1
|
Wang SN, Shi YC, Lin S, He HF. Particulate matter 2.5 accelerates aging: Exploring cellular senescence and age-related diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116920. [PMID: 39208581 DOI: 10.1016/j.ecoenv.2024.116920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Exposure to Particulate matter 2.5 (PM2.5) accelerates aging, causing declines in tissue and organ function, and leading to diseases such as cardiovascular, neurodegenerative, and musculoskeletal disorders. PM2.5 is a major environmental pollutant and an exogenous pathogen in air pollution that is now recognized as an accelerator of human aging and a predisposing factor for several age-related diseases. In this paper, we seek to elucidate the mechanisms by which PM2.5 induces cellular senescence, such as genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, and mitochondrial dysfunction, and age-related diseases. Our goal is to increase awareness among researchers within the field of the toxicity of environmental pollutants and to advocate for personal and public health initiatives to curb their production and enhance population protection. Through these endeavors, we aim to promote longevity and health in older adults.
Collapse
Affiliation(s)
- Sheng-Nan Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yan-Chuan Shi
- Centre of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Australia
| | - Shu Lin
- Centre of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - He-Fan He
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| |
Collapse
|
2
|
Hu R, Fan W, Li S, Zhang G, Zang L, Qin L, Li R, Chen R, Zhang L, Gu W, Zhang Y, Rajagopalan S, Sun Q, Liu C. PM 2.5-induced cellular senescence drives brown adipose tissue impairment in middle-aged mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116423. [PMID: 38705039 DOI: 10.1016/j.ecoenv.2024.116423] [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: 03/05/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Airborne fine particulate matter (PM2.5) exposure is closely associated with metabolic disturbance, in which brown adipose tissue (BAT) is one of the main contributing organs. However, knowledge of the phenotype and mechanism of PM2.5 exposure-impaired BAT is quite limited. In the study, male C57BL/6 mice at three different life phases (young, adult, and middle-aged) were simultaneously exposed to concentrated ambient PM2.5 or filtered air for 8 weeks using a whole-body inhalational exposure system. H&E staining and high-resolution respirometry were used to assess the size of adipocytes and mitochondrial function. Transcriptomics was performed to determine the differentially expressed genes in BAT. Quantitative RT-PCR, immunohistochemistry staining, and immunoblots were performed to verify the transcriptomics and explore the mechanism for BAT mitochondrial dysfunction. Firstly, PM2.5 exposure caused altered BAT morphology and mitochondrial dysfunction in middle-aged but not young or adult mice. Furthermore, PM2.5 exposure increased cellular senescence in BAT of middle-aged mice, accompanied by cell cycle arrest, impaired DNA replication, and inhibited AKT signaling pathway. Moreover, PM2.5 exposure disrupted apoptosis and autophagy homeostasis in BAT of middle-aged mice. Therefore, BAT in middle-aged mice was more vulnerable to PM2.5 exposure, and the cellular senescence-initiated apoptosis, autophagy, and mitochondrial dysfunction may be the mechanism of PM2.5 exposure-induced BAT impairment.
Collapse
Affiliation(s)
- Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200433, China
| | - Sanjay Rajagopalan
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China.
| |
Collapse
|
3
|
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
|
4
|
Yan Q, Zheng R, Li Y, Hu J, Gong M, Lin M, Xu X, Wu J, Sun S. PM 2.5-induced premature senescence in HUVECs through the SIRT1/PGC-1α/SIRT3 pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 921:171177. [PMID: 38402989 DOI: 10.1016/j.scitotenv.2024.171177] [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: 11/21/2023] [Revised: 02/08/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Vascular endothelial cell senescence plays a pivotal role in the development of atherosclerosis. Recent studies have demonstrated that ambient fine particulate matter (PM2.5) induces stress-induced premature senescence (SIPS) in vascular endothelial cells. However, the precise mechanisms underlying this process remain to be fully elucidated. Cellular senescence is closely associated with reactive oxygen species (ROS), and emerging research has established a strong connection between the SIRT1/PGC-1α/SIRT3 signaling pathway and the antioxidant system in vascular endothelial cells. In this study, we aimed to investigate the impact of PM2.5 on vascular endothelial cell senescence and to elucidate the underlying mechanisms. Our findings revealed that PM2.5 exposure led to an increase in senescence-associated β-galactosidase (SA-β-gal) activity and the expression of the cell cycle-blocking proteins P53/P21 and P16 in human umbilical vein endothelial cells (HUVECs). Flow cytometry analysis demonstrated an elevated proportion of cells arrested in the G0/G1 phase after PM2.5 exposure. In addition, PM2.5-induced cellular senescence was attributed to the disruption of the cellular antioxidative defense system through the SIRT1/PGC-1α/SIRT3 signaling pathway. The expression of cellular senescence markers was reduced after targeted scavenging of mitochondrial ROS using MitoQ. Moreover, treatment with SRT1720, a SIRT1-specific activator, upregulated the SIRT1/PGC-1α/SIRT3 signaling pathway, restored the antioxidant system, and attenuated the expression of cellular senescence markers. Taken together, our results suggest that PM2.5 downregulates the SIRT1/PGC-1α/SIRT3 signaling pathway, resulting in impaired antioxidant defenses in HUVECs. This, in turn, allows for the accumulation of ROS, leading to inhibition of endothelial cell cycle progression and the onset of stress-induced senescence in HUVECs.
Collapse
Affiliation(s)
- Qing Yan
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Rao Zheng
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Yi Li
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Juan Hu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Meidi Gong
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Manman Lin
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xuecong Xu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jing Wu
- Department of Toxicology, School of Public Health, Medical College of Soochow University, Suzhou 215123, Jiangsu, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China.
| | - Shikun Sun
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
5
|
Peno-Mazzarino L, Radionov N, Merino M, González S, Mullor JL, Jones J, Caturla N. Protective Potential of a Botanical-Based Supplement Ingredient against the Impact of Environmental Pollution on Cutaneous and Cardiopulmonary Systems: Preclinical Study. Curr Issues Mol Biol 2024; 46:1530-1555. [PMID: 38392217 PMCID: PMC10887869 DOI: 10.3390/cimb46020099] [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: 01/18/2024] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024] Open
Abstract
Air pollution is a growing threat to human health. Airborne pollution effects on respiratory, cardiovascular and skin health are well-established. The main mechanisms of air-pollution-induced health effects involve oxidative stress and inflammation. The present study evaluates the potential of a polyphenol-enriched food supplement ingredient comprising Lippia citriodora, Olea europaea, Rosmarinus officinalis, and Sophora japonica extracts in mitigating the adverse effects of environmental pollution on skin and cardiopulmonary systems. Both in vitro and ex vivo studies were used to assess the blend's effects against pollution-induced damage. In these studies, the botanical blend was found to reduce lipid peroxidation, inflammation (by reducing IL-1α), and metabolic alterations (by regulating MT-1H, AhR, and Nrf2 expression) in human skin explants exposed to a mixture of pollutants. Similar results were also observed in keratinocytes exposed to urban dust. Moreover, the ingredient significantly reduced pollutant-induced ROS production in human endothelial cells and lung fibroblasts, while downregulating the expression of apoptotic genes (bcl-2 and bax) in lung fibroblasts. Additionally, the blend counteracted the effect of urban dust on the heart rate in zebrafish embryos. These results support the potential use of this supplement as an adjuvant method to reduce the impact of environmental pollution on the skin, lungs, and cardiovascular tissues.
