32888613 DOI: 10.1016/j.ecoenv.2020.111044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 05/21/2023]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5) is associated with various adverse health outcomes. Although several mechanisms have been proposed including oxidative stress and inflammatory responses, the exact mechanism is still unknown. Few studies have investigated the mechanism linking PM2.5 and blood pressure (BP). In this study, we measured urinary metabolites and BP -related renin-angiotensin-aldosterone system (RAAS) to investigate the associations between ambient PM2.5 exposure and BP in healthy C57BL/6 mice. METHODS The C57BL/6 mice were exposed to ambient concentrated PM2.5 or filtered air (FA) for 16 weeks. Systolic BP and diastolic BP were measured by noninvasive BP system. The urine metabolites were quantified using the untargeted metabolomics approach. The expression of RAAS-related proteins angiotensin-converting enzyme (ACE)2, angiotensin (Ang) II, Ang (1-7) and aldosterone (ALD) were measured using Western blot and ELISA kits. RESULTS The metabolomics analysis demonstrated that PM2.5 exposure induced significant changes of some metabolites in urine, including stress hormones, amino acids, fatty acids, and lipids. Furthermore, there was an elevation of BP, increase of serous Ang II and ALD, along with the decrease of ACE2 and Ang (1-7) in kidney in the PM2.5-exposed mice compared with FA-exposed mice. CONCLUSIONS The results demonstrated that PM2.5 exposure-induced BP elevation might be associated with RAAS activation. Meanwhile, PM2.5 exposure-induced changes of stress hormone and lipid metabolism might mediate the activation of RAAS. The results suggested that the systemic stress hormone and lipid metabolism was associated with the development of hypertension.
Collapse
Affiliation(s)
- Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Kun Pan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China
| | - Li Zhou
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Wenzhen Ge
- Regeneron Pharmaceuticals Inc., New York, 10591, USA
| | - Renjie Chen
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China; Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China.
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China; Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China.
| |
Collapse
|
32
|
Shi JQ, Wang BR, Jiang T, Gao L, Zhang YD, Xu J. NLRP3 Inflammasome: A Potential Therapeutic Target in Fine Particulate Matter-Induced Neuroinflammation in Alzheimer's Disease. J Alzheimers Dis 2020; 77:923-934. [PMID: 32804134 DOI: 10.3233/jad-200359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
As one of the most harmful air pollutants, fine particulate matter (PM2.5) has been implicated as a risk factor for multiple diseases, which has generated widespread public concern. Accordingly, a growing literature links PM2.5 exposure with Alzheimer's disease (AD). A critical gap in our understanding of the adverse effects of PM2.5 on AD is the mechanism triggered by PM2.5 that contributes to disease progression. Recent evidence has demonstrated that PM2.5 can activate NLRP3 inflammasome-mediated neuroinflammation. In this review, we highlight the novel evidence between PM2.5 exposure and AD incidence, which is collected and summarized from neuropathological, epidemiological, and neuroimaging studies to in-depth deciphering molecular mechanisms. First, neuropathological, epidemiological, and neuroimaging studies will be summarized. Then, the transport pathway for central nervous system delivery of PM2.5 will be presented. Finally, the role of NLRP3 inflammasome-mediated neuroinflammation in PM2.5 induced-effects on AD will be recapitulated.
