1
|
Park JW, Kang M, Kim G, Hyun SY, Shin J, Kim SY, Lee JH, Choi WS, Lee JH, Lee K, Kim SH, Cho WS, Kim HS. The impact of atmospheric ultrafine particulate matter on IgE-mediated type 1 hypersensitivity reaction. JOURNAL OF HAZARDOUS MATERIALS 2025; 484:136705. [PMID: 39637818 DOI: 10.1016/j.jhazmat.2024.136705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
The effect of atmospheric ultrafine particulate matter (UPM) on respiratory allergic diseases has been investigated for decades; however, the precise molecular mechanisms underlying these effects remain poorly understood. In this study, we used a simulated UPM (sUPM) generated via the spark discharge method to refine black carbon, a core particle that closely mimics real-world UPM, including the size (i.e., size of agglomerates: 165 nm) and organic carbon/elemental carbon ratio (i.e., 2.62). When 25 μg/mouse of dispersed sUPM was instilled into the lungs of mice, it promoted the infiltration and degranulation response of pulmonary mast cells, and exposure to sUPM in an immunoglobulin E (IgE)-mediated passive anaphylaxis model intensified the degranulation response of peripheral mast cells. These effects of sUPM were demonstrated to amplify the downstream signaling mechanism of the high-affinity IgE receptor (FcεRI) mediated by IgE when tested using rat basophil leukemia (RBL)-2H3 and mouse bone marrow-derived mast cells (BMMCs) collected from the bone marrow of BALB/c mice. These results indicate that airborne UPM can exacerbate type 1 hypersensitivity reactions by enhancing the IgE-mediated signaling pathways within mast cells. Furthermore, this study provided mechanistic evidence on exacerbated allergic pulmonary diseases induced by UPM inhalation.
Collapse
Affiliation(s)
- Jeong Won Park
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Minseong Kang
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Gyuri Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seung Yeun Hyun
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Juhyun Shin
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seon Young Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jun Ho Lee
- Department of Korean Medicine, College of Korean Medicine, Woosuk University, Jeonju 54986, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do 56212, Republic of Korea
| | - Seok-Ho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea.
| | - Hyuk Soon Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
2
|
Yao Z, Lu Y, Wang P, Chen Z, Zhou L, Sang X, Yang Q, Wang K, Hao M, Cao G. The role of JNK signaling pathway in organ fibrosis. J Adv Res 2024:S2090-1232(24)00431-4. [PMID: 39366483 DOI: 10.1016/j.jare.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Fibrosis is a tissue damage repair response caused by multiple pathogenic factors which could occur in almost every apparatus and leading to the tissue structure damage, physiological abnormality, and even organ failure until death. Up to now, there is still no specific drugs or strategies can effectively block or changeover tissue fibrosis. JNKs, a subset of mitogen-activated protein kinases (MAPK), have been reported that participates in various biological processes, such as genetic expression, DNA damage, and cell activation/proliferation/death pathways. Increasing studies indicated that abnormal regulation of JNK signal pathway has strongly associated with tissue fibrosis. AIM OF REVIEW This review designed to sum up the molecular mechanism progresses in the role of JNK signal pathway in organ fibrosis, hoping to provide a novel therapy strategy to tackle tissue fibrosis. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that JNK signaling pathway could modulates inflammation, immunoreaction, oxidative stress and Multiple cell biological functions in organ fibrosis. Therefore, targeting the JNK pathway may be a useful strategy in cure fibrosis.
Collapse
Affiliation(s)
- Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yandan Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Licheng Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Songyang Research Institute of Zhejiang Chinese Medical University, Songyang, 323400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
3
|
Li D, Zhang J, Jin Y, Zhu Y, Lu X, Huo X, Pan C, Zhong L, Sun K, Yan L, Yan L, Huang P, Li Q, Han JY, Li Y. Silibinin inhibits PM2.5-induced liver triglyceride accumulation through enhancing the function of mitochondrial Complexes I and II. Front Pharmacol 2024; 15:1435230. [PMID: 39351086 PMCID: PMC11440093 DOI: 10.3389/fphar.2024.1435230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Background The standardized extract of milk thistle seeds, known as silibinin, has been utilized in herbal medicine for over two centuries, with the aim of safeguarding the liver against the deleterious effects of various toxic substances. However, the role of silibinin in Particulate Matter (PM2.5)-induced intrahepatic triglyceride accumulation remains unclear. This study seeks to investigate the impact of silibinin on PM2.5-induced intrahepatic triglyceride accumulation and elucidate potential underlying mechanisms. Methods A model of intrahepatic triglyceride accumulation was established in male C57BL/6J mice through intratracheal instillation of PM2.5, followed by assessment of liver weight, body weight, liver index, and measurements of intrahepatic triglycerides and cholesterol after treatment with silibinin capsules. Hep G2 cells were exposed to PM2.5 suspension to create an intracellular triglyceride accumulation model, and after treatment with silibinin, cell viability, intracellular triglycerides and cholesterol, fluorescence staining for Nile Red (lipid droplets), and DCFH-DA (Reactive Oxygen Species, ROS), as well as proteomics, real-time PCR, and mitochondrial function assays, were performed to investigate the mechanisms involved in reducing triglycerides. Results PM2.5 exposure leads to triglyceride accumulation, increased ROS production, elevated expression of inflammatory factors, decreased expression of antioxidant factors, and increased expression of downstream genes of aryl hydrocarbon receptor. Silibinin can partially or fully reverse these factors, thereby protecting cells and animal livers from PM2.5-induced damage. In vitro studies show that silibinin exerts its protective effects by preserving oxidative phosphorylation of mitochondrial complexes I and II, particularly significantly enhancing the function of mitochondrial complex II. Succinate dehydrogenase (mitochondrial complex II) is a direct target of silibinin, but silibinin A and B exhibit different affinities for different subunits of complex II. Conclusion Silibinin improved the accumulation of intrahepatic triglycerides induced by PM2.5, and this was, at least in part, explained by an enhancement of oxidative phosphorylation in mitochondrial Complexes I and II.
