1
|
Qu S, Liang Y, Deng S, Li Y, Yang Y, Liu T, Chen L, Li Y. Pharmacotherapeutic Strategies for Fine Particulate Matter-Induced Lung and Cardiovascular Damage: Marketed Drugs, Traditional Chinese Medicine, and Biological Agents. Cardiovasc Toxicol 2025; 25:666-691. [PMID: 40113640 DOI: 10.1007/s12012-025-09985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Fine particulate matter (PM2.5), defined as airborne particles with a diameter of ≤ 2.5 μm, represents a major constituent of air pollution and has been globally implicated in exacerbating public health burdens by elevating morbidity and mortality rates associated with respiratory and cardiovascular diseases (CVDs). Adverse health effects of PM2.5 exposure manifest across diverse susceptibility profiles and durations of exposure, spanning both acute and chronic timelines. While prior reviews have predominantly focused on elucidating the toxicological mechanisms underlying PM2.5-induced pathologies, there remains a paucity of comprehensive summaries addressing therapeutic interventions for cardiopulmonary damage. This review systematically synthesizes pharmacological agents with potential therapeutic efficacy against PM2.5-induced pulmonary and cardiovascular injury. By integrating mechanistic insights with translational perspectives, this work aims to provide a foundational framework for advancing research into novel therapeutic strategies targeting PM2.5-associated cardiopulmonary disorders.
Collapse
Affiliation(s)
- Shuiqing Qu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing, 102206, China
| | - Yan Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shuoqiu Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yu Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanmin Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Tuo Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lina Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Zhang R, Li X, Li X, Zhang Q, Tang J, Liu Z, Song G, Jiang L, Yang F, Zhou J, Che H, Han Y, Qi X, Chen Y, Zhang S. Characterization of risks and pathogenesis of respiratory diseases caused by rural atmospheric PM 2.5. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169878. [PMID: 38190917 DOI: 10.1016/j.scitotenv.2024.169878] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/17/2023] [Accepted: 01/01/2024] [Indexed: 01/10/2024]
Abstract
Forty-six percent of the world's population resides in rural areas, the majority of whom belong to vulnerable groups. They mainly use cheap solid fuels for cooking and heating, which release a large amount of PM2.5 and cause adverse effects to human health. PM2.5 exhibits urban-rural differences in its health risk to the respiratory system. However, the majority of research on this issue has focused on respiratory diseases induced by atmospheric PM2.5 in urban areas, while rural areas have been ignored for a long time, especially the pathogenesis of respiratory diseases. This is not helpful for promoting environmental equity to aid vulnerable groups under PM2.5 pollution. Thus, this study focuses on rural atmospheric PM2.5 in terms of its chemical components, toxicological effects, respiratory disease types, and pathogenesis, represented by PM2.5 from rural areas in the Sichuan Basin, China (Rural SC-PM2.5). In this study, organic carbon is the most significant component of Rural SC-PM2.5. Rural SC-PM2.5 significantly induces cytotoxicity, oxidative stress, and inflammatory response. Based on multiomics, bioinformatics, and molecular biology, Rural SC-PM2.5 inhibits ribonucleotide reductase regulatory subunit M2 (RRM2) to disrupt the cell cycle, impede DNA replication, and ultimately inhibit lung cell proliferation. Furthermore, this study supplements and supports the epidemic investigation. Through an analysis of the transcriptome and human disease database, it is found that Rural SC-PM2.5 may mainly involve pulmonary hypertension, sarcoidosis, and interstitial lung diseases; in particular, congenital diseases may be ignored by epidemiological surveys in rural areas, including tracheoesophageal fistula, submucous cleft of the hard palate, and congenital hypoplasia of the lung. This study contributes to a greater scientific understanding of the health risks posed by rural PM2.5, elucidates the pathogenesis of respiratory diseases, clarifies the types of respiratory diseases, and promotes environmental equity.
Collapse
Affiliation(s)
- Ronghua Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Xiaomeng Li
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xuan Li
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; School of Public Health, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Qin Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Jiancai Tang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Zhenzhong Liu
- School of Public Health, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Guiqin Song
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Li Jiang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Fumo Yang
- College of Architecture and Environment, Sichuan University, Chengdu 610065, China
| | - Jiawei Zhou
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Hanxiong Che
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Yan Han
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xin Qi
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Yang Chen
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China.
| | - Shumin Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China.
