1
|
Grande G, Wu B, Wu J, Kalpouzos G, Laukka EJ, Bellander T, Rizzuto D. Long-Term Exposure to Ambient Particulate Matter and Structural Brain Changes in Older Adults. Stroke 2025. [PMID: 40353588 DOI: 10.1161/strokeaha.124.048096] [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: 06/07/2024] [Revised: 01/30/2025] [Accepted: 03/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Accumulating evidence links air pollution exposure to late-life cognitive deterioration. Whether air pollution alters brain structure remains poorly understood. Thus, we aimed to quantify the association between long-term exposure to particulate matter ≤2.5 µm and ≤10 µm (PM2.5 and PM10, respectively) and late-life brain structural changes. METHODS In the Swedish National Study on Aging and Care in Kungsholmen, Stockholm, 555 participants free from dementia underwent brain magnetic resonance imaging (MRI) scans at baseline and after 6 years (cohorts <78 years) or after 3 and 6 years (cohorts aged ≥78 years). After the exclusion of participants with neurological conditions (including previous stroke) and suboptimal MRI quality, we had 457 participants with available repeated MRI examinations, where total brain tissue volume, ventricles, hippocampus, and white matter hyperintensities volumes were assessed. PM2.5 and PM10 have been assessed since 1990 using dispersion models at residential addresses. Brain volumes have been standardized using baseline mean and SD. Long-term exposure to PM2.5 and PM10 in relation to the baseline and longitudinal brain MRI volumes were tested through multiadjusted (age, sex, educational level, smoking, socioeconomic status, and neighborhood household mean income) linear regression models. RESULTS At study entry, the mean (SD) age of the participants was 70 (SD, 8.9) years and 41% were males. Individuals who before baseline had been exposed to levels of PM2.5 or PM10 above the median (8.5 and 14.9 μg/m3, respectively) had smaller total brain tissue volume (β, -0.20 [95% CI, -0.33 to -0.06] and β, -0.14 [95% CI, -0.28 to -0.01], respectively) at baseline than those with lower PM2.5 and PM10 levels. Participants exposed during the follow-up to PM2.5>8.7 μg/m3 had on average an annual shrinkage of total brain tissue volume of 0.22 (95% CI, -0.43 to -0.01) and an annual increase of 0.25 (95% CI, 0.07-0.43) of the white matter hyperintensities as compared with participants exposed to PM2.5<8.7 μg/m3. No association was detected between PM10 and an annual rate of change in brain MRI volumes. CONCLUSIONS Long-term exposure to comparatively low levels of PM2.5 was associated with a greater load of structural brain changes, encompassing brain atrophy and vascular pathology. These findings, in a dementia- and cerebrovascular disease-free sample, underscore the importance of addressing air pollution as a modifiable risk factor for late-life cognitive decline, and highlight the need for targeted interventions to prevent its detrimental effects on brain integrity.
Collapse
Affiliation(s)
- Giulia Grande
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
- Stockholm Gerontology Research Centre, Sweden (G.G., E.J.L., D.R.)
| | - Bolin Wu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
| | - Jing Wu
- Institute of Environmental Medicine (IMM), Karolinska Institutet, Sweden (J.W., T.B.)
| | - Grégoria Kalpouzos
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
| | - Erika J Laukka
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
- Stockholm Gerontology Research Centre, Sweden (G.G., E.J.L., D.R.)
| | - Tom Bellander
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
- Institute of Environmental Medicine (IMM), Karolinska Institutet, Sweden (J.W., T.B.)
| | - Debora Rizzuto
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Sweden (G.G., B.W., G.K., E.J.L., T.B., D.R.)
- Stockholm Gerontology Research Centre, Sweden (G.G., E.J.L., D.R.)
| |
Collapse
|
2
|
Kalenik S, Zaczek A, Rodacka A. Air Pollution-Induced Neurotoxicity: The Relationship Between Air Pollution, Epigenetic Changes, and Neurological Disorders. Int J Mol Sci 2025; 26:3402. [PMID: 40244238 PMCID: PMC11989335 DOI: 10.3390/ijms26073402] [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: 03/18/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Air pollution is a major global health threat, responsible for over 8 million deaths in 2021, including 700,000 fatalities among children under the age of five. It is currently the second leading risk factor for mortality worldwide. Key pollutants, such as particulate matter (PM2.5, PM10), ozone, sulfur dioxide, nitrogen oxides, and carbon monoxide, have significant adverse effects on human health, contributing to respiratory and cardiovascular diseases, as well as neurodevelopmental and neurodegenerative disorders. Among these, particulate matter poses the most significant threat due to its highly complex mixture of organic and inorganic compounds with diverse sizes, compositions, and origins. Additionally, it can penetrate deeply into tissues and cross the blood-brain barrier, causing neurotoxicity which contributes to the development of neurodegenerative diseases. Although the link between air pollution and neurological disorders is well documented, the precise mechanisms and their sequence remain unclear. Beyond causing oxidative stress, inflammation, and excitotoxicity, studies suggest that air pollution induces epigenetic changes. These epigenetic alterations may affect the expression of genes involved in stress responses, neuroprotection, and synaptic plasticity. Understanding the relationship between neurological disorders and epigenetic changes induced by specific air pollutants could aid in the early detection and monitoring of central nervous system diseases.