Collapse
Affiliation(s)
- Dexin Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Jingxin Zhang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Yuxin Jin
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Yaoxuan Zhu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Xiaoqing Lu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Xinmei Huo
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Chunshui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Lijun Zhong
- Peking University Medical and Health Analysis Center, Peking University, Beijing, China
| | - Kai Sun
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Lulu Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Ping Huang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Yin Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- The Key Discipline for Integration of Chinese and Western Basic Medicine (Microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing, China
- Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Beijing, China
- Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| |
Collapse
|
4
|
Piotin A, Oulehri W, Charles AL, Tacquard C, Collange O, Mertes PM, Geny B. Oxidative Stress and Mitochondria Are Involved in Anaphylaxis and Mast Cell Degranulation: A Systematic Review. Antioxidants (Basel) 2024; 13:920. [PMID: 39199166 PMCID: PMC11352116 DOI: 10.3390/antiox13080920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Anaphylaxis, an allergic reaction caused by the massive release of active mediators, can lead to anaphylactic shock (AS), the most severe and potentially life-threatening form of anaphylactic reaction. Nevertheless, understanding of its pathophysiology to support new therapies still needs to be improved. We performed a systematic review, assessing the role and the complex cellular interplay of mitochondria and oxidative stress during anaphylaxis, mast cell metabolism and degranulation. After presenting the main characteristics of anaphylaxis, the oxidant/antioxidant balance and mitochondrial functions, we focused this review on the involvement of mitochondria and oxidative stress in anaphylaxis. Then, we discussed the role of oxidative stress and mitochondria following mast cell stimulation by allergens, leading to degranulation, in order to further elucidate mechanistic pathways. Finally, we considered potential therapeutic interventions implementing these findings for the treatment of anaphylaxis. Experimental studies evaluated mainly cardiomyocyte metabolism during AS. Cardiac dysfunction was associated with left ventricle mitochondrial impairment and lipid peroxidation. Studies evaluating in vitro mast cell degranulation, following Immunoglobulin E (IgE) or non-IgE stimulation, revealed that mitochondrial respiratory complex integrity and membrane potential are crucial for mast cell degranulation. Antigen stimulation raises reactive oxygen species (ROS) production from nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and mitochondria, leading to mast cell degranulation. Moreover, mast cell activation involved mitochondrial morphological changes and mitochondrial translocation to the cell surface near exocytosis sites. Interestingly, antioxidant administration reduced degranulation by lowering ROS levels. Altogether, these results highlight the crucial role of oxidative stress and mitochondria during anaphylaxis and mast cell degranulation. New therapeutics against anaphylaxis should probably target oxidative stress and mitochondria, in order to decrease anaphylaxis-induced systemic and major organ deleterious effects.
Collapse
Affiliation(s)
- Anays Piotin
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, 67000 Strasbourg, France
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Walid Oulehri
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Anne-Laure Charles
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Charles Tacquard
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
- Établissement Français du Sang (EFS) Grand Est, French National Institute of Health and Medical Research), (INSERM) BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, 67000 Strasbourg, France
| | - Olivier Collange
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Paul-Michel Mertes
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Bernard Geny
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| |
Collapse
|
5
|
Lin M, Yan J, Tang J, Han S, Guo P, Wu S, Tao L, Xiao H, Chen Y, Tan X. Air Pollutants and Mortality Risk in Patients with Aortic Dissection: Evidence from a Clinical Cohort, Single-Cell Sequencing, and Proteomics. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:6509-6518. [PMID: 38561599 PMCID: PMC11025546 DOI: 10.1021/acs.est.4c00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
We aimed to evaluate the association between air pollutants and mortality risk in patients with acute aortic dissection (AAD) in a longitudinal cohort and to explore the potential mechanisms of adverse prognosis induced by fine particulate matter (PM2.5). Air pollutants data, including PM2.5, PM10.0, nitrogen dioxide (NO2), carbon monoxide (CO), sulfur dioxide (SO2), and ozone (O3), were collected from official monitoring stations, and multivariable Cox regression models were applied. Single-cell sequencing and proteomics of aortic tissue were conducted to explore the potential mechanisms. In total, 1,267 patients with AAD were included. Exposure to higher concentrations of air pollutants was independently associated with an increased mortality risk. The high-PM2.5 group carried approximately 2 times increased mortality risk. There were linear associations of PM10, NO2, CO, and SO2 exposures with long-term mortality risk. Single-cell sequencing revealed an increase in mast cells in aortic tissue in the high-PM2.5 exposure group. Enrichment analysis of the differentially expressed genes identified the inflammatory response as one of the main pathways, with IL-17 and TNF signaling pathways being among the top pathways. Analysis of proteomics also identified these pathways. This study suggests that exposure to higher PM2.5, PM10, NO2, CO, and SO2 are associated with increased mortality risk in patients with AAD. PM2.5-related activation and degranulation of mast cells may be involved in this process.
