1
|
Izard T. Environmental Toxicants and Their Disruption of Integrin Signaling in Lipid Rafts. Bioessays 2025; 47:e202400276. [PMID: 40012268 DOI: 10.1002/bies.202400276] [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: 11/21/2024] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
Talin, a key integrin activator, is essential for cellular adhesion, signal transduction, and mechanical stability. Its transition between autoinhibited and active conformations allows dynamic regulation of adhesion in response to environmental cues. Cholesterol-rich membrane microdomains, such as lipid rafts, organize and stabilize signaling platforms, influencing talin and integrin conformational states. Cholesterol is a switch modulating talin activation, integrin binding, and adhesion. Environmental pollutants, including heavy metals and air toxins, disrupt cholesterol homeostasis, destabilize lipid rafts, and interfere with talin-integrin interactions. These disruptions impair adhesion, tissue repair, and signaling fidelity, contributing to atherosclerosis and cancer metastasis. Understanding talin's interaction with cholesterol-rich domains offers critical insights into adhesion regulation and reveals the broader impact of environmental toxicants on cellular function. This framework emphasizes the importance of membrane composition, particularly cholesterol, in mediating the effects of environmental stressors and suggests potential therapeutic interventions.
Collapse
Affiliation(s)
- Tina Izard
- Cell Adhesion Laboratory, UF Scripps, Jupiter, Florida, USA
- The Skaggs Graduate School, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
2
|
Bentegeac R, Achour D, Grare C, Muntaner M, Gauthier V, Amouyel P, Matran R, Zerimech F, Lo Guidice JM, Dauchet L. Associations between air pollution and biomarkers of oxidative stress and lung damage in a large population-based sample of non-smoking adults in northern France. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2025; 47:166. [PMID: 40220195 PMCID: PMC11993482 DOI: 10.1007/s10653-025-02472-2] [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: 11/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Air pollution is an environmental risk factor associated with lung and cardiovascular disease that may be mediated by physiological pathways such as oxidative stress. Previous studies have identified associations between air pollution and biomarkers of oxidative stress (8-OHdG, 4-HNE, and fluorescent oxidation products (FOPs)), as well as lung health marker CC16, in younger and asthmatic populations. The objective of this study of a large population-based sample of non-smoking adults was to explore the relationship between long-term and short-term atmospheric pollution exposures and plasma or urine levels of these biomarkers. Our study was a post-hoc analysis of the cross-sectional ELISABET study from 2011 to 2013. We included non-smoking inhabitants of Lille, France from the ELISABET study. We assessed mean pluri-annual residential and short-term exposures to atmospheric pollution components (PM10, NO2, and O3) and collected several biomarkers (CC16, 8-OHdG, 4-HNE, and fluorescent oxidation products (FOPs)). We searched for associations between pollutants and biomarkers using log-linear robust multivariate regressions. Our work did not show any association between short- or long-term exposure to air pollution components and CC16, 8-OHdG, 4-HNE or FOP in a large (980 subjects) sample of Lille's general population, despite having sufficient statistical power to replicate previous findings of associations between air pollution and these biomarkers found in younger or asthmatic populations.
Collapse
Affiliation(s)
- Raphaël Bentegeac
- U1167 - RID-AGE, INSERM, Lille, France.
- Institut Pasteur de Lille, Lille, France.
- Lille University Hospital Center, Lille, France.
- Lille University, Lille, France.
| | - Djamal Achour
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- ULR 4483 - IMPECS, Lille University, Lille, France
| | - Céline Grare
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- ULR 4483 - IMPECS, Lille University, Lille, France
| | - Manon Muntaner
- U1167 - RID-AGE, INSERM, Lille, France
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- Lille University, Lille, France
| | - Victoria Gauthier
- U1167 - RID-AGE, INSERM, Lille, France
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- Lille University, Lille, France
| | - Philippe Amouyel
- U1167 - RID-AGE, INSERM, Lille, France
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- Lille University, Lille, France
| | - Regis Matran
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- ULR 4483 - IMPECS, Lille University, Lille, France
| | - Farid Zerimech
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- ULR 4483 - IMPECS, Lille University, Lille, France
| | - Jean-Marc Lo Guidice
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- ULR 4483 - IMPECS, Lille University, Lille, France
| | - Luc Dauchet
- U1167 - RID-AGE, INSERM, Lille, France
- Institut Pasteur de Lille, Lille, France
- Lille University Hospital Center, Lille, France
- Lille University, Lille, France
| |
Collapse
|
3
|
Giammona A, Terribile G, Rainone P, Pellizzer C, Porro D, Cerasa A, Sancini G, Rashid AU, Belloli S, Valtorta S, Lo Dico A, Bertoli G. Effects of particulate air pollution exposure on lung-brain axis and related miRNAs modulation in mouse models. Front Cell Dev Biol 2025; 13:1526424. [PMID: 40248351 PMCID: PMC12003928 DOI: 10.3389/fcell.2025.1526424] [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: 11/11/2024] [Accepted: 02/27/2025] [Indexed: 04/19/2025] Open
Abstract
Particulate matter exposure is linked to numerous health issues, including respiratory, cardiovascular, and neurodegenerative diseases. This review focuses on the biological mechanisms through which air pollution influences the lung-brain axis, highlighting the role of miRNAs in regulating gene pathways affected by PM. Some microRNAs (miRNAs) are identified as key modulators of cellular processes, including inflammation, epithelial-to-mesenchymal transition (EMT), and blood-brain barrier integrity. Using mice models to study these effects allows for controlled experimentation on the systemic distribution of PM across biological barriers. Among the imaging technologies, Positron Emission Tomography is the best approach to monitor the distribution and effects of PM in vivo. The research underscores the importance of miRNA profiles as potential markers for the health effects of PM exposure, suggesting that specific miRNAs could serve as early indicators of damage to the lung-brain axis.
Collapse
Affiliation(s)
- Alessandro Giammona
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Giulia Terribile
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Earth and Environmental Sciences, POLARIS Research Centre, University of Milano-Bicocca, Milano, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Paolo Rainone
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Chiara Pellizzer
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
| | - Danilo Porro
- PhD Program, Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milano, Italy
| | - Antonio Cerasa
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
| | - Giulio Sancini
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Earth and Environmental Sciences, POLARIS Research Centre, University of Milano-Bicocca, Milano, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Ameen-Ur Rashid
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
- PhD Program, Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milano, Italy
| | - Sara Belloli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Silvia Valtorta
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
- NeuroMI - Milan Centre for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Alessia Lo Dico
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Gloria Bertoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| |
Collapse
|
4
|
Coleman PD, Delvaux E, Kordower JH, Boehringer A, Huseby CJ. Massive changes in gene expression and their cause(s) can be a unifying principle in the pathobiology of Alzheimer's disease. Alzheimers Dement 2025; 21:e14555. [PMID: 39912452 PMCID: PMC11851168 DOI: 10.1002/alz.14555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/10/2024] [Accepted: 12/25/2024] [Indexed: 02/07/2025]
Abstract
Understanding of the biology of Alzheimer's disease (AD) has long been fragmented, with various investigators concentrating on amyloid beta (Aβ) or tau, inflammation, cell death pathways, misfolded proteins, glia, and more. Yet data from multiple authors has repeatedly shown altered expression of myriad genes related to these seemingly disparate phenomena. In 2022, Morgan et al. organized the massive data on changes in AD in a meticulous survey of the literature and related these changes to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Their data showed that 91% of the known KEGG pathways are involved in AD and that many of these pathways are represented by the known cellular/molecular phenomena of AD. Such data then raise the fundamental question: What mechanism(s) may be responsible for such widespread changes in gene expression? We review evidence for a unifying model based on sequestrations in stress granules and alteration of nucleocytoplasmic transport in AD. HIGHLIGHTS: In Alzheimer's disease (AD), critical changes take place in neurons before the appearance of plaques or tangles. Addressing these early changes provides a path to early detection and effective intervention in AD.
Collapse
Affiliation(s)
- Paul D. Coleman
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Elaine Delvaux
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Jeffrey H. Kordower
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Ashley Boehringer
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Carol J. Huseby
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| |
Collapse
|
5
|
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
|
6
|
Kritikos M, Zhou JW, Huang C, Gandy S, Pellecchia AC, Santiago-Michels S, Carr MA, Islam S, Yang Y, Horton MK, Lucchini RG, Franceschi AM, Bangiyev L, Vaska P, Clouston SA, Luft BJ. Exposure duration and cerebral amyloidosis in the olfactory cortex of World Trade Center responders: A positron emission tomography and magnetic resonance imaging study. J Alzheimers Dis 2025; 103:383-395. [PMID: 39610293 DOI: 10.1177/13872877241302350] [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] [Indexed: 11/30/2024]
Abstract
BACKGROUND Amyloid-β proteins, a hallmark of Alzheimer's disease, are believed to play an adaptive role in the cerebral immune response. OBJECTIVE Amyloid is believed to play a role in cerebral immune response and could play a similar role in response to air pollution exposures. In the present study, we examined whether WTC exposure duration was associated with cerebral amyloidosis in WTC responders. METHODS WTC responders (aged 44-65 years) who varied in exposure duration but did not use personalized protective equipment were assessed using positron-emission tomography with [18F]-Florbetaben. The outcome was the cortical [18F]-Florbetaben burden, measured using regional standardized uptake value ratios (SUVRs) in 34 Desikan-Killiany regions of interest. Spearman's ρ and generalized linear models were used to estimate correlations between WTC exposure duration and cortical [18F]-Florbetaben SUVR. Cognitive and behavioral symptoms were measured. Magnetic resonance imaging was used to measure cortical thickness and diffusivity. RESULTS The mean age of imaged responders was 56 years old. WTC exposure duration was associated with olfactory [18F]-Florbetaben SUVR (Spearman's ρ = 0.43, p = 0.011), which was in turn associated with elevated [18F]-Florbetaben SUVR in ventral regions (ρ = 0.41, p = 0.016). Cortical [18F]-Florbetaben in ventral regions was associated with reduced response speed (ρ = -0.72, p < 0.001), was co-located with cortical diffusivity across regions in the parietal and frontal lobes and reduced cortical thickness in the isthmus cingulate (ρ = -0.53, p = 0.001). CONCLUSIONS Low-grade amyloidosis in the olfactory and frontal lobes was associated with WTC exposure duration. Future work should examine whether low-grade amyloidosis is correlated with the location or distribution of neurofibrillary tangles in WTC responders.