| |
Collapse
|
3
|
Zhang J, Chen X, Li H, Liu W, Liu X, Song Y, Cong X. Selenium-enriched soybean peptides pretreatment attenuates lung injury in mice induced by fine particulate matters (PM2.5) through inhibition of TLR4/NF-κB/IκBα signaling pathway and inflammasome generation. Food Funct 2022; 13:9459-9469. [PMID: 35979800 DOI: 10.1039/d2fo01585d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study aimed to identify and prepare peptides from selenium (Se)-enriched soybeans and determine whether dietary Se-enriched soybean peptides (Se-SPep) could inhibit lung injury in mice induced by fine particulate matter 2.5 (PM2.5). BALB/c mice were randomly divided into six groups. The mice in the prevention groups were pretreated with 378 mg kg-1 of Se-SPep, soybean peptides (SPep), and Se-enriched soybean protein (Se-SPro), respectively, for four weeks. The mice in the PM2.5 exposure group received concentrated PM2.5 (15 μg per day mice) for 1 h daily from the third week for two weeks. The results showed that the leukocyte and cytokine (IL-1β, IL-6, TNF-α) levels in the bronchoalveolar lavage fluid (BALF) of the PM2.5 exposure group were higher than those in the control group. Se-SPep pretreatment decreased the IL-1β, IL-6, and TNF-α levels compared with the PM2.5 exposure group. Additionally, Se-SPep pretreatment inhibited TLR4/NF-κB/IκBα and NLRP3/ASC/caspase-1 protein expression in the lungs. In conclusion, Se-SPep pretreatment may protect the lungs of the mice against PM2.5-induced inflammation, suggesting that Se-SPep represents a potential preventative agent to inhibit PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Jian Zhang
- National Soybean Processing Industry Technology Innovation Center, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Xinwei Chen
- National Soybean Processing Industry Technology Innovation Center, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - He Li
- National Soybean Processing Industry Technology Innovation Center, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Wanlu Liu
- National Soybean Processing Industry Technology Innovation Center, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Xinqi Liu
- National Soybean Processing Industry Technology Innovation Center, Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Yao Song
- Handan Institute of Innovation, Peking University, Handan 056000, China
| | - Xin Cong
- Enshi Se-Run Health Tech Development Co. Ltd., Enshi 445000, China
| |
Collapse
|
4
|
Ding W, Cai C, Zhu X, Wang J, Jiang Q. Parthenolide ameliorates neurological deficits and neuroinflammation in mice with traumatic brain injury by suppressing STAT3/NF-κB and inflammasome activation. Int Immunopharmacol 2022; 108:108913. [PMID: 35729839 DOI: 10.1016/j.intimp.2022.108913] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) triggers a set of complex inflammation that results in secondary injury. Parthenolide (PTN) is a sesquiterpene lactone extracted from the herb Tanacetum parthenium (Feverfew) and has potent anti-inflammatory, anti-apoptosis and anti-oxidative stress effects in the central nervous system (CNS)-related diseases. This study focuses on investigating the potential neuroprotective effect of PTN on TBI and the related mechanism. METHODS Bv2 microglia, primary microglia were stimulated by LPS, and HT22 neuron cells were stimulated by OGD/R, and they were treated with different doses of PTN. The expression profiles of pro-inflammatory cytokines, proteins, oxidative stress mediators, STAT3/NF-κB pathway, inflammasomes were detected. Forty male/female C57BL/6 mice were randomly divided into the sham, PTN, TBI, and TBI + PTN groups (10 mice per group). A mouse TBI model was set up with a controlled cortical impact (CCI) device. The modified nerve severity score (mNSS) was implemented to check short-term neurological impairment in mice, and the mice's memory and learning were assessed by the Morris water maze test. The water content in the mice's brains was measured by the dry-wet method. Hematoxylin-eosin (H&E) staining, Nissl staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay were applied for neuronal apoptosis. RESULTS PTN dramatically alleviated LPS-induced inflammation in microglia, and OGD-mediated neuronal apoptosis and oxidative stress. In addition, PTN repressed LPS- or OGD-modulated STAT3/NF-κB and NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain containing 4 (NLRC4) inflammasomes activation. Administering the STAT3 inhibitor Stattic or NF-κB inhibitor Bay 11-7082 attenuated PTN-mediated effects. In vivo, PTN treatment relieved neural function deficits, brain edema and neuron apoptosis and improved the memory and learning function of TBI mice. Additionally, PTN impeded microglial activation and reduced the production of pro-inflammatory cytokines in brain lesions of TBI mice. Furthermore, PTN hindered STAT3/NF-κB and inflammasome activation. CONCLUSION PTN can curb microglial activation and neuron apoptosis by dampening the STAT3/NF-κB pathway, thus exerting neuroprotective effects in TBI mice.