Collapse
Affiliation(s)
- Mengyue Lin
- Department
of Cardiology, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
- Shantou
University Medical College, No. 22 Xinling Road, Shantou 515000, China
| | - Jingyi Yan
- Shantou
University Medical College, No. 22 Xinling Road, Shantou 515000, China
| | - Junshuang Tang
- Shantou
University Medical College, No. 22 Xinling Road, Shantou 515000, China
| | - Sirui Han
- Shantou
University Medical College, No. 22 Xinling Road, Shantou 515000, China
| | - Pi Guo
- Department
of Preventive Medicine, Shantou University
Medical College, No. 22 Xinling Road, Shantou 515000, China
| | - Shiwan Wu
- Department
of Cardiology, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
| | - Liang Tao
- Department
of Cardiac Surgery, Wuhan Asia Heart Hospital
Affiliated with Wuhan University of Science and Technology, No. 753 Jinghan Road, Wuhan 430000, China
| | - Hongyan Xiao
- Department
of Cardiac Surgery, Wuhan Asia Heart Hospital
Affiliated with Wuhan University of Science and Technology, No. 753 Jinghan Road, Wuhan 430000, China
| | - Yequn Chen
- Department
of Cardiology, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
- Clinical
Research Center, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
| | - Xuerui Tan
- Department
of Cardiology, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
- Shantou
University Medical College, No. 22 Xinling Road, Shantou 515000, China
- Clinical
Research Center, First Affiliated Hospital
of Shantou University Medical College, No. 57 Changping Road, Shantou 515000, China
| |
Collapse
|
6
|
Zhu J, Zhou Y, Lin Q, Wu K, Ma Y, Liu C, Liu N, Tu T, Liu Q. Causal relationship between particulate matter and COVID-19 risk: A mendelian randomization study. Heliyon 2024; 10:e27083. [PMID: 38439838 PMCID: PMC10909784 DOI: 10.1016/j.heliyon.2024.e27083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
Background Observational studies have linked exposure to fine (PM2.5) and coarse (PM10) particulate matter air pollution with adverse COVID-19 outcomes, including higher incidence and mortality. However, some studies questioned the effect of air pollution on COVID-19 susceptibility, raising questions about the causal nature of these associations. To address this, a less biased method like Mendelian randomization (MR) is utilized, which employs genetic variants as instrumental variables to infer causal relationships in observational data. Method We performed two-sample MR analysis using public genome-wide association studies data. Instrumental variables correlated with PM2.5 concentration, PM2.5 absorbance, PM2.5-10 concentration and PM10 concentration were identified. The inverse variance weighted (IVW), robust adjusted profile score (RAPS) and generalized summary data-based Mendelian randomization (GSMR) methods were used for analysis. Results IVW MR analysis showed PM2.5 concentration [odd ratio (OR) = 3.29, 95% confidence interval (CI) 1.48-7.35, P-value = 0.0036], PM2.5 absorbance (OR = 5.62, 95%CI 1.98-15.94, P-value = 0.0012), and PM10 concentration (OR = 3.74, 95%CI 1.52-9.20, P-value = 0.0041) increased the risk of COVID-19 severity after Bonferroni correction. Further validation confirmed PM2.5 absorbance was associated with heightened COVID-19 severity (OR = 6.05, 95%CI 1.99-18.38, P-value = 0.0015 for RAPS method; OR = 4.91, 95%CI 1.65-14.59, P-value = 0.0042 for GSMR method) and hospitalization (OR = 3.15, 95%CI 1.54-6.47, P-value = 0.0018 for RAPS method). No causal links were observed between particulate matter exposure and COVID-19 susceptibility. Conclusions Our study established a causal relationship between smaller particle pollution, specifically PM2.5, and increased risk of COVID-19 severity and hospitalization. These findings highlight the importance of improving air quality to mitigate respiratory disease progression.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Chan Liu
- International Medical Department, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| |
Collapse
|
7
|
Elieh-Ali-Komi D, Bot I, Rodríguez-González M, Maurer M. Cellular and Molecular Mechanisms of Mast Cells in Atherosclerotic Plaque Progression and Destabilization. Clin Rev Allergy Immunol 2024; 66:30-49. [PMID: 38289515 DOI: 10.1007/s12016-024-08981-9] [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] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Mast cells (MCs) are commonly recognized for their crucial involvement in the pathogenesis of allergic diseases, but over time, it has come to light that they also play a role in the pathophysiology of non-allergic disorders including atherosclerosis. The involvement of MCs in the pathology of atherosclerosis is supported by their accumulation in atherosclerotic plaques upon their progression and the association of intraplaque MC numbers with acute cardiovascular events. MCs that accumulate within the atherosclerotic plaque release a cocktail of mediators through which they contribute to neovascularization, plaque progression, instability, erosion, rupture, and thrombosis. At a molecular level, MC-released proteases, especially cathepsin G, degrade low-density lipoproteins (LDL) and mediate LDL fusion and binding of LDL to proteoglycans (PGs). Through a complicated network of chemokines including CXCL1, MCs promote the recruitment of among others CXCR2+ neutrophils, therefore, aggravating the inflammation of the plaque environment. Additionally, MCs produce extracellular traps which worsen inflammation and contribute to atherothrombosis. Altogether, evidence suggests that MCs actively, via several underlying mechanisms, contribute to atherosclerotic plaque destabilization and acute cardiovascular syndromes, thus, making the study of interventions to modulate MC activation an interesting target for cardiovascular medicine.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Marcus Maurer
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany.