Collapse
Affiliation(s)
- Sebastian Kalenik
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland; (S.K.); (A.Z.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, 21/23 Jana Matejki Street, 90-237 Lodz, Poland
| | - Agnieszka Zaczek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland; (S.K.); (A.Z.)
| | - Aleksandra Rodacka
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska Street, 90-236 Lodz, Poland; (S.K.); (A.Z.)
| |
Collapse
|
3
|
Zeng HX, Qin SJ, Andersson J, Li SP, Zeng QG, Li JH, Wu QZ, Meng WJ, Oudin A, Kanninen KM, Jalava P, Dong GH, Zeng XW. The emerging roles of particulate matter-changed non-coding RNAs in the pathogenesis of Alzheimer's disease: A comprehensive in silico analysis and review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125440. [PMID: 39631655 DOI: 10.1016/j.envpol.2024.125440] [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: 03/22/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Research on epigenetic‒environmental interactions in the development of Alzheimer's disease (AD) has accelerated rapidly in recent decades. Numerous studies have demonstrated the contribution of ambient particulate matter (PM) to the onset of AD. Emerging evidence indicates that non-coding RNAs (ncRNAs), including long non-coding RNAs, circular RNAs, and microRNAs, play a role in the pathophysiology of AD. In this review, we provide an overview of PM-altered ncRNAs in the brain, with emphasis on their potential roles in the pathogenesis of AD. These results suggest that these PM-altered ncRNAs are involved in the regulation of amyloid-beta pathology, microtubule-associated protein Tau pathology, synaptic dysfunction, damage to the blood‒brain barrier, microglial dysfunction, dysmyelination, and neuronal loss. In addition, we utilized in silico analysis to explore the biological functions of PM-altered ncRNAs in the development of AD. This review summarizes the knowns and unknowns of PM-altered ncRNAs in AD pathogenesis and discusses the current dilemma regarding PM-altered ncRNAs as promising biomarkers of AD. Altogether, this is the first thorough review of the connection between PM exposure and ncRNAs in AD pathogenesis, which may offer novel insights into the prevention, diagnosis, and treatment of AD associated with ambient PM exposure.
Collapse
Affiliation(s)
- Hui-Xian Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang-Jian Qin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | | | - Shen-Pan Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Guo Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hui Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Zhen Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen-Jie Meng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Anna Oudin
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Katja M Kanninen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Jalava
- Department of Environmental and Biological Science, University of Eastern Finland, Kuopio, Finland
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Teekaput C, Rachbundit C, Wantaneeyawong C, Teekaput K, Thiankhaw K. Impact of air pollution on the clinical exacerbation of central demyelinating disease: A 10-year data from the Northern Thailand MS and NMOSD registry. Mult Scler Relat Disord 2025; 94:106266. [PMID: 39817949 DOI: 10.1016/j.msard.2025.106266] [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: 05/20/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Particulate matter (PM) 2.5 (PM2.5) and PM10 are implicated in neurological diseases, yet their impact on central demyelinating diseases like multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) remains unclear. This study aimed to determine the association between the levels of PM2.5 and PM10 and the exacerbation of MS and NMOSD. METHODS Patients with clinical exacerbations of MS and NMOSD in the Northern Thailand Registry from 2013 to 2022 were enrolled. Eligible patients were categorized based on air pollution exposure (defined as PM2.5 > 15 μg/m3 and PM10 > 45 μg/m3) or no air pollution exposure. Outcomes assessed included clinical characteristics, Expanded Disability Status Scale (EDSS), and functional outcomes. RESULTS We analyzed 126 exacerbations in the PM2.5 database (mean age: 44.9±14.9 years, 114 NMOSD, 49 first exacerbations) and 135 exacerbations with the PM10 database (mean age: 44.9±14.9 years, 121 NMOSD, 54 first exacerbations), with the highest incidence four months post-peak air pollution. The PM2.5 exposure group had higher severity, showing increased rates of unfavorable EDSS at exacerbation and 90 days (56.3% vs. 23.6%, P < 0.001 and 47.9% vs. 16.4%, P < 0.001, respectively). Gadolinium enhancement in PM2.5 exposure was significantly higher (56.3% vs. 36.4%, P = 0.03). The PM2.5 exposure group also had a higher rate of second-line therapy with plasma exchange (21.1% vs. 7.3%, P = 0.03). PM2.5 exposure, not PM10, was associated with unfavorable EDSS at any time point, active radiological activity, risk of plasma exchange, and prolonged hospitalization. CONCLUSION Environmental pollution, especially PM2.5, significantly impacts MS and NMOSD patients, influencing disease severity, causing permanent disability, and prolonging hospitalization. A national policy on pollution control is imperative, and further data on long-term exposure, together with other pollutants, is still required.
Collapse
Affiliation(s)
- Chutithep Teekaput
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chanita Rachbundit
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chayasak Wantaneeyawong
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kanokkarn Teekaput
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
5
|
Thompson R, Tong X, Shen X, Ran J, Sun S, Yao XI, Shen C. Longitudinal associations between air pollution and incident dementia as mediated by MRI-measured brain volumes in the UK Biobank. ENVIRONMENT INTERNATIONAL 2025; 195:109219. [PMID: 39732110 DOI: 10.1016/j.envint.2024.109219] [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: 05/22/2024] [Revised: 10/04/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Although there is increasing evidence that environmental exposures are associated with the risk of neurodegenerative conditions, there is still limited mechanistic evidence evaluating potential mediators in human populations. METHODS UK Biobank is a large long-term study of 500,000 adults enrolled from 2006 to 2010 age 40-69 years. ICD-10 classified reports of dementia cases up to 2022 (Alzheimer's disease, vascular dementia, dementia in other classified diseases, and unspecified dementia) were identified from health record linkage. Estimates of residential air pollution, traffic noise, and greenspace exposure have been modelled. Structural brain MRI was conducted from 2014 to 2022, with brain volumes relevant to dementia identified a priori. Associations between environmental exposures, brain volumes, and dementia cases (diagnosed post-MRI) were tested using linear and logistic regression and adjusted for age, sex, household income, ethnicity, education, smoking, and area-level deprivation. Mediation of exposure-outcome associations by plausible brain volumes (those associated with both environmental exposure and dementia outcomes) were modelled using the quasi-Bayesian Monte Carlo method (N = 34,817-39,772). RESULTS Small but significant mediating effects (2%-8% of relationships mediated) were observed between PM2.5abs exposure and dementia risk by reduced total brain volume, NOx and Alzheimer's disease risk by reduced peripheral cortical grey matter, PM2.5abs and vascular dementia risk by reduced peripheral cortical grey matter, PM2.5abs and other dementia risk by reduced total grey matter, and PM10 and other dementia risk by reduced total grey matter. Greenspace and noise were not associated with dementia outcomes in the subset of the cohort providing brain imaging data. CONCLUSIONS This study adds to existing evidence of associations between environmental exposures and dementia outcomes. Our findings provide novel evidence that differences in brain volume may mediate these relationships. Future research is required to prove this mechanism and establish the other mechanisms through which exposure to air pollution might increase dementia risk.
