1
|
Sun W, Dong Q, Zhang Y, Wang H, Wang Y, Yuan W, Wang L, Shi X, Feng Y, Wang H, Wang X, Ren Y, Wang L, Lei L, Song W. Evidence of the correlation between air pollution and different types of birth defects: based on a distribution-lag non-linear model. Front Public Health 2025; 13:1562461. [PMID: 40270732 PMCID: PMC12014725 DOI: 10.3389/fpubh.2025.1562461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction As global fertility rates decline, exploring the root causes of birth defects (BDs) becomes urgent. Air pollution, with its ability to penetrate the placental barrier as exogenous toxins, has garnered notable attention in this regard. Methods BD data was collected from five hospitals in Changzhi City birth from 2019 to 2021, air quality data originated from hourly observations at five monitoring stations within the city. Using the distributed lag non-linear model (DLNM), the study aimed to determine the non-linear exposure-lag-effect relationship, evaluating the delayed impact of weekly air pollution on fetal BD risk. During the period under study, the prevalence of BDs was 19.95‰. Results Our findings indicate that exposure to air pollutants during early and mid-pregnancy elevated the risk of BDs. Specifically, for each 10 μg/m3 increase of SO2, NO2, PM10, PM2.5, O3, and CO, the risk of congenital heart defects (CHDs) increased. Peaking at specific gestational weeks: SO2 at week 17, NO2 at week 23, PM10 at week 21, PM2.5 at week 16, O3 at week 8, and CO at week 40. Additionally, a rise of 10 μg/m3 in PM10 during weeks 4-10 of gestation significantly elevated the risk of polydactyly, peaking at week 6. Increases in PM2.5 and CO were associated with an elevated risk of external ear malformations, peaking at week 18 and week 19, respectively. Furthermore, higher concentrations of NOX and NO increased the risk of syndactyly, peaking at week 0 for both pollutants. Finally, increments of 10 μg/m3 in NO2, NOX, NO, and PM10 were all significantly associated with an increased risk of cleft lip and/or palate, peaking at week 3 for NO2, NOX, NO, and PM10. Exposure to air pollutants elevates BD risk, with critical periods during the first and second trimesters. The association between different pollutants and the classification of BDs also varies. Discussion Exposure to pollutants during pregnancy increases the risk of birth defects in newborns, especially SO2, PM10, PM2.5 and O3. In light of these findings, we recommend that, while overall regional air quality improvements remain essential, specific targeted measures should be implemented for pregnant women, who represent a particularly vulnerable population. These targeted recommendations not only aim to reduce exposure risks for pregnant women and their fetuses but also offer practical insights for public health policy and interventions.
Collapse
Affiliation(s)
- Wen Sun
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Shanxi Key Laboratory of Environmental Health Impairment and Prevention, NHC Key Laboratory of Pneumoconiosis, MOE Key Laboratory of Coal Environmental, Taiyuan, Shanxi, China
| | - Qingqing Dong
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Yingying Zhang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Shanxi Key Laboratory of Environmental Health Impairment and Prevention, NHC Key Laboratory of Pneumoconiosis, MOE Key Laboratory of Coal Environmental, Taiyuan, Shanxi, China
| | - Hui Wang
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Youqiang Wang
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Wenjie Yuan
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Leyao Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Shanxi Key Laboratory of Environmental Health Impairment and Prevention, NHC Key Laboratory of Pneumoconiosis, MOE Key Laboratory of Coal Environmental, Taiyuan, Shanxi, China
| | - Xianhong Shi
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Yuhong Feng
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Haiwei Wang
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Xiaodan Wang
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Yingbin Ren
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Lihong Wang
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| | - Lijian Lei
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Shanxi Key Laboratory of Environmental Health Impairment and Prevention, NHC Key Laboratory of Pneumoconiosis, MOE Key Laboratory of Coal Environmental, Taiyuan, Shanxi, China
| | - Wenxia Song
- Changzhi Maternal and Child Health Care Hospital, Changzhi City Key Laboratory of Birth Defect Prevention and Control, Changzhi, Shanxi, China
| |
Collapse
|
2
|
Wang S, Mei Z, Chen J, Zhao K, Kong R, McClements L, Zhang H, Liao A, Liu C. Maternal Immune Activation: Implications for Congenital Heart Defects. Clin Rev Allergy Immunol 2025; 68:36. [PMID: 40175706 DOI: 10.1007/s12016-025-09049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
Congenital heart defects (CHD) are the most common major birth defects and one of the leading causes of death from congenital defects after birth. CHD can arise in pregnancy from the combination of genetic and non-genetic factors. The maternal immune activation (MIA) hypothesis is widely implicated in embryonic neurodevelopmental abnormalities. MIA has been found to be associated with the development of asthma, diabetes mellitus, and other diseases in the offspring. Given the important role of cardiac immune cells and cytokines in embryonic heart development, it is hypothesized that MIA may play a significant role in embryonic heart development. This review aims to stimulate further investigation into the relationship between MIA and CHD and to highlight the gaps in the knowledge. It evaluates the impact of MIA on CHD in the context of pregnancy complications, immune-related diseases, infections, and environmental and lifestyle factors. The review outlines the mechanisms by which immune cells and their secretome indirectly regulate the immuno-microenvironment of the embryonic heart by influencing placental development. Furthermore, the inflammatory cytokines cross the placenta to induce related reactions including oxidative stress in the embryonic heart directly. This review delineates the role of MIA in CHD and underscores the impact of maternal factors, especially immune factors, as well as the embryonic cardiac immuno-microenvironment, on embryonic heart development. This review extends our understanding of the importance of MIA in the pathogenesis of CHD and provides important insights into prenatal prevention and treatment strategies for this congenital condition.