Collapse
Affiliation(s)
- Minos Kritikos
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Juin-Wan Zhou
- Department of Biomedical Engineering, Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Chuan Huang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and Mount Sinai Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison C Pellecchia
- Stony Brook World Trade Center Health and Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | | | - Melissa A Carr
- Stony Brook World Trade Center Health and Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Shabab Islam
- Stony Brook World Trade Center Health and Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Yuan Yang
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Megan K Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto G Lucchini
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
- Department of Biomedical, Metabolic and Neurosciences, University of Modena, Modena MO, Italy
| | | | - Lev Bangiyev
- Department of Radiology, Renaissance School of Medicine at Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Paul Vaska
- Department of Biomedical Engineering, Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Sean Ap Clouston
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| | - Benjamin J Luft
- Stony Brook World Trade Center Health and Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Health Sciences Center, Stony Brook, NY, USA
| |
Collapse
|
7
|
Acharyya S, Kumar SH, Chouksey A, Soni N, Nazeer N, Mishra PK. The enigma of mitochondrial epigenetic alterations in air pollution-induced neurodegenerative diseases. Neurotoxicology 2024; 105:158-183. [PMID: 39374796 DOI: 10.1016/j.neuro.2024.10.002] [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: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The incidence of neurodegenerative diseases is a growing concern worldwide, affecting individuals from diverse backgrounds. Although these pathologies are primarily associated with aging and genetic susceptibility, their severity varies among the affected population. Numerous studies have indicated air pollution as a significant contributor to the increasing prevalence of neurodegeneration. Cohort studies have provided compelling evidence of the association between prolonged exposure to different air toxicants and cognitive decline, behavioural deficits, memory impairment, and overall neuronal health deterioration. Furthermore, molecular research has revealed that air pollutants can disrupt the body's protective mechanisms, participate in neuroinflammatory pathways, and cause neuronal epigenetic modifications. The mitochondrial epigenome is particularly interesting to the scientific community due to its potential to significantly impact our understanding of neurodegenerative diseases' pathogenesis and their release in the peripheral circulation. While protein hallmarks have been extensively studied, the possibility of using circulating epigenetic signatures, such as methylated DNA fragments, miRNAs, and genome-associated factors, as diagnostic tools and therapeutic targets requires further groundwork. The utilization of circulating epigenetic signatures holds promise for developing novel prognostic strategies, creating paramount point-of-care devices for disease diagnosis, identifying therapeutic targets, and developing clinical data-based disease models utilizing multi-omics technologies and artificial intelligence, ultimately mitigating the threat and prevalence of neurodegeneration.
Collapse
Affiliation(s)
- Sayanti Acharyya
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Sruthy Hari Kumar
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Apoorva Chouksey
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Nikita Soni
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Nazim Nazeer
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Pradyumna Kumar Mishra
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India; Faculty of Medical Research, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
8
|
Park HH, Armstrong MJ, Gorin FA, Lein PJ. Air Pollution as an Environmental Risk Factor for Alzheimer's Disease and Related Dementias. MEDICAL RESEARCH ARCHIVES 2024; 12:5825. [PMID: 39822906 PMCID: PMC11736697 DOI: 10.18103/mra.v12i10.5825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Alzheimer's disease and related dementias are a leading cause of morbidity in our aging populations. Although influenced by genetic factors, fewer than 5% of Alzheimer's disease and related dementia cases are due solely to genetic causes. There is growing scientific consensus that these dementias arise from complex gene by environment interactions. The 2020 Lancet Commission on dementia prevention, intervention, and care identified 12 modifiable risk factors of dementia, including lifestyle, educational background, comorbidities, and environmental exposures to environmental contaminants. In this review, we summarize the current understanding and data gaps regarding the role(s) of environmental pollutants in the etiology of Alzheimer's disease and related dementias with a focus on air pollution. In addition to summarizing findings from epidemiological and experimental animal studies that link airborne exposures to environmental contaminants to increased risk and/or severity of Alzheimer's disease and related dementias, we discuss currently hypothesized mechanism(s) underlying these associations, including peripheral inflammation, neuroinflammation and epigenetic changes. Key data gaps in this rapidly expanding investigative field and approaches for addressing these gaps are also addressed.
Collapse
Affiliation(s)
- Heui Hye Park
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Matthew J. Armstrong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Fredric A. Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, and Department of Neurology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
9
|
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
|
10
|
Olloquequi J, Díaz-Peña R, Verdaguer E, Ettcheto M, Auladell C, Camins A. From Inhalation to Neurodegeneration: Air Pollution as a Modifiable Risk Factor for Alzheimer's Disease. Int J Mol Sci 2024; 25:6928. [PMID: 39000036 PMCID: PMC11241587 DOI: 10.3390/ijms25136928] [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: 05/15/2024] [Revised: 06/13/2024] [Accepted: 06/22/2024] [Indexed: 07/14/2024] Open
Abstract
Air pollution, a growing concern for public health, has been linked to various respiratory and cardiovascular diseases. Emerging evidence also suggests a link between exposure to air pollutants and neurodegenerative diseases, particularly Alzheimer's disease (AD). This review explores the composition and sources of air pollutants, including particulate matter, gases, persistent organic pollutants, and heavy metals. The pathophysiology of AD is briefly discussed, highlighting the role of beta-amyloid plaques, neurofibrillary tangles, and genetic factors. This article also examines how air pollutants reach the brain and exert their detrimental effects, delving into the neurotoxicity of air pollutants. The molecular mechanisms linking air pollution to neurodegeneration are explored in detail, focusing on oxidative stress, neuroinflammation, and protein aggregation. Preclinical studies, including in vitro experiments and animal models, provide evidence for the direct effects of pollutants on neuronal cells, glial cells, and the blood-brain barrier. Epidemiological studies have reported associations between exposure to air pollution and an increased risk of AD and cognitive decline. The growing body of evidence supporting air pollution as a modifiable risk factor for AD underscores the importance of considering environmental factors in the etiology and progression of neurodegenerative diseases, in the face of worsening global air quality.
Collapse
Affiliation(s)
- Jordi Olloquequi
- Department of Biochemistry and Physiology, Physiology Section, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Roberto Díaz-Peña
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Ester Verdaguer
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Miren Ettcheto
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Carme Auladell
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Antoni Camins
- Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
11
|
Macé L, Brizais C, Bachelot F, Manoury A, Thomé S, Gloaguen C, Garali I, Magneron V, Monceau V, Sache A, Voyer F, Elie C, Roy L, Gensdarmes F, Klokov D, Block ML, Ibanez C. Exposure to tungsten particles via inhalation triggers early toxicity marker expression in the rat brain. Inhal Toxicol 2024; 36:261-274. [PMID: 38836331 DOI: 10.1080/08958378.2024.2349895] [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: 10/03/2023] [Accepted: 04/26/2024] [Indexed: 06/06/2024]
Abstract
OBJECTIVE Our work is focused on tungsten, considered as an emerging contaminant. Its environmental dispersion is partly due to mining and military activities. Exposure scenario can also be occupational, in areas such as the hard metal industry and specific nuclear facilities. Our study investigated the cerebral effects induced by the inhalation of tungsten particles. METHODS Inhalation exposure campaigns were carried out at two different concentrations (5 and 80 mg/m3) in single and repeated modes (4 consecutive days) in adult rats within a nose-only inhalation chamber. Processes involved in brain toxicity were investigated 24 h after exposure. RESULTS AND DISCUSSION Site-specific effects in terms of neuroanatomy and concentration-dependent changes in specific cellular actors were observed. Results obtained in the olfactory bulb suggest a potential early effect on the survival of microglial cells. Depending on the mode of exposure, these cells showed a decrease in density accompanied by an increase in an apoptotic marker. An abnormal phenotype of the nuclei of mature neurons, suggesting neuronal suffering, was also observed in the frontal cortex, and can be linked to the involvement of oxidative stress. The differential effects observed according to exposure patterns could involve two components: local (brain-specific) and/or systemic. Indeed, tungsten, in addition to being found in the lungs and kidneys, was present in the brain of animals exposed to the high concentration. CONCLUSION Our data question the perceived innocuity of tungsten relative to other metals and raise hypotheses regarding possible adaptive or neurotoxic mechanisms that could ultimately alter neuronal integrity.
Collapse
Affiliation(s)
- Léo Macé
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Chloé Brizais
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Florence Bachelot
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Annabelle Manoury
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Sébastien Thomé
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Céline Gloaguen
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Imène Garali
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Victor Magneron
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Virginie Monceau
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Amandine Sache
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Frédéric Voyer
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Christelle Elie
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - Laurence Roy
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| | - François Gensdarmes
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Sûreté Nucléaire, Service du Confinement et de l'Aérodispersion des Polluants, Gif-sur-YvetteCedex, France
| | - Dmitry Klokov
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Michelle L Block
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, IN University School of Medicine, Indianapolis, IN, USA
| | - Chrystelle Ibanez
- Institut de Radioprotection et de Sûreté Nucléaire, Pôle Santé Environnement, Service de recherche sur les effets biologiques et sanitaires des rayonnements ionisants, Avenue de la Division Leclerc, Fontenay aux Roses, France
| |
Collapse
|
12
|
O'Sharkey K, Meng Q, Mitra S, Paik SA, Liu J, Shen J, Thompson LK, Chow T, Su J, Cockburn M, Weichenthal S, Paulson SE, Jerrett M, Ritz B. Associations between brake and tire wear-related PM 2.5 metal components, particulate oxidative stress potential, and autism spectrum disorder in Southern California. ENVIRONMENT INTERNATIONAL 2024; 185:108573. [PMID: 38484609 DOI: 10.1016/j.envint.2024.108573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/13/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Air pollution is a global health concern, with fine particulate matter (PM2.5) constituents posing potential risks to human health, including children's neurodevelopment. Here we investigated associations between exposure during pregnancy and infancy to specific traffic-related PM2.5 components with Autism Spectrum Disorder (ASD) diagnosis. METHODS For exposure assessment, we estimated PM2.5 components related to traffic exposure (Barium [Ba] as a marker of brake dust and Zinc [Zn] as a tire wear marker, Black Carbon [BC]) and oxidative stress potential (OSP) markers (Hydroxyl Radical [OPOH] formation, Dithiothreitol activity [OPDTT], reactive oxygen species [ROS]) modeled with land use regression with co-kriging based on an intensive air monitoring campaign. We assigned exposures to a cohort of 444,651 children born in Southern California between 2016 and 2019, among whom 11,466 ASD cases were diagnosed between 2018 and 2022, Odds ratios (ORs) and 95% confidence intervals (CIs) were obtained with logistic regression for single pollutant and PM2.5 mass co-adjusted models, also adjusting for sociodemographic characteristics. RESULTS Among PM2.5 components, we found the strongest positive association with ASD for our brake wear marker Ba (ORper IQR = 1.29, 95 % CI: 1.24, 1.34). This was followed by an increased risk for all PM2.5 oxidative stress potential markers; the strongest association was with ROS formation (ORper IQR = 1.22, 95 % CI: 1.18, 1.25). PM2.5 mass was linked to ASD in Hispanic and Black children, but not White children, while traffic-related PM2.5 and OSP markers increased ASD risk across all groups. In neighborhoods with the lowest socioeconomic status (SES), associations with ASD were stronger for all examined pollutants compared to higher SES areas. CONCLUSIONS Our findings suggest that brake wear-related PM2.5 and PM2.5 OSP are associated with ASD diagnosis in Southern California. These results suggest that strategies aimed at reducing the public health impacts of PM2.5 need to consider specific sources.
Collapse
Affiliation(s)
- Karl O'Sharkey
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qi Meng
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sanjali Mitra
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Seung-A Paik
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan Liu
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiaqi Shen
- Department of Atmospheric & Oceanic Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Laura K Thompson
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA 90032, USA
| | - Ting Chow
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jason Su
- Department of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Myles Cockburn
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA 90032, USA
| | - Scott Weichenthal
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec H3A0G4, Canada
| | - Susanne E Paulson
- Department of Atmospheric & Oceanic Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Jerrett
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
13
|
Manzano-Covarrubias AL, Yan H, Luu MDA, Gadjdjoe PS, Dolga AM, Schmidt M. Unravelling the signaling power of pollutants. Trends Pharmacol Sci 2023; 44:917-933. [PMID: 37783643 DOI: 10.1016/j.tips.2023.09.002] [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: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
Exposure to environmental pollutants contributes to diverse pathologies, including pulmonary disease, lower respiratory infections, cancer, and stroke. Pollutants' entry can occur through inhalation, traversing endothelial and epithelial barriers, and crossing the blood-brain barrier, leading to a wide distribution throughout the human body via systemic circulation. Pollutants cause cellular damage by multiple mechanisms encompassing oxidative stress, mitochondrial dysfunction, (neuro)inflammation, and protein instability/proteotoxicity. Sensing pollutants has added a new dimension to disease progression and drug failure. Understanding the molecular pathways and potential receptor binding/signaling that underpin 'sensing' could contribute to ways to combat the detrimental effects of pollutants. We highlight key points of pollutant signaling, crosstalk with receptors acting as drug targets for chronic diseases, and discuss the potential for future therapeutics.