Collapse
Affiliation(s)
| | - Nikita Radionov
- Laboratoire BIO-EC, Chemin de Saulxier 1, 91160 Longjumeau, France
| | - Marián Merino
- Bionos Biotech, S.L. Biopolo La Fe, Av. Fernando Abril Martorell, 106, 46026 Valencia, Spain
| | - Sonia González
- Bionos Biotech, S.L. Biopolo La Fe, Av. Fernando Abril Martorell, 106, 46026 Valencia, Spain
| | - José L Mullor
- Bionos Biotech, S.L. Biopolo La Fe, Av. Fernando Abril Martorell, 106, 46026 Valencia, Spain
| | | | - Nuria Caturla
- Monteloeder SA, Miguel Servet 16, 03203 Elche, Spain
| |
Collapse
|
6
|
Xue Y, Tran M, Diep YN, Shin S, Lee J, Cho H, Kang YJ. Environmental aluminum oxide inducing neurodegeneration in human neurovascular unit with immunity. Sci Rep 2024; 14:744. [PMID: 38185738 PMCID: PMC10772095 DOI: 10.1038/s41598-024-51206-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024] Open
Abstract
Aluminum oxide nanoparticle (AlNP), a ubiquitous neurotoxin highly enriched in air pollution, is often produced as an inevitable byproduct in the manufacturing of industrial products such as cosmetics and metal materials. Meanwhile, ALNP has emerged as a significant public health concern due to its potential association with neurological diseases. However, the studies about the neurotoxic effects of AlNP are limited, partially due to the lack of physiologically relevant human neurovascular unit with innate immunity (hNVUI). Here, we employed our AlNP-treated hNVUI model to investigate the underlying mechanism of AlNP-driven neurodegeneration. First, we validated the penetration of AlNP across a blood-brain barrier (BBB) compartment and found AlNP-derived endothelial cellular senescence through the p16 and p53/p21 pathways. Our study showed that BBB-penetrating AlNP promoted reactive astrocytes, which produced a significant level of reactive oxygen species (ROS). The astrocytic neurotoxic factors caused neuronal damage, including the synaptic impairment, the accumulation of phosphoric-tau proteins, and even neuronal death. Our study suggests that AlNP could be a potential environmental risk factor of neurological disorders mediated by neuroinflammation.
Collapse
Affiliation(s)
- Yingqi Xue
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seonghun Shin
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jinkee Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| | - You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
7
|
Shiwakoti S, Gong D, Sharma K, Kang KW, Schini-Kerth VB, Kim HJ, Ko JY, Oak MH. γ-Oryzanol ameliorates fine dust-induced premature endothelial senescence and dysfunction via attenuating oxidative stress. Food Chem Toxicol 2023; 179:113981. [PMID: 37549806 DOI: 10.1016/j.fct.2023.113981] [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: 05/11/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Various cardiovascular diseases are associated with endothelial senescence, and a recent study showed that fine dust (FD)-induced premature endothelial senescence and dysfunction is associated with increased oxidative stress. The aim of the present study was to investigate protective effect of rice bran extract (RBE) and its major component of γ-Oryzanol (γ-Ory) against FD-induced premature endothelial senescence. Porcine coronary artery endothelial cells (PCAECs) were treated with FD alone or with RBE or γ-Ory. Senescence-associated β-galactosidase (SA-β-gal) activity, expression of cell cycle regulatory proteins, and oxidative stress levels were evaluated. The results indicated that SA-β-gal activity in the FD-treated PCAECs was attenuated by RBE and γ-Ory. Additionally, γ-Ory inhibited FD-induced cell cycle arrest, restored cell proliferation, and reduced the expression of cell cycle regulatory proteins. γ-Ory also inhibited oxidative stress and prevented senescence-associated NADPH oxidase and LAS activity in FD-exposed ECs suggesting that γ-Ory could protect against FD-induced ECs senescence and dysfunction.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Dalseong Gong
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea; Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Kushal Sharma
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ki-Woon Kang
- Division of Cardiology, College of Medicine, Heart Reasearch Institute and Biomedical Research Institute, Chung-Ang University Hospital, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Hyun Jung Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| |
Collapse
|
8
|
Dhakal B, Shiwakoti S, Park EY, Kang KW, Schini-Kerth VB, Park SH, Ji HY, Park JS, Ko JY, Oak MH. SGLT2 inhibition ameliorates nano plastics-induced premature endothelial senescence and dysfunction. Sci Rep 2023; 13:6256. [PMID: 37069192 PMCID: PMC10110533 DOI: 10.1038/s41598-023-33086-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/06/2023] [Indexed: 04/19/2023] Open
Abstract
Nano plastics (NPs) have been a significant threat to human health and are known to cause premature endothelial senescence. Endothelial senescence is considered one of the primary risk factors contributing to numerous cardiovascular disorders. Recent studies have suggested that inhibition of sodium glucose co-transporter (SGLT2) ameliorates endothelial senescence and dysfunction. Therefore, our study intends to explore the role of SGLT2 in NPs-induced endothelial senescence and dysfunction. Porcine coronary artery and its endothelial cells were treated with NPs in the presence or absence of Enavogliflozin (ENA), a SGLT2 inhibitor and then SGLTs expression, senescence markers and vascular function were evaluated. NPs significantly up-regulated SGLT2 and ENA significantly decreased NPs-induced senescence-associated-β-gal activity, cell-cycle arrest, and senescence markers p53 and p21 suggesting that inhibition of SGLT2 prevents NPs-induced endothelial senescence. In addition, ENA decreased the formation of reactive oxygen species with the downregulation of Nox2, and p22phox. Furthermore, SGLT2 inhibition also up regulated the endothelial nitric oxide synthase expression along with improving vascular function. In conclusion, premature endothelial senescence by NPs is, at least in part, associated with SGLT2 and it could be a potential therapeutic target for preventing and/or treating environmental pollutants-induced cardiovascular disorders mediated by endothelial senescence and dysfunction.