Collapse
Affiliation(s)
- Wei Ding
- Department of Neurosurgery, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430000, China; Department of Neurosurgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chen Cai
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaomin Zhu
- Department of Neurology, Guangxi University of Chinese Medicine, Nanning 530200 Guangxi, China
| | - Jing Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
5
|
Fussell JC, Franklin M, Green DC, Gustafsson M, Harrison RM, Hicks W, Kelly FJ, Kishta F, Miller MR, Mudway IS, Oroumiyeh F, Selley L, Wang M, Zhu Y. A Review of Road Traffic-Derived Non-Exhaust Particles: Emissions, Physicochemical Characteristics, Health Risks, and Mitigation Measures. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:6813-6835. [PMID: 35612468 PMCID: PMC9178796 DOI: 10.1021/acs.est.2c01072] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 05/22/2023]
Abstract
Implementation of regulatory standards has reduced exhaust emissions of particulate matter from road traffic substantially in the developed world. However, nonexhaust particle emissions arising from the wear of brakes, tires, and the road surface, together with the resuspension of road dust, are unregulated and exceed exhaust emissions in many jurisdictions. While knowledge of the sources of nonexhaust particles is fairly good, source-specific measurements of airborne concentrations are few, and studies of the toxicology and epidemiology do not give a clear picture of the health risk posed. This paper reviews the current state of knowledge, with a strong focus on health-related research, highlighting areas where further research is an essential prerequisite for developing focused policy responses to nonexhaust particles.
Collapse
Affiliation(s)
- Julia C. Fussell
- National
Institute for Health Research Health Protection Research Unit in Environmental
Exposures and Health, School of Public Health, Imperial College London, London, W12 0BZ, U.K.
| | - Meredith Franklin
- Department
of Statistical Sciences, University of Toronto, Toronto, Ontario M5G 1Z5, Canada
| | - David C. Green
- National
Institute for Health Research Health Protection Research Unit in Environmental
Exposures and Health, School of Public Health, Imperial College London, London, W12 0BZ, U.K.
| | - Mats Gustafsson
- Swedish
National Road and Transport Research Institute (VTI), SE-581 95, Linköping, Sweden
| | - Roy M. Harrison
- School
of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, B15 2TT, U.K.
- Department
of Environmental Sciences / Centre of Excellence in Environmental
Studies, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - William Hicks
- National
Institute for Health Research Health Protection Research Unit in Environmental
Exposures and Health, School of Public Health, Imperial College London, London, W12 0BZ, U.K.
| | - Frank J. Kelly
- National
Institute for Health Research Health Protection Research Unit in Environmental
Exposures and Health, School of Public Health, Imperial College London, London, W12 0BZ, U.K.
| | - Franceska Kishta
- Centre
for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, U.K.
| | - Mark R. Miller
- Centre
for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, U.K.
| | - Ian S. Mudway
- National
Institute for Health Research Health Protection Research Unit in Environmental
Exposures and Health, School of Public Health, Imperial College London, London, W12 0BZ, U.K.
| | - Farzan Oroumiyeh
- Department
of Environmental Health Sciences, Jonathan and Karin Fielding School
of Public Health, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Liza Selley
- MRC
Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge,CB2 1QR, U.K.
| | - Meng Wang
- University
at Buffalo, School of Public
Health and Health Professions, Buffalo, New York 14214, United States
| | - Yifang Zhu
- Department
of Environmental Health Sciences, Jonathan and Karin Fielding School
of Public Health, University of California,
Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
6
|
Zhan B, Shen J. Mitochondria and their potential role in acute lung injury (Review). Exp Ther Med 2022; 24:479. [PMID: 35761815 PMCID: PMC9214601 DOI: 10.3892/etm.2022.11406] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Biao Zhan
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
7
|
Sharma A, Ahmad S, Ahmad T, Ali S, Syed MA. Mitochondrial dynamics and mitophagy in lung disorders. Life Sci 2021; 284:119876. [PMID: 34389405 DOI: 10.1016/j.lfs.2021.119876] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
Mitochondria are biosynthetic, bioenergetic, and signaling organelles which are critical for physiological adaptations and cellular stress responses to the environment. Various endogenous and environmental stress affects critical processes in mitochondrial homeostasis such as oxidative phosphorylation, biogenesis, mitochondrial redox system which leads to the formation of reactive oxygen species (ROS) and free radicals. The state of function of the mitochondrion is particularly dependent on the dynamic balance between mitochondrial biogenesis, fusion and fission, and degradation of damaged mitochondria by mitophagy. Increasing evidence has suggested a prominent role of mitochondrial dysfunction in the onset and progression of various lung pathologies, ranging from acute to chronic disorders. In this comprehensive review, we discuss the emerging findings of multifaceted regulations of mitochondrial dynamics and mitophagy in normal lung homeostasis as well as the prominence of mitochondrial dysfunction as a determining factor in different lung disorders such as lung cancer, COPD, IPF, ALI/ARDS, BPD, and asthma. The review will contribute to the existing understanding of critical molecular machinery regulating mitochondrial dynamic state during these pathological states. Furthermore, we have also highlighted various molecular checkpoints involved in mitochondrial dynamics, which may serve as hopeful therapeutic targets for the development of potential therapies for these lung disorders.