| |
Collapse
|
8
|
Wu S, Guo W, Chen L, Lin X, Tang M, Lin C, Guo H, Zhang T, Gao Y. Downregulation of Gadd45β alleviates osteoarthritis by repressing lipopolysaccharide-induced fibroblast-like synoviocyte inflammation, proliferation and migration. Int Immunopharmacol 2024; 126:111202. [PMID: 37988908 DOI: 10.1016/j.intimp.2023.111202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE Gadd45β have a regulatory role in cellular inflammation, proliferation and migration. However, the role of Gadd45β in synovial inflammation in osteoarthritis (OA) remains to be explored. This study aimed to ascertain whether Gadd45β is involved in OA synovial inflammation. METHODS The rat model was induced by sodium iodoacetate and the cellular model was constructed with lipopolysaccharide (LPS)-induced fibroblast-like synoviocytes (FLSs). siRNA was applied to interfere with the expression of intracellular Gadd45β. Real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to detect the expression of Gadd45β mRNA and protein. The inflammation, proliferation, and migration of OA-FLSs were detected by enzyme-linked immunosorbent assay, cell scratch assay, 5-ethynyl-2'-deoxyuridine assay, etc. The effect of downregulation of Gadd45β on the nuclear factor-κB (NF-κB) pathway was investigated. RESULTS Expression of Gadd45β in OA rat synovial tissues and OA-FLSs was increased, and LPS treatment promoted cell proliferation and enhanced cell migration. Gadd45β interference inhibited the inflammation, proliferation and migration of cells induced by LPS. LPS promoted P65 expression in the nucleus and activated the NF-κB signaling pathway, whereas si-Gadd45β reversed this situation. CONCLUSIONS si-Gadd45β inhibited the inflammatory response, proliferation and migration of FLSs, and activation of the NF-κB signaling pathway, which could delay the progression of OA. Hence, it may become a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Suyu Wu
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Wenwen Guo
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Ling Chen
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Xinxin Lin
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China; Department of Pathology, Fuzhou Second Hospital, Fuzhou 350007, Fujian, China
| | - Minjie Tang
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Cheng Lin
- The School of Health, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Hanzhi Guo
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Tianwen Zhang
- Fujian Fishery Resources Monitoring Center, Fuzhou 350003, Fujian, China
| | - Yao Gao
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China; Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou 350004, Fujian, China.
| |
Collapse
|
9
|
Park S, Lim J, Kim S, Jeon M, Baek H, Park W, Park J, Kim SN, Kang NG, Park CG, Kim JW. Anti-Inflammatory Artificial Extracellular Vesicles with Notable Inhibition of Particulate Matter-Induced Skin Inflammation and Barrier Function Impairment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59199-59208. [PMID: 37983083 DOI: 10.1021/acsami.3c14377] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Particulate matter (PM) exposure disrupts the skin barrier, causing cutaneous inflammation that may eventually contribute to the development of various skin diseases. Herein, we introduce anti-inflammatory artificial extracellular vesicles (AEVs) fabricated through cell extrusion using the biosurfactant PEGylated mannosylerythritol lipid (P-MEL), hereafter named AEVP-MEL. The P-MEL has anti-inflammatory abilities with demonstrated efficacy in inhibiting the secretion of pro-inflammatory mediators. Mechanistically, AEVP-MEL enhanced anti-inflammatory response by inhibiting the mitogen-activated protein kinase (MAPK) pathway and decreasing the release of inflammatory mediators such as reactive oxygen species (ROS), cyclooxygenase-2 (COX-2), and pro-inflammatory cytokines in human keratinocytes. Moreover, AEVP-MEL promoted increased expression levels of skin barrier proteins (e.g., involucrin, IVL) and water-proteins (e.g., aquaporin 3, AQP3). In vivo studies revealed that repeated PM exposure to intact skin resulted in cutaneous inflammatory responses, including increased skin thickness (hyperkeratosis) and mast cell infiltration. Importantly, our data showed that the AEVP-MEL treatment significantly restored immune homeostasis in the skin affected by PM-induced inflammation and enhanced the intrinsic skin barrier function. This study highlights the potential of the AEVP-MEL in promoting skin health against PM exposure and its promising implications for the prevention and treatment of PM-related skin disorders.
Collapse
Affiliation(s)
- Simon Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jaesung Lim
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seulgi Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minha Jeon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai'i at Manoa, Honolulu 96813, United States
| | - Se Na Kim
- Research and Development Center, MediArk Inc.,Cheongju 28644, Republic of Korea
- Department of Industrial Cosmetic Science, College of Bio-Health University System, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Nae-Gyu Kang
- R&D Campus, LG Household & Health Care, Seoul 07795, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
10
|
Chao L, Feng B, Liang H, Zhao X, Song J. Particulate matter and inflammatory skin diseases: From epidemiological and mechanistic studies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167111. [PMID: 37716690 DOI: 10.1016/j.scitotenv.2023.167111] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
Epidemiological and toxicological studies have confirmed that exposure to atmospheric particulate matter (PM) could affect our cardiovascular and respiratory systems. Recent studies have shown that PM can penetrate the skin and cause skin inflammation, but the evidence is limited and contradictory. As the largest outermost surface of the human body, the skin is constantly exposed to the environment. The aim of this study was to assess the relationship between PM and inflammatory skin diseases. Most epidemiological studies have provided positive evidence for outdoor, indoor, and wildfire PM and inflammatory skin diseases. The effects of PM exposure during pregnancy and inflammatory skin diseases in offspring are heterogeneous. Skin barrier dysfunction, Oxidative stress, and inflammation may play a critical role in the underlying mechanisms. Finally, we summarize some interventions to alleviate PM-induced inflammatory skin diseases, which may contribute to public health welfare. Overall, PM is related to inflammatory skin diseases via skin barrier dysfunction, oxidative stress, and inflammation. Appropriate government interventions are beneficial.