Collapse
Affiliation(s)
- Jian-Quan Shi
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Bian-Rong Wang
- Department of Neurology, Geriatric Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Li Gao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Jun Xu
- Department of Cognitive Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Dijkhoff IM, Drasler B, Karakocak BB, Petri-Fink A, Valacchi G, Eeman M, Rothen-Rutishauser B. Impact of airborne particulate matter on skin: a systematic review from epidemiology to in vitro studies. Part Fibre Toxicol 2020; 17:35. [PMID: 32711561 PMCID: PMC7382801 DOI: 10.1186/s12989-020-00366-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Air pollution is killing close to 5 million people a year, and harming billions more. Air pollution levels remain extremely high in many parts of the world, and air pollution-associated premature deaths have been reported for urbanized areas, particularly linked to the presence of airborne nano-sized and ultrafine particles. MAIN TEXT To date, most of the research studies did focus on the adverse effects of air pollution on the human cardiovascular and respiratory systems. Although the skin is in direct contact with air pollutants, their damaging effects on the skin are still under investigation. Epidemiological data suggested a correlation between exposure to air pollutants and aggravation of symptoms of chronic immunological skin diseases. In this study, a systematic literature review was conducted to understand the current knowledge on the effects of airborne particulate matter on human skin. It aims at providing a deeper understanding of the interactions between air pollutants and skin to further assess their potential risks for human health. CONCLUSION Particulate matter was shown to induce a skin barrier dysfunction and provoke the formation of reactive oxygen species through direct and indirect mechanisms, leading to oxidative stress and induced activation of the inflammatory cascade in human skin. Moreover, a positive correlation was reported between extrinsic aging and atopic eczema relative risk with increasing particulate matter exposure.
Collapse
Affiliation(s)
- Irini M Dijkhoff
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Bedia Begum Karakocak
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
- Department of Animal Sciences, PHHI NCRC, North Carolina State University, Kannapolis, NC, USA
| | | | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
34
|
Gangwar RS, Bevan GH, Palanivel R, Das L, Rajagopalan S. Oxidative stress pathways of air pollution mediated toxicity: Recent insights. Redox Biol 2020; 34:101545. [PMID: 32505541 PMCID: PMC7327965 DOI: 10.1016/j.redox.2020.101545] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/01/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023] Open
Abstract
Ambient air pollution is a leading environmental cause of morbidity and mortality globally with most of the outcomes of cardiovascular origin. While numerous mechanisms are proposed to explain the link between air pollutants and cardiovascular events, the evidence supports a role for oxidative stress as a critical intermediary pathway in the transduction of systemic responses in the cardiovascular system. Indeed, alterations in vascular function are a critical step in the development of cardiometabolic disorders such as hypertension, diabetes, and atherosclerosis. This review will provide an overview of the impact of particulate and gaseous pollutants on oxidative stress from human and animal studies published in the last five years. We discuss current gaps in knowledge and evidence to date implicating the role of oxidative stress with an emphasis on inhalational exposures. We conclude with the identification of gaps, and an exhortation for further studies to elucidate the impact of oxidative stress in air pollution mediated effects. Particulate matter air pollution is the leading risk factor for cardiovascular morbidity and mortality globally. Mechanisms of oxidative stress mediated pathways. How does lung inflammation crucial to inhalational exposure mediate systemic toxicity? Review of recent animal and human exposure studies providing insights into oxidative stress pathways.
Collapse
Affiliation(s)
- Roopesh Singh Gangwar
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Graham H Bevan
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rengasamy Palanivel
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lopa Das
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
35
|
Ferrara F, Pambianchi E, Pecorelli A, Woodby B, Messano N, Therrien JP, Lila MA, Valacchi G. Redox regulation of cutaneous inflammasome by ozone exposure. Free Radic Biol Med 2020; 152:561-570. [PMID: 31778733 DOI: 10.1016/j.freeradbiomed.2019.11.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
Several pollutants have been shown to affect skin physiology, among which ozone (O3) is one of the most toxic. Prolonged exposure to O3 leads to increased oxidative damage and cutaneous inflammation. The correlation between O3 exposure and inflammatory cutaneous conditions (atopic dermatitis, psoriasis, acne and eczema) has been already suggested, although the mechanism involved is still unclear. In the last few decades, a new multiprotein complex, the inflammasome, has been discovered and linked to tissue inflammation, including inflammatory skin conditions. The inflammasome activates inflammatory responses and contributes to the maturation of cytokines such as interleukin 1β (IL-1β) and interleukin 18. This complex is also responsive to reactive oxygen species (ROS), which plays a role in triggering the activation of the complex. On this basis it is possible hypothesize that the activation of the inflammasome could be the link between the inflammatory skin conditions associated to O3 exposure. In the present work, the ability of O3 to induce inflammasome activation was determined in different skin models, ranging from 2D (human keratinocytes) to 3D models in vitro and ex vivo. Results clearly showed that O3 exposure increased both transcript and protein levels of the main inflammasome complex, such as ASC and caspase-1. Furthermore, by using both immunofluorescence and an ASC oligomerization assay the formation of the complex was determined together with increased secreted levels of both IL-18 and IL-1β. Of note is that H2O2 and to a less extent 4HNE (both considered the main mediators of O3 interaction with cellular membranes) were also able to activate skin inflammasome while the use of catalase prevents the activation. This study demonstrated that O3 can activate cutaneous inflammasome in a redox dependent manner suggesting a possible role of this new pathway in pollution induced inflammatory skin conditions.