Collapse
Affiliation(s)
- Archana Sharma
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shaniya Ahmad
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advance Research and Studies, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
8
|
Liu M, Shi Z, Yin Y, Wang Y, Mu N, Li C, Ma H, Wang Q. Particulate matter 2.5 triggers airway inflammation and bronchial hyperresponsiveness in mice by activating the SIRT2-p65 pathway. Front Med 2021; 15:750-766. [PMID: 34181194 DOI: 10.1007/s11684-021-0839-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Exposure to particulate matter 2.5 (PM2.5) potentially triggers airway inflammation by activating nuclear factor-κB (NF-κB). Sirtuin 2 (SIRT2) is a key modulator in inflammation. However, the function and specific mechanisms of SIRT2 in PM2.5-induced airway inflammation are largely understudied. Therefore, this work investigated the mechanisms of SIRT2 in regulating the phosphorylation and acetylation of p65 influenced by PM2.5-induced airway inflammation and bronchial hyperresponsiveness. Results revealed that PM2.5 exposure lowered the expression and activity of SIRT2 in bronchial tissues. Subsequently, SIRT2 impairment promoted the phosphorylation and acetylation of p65 and activated the NF-κB signaling pathway. The activation of p65 triggered airway inflammation, increment of mucus secretion by goblet cells, and acceleration of tracheal stenosis. Meanwhile, p65 phosphorylation and acetylation, airway inflammation, and bronchial hyperresponsiveness were deteriorated in SIRT2 knockout mice exposed to PM2.5. Triptolide (a specific p65 inhibitor) reversed p65 activation and ameliorated PM2.5-induced airway inflammation and bronchial hyperresponsiveness. Our findings provide novel insights into the molecular mechanisms underlying the toxicity of PM2.5 exposure. Triptolide inhibition of p65 phosphorylation and acetylation could be an effective therapeutic approach in averting PM2.5-induced airway inflammation and bronchial hyperresponsiveness.
Collapse
Affiliation(s)
- Manling Liu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhaoling Shi
- Department of Pediatrics, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Li
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Heng Ma
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Qiong Wang
- Department of Cardiovascular Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
9
|
Wang YW, Wu YH, Zhang JZ, Tang JH, Fan RP, Li F, Yu BY, Kou JP, Zhang YY. Ruscogenin attenuates particulate matter-induced acute lung injury in mice via protecting pulmonary endothelial barrier and inhibiting TLR4 signaling pathway. Acta Pharmacol Sin 2021; 42:726-734. [PMID: 32855531 PMCID: PMC8114925 DOI: 10.1038/s41401-020-00502-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022]
Abstract
The inhalation of particulate matter (PM) is closely related to respiratory damage, including acute lung injury (ALI), characterized by inflammatory fluid edema and disturbed alveolar-capillary permeability. Ruscogenin (RUS), the main active ingredient in the traditional Chinese medicine Ophiopogonis japonicus, has been found to exhibit anti-inflammatory activity and rescue LPS-induced ALI. In this study, we investigated whether and how RUS exerted therapeutic effects on PM-induced ALI. RUS (0.1, 0.3, 1 mg·kg-1·d-1) was orally administered to mice prior to or after intratracheal instillation of PM suspension (50 mg/kg). We showed that RUS administration either prior to or after PM challenge significantly attenuated PM-induced pathological injury, lung edema, vascular leakage and VE-cadherin expression in lung tissue. RUS administration significantly decreased the levels of cytokines IL-6 and IL-1β, as well as the levels of NO and MPO in both bronchoalveolar lavage fluid (BALF) and serum. RUS administration dose-dependently suppressed the phosphorylation of NF-κB p65 and the expression of TLR4 and MyD88 in lung tissue. Furthermore, TLR4 knockout partly diminished PM-induced lung injury, and abolished the protective effects of RUS in PM-instilled mice. In conclusion, RUS effectively alleviates PM-induced ALI probably by inhibition of vascular leakage and TLR4/MyD88 signaling. TLR4 might be crucial for PM to initiate pulmonary lesion and for RUS to exert efficacy against PM-induced lung injury.