Collapse
Affiliation(s)
- Ling Chao
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Bin Feng
- Environmental Health Section, Xinxiang Health Technology Supervision Center, School of Management, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Haiyan Liang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Xiangmei Zhao
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Song
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
11
|
Atiakshin D, Kostin A, Volodkin A, Nazarova A, Shishkina V, Esaulenko D, Buchwalow I, Tiemann M, Noda M. Mast Cells as a Potential Target of Molecular Hydrogen in Regulating the Local Tissue Microenvironment. Pharmaceuticals (Basel) 2023; 16:817. [PMID: 37375765 DOI: 10.3390/ph16060817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Knowledge of the biological effects of molecular hydrogen (H2), hydrogen gas, is constantly advancing, giving a reason for the optimism in several healthcare practitioners regarding the management of multiple diseases, including socially significant ones (malignant neoplasms, diabetes mellitus, viral hepatitis, mental and behavioral disorders). However, mechanisms underlying the biological effects of H2 are still being actively debated. In this review, we focus on mast cells as a potential target for H2 at the specific tissue microenvironment level. H2 regulates the processing of pro-inflammatory components of the mast cell secretome and their entry into the extracellular matrix; this can significantly affect the capacity of the integrated-buffer metabolism and the structure of the immune landscape of the local tissue microenvironment. The analysis performed highlights several potential mechanisms for developing the biological effects of H2 and offers great opportunities for translating the obtained findings into clinical practice.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Artem Volodkin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Anna Nazarova
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Viktoriya Shishkina
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Dmitry Esaulenko
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Igor Buchwalow
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 816-0811, Japan
| |
Collapse
|
12
|
Garcia A, Santa-Helena E, De Falco A, de Paula Ribeiro J, Gioda A, Gioda CR. Toxicological Effects of Fine Particulate Matter (PM 2.5): Health Risks and Associated Systemic Injuries-Systematic Review. WATER, AIR, AND SOIL POLLUTION 2023; 234:346. [PMID: 37250231 PMCID: PMC10208206 DOI: 10.1007/s11270-023-06278-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 03/29/2023] [Indexed: 05/31/2023]
Abstract
Previous studies focused on investigating particulate matter with aerodynamic diameter ≤ 2.5 µm (PM2.5) have shown the risk of disease development, and association with increased morbidity and mortality rates. The current review investigate epidemiological and experimental findings from 2016 to 2021, which enabled the systemic overview of PM2.5's toxic impacts on human health. The Web of Science database search used descriptive terms to investigate the interaction among PM2.5 exposure, systemic effects, and COVID-19 disease. Analyzed studies have indicated that cardiovascular and respiratory systems have been extensively investigated and indicated as the main air pollution targets. Nevertheless, PM2.5 reaches other organic systems and harms the renal, neurological, gastrointestinal, and reproductive systems. Pathologies onset and/or get worse due to toxicological effects associated with the exposure to this particle type, since it can trigger several reactions, such as inflammatory responses, oxidative stress generation and genotoxicity. These cellular dysfunctions lead to organ malfunctions, as shown in the current review. In addition, the correlation between COVID-19/Sars-CoV-2 and PM2.5 exposure was also assessed to help better understand the role of atmospheric pollution in the pathophysiology of this disease. Despite the significant number of studies about PM2.5's effects on organic functions, available in the literature, there are still gaps in knowledge about how this particulate matter can hinder human health. The current review aimed to approach the main findings about the effect of PM2.5 exposure on different systems, and demonstrate the likely interaction of COVID-19/Sars-CoV-2 and PM2.5.
Collapse
Affiliation(s)
- Amanda Garcia
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Eduarda Santa-Helena
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Anna De Falco
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Joaquim de Paula Ribeiro
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Adriana Gioda
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Carolina Rosa Gioda
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| |
Collapse
|
13
|
Liu HM, Cheng MY, Xun MH, Zhao ZW, Zhang Y, Tang W, Cheng J, Ni J, Wang W. Possible Mechanisms of Oxidative Stress-Induced Skin Cellular Senescence, Inflammation, and Cancer and the Therapeutic Potential of Plant Polyphenols. Int J Mol Sci 2023; 24:ijms24043755. [PMID: 36835162 PMCID: PMC9962998 DOI: 10.3390/ijms24043755] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
As the greatest defense organ of the body, the skin is exposed to endogenous and external stressors that produce reactive oxygen species (ROS). When the antioxidant system of the body fails to eliminate ROS, oxidative stress is initiated, which results in skin cellular senescence, inflammation, and cancer. Two main possible mechanisms underlie oxidative stress-induced skin cellular senescence, inflammation, and cancer. One mechanism is that ROS directly degrade biological macromolecules, including proteins, DNA, and lipids, that are essential for cell metabolism, survival, and genetics. Another one is that ROS mediate signaling pathways, such as MAPK, JAK/STAT, PI3K/AKT/mTOR, NF-κB, Nrf2, and SIRT1/FOXO, affecting cytokine release and enzyme expression. As natural antioxidants, plant polyphenols are safe and exhibit a therapeutic potential. We here discuss in detail the therapeutic potential of selected polyphenolic compounds and outline relevant molecular targets. Polyphenols selected here for study according to their structural classification include curcumin, catechins, resveratrol, quercetin, ellagic acid, and procyanidins. Finally, the latest delivery of plant polyphenols to the skin (taking curcumin as an example) and the current status of clinical research are summarized, providing a theoretical foundation for future clinical research and the generation of new pharmaceuticals and cosmetics.
