1
|
Wāng Y, Wang C, Jiang Y, Wang T, Wu T, Tang M. Carbonaceous cores serve as surrogates for environmental particulate matter inducing vascular endothelial inflammation via inflammasome activation. JOURNAL OF HAZARDOUS MATERIALS 2024; 486:137011. [PMID: 39736255 DOI: 10.1016/j.jhazmat.2024.137011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Ambient particulate matter (PM) exposure is a known risk factor for cardiovascular diseases. Epidemiological studies have shown the association between PM exposure and vascular complications, including vasculitis, embolism, hypertension, stroke, and atherosclerosis. However, the exact mechanisms underlying its vascular toxicity, especially in relation to short-term exposures, remain incompletely understood. This study investigates the role of PM and its carbonaceous cores in driving vascular endothelial inflammation via inflammasome activation. We hypothesized that PM SRM1648a exposure induces vascular endothelial inflammation through oxidative stress and inflammasome activation. Short-term exposure to PM SRM1648a was assessed in BALB/c mice for systemic inflammation and oxidative stress biomarkers, alongside in vitro studies in HUVECs and EA.hy926 endothelial cells to elucidate inflammasome activation pathways. PM SRM1648a exposure significantly altered redox balance and cytokine profiles in mice and upregulated NLRP3/NLRC4 inflammasomes in vascular endothelial cells, leading to caspase-1/IL-1β activation. Intriguingly, pyroptosis was not the primary mode of cell death. In vitro studies demonstrated that antioxidants glutathione monoethyl ester effectively mitigated oxidative stress and inflammasome activation in endothelial cells. This study highlights the critical role of ROS-mediated inflammasome activation in vascular inflammation induced by PM SRM1648a, with carbon-based cores as key contributors.
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.
| | - Chunzhi Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yang Jiang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Wang
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui 231100, China
| | - Tianshu Wu
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Meng Tang
- Key laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
2
|
Wāng Y. Ambient fine particulate matter provokes multiple modalities of cell death via perturbation of subcellular structures. ENVIRONMENT INTERNATIONAL 2024; 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] [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
|
3
|
Torrico-Lavayen R, Posadas-Sánchez R, Osorio-Yáñez C, Sanchez-Guerra M, Texcalac-Sangrador JL, Ortiz-Panozo E, De Vizcaya-Ruiz A, Botello-Taboada V, Hernández-Rodríguez EA, Gutiérrez-Avila I, Vargas-Alarcón G, Riojas-Rodríguez H. Fine particulate matter and intima media thickness: Role of endothelial function biomarkers. Environ Epidemiol 2024; 8:e356. [PMID: 39600525 PMCID: PMC11596520 DOI: 10.1097/ee9.0000000000000356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background Ambient fine particulate matter (PM2.5) is a risk factor for atherosclerosis disease. We aimed to assess whether nitric oxide stable metabolites (NOx) and l-arginine mediate the association between PM2.5 and carotid intima media thickness (cIMT) increase. Methods We selected 251 participants from the control group of GEA (Genetics of Atheroslerosis Disease Mexican) study (2008-2013) in Mexico City. Mediation models were carried out using pathway analyses, a special case of structural equation models. Results The median concentration of PM2.5 area under the curve (auc) was 25.2 µg/m3 (interquartile range: 24.2-26.4 µg/m3). Employing participants with observed values for both biomarkers (n = 117), the total effect of PM2.5auc on mean cIMT at bilateral, right, and left was 19.27 µm (95% confidence interval [CI]: 5.77, 32.78; P value = 0.005), 12.69 µm (95% CI: 0.67, 24.71; P value = 0.039), and 25.86 µm (95% CI: 3.18, 48.53; P value = 0.025) per each 1 µg/m3 increase of PM2.5auc. The direct effect of PM2.5auc (per 1 µg/m3 increase) was 18.89 µm (95% CI: 5.37, 32.41; P value = 0.006) for bilateral, 13.65 µm (95% CI: 0.76, 26.55; P value = 0.038) for right, and 24.13 µm (95% CI: 3.22, 45.03; P value = 0.024) for left. The indirect effects of NOx and l-arginine were not statistically significant showing that endothelial function biomarkers did not mediate PM2.5 and cIMT associations. Although l-arginine was not a mediator in the PM2.5 and cIMT pathway, a decrease in l-arginine was significantly associated with PM2.5auc. Conclusions In this study of adults from Mexico City, we found that PM2.5 was associated with an increase in cIMT at bilateral, left, and right, and these associations were not mediated by endothelial function biomarkers (l-arginine and NOx).
Collapse
Affiliation(s)
- Rocio Torrico-Lavayen
- Departamento de Patología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Department of Environmental Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Citlalli Osorio-Yáñez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación en Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | | | - Eduardo Ortiz-Panozo
- Center of Population Health Research, National Institute of Public Health, Cuernavaca, Mexico
- Department of Epidemiology, Harvard T.H. Chan School of Public Health. Boston, Massachusetts
| | - Andrea De Vizcaya-Ruiz
- Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, California
| | - Viridiana Botello-Taboada
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación en Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Elihu Alexander Hernández-Rodríguez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación en Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Iván Gutiérrez-Avila
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | |
Collapse
|
4
|
Deng S, Lao M, Zheng H, Hao J. Forkhead box P1 transcriptionally activates IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome. Biogerontology 2024; 26:15. [PMID: 39585426 DOI: 10.1007/s10522-024-10151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Endothelial cell (EC) senescence is a major contributor in atherosclerosis (AS) development. Herein, the role of forkhead box P transcription factor 1 (FOXP1) and insulin-like growth factor (IGF)-1 in regulating EC senescence during AS progression was investigated. The mRNA and protein expressions were assessed using qRT-PCR and western blot. IL-1β and IL-18 secretion levels were analyzed by ELISA. Cell viability and pyroptosis were determined by MTT assay and flow cytometry, respectively. SA β-Gal staining was used to measure cell senescence. Tube formation assay was adopted to detect the angiogenesis ability. Dual-luciferase reporter and ChIP assays were used to investigate the relationship between FOXP1 and IGF‑1. ox-LDL stimulation significantly reduced FOXP1 and IGF-1 expression levels in human aortic endothelial cells (HAECs). FOXP1 or IGF-1 overexpression both mitigated ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. It was subsequently revealed that FOXB1 transcriptionally activated IGF-1 expression in HAECs by binding to IGF-1 promoter. Rescue experiments demonstrated that IGF-1 silencing abolished the inhibitory impact of FOXP1 overexpression on ox-LDL-induced cellular senescence and NLRP3 activation in HAECs. FOXP1 transcriptionally activated IGF-1 to lighten ox-LDL-induced endothelial cellular senescence by inactivating NLRP3 inflammasome.
Collapse
Affiliation(s)
- Siqi Deng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Meili Lao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Huihui Zheng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China
| | - Jingwen Hao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, No. 3, Xueyuan Road, Longhua District, Haikou, 571199, Hainan Province, China.
| |
Collapse
|
5
|
Meng T, He J, Huo Q, Wang Y, Ren Q, Kang Y. Association of Stress Defense System With Fine Particulate Matter Exposure: Mechanism Analysis and Application Prospects. J Appl Toxicol 2024. [PMID: 39538419 DOI: 10.1002/jat.4724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The association between the stress defense system and exposure to fine particulate matter (PM2.5) is a hot topic in the field of environmental health. PM2.5 pollution is an increasingly serious issue, and its impact on health cannot be ignored. The stress defense system is an important biological mechanism for maintaining cell and internal environment homeostasis, playing a crucial role in PM2.5-induced damage and diseases. The association between PM2.5 exposure and activation of the stress defense system has been reported. Moderate PM2.5 exposure rapidly mobilizes the stress defense system, while excessive PM2.5 exposure may exceed its compensatory and coping abilities, resulting in system imbalance and dysfunction that triggers pathological changes in cells and tissues, thereby increasing the risk of chronic diseases, such as respiratory diseases, cardiovascular diseases, and cancer. This detailed review focuses on the composition, function, and regulatory mechanisms of the antioxidant defense system, autophagy system, ubiquitin-proteasome system, and inflammatory response system, which are all components of the stress defiance system. In particular, the influence of PM2.5 exposure on each of these defense systems and their roles in responding to PM2.5-induced damage was investigated to provide an in-depth understanding of the pathogenesis of PM2.5 exposure, accurately assess potential hazards, and formulate prevention and intervention strategies for health damage caused by PM2.5 exposure.