Collapse
Affiliation(s)
- Yu-Wei Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yun-Hao Wu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Zhi Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Hui Tang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Rui-Ping Fan
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jun-Ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Yuan-Yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
10
|
Chen X, Deng T, Huo T, Dong F, Deng J. MiR-140-5p/TLR4 /NF-κB signaling pathway: Crucial role in inflammatory response in 16HBE cells induced by dust fall PM 2.5. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111414. [PMID: 33080424 DOI: 10.1016/j.ecoenv.2020.111414] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 06/11/2023]
Abstract
Fine atmospheric particles with a diameter of 2.5 µm or less (PM2.5) have a large specific surface area, and carry a variety of organic matter, heavy metals, minerals and bacteria. They are an important risk factor in human non-communicable disease. To explore the molecular regulatory mechanism of the airway inflammation caused by PM2.5, an in vitro human bronchial epithelial (16HBE) cells poisoning model was deployed. Results showed that PM2.5 had a strong inhibitory effect on cells viability, and induced cells to secrete high levels of IL-6 and CXCL 8. These two biomarkers of inflammation were significantly reduced in the presence of TAK 242. TLR4, MyD88, IKK, and p-p65 proteins were highly expressed on exposure to PM2.5. Pretreatment with TAK 242 interfered with the activation of the TLR4 signaling pathway. By detecting the presence of lipopolysaccharides (LPS) in PM2.5 which had been autoclaved, it was speculated that the activation of the TLR4/NF-κB signaling pathway may be mediated by LPS. It was demonstrated using gain- and loss- function experiments that miR-140-5p negatively regulated TLR4 to mediate inflammation in 16HBE cells. The dual-luciferase reporter assay confirmed that miR-140-5p directly binds to the 3' untranslated region (3' UTR) of TLR4 to initiate biological activity. In conclusion, this study revealed a new mechanism by which the miR-140-5p/TLR4 signaling pathway mediated the inflammatory response of 16HBE cells induced by PM2.5. Differential expression of miRNA, and the activation of the TLR4/NF-κB signaling pathway induced by PM2.5 implicates PM2.5 in the pathogenesis of airway inflammation.
Collapse
Affiliation(s)
- Xiangwa Chen
- Department of Laboratory Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tao Deng
- Southwest University of Science and Technology, Mianyang 621010, China
| | - Tingting Huo
- Southwest University of Science and Technology, Mianyang 621010, China
| | - Faqin Dong
- Southwest University of Science and Technology, Mianyang 621010, China.
| | - Jianjun Deng
- Department of Laboratory Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Mianyang 404 Hospital, Mianyang 621000, China.
| |
Collapse
|
11
|
Lan Y, Ng CT, Ong CN, Yu LE, Bay BH. Transcriptomic analysis identifies dysregulated genes and functional networks in human small airway epithelial cells exposed to ambient PM 2.5. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111702. [PMID: 33396033 DOI: 10.1016/j.ecoenv.2020.111702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Cellular models exhibiting human physiological features of pseudostratified columnar epithelia, provide a more realistic approach for elucidating detailed mechanisms underlying PM2.5-induced pulmonary toxicity. In this study, we characterized the barrier and mucociliary functions of differentiated human small airway epithelial cells (SAECs), cultured at the air-liquid interface (ALI). Due to the presence of mucociliary protection, particle internalization was reduced, with a concomitant decrease in cytotoxicity in differentiated S-ALI cells, as compared to conventional submerged SAEC cultures. After 24-hour exposure to PM2.5 surrogates, 117 up-regulated genes and 156 down-regulated genes were detected in S-ALI cells, through transcriptomic analysis using the Affymetrix Clariom™ S Human Array. Transcription-level changes in >60 signaling pathways, were revealed by functional annotation of the 273 differentially expressed genes, using the PANTHER Gene List Analysis. These pathways are involved in multiple cellular processes, that include inflammation and apoptosis. Exposure to urban PM2.5 led to complex responses in airway epithelia, including a net induction of downstream pro-inflammatory and pro-apoptotic responses. Collectively, this study highlights the importance of using the more advanced ALI model rather than the undifferentiated submerged model, to avoid over-assessment of inhaled particle toxicity in human. The results of our study also suggest that reduction of ambient PM2.5 concentrations would have a protective effect on respiratory health in humans.