Collapse
Affiliation(s)
- Ana L Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Phoeja S Gadjdjoe
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
14
|
Marques-da-Silva D, Lagoa R. Rafting on the Evidence for Lipid Raft-like Domains as Hubs Triggering Environmental Toxicants' Cellular Effects. Molecules 2023; 28:6598. [PMID: 37764374 PMCID: PMC10536579 DOI: 10.3390/molecules28186598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The plasma membrane lipid rafts are cholesterol- and sphingolipid-enriched domains that allow regularly distributed, sub-micro-sized structures englobing proteins to compartmentalize cellular processes. These membrane domains can be highly heterogeneous and dynamic, functioning as signal transduction platforms that amplify the local concentrations and signaling of individual components. Moreover, they participate in cell signaling routes that are known to be important targets of environmental toxicants affecting cell redox status and calcium homeostasis, immune regulation, and hormonal functions. In this work, the evidence that plasma membrane raft-like domains operate as hubs for toxicants' cellular actions is discussed, and suggestions for future research are provided. Several studies address the insertion of pesticides and other organic pollutants into membranes, their accumulation in lipid rafts, or lipid rafts' disruption by polychlorinated biphenyls (PCBs), benzo[a]pyrene (B[a]P), and even metals/metalloids. In hepatocytes, macrophages, or neurons, B[a]P, airborne particulate matter, and other toxicants caused rafts' protein and lipid remodeling, oxidative changes, or amyloidogenesis. Different studies investigated the role of the invaginated lipid rafts present in endothelial cells in mediating the vascular inflammatory effects of PCBs. Furthermore, in vitro and in vivo data strongly implicate raft-localized NADPH oxidases, the aryl hydrocarbon receptor, caveolin-1, and protein kinases in the toxic mechanisms of occupational and environmental chemicals.
Collapse
Affiliation(s)
- Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
15
|
Lamorie‐Foote K, Liu Q, Shkirkova K, Ge B, He S, Morgan TE, Mack WJ, Sioutas C, Finch CE, Mack WJ. Particulate matter exposure and chronic cerebral hypoperfusion promote oxidative stress and induce neuronal and oligodendrocyte apoptosis in male mice. J Neurosci Res 2023; 101:384-402. [PMID: 36464774 PMCID: PMC10107949 DOI: 10.1002/jnr.25153] [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: 01/06/2022] [Revised: 10/16/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) may amplify the neurotoxicity of nanoscale particulate matter (nPM), resulting in white matter injury. This study characterized the joint effects of nPM (diameter ≤ 200 nm) and CCH secondary to bilateral carotid artery stenosis (BCAS) exposure on neuronal and white matter injury in a murine model. nPM was collected near a highway and re-aerosolized for exposure. Ten-week-old C57BL/6 male mice were randomized into four groups: filtered air (FA), nPM, FA + BCAS, and nPM + BCAS. Mice were exposed to FA or nPM for 10 weeks. BCAS surgeries were performed. Markers of inflammation, oxidative stress, and apoptosis were examined. nPM + BCAS exposure increased brain hemisphere TNFα protein compared to FA. iNOS and HNE immunofluorescence were increased in the corpus callosum and cerebral cortex of nPM + BCAS mice compared to FA. While nPM exposure alone did not decrease cortical neuronal cell count, nPM decreased corpus callosum oligodendrocyte cell count. nPM exposure decreased mature oligodendrocyte cell count and increased oligodendrocyte precursor cell count in the corpus callosum. nPM + BCAS mice exhibited a 200% increase in cortical neuronal TUNEL staining and a 700% increase in corpus callosum oligodendrocyte TUNEL staining compared to FA. There was a supra-additive interaction between nPM and BCAS on cortical neuronal TUNEL staining (2.6× the additive effects of nPM + BCAS). nPM + BCAS exposure increased apoptosis, neuroinflammation, and oxidative stress in the cerebral cortex and corpus callosum. nPM + BCAS exposure increased neuronal apoptosis above the separate responses to each exposure. However, oligodendrocytes in the corpus callosum demonstrated a greater susceptibility to the combined neurotoxic effects of nPM + BCAS exposure.
Collapse
Affiliation(s)
- Krista Lamorie‐Foote
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Neurological Surgery, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Qinghai Liu
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kristina Shkirkova
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Brandon Ge
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Shannon He
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Todd E. Morgan
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Wendy J. Mack
- Department of Population and Public Health SciencesUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Caleb E. Finch
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - William J. Mack
- Zilkha Neurogenetic InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Neurological Surgery, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
16
|
Kritikos M, Diminich ED, Meliker J, Mielke M, Bennett DA, Finch CE, Gandy SE, Carr MA, Yang X, Kotov R, Kuan P, Bromet EJ, Clouston SAP, Luft BJ. Plasma amyloid beta 40/42, phosphorylated tau 181, and neurofilament light are associated with cognitive impairment and neuropathological changes among World Trade Center responders: A prospective cohort study of exposures and cognitive aging at midlife. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12409. [PMID: 36911360 PMCID: PMC9994167 DOI: 10.1002/dad2.12409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/09/2023] [Accepted: 01/19/2023] [Indexed: 03/14/2023]
Abstract
Introduction World Trade Center (WTC) responders are experiencing a high risk of mild cognitive impairment (MCI) and dementia, though the etiology remains inadequately characterized. This study investigated whether WTC exposures and chronic post-traumatic stress disorder (PTSD) were correlated with plasma biomarkers characteristic of Alzheimer's disease (AD) neuropathology. Methods Eligible participants included WTC-exposed individuals with a baseline cognitive assessment and available plasma sample. We examined levels of the amyloid beta (Aβ)40/42 ratio, phosphorylated tau 181 (p-tau181), and neurofilament light chain (NfL) and associations with a WTC exposures (duration on site ≥15 weeks, dust cloud), the PTSD Symptom Checklist for Diagnostic and Statistical Manual of Mental Disorders, 4th edition PTSD, and classification of amyloid/tau/neurodegeneration (AT[N]) profiles. Multinomial logistic regressions assessed whether biomarkers predicted increased risk of MCI or dementia. Results Of 1179 eligible responders, 93.0% were male, mean (standard deviation) age 56.6 years (7.8). Aβ40/42, p-tau181, and NfL intercorrelated and increased with age. In subgroup analyses of responders with available neuroimaging data (n = 75), Aβ40/42 and p-tau181 were further associated with decreased hippocampal volume (Spearman's ρ = -0.3). Overall, 58.08% of responders with dementia had ≥1 elevated biomarker, and 3.45% had elevations across all biomarkers. In total, 248 (21.05%) had MCI and 70 (5.94%) had dementia. Increased risk of dementia was associated with plasma AT(N) profile T+ or A+N+. Exposure on site ≥15 weeks was independently associated with T+ (adjusted risk ratio [aRR] = 1.03 [1.01-1.05], P = 0.009), and T+N+ profile (aRR = 2.34 [1.12-4.87]). The presence of PTSD was independently associated with risk of A+ (aRR = 1.77 [1.11-2.82]). Discussion WTC exposures and chronic PTSD are associated with plasma biomarkers consistent with neurodegenerative disease.
Collapse
Affiliation(s)
- Minos Kritikos
- Program in Public Health and Department of FamilyPopulation, and Preventive MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Erica D. Diminich
- Program in Public Health and Department of FamilyPopulation, and Preventive MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Jaymie Meliker
- Program in Public Health and Department of FamilyPopulation, and Preventive MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Michelle Mielke
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush UniversityChicagoIllinoisUSA
| | - Caleb E. Finch
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Sam E. Gandy
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Psychiatry and Mount Sinai Alzheimer's Disease Research CenterIcahn School of Medicine, Mount SinaiNew YorkNew YorkUSA
| | - Melissa A. Carr
- Department of MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Xiaohua Yang
- Department of MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Roman Kotov
- Department of PsychiatryRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Pei‐Fen Kuan
- Department of Applied MathematicsRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Evelyn J. Bromet
- Department of PsychiatryRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Sean A. P. Clouston
- Program in Public Health and Department of FamilyPopulation, and Preventive MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| | - Benjamin J. Luft
- Department of MedicineRenaissance School of Medicine at Stony Brook UniversityStony BrookNew YorkUSA
| |
Collapse
|
17
|
Franz CE, Gustavson DE, Elman JA, Fennema-Notestine C, Hagler DJ, Baraff A, Tu XM, Wu TC, DeAnda J, Beck A, Kaufman JD, Whitsel N, Finch CE, Chen JC, Lyons MJ, Kremen WS. Associations Between Ambient Air Pollution and Cognitive Abilities from Midlife to Early Old Age: Modification by APOE Genotype. J Alzheimers Dis 2023; 93:193-209. [PMID: 36970897 PMCID: PMC10827529 DOI: 10.3233/jad-221054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
BACKGROUND Fine particulate matter (PM2.5) and nitrogen dioxide (NO2) measures of ambient air pollution are associated with accelerated age-related cognitive impairment, and Alzheimer's disease and related dementias (ADRD). OBJECTIVE We examined associations between air pollution, four cognitive factors, and the moderating role of apolipoprotein E (APOE) genotype in the understudied period of midlife. METHODS Participants were ∼1,100 men in the Vietnam Era Twin Study of Aging. Baseline cognitive assessments were from 2003 to 2007. Measures included past (1993-1999) and recent (3 years prior to baseline assessment) PM2.5 and NO2 exposure, in-person assessment of episodic memory, executive function, verbal fluency, and processing speed, and APOE genotype. Average baseline age was 56 years with a 12-year follow-up. Analyses adjusted for health and lifestyle covariates. RESULTS Performance in all cognitive domains declined from age 56 to 68. Higher PM2.5 exposures were associated with worse general verbal fluency. We found significant exposure-by-APOE genotype interactions for specific cognitive domains: PM2.5 with executive function and NO2 with episodic memory. Higher PM2.5 exposure was related to worse executive function in APOE ɛ4 carriers, but not in non-carriers. There were no associations with processing speed. CONCLUSION These results indicate negative effects of ambient air pollution exposure on fluency alongside intriguing differential modifications of cognitive performance by APOE genotype. APOE ɛ4 carriers appeared more sensitive to environmental differences. The process by which air pollution and its interaction with genetic risk for ADRD affects risk for later life cognitive decline or progression to dementia may begin in midlife.