Collapse
Affiliation(s)
- Tao Meng
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
- Doctoral Innovation Station of Shanxi Province, Key Laboratory of TCM Prevention and Treatment of Dementia Disease, The Fifth People's Hospital of Datong, Datong, China
| | - Jing He
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qianru Huo
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yajie Wang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qingchun Ren
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yihui Kang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| |
Collapse
|
6
|
Zhou SY, Du JM, Li WJ, Liu QY, Zhang QY, Su GH, Li Y. The roles and regulatory mechanisms of cigarette smoke constituents in vascular remodeling. Int Immunopharmacol 2024; 140:112784. [PMID: 39083928 DOI: 10.1016/j.intimp.2024.112784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Vascular remodeling is a dynamic process involving cellular and molecular changes, including cell proliferation, migration, apoptosis and extracellular matrix (ECM) synthesis or degradation, which disrupt the homeostasis of endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). Cigarette smoke exposure (CSE) is thought to promote vascular remodeling, but the components are complex and the mechanisms are unclear. In this review, we overview the progression of major components of cigarette smoke (CS), such as nicotine and acrolein, involved in vascular remodeling in terms of ECs injury, VSMCs proliferation, migration, apoptosis, and ECM disruption. The aim was to elucidate the effects of different components of CS on different cells of the vascular system, to discover the relevance of their actions, and to provide new references for future studies.
Collapse
Affiliation(s)
- Si-Yuan Zhou
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Jia-Min Du
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Wen-Jing Li
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qi-Yun Liu
- Department of Cardiology, Shandong Second Medical University, Weifang, China
| | - Qun-Ye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China
| | - Guo-Hai Su
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Li
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
7
|
Yuan Z, Wang Y, Du Z, Lin S, Zeng J, Sun X, Zhang Y, Deng X, Sun Y, Wei J, Zhang M, Sun J, Gu J, Zhang W, Hao Y. Association of long-term exposure to PM 2.5 and its chemical components with the reduced quality of sleep. Sleep Med 2024; 121:251-257. [PMID: 39024779 DOI: 10.1016/j.sleep.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/22/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
Poor sleep quality is a widespread concern. While the influence of particle exposure on sleep disturbances has received considerable attention, research exploring other dimensions of sleep quality and the chemical components of the particles remains limited. We employed a marginal structural model to explore the association of long-term exposure to PM2.5 and its chemical components with poor sleep quality. The odds ratio (95 % CI) for poor sleep quality was 1.335 (1.292-1.378), 1.097 (1.080-1.113), 1.137 (1.100-1.174), 1.197 (1.156-1.240), and 1.124 (1.107-1.140) per IQR increase in the concentration of PM2.5, SO42-, NO3-, NH4+, and BC, respectively. The score (and 95 % CI) of sleep latency, use of sleep medication, habitual sleep efficiency, subjective sleep quality, and daytime dysfunction were affected by PM2.5, with an increase of 0.059 (0.050-0.069), 0.054 (0.049-0.059), 0.011 (0.008-0.014), 0.011 (0.005-0.018), and 0.026 (0.018-0.034) per IQR increase in PM2.5 concentrations, respectively. This study supports the association of long-term exposure to PM2.5 and its chemical components with poor sleep quality.
Collapse
Affiliation(s)
- Zhupei Yuan
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ying Wang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhicheng Du
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shao Lin
- Department of Environmental Health Sciences, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Jincai Zeng
- Department of Preventive Health, Dongguan People's Hospital, Dongguan, China
| | - Xurui Sun
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuqin Zhang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xinlei Deng
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Yongqing Sun
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jing Wei
- Department of Atmospheric and Oceanic Science, Earth System Science Interdisciplinary Center, University of Maryland, College Park, MD, 20740, USA
| | - Man Zhang
- Department of nosocomial infection management, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jie Sun
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jing Gu
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wangjian Zhang
- Department of Medical Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Yuantao Hao
- Peking University Center for Public Health and Epidemic Preparedness & Response, Peking, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, China.
| |
Collapse
|
8
|
Sun X, Jia X, Tan Z, Fan D, Chen M, Cui N, Liu A, Liu D. Oral Nanoformulations in Cardiovascular Medicine: Advances in Atherosclerosis Treatment. Pharmaceuticals (Basel) 2024; 17:919. [PMID: 39065770 PMCID: PMC11279631 DOI: 10.3390/ph17070919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis (AS) is the formation of atherosclerotic plaques on the walls of the arteries, causing them to narrow. If this occurs in the coronary arteries, the blood vessels may be completely blocked, resulting in myocardial infarction; if it occurs in the blood vessels of the brain, the blood vessels may be blocked, resulting in cerebral infarction, i.e., stroke. Studies have shown that the pathogenesis of atherosclerosis involves the processes of inflammation, lipid infiltration, oxidative stress, and endothelial damage, etc. SIRT, as a key factor regulating the molecular mechanisms of oxidative stress, inflammation, and aging, has an important impact on the pathogenesis of plaque formation, progression, and vulnerability. Statistics show that AS accounts for about 50 per cent of deaths in Western countries. Currently, oral medication is the mainstay of AS treatment, but its development is limited by side effects, low bioavailability and other unfavourable factors. In recent years, with the rapid development of nano-preparations, researchers have combined statins and natural product drugs within nanopreparations to improve their bioavailability. Based on this, this paper summarises the main pathogenesis of AS and also proposes new oral nanoformulations such as liposomes, nanoparticles, nanoemulsions, and nanocapsules to improve their application in the treatment of AS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aidong Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| |
Collapse
|
9
|
Ding R, Huang L, Yan K, Sun Z, Duan J. New insight into air pollution-related cardiovascular disease: an adverse outcome pathway framework of PM2.5-associated vascular calcification. Cardiovasc Res 2024; 120:699-707. [PMID: 38636937 DOI: 10.1093/cvr/cvae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| |
Collapse
|
10
|
Koh H, Kang W, Mao YY, Park J, Kim S, Hong SH, Lee JH. Employment of diverse in vitro systems for analyzing multiple aspects of disease, hereditary hemorrhagic telangiectasia (HHT). Cell Biosci 2024; 14:65. [PMID: 38778363 PMCID: PMC11110195 DOI: 10.1186/s13578-024-01247-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND In vitro disease modeling enables translational research by providing insight into disease pathophysiology and molecular mechanisms, leading to the development of novel therapeutics. Nevertheless, in vitro systems have limitations for recapitulating the complexity of tissues, and a single model system is insufficient to gain a comprehensive understanding of a disease. RESULTS Here we explored the potential of using several models in combination to provide mechanistic insight into hereditary hemorrhagic telangiectasia (HHT), a genetic vascular disorder. Genome editing was performed to establish hPSCs (H9) with ENG haploinsufficiency and several in vitro models were used to recapitulate the functional aspects of the cells that constitute blood vessels. In a 2D culture system, endothelial cells showed early senescence, reduced viability, and heightened susceptibility to apoptotic insults, and smooth muscle cells (SMCs) exhibited similar behavior to their wild-type counterparts. Features of HHT were evident in 3D blood-vessel organoid systems, including thickening of capillary structures, decreased interaction between ECs and surrounding SMCs, and reduced cell viability. Features of ENG haploinsufficiency were observed in arterial and venous EC subtypes, with arterial ECs showing significant impairments. Molecular biological approaches confirmed the significant downregulation of Notch signaling in HHT-ECs. CONCLUSIONS Overall, we demonstrated refined research strategies to enhance our comprehension of HHT, providing valuable insights for pathogenic analysis and the exploration of innovative therapeutic interventions. Additionally, these results underscore the importance of employing diverse in vitro systems to assess multiple aspects of disease, which is challenging using a single in vitro system.
Collapse
Affiliation(s)
- Hyebin Koh
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Woojoo Kang
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Ying-Ying Mao
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, Republic of Korea
| | - Jisoo Park
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Republic of Korea
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea.