Collapse
Affiliation(s)
- Yang Lan
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Cheng Teng Ng
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore
| | - Choon Nam Ong
- NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Liya E Yu
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore 117576, Singapore; NUS Environmental Research Institute, National University of Singapore, Singapore 117411, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
12
|
Fine particulate matter exposure induces DNA damage by downregulating Rad51 expression in human bronchial epithelial Beas-2B cells in vitro. Toxicology 2020; 444:152581. [DOI: 10.1016/j.tox.2020.152581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
|
13
|
Pompilio A, Di Bonaventura G. Ambient air pollution and respiratory bacterial infections, a troubling association: epidemiology, underlying mechanisms, and future challenges. Crit Rev Microbiol 2020; 46:600-630. [PMID: 33059504 DOI: 10.1080/1040841x.2020.1816894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The World Health Organization attributed more than four million premature deaths to ambient air pollution in 2016. Numerous epidemiologic studies demonstrate that acute respiratory tract infections and exacerbations of pre-existing chronic airway diseases can result from exposure to ambient (outdoor) air pollution. In this context, the atmosphere contains both chemical and microbial pollutants (bioaerosols), whose impact on human health remains unclear. Therefore, this review: summarises the findings from recent studies on the association between exposure to air pollutants-especially particulate matter and ozone-and onset or exacerbation of respiratory infections (e.g. pneumonia, cystic fibrosis lung infection, and tuberculosis); discusses the mechanisms underlying the relationship between air pollution and respiratory bacterial infections, which is necessary to define prevention and treatment strategies; demonstrates the relevance of air pollution modelling in investigating and preventing the impact of exposure to air pollutants on human health; and outlines future actions required to improve air quality and reduce morbidity and mortality related to air pollution.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
14
|
Dai MY, Chen FF, Wang Y, Wang MZ, Lv YX, Liu RY. Particulate matters induce acute exacerbation of allergic airway inflammation via the TLR2/NF-κB/NLRP3 signaling pathway. Toxicol Lett 2019; 321:146-154. [PMID: 31836503 DOI: 10.1016/j.toxlet.2019.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Exposure to particulate matters (PMs) can lead to an acute exacerbation of allergic airway diseases, increasing the severity of symptoms and mortality. However, little is known about the underlying molecular mechanism. This study aimed to investigate the effects of PMs on acute exacerbation of allergic airway inflammation and seek potential therapeutic targets. METHODS Non-allergic control and ovalbumin (OVA)-allergic wide-type (WT) and Toll-like receptor 2 knockout (Tlr2-/-) mice were exposed to 100 μg of PM (diameter 5.85 μm) or saline by the oropharyngeal instillation. The responses were examined three days after exposure. In the RAW264.7 macrophage cell line, Tlr2 was knocked down by small-interfering RNA or the NF-κB inhibitor JSH-23 was used, and then the cells were stimulated with PMs for 12 h before comparison of the inflammatory responses. RESULTS PM exposure led to increased inflammatory cell recruitment and airway intensity of PAS + staining in OVA-allergic WT mice, accompanied with an accumulation of inflammatory cells and elevated inflammatory cytokines, such as IL-6 and IL-18, in the bronchoalveolar lavage fluid (BALF). Furthermore, the protein levels of TLR2 and the NLRP3 inflammasome were elevated concomitantly with the airway inflammation post-OVA/PMs challenge. Tlr2 deficiency effectively inhibited the airway inflammation, including pulmonary inflammatory cell recruitment, mucus secretion, serum OVA-specific immunoglobulin E (IgE), and BALF inflammatory cytokine production. Additionally, the P-induced NLRP3 activation in the RAW 264.7 cell line was diminished by the knockdown of Tlr2 or JSH-23 treatment in vitro. CONCLUSION Our results indicated that PMs exacerbate the allergic airway inflammation mediated by the TLR2/ NF-κB/NLRP3 signaling pathway. Inhibition of NF-κB seems to be a possible treatment.