Collapse
Affiliation(s)
- Rhiannon Thompson
- National Institute for Health Research (NIHR) School for Public Health Research (NIHR SPHR), UK; MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Xinning Tong
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, China
| | - Xueyi Shen
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Jinjun Ran
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengzhi Sun
- School of Public Health, Capital Medical University, Beijing, China
| | - Xiaoxin Iris Yao
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, China.
| | - Chen Shen
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK; National Institute for Health Research Health Protection Research Unit in Chemical and Radiation Threats and Hazards, School of Public Health, Imperial College London, UK.
| |
Collapse
|
6
|
Ru D, Zhang Z, Liu M, Fan X, Wang Y, Yan Y, Wang E. Downregulation of Notch Signaling-Stimulated Genes in Neurovascular Unit Alterations Induced by Chronic Cerebral Hypoperfusion. Immun Inflamm Dis 2024; 12:e70082. [PMID: 39607309 PMCID: PMC11603426 DOI: 10.1002/iid3.70082] [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: 07/06/2024] [Revised: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a key contributor to vascular cognitive impairment (VCI) and is typically associated with blood-brain barrier (BBB) damage. This study investigates the pathological mechanisms underlying CCH-induced neurovascular unit (NVU) alterations. METHODS A mouse model of CCH was established using the bilateral common carotid artery stenosis (BCAS) procedure. Decreased cerebral blood flow (CBF) and impaired BBB integrity were assessed. Brain microvessel (BMV)-specific transcriptome profiles were analyzed using RNA-seq, supplemented with published single-cell RNA-seq data. RESULTS RNA-seq revealed neuroinflammation-related gene activation and significant downregulation of Notch signaling pathway genes in BMVs post-BCAS. Upregulated differentially expressed genes (DEGs) were enriched in microglia/macrophages, while downregulated DEGs were prominent in endothelial cells and pericytes. Enhanced activation of vascular-associated microglia (VAM) was linked to neurovascular alterations. CONCLUSION CCH induces significant NVU changes, marked by microglia-associated neuroinflammation and Notch signaling downregulation. These insights highlight potential therapeutic targets for treating neuroinflammatory and vascular-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Dewen Ru
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zengyu Zhang
- Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Neurology, Minhang HospitalFudan UniversityShanghaiChina
| | - Meng Liu
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Xuhui Fan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yuqi Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yufeng Yan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Ersong Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Cho S, Park DH, Park EK, Bae JS. The beneficial effects of lupeol on particulate matter-mediated pulmonary inflammation. Food Chem Toxicol 2024; 191:114893. [PMID: 39067743 DOI: 10.1016/j.fct.2024.114893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/07/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Particulate matter (PM) poses significant health risks, especially fine particles (PM2.5) that can cause severe lung injuries. Lupeol, a phytosterol from medicinal plants, has potential anti-cancer properties. This study investigated lupeol's protective effects against PM2.5-induced lung damage. Mice received lupeol following intratracheal PM2.5 exposure. Results showed lupeol reduced lung damage, lowered wet/dry (W/D) weight ratio, and suppressed increased permeability caused by PM2.5. Additionally, lupeol decreased plasma inflammatory cytokines, total protein concentration in bronchoalveolar lavage fluid (BALF), and PM2.5-induced lymphocyte proliferation. Lupeol also reduced expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and autophagy-related proteins microtubule-associated protein 1 A/1 B-light chain 3 (LC3) II and Beclin 1, while increasing phosphorylated mammalian target of rapamycin (mTOR) phosphorylation. These findings suggest lupeol's potential as a therapeutic agent for PM2.5-induced lung damage via modulation of the TLR4-MyD88 and mTOR-autophagy pathways.
Collapse
Affiliation(s)
- Sanghee Cho
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, 41940, South Korea
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
8
|
Godoy‐Lugo JA, Thorwald MA, Cacciottolo M, D'Agostino C, Chakhoyan A, Sioutas C, Tanzi RE, Rynearson KD, Finch CE. Air pollution amyloidogenesis is attenuated by the gamma-secretase modulator GSM-15606. Alzheimers Dement 2024; 20:6107-6114. [PMID: 39132765 PMCID: PMC11497728 DOI: 10.1002/alz.14086] [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: 02/07/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Chronic air pollution (AirPoll) is associated with accelerated cognitive decline and risk of Alzheimer's disease (AD). Correspondingly, wild-type and AD-transgenic rodents exposed to AirPoll have increased amyloid peptides and behavioral impairments. METHODS We examined the γ-secretase modulator GSM-15606 for potential AirPoll protection by its attenuating of amyloid beta (Aβ)42 peptide production. Male and female wild-type mice were fed GSM-15606 during an 8-week inhalation exposure to AirPoll subfractions, ambient nanoparticulate matter (nPM), and diesel exhaust particles (DEP). RESULTS GSM-15606 decreased Aβ42 during nPM and DEP exposure without changing beta- or gamma-secretase activity or BACE1 and PS1 protein levels. DEP increased lateral ventricle volume by 25%. DISCUSSION These enzyme responses are relevant to AD drug treatments, as well as to the physiological functions of the Aβ42 peptide. GSM-15606 attenuation of Aβ42 may benefit human exposure to AirPoll. HIGHLIGHTS Gamma-secretase modulator (GSM-15606) attenuates the amyloidogenic amyloid beta (Aβ)42 peptide during exposure to air pollution, which may be a mechanism by which air pollution increases Alzheimer's disease (AD) risk. AD drug treatments may also consider Aβ homeostasis among the chronic effects of GSM-15606 and other amyloid reduction treatments on secretase enzymes.