Collapse
Affiliation(s)
- Carol E. Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
| | - Daniel E. Gustavson
- Institute for Behavior Genetics, University of Colorado Boulder, Boulder, CO
| | - Jeremy A. Elman
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
| | - Christine Fennema-Notestine
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
- Department of Radiology, University of California, San Diego, La Jolla, CA
| | - Donald J. Hagler
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
- Department of Radiology, University of California, San Diego, La Jolla, CA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Aaron Baraff
- Vietnam Era Twin Registry, VA Puget Sound Health Care, Seattle, WA
| | - Xin M. Tu
- Herbert Wertheim School of Public Health & Human Longevity Science, University of California San Diego, CA
| | - Tsung-Chin Wu
- Herbert Wertheim School of Public Health & Human Longevity Science, University of California San Diego, CA
| | - Jaden DeAnda
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
- Department of Psychology, San Diego State University, San Diego, CA
| | - Asad Beck
- Graduate Program in Neuroscience, University of Washington, Seattle, WA
| | - Joel D. Kaufman
- Epidemiology, Environmental and Occupational Health Sciences, and General Internal Medicine, University of Washington, Seattle, WA
| | - Nathan Whitsel
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Jiu-Chiuan Chen
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA
| | - Michael J. Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA
| | - William S. Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA
| |
Collapse
|
18
|
Long-term particulate matter 2.5 exposure and dementia: a systematic review and meta-analysis. Public Health 2022; 212:33-41. [DOI: 10.1016/j.puhe.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/26/2022] [Accepted: 08/16/2022] [Indexed: 11/11/2022]
|
19
|
Lee SH, Lin CY, Chen TF, Chou CCK, Chiu MJ, Tee BL, Liang HJ, Cheng TJ. Distinct brain lipid signatures in response to low-level PM 2.5 exposure in a 3xTg-Alzheimer's disease mouse inhalation model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156456. [PMID: 35660587 DOI: 10.1016/j.scitotenv.2022.156456] [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: 02/22/2022] [Revised: 05/11/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) poses a significant risk to human health. The molecular mechanisms underlying low-level PM2.5-induced neurotoxicity in the central nervous system remain unclear. In addition, changes in lipids in response to PM2.5 exposure have not yet been fully elucidated. In this study, 3xTg-Alzheimer's disease (AD) mice experienced continuous whole-body exposure to non-concentrated PM2.5 for three consecutive months, while control mice inhaled particulate matter-filtered air over the same time span. A liquid chromatography-mass spectrometry-based lipidomic platform was used to determine the distinct lipid profiles of various brain regions. The average PM2.5 concentration during the exposure was 11.38 μg/m3, which was close to the regulation limits of USA and Taiwan. The partial least squares discriminant analysis model showed distinct lipid profiles in the cortex, hippocampus, and olfactory bulb, but not the cerebellum, of mice in the exposure group. Increased levels of fatty acyls, glycerolipids, and sterol lipids, as well as the decreased levels of glycerophospholipids and sphingolipids in PM2.5-exposed mouse brains may be responsible for the increased energy demand, membrane conformation, neuronal loss, antioxidation, myelin function, and cellular signaling pathways associated with AD development. Our research suggests that subchronic exposure to low levels of PM2.5 may cause neurotoxicity by changing the lipid profiles in a susceptible model. Lipidomics is a powerful tool to study the early effects of PM2.5-induced AD toxicity.
Collapse
Affiliation(s)
- Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ching-Yu Lin
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Charles C-K Chou
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA, USA
| | - Hao-Jan Liang
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
20
|
O'Piela DR, Durisek GR, Escobar YNH, Mackos AR, Wold LE. Particulate matter and Alzheimer's disease: an intimate connection. Trends Mol Med 2022; 28:770-780. [PMID: 35840480 PMCID: PMC9420776 DOI: 10.1016/j.molmed.2022.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/09/2022] [Accepted: 06/10/2022] [Indexed: 10/17/2022]
Abstract
The environmental role in disease progression has been appreciated for decades; however, understanding how airborne toxicant exposure can affect organs beyond the lungs is an underappreciated area of scientific inquiry. Particulate matter (PM) includes various gases, liquids, and particles in suspension and is produced by industrial activities such as fossil fuel combustion and natural events including wildfires and volcanic eruptions. Although agencies have attempted to reduce acceptable airborne particulate levels, with urbanization and population growth, these policies have been only moderately effective in mitigating disease progression. A growing area of research is focused on the role of PM exposure in the progression of Alzheimer's disease (AD). This review will summarize the knowns and unknowns of this expanding field.
Collapse
Affiliation(s)
- Devin R O'Piela
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - George R Durisek
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Yael-Natalie H Escobar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Amy R Mackos
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
21
|
Shkirkova K, Lamorie-Foote K, Zhang N, Li A, Diaz A, Liu Q, Thorwald MA, Godoy-Lugo JA, Ge B, D'Agostino C, Zhang Z, Mack WJ, Sioutas C, Finch CE, Mack WJ, Zhang H. Neurotoxicity of Diesel Exhaust Particles. J Alzheimers Dis 2022; 89:1263-1278. [PMID: 36031897 DOI: 10.3233/jad-220493] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Air pollution particulate matter (PM) is strongly associated with risks of accelerated cognitive decline, dementia and Alzheimer's disease. Ambient PM batches have variable neurotoxicity by collection site and season, which limits replicability of findings within and between research groups for analysis of mechanisms and interventions. Diesel exhaust particles (DEP) offer a replicable model that we define in further detail. OBJECTIVE Define dose- and time course neurotoxic responses of mice to DEP from the National Institute of Science and Technology (NIST) for neurotoxic responses shared by DEP and ambient PM. METHODS For dose-response, adult C57BL/6 male mice were exposed to 0, 25, 50, and 100μg/m3 of re-aerosolized DEP (NIST SRM 2975) for 5 h. Then, mice were exposed to 100μg/m3 DEP for 5, 100, and 200 h and assayed for amyloid-β peptides, inflammation, oxidative damage, and microglial activity and morphology. RESULTS DEP exposure at 100μg/m3 for 5 h, but not lower doses, caused oxidative damage, complement and microglia activation in cerebral cortex and corpus callosum. Longer DEP exposure for 8 weeks/200 h caused further oxidative damage, increased soluble Aβ, white matter injury, and microglial soma enlargement that differed by cortical layer. CONCLUSION Exposure to 100μg/m3 DEP NIST SRM 2975 caused robust neurotoxic responses that are shared with prior studies using DEP or ambient PM0.2. DEP provides a replicable model to study neurotoxic mechanisms of ambient PM and interventions relevant to cognitive decline and dementia.
Collapse
Affiliation(s)
- Kristina Shkirkova
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Krista Lamorie-Foote
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nathan Zhang
- Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Andrew Li
- Dornsife College, University of Southern California, Los Angeles, CA, USA
| | - Arnold Diaz
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Qinghai Liu
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jose A Godoy-Lugo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Brandon Ge
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carla D'Agostino
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Zijiao Zhang
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Wendy J Mack
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Constantinos Sioutas
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Dornsife College, University of Southern California, Los Angeles, CA, USA.,Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - William J Mack
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Song J, Han K, Wang Y, Qu R, Liu Y, Wang S, Wang Y, An Z, Li J, Wu H, Wu W. Microglial Activation and Oxidative Stress in PM2.5-Induced Neurodegenerative Disorders. Antioxidants (Basel) 2022; 11:antiox11081482. [PMID: 36009201 PMCID: PMC9404971 DOI: 10.3390/antiox11081482] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Fine particulate matter (PM2.5) pollution remains a prominent environmental problem worldwide, posing great threats to human health. The adverse effects of PM2.5 on the respiratory and cardiovascular systems have been extensively studied, while its detrimental effects on the central nervous system (CNS), specifically neurodegenerative disorders, are less investigated. Neurodegenerative disorders are characterized by reduced neurogenesis, activated microglia, and neuroinflammation. A variety of studies involving postmortem examinations, epidemiological investigations, animal experiments, and in vitro cell models have shown that PM2.5 exposure results in neuroinflammation, oxidative stress, mitochondrial dysfunction, neuronal apoptosis, and ultimately neurodegenerative disorders, which are strongly associated with the activation of microglia. Microglia are the major innate immune cells of the brain, surveilling and maintaining the homeostasis of CNS. Upon activation by environmental and endogenous insults, such as PM exposure, microglia can enter an overactivated state that is featured by amoeboid morphology, the over-production of reactive oxygen species, and pro-inflammatory mediators. This review summarizes the evidence of microglial activation and oxidative stress and neurodegenerative disorders following PM2.5 exposure. Moreover, the possible mechanisms underlying PM2.5-induced microglial activation and neurodegenerative disorders are discussed. This knowledge provides certain clues for the development of therapies that may slow or halt the progression of neurodegenerative disorders induced by ambient PM.
Collapse
Affiliation(s)
- Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Keyang Han
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Ya Wang
- Nursing School, Zhenjiang College, Zhenjiang 212028, China;
| | - Rongrong Qu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Yuan Liu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Shaolan Wang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Yinbiao Wang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Hui Wu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
- Correspondence:
| |
Collapse
|
23
|
Garcia Manriquez BA, Kikis EA. Genetic background modulates the sensitivity of Caenorhabditis elegans to the toxic effects of vehicular air pollution. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000613. [PMID: 35903775 PMCID: PMC9315412 DOI: 10.17912/micropub.biology.000613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/07/2022]
Abstract
Vehicular air pollution is an environmental toxicant that can have several health consequences, such as decreased respiratory and cardiovascular function and an increased incidence of age-related dementia and neurodegenerative diseases such as Alzheimer's disease. C. elegans has been previously shown to be a valuable animal model to study the effects of air pollution due to its tendency to respond to acute exposure to nano-sized particulate matter (nPM) produced by vehicular emissions. Specifically, nPM causes delayed development resulting in smaller animal size and induction of the SKN-1-mediated oxidative stress response. Here we show that various wild isolates demonstrate differential susceptibility to nPM, as measured by body size. Specifically, the Hawaiian isolate, CB4856, displayed the highest sensitivity, equivalent to its sensitivity to the potent oxidant paraquat. The findings described herein suggest that C. elegans may be a useful genetic tool for identifying nPM-susceptibility loci.
Collapse
Affiliation(s)
| | - Elise A. Kikis
- Biology Department, The University of the South, Sewanee, Tennessee, USA
,
Correspondence to: Elise A. Kikis (
)
| |
Collapse
|
24
|
Raichlen DA, Furlong M, Klimentidis YC, Sayre MK, Parra KL, Bharadwaj PK, Wilcox RR, Alexander GE. Association of Physical Activity with Incidence of Dementia Is Attenuated by Air Pollution. Med Sci Sports Exerc 2022; 54:1131-1138. [PMID: 35704438 PMCID: PMC9204780 DOI: 10.1249/mss.0000000000002888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Physical activity (PA) is recognized as one of the key lifestyle behaviors that reduces risk of developing dementia late in life. However, PA also leads to increased respiration, and in areas with high levels of air pollution, PA may increase exposure to pollutants linked with higher risk of developing dementia. Here, we investigate whether air pollution attenuates the association between PA and dementia risk. METHODS This prospective cohort study included 35,562 adults 60 yrs and older from the UK Biobank. Average acceleration magnitude (ACCave) from wrist-worn accelerometers was used to assess PA levels. Air pollution levels (NO, NO2, PM10, PM2.5, PM2.5-10, and PM2.5 absorbance) were estimated with land use regression methods. Incident all-cause dementia was derived from inpatient hospital records and death registry data. RESULTS In adjusted models, ACCave was associated with reduced risk of developing dementia (HR = 0.71, 95% confidence interval [CI] = 0.60-0.83), whereas air pollution variables were not associated with dementia risk. There were significant interactions between ACCave and PM2.5 (HRinteraction = 1.33, 95% CI = 1.13-1.57) and PM2.5 absorbance (HRinteraction = 1.24, 95% CI = 1.07-1.45) on incident dementia. At the lowest tertiles of pollution, ACCave was associated with reduced risk of incident dementia (HRPM 2.5 = 0.66, 95% CI = 0.49-0.91; HRPM 2.5 absorbance = 0.60, 95% CI = 0.44-0.81). At the highest tertiles of these pollutants, there was no significant association of ACCave with incident dementia (HRPM 2.5 = 0.88, 95% CI = 0.68-1.14; HRPM 2.5 absorbance = 0.79, 95% CI = 0.60-1.04). CONCLUSIONS PA is associated with reduced risk of developing all-cause dementia. However, exposure to even moderate levels of air pollution attenuates the benefits of PA on risk of dementia.