Collapse
Affiliation(s)
- Bikalpa Dhakal
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-ro, Cheonggye-Myeonn, Muan-Gun, Jeonnam, 58554, Republic of Korea
| | - Saugat Shiwakoti
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-ro, Cheonggye-Myeonn, Muan-Gun, Jeonnam, 58554, Republic of Korea
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-ro, Cheonggye-Myeonn, Muan-Gun, Jeonnam, 58554, Republic of Korea
| | - Ki-Woon Kang
- Division of Cardiology, College of Medicine, Heart Reasearch Institute and Biomedical Research Institute, Chung-Ang University Hospital, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260 INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Sun-Hwa Park
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, 17028, Republic of Korea
| | - Hye-Young Ji
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, 17028, Republic of Korea
| | - Joon Seok Park
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, 17028, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-ro, Cheonggye-Myeonn, Muan-Gun, Jeonnam, 58554, Republic of Korea.
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-ro, Cheonggye-Myeonn, Muan-Gun, Jeonnam, 58554, Republic of Korea.
| |
Collapse
|
9
|
Afsar B, Afsar RE. Hypertension and cellular senescence. Biogerontology 2023:10.1007/s10522-023-10031-4. [PMID: 37010665 DOI: 10.1007/s10522-023-10031-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
Essential or primary hypertension is a wordwide health problem. Elevated blood pressure (BP) is closely associated not only with increased chronological aging but also with biological aging. There are various common pathways that play a role in cellular aging and BP regulation. These include but not limited to inflammation, oxidative stress, mitochondrial dysfunction, air pollution, decreased klotho activity increased renin angiotensin system activation, gut dysbiosis etc. It has already been shown that some anti-hypertensive drugs have anti-senescent actions and some senolytic drugs have BP lowering effects. In this review, we have summarized the common mechanisms underlying cellular senescence and HT and their relationships. We further reviewed the effect of various antihypertensive medications on cellular senescence and suggest further issues to be studied.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
10
|
Olesiejuk K, Chałubiński M. How does particulate air pollution affect barrier functions and inflammatory activity of lung vascular endothelium? Allergy 2023; 78:629-638. [PMID: 36588285 DOI: 10.1111/all.15630] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/03/2023]
Abstract
Both particulate matter and gaseous components of air pollution have already been shown to increase cardiovascular mortality in numerous studies. It is, however, important to note that on their way to the bloodstream the polluting agents pass the lung barrier. Inside the alveoli, particles of approximately 0.4-1 μm are most efficiently deposited and commonly undergo phagocytosis by lung macrophages. Not only the soluble agents, but also particles fine enough to leave the alveoli enter the bloodstream in this finite part of the endothelium, reaching thus higher concentrations in close proximity of the alveoli and endothelium. Additionally, deposits of particulate matter linger in direct proximity of the endothelial cells and may induce inflammation, immune responses, and influence endothelial barrier dysfunction thus increasing PM bioavailability in positive feedback. The presented discussion provides an overview of possible components of indoor PM and how endothelium is thus influenced, with emphasis on lung vascular endothelium and clinical perspectives.
Collapse
Affiliation(s)
- Krzysztof Olesiejuk
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Lodz, Poland
| | - Maciej Chałubiński
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Fossel M, Bean J, Khera N, Kolonin MG. A Unified Model of Age-Related Cardiovascular Disease. BIOLOGY 2022; 11:1768. [PMID: 36552277 PMCID: PMC9775230 DOI: 10.3390/biology11121768] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/18/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Despite progress in biomedical technologies, cardiovascular disease remains the main cause of mortality. This is at least in part because current clinical interventions do not adequately take into account aging as a driver and are hence aimed at suboptimal targets. To achieve progress, consideration needs to be given to the role of cell aging in disease pathogenesis. We propose a model unifying the fundamental processes underlying most age-associated cardiovascular pathologies. According to this model, cell aging, leading to cell senescence, is responsible for tissue changes leading to age-related cardiovascular disease. This process, occurring due to telomerase inactivation and telomere attrition, affects all components of the cardiovascular system, including cardiomyocytes, vascular endothelial cells, smooth muscle cells, cardiac fibroblasts, and immune cells. The unified model offers insights into the relationship between upstream risk factors and downstream clinical outcomes and explains why interventions aimed at either of these components have limited success. Potential therapeutic approaches are considered based on this model. Because telomerase activity can prevent and reverse cell senescence, telomerase gene therapy is discussed as a promising intervention. Telomerase gene therapy and similar systems interventions based on the unified model are expected to be transformational in cardiovascular medicine.
Collapse
Affiliation(s)
| | - Joe Bean
- University of Missouri School of Medicine, Kansas City, MO 65211, USA
| | - Nina Khera
- Buckingham Browne and Nichols School, Wellesley, MA 02138, USA
| | - Mikhail G. Kolonin
- University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
12
|
Al-Azab M, Safi M, Idiiatullina E, Al-Shaebi F, Zaky MY. Aging of mesenchymal stem cell: machinery, markers, and strategies of fighting. Cell Mol Biol Lett 2022; 27:69. [PMID: 35986247 PMCID: PMC9388978 DOI: 10.1186/s11658-022-00366-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are primary multipotent cells capable of differentiating into osteocytes, chondrocytes, and adipocytes when stimulated under appropriate conditions. The role of MSCs in tissue homeostasis, aging-related diseases, and cellular therapy is clinically suggested. As aging is a universal problem that has large socioeconomic effects, an improved understanding of the concepts of aging can direct public policies that reduce its adverse impacts on the healthcare system and humanity. Several studies of aging have been carried out over several years to understand the phenomenon and different factors affecting human aging. A reduced ability of adult stem cell populations to reproduce and regenerate is one of the main contributors to the human aging process. In this context, MSCs senescence is a major challenge in front of cellular therapy advancement. Many factors, ranging from genetic and metabolic pathways to extrinsic factors through various cellular signaling pathways, are involved in regulating the mechanism of MSC senescence. To better understand and reverse cellular senescence, this review highlights the underlying mechanisms and signs of MSC cellular senescence, and discusses the strategies to combat aging and cellular senescence.