Collapse
Affiliation(s)
- Jose A. Godoy‐Lugo
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Max A. Thorwald
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mafalda Cacciottolo
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Capricor Therapeutics, Inc.Beverly HillsCaliforniaUSA
| | - Carla D'Agostino
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ararat Chakhoyan
- Department of Physiology and NeuroscienceKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Zilkha Neurogenetic InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Constantinos Sioutas
- Civil and Environmental EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Rudolph E. Tanzi
- Department of NeurologyGenetics and Aging Research UnitMassachusetts General Hospital and Harvard Medical SchoolCharlestownMassachusettsUSA
- Henry and Allison McCance Center for Brain HealthMassachusetts General HospitalBostonMassachusettsUSA
| | - Kevin D. Rynearson
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Caleb E. Finch
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Dornsife CollegeUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
9
|
Sun X, Ma S, Guo Y, Chen C, Pan L, Cui Y, Chen Z, Dijkhuizen RM, Zhou Y, Boltze J, Yu Z, Li P. The association between air pollutant exposure and cerebral small vessel disease imaging markers with modifying effects of PRS-defined genetic susceptibility. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116638. [PMID: 38944013 DOI: 10.1016/j.ecoenv.2024.116638] [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: 04/09/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
Studies have highlighted a possible link between air pollution and cerebral small vessel disease (CSVD) imaging markers. However, the exact association and effects of polygenic risk score (PRS) defined genetic susceptibility remains unclear. This cross-sectional study used data from the UK Biobank. Participants aged 40-69 years were recruited between the year 2006 and 2010. The annual average concentrations of NOX, NO2, PM2.5, PM2.5-10, PM2.5 absorbance, and PM10, were estimated, and joint exposure to multiple air pollutants was reflected in the air pollution index (APEX). Air pollutant exposure was classified into the low (T1), intermediate (T2), and high (T3) tertiles. Three CSVD markers were used: white matter hyper-intensity (WMH), mean diffusivity (MD), and fractional anisotropy (FA). The first principal components of the MD and FA measures in the 48 white matter tracts were analysed. The sample consisted of 44,470 participants from the UK Biobank. The median (T1-T3) concentrations of pollutants were as follows: NO2, 25.5 (22.4-28.7) μg/m3; NOx, 41.3 (36.2-46.7) μg/m3; PM10, 15.9 (15.4-16.4) μg/m3; PM2.5, 9.9 (9.5-10.3) μg/m3; PM2.5 absorbance, 1.1 (1.0-1.2) per metre; and PM2.5-10, 6.1 (5.9-6.3) μg/m3. Compared with the low group, the high group's APEX, NOX, and PM2.5 levels were associated with increased WMH volumes, and the estimates (95 %CI) were 0.024 (0.003, 0.044), 0.030 (0.010, 0.050), and 0.032 (0.011, 0.053), respectively, after adjusting for potential confounders. APEX, PM10, PM2.5 absorbance, and PM2.5-10 exposure in the high group were associated with increased FA values compared to that in the low group. Sex-specific analyses revealed associations only in females. Regarding the combined associations of air pollutant exposure and PRS-defined genetic susceptibility with CSVD markers, the associations of NO2, NOX, PM2.5, and PM2.5-10 with WMH were more profound in females with low PRS-defined genetic susceptibility, and the associations of PM10, PM2.5, and PM2.5 absorbance with FA were more profound in females with higher PRS-defined genetic susceptibility. Our study demonstrated that air pollutant exposure may be associated with CSVD imaging markers, with females being more susceptible, and that PRS-defined genetic susceptibility may modify the associations of air pollutants.
Collapse
Affiliation(s)
- Xiaowei Sun
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shiyang Ma
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yunlu Guo
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Caiyang Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lijun Pan
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yidan Cui
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zengai Chen
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Yan Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK.
| | - Zhangsheng Yu
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Peiying Li
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands; Outcomes Research Consortium, Cleveland, OH, United States.
| |
Collapse
|
10
|
Finch CE, Thorwald MA. Inhaled Pollutants of the Gero-Exposome and Later-Life Health. J Gerontol A Biol Sci Med Sci 2024; 79:glae107. [PMID: 38644649 PMCID: PMC11170295 DOI: 10.1093/gerona/glae107] [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: 12/07/2023] [Indexed: 04/23/2024] Open
Abstract
Inhaled air pollutants (AirP) comprise extraordinarily diverse particles, volatiles, and gases from traffic, wildfire, cigarette smoke, dust, and various other sources. These pollutants contain numerous toxic components, which collectively differ in relative levels of components, but broadly share chemical classes. Exposure and health outcomes from AirP are complex, depending on pollutant source, duration of exposure, and socioeconomic status. We discuss examples in the current literature on organ responses to AirP, with a focus on lung, arteries, and brain. Some transcriptional responses are shared. It is well accepted that AirP contributes to Alzheimer's disease and other neurodegenerative conditions in the Gero-Exposome. However, we do not know which chemical compounds initiate these changes and how activation of these transcriptional pathways is further modified by genetics and prenatal development.