Collapse
Affiliation(s)
- Francesca Ferrara
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA; Dept. of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Erika Pambianchi
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA
| | - Alessandra Pecorelli
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA
| | - Brittany Woodby
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA
| | - Nicolo' Messano
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA
| | | | - Mary Ann Lila
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA
| | - Giuseppe Valacchi
- Plants for Human Health Institute, NC Research Campus, NC State University, NC, USA; Dept. of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
36
|
Su X, Tian J, Li B, Zhou L, Kang H, Pei Z, Zhang M, Li C, Wu M, Wang Q, Han B, Chu C, Pang Y, Ning J, Zhang B, Niu Y, Zhang R. Ambient PM2.5 caused cardiac dysfunction through FoxO1-targeted cardiac hypertrophy and macrophage-activated fibrosis in mice. CHEMOSPHERE 2020; 247:125881. [PMID: 31978653 DOI: 10.1016/j.chemosphere.2020.125881] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 06/10/2023]
Abstract
Plenty of epidemiological evidences have shown that ambient particulate matter (PM2.5) exposure increased the prevalence of cardiovascular disease, but the potential mechanism has not been known clearly. We established mice models by ambient PM2.5 exposure system to explore the adverse effects of PM2.5 on cardiac function in mice. Forty-eight C57BL/6 mice were randomly divided into 3 groups and exposed to filtered air (FA), unfiltered air (UA) and concentrated PM2.5 air (CA) for 8 or 16 weeks, 6 hours per day, 7 days per week, respectively. The changes of cardiac structure and function, histological analysis and related mechanism were investigated. The main manifestations of cardiac structure were cardiac hypertrophy and fibrosis in a dose- and time-dependent manner after PM2.5 exposure, which led to the decrease of cardiac systolic function. Cardiac hypertrophy in mice might be regulated by PI3K/Akt/FoxO1 signal. Cardiac fibrosis might be attributed to inflammatory infiltration caused by macrophage activation. Consequently, our data indicated that cardiac hypertrophy and fibrosis might be important factors of PM2.5-induced cardiac dysfunction in mice.
Collapse
Affiliation(s)
- Xuan Su
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Junzhi Tian
- Department of Physical Examination, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
| | - Binghua Li
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Lixiao Zhou
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Hui Kang
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zijie Pei
- Department of Pathology, Medical School, China Three Gorge University, Yichang, 443002, PR China
| | - Mengyue Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Chen Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mengqi Wu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Qian Wang
- Department of Experimental Center, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Bin Han
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Chen Chu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yaxian Pang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Jie Ning
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Boyuan Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yujie Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China.