Collapse
Affiliation(s)
- Sixing Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Second Clinical Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zilin Mei
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jin Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ruize Kong
- Department of Vascular Surgery, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China First People'S Hospital of Yunnan Province, Kunming, PR China
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Aihua Liao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
3
|
Wāng Y. Ambient fine particulate matter provokes multiple modalities of cell death via perturbation of subcellular structures. ENVIRONMENT INTERNATIONAL 2025; 195:109193. [PMID: 39721566 DOI: 10.1016/j.envint.2024.109193] [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: 09/24/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
Fine particulate matter (PM2.5) is increasingly recognized for its detrimental effects on human health, with substantial evidence linking exposure to various forms of cell death and dysfunction across multiple organ systems. This review examines key cell death mechanisms triggered by PM2.5, including PANoptosis, necroptosis, autophagy, and ferroptosis, while other forms such as oncosis, paraptosis, and cuprotosis remain unreported in relation to PM2.5 exposure. Mitochondria, endoplasmic reticulum, and lysosomes emerge as pivotal organelles in the disruption of cellular homeostasis, with mitochondrial dysfunction particularly implicated in metabolic dysregulation and the activation of pro-apoptotic pathways. Although PM2.5 primarily affects the nucleus, cytoskeleton, mitochondria, endoplasmic reticulum, and lysosomes, other organelles like ribosomes, Golgi apparatus, and peroxisomes have received limited attention. Interactions between these organelles, such as endoplasmic reticulum-associated mitochondrial membranes, lysosome-associated mitophagy, and mitochondria-nuclei retro-signaling may significantly contribute to the cytotoxic effects of PM2.5. The mechanisms of PM2.5 toxicity, encompassing oxidative stress, inflammatory responses, and metabolic imbalances, are described in detail. Notably, PM2.5 activates the NLRP3 inflammasome, amplifying inflammatory responses and contributing to chronic diseases. Furthermore, PM2.5 exposure disrupts genetic and epigenetic regulation, often resulting in cell cycle arrest and exacerbating cellular damage. The composition, concentration, and seasonal variability of PM2.5 modulate these effects, underscoring the complexity of PM2.5-induced cellular dysfunction. Despite significant advances in understanding these pathways, further research is required to elucidate the long-term effects of chronic PM2.5 exposure, the role of epigenetic regulation, and potential strategies to mitigate its harmful impact on human health.
Collapse
Affiliation(s)
- Yán Wāng
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
4
|
Zheng S, Zhao N, Lin X, Qiu L. Impacts and potential mechanisms of fine particulate matter (PM 2.5) on male testosterone biosynthesis disruption. REVIEWS ON ENVIRONMENTAL HEALTH 2024; 39:777-789. [PMID: 37651650 DOI: 10.1515/reveh-2023-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Exposure to PM2.5 is the most significant air pollutant for health risk. The testosterone level in male is vulnerable to environmental toxicants. In the past, researchers focused more attention on the impacts of PM2.5 on respiratory system, cardiovascular system, and nervous system, and few researchers focused attention on the reproductive system. Recent studies have reported that PM2.5 involved in male testosterone biosynthesis disruption, which is closely associated with male reproductive health. However, the underlying mechanisms by which PM2.5 causes testosterone biosynthesis disruption are still not clear. To better understand its potential mechanisms, we based on the existing scientific publications to critically and comprehensively reviewed the role and potential mechanisms of PM2.5 that are participated in testosterone biosynthesis in male. In this review, we summarized the potential mechanisms of PM2.5 triggering the change of testosterone level in male, which involve in oxidative stress, inflammatory response, ferroptosis, pyroptosis, autophagy and mitophagy, microRNAs (miRNAs), endoplasmic reticulum (ER) stress, and N6-methyladenosine (m6A) modification. It will provide new suggestions and ideas for prevention and treatment of testosterone biosynthesis disruption caused by PM2.5 for future research.