Collapse
|
13
|
The macrophage senescence hypothesis: the role of poor heat shock response in pulmonary inflammation and endothelial dysfunction following chronic exposure to air pollution. Inflamm Res 2022; 71:1433-1448. [PMID: 36264363 DOI: 10.1007/s00011-022-01647-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/18/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Cardiovascular diseases (CVD) have been associated with high exposure to fine particulate air pollutants (PM2.5). Alveolar macrophages are the first defense against inhaled particles. As soon as they phagocytize the particles, they reach an inflammatory phenotype, which affects the surrounding cells and associates with CVD. Not coincidentally, CVD are marked by a depleted heat shock response (HSR), defined by a deficit in inducing 70-kDa heat shock protein (HSP70) expression during stressful conditions. HSP70 is a powerful anti-inflammatory chaperone, whose reduced levels trigger a pro-inflammatory milieu, cellular senescence, and a senescence-associated secretory phenotype (SASP). However, whether macrophage senescence is the main mechanism by which PM2.5 propagates low-grade inflammation remains unclear. OBJECTIVE AND DESIGN In this article, we review evidence supporting that chronic exposure to PM2.5 depletes HSR and determines the ability to solve the initial stress. RESULTS AND DISCUSSION When exposed to PM2.5, macrophages increase the production of reactive oxygen species, which activate nuclear factor-kappa B (NF-κB). NF-κB is naturally a pro-inflammatory factor that drives prostaglandin E2 (PGE2) synthesis and causes fever. PGE2 can be converted into prostaglandin A2, a powerful inducer of HSR. Therefore, when transiently activated, NF-κB can trigger the anti-inflammatory response through negative feedback, by inducing HSP70 expression. However, when chronically activated, NF-κB heads a set of pathways involved in mitochondrial dysfunction, endoplasmic reticulum stress, unfolded protein response, inflammasome activation, and apoptosis. During chronic exposure to PM2.5, cells cannot properly express sirtuin-1 or activate heat shock factor-1 (HSF-1), which delays the resolution phase of inflammation. Since alveolar macrophages are the first immune defense against PM2.5, we suppose that the pollutant impairs HSR and, consequently, induces cellular senescence. Accordingly, senescent macrophages change its secretory phenotype to a more inflammatory one, known as SASP. Finally, macrophages' SASP would propagate the systemic inflammation, leading to endothelial dysfunction and atherosclerosis.
Collapse
|
14
|
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: 16] [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
|
15
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
16
|
Shiwakoti S, Ko JY, Gong D, Dhakal B, Lee JH, Adhikari R, Gwak Y, Park SH, Jun Choi I, Schini-Kerth VB, Kang KW, Oak MH. Effects of polystyrene nanoplastics on endothelium senescence and its underlying mechanism. ENVIRONMENT INTERNATIONAL 2022; 164:107248. [PMID: 35461096 DOI: 10.1016/j.envint.2022.107248] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/03/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Global plastic use has increased rapidly, and environmental pollution associated with nanoplastics (NPs) has been a growing concern recently. However, the impact and biological mechanism of NPs on the cardiovascular system are not well characterized. This study aimed to assess the possibility that NPs exposure promotes premature endothelial cell (EC) senescence in porcine coronary artery ECs and, if so, to elucidate the underlying mechanism. Treatment of ECs with NPs promoted the acquisition of senescence markers, senescence-associated β-galactosidase activity, and p53, p21, and p16 protein expression, resulting in the inhibition of proliferation. In addition, NPs impaired endothelium-dependent vasorelaxation associated with decreased endothelial nitric oxide synthase (eNOS) expression. NPs enhanced reactive oxygen species formation in ECs, and increased oxidative stress levels were associated with the induction of NADPH oxidases expression, followed by the subsequent downregulation of Sirt1 expression. The characteristics of EC senescence and dysfunction caused by NPs are prevented by an antioxidant (N-acetylcysteine), an NADPH oxidase inhibitor (apocynin), and a Sirt1 activator (resveratrol). These findings indicate that NPs induced premature EC senescence, at least in part, through the redox-sensitive eNOS/Sirt1 signaling pathway. This study suggested the effects and underlying mechanism of NPs on the cardiovascular system, which may provide pharmacological targets to prevent NPs-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Dalseong Gong
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea; Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260 INSERM (French National Institute of Health and Medical Research), University of Strasbourg, Strasbourg, France
| | - Bikalpa Dhakal
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Jeong-Hye Lee
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Radhika Adhikari
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Yeonhyang Gwak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Sin-Hee Park
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ik Jun Choi
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260 INSERM (French National Institute of Health and Medical Research), University of Strasbourg, Strasbourg, France
| | - Ki-Woon Kang
- Division of Cardiology, Cardiovascular and Arrhythmia Center, Chung-Ang University, Seoul, Republic of Korea
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea.
| |
Collapse
|
17
|
Xu Y, Bu H, Jiang Y, Zhuo X, Hu K, Si Z, Chen Y, Liu Q, Gong X, Sun H, Zhu Q, Cui L, Ma X, Cui Y. N‑acetyl cysteine prevents ambient fine particulate matter‑potentiated atherosclerosis via inhibition of reactive oxygen species‑induced oxidized low density lipoprotein elevation and decreased circulating endothelial progenitor cell. Mol Med Rep 2022; 26:236. [PMID: 35621139 PMCID: PMC9185698 DOI: 10.3892/mmr.2022.12752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/09/2022] [Indexed: 11/06/2022] Open
Abstract
Ambient fine particulate matter (PM) serves an important role in the development of cardiovascular disease, including atherosclerosis. Antioxidant N‑acetyl cysteine (NAC) has protective effects in the cardiovascular system. However, it is unknown if NAC prevents PM‑potentiated atherosclerosis in hyperlipidemia. Low‑density lipoprotein (LDL) receptor knockout mice were pretreated with 1 mg/ml NAC in drinking water for 1 week and continued to receive NAC, high‑fat diet and intranasal instillation of PM for 1 week or 6 months. Blood plasma was collected for lipid profile, oxidized (ox‑)LDL, blood reactive oxygen species (ROS) and inflammatory cytokine (TNF‑α, IL‑1β and IL‑6) measurement. Blood cells were harvested for endothelial progenitor cell (EPC) population and intracellular ROS analysis. Murine aorta was isolated for atherosclerotic plaque ratio calculation. NAC treatment maintained circulating EPC level and significantly decreased blood ox‑LDL and ROS, inflammatory cytokines, mononuclear and EPC intracellular ROS levels as well as aortic plaque ratio. NAC prevented PM‑potentiated atherosclerosis by inhibiting plasma ROS‑induced ox‑LDL elevation, mononuclear cell and EPC intracellular ROS‑induced circulating EPC reduction and inflammatory cytokine production.