Collapse
Affiliation(s)
- Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
Bottenhorn KL, Sukumaran K, Cardenas-Iniguez C, Habre R, Schwartz J, Chen JC, Herting MM. Air pollution from biomass burning disrupts early adolescent cortical microarchitecture development. ENVIRONMENT INTERNATIONAL 2024; 189:108769. [PMID: 38823157 PMCID: PMC11878718 DOI: 10.1016/j.envint.2024.108769] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
Exposure to outdoor particulate matter (PM2.5) represents a ubiquitous threat to human health, and particularly the neurotoxic effects of PM2.5 from multiple sources may disrupt neurodevelopment. Studies addressing neurodevelopmental implications of PM exposure have been limited by small, geographically limited samples and largely focus either on macroscale cortical morphology or postmortem histological staining and total PM mass. Here, we leverage residentially assigned exposure to six, data-driven sources of PM2.5 and neuroimaging data from the longitudinal Adolescent Brain Cognitive Development Study (ABCD Study®), collected from 21 different recruitment sites across the United States. To contribute an interpretable and actionable assessment of the role of air pollution in the developing brain, we identified alterations in cortical microstructure development associated with exposure to specific sources of PM2.5 using multivariate, partial least squares analyses. Specifically, average annual exposure (i.e., at ages 8-10 years) to PM2.5 from biomass burning was related to differences in neurite development across the cortex between 9 and 13 years of age.
Collapse
Affiliation(s)
- Katherine L Bottenhorn
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA; Department of Psychology, Florida International University, Miami, FL, USA.
| | - Kirthana Sukumaran
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Carlos Cardenas-Iniguez
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Rima Habre
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA; Spatial Sciences Institute, University of Southern California, Los Angeles, CA, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jiu-Chiuan Chen
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Megan M Herting
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Bottenhorn KL, Sukumaran K, Cardenas-Iniguez C, Habre R, Schwartz J, Chen JC, Herting MM. Air pollution from biomass burning disrupts early adolescent cortical microarchitecture development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.21.563430. [PMID: 38798573 PMCID: PMC11118378 DOI: 10.1101/2023.10.21.563430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Exposure to outdoor particulate matter (PM 2.5 ) represents a ubiquitous threat to human health, and particularly the neurotoxic effects of PM 2.5 from multiple sources may disrupt neurodevelopment. Studies addressing neurodevelopmental implications of PM exposure have been limited by small, geographically limited samples and largely focus either on macroscale cortical morphology or postmortem histological staining and total PM mass. Here, we leverage residentially assigned exposure to six, data-driven sources of PM 2.5 and neuroimaging data from the longitudinal Adolescent Brain Cognitive Development Study (ABCD Study®), collected from 21 different recruitment sites across the United States. To contribute an interpretable and actionable assessment of the role of air pollution in the developing brain, we identified alterations in cortical microstructure development associated with exposure to specific sources of PM 2.5 using multivariate, partial least squares analyses. Specifically, average annual exposure (i.e., at ages 8-10 years) to PM 2.5 from biomass burning was related to differences in neurite development across the cortex between 9 and 13 years of age.
Collapse
|
13
|
Shkirkova K, Demetriou AN, Sizdahkhani S, Lamorie-Foote K, Zhang H, Morales M, Chen S, Zhao L, Diaz A, Godoy-Lugo JA, Zhou B, Zhang N, Li A, Mack WJ, Sioutas C, Thorwald MA, Finch CE, Pike C, Mack WJ. Microglial TLR4 Mediates White Matter Injury in a Combined Model of Diesel Exhaust Exposure and Cerebral Hypoperfusion. Stroke 2024; 55:1090-1093. [PMID: 38299349 PMCID: PMC10978264 DOI: 10.1161/strokeaha.124.046412] [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: 01/07/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Air pollution particulate matter exposure and chronic cerebral hypoperfusion (CCH) contribute to white matter toxicity through shared mechanisms of neuroinflammation, oxidative stress, and myelin breakdown. Prior studies showed that exposure of mice to joint particulate matter and CCH caused supra-additive injury to corpus callosum white matter. This study examines the role of TLR4 (toll-like receptor 4) signaling in mediating neurotoxicity and myelin damage observed in joint particulate matter and CCH exposures. METHODS Experiments utilized a novel murine model of inducible monocyte/microglia-specific TLR4 knockout (i-mTLR4-ko). Bilateral carotid artery stenosis (BCAS) was induced surgically to model CCH. TLR4-intact (control) and i-mTLR4-ko mice were exposed to 8 weeks of either aerosolized diesel exhaust particulate (DEP) or filtered air (FA) in 8 experimental groups: (1) control/FA (n=10), (2) control/DEP (n=10), (3) control/FA+BCAS (n=9), (4) control/DEP+BCAS (n=10), (5) i-mTLR4-ko/FA (n=9), (6) i-mTLR4-ko/DEP (n=8), (7) i-mTLR4-ko/FA+BCAS (n=8), and (8) i-mTLR4-ko/DEP+BCAS (n=10). Corpus callosum levels of 4-hydroxynonenal, 8-Oxo-2'-deoxyguanosine, Iba-1 (ionized calcium-binding adapter molecule 1), and dMBP (degraded myelin basic protein) were assayed via immunofluorescence to measure oxidative stress, neuroinflammation, and myelin breakdown, respectively. RESULTS Compared with control/FA mice, control/DEP+BCAS mice exhibited increased dMBP (41%; P<0.01), Iba-1 (51%; P<0.0001), 4-hydroxynonenal (100%; P<0.0001), and 8-Oxo-2'-deoxyguanosine (65%; P<0.05). I-mTLR4 knockout attenuated responses to DEP/BCAS for all markers. CONCLUSIONS i-mTLR4-ko markedly reduced neuroinflammation and oxidative stress and attenuated white matter degradation following DEP and CCH exposures. This suggests a potential role for targeting TLR4 signaling in individuals with vascular cognitive impairment, particularly those exposed to substantial ambient air pollution.