Collapse
Affiliation(s)
- David A Raichlen
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, CA
| | - Melissa Furlong
- Department of Community, Environment, and Policy, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | | | - M Katherine Sayre
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, CA
| | - Kimberly L Parra
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | | | - Rand R Wilcox
- Department of Psychology, University of Southern California, CA
| | | |
Collapse
|
25
|
Chen TF, Lee SH, Zheng WR, Hsu CC, Cho KH, Kuo LW, Chou CCK, Chiu MJ, Tee BL, Cheng TJ. White matter pathology in alzheimer's transgenic mice with chronic exposure to low-level ambient fine particulate matter. Part Fibre Toxicol 2022; 19:44. [PMID: 35768852 PMCID: PMC9245233 DOI: 10.1186/s12989-022-00485-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/29/2022] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Air pollution, especially fine particulate matter (PM), can cause brain damage, cognitive decline, and an increased risk of neurodegenerative disease, especially alzheimer's disease (AD). Typical pathological findings of amyloid and tau protein accumulation have been detected in the brain after exposure in animal studies. However, these observations were based on high levels of PM exposure, which were far from the WHO guidelines and those present in our environment. In addition, white matter involvement by air pollution has been less reported. Thus, this experiment was designed to simulate the true human world and to discuss the possible white matter pathology caused by air pollution. RESULTS 6 month-old female 3xTg-AD mice were divided into exposure and control groups and housed in the Taipei Air Pollutant Exposure System (TAPES) for 5 months. The mice were subjected to the Morris water maze test after exposure and were then sacrificed with brain dissection for further analyses. The mean mass concentration of PM2.5 during the exposure period was 13.85 μg/m3. After exposure, there was no difference in spatial learning function between the two groups, but there was significant decay of memory in the exposure group. Significantly decreased total brain volume and more neuronal death in the cerebral and entorhinal cortex and demyelination of the corpus callosum were noted by histopathological staining after exposure. However, there was no difference in the accumulation of amyloid or tau on immunohistochemistry staining. For the protein analysis, amyloid was detected at significantly higher levels in the cerebral cortex, with lower expression of myelin basic protein in the white matter. A diffuse tensor image study also revealed insults in multiple white matter tracts, including the optic tract. CONCLUSIONS In conclusion, this pilot study showed that even chronic exposure to low PM2.5 concentrations still caused brain damage, such as gross brain atrophy, cortical neuron damage, and multiple white matter tract damage. Typical amyloid cascade pathology did not appear prominently in the vulnerable brain region after exposure. These findings imply that multiple pathogenic pathways induce brain injury by air pollution, and the optic nerve may be another direct invasion route in addition to olfactory nerve.
Collapse
Affiliation(s)
- Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Wan-Ru Zheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Ching-Chou Hsu
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Kuan-Hung Cho
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Charles C-K Chou
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA, USA
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Room 720, No. 17, Xuzhou Rd, Taipei, 100, Taiwan.
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
26
|
Clouston SAP, Hall CB, Kritikos M, Bennett DA, DeKosky S, Edwards J, Finch C, Kreisl WC, Mielke M, Peskind ER, Raskind M, Richards M, Sloan RP, Spiro A, Vasdev N, Brackbill R, Farfel M, Horton M, Lowe S, Lucchini RG, Prezant D, Reibman J, Rosen R, Seil K, Zeig-Owens R, Deri Y, Diminich ED, Fausto BA, Gandy S, Sano M, Bromet EJ, Luft BJ. Cognitive impairment and World Trade Centre-related exposures. Nat Rev Neurol 2022; 18:103-116. [PMID: 34795448 PMCID: PMC8938977 DOI: 10.1038/s41582-021-00576-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 02/03/2023]
Abstract
On 11 September 2001 the World Trade Center (WTC) in New York was attacked by terrorists, causing the collapse of multiple buildings including the iconic 110-story 'Twin Towers'. Thousands of people died that day from the collapse of the buildings, fires, falling from the buildings, falling debris, or other related accidents. Survivors of the attacks, those who worked in search and rescue during and after the buildings collapsed, and those working in recovery and clean-up operations were exposed to severe psychological stressors. Concurrently, these 'WTC-affected' individuals breathed and ingested a mixture of organic and particulate neurotoxins and pro-inflammogens generated as a result of the attack and building collapse. Twenty years later, researchers have documented neurocognitive and motor dysfunctions that resemble the typical features of neurodegenerative disease in some WTC responders at midlife. Cortical atrophy, which usually manifests later in life, has also been observed in this population. Evidence indicates that neurocognitive symptoms and corresponding brain atrophy are associated with both physical exposures at the WTC and chronic post-traumatic stress disorder, including regularly re-experiencing traumatic memories of the events while awake or during sleep. Despite these findings, little is understood about the long-term effects of these physical and mental exposures on the brain health of WTC-affected individuals, and the potential for neurocognitive disorders. Here, we review the existing evidence concerning neurological outcomes in WTC-affected individuals, with the aim of contextualizing this research for policymakers, researchers and clinicians and educating WTC-affected individuals and their friends and families. We conclude by providing a rationale and recommendations for monitoring the neurological health of WTC-affected individuals.
Collapse
Affiliation(s)
- Sean A P Clouston
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| | - Charles B Hall
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Minos Kritikos
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, IL, USA
| | - Steven DeKosky
- Evelyn F. and William L. McKnight Brain Institute and Florida Alzheimer's Disease Research Center, Department of Neurology and Neuroscience, University of Florida, Gainesville, FL, USA
| | - Jerri Edwards
- Department of Psychiatry and Behavioral Neuroscience, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Caleb Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
| | - Michelle Mielke
- Specialized Center of Research Excellence on Sex Differences, Department of Neurology, Department of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Elaine R Peskind
- Veteran's Association VISN 20 Northwest Mental Illness Research, Education, and Clinical Center, Veteran's Affairs Puget Sound Health Care System, Seattle, WA, USA
- Alzheimer's Disease Research Center, Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Murray Raskind
- Veteran's Association VISN 20 Northwest Mental Illness Research, Education, and Clinical Center, Veteran's Affairs Puget Sound Health Care System, Seattle, WA, USA
- Alzheimer's Disease Research Center, Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Marcus Richards
- Medical Research Council Unit for Lifelong Health and Ageing, Population Health Sciences, University College London, London, UK
| | - Richard P Sloan
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Avron Spiro
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Department of Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Center, Center for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Robert Brackbill
- World Trade Center Health Registry, New York Department of Health and Mental Hygiene, New York, NY, USA
| | - Mark Farfel
- World Trade Center Health Registry, New York Department of Health and Mental Hygiene, New York, NY, USA
| | - Megan Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandra Lowe
- The World Trade Center Mental Health Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto G Lucchini
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA
| | - David Prezant
- World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joan Reibman
- Department of Environmental Medicine, New York University Langone Health, New York, NY, USA
| | - Rebecca Rosen
- World Trade Center Environmental Health Center, Department of Psychiatry, New York University, New York, NY, USA
| | - Kacie Seil
- World Trade Center Health Registry, New York Department of Health and Mental Hygiene, New York, NY, USA
| | - Rachel Zeig-Owens
- World Trade Center Health Program, Fire Department of the City of New York, Brooklyn, NY, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yael Deri
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Psychiatry, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Erica D Diminich
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Bernadette A Fausto
- Center for Molecular & Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Sam Gandy
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Department of Psychiatry, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Mary Sano
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Department of Psychiatry, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Evelyn J Bromet
- Department of Psychiatry, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Benjamin J Luft
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
27
|
Younan D, Wang X, Millstein J, Petkus AJ, Beavers DP, Espeland MA, Chui HC, Resnick SM, Gatz M, Kaufman JD, Wellenius GA, Whitsel EA, Manson JE, Rapp SR, Chen JC. Air quality improvement and cognitive decline in community-dwelling older women in the United States: A longitudinal cohort study. PLoS Med 2022; 19:e1003893. [PMID: 35113870 PMCID: PMC8812844 DOI: 10.1371/journal.pmed.1003893] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 12/15/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Late-life exposure to ambient air pollution is a modifiable risk factor for dementia, but epidemiological studies have shown inconsistent evidence for cognitive decline. Air quality (AQ) improvement has been associated with improved cardiopulmonary health and decreased mortality, but to the best of our knowledge, no studies have examined the association with cognitive function. We examined whether AQ improvement was associated with slower rate of cognitive decline in older women aged 74 to 92 years. METHODS AND FINDINGS We studied a cohort of 2,232 women residing in the 48 contiguous US states that were recruited from more than 40 study sites located in 24 states and Washington, DC from the Women's Health Initiative (WHI) Memory Study (WHIMS)-Epidemiology of Cognitive Health Outcomes (WHIMS-ECHO) study. They were predominantly non-Hispanic White women and were dementia free at baseline in 2008 to 2012. Measures of annual (2008 to 2018) cognitive function included the modified Telephone Interview for Cognitive Status (TICSm) and the telephone-based California Verbal Learning Test (CVLT). We used regionalized universal kriging models to estimate annual concentrations (1996 to 2012) of fine particulate matter (PM2.5) and nitrogen dioxide (NO2) at residential locations. Estimates were aggregated to the 3-year average immediately preceding (recent exposure) and 10 years prior to (remote exposure) WHIMS-ECHO enrollment. Individual-level improved AQ was calculated as the reduction from remote to recent exposures. Linear mixed effect models were used to examine the associations between improved AQ and the rates of cognitive declines in TICSm and CVLT trajectories, adjusting for sociodemographic (age; geographic region; race/ethnicity; education; income; and employment), lifestyle (physical activity; smoking; and alcohol), and clinical characteristics (prior hormone use; hormone therapy assignment; depression; cardiovascular disease (CVD); hypercholesterolemia; hypertension; diabetes; and body mass index [BMI]). For both PM2.5 and NO2, AQ improved significantly over the 10 years before WHIMS-ECHO enrollment. During a median of 6.2 (interquartile range [IQR] = 5.0) years of follow-up, declines in both general cognitive status (β = -0.42/year, 95% CI: -0.44, -0.40) and episodic memory (β = -0.59/year, 95% CI: -0.64, -0.54) were observed. Greater AQ improvement was associated with slower decline in TICSm (βPM2.5improvement = 0.026 per year for improved PM2.5 by each IQR = 1.79 μg/m3 reduction, 95% CI: 0.001, 0.05; βNO2improvement = 0.034 per year for improved NO2 by each IQR = 3.92 parts per billion [ppb] reduction, 95% CI: 0.01, 0.06) and CVLT (βPM2.5 improvement = 0.070 per year for improved PM2.5 by each IQR = 1.79 μg/m3 reduction, 95% CI: 0.02, 0.12; βNO2improvement = 0.060 per year for improved NO2 by each IQR = 3.97 ppb reduction, 95% CI: 0.005, 0.12) after adjusting for covariates. The respective associations with TICSm and CVLT were equivalent to the slower decline rate found with 0.9 to 1.2 and1.4 to 1.6 years of younger age and did not significantly differ by age, region, education, Apolipoprotein E (ApoE) e4 genotypes, or cardiovascular risk factors. The main limitations of this study include measurement error in exposure estimates, potential unmeasured confounding, and limited generalizability. CONCLUSIONS In this study, we found that greater improvement in long-term AQ in late life was associated with slower cognitive declines in older women. This novel observation strengthens the epidemiologic evidence of an association between air pollution and cognitive aging.