Collapse
Affiliation(s)
- Yixin Xu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Haoran Bu
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yufan Jiang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaoqing Zhuo
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ke Hu
- Department of Emergency, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| | - Zhihua Si
- Department of Emergency, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, P.R. China
| | - Yong Chen
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qiwei Liu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xianwei Gong
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Haihui Sun
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Qingyi Zhu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaochun Ma
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Yuqi Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
18
|
Zhang Y, Yu G, Jin R, Zhang Y, Dong K, Cheng T, Wang B. Water vapor distribution and particle condensation growth in turbulent pipe flow. POWDER TECHNOL 2022. [DOI: 10.1016/j.powtec.2022.117401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Seong EH, Gong DS, Shiwakoti S, Adhikari D, Kim HJ, Oak MH. Taxifolin as a Major Bioactive Compound in the Vasorelaxant Effect of Different Pigmented Rice Bran Extracts. Front Pharmacol 2022; 13:799064. [PMID: 35387354 PMCID: PMC8979019 DOI: 10.3389/fphar.2022.799064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality in recent years. The intake of polyphenol rich diets has been associated with improved cardiovascular function and reduced cardiovascular risks. Oryza sativa L. is one of the most common cereals worldwide. Rice bran, a byproduct of the rice milling process, contains many bioactive ingredients, including polyphenols, polysaccharides, proteins, and micronutrients. It is also consumed as a healthy diet in the form of rice bran oil and powder in many Asian countries like Japan, South Korea, and India for its several health benefits as a natural antioxidant. Thus, this study evaluated the vasorelaxant effect of ethanolic extracts of brown, green, red, and black rice bran and investigated its underlying vasorelaxant mechanism. Among the four rice bran extracts (RBEs) examined, the red rice bran extract (RRBE) had a strong endothelium-dependent vasorelaxant effect, which was markedly prevented by N-ω-nitro-L-arginine [endothelial nitric oxide synthase (eNOS) inhibitor], wortmannin [phosphoinositide-3 kinase (PI3K) inhibitor], and 1H-[1,2,4]oxadiazole[4,3-alpha]quinoxalin-1-one (inhibitor of guanylate cyclase). Likewise, RRBE induced the phosphorylation of eNOS and Src in cultured endothelial cells, thereby stimulating NO formation. Altogether, these findings propose that RRBE induces endothelium-dependent relaxation, involving at least in part, NO-mediated signaling through the PI3K/eNOS pathway. Further, LC-PDA analysis conducted on the four RBEs also revealed that RRBE highly contained taxifolin, which is an active flavanonol that induces endothelium-dependent vasorelaxation, compared to other RBEs. Subsequently, the underlying mechanism of taxifolin was assessed through vascular reactivity studies with pharmacological inhibitors similar to that of RRBE. These findings deciphered a distinct difference in vasorelaxant effects between RRBE and the other RBEs. We also observed that RRBE induced a potent endothelium-dependent NO-mediated relaxation in coronary artery rings, which involved the Src/PI3K pathway that activates eNOS. Additionally, taxifolin exhibited, at least in part, similar vasoprotective effects of RRBE. Therefore, we propose that RRBE may serve as natural sources of functional phytochemicals that improve cardiovascular diseases associated with disturbed NO production and endothelial dysfunction.
Collapse
Affiliation(s)
- Eun-Hee Seong
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| | - Dal-Seong Gong
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| | - Saugat Shiwakoti
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| | - Deepak Adhikari
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| | - Hyun Jung Kim
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, Mokpo, South Korea
| |
Collapse
|
20
|
Chang JH, Lee YL, Laiman V, Han CL, Jheng YT, Lee KY, Yeh CT, Kuo HP, Chung KF, Heriyanto DS, Hsiao TC, Wu SM, Ho SC, Chuang KJ, Chuang HC. Air pollution-regulated E-cadherin mediates contact inhibition of proliferation via the hippo signaling pathways in emphysema. Chem Biol Interact 2022; 351:109763. [PMID: 34852269 DOI: 10.1016/j.cbi.2021.109763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 01/07/2023]
Abstract
Air pollution has been linked to emphysema in chronic obstruction pulmonary disease (COPD). However, the underlying mechanisms in the development of emphysema due to air pollution remain unclear. The objective of this study was to investigate the role of components of the Hippo signaling pathway for E-cadherin-mediated contact inhibition of proliferation in the lungs after air pollution exposure. E-Cadherin-mediated contact inhibition of proliferation via the Hippo signaling pathway was investigated in Sprague-Dawley (SD) rats whole-body exposed to air pollution, and in alveolar epithelial A549 cells exposed to diesel exhaust particles (DEPs), E-cadherin-knockdown, and high-mobility group box 1 (HMGB1) treatment. Underlying epithelial differentiation, apoptosis, and senescence were also examined, and the interaction network among these proteins was examined. COPD lung sections were used to confirm the observations in rats. Expressions of HMGB1 and E-cadherin were negatively regulated in the lungs and A549 cells by air pollution, and this was confirmed by knockdown of E-cadherin and by treating A549 cells with HMGB1. Depletion of phosphorylated (p)-Yap occurred after exposure to air pollution and E-cadherin-knockdown, which resulted in decreases of SPC and T1α. Exposure to air pollution and E-cadherin-knockdown respectively downregulated p-Sirt1 and increased p53 levels in the lungs and in A549 cells. Moreover, the protein interaction network suggested that E-cadherin is a key activator in regulating Sirt1 and p53, as well as alveolar epithelial cell differentiation by SPC and T1α. Consistently, downregulation of E-cadherin, p-Yap, SPC, and T1α was observed in COPD alveolar regions with particulate matter (PM) deposition. In conclusion, our results indicated that E-cadherin-mediated cell-cell contact directly regulates the Hippo signaling pathway to control differentiation, cell proliferation, and senescence due to air pollution. Exposure to air pollution may initiate emphysema in COPD patients.
Collapse
Affiliation(s)
- Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Vincent Laiman
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Chia-Li Han
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Teng Jheng
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Han-Pin Kuo
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
21
|
The cardiovascular effects of air pollution: Prevention and reversal by pharmacological agents. Pharmacol Ther 2021; 232:107996. [PMID: 34571110 PMCID: PMC8941724 DOI: 10.1016/j.pharmthera.2021.107996] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
Air pollution is associated with staggering levels of cardiovascular morbidity and mortality. Airborne particulate matter (PM), in particular, has been associated with a wide range of detrimental cardiovascular effects, including impaired vascular function, raised blood pressure, alterations in cardiac rhythm, blood clotting disorders, coronary artery disease, and stroke. Considerable headway has been made in elucidating the biological processes underlying these associations, revealing a labyrinth of multiple interacting mechanistic pathways. Several studies have used pharmacological agents to prevent or reverse the cardiovascular effects of PM; an approach that not only has the advantages of elucidating mechanisms, but also potentially revealing therapeutic agents that could benefit individuals that are especially susceptible to the effects of air pollution. This review gathers investigations with pharmacological agents, offering insight into the biology of how PM, and other air pollutants, may cause cardiovascular morbidity.