| |
Collapse
|
37
|
Du X, Zeng X, Pan K, Zhang J, Song L, Zhou J, Chen R, Xie Y, Sun Q, Zhao J, Kan H. Metabolomics analysis of urine from healthy wild type mice exposed to ambient PM 2.5. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 714:136790. [PMID: 31982767 DOI: 10.1016/j.scitotenv.2020.136790] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/12/2020] [Accepted: 01/17/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Ambient fine particulate matter (PM2.5) exposure has been linked with various adverse health outcomes. However, the urine metabolomics changes impacted by PM2.5 have not been well elucidated. METHODS The normal healthy C57BL/6 mice were exposed to concentrated ambient PM2.5 (PM) or filtered air (FA) for four weeks using "Shanghai-METAS". The urinary metabolome was quantified using liquid/gas chromatography coupled with mass spectrometry. RESULTS There were 2213 metabolites identified in total and 163 of them were significantly different between FA- and PM-exposed mice. The KEGG pathway analysis suggested that there were nine perturbed metabolic pathways related to amino acid metabolism. The amino acid metabolism what mainly impacted by PM2.5 were beta-alanine, arginine, proline, alanine, aspartate, glutamate, phenylalanine, glycine, serine, threonine and tyrosine metabolism. Meanwhile, nineteen differential metabolites related to lipid metabolism and seven differential metabolites related to glucose homeostasis were different between FA and PM mice. Furthermore, the glucose and its metabolites were significantly increased in the PM mice compared with the FA mice. CONCLUSION The current study provided a critical information on evaluating the systemic toxicity of PM2.5. The results demonstrated that there were significant alterations in urine metabolome by short-term exposure to PM, including amino acid metabolism, lipid metabolism and glucose metabolism. The metabolomics approach might be an effective tool to evaluate the potential mechanism of PM2.5 in inducing adverse health outcomes.
Collapse
Affiliation(s)
- Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Kun Pan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China
| | - Renjie Chen
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China; Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China.
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Pan K, Jiang S, Du X, Zeng X, Zhang J, Song L, Zhou J, Kan H, Sun Q, Xie Y, Zhao J. AMPK activation attenuates inflammatory response to reduce ambient PM 2.5-induced metabolic disorders in healthy and diabetic mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 179:290-300. [PMID: 31071567 DOI: 10.1016/j.ecoenv.2019.04.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
Epidemiological and experimental studies have indicated that ambient fine particulate matter (PM2.5) exposure is associated with the occurrence and development of metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM). However, the mechanism is not clear yet, and there are few studies to explore the possible prevention measure. In this study, C57BL/6 and db/db mice were exposed to concentrated PM2.5 or filtered air using Shanghai Meteorological and Environmental Animal Exposure System (Shanghai-METAS) for 12 weeks. From week 11, some of the mice were assigned to receive a subcutaneous injection of AMPK activator (AICAR). Lipid metabolism, glucose tolerance, insulin sensitivity and energy homeostasis were measured. Meanwhile, the respiratory, systemic and visceral fat inflammatory response was detected. The results showed that PM2.5 exposure induced the impairments of glucose tolerance, insulin resistance, lipid metabolism disorders and disturbances of energy metabolism in both C57BL/6 and db/db mice. These impairments might be consistent with the increased respiratory, circulating and visceral adipose tissue (VAT) inflammatory response, which was characterized by the release of IL-6 and TNF-α in lung, serum and VAT. More importantly, AICAR administration led to the significant enhancement of energy metabolism, elevation of AMPK as well as the decreased IL-6 and TNF-α in VAT of PM2.5-exposed mice, which suggesting that AMPK activation might attenuate the inflammatory responses in VAT via the inhibition of MAPKs and NFκB. The study indicated that exposure to ambient PM2.5 under the concentration which is often seen in some developing countries could induce the occurrence of metabolic disorders in normal healthy mice and exacerbate metabolic disorders in diabetic mice. The adverse impacts of PM2.5 on insulin sensitivity, energy homeostasis, lipid metabolism and inflammatory response were associated with AMPK inhibition. AMPK activation might inhibit PM2.5-induced metabolic disorders via inhibition of inflammatory cytokines release. These findings suggested that AMPK activation is a potential therapy to prevent some of the metabolic disorders attributable to air pollution exposure.
Collapse
Affiliation(s)
- Kun Pan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Liying Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Yuquan Xie
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200127, China.
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China; Shanghai Key Laboratory of Meteorology and Health, Shanghai, China.
| |
Collapse
|