Collapse
Affiliation(s)
- Shaokai Zheng
- School of Public Health, Nantong University, Nantong, P.R. China
| | - Nannan Zhao
- School of Public Health, Nantong University, Nantong, P.R. China
| | - Xiaojun Lin
- School of Public Health, Nantong University, Nantong, P.R. China
| | - Lianglin Qiu
- School of Public Health, Nantong University, Nantong, P.R. China
| |
Collapse
|
5
|
Chanda F, Lin KX, Chaurembo AI, Huang JY, Zhang HJ, Deng WH, Xu YJ, Li Y, Fu LD, Cui HD, Shu C, Chen Y, Xing N, Lin HB. PM 2.5-mediated cardiovascular disease in aging: Cardiometabolic risks, molecular mechanisms and potential interventions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176255. [PMID: 39276993 DOI: 10.1016/j.scitotenv.2024.176255] [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: 06/12/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Air pollution, particularly fine particulate matter (PM2.5) with <2.5 μm in diameter, is a major public health concern. Studies have consistently linked PM2.5 exposure to a heightened risk of cardiovascular diseases (CVDs) such as ischemic heart disease (IHD), heart failure (HF), and cardiac arrhythmias. Notably, individuals with pre-existing age-related cardiometabolic conditions appear more susceptible. However, the specific impact of PM2.5 on CVDs susceptibility in older adults remains unclear. Therefore, this review addresses this gap by discussing the factors that make the elderly more vulnerable to PM2.5-induced CVDs. Accordingly, we focused on physiological aging, increased susceptibility, cardiometabolic risk factors, CVDs, and biological mechanisms. This review concludes by examining potential interventions to reduce exposure and the adverse health effects of PM2.5 in the elderly population. The latter includes dietary modifications, medications, and exploration of the potential benefits of supplements. By comprehensively analyzing these factors, this review aims to provide a deeper understanding of the detrimental effects of PM2.5 on cardiovascular health in older adults. This knowledge can inform future research and guide strategies to protect vulnerable populations from the adverse effects of air pollution.
Collapse
Affiliation(s)
- Francis Chanda
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kai-Xuan Lin
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Abdallah Iddy Chaurembo
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jian-Yuan Huang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui-Juan Zhang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wen-Hui Deng
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yun-Jing Xu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Li-Dan Fu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Hao-Dong Cui
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chi Shu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; Food Science College, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yang Chen
- University of Chinese Academy of Sciences, Beijing, China; Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
| | - Na Xing
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China.
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
6
|
Chen W, Ge P, Lu Z, Liu X, Cao M, Yan Z, Chen M. Acute exposure to seasonal PM 2.5 induces toxicological responses in A549 cells cultured at the air-liquid interface mediated by oxidative stress and endoplasmic reticulum stress. ENVIRONMENTAL RESEARCH 2024; 248:118283. [PMID: 38253190 DOI: 10.1016/j.envres.2024.118283] [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: 12/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Atmospheric fine particulate matter (PM2.5) enters the human body through respiration and poses a threat to human health. This is not only dependent on its mass concentration in the atmosphere, but also related to seasonal variations in its chemical components, which makes it important to study the cytotoxicity of PM2.5 in different seasons. Traditional immersion exposure cannot simulate the living environment of human epithelial cells in the human body, making this method unsuitable for evaluating the inhalation toxicity of PM2.5. In this study, a novel air-liquid interface (ALI) particulate matter exposure device (VITROCELL Cloud 12 system) was used to evaluate the toxic effects and potential mechanisms of human lung epithelial cells (A549) after exposure to seasonal PM2.5. PM2.5 samples from four seasons were collected and analyzed for chemical components. After 6 h of exposure to seasonal PM2.5, winter PM2.5 exhibited the highest cytotoxicity among most toxicity indicators, especially apoptosis rate, reactive oxygen species (ROS), inflammatory responses and DNA damage (γ-H2AX). The effect of autumn PM2.5 on apoptosis rate was significantly higher than that in spring, and there was no significant difference in other toxicity indicators between spring and autumn. The cytotoxicity of summer PM2.5 was the lowest among the four seasons. It should be noted that even exposure to low doses of summer PM2.5 leads to significant DNA damage in A459 cells. Correlation analysis results showed that water-soluble ions, metallic elements, and polycyclic aromatic hydrocarbons (PAHs) were associated with most toxicological endpoints. Inhibitors of oxidative stress and endoplasmic reticulum (ER) stress significantly inhibited cellular damage, indicating that PM2.5-induced cytotoxicity may be related to the generation of ROS and ER stress. In addition, PM2.5 can induce ER stress through oxidative stress, which ultimately leads to apoptosis.
Collapse
Affiliation(s)
- Wankang Chen
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Pengxiang Ge
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Zhenyu Lu
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Xiaoming Liu
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Maoyu Cao
- School of Atmospheric Sciences, Nanjing University, Nanjing, 210023, China
| | - Zhansheng Yan
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Mindong Chen
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
7
|
Wen J, Zhang J, Zhang H, Zhang N, Lei R, Deng Y, Cheng Q, Li H, Luo P. Large-scale genome-wide association studies reveal the genetic causal etiology between air pollutants and autoimmune diseases. J Transl Med 2024; 22:392. [PMID: 38685026 PMCID: PMC11057084 DOI: 10.1186/s12967-024-04928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/23/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Epidemiological evidence links a close correlation between long-term exposure to air pollutants and autoimmune diseases, while the causality remained unknown. METHODS Two-sample Mendelian randomization (TSMR) was used to investigate the role of PM10, PM2.5, NO2, and NOX (N = 423,796-456,380) in 15 autoimmune diseases (N = 14,890-314,995) using data from large European GWASs including UKB, FINNGEN, IMSGC, and IPSCSG. Multivariable Mendelian randomization (MVMR) was conducted to investigate the direct effect of each air pollutant and the mediating role of common factors, including body mass index (BMI), alcohol consumption, smoking status, and household income. Transcriptome-wide association studies (TWAS), two-step MR, and colocalization analyses were performed to explore underlying mechanisms between air pollution and autoimmune diseases. RESULTS In TSMR, after correction of multiple testing, hypothyroidism was causally associated with higher exposure to NO2 [odds ratio (OR): 1.37, p = 9.08 × 10-4] and NOX [OR: 1.34, p = 2.86 × 10-3], ulcerative colitis (UC) was causally associated with higher exposure to NOX [OR: 2.24, p = 1.23 × 10-2] and PM2.5 [OR: 2.60, p = 5.96 × 10-3], rheumatoid arthritis was causally associated with higher exposure to NOX [OR: 1.72, p = 1.50 × 10-2], systemic lupus erythematosus was causally associated with higher exposure to NOX [OR: 4.92, p = 6.89 × 10-3], celiac disease was causally associated with lower exposure to NOX [OR: 0.14, p = 6.74 × 10-4] and PM2.5 [OR: 0.17, p = 3.18 × 10-3]. The risky effects of PM2.5 on UC remained significant in MVMR analyses after adjusting for other air pollutants. MVMR revealed several common mediators between air pollutants and autoimmune diseases. Transcriptional analysis identified specific gene transcripts and pathways interconnecting air pollutants and autoimmune diseases. Two-step MR revealed that POR, HSPA1B, and BRD2 might mediate from air pollutants to autoimmune diseases. POR pQTL (rs59882870, PPH4=1.00) strongly colocalized with autoimmune diseases. CONCLUSION This research underscores the necessity of rigorous air pollutant surveillance within public health studies to curb the prevalence of autoimmune diseases.