Collapse
|
22
|
Hehua Z, Qing C, Yuhong Z. Association between ambient particulate matter exposure and platelet counts in adults: a retrospective cohort study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:31268-31275. [PMID: 33599925 DOI: 10.1007/s11356-021-12865-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/04/2021] [Indexed: 06/12/2023]
Abstract
Associations between ambient particulate matter exposure and platelet counts are inconsistent in previous studies, and study on the effect of long-term exposure especially in Asian populations is limited. We explored the associations between long-term PM2.5 (particulate matter < 2.5 μm) exposure and platelet counts using a prospective cohort study in Northeast China. We used a logistic regression model to analyze the effects of different PM2.5 increments and platelet count elevation. Mixed linear models were used to analyze the association between PM2.5 concentration and platelet counts. Interaction and sub-group analyses were also conducted. Results showed that every 1 μg/m3 increment of PM2.5 exposure was associated with 0.29% (95%CI: 0.25-0.32%) increase in platelet counts and 10% (95%CI: 8-12%) higher risk of platelet elevation. Effects of long-term PM2.5 exposure on platelet elevation were stronger in male participants, of Han ethnicity, and without diabetes. Long-term PM2.5 exposure would increase platelet counts in adults in Northeast area of China, which might add more evidence to the potential biological mechanisms responsible for the effect of air pollution exposure on cardiovascular disease.
Collapse
Affiliation(s)
- Zhang Hehua
- Clinical Research Center, Shengjing Hospital of China Medical University, Huaxiang Road No. 39, Tiexi District, Shenyang, China
| | - Chang Qing
- Clinical Research Center, Shengjing Hospital of China Medical University, Huaxiang Road No. 39, Tiexi District, Shenyang, China
| | - Zhao Yuhong
- Clinical Research Center, Department of Clinical Epidemiology, Shenjing Hospital of China Medical University, Sanhao Street, No. 36, Heping District, Shenyang, China.
| |
Collapse
|
23
|
Li A, Mei Y, Zhao M, Xu J, Li R, Zhao J, Zhou Q, Ge X, Xu Q. Associations between air pollutant exposure and renal function: A prospective study of older adults without chronic kidney disease. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 277:116750. [PMID: 33676338 DOI: 10.1016/j.envpol.2021.116750] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
We used real-world exposure scenarios to evaluate the effect of six ambient air pollutant (PM2.5, PM10, NO2, SO2, CO, and O3) exposure on renal function among older adults without chronic kidney disease (CKD). We recruited 169 older adults without CKD in Beijing, China, for a longitudinal study from 2016 to 2018. The Modification of Diet in Renal Disease (MDRD) and the Chronic Kidney Disease Epidemiology Collaboration (EPI) equations were employed to derive the estimated glomerular filtration rate (eGFR). A linear mixed-effects model with random intercepts for participants was employed to determine the effects of air pollutants on renal function evaluated on the basis of eGFR and urinary albumin/creatinine ratio at different exposure windows (1-, 2-, 3-, 5-, 7-, 14-, 28-, 45-, and 60-days moving averages). An interquartile range (IQR) increase in NO2 for was associated with significant decreases of in eGFR (MDRD equation) [percentage changes: -4.49 (95% confidence interval: -8.44, -0.37), -5.51 (-10.43, -0.33), -2.26 (-4.38, -0.08), -3.71 (-6.67, -0.65), -5.44 (-9.58, -1.11), -5.50 (-10.24, -0.51), -6.15 (-10.73, -1.33), and -6.34 (-11.17, -1.25) for 1-, 2-, 5-, 7-, 14-, 28-, 45-, and 60-days moving averages, respectively] and in eGFR (EPI equation) [percentage changes: -5.04 (-7.09, -2.94), -6.25 (-8.81, -3.62), -5.16 (-7.34, -2.92), -5.10 (-7.85, -2.28), -5.83 (-8.23, -3.36), -6.04 (-8.55, -3.47) for 1-, 2-, 14-, 28-, 45-, and 60-days moving averages, respectively]. In two-pollutant model, only the association of NO2 exposure with eGFR remained robust after adjustment for any other pollutant. This association was stronger for individuals with hypertension for the EPI equation or BMI <25 kg/m2 for the MDRD equation at lags 1 and 1-2. Our findings suggest that NO2 exposure is associated with eGFR reduction among older adults without CKD for short (1-, 2-days) and medium (14-, 28-, 45-, 60-days) term exposure periods; thus, NO2 exposure may contribute to renal impairment.
Collapse
Affiliation(s)
- Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Yayuan Mei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Runkui Li
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China; State Key Laboratory of Resources and Environmental Information System, Institute of Geographic Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaxin Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Quan Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Xiaoyu Ge
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
24
|
Costa-Beber LC, Goettems-Fiorin PB, Dos Santos JB, Friske PT, Frizzo MN, Heck TG, Hirsch GE, Ludwig MS. Ovariectomy enhances female rats' susceptibility to metabolic, oxidative, and heat shock response effects induced by a high-fat diet and fine particulate matter. Exp Gerontol 2020; 145:111215. [PMID: 33340683 DOI: 10.1016/j.exger.2020.111215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/20/2020] [Accepted: 12/12/2020] [Indexed: 01/21/2023]
Abstract
Obesity and exposure to fine particulate matter (air pollutant PM2.5) are important risk factors for metabolic and cardiovascular diseases. They are also related to early menopause. The reduction of 17β-estradiol (E2) levels during female climacteric, marked by menopause, is of significant concern because of its imminent influence on metabolism, redox and inflammatory status. This complex homeostasis-threatening scenario may induce a heat shock response (HSR) in cells, enhancing the expression of the 70 kDa heat shock protein (HSP70). A failure in this mechanism could predispose women to cardiovascular diseases. In this study, we evaluated if the climacteric could represent an additional risk among obese rats exposed to PM2.5 by worsening lipid, oxidative, and inflammatory parameters and HSP70 in cardiac tissue. We induced obesity in female Wistar rats using a high-fat diet (HFD) (58.3% as fats) and exposed them to 50 μL of saline 0.9% (control, n = 15) or 250 μg residual oil fly ash (ROFA, the inorganic portion of PM2.5) (polluted, n = 15) by intranasal instillation, 5 days/w for 12 weeks. At the 12th week, we subdivided these animals into four groups: control (n = 6), OVX (n = 9), polluted (n = 6) and polluted + OVX (n = 9). OVX and polluted + OVX were submitted to a bilateral ovariectomy (OVX), a surgical model for menopause, while control and polluted received a false surgery (sham). ROFA exposure and HFD consumption were continued for 12 additional weeks, after which the animals were euthanized. ROFA enhanced the susceptibility to ovariectomy-induced dyslipidemia, while ovariectomy predisposed female rats to the ROFA-induced decrease of cardiac iHSP70 expression. Ovariectomy also decreased the IL-6 levels and IL-6/IL-10 in obese animals, reinforcing a metabolic impairment and a failure to respond to unfavorable conditions. Our results support the hypothesis that obese ovariectomized animals are predisposed to a metabolic worsening under polluted conditions and are at higher risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Lílian Corrêa Costa-Beber
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil.