- KW-Bio Co., Ltd, Chuncheon, South Korea.
| | - Jong-Hee Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea.
| |
Collapse
|
11
|
Wass SY, Hahad O, Asad Z, Li S, Chung MK, Benjamin EJ, Nasir K, Rajagopalan S, Al-Kindi SG. Environmental Exposome and Atrial Fibrillation: Emerging Evidence and Future Directions. Circ Res 2024; 134:1029-1045. [PMID: 38603473 PMCID: PMC11060886 DOI: 10.1161/circresaha.123.323477] [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] [Indexed: 04/13/2024]
Abstract
There has been increased awareness of the linkage between environmental exposures and cardiovascular health and disease. Atrial fibrillation is the most common sustained cardiac arrhythmia, affecting millions of people worldwide and contributing to substantial morbidity and mortality. Although numerous studies have explored the role of genetic and lifestyle factors in the development and progression of atrial fibrillation, the potential impact of environmental determinants on this prevalent condition has received comparatively less attention. This review aims to provide a comprehensive overview of the current evidence on environmental determinants of atrial fibrillation, encompassing factors such as air pollution, temperature, humidity, and other meteorologic conditions, noise pollution, greenspace, and the social environment. We discuss the existing evidence from epidemiological and mechanistic studies, critically evaluating the strengths and limitations of these investigations and the potential underlying biological mechanisms through which environmental exposures may affect atrial fibrillation risk. Furthermore, we address the potential implications of these findings for public health and clinical practice and identify knowledge gaps and future research directions in this emerging field.
Collapse
Affiliation(s)
- Sojin Youn Wass
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, OH (M.K.C., S.Y.W.)
| | - Omar Hahad
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Germany (O.H.)
| | - Zain Asad
- Division of Cardiovascular Medicine, University of Oklahoma Medical Center, Oklahoma City (Z.A.)
| | - Shuo Li
- Biomedical Engineering, Case Western Reserve University, Cleveland, OH (S.L.)
| | - Mina K Chung
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, OH (M.K.C., S.Y.W.)
| | - Emelia J Benjamin
- Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine and Department of Epidemiology, Boston University School of Public Health, MA (E.J.B.)
| | - Khurram Nasir
- Cardiovascular Prevention and Wellness, DeBakey Heart and Vascular Center, Houston Methodist, TX (K.N., S.G.A.-K.)
| | - Sanjay Rajagopalan
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH (S.R.)
- Case Western Reserve University School of Medicine, Cleveland, OH (S.R.)
| | - Sadeer G Al-Kindi
- Cardiovascular Prevention and Wellness, DeBakey Heart and Vascular Center, Houston Methodist, TX (K.N., S.G.A.-K.)
| |
Collapse
|
12
|
Mei H, Wu D, Yong Z, Cao Y, Chang Y, Liang J, Jiang X, Xu H, Yang J, Shi X, Xie R, Zhao W, Wu Y, Liu Y. PM 2.5 exposure exacerbates seizure symptoms and cognitive dysfunction by disrupting iron metabolism and the Nrf2-mediated ferroptosis pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 910:168578. [PMID: 37981141 DOI: 10.1016/j.scitotenv.2023.168578] [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: 08/23/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
In recent years, air pollution has garnered global attention due to its ability to traverse borders and regions, thereby impacting areas far removed from the emission sources. While prior studies predominantly focused on the deleterious effects of PM2.5 on the respiratory and cardiovascular systems, emerging evidence has highlighted the potential risks of PM2.5 exposure to the central nervous system. Nonetheless, research elucidating the potential influences of PM2.5 exposure on seizures, specifically in relation to neuronal ferroptosis, remains limited. In this study, we investigated the potential effects of PM2.5 exposure on seizure symptoms and seizures-induced hippocampal neuronal ferroptosis. Our findings suggest that seizure patients residing in regions with high PM2.5 levels are more likely to disturb iron homeostasis and the Nrf2 dependent ferroptosis pathway compared to those living in areas with lower PM2.5 levels. The Morris Water Maze test, Racine scores, and EEG recordings in epileptic mice suggest that PM2.5 exposure can exacerbate seizure symptoms and cognitive dysfunction. Neurotoxic effects of PM2.5 exposure were demonstrated via Nissl staining and CCK-8 assays. Direct evidence of PM2.5-induced hippocampal neuronal ferroptosis was provided through TEM images. Additionally, increased Fe2+ and lipid ROS levels indirectly supported the notion of PM2.5-induced hippocampal ferroptosis. Therefore, our study underscores the necessity of preventing and controlling PM2.5 levels, particularly for patients with seizures.
Collapse
Affiliation(s)
- Huiya Mei
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongqin Wu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zenghua Yong
- Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingsi Cao
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuanjin Chang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Junjie Liang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaofan Jiang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hua Xu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jiatao Yang
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xian Shi
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ruijin Xie
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Wenjing Zhao
- Yangzhou Key Laboratory of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| | - Yu Wu
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yueying Liu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
13
|
Krittanawong C, Qadeer YK, Hayes RB, Wang Z, Thurston GD, Virani S, Lavie CJ. PM 2.5 and cardiovascular diseases: State-of-the-Art review. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2023; 19:200217. [PMID: 37869561 PMCID: PMC10585625 DOI: 10.1016/j.ijcrp.2023.200217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/09/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023]
Abstract
Air pollution, especially exposure to particulate matter 2.5 (PM2.5), has been associated with an increase in morbidity and mortality around the world. Specifically, it seems that PM2.5 promotes the development of cardiovascular risk factors such as hypertension and atherosclerosis, while being associated with an increased risk of cardiovascular diseases, including myocardial infarction (MI), stroke, heart failure, and arrhythmias. In this review, we seek to elucidate the pathophysiological mechanisms by which exposure to PM2.5 can result in adverse cardiovascular outcomes, in addition to understanding the link between exposure to PM2.5 and cardiovascular events. It is hypothesized that PM2.5 functions via 3 mechanisms: increased oxidative stress, activation of the inflammatory pathway of the immune system, and stimulation of the autonomic nervous system which ultimately promote endothelial dysfunction, atherosclerosis, and systemic inflammation that can thus lead to cardiovascular events. It is important to note that the various cardiovascular associations of PM2.5 differ regarding the duration of exposure (short vs long) to PM2.5, the source of PM2.5, and regulations regarding air pollution in the area where PM2.5 is prominent. Current strategies to reduce PM2.5 exposure include personal strategies such as avoiding high PM2.5 areas such as highways or wearing masks outdoors, to governmental policies restricting the amount of PM2.5 produced by organizations. This review, by highlighting the significant impact between PM2.5 exposure and cardiovascular health will hopefully bring awareness and produce significant change regarding dealing with PM2.5 levels worldwide.
Collapse
Affiliation(s)
| | | | - Richard B. Hayes
- Division of Epidemiology, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Zhen Wang
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
- Division of Health Care Policy and Research, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - George D. Thurston
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Salim Virani
- Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
- The Aga Khan University, Karachi, Pakistan
| | - Carl J. Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA, USA
| |
Collapse
|
14
|
Chen W, Luo Y, Quan J, Zhou J, Yi B, Huang Z. PM 2.5 induces renal tubular injury by activating NLRP3-mediated pyroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115490. [PMID: 37742582 DOI: 10.1016/j.ecoenv.2023.115490] [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: 05/03/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
Fine particulate matter (PM2.5)-related health issues have received increasing attention as a worldwide public health problem, and PM2.5-related chronic kidney disease (CKD) has been emerging over the years. Limited research has focused on the mechanism of PM2.5-induced kidney disease. To investigate the impact of PM2.5 on the kidney and its potential mechanism, we generated a PM2.5-exposed C57BL/6 mouse model by using Shanghai Meteorological and Environment Animal Exposure System (Shanghai-METAS) for 12 weeks, urine, blood and kidney tissues were collected. The pathological changes and the function of the kidney were measured after PM2.5 exposure for 12 weeks. Along with glomerular damage, tubular damage was also severe in PM2.5-induced mice. The results of mRNA-seq indicate that pyroptosis is involved. Pyroptosis is defined as caspase-1-dependent programmed cell death in response to insults. The expression of the nucleotide-binding and oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3), Caspase-1, gasdermin D (GSDMD) and IL-1β was detected. NLRP3 inflammasome activation and subsequent pyroptosis were observed in PM2.5-exposed kidney tissues and PM2.5-exposed Bumpt cells too. At the meantime, the inhibitors of NLRP3 and caspase-1 were applied to the PM2.5 exposed Bumpt cells. It turned out to have a significant rescue effect of the inhibitors. This study revealed new insights into PM2.5-induced kidney injury and specific kidney pathological damage, as well as morphological changes, and defined the important role of pyroptosis in PM2.5-induced kidney dysfunction.