Collapse
Affiliation(s)
- Jie Wen
- The Animal Laboratory Center, Hunan Cancer Hospital, and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ruoyan Lei
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yujia Deng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- First Clinical Department, Changsha Medical University, Changsha, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - He Li
- The Animal Laboratory Center, Hunan Cancer Hospital, and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Yan R, Ma D, Liu Y, Wang R, Fan L, Yan Q, Chen C, Wang W, Ren Z, Ku T, Ning X, Sang N. Developmental Toxicity of Fine Particulate Matter: Multifaceted Exploration from Epidemiological and Laboratory Perspectives. TOXICS 2024; 12:274. [PMID: 38668497 PMCID: PMC11054511 DOI: 10.3390/toxics12040274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/29/2024]
Abstract
Particulate matter of size ≤ 2.5 μm (PM2.5) is a critical environmental threat that considerably contributes to the global disease burden. However, accompanied by the rapid research progress in this field, the existing research on developmental toxicity is still constrained by limited data sources, varying quality, and insufficient in-depth mechanistic analysis. This review includes the currently available epidemiological and laboratory evidence and comprehensively characterizes the adverse effects of PM2.5 on developing individuals in different regions and various pollution sources. In addition, this review explores the effect of PM2.5 exposure to individuals of different ethnicities, genders, and socioeconomic levels on adverse birth outcomes and cardiopulmonary and neurological development. Furthermore, the molecular mechanisms involved in the adverse health effects of PM2.5 primarily encompass transcriptional and translational regulation, oxidative stress, inflammatory response, and epigenetic modulation. The primary findings and novel perspectives regarding the association between public health and PM2.5 were examined, highlighting the need for future studies to explore its sources, composition, and sex-specific effects. Additionally, further research is required to delve deeper into the more intricate underlying mechanisms to effectively prevent or mitigate the harmful effects of air pollution on human health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China; (R.Y.); (D.M.); (Y.L.); (R.W.); (L.F.); (Q.Y.); (C.C.); (W.W.); (Z.R.); (X.N.); (N.S.)
| | | | | |
Collapse
|
9
|
Guo W, Kang C, Wang X, Zhang H, Yuan L, Wei X, Xiao Q, Hao W. Chlorocholine chloride exposure induced spermatogenic dysfunction via iron overload caused by AhR/PERK axis-dependent ferritinophagy activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116193. [PMID: 38460407 DOI: 10.1016/j.ecoenv.2024.116193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Chlorocholine chloride (CCC) is a plant growth regulator used worldwide that is detectable in cereals, fruits and animal products. The health effects of CCC exposure have raised public concern. Our previous research showed that CCC exposure decreased testosterone synthesis in pubertal rats. However, little is known about whether and how pubertal CCC exposure impacts spermatogenesis. In this study, we used BALB/c mice and spermatogonia-derived GC-1 cells to examine CCC-induced spermatogenic dysfunction. In vivo, pubertal CCC exposure led to decreased testicular weight, decreased testicular germ cells and poor sperm quality. This effect worsened after cessation of CCC exposure for the next 30 days. RNA-seq and western blot analysis revealed that CCC induced aryl hydrocarbon receptor (AhR) signaling, endoplasmic reticulum stress (ERS) and ferritinophagy. Increased iron content and lipid peroxidation levels were also observed in CCC-treated testes. In vitro, it was identified that iron overload mediated by enhanced ferritinophagy occurred in CCC-treated GC-1 cells, which might be attributed to the PERK pathway in ERS. Further, for the first time, our study elucidated the involvement of AhR in CCC-induced iron overload, which aggravated testicular oxidative damage via lipid peroxidation. Considering the adverse impact of CCC exposure on rodents, supportive evidence from GC-1 cells, and the critical importance of spermatogenesis on male development, the effects of CCC on the male reproduction warrant increased attention.