Collapse
Affiliation(s)
| | | | - Saman Sizdahkhani
- Zilkha Neurogenetic Institute, University of Southern California (USC)
| | | | | | - Manuel Morales
- Zilkha Neurogenetic Institute, University of Southern California (USC)
| | - Selena Chen
- Zilkha Neurogenetic Institute, University of Southern California (USC)
| | - Lifu Zhao
- Zilkha Neurogenetic Institute, University of Southern California (USC)
| | | | | | - Beryl Zhou
- Leonard Davis School of Gerontology, USC
| | | | - Andrew Li
- Leonard Davis School of Gerontology, USC
| | - Wendy J. Mack
- Keck School of Medicine, Department of Population and Public Health Sciences, USC
| | | | | | | | | | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California (USC)
| |
Collapse
|
14
|
Cho S, Choi HJ, Song GY, Bae JS. Therapeutic effects of hederacolchiside A1 on particulate matter-induced pulmonary injury. Toxicon 2024; 241:107650. [PMID: 38360299 DOI: 10.1016/j.toxicon.2024.107650] [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: 09/21/2023] [Revised: 01/21/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Particulate matter (PM) comprises a hazardous mixture of inorganic and organic particles that carry health risks. Inhaling fine PM particles with a diameter of ≤2.5 μm (PM2.5) can promote significant lung damage. Hederacolchiside A1 (HA1) exhibits notable in vivo antitumor effects against various solid tumors. However, our understanding of its therapeutic potential for individuals with PM2.5-induced lung injuries remains limited. Here, we explored the protective properties of HA1 against lung damage caused by PM2.5 exposure. HA1 was administered to the mice 30 min after intratracheal tail vein injection of PM2.5. Various parameters, such as changes in lung tissue wet/dry (W/D) weight ratio, total protein/total cell ratio, lymphocyte counts, inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF), vascular permeability, and histology, were assessed in mice exposed to PM2.5. Our data showed that HA1 mitigated lung damage, reduced the W/D weight ratio, and suppressed hyperpermeability caused by PM2.5 exposure. Moreover, HA1 effectively decreased plasma levels of inflammatory cytokines in those exposed to PM2.5, including tumor necrosis factor-α, interleukin-1β, and nitric oxide, while also lowering the total protein concentration in BALF and successfully alleviating PM2.5-induced lymphocytosis. Furthermore, HA1 significantly decreased the expression levels of toll-like receptor 4 (TLR4), myeloid differentiation primary response (MyD) 88, and autophagy-related proteins LC3 II and Beclin 1 but increased the protein phosphorylation of the mammalian target of rapamycin (mTOR). The anti-inflammatory characteristics of HA1 highlights its potential as a promising therapeutic agent for mitigating PM2.5-induced lung injuries by modulating the TLR4-MyD88 and mTOR-autophagy pathways.
Collapse
Affiliation(s)
- Sanghee Cho
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hui Ji Choi
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea
| | - Gyu Yong Song
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea.
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
15
|
Kalaria RN, Akinyemi RO, Paddick SM, Ihara M. Current perspectives on prevention of vascular cognitive impairment and promotion of vascular brain health. Expert Rev Neurother 2024; 24:25-44. [PMID: 37916306 PMCID: PMC10872925 DOI: 10.1080/14737175.2023.2273393] [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: 07/21/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
INTRODUCTION The true global burden of vascular cognitive impairment (VCI) is unknown. Reducing risk factors for stroke and cardiovascular disease would inevitably curtail VCI. AREAS COVERED The authors review current diagnosis, epidemiology, and risk factors for VCI. VCI increases in older age and by inheritance of known genetic traits. They emphasize modifiable risk factors identified by the 2020 Lancet Dementia Commission. The most profound risks for VCI also include lower education, cardiometabolic factors, and compromised cognitive reserve. Finally, they discuss pharmacological and non-pharmacological interventions. EXPERT OPINION By virtue of the high frequencies of stroke and cardiovascular disease the global prevalence of VCI is expectedly higher than prevalent neurodegenerative disorders causing dementia. Since ~ 90% of the global burden of stroke can be attributed to modifiable risk factors, a formidable opportunity arises to reduce the burden of not only stroke but VCI outcomes including progression from mild to the major in form of vascular dementia. Strict control of vascular risk factors and secondary prevention of cerebrovascular disease via pharmacological interventions will impact on burden of VCI. Non-pharmacological measures by adopting healthy diets and encouraging physical and cognitive activities and urging multidomain approaches are important for prevention of VCI and preservation of vascular brain health.
Collapse
Affiliation(s)
- Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Stella-Maria Paddick
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Centre, Osaka, Japan
| |
Collapse
|
16
|
Blanco MN, Shaffer RM, Li G, Adar SD, Carone M, Szpiro AA, Kaufman JD, Larson TV, Hajat A, Larson EB, Crane PK, Sheppard L. Traffic-related air pollution and dementia incidence in the Adult Changes in Thought Study. ENVIRONMENT INTERNATIONAL 2024; 183:108418. [PMID: 38185046 PMCID: PMC10873482 DOI: 10.1016/j.envint.2024.108418] [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: 10/13/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND While epidemiologic evidence links higher levels of exposure to fine particulate matter (PM2.5) to decreased cognitive function, fewer studies have investigated links with traffic-related air pollution (TRAP), and none have examined ultrafine particles (UFP, ≤100 nm) and late-life dementia incidence. OBJECTIVE To evaluate associations between TRAP exposures (UFP, black carbon [BC], and nitrogen dioxide [NO2]) and late-life dementia incidence. METHODS We ascertained dementia incidence in the Seattle-based Adult Changes in Thought (ACT) prospective cohort study (beginning in 1994) and assessed ten-year average TRAP exposures for each participant based on prediction models derived from an extensive mobile monitoring campaign. We applied Cox proportional hazards models to investigate TRAP exposure and dementia incidence using age as the time axis and further adjusting for sex, self-reported race, calendar year, education, socioeconomic status, PM2.5, and APOE genotype. We ran sensitivity analyses where we did not adjust for PM2.5 and other sensitivity and secondary analyses where we adjusted for multiple pollutants, applied alternative exposure models (including total and size-specific UFP), modified the adjustment covariates, used calendar year as the time axis, assessed different exposure periods, dementia subtypes, and others. RESULTS We identified 1,041 incident all-cause dementia cases in 4,283 participants over 37,102 person-years of follow-up. We did not find evidence of a greater hazard of late-life dementia incidence with elevated levels of long-term TRAP exposures. The estimated hazard ratio of all-cause dementia was 0.98 (95 % CI: 0.92-1.05) for every 2000 pt/cm3 increment in UFP, 0.95 (0.89-1.01) for every 100 ng/m3 increment in BC, and 0.96 (0.91-1.02) for every 2 ppb increment in NO2. These findings were consistent across sensitivity and secondary analyses. DISCUSSION We did not find evidence of a greater hazard of late-life dementia risk with elevated long-term TRAP exposures in this population-based prospective cohort study.