Collapse
Affiliation(s)
- Hui-Min Liu
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
- Engineering Research Center of Perfume & Aroma and Cosmetics, Ministry of Education, Shanghai 201418, China
| | - Ming-Yan Cheng
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Meng-Han Xun
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhi-Wei Zhao
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yun Zhang
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wei Tang
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jun Cheng
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jia Ni
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wei Wang
- School of Perfume & Aroma and Cosmetics, Shanghai Institute of Technology, Shanghai 201418, China
- Engineering Research Center of Perfume & Aroma and Cosmetics, Ministry of Education, Shanghai 201418, China
- Correspondence: ; Tel.: +86-18918830550
| |
Collapse
|
14
|
Wang X, Niu L, Kang A, Pang Y, Zhang Y, Wang W, Zhang Y, Huang X, Liu Q, Geng Z, He L, Niu Y, Zhang R. Effects of ambient PM 2.5 on development of psoriasiform inflammation through KRT17-dependent activation of AKT/mTOR/HIF-1α pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:114008. [PMID: 36029575 DOI: 10.1016/j.ecoenv.2022.114008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Exposure to fine particulate matter (PM2.5) has significant effects on human skin health, mainly disrupting skin homeostasis and accelerating aging. To date, the effects of PM2.5 on psoriasis (PSO) have not been elucidated. An ambient particulate matter exposed and well characterized imiquimod (IMQ)-induced psoriasis mouse model was established. Thirty male C57BL/6 mice aged 8 weeks were randomly divided into three groups: filtered air (FA) group (Control group), PSO+ FA group and PSO + PM2.5 group. A KRT17 knockdown (KRT17-KD) mouse model was simultaneously established by subcutaneously injecting KRT17-KD lentivirus. Forty male C57BL/6 mice were randomly divided into four groups: PSO + FA + KRT17-RNAi negative control lentivirus (KRT17-NC) group, PSO+ FA+ KRT17-KD group, PSO + PM2.5 + KRT17-NC group and PSO + PM2.5 + KRT17-KD group. PM2.5 exposure continued for 8 weeks. Psoriasis was induced by topically applying IMQ on the dorsal skin of the mice for 6 days during week 8. Morphometric and histological analyses were performed to investigate the changes in psoriatic lesions. Differentially expressed genes and enriched pathways were explored using bioinformatics analysis and showed that KRT17 gene and the vascular endothelial growth factor receptor signaling pathway were associated with psoriasis. HaCaT cells were stimulated with interleukin-17A and infected with KRT17-KD lentivirus to establish an in vitro KRT17 knockdown psoriasis cell model. Notably, PM2.5 exposure increased the expression of KRT17 protein and activated AKT/mTOR/HIF-1α signaling pathway in vivo. Moreover, specific agonist of AKT (740Y-P) reversed the decreased neovascularization induced by KRT17 knockdown through AKT/mTOR/HIF-1α signaling pathway in vitro. Consequently, PM2.5 exposure could promote the development and progression of psoriasis through KRT17-dependent activation of AKT/mTOR/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Xueliang Wang
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China; Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Linpeng Niu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Aijuan Kang
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Yaling Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Wenqing Wang
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Yan Zhang
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Xiaoyan Huang
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Zihan Geng
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Liyi He
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Yujie Niu
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, People's Republic of China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, People's Republic of China.
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, People's Republic of China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, People's Republic of China.
| |
Collapse
|
15
|
Yang YS, Cao MD, Wang A, Liu QM, Zhu DX, Zou Y, Ma LL, Luo M, Shao Y, Xu DD, Wei JF, Sun JL. Nano-silica particles synergistically IgE-mediated mast cell activation exacerbating allergic inflammation in mice. Front Immunol 2022; 13:911300. [PMID: 35936002 PMCID: PMC9355306 DOI: 10.3389/fimmu.2022.911300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/27/2022] [Indexed: 12/05/2022] Open
Abstract
Background Allergic respiratory diseases have increased dramatically due to air pollution over the past few decades. However, studies are limited on the effects of inorganic components and particulate matter with different particle sizes in smog on allergic diseases, and the possible molecular mechanism of inducing allergies has not been thoroughly studied. Methods Four common mineral elements with different particle sizes in smog particles were selected, including Al2O3, TiO2, Fe2O3, and SiO2. We studied the relationship and molecular mechanism of smog particle composition, particle size, and allergic reactions using mast cells, immunoglobulin E (IgE)-mediated passive cutaneous anaphylaxis (PCA) model, and an ovalbumin (OVA)-induced asthmatic mouse model in vitro and in vivo, combined with transmission electron microscopy, scanning transmission X-ray microscopy analysis, and transcriptome sequencing. Results Only 20 nm SiO2 particles significantly increased β-hexosaminidase release, based on dinitrophenol (DNP)-human serum albumin (HSA) stimulation, from IgE-sensitized mast cells, while other particles did not. Meanwhile, the PCA model showed that Evan’s blue extravasation in mice was increased after treatment with nano-SiO2 particles. Nano-SiO2 particles exposure in the asthmatic mouse model caused an enhancement of allergic airway inflammation as manifested by OVA-specific serum IgE, airway hyperresponsiveness, lung inflammation injury, mucous cell metaplasia, cytokine expression, mast cell activation, and histamine secretion, which were significantly increased. Nano-SiO2 particles exposure did not affect the expression of FcϵRI or the ability of mast cells to bind IgE but synergistically activated mast cells by enhancing the mitogen-activated protein kinase (MAPK) signaling pathway, especially the phosphorylation levels of the extracellular signal-regulated kinase (ERK)1/2. The ERK inhibitors showed a significant inhibitory effect in reducing β-hexosaminidase release. Conclusion Our results indicated that nano-SiO2 particles stimulation might synergistically activate IgE-sensitized mast cells by enhancing the MAPK signaling pathway and that nano-SiO2 particles exposure could exacerbate allergic inflammation. Our experimental results provide useful information for preventing and treating allergic diseases.