Collapse
Affiliation(s)
- Wanqian Guo
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Chenping Kang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Haoran Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Lilan Yuan
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China.
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China.
| |
Collapse
|
10
|
Jiang Y, Zhang M, Li J, Hu K, Chen T. AHR/cyp1b1 signaling-mediated extrinsic apoptosis contributes to 6PPDQ-induced cardiac dysfunction in zebrafish embryos. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123467. [PMID: 38311157 DOI: 10.1016/j.envpol.2024.123467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPDQ) has raised significant concerns due to its widespread distribution and high toxicity to aquatic organisms. However, the cardiac developmental toxicity of 6PPDQ and the underlying mechanisms remain unclear. In this study, we observed no notable alterations in heart morphology or embryo survival in zebrafish embryos exposed to 6PPDQ (0.2-2000 μg/L) up to 3 days post-fertilization (dpf). However, concentrations at 2 μg/L or higher induced cardiac dysfunctions, leading to lethal effects at later stages (6-8 dpf). We further found that the aryl hydrocarbon receptor (AHR) inhibitor CH22351 attenuated 6PPDQ-induced cardiac dysfunctions, implicating the involvement of AHR signal pathway. Moreover, 6PPDQ exposure led to an overproduction of reactive oxygen species (ROS) and an upregulation of genes associated with oxidative stress (sod1, sod2, and nrf2a). This was accompanied by an increase in oxidative DNA damage and the induction of p53-dependent extrinsic apoptosis. Co-exposure to the ROS scavenger N-acetylcysteine effectively counteracted the DNA damage and apoptosis induced by 6PPDQ. Importantly, inhibition of AHR or its downstream target cyp1b1 attenuated 6PPDQ-induced oxidative stress, DNA damage, and apoptosis. In conclusion, our results provide evidence that 6PPDQ induces oxidative stress through the AHR/cyp1b1 signaling pathway, leading to DNA damage and extrinsic apoptosis, ultimately resulting in cardiac dysfunction.
Collapse
Affiliation(s)
- Yan Jiang
- Suzhou Medical College of Soochow University, Suzhou, China; MOE Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China
| | - Mingxuan Zhang
- Suzhou Medical College of Soochow University, Suzhou, China; MOE Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China
| | - Jinhao Li
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Keqi Hu
- Department of Science and Education, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Tao Chen
- Suzhou Medical College of Soochow University, Suzhou, China; MOE Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China.
| |
Collapse
|
11
|
Li X, Jiang X, Zeng R, Lai X, Wang J, Liu H, Wu H, He J, Liu L, Zhu Z, Li J, Liang X. Formononetin attenuates cigarette smoke-induced COPD in mice by suppressing inflammation, endoplasmic reticulum stress, and apoptosis in bronchial epithelial cells via AhR/CYP1A1 and AKT/mTOR signaling pathways. Phytother Res 2024; 38:1278-1293. [PMID: 38191199 DOI: 10.1002/ptr.8104] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/16/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic, progressive, and lethal lung disease with few treatments. Formononetin (FMN) is a clinical preparation extract with extensive pharmacological actions. However, its effect on COPD remains unknown. This study aimed to explore the effect and underlying mechanisms of FMN on COPD. A mouse model of COPD was established by exposure to cigarette smoke (CS) for 24 weeks. In addition, bronchial epithelial BEAS-2B cells were treated with CS extract (CSE) for 24 h to explore the in vitro effect of FMN. FMN significantly improved lung function and attenuated pathological lung damage. FMN treatment reduced inflammatory cell infiltration and pro-inflammatory cytokines secretion. FMN also suppressed apoptosis by regulating apoptosis-associated proteins. Moreover, FMN relieved CS-induced endoplasmic reticulum (ER) stress in the mouse lungs. In BEAS-2B cells, FMN treatment reduced CSE-induced inflammation, ER stress, and apoptosis. Mechanistically, FMN downregulated the CS-activated AhR/CYP1A1 and AKT/mTOR signaling pathways in vivo and in vitro. FMN can attenuate CS-induced COPD in mice by suppressing inflammation, ER stress, and apoptosis in bronchial epithelial cells via the inhibition of AhR/CYP1A1 and AKT/mTOR signaling pathways, suggesting a new therapeutic potential for COPD treatment.