Collapse
Affiliation(s)
- Magali N Blanco
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Rachel M Shaffer
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Ge Li
- VA Northwest Network Mental Illness Research, Education, and Clinical Center, Virginia Puget Sound Health Care System, Seattle, WA, USA; Geriatric Research, Education, and Clinical Center, Virginia Puget Sound Health Care System, Seattle, WA, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Sara D Adar
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Adam A Szpiro
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Joel D Kaufman
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Timothy V Larson
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Civil & Environmental Engineering, University of Washington, Seattle, WA, USA
| | - Anjum Hajat
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lianne Sheppard
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Cho S, Park YJ, Bae JS. Therapeutic Effects of (+)-Afzelechin on Particulate Matter-Induced Pulmonary Injury. Biomol Ther (Seoul) 2024; 32:162-169. [PMID: 38148560 PMCID: PMC10762276 DOI: 10.4062/biomolther.2023.187] [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: 10/30/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/28/2023] Open
Abstract
Particulate matter (PM) constitutes a hazardous blend of organic and inorganic particles that poses health risks. Inhalation of fine airborne PM with a diameter of ≤ 2.5 μm (PM2.5) can lead to significant lung impairments. (+)-afzelechin (AZC), a natural compound sourced from Bergenia ligulata, boasts a range of attributes, including antioxidant, antimicrobial, anticancer, and cardiovascular effects. However, knowledge about the therapeutic potential of AZC for patients with PM2.5-induced lung injuries remains limited. Thus, in this study, we investigated the protective attributes of AZC against lung damage caused by PM2.5 exposure. AZC was administered to the mice 30 min after intratracheal instillation of PM2.5. Various parameters, such as changes in lung tissue wet/dry (W/D) weight ratio, total protein/total cell ratio, lymphocyte counts, levels of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), vascular permeability, and histology, were evaluated in mice exposed to PM2.5. Data demonstrated that AZC mitigated lung damage, reduced W/D weight ratio, and curbed hyperpermeability induced by PM2.5 exposure. Furthermore, AZC effectively lowered plasma levels of inflammatory cytokines produced by PM2.5 exposure. It reduced the total protein concentration in BALF and successfully alleviated PM2.5-induced lymphocytosis. Additionally, AZC substantially diminished the expression levels of Toll-like receptors 4 (TLR4), MyD88, and autophagy-related proteins LC3 II and Beclin 1. In contrast, it elevated the protein phosphorylation of the mammalian target of rapamycin (mTOR). Consequently, the anti-inflammatory attribute of AZC positions it as a promising therapeutic agent for mitigating PM2.5-induced lung injuries by modulating the TLR4-MyD88 and mTOR-autophagy pathways.
Collapse
Affiliation(s)
- Sanghee Cho
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yun Jin Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
18
|
Finch CE. Air pollution, dementia, and lifespan in the socio-economic gradient of aging: perspective on human aging for planning future experimental studies. FRONTIERS IN AGING 2023; 4:1273303. [PMID: 38034419 PMCID: PMC10683094 DOI: 10.3389/fragi.2023.1273303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/12/2023] [Indexed: 12/02/2023]
Abstract
Air pollution (AirPoll) accelerates human aging, as assessed by increased adult mortality and earlier onset of cardiovascular diseases, and dementia. Socio-economic strata (SES) of wealth and education have parallel differences of mortality and these diseases. Children from impoverished homes differ in brain development at birth and in risk of early fat excess and hypertension. To further enhance the healthspan, biogerontologists may consider a wider range of environmental exposures from gestation through later life morbidity that comprise the Gero-Exposome. Experimental studies with rodents and nematodes document shared transcriptional responses to AirPoll. In rodents, AirPoll exposure activates gene systems for body-wide detoxification through Nrf2 and NFkB transcription factors that mediate multiple aging processes. Gestational environmental factors include maternal diet and exposure to AirPoll and cigarette smoke. Correspondingly, gestational exposure of mice to AirPoll increased adult body fat, impaired glucose clearance, and decreased adult neurogenesis in the hippocampus, a brain region damaged in dementia. Nematode larvae also respond to AirPoll with Alzheimer relevant responses. These experimental approaches could identify to interventions for expanded human health and longevity across SES gradients.