Collapse
Affiliation(s)
- Weilin Chen
- Department of Nephrology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; The Critical Kidney Disease Research Center of Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China
| | - Yanfang Luo
- Department of Nephrology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; The Critical Kidney Disease Research Center of Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China
| | - Jingjing Quan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; The Critical Kidney Disease Research Center of Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China
| | - Ji Zhou
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai 200030, China
| | - Bin Yi
- Department of Nephrology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; The Critical Kidney Disease Research Center of Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; Furong Laboratory, Changsha 410013, Hunan, China.
| | - Zhijun Huang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410013, Hunan, China; Furong Laboratory, Changsha 410013, Hunan, China; Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
15
|
Wang Y, Qiu X, Wei Y, Schwartz JD. Long-Term Exposure to Ambient PM 2.5 and Hospitalizations for Myocardial Infarction Among US Residents: A Difference-in-Differences Analysis. J Am Heart Assoc 2023; 12:e029428. [PMID: 37702054 PMCID: PMC10547266 DOI: 10.1161/jaha.123.029428] [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: 01/09/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023]
Abstract
Background Air pollution has been recognized as an untraditional risk factor for myocardial infarction (MI). However, the MI risk attributable to long-term exposure to fine particulate matter ≤2.5 μm in aerodynamic diameter (PM2.5) is unclear, especially in younger populations, and few studies have represented the general population or had power to examine comorbidities. Methods and Results We applied the difference-in-differences approach to estimate the relationship between annual PM2.5 exposure and hospitalizations for MI among US residents and further identified potential susceptible subpopulations. All hospital admissions for MI in 10 US states over the period 2002 to 2016 were obtained from the Healthcare Cost and Utilization Project State Inpatient Database. In total, 1 914 684 MI hospital admissions from 8106 zip codes were included in this study. We observed a 1.35% (95% CI, 1.11-1.59) increase in MI hospitalization rate for 1-μg/m3 increase in annual PM2.5 exposure. The estimate was robust to adjustment for surface pressure, relative humidity, and copollutants. In the population exposed to ≤12 μg/m3, there was a larger increment of 2.17% (95% CI, 1.79-2.56) in hospitalization rate associated with 1-μg/m3 increase in PM2.5. Young people (0-34 years of age) and elderly people (≥75 years of age) were the 2 most susceptible age groups. Residents living in more densely populated or poorer areas and individuals with comorbidities were observed to be at a greater risk. Conclusions This study indicates long-term residential exposure to PM2.5 could increase risk of MI among the general US population, people with comorbidities, and poorer individuals. The association persists below current standards.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMA
| | - Xinye Qiu
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMA
| | - Yaguang Wei
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMA
| | - Joel D. Schwartz
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMA
| |
Collapse
|
16
|
Torrico-Lavayen R, Vargas-Alarcón G, Riojas-Rodriguez H, Sánchez-Guerra M, Texcalac-Sangrador JL, Ortiz-Panozo E, Gutiérrez-Avila I, De Vizcaya-Ruiz A, Cardenas A, Posadas-Sánchez R, Osorio-Yáñez C. Long-term exposure to ambient fine particulate matter and carotid intima media thickness at bilateral, left and right in adults from Mexico City: Results from GEA study. CHEMOSPHERE 2023; 335:139009. [PMID: 37245594 DOI: 10.1016/j.chemosphere.2023.139009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/13/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND PM2.5 exposure has been associated with intima-media thickness (cIMT) increase. However, very few studies distinguished between left and right cIMT in relation to PM2.5 exposure. AIM To evaluate associations between chronic exposure to PM2.5 and cIMT at bilateral, left, and right in adults from Mexico City. METHODS This study comprised 913 participants from the control group, participants without personal or family history of cardiovascular disease, of the Genetics of Atherosclerosis Disease Mexican study (GEA acronym in Spanish), recruited at the Instituto Nacional de Cardiología Ignacio Chávez from June 2008 to January 2013. To assess the associations between chronic exposure to PM2.5 (per 5 μg/m3 increase) at different lag years (1-4 years) and cIMT (bilateral, left, and right) we applied distributed lag non-linear models (DLNMs). RESULTS The median and interquartile range for cIMT at bilateral, left, and right, were 630 (555, 735), 640 (550, 750), and 620 (530, 720) μm, respectively. Annual average PM2.5 exposure was 26.64 μg/m3, with median and IQR, of 24.46 (23.5-25.46) μg/m3. Results from DLNMs adjusted for age, sex, body mass index, low-density lipoproteins, and glucose, showed that PM2.5 exposure for year 1 and 2, were positively and significantly associated with right-cIMT [6.99% (95% CI: 3.67; 10.42) and 2.98% (0.03; 6.01), respectively]. Negative associations were observed for PM2.5 at year 3 and 4 and right-cIMT; however only year 3 was statistically significant [-2.83% (95% CI: 5.12; -0.50)]. Left-cIMT was not associated with PM2.5 exposure at any lag year. The increase in bilateral cIMT followed a similar pattern as that observed for right-cIMT, but with lower estimates. CONCLUSIONS Our results suggest different susceptibility between left and right cIMT associated with PM2.5 exposure highlighting the need of measuring both, left and right cIMT, regarding ambient air pollution in epidemiological studies.
Collapse
Affiliation(s)
- Rocio Torrico-Lavayen
- Departamento de Patología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico; Department of Environmental Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | | | | | | | - Eduardo Ortiz-Panozo
- Center of Population Health Research, National Institute of Public Health, Cuernavaca, Mexico; Department of Epidemiology, Harvard T.H. Chan School of Public Health. Boston, United States
| | - Iván Gutiérrez-Avila
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Andrea De Vizcaya-Ruiz
- Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, CA, United States
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, United States
| | - Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | - Citlalli Osorio-Yáñez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, 04510, Mexico; Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación en Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico.
| |
Collapse
|
17
|
Guan L, Shi H, Tian J, Wang X, Liu N, Wang C, Zhang Z. PM 2.5 induces the inflammatory response in rat spleen lymphocytes through autophagy activation of NLRP3 inflammasome. Mol Immunol 2023; 161:74-81. [PMID: 37506549 DOI: 10.1016/j.molimm.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023]
Abstract
Recent evidence has suggested that fine particulate matter (PM2.5) can induce inflammatory injury in spleen. However, the underlying mechanisms of injury remain enigmatic. In this study, we aim to clarify the inflammatory injury mechanisms of PM2.5 through investigating the crosstalk between autophagy and nod-like receptor protein 3 (NLRP3) inflammasome. The spleen lymphocytes were extracted from SD rats and subjected to PM2.5 and its water-soluble components. The CCK-8 assay was utilized to explore the effects of PM2.5 and its water-soluble components on lymphocytes. Then, the effects of PM2.5 and its water-soluble components exposure on autophagy and NLRP3 inflammasome were detected by qRT-PCR, western blotting, and immunofluorescence staining. The autophagosome production was observed under the transmission electron microscope. The autophagy inhibitor 3-methyladenine (3MA) following PM2.5 water-soluble components was used to investigate the regulation of NLRP3 inflammasome by autophagy. We found that PM2.5 and its water-soluble components decreased the viability of spleen lymphocytes in a dose-dependent manner. PM2.5 exposure and its water-soluble components exposure activated the autophagy and NlRP3 inflammasome, as indicated by an increased expression of LC3, P62, NLRP3, Caspase-1 p10, and increased release of IL-1β. Furthermore, the treatment with autophagy inhibitor 3MA attenuated the production of autophagosome and NLRP3 inflammasome induced by PM2.5 water-soluble components with decreased expression of NLRP3, Caspase-1 p10, and diminished production of IL-1β. These results suggested that PM2.5 and its water-soluble components could induce autophagy and inflammatory response through NLRP3 inflammasome in spleen lymphocytes, while the NLRP3 inflammasome induced by PM2.5 could be significantly alleviated by inhibition of autophagy, further providing new insights for the understanding of spleen injury caused by PM2.5.