Collapse
Affiliation(s)
- Xiaomei Li
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xianhan Jiang
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Runhao Zeng
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiujuan Lai
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Wang
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Liu
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huihui Wu
- Department of Endocrinology and Metabolism, Jing'an District Center Hospital of Shanghai, Shanghai, China
| | - Jiaxun He
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lian Liu
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiying Zhu
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingpei Li
- Department of Thoracic Surgery/Oncology, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xue Liang
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Song C, Wang Z, Cao J, Dong Y, Chen Y. Hesperetin protects hippocampal neurons from the neurotoxicity of Aflatoxin B1 in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115782. [PMID: 38056121 DOI: 10.1016/j.ecoenv.2023.115782] [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: 08/02/2023] [Revised: 11/01/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
Aflatoxin B1 (AFB1) is a major food and feed pollutant that endangers public health. Previous studies have shown that exposure to AFB1 causes neurotoxicity in the body. However, the mechanism of neurotoxicity caused by AFB1 is not well understood, and finding a workable and practical method to safeguard animals from AFB1 toxicity is essential. This study confirmed that AFB1 caused endoplasmic reticulum stress (ER stress) and apoptosis in hippocampal neurons using C57BL/6 J mice and HT22 cells as models. In vitro experiments showed that the aryl hydrocarbon receptor (AHR) plays a significant role in the cytotoxicity of AFB1. Finally, we assessed how hesperetin protecting against the neurotoxicity caused by AFB1. Our findings demonstrated that AFB1 increased the levels of BAX and Cleaved-Caspase3 proteins, while decreasing the levels of BCL2 protein in the CA1 and CA3 regions of the hippocampus. The AFB1 increased the expression of AHR and activated nuclear translocation. It also elevated the expression levels of Chop, GRP78, p-IRE1/ Xbp1s, and p-PERK/p-EIF2a. Importantly, we also discovered for the first time that blocking AHR in HT22 cells dramatically reduced the level of ER stress and apoptosis caused by AFB1. In vivo and in vitro studies, supplementation of hesperetin effectively reversed AFB1-induced cytotoxicity. We have demonstrated that hesperetin effectively restored the imbalance in the GSH/GST system in HT22 cells treated with AFB1. Furthermore, we observed that elevated GSH levels facilitated the formation of AFB1-GSH complexes, which enhanced the excretion of AFB1. Therefore, hesperetin improves ER stress-induced apoptosis by reducing AFB1 activation of AHR.
Collapse
Affiliation(s)
- Chao Song
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Jing Cao
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China; Department of Nutrition and Health, China Agricultural University, Haidian, Beijing 100193, China.
| |
Collapse
|
13
|
Zou H, Zhang M, Chen J, Aniagu S, Jiang Y, Chen T. AHR-mediated DNA damage contributes to BaP-induced cardiac malformations in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167636. [PMID: 37806592 DOI: 10.1016/j.scitotenv.2023.167636] [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: 09/06/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Benzo[a]pyrene (BaP) is a representative polycyclic aromatic hydrocarbon widely present in the environment. We previously reported that the aryl hydrocarbon receptor (AHR) mediates BaP-induced apoptosis and cardiac malformations in zebrafish embryos, but the underlying molecular mechanisms were unclear. Since BaP is a mutagenetic compound, we hypothesize that BaP induces apoptosis and heart defects via AHR-mediated DNA damage. In this study, zebrafish embryos were exposed to BaP at a concentration of 0.1 μM from 2 to 72 h post fertilization, either with or without inhibitors/agonists. AHR activity and levels of reactive oxygen species (ROS) were examined under a fluorescence microscope. mRNA expression levels were quantified by qPCR. DNA damage and apoptosis were detected by immunofluorescence. Our findings revealed that BaP exposure significantly increased BPDE-DNA adducts, mitochondrial damage, apoptosis and heart defects in zebrafish embryos. These effects were counteracted by inhibiting AHR/cyp1a1 using pharmaceutical inhibitors or genetic knockdown. Furthermore, we observed that spironolactone, an antagonist of nucleotide excision repair (NER), significantly enhanced BaP-induced BPDE-DNA adducts, mitochondrial damage, apoptosis and heart malformation rates. Conversely, SRT1720, a SIRT1 agonist, reduced the adverse effects of BaP. Supplementation with spironolactone also enhanced γ-H2AX signals in the heart of zebrafish embryos exposed to BaP. Additional experiments demonstrated that BaP suppressed the expression of SIRT1. We further established that AHR, when activated by BaP, directly inhibited SIRT1 transcription, leading to downregulation of XPC and XPA, which are essential NER genes involved in the recognition and verification steps of the NER process. Taken together, our results indicate that AHR mediates BaP-induced DNA damage in the heart of zebrafish embryos by inducing BPDE-DNA adduct formation via the AHR/Cyp1a1 signalling pathway, as well as suppressing NER via AHR-mediated inhibition of SIRT1.
Collapse
Affiliation(s)
- Hongmei Zou
- Suzhou Medical College, Soochow University, Suzhou, China; Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China
| | - Mingxuan Zhang
- Suzhou Medical College, Soochow University, Suzhou, China; Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China
| | - Jin Chen
- Suzhou Medical College, Soochow University, Suzhou, China; Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China
| | - Stanley Aniagu
- Toxicology, Risk Assessment, and Research Division, Texas Commission on Environmental Quality, 12015 Park 35 Cir, Austin, TX, USA
| | - Yan Jiang
- Suzhou Medical College, Soochow University, Suzhou, China; Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China.
| | - Tao Chen
- Suzhou Medical College, Soochow University, Suzhou, China; Education Key Laboratory of Geriatric Diseases and Immunology, Suzhou, China.