Collapse
Affiliation(s)
- Caleb E. Finch
- Leonard Davis School of Gerontology and Dornsife College, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
20
|
Chakhoyan A, Shkrkova K, Sizdahkhani S, Huuskonen MT, Lamorie-Foote K, Diaz A, Chen S, Liu Q, D'Agostino C, Zhang H, Mack WJ, Sioutas C, Finch CE, Zlokovic B, Mack WJ. Magnetic resonance imaging of white matter response to diesel exhaust particles. RESEARCH SQUARE 2023:rs.3.rs-3087503. [PMID: 37503159 PMCID: PMC10371072 DOI: 10.21203/rs.3.rs-3087503/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Air pollution is associated with risks of dementia and accelerated cognitive decline. Rodent air pollution models have shown white matter vulnerability. This study uses diffusion tensor imaging (DTI) to quantify changes to white matter microstructure and tractography in multiple myelinated regions after exposure to diesel exhaust particulate (DEP). Adult C57BL/6 male mice were exposed to re-aerosolized DEP (NIST SRM 2975) at a concentration of 100 ug/m3 for 200 hours. Ex-vivo MRI analysis and fractional anisotropy (FA)-aided white matter tractography were conducted to study the effect of DEP exposure on the brain white matter tracts. Immunohistochemistry was used to assess myelin and axonal structure. DEP exposure for 8 weeks altered myelin composition in multiple regions. Diffusion tensor imaging (DTI) showed decreased FA in the corpus callosum (30%), external capsule (15%), internal capsule (15%), and cingulum (31 %). Separate immunohistochemistry analyses confirmed prior findings. Myelin basic protein (MBP) was decreased (corpus callosum: 28%, external capsule: 29%), and degraded MPB increased (corpus callosum: 32%, external capsule: 53%) in the DEP group. White matter is highly susceptible to chronic DEP exposure. This study demonstrates the utility of DTI as a neuroanatomical tool in the context of air pollution and white matter myelin vulnerability.
Collapse
Affiliation(s)
- Ararat Chakhoyan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Kristina Shkrkova
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Saman Sizdahkhani
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Mikko T Huuskonen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Arnold Diaz
- Leonard Davis School of Gerontology, University of Southern California
| | - Selena Chen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - Carla D'Agostino
- Leonard Davis School of Gerontology, University of Southern California
| | - Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California
| | - Wendy J Mack
- Department of Population and Public Health Sciences, Keck School of Medicine
| | | | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California
| | - Berislav Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | - William J Mack
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| |
Collapse
|
21
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [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: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
Collapse
|
22
|
Therapeutic Effects of Cornuside on Particulate Matter-Induced Lung Injury. Int J Mol Sci 2023; 24:ijms24054979. [PMID: 36902409 PMCID: PMC10002561 DOI: 10.3390/ijms24054979] [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: 02/02/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Particulate matter (PM) is a mixture comprising both organic and inorganic particles, both of which are hazardous to health. The inhalation of airborne PM with a diameter of ≤2.5 μm (PM2.5) can cause considerable lung damage. Cornuside (CN), a natural bisiridoid glucoside derived from the fruit of Cornus officinalis Sieb, exerts protective properties against tissue damage via controlling the immunological response and reducing inflammation. However, information regarding the therapeutic potential of CN in patients with PM2.5-induced lung injury is limited. Thus, herein, we examined the protective properties of CN against PM2.5-induced lung damage. Mice were categorized into eight groups (n = 10): a mock control group, a CN control group (0.8 mg/kg mouse body weight), four PM2.5+CN groups (0.2, 0.4, 0.6, and 0.8 mg/kg mouse body weight), and a PM2.5+CN group (0.2, 0.4, 0.6, and 0.8 mg/kg mouse body weight). The mice were administered with CN 30 min following intratracheal tail vein injection of PM2.5. In mice exposed to PM2.5, different parameters including changes in lung tissue wet/dry (W/D) lung weight ratio, total protein/total cell ratio, lymphocyte counts, inflammatory cytokine levels in the bronchoalveolar lavage fluid (BALF), vascular permeability, and histology were examined. Our findings revealed that CN reduced lung damage, the W/D weight ratio, and hyperpermeability caused by PM2.5. Moreover, CN reduced the plasma levels of inflammatory cytokines produced because of PM2.5 exposure, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide, as well as the total protein concentration in the BALF, and successfully attenuated PM2.5-associated lymphocytosis. In addition, CN substantially reduced the expression levels of Toll-like receptors 4 (TLR4), MyD88, and autophagy-related proteins LC3 II and Beclin 1, and increased protein phosphorylation of the mammalian target of rapamycin (mTOR). Thus, the anti-inflammatory property of CN renders it a potential therapeutic agent for treating PM2.5-induced lung injury by controlling the TLR4-MyD88 and mTOR-autophagy pathways.
Collapse
|
23
|
Zhu Z, Zhao X, Zhu L, Xiong Y, Cong S, Zhou M, Zhang M, Cheng M, Luo X. Effects of short-term waterfall forest aerosol air exposure on rat lung proteomics. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1223. [PMID: 36544689 PMCID: PMC9761115 DOI: 10.21037/atm-22-4813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
Background Chronic exposure to airborne microparticles has been shown to increase the incidence of several chronic diseases. Previous studies have found that waterfall forest aerosols contribute to a diminished immune stress response in patients with asthma. However, the specific effects of short-term waterfall forest aerosol exposure on lung proteins have not been fully elucidated. Methods This study used liquid chromatography-tandem mass spectrometry (LC-MS) to analyze changes in protein expression in the lungs of rats exposed to short-term waterfall forest aerosol environments. Specific protein markers were identified using bioconductivity analysis screening and validated using immunohistochemistry. Results Waterfall forest aerosol environment exposure on day 5 downregulated the expression of the classical inflammatory pathway nuclear factor κB (NF-κB) signaling pathway. As the waterfall forest aerosol environment increased due to the duration of exposure, it was involved in oxidative phosphorylation and then hormone signaling in lung cells from the very beginning. In contrast, at day 15 of exposure, there is an effect on the regulation of the immune-related high-affinity IgE receptor pathway. In addition, iron-sulfur Rieske protein (Uqcrfs1), mitochondrial Tu translation elongation factor (Tufm) and ribosomal protein L4 (Rpl4) were identified as possible bioindicators for the evaluation of air quality. Conclusions These results provide a comprehensive proteomic analysis that supports the positive contribution of a good air quality environment to lung health.
Collapse
Affiliation(s)
- Zixin Zhu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xueke Zhao
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lili Zhu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Xiong
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Shuo Cong
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Mingyu Zhou
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Manman Zhang
- Department of Gastroenterology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Mingliang Cheng
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xinhua Luo
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
24
|
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: 32] [Impact Index Per Article: 10.7] [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
|