Collapse
Affiliation(s)
- Meng-Yuan Dai
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fang-Fang Chen
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yong Wang
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mu-Zi Wang
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yun-Xiang Lv
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Rong-Yu Liu
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
15
|
Pan Y, Wang M, Wang L, Xu G, Baloch AR, Kashif J, Fan J, Yu S. Interleukin-1 beta induces autophagy of mouse preimplantation embryos and improves blastocyst quality. J Cell Biochem 2019; 121:1087-1100. [PMID: 31453635 DOI: 10.1002/jcb.29345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/13/2019] [Indexed: 01/08/2023]
Abstract
Autophagy is one of the basic cellular mechanism during preimplantation development of mammalian embryos, and it plays crucial role in several physiological processes. It is induced by interleukin (IL)-1β in mammalian cells. Our present study shows that IL-1β is important for autophagy activation in embryo development. Our in vitro culture system analysis shows effect of IL-1β in medium on the development of mouse embryos and it was found to be concentration dependent. A preimplantation embryo culture using medium containing IL-1β did not improve cleavage and blastocyst development rates of mouse embryos; however, blastocyst quality was significantly improved by increasing total cell number, especially in supplementary 20 ng/mL IL-1β. Furthermore, autophagy activation mainly occurs in 2 to 4 cell embryo and blastocyst, 20 ng/mL IL-1β into culture medium can effectively enhance levels of messenger RNA and protein of autophagy-related-factors in 2 to 4 cell embryos and blastocyst, while these factors reduce in VGX-1027 (IL-1β inhibitor) groups that also reduce the quality of blastocyst. Effects of IL-1β on the development of embryo reduced in 20 ng/mL IL-1β supplemented group when 5 mM 3-methyladenine (3-MA) was also added, which used to inhibit autophagy activation in endogenous PtdIns3Ks signal pathway. Our current results show that exogenous IL-1β can effectively induce autophagy in mouse embryos at stages of 2 to 8 cell and blastocyst, that also help to improve the quality of blastocyst.
Collapse
Affiliation(s)
- Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Libin Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Gengquan Xu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Abdul Rasheed Baloch
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Jam Kashif
- Department of Veterinary Medicine, Sindh Agriculture University, Tandojam, Pakistan
| | - Jiangfeng Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Xu M, Wang L, Wang M, Wang H, Zhang H, Chen Y, Wang X, Gong J, Zhang JJ, Adcock IM, Chung KF, Li F. Mitochondrial ROS and NLRP3 inflammasome in acute ozone-induced murine model of airway inflammation and bronchial hyperresponsiveness. Free Radic Res 2019; 53:780-790. [PMID: 31185753 DOI: 10.1080/10715762.2019.1630735] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a key mechanism underlying ozone-induced lung injury. Mitochondria can release mitochondrial reactive oxidative species (mtROS), which may lead to the activation of NLRP3 inflammasome. The goal of this study was to examine the roles of mtROS and NLRP3 inflammasome in acute ozone-induced airway inflammation and bronchial hyperresponsiveness (BHR). C57/BL6 mice (n = 8/group) were intraperitoneally treated with vehicle (phosphate buffered saline, PBS) or mitoTEMPO (mtROS inhibitor, 20 mg/kg), or orally treated with VX-765 (caspse-1 inhibitor, 100 mg/kg) 1 h before the ozone exposure (2.5 ppm, 3 h). Compared to the PBS-treated ozone-exposed mice, mitoTEMPO reduced the level of total malondialdehyde in bronchoalveolar lavage (BAL) fluid and increased the expression of mitochondrial complexes II and IV in the lung 24 h after single ozone exposure. VX-765 inhibited ozone-induced BHR, BAL total cells including neutrophils and eosinophils, and BAL inflammatory cytokines including IL-1α, IL-1β, KC, and IL-6. Both mitoTEMPO and VX-765 reduced ozone-induced mtROS and inhibited capase-1 activity in lung tissue whilst VX-765 further inhibited DRP1 and MFF expression, increased MFN2 expression, and down-regulated caspase-1 expression in the lung tissue. These results indicate that acute ozone exposure induces mitochondrial dysfunction and NLRP3 inflammasome activation, while the latter has a critical role in the pathogenesis of ozone-induced airway inflammation and BHR.