Collapse
Affiliation(s)
- Linlin Guan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of Clinical Laboratory, Shanxi Provincial People's Hospital,Taiyuan, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of public health, Linyi Cancer Hospital, Linyi, China
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China.
| |
Collapse
|
18
|
Gałuszka-Bulaga A, Tkacz K, Węglarczyk K, Siedlar M, Baran J. Air pollution induces pyroptosis of human monocytes through activation of inflammasomes and Caspase-3-dependent pathways. J Inflamm (Lond) 2023; 20:26. [PMID: 37563611 PMCID: PMC10416410 DOI: 10.1186/s12950-023-00353-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
According to the World Health Organization (WHO), air pollution is one of the most serious threats for our planet. Despite a growing public awareness of the harmful effects of air pollution on human health, the specific influence of particulate matter (PM) on human immune cells remains poorly understood. In this study, we investigated the effect of PM on peripheral blood monocytes in vitro. Monocytes from healthy donors (HD) were exposed to two types of PM: NIST (SRM 1648a, standard urban particulate matter from the US National Institute for Standards and Technology) and LAP (SRM 1648a with the organic fraction removed). The exposure to PM-induced mitochondrial ROS production followed by the decrease of mitochondrial membrane potential and activation of apoptotic protease activating factor 1 (Apaf-1), Caspase-9, and Caspase-3, leading to the cleavage of Gasdermin E (GSDME), and initiation of pyroptosis. Further analysis showed a simultaneous PM-dependent activation of inflammasomes, including NLRP3 (nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3) and Caspase-1, followed by cleavage of Gasdermin D (GSDMD) and secretion of IL-1β. These observations suggest that PM-treated monocytes die by pyroptosis activated by two parallel signaling pathways, related to the inorganic and organic PM components. The release of IL-1β and expression of danger-associated molecular patterns (DAMPs) by pyroptotic cells further activated the remnant viable monocytes to produce inflammatory cytokines (TNF-α, IL-6, IL-8) and protected them from death induced by the second challenge with PM.In summary, our report shows that PM exposure significantly impacts monocyte function and induces their death by pyroptosis. Our observations indicate that the composition of PM plays a crucial role in this process-the inorganic fraction of PM is responsible for the induction of the Caspase-3-dependent pyroptotic pathway. At the same time, the canonical inflammasome path is activated by the organic components of PM, including LPS (Lipopolysaccharide/endotoxin). PM-induced pyroptosis of human monocytes. Particulate matter (PM) treatment affects monocytes viability already after 15 min of their exposure to NIST or LAP in vitro. The remnant viable monocytes in response to danger-associated molecular patterns (DAMPs) release pro-inflammatory cytokines and activate Th1 and Th17 cells. The mechanism of PM-induced cell death includes the increase of reactive oxygen species (ROS) production followed by collapse of mitochondrial membrane potential (ΔΨm), activation of Apaf-1, Caspase-9 and Caspase-3, leading to activation of Caspase-3-dependent pyroptotic pathway, where Caspase-3 cleaves Gasdermin E (GSDME) to produce a N-terminal fragment responsible for the switch from apoptosis to pyroptosis. At the same time, PM activates the canonical inflammasome pathway, where activated Caspase-1 cleaves the cytosolic Gasdermin D (GSDMD) to produce N-terminal domain allowing IL-1β secretion. As a result, PM-treated monocytes die by pyroptosis activated by two parallel pathways-Caspase-3-dependent pathway related to the inorganic fraction of PM and the canonical inflammasome pathway dependent on the organic components of PM.
Collapse
Affiliation(s)
- Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
| | - Karolina Tkacz
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| |
Collapse
|
19
|
Nemmar A, Beegam S, Zaaba NE, Elzaki O, Pathan A, Ali BH. Waterpipe smoke inhalation induces lung injury and aortic endothelial dysfunction in mice. Physiol Res 2023; 72:337-347. [PMID: 37449747 PMCID: PMC10669000 DOI: 10.33549/physiolres.935042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/28/2023] [Indexed: 08/26/2023] Open
Abstract
Waterpipe tobacco smoking (WPS) inhalation has been shown to trigger endothelial dysfunction and atherosclerosis. However, the mechanisms underlying these effects are still unknown. Here, we assessed the impact and underlying mechanism of WPS exposure for one month on endothelial dysfunction using aortic tissue of mice. The duration of the session was 30 min/day and 5 days/week. Control mice were exposed to air. Inhalation of WPS induced an increase in the number of macrophages and neutrophils and the concentrations of protein, tumor necrosis factor alpha (TNF alpha), interleukin (IL)-1beta, and glutathione in bronchoalveolar lavage fluid. Moreover, the concentrations of proinflammatory cytokines (TNF alpha, IL-6 and IL-1beta), adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, E-selectin and P-selectin) and markers of oxidative stress (lipid peroxidation, glutathione, superoxide dismutase and nitric oxide) in aortic homogenates of mice exposed to WPS were significantly augmented compared with air exposed mice. Likewise, the concentration of galectin-3 was significantly increased in the aortic homogenates of mice exposed to WPS compared with control group. WPS inhalation induced vascular DNA damage assessed by comet assay and apoptosis characterized by a significant increase in cleaved caspase-3. While the aortic expression of phosphorylated nuclear factor kappaB (NF-kappaB) was significantly increased following WPS inhalation, the concentration of sirtuin 1 (SIRT1) was significantly decreased in WPS group compared with air-exposed group. In conclusion, our study provided evidence that WPS inhalation triggers lung injury and endothelial inflammation, oxidative stress and apoptosis which were associated with nuclear factor-kappaB activation and SIRT1 down-regulation.
Collapse
Affiliation(s)
- A Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates. and
| | | | | | | | | | | |
Collapse
|
20
|
Zhen AX, Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Cho SJ, Hyun JW. 3-Bromo-4,5-dihydroxybenzaldehyde Protects Keratinocytes from Particulate Matter 2.5-Induced Damages. Antioxidants (Basel) 2023; 12:1307. [PMID: 37372037 DOI: 10.3390/antiox12061307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence can be activated by several stimuli, including ultraviolet radiation and air pollutants. This study aimed to evaluate the protective effect of marine algae compound 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on particulate matter 2.5 (PM2.5)-induced skin cell damage in vitro and in vivo. The human HaCaT keratinocyte was pre-treated with 3-BDB and then with PM2.5. PM2.5-induced reactive oxygen species (ROS) generation, lipid peroxidation, mitochondrial dysfunction, DNA damage, cell cycle arrest, apoptotic protein expression, and cellular senescence were measured using confocal microscopy, flow cytometry, and Western blot. The present study exhibited PM2.5-generated ROS, DNA damage, inflammation, and senescence. However, 3-BDB ameliorated PM2.5-induced ROS generation, mitochondria dysfunction, and DNA damage. Furthermore, 3-BDB reversed the PM2.5-induced cell cycle arrest and apoptosis, reduced cellular inflammation, and mitigated cellular senescence in vitro and in vivo. Moreover, the mitogen-activated protein kinase signaling pathway and activator protein 1 activated by PM2.5 were inhibited by 3-BDB. Thus, 3-BDB suppressed skin damage induced by PM2.5.