Collapse
Affiliation(s)
- Diana Younan
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Xinhui Wang
- Department of Neurology, University of Southern California, Los Angeles, California, United States of America
| | - Joshua Millstein
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Andrew J. Petkus
- Department of Neurology, University of Southern California, Los Angeles, California, United States of America
| | - Daniel P. Beavers
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Mark A. Espeland
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Helena C. Chui
- Department of Neurology, University of Southern California, Los Angeles, California, United States of America
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, California, United States of America
| | - Joel D. Kaufman
- Departments of Environmental & Occupational Health Sciences, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Gregory A. Wellenius
- Department of Environmental Health, Boston University, Boston, Massachusetts, United States of America
| | - Eric A. Whitsel
- Departments of Epidemiology and Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - JoAnn E. Manson
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephen R. Rapp
- Departments of Psychiatry and Behavioral Medicine and Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jiu-Chiuan Chen
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, United States of America
- Department of Neurology, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
28
|
Adivi A, JoAnn L, Simpson N, McDonald JD, Lund AK. Traffic-generated air pollution - Exposure mediated expression of factors associated with demyelination in a female apolipoprotein E -/- mouse model. Neurotoxicol Teratol 2022; 90:107071. [PMID: 35016995 PMCID: PMC8904307 DOI: 10.1016/j.ntt.2022.107071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 12/31/2021] [Accepted: 01/06/2022] [Indexed: 11/29/2022]
Abstract
Epidemiology studies suggest that exposure to ambient air pollution is associated with demyelinating diseases in the central nervous system (CNS), including multiple sclerosis (MS). The pathophysiology of MS results from an autoimmune response involving increased inflammation and demyelination in the CNS, which is higher in young (adult) females. Exposure to traffic-generated air pollution is associated with neuroinflammation and other detrimental outcomes in the CNS; however, its role in the progression of pathologies associated with demyelinating diseases has not yet been fully characterized in a female model. Thus, we investigated the effects of inhalation exposure to mixed vehicle emissions (MVE) in the brains of both ovary-intact (ov+) and ovariectomized (ov-) female Apolipoprotein (ApoE-/-) mice. Ov + and ov- ApoE-/- mice were exposed via whole-body inhalation to either filtered air (FA, controls) or mixed gasoline and diesel vehicle emissions (MVE: 200 PM μg/m3) for 6 h/d, 7 d/wk., for 30 d. We then analyzed MVE-exposure mediated alterations in myelination, the presence of CD4+ and CD8+ T cells, reactive oxygen species (ROS), myelin oligodendrocyte protein (MOG), and expression of estrogen (ERα and ERβ) and progesterone (PROA/B) receptors in the CNS. MVE-exposure mediated significant alterations in myelination across multiple regions in the cerebrum, as well as increased CD4+ and CD8+ staining. There was also an increase in ROS production in the CNS of MVE-exposed ov- and ov + ApoE-/- mice. Ov- mice displayed a reduction in cerebral ERα mRNA expression, compared to ov + mice; however, MVE exposure resulted in an even further decrease in ERα expression, while ERβ and PRO A/B were unchanged across groups. These findings collectively suggest that inhaled MVE-exposure may mediate estrogen receptor expression alterations associated with increased CD4+/CD8+ infiltration, regional demyelination, and ROS production in the CNS of female ApoE-/- mice.
Collapse
Affiliation(s)
- Anna Adivi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA, 76201
| | - Lucero JoAnn
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA, 76201
| | - Nicholas Simpson
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA, 76201
| | - Jacob D McDonald
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, USA, 87108
| | - Amie K. Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA, 76201.,Corresponding author at: University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76201,
| |
Collapse
|
29
|
Alemany S, Crous-Bou M, Vilor-Tejedor N, Milà-Alomà M, Suárez-Calvet M, Salvadó G, Cirach M, Arenaza-Urquijo EM, Sanchez-Benavides G, Grau-Rivera O, Minguillon C, Fauria K, Kollmorgen G, Domingo Gispert J, Gascón M, Nieuwenhuijsen M, Zetterberg H, Blennow K, Sunyer J, Luis Molinuevo J. Associations between air pollution and biomarkers of Alzheimer's disease in cognitively unimpaired individuals. ENVIRONMENT INTERNATIONAL 2021; 157:106864. [PMID: 34537521 DOI: 10.1016/j.envint.2021.106864] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/10/2021] [Accepted: 09/02/2021] [Indexed: 05/12/2023]
Abstract
BACKGROUND Air quality contributes to incidence of Alzheimer's disease (AD) although the underlying neurobiological mechanisms are unclear. This study was aimed to examine the association between air pollution and concentrations of cerebrospinal fluid (CSF) AD biomarkers and amyloid-β (Aβ) deposition. Participants and methods The sample included 156 cognitively unimpaired adults aged 57 years (61 at biomarkers assessment) with increased risk of AD from the ALFA + Study. We examined CSF levels of Aβ42, Aβ40, p-Tau, t-Tau, neurofilament light (NfL) and cerebral amyloid load (Centiloid). A Land Use Regression model from 2009 was used to estimate residential exposure to air pollutants including nitrogen dioxide (NO2,) and particulate matter (PM2.5, PM2.5 abs, PM10). This model was considered a surrogate of long-term exposure until time of data collection in 2013-2014. Participants have resided in the same residence for at least the previous 3 years. Multiple linear regression models were used to estimate associations between air pollutants and biomarkers. The effect modification by CSF Aβ status and APOE-ε4 carriership was also assessed. RESULTS A consistent pattern of results indicated that greater exposure to NO2 and PM2.5 absorbance was associated with higher levels of brain Aβ deposition, while greater exposure to PM10 and PM2.5was associated with higher levels of CSF NfL. Most associations were driven by individuals that were Aβ-positive. Although APOE-ε4 status did not significantly modify these associations, the effect of air pollutants exposure on CSF NfL levels was stronger in APOE-ε4 carriers. CONCLUSION In a population of cognitively unimpaired adults with increased risk of AD, long-term exposure to air pollution was associated with higher levels in biomarkers of AD pathology. While further research is granted to elucidate the mechanisms involved in such associations, our results reinforce the role of air pollution as an environmental risk factor for AD.
Collapse
Affiliation(s)
- Silvia Alemany
- ISGlobal, Barcelona Institute for Global Health - Campus MAR, Barcelona Biomedical Research Park. Barcelona, Spain; Universitat Pompeu Fabra (UPF). Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP). Madrid, Spain
| | - Marta Crous-Bou
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO) - Bellvitge Biomedical Research Institute (IDIBEL). L'Hospitalet de Llobregat, Barcelona, Spain; Department of Epidemiology, Harvard T.H. Chan School of Public Health. Boston, MA, USA.
| | - Natalia Vilor-Tejedor
- Universitat Pompeu Fabra (UPF). Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centre for Genomic Regulation (CRG). The Barcelona Institute for Science and Technology, Barcelona, Spain; Erasmus MC. University Medical Center Rotterdam. Department of Clinical Genetics.Center, Rotterdam, the Netherlands
| | - Marta Milà-Alomà
- Universitat Pompeu Fabra (UPF). Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain
| | - Marta Cirach
- ISGlobal, Barcelona Institute for Global Health - Campus MAR, Barcelona Biomedical Research Park. Barcelona, Spain; Universitat Pompeu Fabra (UPF). Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP). Madrid, Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain
| | - Gonzalo Sanchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBER-FES). Madrid, Spain
| | | | - Juan Domingo Gispert
- Universitat Pompeu Fabra (UPF). Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Mireia Gascón
- ISGlobal, Barcelona Institute for Global Health - Campus MAR, Barcelona Biomedical Research Park. Barcelona, Spain; Universitat Pompeu Fabra (UPF). Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP). Madrid, Spain
| | - Mark Nieuwenhuijsen
- ISGlobal, Barcelona Institute for Global Health - Campus MAR, Barcelona Biomedical Research Park. Barcelona, Spain; Universitat Pompeu Fabra (UPF). Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP). Madrid, Spain
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jordi Sunyer
- ISGlobal, Barcelona Institute for Global Health - Campus MAR, Barcelona Biomedical Research Park. Barcelona, Spain; Universitat Pompeu Fabra (UPF). Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP). Madrid, Spain; IMIM - Hospital del Mar Medical Research Institute. Barcelona, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.
| |
Collapse
|
30
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
31
|
Qamar AY, Hussain T, Rafique MK, Bang S, Tanga BM, Seong G, Fang X, Saadeldin IM, Cho J. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility. Cells 2021; 10:2460. [PMID: 34572109 PMCID: PMC8468931 DOI: 10.3390/cells10092460] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Infertility is a globally recognized issue caused by different reproductive disorders. To date, various therapeutic approaches to restore fertility have been attempted including etiology-specific medication, hormonal therapies, surgical excisions, and assisted reproductive technologies. Although these approaches produce results, however, fertility restoration is not achieved in all cases. Advances in using stem cell (SC) therapy hold a great promise for treating infertile patients due to their abilities to self-renew, differentiate, and produce different paracrine factors to regenerate the damaged or injured cells and replenish the affected germ cells. Furthermore, SCs secrete extracellular vesicles (EVs) containing biologically active molecules including nucleic acids, lipids, and proteins. EVs are involved in various physiological and pathological processes and show promising non-cellular therapeutic uses to combat infertility. Several studies have indicated that SCs and/or their derived EVs transplantation plays a crucial role in the regeneration of different segments of the reproductive system, oocyte production, and initiation of sperm production. However, available evidence triggers the need to testify the efficacy of SC transplantation or EVs injection in resolving the infertility issues of the human population. In this review, we highlight the recent literature covering the issues of infertility in females and males, with a special focus on the possible treatments by stem cells or their derived EVs.
Collapse
Affiliation(s)
- Ahmad Yar Qamar
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- College of Veterinary and Animal Sciences, Jhang, Sub-Campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Tariq Hussain
- College of Veterinary and Animal Sciences, Jhang, Sub-Campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Muhammad Kamran Rafique
- College of Veterinary and Animal Sciences, Jhang, Sub-Campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Seonggyu Bang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Bereket Molla Tanga
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Faculty of Veterinary Medicine, Hawassa University, Hawassa 05, Ethiopia
| | - Gyeonghwan Seong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Xun Fang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Islam M Saadeldin
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jongki Cho
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
32
|
Finch CE, Haghani A. Gene-Environment Interactions and Stochastic Variations in the Gero-Exposome. J Gerontol A Biol Sci Med Sci 2021; 76:1740-1747. [PMID: 33580247 PMCID: PMC8436990 DOI: 10.1093/gerona/glab045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
The limited heritability of human life spans suggests an important role for gene-environment (G × E) interactions across the life span (T), from gametes to geronts. Multilevel G × E × T interactions of aging phenotypes are conceptualized in the Gero-Exposome as Exogenous and Endogenous domains. Stochastic variations in the Endogenous domain contribute to the diversity of aging phenotypes, shown for the diversity of inbred Caenorhabditis elegans life spans in the same culture environment, and for variegated gene expression of somatic cells in nematodes and mammals. These phenotypic complexities can be analyzed as 3-way interactions of gene, environment, and stochastic variations, the Tripartite Phenotype of Aging. Single-cell analyses provide tools to explore this broadening frontier of biogerontology.