Collapse
Affiliation(s)
- Mengmeng Xu
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Lei Wang
- b Department of Otorhinolaryngology and Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Muyun Wang
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Hanying Wang
- b Department of Otorhinolaryngology and Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Hai Zhang
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Yuqing Chen
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Xiaohui Wang
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| | - Jicheng Gong
- c Duke Global Health Institute and Nicholas School of the Environment, Duke University , Durham , NC , USA.,d College of Environmental Sciences and Engineering and BIC-ESAT, Peking University , Beijing , PR China
| | - Junfeng Jim Zhang
- d College of Environmental Sciences and Engineering and BIC-ESAT, Peking University , Beijing , PR China.,e Global Health Research Center, Duke Kun Shan University , Kunshan , PR China
| | - Ian M Adcock
- f Airway Disease Section, National Heart and Lung Institute, Imperial College London , London , UK.,g Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, University of Newcastle , Newcastle , Australia
| | - Kian Fan Chung
- f Airway Disease Section, National Heart and Lung Institute, Imperial College London , London , UK
| | - Feng Li
- a Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University , Shanghai , PR China
| |
Collapse
|
17
|
Zhang J, Wu J, Liu F, Tong L, Chen Z, Chen J, He H, Xu R, Ma Y, Huang C. Neuroprotective effects of anthocyanins and its major component cyanidin-3-O-glucoside (C3G) in the central nervous system: An outlined review. Eur J Pharmacol 2019; 858:172500. [PMID: 31238064 DOI: 10.1016/j.ejphar.2019.172500] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 01/02/2023]
Abstract
Anthocyanins, a class of water soluble flavonoids extracted from plants like berries and soybean seed, have been shown to display obvious anti-oxidative, anti-inflammatory, and anti-apoptotic activities. They are recommended as a supplementation for prevention and/or treatment of disorders ranging from cardiovascular disease, metabolic syndrome, and cancer. In the central nervous system (CNS), anthocyanins and its major component cyanidin-3-O-glucoside (C3G) have been reported to produce preventive and/or therapeutic activities in a wide range of disorders, such as cerebral ischemia, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and glioblastoma. Both anthocyanins and C3G can also affect some important processes in aging, including neuronal apoptosis and death as well as learning and memory impairment. Further, the anthocyanins and C3G have been shown to prevent neuro-toxicities induced by different toxic factors, such as lipopolysaccharide, hydrogen peroxide, ethanol, kainic acid, acrolein, glutamate, and scopolamine. Mechanistic studies have shown that inhibition of oxidative stress and neuroinflammation are two critical mechanisms by which anthocyanins and C3G produce protective effects in CNS disorder prevention and/or treatment. Other mechanisms, including suppression of c-Jun N-terminal kinase (JNK) activation, amelioration of cellular degeneration, activation of the brain-derived neurotrophic factor (BDNF) signaling, and restoration of Ca2+ and Zn2+ homeostasis, may also mediate the neuroprotective effects of anthocyanins and C3G. In this review, we summarize the pharmacological effects of anthocyanins and C3G in CNS disorders as well as their possible mechanisms, aiming to get a clear insight into the role of anthocyanins in the CNS.
Collapse
Affiliation(s)
- Jinlin Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, #30 Tongyang North Road, Nantong, 226361, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou, 215021, Jiangsu, China
| | - Fengguo Liu
- Department of Neurology, Danyang People's Hospital, Danyang, 212300, Jiangsu, China
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong, 226001, Jiangsu, China
| | - Jinliang Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Haiyan He
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Rong Xu
- Department of Pharmacy and Medical Technology, Nantong Health College of Jiangsu Province, #288, Zhenxing East Road, Nantong Economic Development Zone, Nantong, 226009, Jiangsu, China
| | - Yaoying Ma
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
18
|
TRPV1 and TRPA1 in Lung Inflammation and Airway Hyperresponsiveness Induced by Fine Particulate Matter (PM 2.5). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7450151. [PMID: 31281589 PMCID: PMC6589236 DOI: 10.1155/2019/7450151] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/02/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022]
Abstract
Exposure to fine particulate matter (PM2.5) has been associated with lung inflammation and airway hyperresponsiveness (AHR). Transient receptor potential (TRP) vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1) both may play important roles in lung inflammation and AHR. We investigated whether PM2.5-induced lung inflammation and AHR could be prevented by blocking TRPV1 and TRPA1 channels. Mice were injected intraperitoneally with AMG9810 (30 mg/kg, a TRPV1 antagonist) or A967079 (30 mg/kg, a TRPA1 antagonist) or their combination or vehicle (PBS) one hour before intranasal instillation of PM2.5 (7.8 mg/kg) or vehicle (PBS) for two consecutive days, and then the mice were studied 24 h later. All pretreatments inhibited PM2.5-induced AHR and inflammatory infiltration in the lung tissue and decreased inflammatory cytokine levels in the bronchoalveolar lavage fluid, together with oxidant levels in the lung. AMG9810 inhibited MFF expression and increased MFN2 expression while A967079 inhibited DRP1 expression and increased OPA1 expression; combined pretreatment reduced MFF and DPR1 expression and increased MFN2 and OPA1 expression. All pretreatments inhibited the activation of the TLR4/NF-κB pathway, while A967079 alone, and combined with AMG9810 also reduced the activation of the NLRP3/caspase-1 pathway. Both TRPV1 and TRPA1 channels play an important role in PM2.5-induced lung inflammation and AHR. However, inhibition of the TRPA1 channel or combined inhibition of TRPA1 and TRPV1 channels resulted in greater inhibitory effect on PM2.5-induced lung injury through regulating the mitochondrial fission/fusion proteins and inhibiting the TLR4/NF-κB and NLRP3/caspase-1 pathways.
Collapse
|