Collapse
Affiliation(s)
- Ao-Xuan Zhen
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei-Jing Piao
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung-Ah Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Suk-Ju Cho
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
21
|
Jia R, Wei M, Lei J, Meng X, Du R, Yang M, Lu X, Jiang Y, Cao R, Wang L, Song L. PM 2.5 induce myocardial injury in hyperlipidemic mice through ROS-pyroptosis signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114699. [PMID: 36889212 DOI: 10.1016/j.ecoenv.2023.114699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/07/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Exposure to particulate matters with diameters below 2.5 µm (PM2.5) is considered a major risk factor for cardiovascular diseases (CVDs). The closest associations between PM2.5 and CVDs have been observed in hyperbetalipoproteinemia cases, although the detailed underpinning mechanism remains undefined. In this work, hyperlipidemic mice and H9C2 cells were used to examine the effects of PM2.5 on myocardial injury and their underlying mechanisms. The results revealed that PM2.5 exposure caused severe myocardial damage in the high-fat mouse model. Oxidative stress and pyroptosis were also observed along with myocardial injury. After inhibiting pyroptosis with disulfiram (DSF), the level of pyroptosis was effectively reduced as well as myocardial injury, suggesting that PM2.5 induced the pyroptosis pathway and further caused myocardial injury and cell death. Afterwards, by suppressing PM2.5-induced oxidative stress with N-acetyl-L-cysteine (NAC), myocardial injury was markedly ameliorated, and the upregulation of pyroptosis markers was reversed, which indicated that PM2.5-pyroptosis was also improved. Taken together, this study revealed that PM2.5 induce myocardial injury through the ROS-pyroptosis signaling pathway in hyperlipidemia mice models, providing a potential approach for clinical interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China; Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, People's Republic of China
| | - Min Wei
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China
| | - Jinrong Lei
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China
| | - Xianzong Meng
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China; Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Rui Du
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China
| | - Mengxin Yang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning Province 116023, People's Republic of China
| | - Xinjun Lu
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning Province 116000, People's Republic of China
| | - Yizhu Jiang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning Province 116023, People's Republic of China
| | - Ran Cao
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning Province 116023, People's Republic of China
| | - Lili Wang
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, People's Republic of China.
| | - Laiyu Song
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning Province 116044, People's Republic of China.
| |
Collapse
|
22
|
Chen D, Du Y, Ye S, Yu J. Astragaloside IV protects against oxidized low-density lipoprotein-induced injury in human umbilical vein endothelial cells via the histone deacetylase 9 (HDAC9)/NF-κB axis. ENVIRONMENTAL TOXICOLOGY 2023; 38:534-544. [PMID: 36322813 DOI: 10.1002/tox.23696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Atherosclerosis is a main cause of multiple cardiovascular diseases, and cell damage of human umbilical vein endothelial cells (HUVECs) was reported to participate in the development of atherosclerosis. In this study, we aimed to study the action of Astragaloside IV (ASV) on AS development using in vitro AS cell model. METHODS MTT assay, EdU staining assay, and flow cytometry were utilized for detection of cell proliferation and apoptosis, respectively. The protein expression of histone deacetylase 9 (HDAC9), Bax, Bcl-2, p-P65, P65, p-IκBα, and IκBα was gaged using western blot. The angiogenesis was evaluated by tube formation assay. The inflammatory response was evaluated by ELISA kits. SOD activity and MDA level were detected using the matched commercial kits. RT-qPCR was used for HDAC9 mRNA expression measurement. RESULTS Oxidized low-density lipoprotein (ox-LDL) significantly repressed cell proliferation, angiogenesis, and enhanced apoptosis, inflammation, and oxidative stress in HUVECs. ASV addition could alleviate ox-LDL-caused cell damage in HUVECs. Moreover, HDAC9 was overexpressed in AS patients and AS cell model. Functionally, HDAC9 knockdown also exhibited the protective role in ox-LDL-treated HUVECs. In addition, ASV treatment protected against ox-LDL-induced damage in HUVECs via targeting HDAC9. ASV could inactivate the NF-κB pathway via regulating HDAC9 in AS cell model. CONCLUSION ASV exerted the protective effects on ox-LDL-induced damage in HUVECs through the HDAC9/NF-κB axis.
Collapse
Affiliation(s)
- Decai Chen
- Department of Vascular Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China
| | - Yan Du
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| | - Shouwan Ye
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| | - Jinsong Yu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| |
Collapse
|
23
|
Zhao T, Li X, Qian H, Miao X, Zhu Y, Wang J, Hui J, Zhou L, Ye L. PM 2.5 induces the abnormal lipid metabolism and leads to atherosclerosis via Notch signaling pathway in rats. Toxicology 2023; 485:153415. [PMID: 36603807 DOI: 10.1016/j.tox.2022.153415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
PM2.5 can affect the lipid metabolism and cause atherosclerosis. Abnormal lipid metabolism is a sever risk factor of atherosclerosis and the underlying molecular mechanism still remains unclear. In this study, GPL16956 Agilent-045997 Arraystar human lncRNA microarray V3 (Probe Name Version) platform was used to detect the different genes of lipid metabolism between the normal arterial intima and advanced atherosclerotic plaque, which were downloaded from GEO database. A high-fat diet and vitamin D3 were administered to Wistar rats to establish the atherosclerotic model and another normal healthy 56 rats were used as the non-atherosclerotic exposure groups. The atherosclerotic rats and non-atherosclerotic rats were randomly divided into 4 PM2.5 groups (0, 1.5, 7.5, 37.5 mg/kg), respectively. The results of bioinformatics showed changes in the Notch1, Dll1, Hes1, LDLR and ABCG1 levels. PM2.5 exposure could produce damage to the physiological structure of the aorta, and aggravate atherosclerosis in rats from both non-atherosclerotic and atherosclerotic groups. With the increase of the exposure dose, the levels of TC and TG significantly increased. PM2.5 exposure significantly affected the expression levels of PPARγ, ABCA1, LDLR, CD36, SR-BI and SREBP2. PM2.5 exposure could also affect the expression levels of the Notch signaling pathways which was significantly correlated with the levels of TC and TG. The results proved that PM2.5 exposure could induce and aggravate the atherosclerosis in rats by disrupting lipid metabolism in which Notch signaling pathway may play a significant role.
Collapse
Affiliation(s)
- Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Xu Li
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Honghao Qian
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Xiaohan Miao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Ying Zhu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Jia Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Ju Hui
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China.
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China.
| |
Collapse
|
24
|
Fathieh S, Grieve SM, Negishi K, Figtree GA. Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution-A State-of-the-Art Review. Heart Lung Circ 2023; 32:26-42. [PMID: 36585310 DOI: 10.1016/j.hlc.2022.11.014] [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: 03/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 12/29/2022]
Abstract
Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm-10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual's exposure, response, and likely risk.
Collapse
Affiliation(s)
- Sina Fathieh
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Stuart M Grieve
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia; Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan; Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia.
| |
Collapse
|
25
|
Zeng Y, Jin H, Wang J, Guo C, Chen W, Tan Y, Wang L, Zhou Z. An optimized method for intratracheal instillation in mice. J Pharmacol Toxicol Methods 2022; 118:107230. [DOI: 10.1016/j.vascn.2022.107230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/12/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022]
|
26
|
Chen Z, Liu P, Xia X, Wang L, Li X. The underlying mechanism of PM2.5-induced ischemic stroke. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 310:119827. [PMID: 35917837 DOI: 10.1016/j.envpol.2022.119827] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Under the background of global industrialization, PM2.5 has become the fourth-leading risk factor for ischemic stroke worldwide, according to the 2019 GBD estimates. This highlights the hazards of PM2.5 for ischemic stroke, but unfortunately, PM2.5 has not received the attention that matches its harmfulness. This article is the first to systematically describe the molecular biological mechanism of PM2.5-induced ischemic stroke, and also propose potential therapeutic and intervention strategies. We highlight the effect of PM2.5 on traditional cerebrovascular risk factors (hypertension, hyperglycemia, dyslipidemia, atrial fibrillation), which were easily overlooked in previous studies. Additionally, the effects of PM2.5 on platelet parameters, megakaryocytes activation, platelet methylation, and PM2.5-induced oxidative stress, local RAS activation, and miRNA alterations in endothelial cells have also been described. Finally, PM2.5-induced ischemic brain pathological injury and microglia-dominated neuroinflammation are discussed. Our ultimate goal is to raise the public awareness of the harm of PM2.5 to ischemic stroke, and to provide a certain level of health guidance for stroke-susceptible populations, as well as point out some interesting ideas and directions for future clinical and basic research.