Collapse
Affiliation(s)
- Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
- Dornsife College, University of Southern California, Los Angeles, CA
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
33
|
Wang X, Younan D, Petkus AJ, Beavers DP, Espeland MA, Chui HC, Resnick SM, Gatz M, Kaufman JD, Wellenius GA, Whitsel EA, Manson JE, Chen JC. Ambient Air Pollution and Long-Term Trajectories of Episodic Memory Decline among Older Women in the WHIMS-ECHO Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:97009. [PMID: 34516296 PMCID: PMC8437247 DOI: 10.1289/ehp7668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Episodic memory decline varies by age and underlying neuropathology. Whether ambient air pollution contributes to the heterogeneity of episodic memory decline in older populations remains unclear. OBJECTIVES We estimated associations between air pollution exposures and episodic memory decline according to pollutant, exposure time window, age, and latent class subgroups defined by episodic memory trajectories. METHODS Participants were from the Women's Health Initiative Memory Study-Epidemiology of Cognitive Health Outcomes. Older women (n = 2,056 ; 74-92 years of age) completed annual (2008-2018) episodic memory assessments using the telephone-based California Verbal Learning Test (CVLT). We estimated 3-y average fine particulate matter [PM with an aerodynamic diameter of ≤ 2.5 μ m (PM 2.5 )] and nitrogen dioxide (NO 2 ) exposures at baseline and 10 y earlier (recent and remote exposures, respectively), using regionalized national universal kriging. Separate latent class mixed models were used to estimate associations between interquartile range increases in exposures and CVLT trajectories in women ≤ 80 and > 80 years of age , adjusting for covariates. RESULTS Two latent classes were identified for women ≤ 80 years of age (n = 828 ), "slow-decliners" {slope = - 0.12 / y [95% confidence interval (CI): - 0.23 , - 0.01 ] and "fast-decliners" [slope = - 1.79 / y (95% CI: - 2.08 , - 1.50 )]}. In the slow-decliner class, but not the fast-decliner class, PM 2.5 exposures were associated with a greater decline in CVLT scores over time, with a stronger association for recent vs. remote exposures [- 0.16 / y (95% CI: - 2.08 , - 0.03 ) per 2.88 μ g / m 3 and - 0.11 / y (95% CI: - 0.22 , 0.01) per 3.27 μ g / m 3 , respectively]. Among women ≥ 80 years of age (n = 1,128 ), the largest latent class comprised "steady-decliners" [slope = - 1.35 / y (95% CI: - 1.53 , - 1.17 )], whereas the second class, "cognitively resilient", had no decline in CVLT on average. PM 2.5 was not associated with episodic memory decline in either class. A 6.25 -ppb increase in recent NO 2 was associated with nonsignificant acceleration of episodic memory decline in the ≤ 80 -y-old fast-decliner class [- 0.21 / y (95% CI: - 0.45 , 0.04)], and in the > 80 -y-old cognitively resilient class [- 0.10 / y (95% CI: - 0.24 , 0.03)] and steady-decliner class [- 0.11 / y (95% CI: - 0.27 , 0.05)]. Associations with recent NO 2 exposure in women > 80 years of age were stronger and statistically significant when 267 women with incident probable dementia were excluded [e.g., - 0.12 / y (95% CI: - 0.22 , - 0.02 ) for the cognitively resilient class]. In contrast with changes in CVLT over time, there were no associations between exposures and CVLT scores during follow-up in any subgroup. DISCUSSION In a community-dwelling U.S. population of older women, associations between late-life exposure to ambient air pollution and episodic memory decline varied by age-related cognitive trajectories, exposure time windows, and pollutants. https://doi.org/10.1289/EHP7668.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Diana Younan
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| | - Andrew J Petkus
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Daniel P Beavers
- Department of Biostatistics and Data Sciences, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Mark A Espeland
- Department of Biostatistics and Data Sciences, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Helena C Chui
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland, USA
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, California, USA
| | - Joel D Kaufman
- Departments of Environmental & Occupational Health Sciences, Medicine (General Internal Medicine), and Epidemiology, University of Washington, Seattle, Washington, USA
| | - Gregory A Wellenius
- Department of Environmental Health, Boston University, Boston, Massachusetts, USA
| | - Eric A Whitsel
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - JoAnn E Manson
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jiu-Chiuan Chen
- Department of Neurology, University of Southern California, Los Angeles, California, USA
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
34
|
Lee SH, Chen YH, Chien CC, Yan YH, Chen HC, Chuang HC, Hsieh HI, Cho KH, Kuo LW, Chou CCK, Chiu MJ, Tee BL, Chen TF, Cheng TJ. Three month inhalation exposure to low-level PM2.5 induced brain toxicity in an Alzheimer's disease mouse model. PLoS One 2021; 16:e0254587. [PMID: 34437570 PMCID: PMC8389369 DOI: 10.1371/journal.pone.0254587] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Although numerous epidemiological studies revealed an association between ambient fine particulate matter (PM2.5) exposure and Alzheimer's disease (AD), the PM2.5-induced neuron toxicity and associated mechanisms were not fully elucidated. The present study assessed brain toxicity in 6-month-old female triple-transgenic AD (3xTg-AD) mice following subchronic exposure to PM2.5 via an inhalation system. The treated mice were whole-bodily and continuously exposed to real-world PM2.5 for 3 months, while the control mice inhaled filtered air. Changes in cognitive and motor functions were evaluated using the Morris Water Maze and rotarod tests. Magnetic resonance imaging analysis was used to record gross brain volume alterations, and tissue staining with hematoxylin and eosin, Nissl, and immunohistochemistry methods were used to monitor pathological changes in microstructures after PM2.5 exposure. The levels of AD-related hallmarks and the oxidative stress biomarker malondialdehyde (MDA) were assessed using Western blot analysis and liquid chromatography-mass spectrometry, respectively. Our results showed that subchronic exposure to environmental levels of PM2.5 induced obvious neuronal loss in the cortex of exposed mice, but without significant impairment of cognitive and motor function. Increased levels of phosphorylated-tau and MDA were also observed in olfactory bulb or hippocampus after PM2.5 exposure, but no amyloid pathology was detected, as reported in previous studies. These results revealed that a relatively lower level of PM2.5 subchronic exposure from the environmental atmosphere still induced certain neurodegenerative changes in the brains of AD mice, especially in the olfactory bulb, entorhinal cortex and hippocampus, which is consistent with the nasal entry and spreading route for PM exposure. Systemic factors may also contribute to the neuronal toxicity. The effects of PM2.5 after a more prolonged exposure period are needed to establish a more comprehensive picture of the PM2.5-mediated development of AD.
Collapse
Affiliation(s)
- Sheng-Han Lee
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsuan Chen
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chu-Chun Chien
- Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Horng Yan
- Department of Endocrinology and Metabolism, Kuang Tien General Hospital, Taichung, Taiwan
- Department of Nutrition and Institute of Biomedical Nutrition, Hung Kuang University, Taichung, Taiwan
| | - Hsin-Chang Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-I Hsieh
- Department of Occupational Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Kuan-Hung Cho
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Boon Lead Tee
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, California, United States of America
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
35
|
Younan D, Wang X, Gruenewald T, Gatz M, Serre ML, Vizuete W, Braskie MN, Woods NF, Kahe K, Garcia L, Lurmann F, Manson JE, Chui HC, Wallace RB, Espeland MA, Chen JC. Racial/Ethnic Disparities in Alzheimer's Disease Risk: Role of Exposure to Ambient Fine Particles. J Gerontol A Biol Sci Med Sci 2021; 77:977-985. [PMID: 34383042 PMCID: PMC9071399 DOI: 10.1093/gerona/glab231] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Whether racial/ethnic disparities in Alzheimer's disease (AD) risk may be explained by ambient fine particles (PM2.5) has not been studied. METHOD We conducted a prospective, population-based study on a cohort of Black (n = 481) and White (n = 6 004) older women (aged 65-79) without dementia at enrollment (1995-1998). Cox models accounting for competing risk were used to estimate the hazard ratio (HR) for racial/ethnic disparities in AD (1996-2010) defined by Diagnostic and Statistical Manual of Mental Disorders, 4th edition and the association with time-varying annual average PM2.5 (1999-2010) estimated by spatiotemporal model. RESULTS Over an average follow-up of 8.3 (±3.5) years with 158 incident cases (21 in Black women), the racial disparities in AD risk (range of adjusted HRBlack women = 1.85-2.41) observed in various models could not be explained by geographic region, age, socioeconomic characteristics, lifestyle factors, cardiovascular risk factors, and hormone therapy assignment. Estimated PM2.5 exposure was higher in Black (14.38 ± 2.21 µg/m3) than in White (12.55 ± 2.76 µg/m3) women, and further adjustment for the association between PM2.5 and AD (adjusted HRPM2.5 = 1.18-1.28) slightly reduced the racial disparities by 2%-6% (HRBlack women = 1.81-2.26). The observed association between PM2.5 and AD risk was ~2 times greater in Black (HRPM2.5 = 2.10-2.60) than in White (HRPM2.5 = 1.07-1.15) women (range of interaction ps: <.01-.01). We found similar results after further adjusting for social engagement (social strain, social support, social activity, living alone), stressful life events, Women's Health Initiative's clinic sites, and neighborhood socioeconomic characteristics. CONCLUSIONS PM2.5 may contribute to racial/ethnic disparities in AD risk and its associated increase in AD risk was stronger among Black women.
Collapse
Affiliation(s)
- Diana Younan
- University of Southern California, Los Angeles, USA
| | - Xinhui Wang
- University of Southern California, Los Angeles, USA
| | | | | | | | | | | | - Nancy F Woods
- University of Washington School of Nursing, Seattle, USA
| | - Ka Kahe
- Columbia University Irving Medical Center, New York, New York, USA
| | | | - Fred Lurmann
- Sonoma Technology, Inc., Petaluma, California, USA
| | - JoAnn E Manson
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Mark A Espeland
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jiu-Chiuan Chen
- Address correspondence to: Jiu-Chiuan Chen, MD, ScD, University of Southern California, 2001 N Soto Street, Los Angeles, CA 90032, USA. E-mail:
| |
Collapse
|
36
|
Liu Q, Shkirkova K, Lamorie-Foote K, Connor M, Patel A, Babadjouni R, Huuskonen M, Montagne A, Baertsch H, Zhang H, Chen JC, Mack WJ, Walcott BP, Zlokovic BV, Sioutas C, Morgan TE, Finch CE, Mack WJ. Air Pollution Particulate Matter Exposure and Chronic Cerebral Hypoperfusion and Measures of White Matter Injury in a Murine Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:87006. [PMID: 34424052 PMCID: PMC8382048 DOI: 10.1289/ehp8792] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Exposure to ambient air pollution particulate matter (PM) is associated with increased risk of dementia and accelerated cognitive loss. Vascular contributions to cognitive impairment are well recognized. Chronic cerebral hypoperfusion (CCH) promotes neuroinflammation and blood-brain barrier weakening, which may augment neurotoxic effects of PM. OBJECTIVES This study examined interactions of nanoscale particulate matter (nPM; fine particulate matter with aerodynamic diameter ≤ 200 nm ) and CCH secondary to bilateral carotid artery stenosis (BCAS) in a murine model to produce white matter injury. Based on other air pollution interactions, we predicted synergies of nPM with BCAS. METHODS nPM was collected using a particle sampler near a Los Angeles, California, freeway. Mice were exposed to 10 wk of reaerosolized nPM or filtered air (FA) for 150 h. CCH was induced by BCAS surgery. Mice (C57BL/6J males) were randomized to four exposure paradigms: a) FA, b) nPM, c) FA + BCAS , and d) nPM + BCAS . Behavioral outcomes, white matter injury, glial cell activation, inflammation, and oxidative stress were assessed. RESULTS The joint nPM + BCAS group exhibited synergistic effects on white matter injury (2.3× the additive nPM and FA + BCAS scores) with greater loss of corpus callosum volume on T2 magnetic resonance imaging (MRI) (30% smaller than FA group). Histochemical analyses suggested potential microglial-specific inflammatory responses with synergistic effects on corpus callosum C5 immunofluorescent density and whole brain nitrate concentrations (2.1× and 3.9× the additive nPM and FA + BCAS effects, respectively) in the joint exposure group. Transcriptomic responses (RNA-Seq) showed greater impact of nPM + BCAS than individual additive effects, consistent with changes in proinflammatory pathways. Although nPM exposure alone did not alter working memory, the nPM + BCAS cohort demonstrated impaired working memory when compared to the FA + BCAS group. DISCUSSION Our data suggest that nPM and CCH contribute to white matter injury in a synergistic manner in a mouse model. Adverse neurological effects may be aggravated in a susceptible population exposed to air pollution. https://doi.org/10.1289/EHP8792.