| |
Collapse
|
14
|
Huttner IG, Santiago CF, Jacoby A, Cheng D, Trivedi G, Cull S, Cvetkovska J, Chand R, Berger J, Currie PD, Smith KA, Fatkin D. Loss of Sec-1 Family Domain-Containing 1 ( scfd1) Causes Severe Cardiac Defects and Endoplasmic Reticulum Stress in Zebrafish. J Cardiovasc Dev Dis 2023; 10:408. [PMID: 37887855 PMCID: PMC10607167 DOI: 10.3390/jcdd10100408] [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: 03/28/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder that frequently leads to heart failure, arrhythmias, and death. While DCM is often heritable, disease-causing mutations are identified in only ~30% of cases. In a forward genetic mutagenesis screen, we identified a novel zebrafish mutant, heart and head (hahvcc43), characterized by early-onset cardiomyopathy and craniofacial defects. Linkage analysis and next-generation sequencing identified a nonsense variant in the highly conserved scfd1 gene, also known as sly1, that encodes sec1 family domain-containing 1. Sec1/Munc18 proteins, such as Scfd1, are involved in membrane fusion regulating endoplasmic reticulum (ER)/Golgi transport. CRISPR/Cas9-engineered scfd1vcc44 null mutants showed severe cardiac and craniofacial defects and embryonic lethality that recapitulated the phenotype of hahvcc43 mutants. Electron micrographs of scfd1-depleted cardiomyocytes showed reduced myofibril width and sarcomere density, as well as reticular network disorganization and fragmentation of Golgi stacks. Furthermore, quantitative PCR analysis showed upregulation of ER stress response and apoptosis markers. Both heterozygous hahvcc43 mutants and scfd1vcc44 mutants survived to adulthood, showing chamber dilation and reduced ventricular contraction. Collectively, our data implicate scfd1 loss-of-function as the genetic defect at the hahvcc43 locus and provide new insights into the role of scfd1 in cardiac development and function.
Collapse
Affiliation(s)
- Inken G. Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Celine F. Santiago
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Arie Jacoby
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Delfine Cheng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Gunjan Trivedi
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Stephen Cull
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Jasmina Cvetkovska
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Renee Chand
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Kelly A. Smith
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
- Cardiology Department, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
15
|
Sun S, Zhang C, Zhang Q, Li C, Huang D, Ding R, Cao J, Hao J. Role of ROS-mediated PERK/ATF4 signaling activation in extracorporeal tube formation injury of human umbilical vein endothelial cells induced by cooking oil fume PM 2.5 exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115332. [PMID: 37611476 DOI: 10.1016/j.ecoenv.2023.115332] [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: 04/06/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023]
Abstract
Cooking oil fume-derived PM2.5 (COF-PM2.5) is a major source of indoor air contamination in China, which has been demonstrated to be a hazard factor of cardiovascular and cerebrovascular diseases. This study aimed to investigate the role of ROS-mediated PERK/ATF4 signaling activation in COF-PM2.5-inhibited extracorporeal tube formation in human umbilical vein endothelial cells (HUVECs). HUVECs were treated with 100 μg/mL COF-PM2.5 at different times, with or without 100 nM PERK activity inhibitor GSK2606414 (GSK) or 200 μM antioxidant N-acetylcysteine (NAC) pretreatment. Our results showed that COF-PM2.5 exposure can inhibit extracorporeal tube formation and down-regulate VEGFR2 expression in HUVECs. Furthermore, our data indicated that COF-PM2.5 exposure can activate the PERK/ATF4 signaling in HUVECs. Mechanistically, pretreatment with GSK interdicted PERK/ATF4 signaling, thereby reversing COF-PM2.5-downregulated VEGFR2 protein expression in HUVECs. Furthermore, NAC reversed VEGFR2 expression downregulated induced by COF-PM2.5 by inhibiting the upregulation of intracellular ROS levels and PERK/ATF4 signaling in HUVECs. As above, COF-PM2.5 exposure could induce ROS release from HUVECs, which in turn activate the endoplasmic reticulum PERK/ATF4 signaling and inhibit tube formation of HUVECs.
Collapse
Affiliation(s)
- Shu Sun
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Chao Zhang
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Qi Zhang
- Hefei Institutes of Physical Science Chinese Academy of Sciences, No 350 Shushanhu Road, Hefei 230001, Anhui, China
| | - Changlian Li
- Department of Environmental Health, Hefei Center for Disease Control and Prevention, No 86 Lu'an Road, Hefei 230061, Anhui, China
| | - Dan Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Jiyu Cao
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Jiahu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
16
|
Shao X, Meng C, Song W, Zhang T, Chen Q. Subcellular visualization: Organelle-specific targeted drug delivery and discovery. Adv Drug Deliv Rev 2023; 199:114977. [PMID: 37391014 DOI: 10.1016/j.addr.2023.114977] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Organelles perform critical biological functions due to their distinct molecular composition and internal environment. Disorders in organelles or their interacting networks have been linked to the incidence of numerous diseases, and the research of pharmacological actions at the organelle level has sparked pharmacists' interest. Currently, cell imaging has evolved into a critical tool for drug delivery, drug discovery, and pharmacological research. The introduction of advanced imaging techniques in recent years has provided researchers with richer biological information for viewing and studying the ultrastructure of organelles, protein interactions, and gene transcription activities, leading to the design and delivery of precision-targeted drugs. Therefore, this reviews the research on organelles-targeted drugs based upon imaging technologies and development of fluorescent molecules for medicinal purposes. We also give a thorough analysis of a number of subcellular-level elements of drug development, including subcellular research instruments and methods, organelle biological event investigation, subcellular target and drug identification, and design of subcellular delivery systems. This review will make it possible to promote drug research from the individual/cellular level to the subcellular level, as well as give a new focus based on newly found organelle activities.