Collapse
Affiliation(s)
- Zhuangzhuang Chen
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Peilin Liu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaoshuang Xia
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Interdisciplinary Innovation Centre for Health and Meteorology, Tianjin, China
| | - Lin Wang
- Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Interdisciplinary Innovation Centre for Health and Meteorology, Tianjin, China
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Interdisciplinary Innovation Centre for Health and Meteorology, Tianjin, China.
| |
Collapse
|
27
|
Wang B, Sun T, Sun L, Li L, Wan H, Ding Z, Ye X. Amygdalin attenuates PM2.5-induced human umbilical vein endothelial cell injury via the TLR4/NF-κB and Bcl-2/Bax signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1476-1485. [PMID: 36178164 PMCID: PMC9828314 DOI: 10.3724/abbs.2022136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/17/2022] [Indexed: 12/29/2022] Open
Abstract
Mounting evidence supports that long-term exposure to fine particle pollutants (PM2.5) is closely implicated in cardiovascular diseases, especially atherosclerosis. Amygdalin is reported to attenuate external stimuli-induced cardiovascular diseases. However, the underlying mechanisms are still not understood. In this study, we aim to explore the protective effects of amygdalin on PM2.5-induced human umbilical vein endothelial cell (HUVEC) injury and unravel the specific mechanisms by MTT, DCFH-DA, biochemical, immunofluorescence, ELISA, RT-qPCR, flow cytometry, TUNEL and western blot analysis. The results reveal that amygdalin reverses PM2.5-induced cytotoxicity and attenuates intracellular ROS production. Moreover, amygdalin increases the levels of SOD and GSH and alleviates the MDA content. Additionally, amygdalin causes a decline of IL-6, IL-1β, TNF-α and COX-2 levels. Moreover, amygdalin inhibits NF-κB p50 and TLR4 protein expressions and NF-κB p65 nuclear translocation. Concomitantly, a decline of phospho-NF-κB p65/NF-κB p65 and phospho-IκB-α/IκB-α is detected. Meanwhile, amygdalin pretreatment reduces HUVEC apoptosis. In addition, amygdalin triggers an upregulation of Bcl-2 and a downregulation of Bax after stimulation with PM2.5. Collectively, these results suggest that amygdalin suppresses PM2.5-induced HUVEC injury by regulating the TLR4/NF-κB and Bcl-2/Bax signaling pathways, indicating that amygdalin may be a novel target for atherosclerosis treatments.
Collapse
Affiliation(s)
- Bixu Wang
- School of Medical Technology and Information EngineeringZhejiang Chinese Medical UniversityHangzhou310053China
| | - Tong Sun
- School of Medical Technology and Information EngineeringZhejiang Chinese Medical UniversityHangzhou310053China
| | - Ling Sun
- School of Medical Technology and Information EngineeringZhejiang Chinese Medical UniversityHangzhou310053China
| | - Lan Li
- School of Life SciencesZhejiang Chinese Medical UniversityHangzhou310053China
| | - Haitong Wan
- School of Life SciencesZhejiang Chinese Medical UniversityHangzhou310053China
| | - Zhishan Ding
- School of Medical Technology and Information EngineeringZhejiang Chinese Medical UniversityHangzhou310053China
| | - Xiaoqing Ye
- School of Medical Technology and Information EngineeringZhejiang Chinese Medical UniversityHangzhou310053China
| |
Collapse
|
28
|
Li W, Lin G, Xiao Z, Zhang Y, Li B, Zhou Y, Ma Y, Chai E. A review of respirable fine particulate matter (PM2.5)-induced brain damage. Front Mol Neurosci 2022; 15:967174. [PMID: 36157076 PMCID: PMC9491465 DOI: 10.3389/fnmol.2022.967174] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Respirable fine particulate matter (PM2.5) has been one of the most widely publicized indicators of pollution in recent years. Epidemiological studies have established a strong association between PM2.5, lung disease, and cardiovascular disease. Recent studies have shown that PM2.5 is also strongly associated with brain damage, mainly cerebrovascular damage (stroke) and neurological damage to the brain (changes in cognitive function, dementia, psychiatric disorders, etc.). PM2.5 can pass through the lung–gas–blood barrier and the “gut–microbial–brain” axis to cause systemic oxidative stress and inflammation, or directly enter brain tissue via the olfactory nerve, eventually damaging the cerebral blood vessels and brain nerves. It is worth mentioning that there is a time window for PM2.5-induced brain damage to repair itself. However, the exact pathophysiological mechanisms of brain injury and brain repair are not yet fully understood. This article collects and discusses the mechanisms of PM2.5-induced brain injury and self-repair after injury, which may provide new ideas for the prevention and treatment of cerebrovascular and cerebral neurological diseases.
Collapse
Affiliation(s)
- Wei Li
- The First Clinical Medical College of Gansu University of Chinese Medical, Lan Zhou, China
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
| | - Guohui Lin
- Day Treatment Center II of Gansu Provincial Maternity and Child-Care Hospital, Lan Zhou, China
| | - Zaixing Xiao
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
| | - Yichuan Zhang
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
| | - Bin Li
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
| | - Yu Zhou
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
- The First School of Clinical Medicine of Lanzhou University, Lan Zhou, China
| | - Yong Ma
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
| | - Erqing Chai
- Cerebrovascular Disease Center of Gansu Provincial People's Hospital, Lan Zhou, China
- Key Laboratory of Cerebrovascular Diseases in Gansu Province, Lan Zhou, China
- *Correspondence: Erqing Chai
| |
Collapse
|
29
|
Gałuszka-Bulaga A, Hajto J, Borczyk M, Gołda S, Piechota M, Korostyński M, Rutkowska-Zapała M, Latacz P, Guła Z, Korkosz M, Pera J, Słowik A, Siedlar M, Baran J. Transcriptional Response of Blood Mononuclear Cells from Patients with Inflammatory and Autoimmune Disorders Exposed to "Krakow Smog". Cells 2022; 11:cells11162586. [PMID: 36010662 PMCID: PMC9406644 DOI: 10.3390/cells11162586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the general awareness of the need to reduce air pollution, the efforts were undertaken in Poland to eliminate the pollutants and their harmful effect on human health seem to be insufficient. Moreover, the latest data indicate that the city of Krakow is at the forefront of the most polluted cities worldwide. Hence, in this report, we investigated the impact of particulate matter isolated from the air of Krakow (PM KRK) on the gene expression profile of peripheral blood mononuclear cells (PBMCs) in healthy donors (HD) and patients with atherosclerosis (AS), rheumatoid arthritis (RA) and multiple sclerosis (MS), after in vitro exposure. Blood samples were collected in two seasons, differing in the concentration of PM in the air (below or above a daily limit of 50 µg/m3 for PM 10). Data show that PBMCs exposed in vitro to PM KRK upregulated the expression of genes involved, among others, in pro-inflammatory response, cell motility, and regulation of cell metabolism. The transcriptional effects were observed predominantly in the group of patients with AS and MS. The observed changes seem to be dependent on the seasonal concentration of PM in the air of Krakow and may suggest their important role in the progression of AS, MS, and RA in the residents of Krakow.
Collapse
Affiliation(s)
- Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Małgorzata Borczyk
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Sławomir Gołda
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Marcin Piechota
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Michał Korostyński
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Magdalena Rutkowska-Zapała
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
| | - Paweł Latacz
- Department of Clinical Neurology, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Zofia Guła
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Joanna Pera
- Department of Clinical Neurology, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Agnieszka Słowik
- Department of Clinical Neurology, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Correspondence: ; Tel.: +48-12-65-82-011
| |
Collapse
|
30
|
Basith S, Manavalan B, Shin TH, Park CB, Lee WS, Kim J, Lee G. The Impact of Fine Particulate Matter 2.5 on the Cardiovascular System: A Review of the Invisible Killer. NANOMATERIALS 2022; 12:nano12152656. [PMID: 35957086 PMCID: PMC9370264 DOI: 10.3390/nano12152656] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/26/2022]
Abstract
Air pollution exerts several deleterious effects on the cardiovascular system, with cardiovascular disease (CVD) accounting for 80% of all premature deaths caused by air pollution. Short-term exposure to particulate matter 2.5 (PM2.5) leads to acute CVD-associated deaths and nonfatal events, whereas long-term exposure increases CVD-associated risk of death and reduces longevity. Here, we summarize published data illustrating how PM2.5 may impact the cardiovascular system to provide information on the mechanisms by which it may contribute to CVDs. We provide an overview of PM2.5, its associated health risks, global statistics, mechanistic underpinnings related to mitochondria, and hazardous biological effects. We elaborate on the association between PM2.5 exposure and CVD development and examine preventive PM2.5 exposure measures and future strategies for combating PM2.5-related adverse health effects. The insights gained can provide critical guidelines for preventing pollution-related CVDs through governmental, societal, and personal measures, thereby benefitting humanity and slowing climate change.