Collapse
Affiliation(s)
- Yong-Shi Yang
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Meng-Da Cao
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - An Wang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Qing-Mei Liu
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Dan-Xuan Zhu
- Women and Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Zou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Ling-Ling Ma
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Min Luo
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Yang Shao
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Dian-Dou Xu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Jin-Lyu Sun, ; Ji-Fu Wei, ; Dian-Dou Xu,
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Jin-Lyu Sun, ; Ji-Fu Wei, ; Dian-Dou Xu,
| | - Jin-Lyu Sun
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Jin-Lyu Sun, ; Ji-Fu Wei, ; Dian-Dou Xu,
| |
Collapse
|
16
|
Kwack MH, Bang JS, Lee WJ. Preventative Effects of Antioxidants against PM 10 on Serum IgE Concentration, Mast Cell Counts, Inflammatory Cytokines, and Keratinocyte Differentiation Markers in DNCB-Induced Atopic Dermatitis Mouse Model. Antioxidants (Basel) 2022; 11:1334. [PMID: 35883825 PMCID: PMC9311925 DOI: 10.3390/antiox11071334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 12/02/2022] Open
Abstract
Particulate matter (PM) can cause oxidative stress, inflammation, and skin aging. We investigated the effects of antioxidants such as dieckol, punicalagin, epigallocatechin gallate (EGCG), resveratrol, and Siegesbeckiae Herba extract (SHE) against PM < 10 μm (PM10) on serum IgE concentration, mast cell counts, inflammatory cytokines, and keratinocyte differentiation markers in a 2,4-Dinitrochlorobenzene (DNCB)-induced atopic dermatitis mouse model. Seven-week-old BALB/c mice were sensitized with 2% DNCB. Atopic dermatitis-like lesions were induced on the mice with 0.2% DNCB. Antioxidants and PM10 were applied to the mice for 4 weeks. PM10 increased the serum IgE concentration and spleen weight in mice, and all antioxidants downregulated these parameters. Histological examination showed an increase in epidermal thickness and mast cell counts in response to PM10, and all antioxidants showed a decrease. PM10 upregulates the expression of inflammatory cytokines, including interleukin (IL)-1β, IL-4, IL-6, IL-17α, IL-25, IL-31 and thymic stromal lymphopoietin (TSLP) in mice, and all antioxidants inhibited the upregulation of inflammatory cytokines. ELISA showed the same results as real-time PCR. PM10 downregulates the expression of keratinocyte differentiation markers, including loricrin and filaggrin, in mouse keratinocytes and antioxidants prevented the downregulation of the keratinocyte differentiation markers. Conclusively, PM10 aggravated the DNCB-induced mouse model in serum IgE concentration, mast cell counts, inflammatory cytokine, and keratinocyte differentiation markers. In addition, antioxidants modulated changes in the DNCB-induced mouse model caused by PM10.
Collapse
Affiliation(s)
- Mi Hee Kwack
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Jin Seon Bang
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Weon Ju Lee
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| |
Collapse
|
17
|
Connecting the Dots in Emerging Mast Cell Research: Do Factors Affecting Mast Cell Activation Provide a Missing Link between Adverse COVID-19 Outcomes and the Social Determinants of Health? Med Sci (Basel) 2022; 10:medsci10020029. [PMID: 35736349 PMCID: PMC9228930 DOI: 10.3390/medsci10020029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/09/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023] Open
Abstract
Evidence continues to emerge that the social determinants of health play a role in adverse outcomes related to COVID-19, including increased morbidity and mortality, increased risk of long COVID, and vaccine adverse effects. Therefore, a more nuanced understanding of the biochemical and cellular pathways of illnesses commonly associated with adverse social determinants of health is urgently needed. We contend that a commitment to understanding adverse outcomes in historically marginalized communities will increase community-level confidence in public health measures. Here, we synthesize emerging literature on mast cell disease, and the role of mast cells in chronic illness, alongside emerging research on mechanisms of COVID illness and vaccines. We propose that a focus on aberrant and/or hyperactive mast cell behavior associated with chronic underlying health conditions can elucidate adverse COVID-related outcomes and contribute to the pandemic recovery. Standards of care for mast cell activation syndrome (MCAS), as well as clinical reviews, experimental research, and case reports, suggest that effective and cost-efficient remedies are available, including antihistamines, vitamin C, and quercetin, among others. Primary care physicians, specialists, and public health workers should consider new and emerging evidence from the biomedical literature in tackling COVID-19. Specialists and researchers note that MCAS is likely grossly under-diagnosed; therefore, public health agencies and policy makers should urgently attend to community-based experiences of adverse COVID outcomes. It is essential that we extract and examine experiential evidence of marginalized communities from the broader political–ideological discourse.