Collapse
Affiliation(s)
- Xintian Shao
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Caicai Meng
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Wenjing Song
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China; School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Tao Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 250014, PR China
| | - Qixin Chen
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| |
Collapse
|
17
|
Grishanova AY, Klyushova LS, Perepechaeva ML. AhR and Wnt/β-Catenin Signaling Pathways and Their Interplay. Curr Issues Mol Biol 2023; 45:3848-3876. [PMID: 37232717 DOI: 10.3390/cimb45050248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
As evolutionarily conserved signaling cascades, AhR and Wnt signaling pathways play a critical role in the control over numerous vital embryonic and somatic processes. AhR performs many endogenous functions by integrating its signaling pathway into organ homeostasis and into the maintenance of crucial cellular functions and biological processes. The Wnt signaling pathway regulates cell proliferation, differentiation, and many other phenomena, and this regulation is important for embryonic development and the dynamic balance of adult tissues. AhR and Wnt are the main signaling pathways participating in the control of cell fate and function. They occupy a central position in a variety of processes linked with development and various pathological conditions. Given the importance of these two signaling cascades, it would be interesting to elucidate the biological implications of their interaction. Functional connections between AhR and Wnt signals take place in cases of crosstalk or interplay, about which quite a lot of information has been accumulated in recent years. This review is focused on recent studies about the mutual interactions of key mediators of AhR and Wnt/β-catenin signaling pathways and on the assessment of the complexity of the crosstalk between the AhR signaling cascade and the canonical Wnt pathway.
Collapse
Affiliation(s)
- Alevtina Y Grishanova
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630117, Russia
| | - Lyubov S Klyushova
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630117, Russia
| | - Maria L Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630117, Russia
| |
Collapse
|
18
|
Nan N, Yan Z, Zhang Y, Chen R, Qin G, Sang N. Overview of PM 2.5 and health outcomes: Focusing on components, sources, and pollutant mixture co-exposure. CHEMOSPHERE 2023; 323:138181. [PMID: 36806809 DOI: 10.1016/j.chemosphere.2023.138181] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
PM2.5 varies in source and composition over time and space as a complicated mixture. Consequently, the health effects caused by PM2.5 varies significantly over time and generally exhibit significant regional variations. According to numerous studies, a notable relationship exists between PM2.5 and the occurrence of many diseases, such as respiratory, cardiovascular, and nervous system diseases, as well as cancer. Therefore, a comprehensive understanding of the effect of PM2.5 on human health is critical. The toxic effects of various PM2.5 components, as well as the overall toxicity of PM2.5 are discussed in this review to provide a foundation for precise PM2.5 emission control. Furthermore, this review summarizes the synergistic effect of PM2.5 and other pollutants, which can be used to draft effective policies.
Collapse
Affiliation(s)
- Nan Nan
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Zhipeng Yan
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Yaru Zhang
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Rui Chen
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, 100054, PR China; Beijing City University, Beijing, 11418, PR China.
| | - Guohua Qin
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| |
Collapse
|
19
|
Jiang Y, Zhao X, Chen J, Aniagu S, Chen T. PM2.5 induces cardiac malformations via PI3K/akt2/mTORC1 signaling pathway in zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 323:121306. [PMID: 36804889 DOI: 10.1016/j.envpol.2023.121306] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/12/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Growing evidence indicates that maternal fine particulate matter (PM2.5) exposure is linked with congenital heart diseases in the offspring. To explore the underlying molecular mechanisms, we tested the effects of a number of pharmaceutical inhibitors, and found that suppressing the PI3K/akt signaling pathway had a protective effect against cardiac defects in zebrafish larvae exposed to extractable organic matter (EOM) from PM2.5. Using genetic knockdown and a specific akt2 pharmacological inhibitor, CCT128930, we demonstrated that akt2 activation is essential to EOM-induced heart malformations. Next, we found that the EOM-induced akt2 overactivation enhances intracellular reactive oxygen species (ROS)/mitochondrial ROS production, decreases mitochondrial membrane potential levels, and elicits intrinsic apoptosis in the heart of zebrafish embryos. In addition, EOM-induced akt2 activation decreased active β-catenin levels and inhibited the expression of Wnt target genes axin2 and nkx2.5. We further demonstrated that mTORC1 phosphorylation mediates the adverse effects of akt2 on intrinsic apoptosis and canonical Wnt signaling in the heart of zebrafish larvae exposed to EOM. Moreover, EOM-induced akt2 activation is mediated via aryl hydrocarbon receptor (AHR)/ROS-induced PTEN inhibition. In conclusion, our results indicate that PM2.5 activates PI3K/akt2/mTORC1 signaling via AHR/ROS-induced PTEN suppression, which leads to mitochondrial-mediated intrinsic apoptosis and Wnt signaling suppression, resulting in cardiac defects in zebrafish larvae.
Collapse
Affiliation(s)
- Yan Jiang
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Xiahao Zhao
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Jin Chen
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Stanley Aniagu
- Toxicology, Risk Assessment, and Research Division, Texas Commission on Environmental Quality, 12015 Park 35 Cir, Austin, TX, USA
| | - Tao Chen
- Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China.
| |
Collapse
|