Collapse
Affiliation(s)
- Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (S.B.); (T.H.S.); (C.B.P.)
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (S.B.); (T.H.S.); (C.B.P.)
| | - Chan Bae Park
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (S.B.); (T.H.S.); (C.B.P.)
| | - Wang-Soo Lee
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Chung-Ang University, Seoul 06973, Korea;
| | - Jaetaek Kim
- Department of Internal Medicine, Division of Endocrinology and Metabolism, College of Medicine, Chung-Ang University, Seoul 06973, Korea
- Correspondence: (J.K.); (G.L.)
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (S.B.); (T.H.S.); (C.B.P.)
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: (J.K.); (G.L.)
| |
Collapse
|
31
|
Huang D, Shi S, Wang Y, Wang X, Shen Z, Wang M, Pei C, Wu Y, He Y, Wang Z. Astragaloside IV alleviates PM2.5-caused lung toxicity by inhibiting inflammasome-mediated pyroptosis via NLRP3/caspase-1 axis inhibition in mice. Biomed Pharmacother 2022; 150:112978. [PMID: 35462332 DOI: 10.1016/j.biopha.2022.112978] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Exposure to particulate matter (PM)2.5 in air pollution is a serious health issue worldwide. At present, effective prevention measures and modalities of treatment for PM2.5-caused lung toxicity are lacking. This study elucidated the protective effect of astragaloside IV (Ast), a natural product from Astragalus membranaceous Bunge, against PM2.5-caused lung toxicity and its possible molecular mechanisms. The mice model of lung toxicity was performed by intratracheal instillation of PM2.5 dust suspension. The investigation was performed with Ast or in combination with nigericin, which is a NOD-like receptor protein 3 (NLRP3) activator. The results revealed that PM2.5 lead significant lung inflammation and promoted the pyroptosis pattern of cell death by upregulating pro-inflammatory cytokines and causing oxidative stress related to the NLRP3 inflammasome-mediated pyroptosis pathway. Ast protected against PM2.5 resulted lung toxicity via suppressing NLRP3 inflammasome-mediated pyroptosis via NLRP3/caspase-1 axis inhibition, thereby protecting the lung against PM2.5-induced lung inflammation and oxidative damage, eventually resulting in prolonged survival in mice. Nigericin partially reversed the protective effects of Ast. The present research provides new insights into the therapeutic potential of Ast, demonstrating that it might be a possible candidate for the prevention of PM2.5-caused respiratory diseases. Targeting the NLRP3 inflammasome might be a novel therapeutic tactic for PM2.5-caused respiratory diseases.
Collapse
Affiliation(s)
- Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Mingjie Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yongcan Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
32
|
Jiang H, Zhou Y, Nabavi SM, Sahebkar A, Little PJ, Xu S, Weng J, Ge J. Mechanisms of Oxidized LDL-Mediated Endothelial Dysfunction and Its Consequences for the Development of Atherosclerosis. Front Cardiovasc Med 2022; 9:925923. [PMID: 35722128 PMCID: PMC9199460 DOI: 10.3389/fcvm.2022.925923] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/13/2022] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis is an immuno-metabolic disease involving chronic inflammation, oxidative stress, epigenetics, and metabolic dysfunction. There is compelling evidence suggesting numerous modifications including the change of the size, density, and biochemical properties in the low-density lipoprotein (LDL) within the vascular wall. These modifications of LDL, in addition to LDL transcytosis and retention, contribute to the initiation, development and clinical consequences of atherosclerosis. Among different atherogenic modifications of LDL, oxidation represents a primary modification. A series of pathophysiological changes caused by oxidized LDL (oxLDL) enhance the formation of foam cells and atherosclerotic plaques. OxLDL also promotes the development of fatty streaks and atherogenesis through induction of endothelial dysfunction, formation of foam cells, monocyte chemotaxis, proliferation and migration of SMCs, and platelet activation, which culminate in plaque instability and ultimately rupture. This article provides a concise review of the formation of oxLDL, enzymes mediating LDL oxidation, and the receptors and pro-atherogenic signaling pathways of oxLDL in vascular cells. The review also explores how oxLDL functions in different stages of endothelial dysfunction and atherosclerosis. Future targeted pathways and therapies aiming at reducing LDL oxidation and/or lowering oxLDL levels and oxLDL-mediated pro-inflammatory responses are also discussed.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yongwen Zhou
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peter J. Little
- School of Health and Behavioural Sciences, Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Suowen Xu ; orcid.org/0000-0002-5488-5217
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Jianping Weng ; orcid.org/0000-0002-7889-1697
| | - Jianjun Ge
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Jianjun Ge ; orcid.org/0000-0002-9424-6049
| |
Collapse
|
33
|
Zeng X, Liu D, Wu W, Huo X. PM 2.5 exposure inducing ATP alteration links with NLRP3 inflammasome activation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:24445-24456. [PMID: 35064883 PMCID: PMC8783591 DOI: 10.1007/s11356-021-16405-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/04/2021] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) has been the primary air pollutant and the fourth leading risk factor for disease and death in the world. Exposure to PM2.5 is related to activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, but the mechanism of PM2.5 affecting the NLRP3 inflammasome is still unclear. Previous studies have shown that PM2.5 can cause alterations in adenosine triphosphate (ATP), and an increase in extracellular ATP and a decrease in intracellular ATP can trigger the activation process of the NLRP3 inflammasome. Therefore, we emphasize that ATP changes may be the central link and key mechanism of PM2.5 exposure that activates the NLRP3 inflammasome. This review briefly elucidates and summarizes how PM2.5 acts on ATP and subsequently further impacts the NLRP3 inflammasome. Investigation of ATP changes due to exposure to PM2.5 may be essential to regulate NLRP3 inflammasome activation and treat inflammation-related diseases such as coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| | - Dongling Liu
- Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Weidong Wu
- Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
34
|
Wang W, Zhang J, Li Z, Gu J, Qin J, Li J, Zhang X, Ru S. Bisphenol S exposure accelerates the progression of atherosclerosis in zebrafish embryo-larvae. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128042. [PMID: 34942454 DOI: 10.1016/j.jhazmat.2021.128042] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
Bisphenol S (BPS), widely utilized in manufacturing of daily necessities, is a toxicant with potential to induce atherosclerotic cardiovascular disease (ASCVD). However, the mode of action by which BPS exposure induces ASCVD remains unknown. Here, macrophages that were exposed to BPS in combination with oxidized low-density lipoprotein (oxLDL) exhibited enhanced formation of foam cells, a hallmark of ASCVD. Furthermore, zebrafish embryo-larvae were exposed to BPS (0, 1, 10 and 100 μg/L) for 15 days (d) and the characteristic symptoms of ASCVD including an inflammatory response, macrophage recruitment around blood vessels, and accumulation of oxLDL on vascular endothelium, were induced in 15-d larvae. After zebrafish were exposed to BPS for 45 d, BPS mobilized fatty acid metabolism and activated peroxisome proliferator-activated receptor signaling in larval liver, the hub of endogenous lipid metabolism, causing an increase in plasma LDL. Driven by high plasma LDL levels, the caudal artery of zebrafish larvae exhibited lipid accumulation and a thickened area with a large number of collagen fibers, accompanied by characteristic lesions, as well as hyperlipidemia, erythrocyte aggregation, thinner blood vessel walls and increased levels of leukocytes and thromboocytes in plasma. Our data demonstrate that BPS accelerates the progression of ASCVD using zebrafish embryo-larvae as a model.
Collapse
Affiliation(s)
- Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Zhang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Gu
- Nanjing Institute of Environmental Sciences, Nanjing 210000, China
| | - Jingyu Qin
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xiaona Zhang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| |
Collapse
|