Collapse
|
18
|
Mishra PK, Bhargava A, Kumari R, Bunkar N, Chauhan P, Mukherjee S, Shandilya R, Singh RD, Tiwari R, Chaudhury K. Integrated mitoepigenetic signalling mechanisms associated with airborne particulate matter exposure: A cross-sectional pilot study. ATMOSPHERIC POLLUTION RESEARCH 2022; 13:101399. [DOI: 10.1016/j.apr.2022.101399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
19
|
Qu S, Deng S, Yang T, Yang Y, Zhang Y, Zheng Z, Chen L, Li Y. Shengmai Yin alleviated plaque vulnerability and ischemic myocardial damage in diesel exhaust particle-aggravated atherosclerosis with myocardial ischemia. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113379. [PMID: 35278994 DOI: 10.1016/j.ecoenv.2022.113379] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/17/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
Exposure to diesel exhaust particles (DEP) increases the risk of ischemic heart disease, especially heart attacks and ischemic/thrombotic strokes. Shengmai Yin (SMY) is a traditional Chinese medicine used to treat coronary heart disease. The aim of this study was to determine the protective role of SMY and the mechanism by which SMY affects DEP-induced cardiovascular injury. This study is expected to provide the basis for the development of an adaptive signature of SMY in the prevention of atherosclerotic cardiovascular disease and premature death from global air pollution exposure. We developed animal models of myocardial ischemia and atherosclerosis (AS) in response to DEP exposure. After SMY treatment, serum lipids returned to normal. Aortic plaque area and MMP9 expression were significantly reduced and collagen fiber expression increased after SMY treatment compared to DEP exposure alone. Thus, the risk of plaque formation and vulnerability is reduced. In addition, SMY improved left ventricular structure, morphology, function, blood flow, infarct area, myocardial damage, and ROS accumulation to varying degrees in ApoE-/- mice. These results indicate that the use of SMY is effective, to varying degrees, for the treatment of dyslipidemia, atherosclerosis, myocardial ischemia, and oxidative stress in ApoE-/- mice. SMY has a potential protective effect in DEP-aggravated AS in people with myocardial ischemia.
Collapse
Affiliation(s)
- Shuiqing Qu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuoqiu Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ting Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanmin Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyuan Zheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
20
|
Choi JY, Bae JE, Kim JB, Jo DS, Park NY, Kim YH, Lee HJ, Kim SH, Kim SH, Jeon HB, Na HW, Choi H, Ryu HY, Ryoo ZY, Lee HS, Cho DH. 2-IPMA Ameliorates PM2.5-Induced Inflammation by Promoting Primary Ciliogenesis in RPE Cells. Molecules 2021; 26:molecules26175409. [PMID: 34500843 PMCID: PMC8433925 DOI: 10.3390/molecules26175409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
Primary cilia mediate the interactions between cells and external stresses. Thus, dysregulation of primary cilia is implicated in various ciliopathies, e.g., degeneration of the retina caused by dysregulation of the photoreceptor primary cilium. Particulate matter (PM) can cause epithelium injury and endothelial dysfunction by increasing oxidative stress and inflammatory responses. Previously, we showed that PM disrupts the formation of primary cilia in retinal pigment epithelium (RPE) cells. In the present study, we identified 2-isopropylmalic acid (2-IPMA) as a novel inducer of primary ciliogenesis from a metabolite library screening. Both ciliated cells and primary cilium length were increased in 2-IPMA-treated RPE cells. Notably, 2-IPMA strongly promoted primary ciliogenesis and restored PM2.5-induced dysgenesis of primary cilia in RPE cells. Both excessive reactive oxygen species (ROS) generation and activation of a stress kinase, JNK, by PM2.5 were reduced by 2-IPMA. Moreover, 2-IPMA inhibited proinflammatory cytokine production, i.e., IL-6 and TNF-α, induced by PM2.5 in RPE cells. Taken together, our data suggest that 2-IPMA ameliorates PM2.5-induced inflammation by promoting primary ciliogenesis in RPE cells.
Collapse
Affiliation(s)
- Ji Yeon Choi
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Ji-Eun Bae
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea;
| | - Joon Bum Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Doo Sin Jo
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Na Yeon Park
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Yong Hwan Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Ha Jung Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Seong Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - So Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Hong Bae Jeon
- Stem Cell Institute, ENCell Co. Ltd., Seoul 06072, Korea;
| | - Hye-Won Na
- R&D Center AMOREPACIFIC Corporation, Yongin 17074, Gyeonggi-do, Korea; (H.-W.N.); (H.C.)
| | - Hyungjung Choi
- R&D Center AMOREPACIFIC Corporation, Yongin 17074, Gyeonggi-do, Korea; (H.-W.N.); (H.C.)
| | - Hong-Yeoul Ryu
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Zae Young Ryoo
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Hyun-Shik Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
| | - Dong-Hyung Cho
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (J.Y.C.); (J.B.K.); (D.S.J.); (N.Y.P.); (Y.H.K.); (H.J.L.); (S.H.K.); (S.H.K.); (H.-Y.R.); (Z.Y.R.); (H.-S.L.)
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea;
- Correspondence:
| |
Collapse
|