| | - Pauline Brendler Goettems-Fiorin
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Atmospheric Pollution Laboratory, Postgraduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245 Porto Alegre, RS, Brazil
| | - Jaíne Borges Dos Santos
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil
| | - Paula Taís Friske
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil
| | - Matias Nunes Frizzo
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Atmospheric Pollution Laboratory, Postgraduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245 Porto Alegre, RS, Brazil; Medicine Course, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Thiago Gomes Heck
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil; Medicine Course, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Gabriela Elisa Hirsch
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Mirna Stela Ludwig
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Rua do Comércio, 3000 - Bairro Universitário, Ijuí, RS, Brazil; Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil; Medicine Course, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| |
Collapse
|
25
|
Shiwakoti S, Adhikari D, Lee JP, Kang KW, Lee IS, Kim HJ, Oak MH. Prevention of Fine Dust-Induced Vascular Senescence by Humulus lupulus Extract and Its Major Bioactive Compounds. Antioxidants (Basel) 2020; 9:E1243. [PMID: 33297587 PMCID: PMC7762380 DOI: 10.3390/antiox9121243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022] Open
Abstract
Both short- and long-term exposure to fine dust (FD) from air pollution has been linked to various cardiovascular diseases (CVDs). Endothelial cell (EC) senescence is an important risk factor for CVDs, and recent evidence suggests that FD-induced premature EC senescence increases oxidative stress levels. Hop plant (Humulus lupulus) is a very rich source of polyphenols known to have nutritional and therapeutic properties, including antioxidant behavior. The aims of this study were to evaluate whether Humulus lupulus extract prevents FD-induced vascular senescence and dysfunction and, if so, to characterize the underlying mechanisms and active components. Porcine coronary arteries and endothelial cells were treated with FD in the presence or absence of hop extract (HOP), and the senescence-associated-beta galactosidase (SA-β-gal) activity, cell-cycle progression, expression of senescence markers, oxidative stress level, and vascular function were evaluated. Results indicated that HOP inhibited FD-induced SA-β-gal activity, cell-cycle arrest, and oxidative stress, suggesting that HOP prevents premature induction of senescence by FD. HOP also ameliorated FD-induced vascular dysfunction. Additionally, xanthohumol (XN) and isoxanthohumol (IX) were found to produce the protective effects of HOP. Treatment with HOP and its primary active components XN and IX downregulated the expression of p22phox, p53, and angiotensin type 1 receptor, which all are known FD-induced redox-sensitive EC senescence inducers. Taken together, HOP and its active components protect against FD-induced endothelial senescence most likely via antioxidant activity and may be a potential therapeutic agent for preventing and/or treating air-pollution-associated CVDs.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Deepak Adhikari
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Jeong Pyo Lee
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Ki-Woon Kang
- Division of Cardiology, Eulji University Hospital, Eulji University School of Medicine, Daejeon 34824, Korea;
| | - Ik-Soo Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea;
| | - Hyun Jung Kim
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| |
Collapse
|
26
|
Liang S, Zhang J, Ning R, Du Z, Liu J, Batibawa JW, Duan J, Sun Z. The critical role of endothelial function in fine particulate matter-induced atherosclerosis. Part Fibre Toxicol 2020; 17:61. [PMID: 33276797 PMCID: PMC7716453 DOI: 10.1186/s12989-020-00391-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
Ambient and indoor air pollution contributes annually to approximately seven million premature deaths. Air pollution is a complex mixture of gaseous and particulate materials. In particular, fine particulate matter (PM2.5) plays a major mortality risk factor particularly on cardiovascular diseases through mechanisms of atherosclerosis, thrombosis and inflammation. A review on the PM2.5-induced atherosclerosis is needed to better understand the involved mechanisms. In this review, we summarized epidemiology and animal studies of PM2.5-induced atherosclerosis. Vascular endothelial injury is a critical early predictor of atherosclerosis. The evidence of mechanisms of PM2.5-induced atherosclerosis supports effects on vascular function. Thus, we summarized the main mechanisms of PM2.5-triggered vascular endothelial injury, which mainly involved three aspects, including vascular endothelial permeability, vasomotor function and vascular reparative capacity. Then we reviewed the relationship between PM2.5-induced endothelial injury and atherosclerosis. PM2.5-induced endothelial injury associated with inflammation, pro-coagulation and lipid deposition. Although the evidence of PM2.5-induced atherosclerosis is undergoing continual refinement, the mechanisms of PM2.5-triggered atherosclerosis are still limited, especially indoor PM2.5. Subsequent efforts of researchers are needed to improve the understanding of PM2.5 and atherosclerosis. Preventing or avoiding PM2.5-induced endothelial damage may greatly reduce the occurrence and development of atherosclerosis.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Jingyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Ruihong Ning
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Zhou Du
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Jiangyan Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Joe Werelagi Batibawa
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| |
Collapse
|
27
|
Vasorelaxant Effect of Boesenbergia rotunda and Its Active Ingredients on an Isolated Coronary Artery. PLANTS 2020; 9:plants9121688. [PMID: 33271853 PMCID: PMC7760037 DOI: 10.3390/plants9121688] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 11/16/2022]
Abstract
Cardiovascular diseases are a major cause of death in developed countries. The regulation of vascular tone is a major approach to prevent and ameliorate vascular diseases. As part of our ongoing screening for cardioprotective natural compounds, we investigated the vasorelaxant effect of rhizomes from Boesenbergia rotunda (L.) Mansf. [Boesenbergia pandurata (Roxb.) Schltr.] used as a spice and herbal medicine in Asian countries. The methanol extract of B. rotunda rhizomes (BRE) exhibited significant vasorelaxation effects ex vivo at EC50 values of 13.4 ± 6.1 μg/mL and 40.9 ± 7.9 μg/mL, respectively, with and without endothelium in the porcine coronary artery ring. The intrinsic mechanism was evaluated by treating with specific inhibitors or activators that typically affect vascular reactivity. The results suggested that BRE induced relaxation in the coronary artery rings via an endothelium-dependent pathway involving NO-cGMP, and also via an endothelium-independent pathway involving the blockade of Ca2+ channels. Vasorelaxant principles in BRE were identified by subsequent chromatographic methods, which revealed that flavonoids regulate vasorelaxant activity in BRE. One of the flavonoids was a Diels-Alder type adduct, 4-hydroxypanduratin A, which showed the most potent vasorelaxant effect on porcine coronary artery with an EC50 of 17.8 ± 2.5 μM. Our results suggest that rhizomes of B. rotunda might be of interest as herbal medicine against cardiovascular diseases.
Collapse
|
28
|
Mescoli A, Maffei G, Pillo G, Bortone G, Marchesi S, Morandi E, Ranzi A, Rotondo F, Serra S, Vaccari M, Zauli Sajani S, Mascolo MG, Jacobs MN, Colacci A. The Secretive Liaison of Particulate Matter and SARS-CoV-2. A Hypothesis and Theory Investigation. Front Genet 2020; 11:579964. [PMID: 33240326 PMCID: PMC7680895 DOI: 10.3389/fgene.2020.579964] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/11/2020] [Indexed: 12/29/2022] Open
Abstract
As the novel coronavirus disease sweeps across the world, there is growing speculation on the role that atmospheric factors may have played on the different distribution of SARS-CoV-2, and on the epidemiological characteristics of COVID-19. Knowing the role that environmental factors play in influenza virus outbreaks, environmental pollution and, in particular, atmospheric airborne (particulate matter, PM) has been considered as a potential key factor in the spread and mortality of COVID-19. A possible role of the PM as the virus carrier has also been debated. The role of PM in exacerbating respiratory and cardiovascular disease has been well recognized. Accumulating evidence support the hypothesis that PM can trigger inflammatory response at molecular, cellular and organ levels. On this basis, we developed the hypothesis that PM may play a role as a booster of COVID-19 rather than as a carrier of SARS-CoV-2. To support our hypothesis, we analyzed the molecular signatures detected in cells exposed to PM samples collected in one of the most affected areas by the COVID-19 outbreak, in Italy. T47D human breast adenocarcinoma cells were chosen to explore the global gene expression changes induced by the treatment with organic extracts of PM 2.5. The analysis of the KEGG's pathways showed modulation of several gene networks related to the leucocyte transendothelial migration, cytoskeleton and adhesion system. Three major biological process were identified, including coagulation, growth control and immune response. The analysis of the modulated genes gave evidence for the involvement of PM in the endothelial disease, coagulation disorders, diabetes and reproductive toxicity, supporting the hypothesis that PM, directly or through molecular interplay, affects the same molecular targets as so far known for SARS-COV-2, contributing to the cytokines storm and to the aggravation of the symptoms triggered by COVID-19. We provide evidence for a plausible cooperation of receptors and transmembrane proteins, targeted by PM and involved in COVID-19, together with new insights into the molecular interplay of chemicals and pathogens that could be of importance for sustaining public health policies and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Ada Mescoli
- Department of Experimental, Diagnostic and Specialty Medicine, Section of Cancerology, University of Bologna, Bologna, Italy
| | - Giangabriele Maffei
- Department of Experimental, Diagnostic and Specialty Medicine, Section of Cancerology, University of Bologna, Bologna, Italy
| | - Gelsomina Pillo
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Giuseppe Bortone
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Stefano Marchesi
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Elena Morandi
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Andrea Ranzi
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Francesca Rotondo
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Stefania Serra
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | - Monica Vaccari
- Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| | | | | | - Miriam Naomi Jacobs
- Department of Toxicology, Centre for Radiation, Chemical and Environmental Hazards Public Health England, Chilton, United Kingdom
| | - Annamaria Colacci
- Department of Experimental, Diagnostic and Specialty Medicine, Section of Cancerology, University of Bologna, Bologna, Italy.,Agency for Prevention, Environment and Energy (Arpae), Emilia-Romagna, Italy
| |
Collapse
|
29
|
Microparticles-Mediated Vascular Inflammation and its Amelioration by Antioxidant Activity of Baicalin. Antioxidants (Basel) 2020; 9:antiox9090890. [PMID: 32962240 PMCID: PMC7555600 DOI: 10.3390/antiox9090890] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/14/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Microparticles (MPs) are extracellular vesicles (0.1–1.0 μm in size), released in response to cell activation or apoptosis. Endothelial microparticles (EC-MP), vascular smooth muscle cell microparticles (VSMC-MP), and macrophage microparticles (MØ-MP) are key hallmarks of atherosclerosis progression. In our current study, we investigated the potent antioxidant activity of baicalin to ameliorate MP-induced vascular smooth muscle cell (VSMC) dysfunction and endothelial cell (EC) dysfunction, as well as the production of inflammatory mediators in macrophage (RAW264.7). In our study, baicalin suppressed the apoptosis, reactive oxygen species (ROS) generation, NO production, foam cell formation, protein expression of inducible nitric oxide synthase and cyclooxygenase-2 in MØ-MP-induced RAW264.7. In addition, VSMC migration induced by VSMC-MP was dose-dependently inhibited by baicalin. Likewise, baicalin inhibits metalloproteinase-9 expression and suppresses VSMC-MP-induced VSMC proliferation by down-regulation of mitogen-activated protein kinase and proliferating cell nuclear antigen protein expressions. Baicalin also inhibited ROS production and apoptosis in VSMC. In EC, the marker of endothelial dysfunction (endothelial senescence, upregulation of ICAM, and ROS production) induced by EC-MP was halted by baicalin. Our results suggested that baicalin exerts potent biological activity to restore the function of EC and VSMC altered by their corresponding microparticles and inhibits the release of inflammation markers from activated macrophages.
Collapse
|
30
|
Miller MR. Oxidative stress and the cardiovascular effects of air pollution. Free Radic Biol Med 2020; 151:69-87. [PMID: 31923583 PMCID: PMC7322534 DOI: 10.1016/j.freeradbiomed.2020.01.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
Abstract
Cardiovascular causes have been estimated to be responsible for more than two thirds of the considerable mortality attributed to air pollution. There is now a substantial body of research demonstrating that exposure to air pollution has many detrimental effects throughout the cardiovascular system. Multiple biological mechanisms are responsible, however, oxidative stress is a prominent observation at many levels of the cardiovascular impairment induced by pollutant exposure. This review provides an overview of the evidence that oxidative stress is a key pathway for the different cardiovascular actions of air pollution.
Collapse
Affiliation(s)
- Mark R Miller
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH4 3RL, United Kingdom.
| |
Collapse
|