Collapse
Affiliation(s)
- Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arati Patel
- Department of Neurological Surgery, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Robin Babadjouni
- Department of Neurological Surgery, Cedars-Sinai, Los Angeles, California, USA
| | - Mikko Huuskonen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hongqiao Zhang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Jiu-Chiuan Chen
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Wendy J. Mack
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Brian P. Walcott
- Department of Neurosurgery, Northshore Neurological Institute, Evanston, Illinois, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
37
|
Dos Santos NV, Yariwake VY, Marques KDV, Veras MM, Fajersztajn L. Air Pollution: A Neglected Risk Factor for Dementia in Latin America and the Caribbean. Front Neurol 2021; 12:684524. [PMID: 34367051 PMCID: PMC8339300 DOI: 10.3389/fneur.2021.684524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
The risk of dementia and Alzheimer's disease in Latin America and the Caribbean (LAC) rises with increasing age and polluted air. Currently, at least 172 million people breathe unhealthy levels of air pollution in LAC countries. Several cohort studies have indicated that air pollution increases the risk of developing dementia and neurodegenerative diseases, but the mechanisms underlying the association are still not clear. Air pollution causes and aggravates five established risk factors for dementia (obesity, hypertension, stroke, diabetes mellitus, and heart diseases) and is linked to three other risk factors (physical inactivity, cognitive inactivity, and depression). Some of these risk factors could be mediating the association between air pollution and dementia. Reducing the risks for dementia is crucial and urgently needed in LAC countries. There is room for improving air quality in many urban areas in the LAC region and other low- and middle-income countries (LMICs), a routealready explored by many urban areas in developing regions. Moreover, reducing air pollution has proved to improve health outcomes before. In this article, we propose that despite the ongoing and valid scientific discussion, if air pollution can or cannot directly affect the brain and cause or aggravate dementia, we are ready to consider air pollution as a potentially modifiable risk factor for dementia in LAC and possibly in other LMICs. We suggest that controlling and reducing current air pollution levels in LAC and other LMIC regions now could strongly contribute.
Collapse
Affiliation(s)
- Nathália Villa Dos Santos
- Laboratório de Poluição Ambiental, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Saude Ambiental, Faculdade de Saude Publica, Universidade de São Paulo, São Paulo, Brazil
| | - Victor Yuji Yariwake
- Laboratório de Poluição Ambiental, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Mariana Matera Veras
- Laboratório de Poluição Ambiental, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Laís Fajersztajn
- Laboratório de Poluição Ambiental, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Liu RM, Chong Z, Chen JC. Ozone and Particulate Matter Exposure and Alzheimer's Disease: A Review of Human and Animal Studies. J Alzheimers Dis 2021; 76:807-824. [PMID: 32568209 DOI: 10.3233/jad-200435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), an aging-related neurodegenerative disease, is a major cause of dementia in the elderly. Although the early-onset (familial) AD is attributed to mutations in the genes coding for amyloid-β protein precursor (AβPP) and presenilin1/presenilin 2 (PS1/PS2), the cause for the late-onset AD (LOAD), which accounts for more than 95% of AD cases, remains unclear. Aging is the greatest risk factor for LOAD, whereas the apolipo protein E4 allele (APOEɛ4) is believed to be a major genetic risk factor in acquiring LOAD, with female APOEɛ4 carriers at highest risk. Nonetheless, not all the elderly, even older female APOEɛ4 carriers, develop LOAD, suggesting that other factors, including environmental exposure, must play a role. This review summarizes recent studies that show a potential role of environmental exposure, especially ozone and particulate matter exposure, in the development of AD. Interactions between environmental exposure, genetic risk factor (APOEɛ4), and sex in AD pathophysiology are also discussed briefly. Identification of environmental risk factor(s) and elucidation of the complex interactions between genetic and environmental risk factors plus aging and female sex in the onset of AD will be a key to our understanding of the etiology and pathogenesis of AD and the development of the strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zechen Chong
- Department of Genetics, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jiu-Chiuan Chen
- Department of Biostatistics and Data Science, The University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Haghani A, Morgan TE, Forman HJ, Finch CE. Air Pollution Neurotoxicity in the Adult Brain: Emerging Concepts from Experimental Findings. J Alzheimers Dis 2021; 76:773-797. [PMID: 32538853 DOI: 10.3233/jad-200377] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epidemiological studies are associating elevated exposure to air pollution with increased risk of Alzheimer's disease and other neurodegenerative disorders. In effect, air pollution accelerates many aging conditions that promote cognitive declines of aging. The underlying mechanisms and scale of effects remain largely unknown due to its chemical and physical complexity. Moreover, individual responses to air pollution are shaped by an intricate interface of pollutant mixture with the biological features of the exposed individual such as age, sex, genetic background, underlying diseases, and nutrition, but also other environmental factors including exposure to cigarette smoke. Resolving this complex manifold requires more detailed environmental and lifestyle data on diverse populations, and a systematic experimental approach. Our review aims to summarize the modest existing literature on experimental studies on air pollution neurotoxicity for adult rodents and identify key gaps and emerging challenges as we go forward. It is timely for experimental biologists to critically understand prior findings and develop innovative approaches to this urgent global problem. We hope to increase recognition of the importance of air pollution on brain aging by our colleagues in the neurosciences and in biomedical gerontology, and to support the immediate translation of the findings into public health guidelines for the regulation of remedial environmental factors that accelerate aging processes.
Collapse
Affiliation(s)
- Amin Haghani
- Leonard Davis School of Gerontology, USC, Los Angeles, CA, USA
| | - Todd E Morgan
- Leonard Davis School of Gerontology, USC, Los Angeles, CA, USA
| | | | - Caleb E Finch
- Leonard Davis School of Gerontology, USC, Los Angeles, CA, USA.,Dornsife College, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Patten KT, Valenzuela AE, Wallis C, Berg EL, Silverman JL, Bein KJ, Wexler AS, Lein PJ. The Effects of Chronic Exposure to Ambient Traffic-Related Air Pollution on Alzheimer's Disease Phenotypes in Wildtype and Genetically Predisposed Male and Female Rats. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:57005. [PMID: 33971107 PMCID: PMC8110309 DOI: 10.1289/ehp8905] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
BACKGROUND Epidemiological data link traffic-related air pollution (TRAP) to increased risk of Alzheimer's disease (AD). Preclinical data corroborating this association are largely from studies of male animals exposed acutely or subchronically to high levels of isolated fractions of TRAP. What remains unclear is whether chronic exposure to ambient TRAP modifies AD risk and the influence of sex on this interaction. OBJECTIVES This study sought to assess effects of chronic exposure to ambient TRAP on the time to onset and severity of AD phenotypes in a preclinical model and to determine whether sex or genetic susceptibility influences outcomes. METHODS Male and female TgF344-AD rats that express human AD risk genes and wildtype littermates were housed in a vivarium adjacent to a heavily trafficked tunnel in Northern California and exposed for up to 14 months to filtered air (FA) or TRAP drawn from the tunnel and delivered to animals unchanged in real time. Refractive particles in the brain and AD phenotypes were quantified in 3-, 6-, 10-, and 15-month-old animals using hyperspectral imaging, behavioral testing, and neuropathologic measures. RESULTS Particulate matter (PM) concentrations in TRAP exposure chambers fluctuated with traffic flow but remained below 24-h PM with aerodynamic diameter less than or equal to 2.5 micrometers (PM2.5) U.S. National Ambient Air Quality Standards limits. Ultrafine PM was a predominant component of TRAP. Nano-sized refractive particles were detected in the hippocampus of TRAP animals. TRAP-exposed animals had more amyloid plaque deposition, higher hyperphosphorylated tau levels, more neuronal cell loss, and greater cognitive deficits in an age-, genotype-, and sex-dependent manner. TRAP-exposed animals also had more microglial cell activation, but not astrogliosis. DISCUSSION These data demonstrate that chronic exposure to ambient TRAP promoted AD phenotypes in wildtype and genetically susceptible rats. TRAP effects varied according to age, sex, and genotype, suggesting that AD progression depends on complex interactions between environment and genetics. These findings suggest current PM2.5 regulations are insufficient to protect the aging brain. https://doi.org/10.1289/EHP8905.
Collapse
Affiliation(s)
- Kelley T. Patten
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
| | - Anthony E. Valenzuela
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
| | | | - Elizabeth L. Berg
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, California, USA
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, California, USA
- The MIND Institute, UC Davis School of Medicine, Sacramento, California, USA
| | - Keith J. Bein
- Air Quality Research Center, UC Davis, Davis, California, USA
- Center for Health and the Environment, UC Davis, Davis, California, USA
| | - Anthony S. Wexler
- Air Quality Research Center, UC Davis, Davis, California, USA
- Mechanical and Aerospace Engineering, Civil and Environmental Engineering, and Land, Air and Water Resources, UC Davis, Davis, California, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California Davis (UC Davis) School of Veterinary Medicine, Davis, California, USA
- The MIND Institute, UC Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
41
|
Clouston SA, Kritikos M, Deri Y, Horton M, Pellecchia AC, Santiago-Michels S, Carr MA, Gandy S, Sano M, Bromet EJ, Lucchini RG, Luft BJ. A cortical thinning signature to identify World Trade Center responders with possible dementia. INTELLIGENCE-BASED MEDICINE 2021; 5:100032. [PMID: 35991958 PMCID: PMC9387912 DOI: 10.1016/j.ibmed.2021.100032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Introduction World Trade Center (WTC) responders have a high risk of early-onset cognitive impairment (CI), but little is known about the etiology including the extent to which CI in WTC responders is accompanied by cortical atrophy as is common in progressive diseases causing age-related CI such as Alzheimer's disease and related dementias. In the current study, we entrained an artificial neural network (ANN) to determine the accuracy of cortical thickness (CTX) on magnetic resonance imaging to identify World Trade Center responders at midlife (aged 44-65 years) with possible dementia. Methods A total of 119 WTC responders (57 with CI and 62 with intact cognition) underwent a structural MRI scanning protocol including T1-weighted MPRAGE as part of two imaging studies. The discovery study was divided into training and validation samples, while a second replication sample was used. An ANN was trained using regional CTX measured across 34 unilateral regions of interest (ROIs) using Freesurfer software and 'Desikan-Killiany' brain atlas. The discovery sample was used for model development, and the replication sample was used to evaluate predictive accuracy. Results In the WTC responder cohort, the ANN algorithm showed high discrimination performance for CI. The ANN model using regional CTX data from both hemispheres achieved an area under the receiver operating characteristic curve (AUC) of 0.96 95% C.I. = [0.91-1.00] (Accuracy = 96.0%, Precision = 97.8%, Recall = 95.8%, Sensitivity = 95.8%, Specificity = 98.0%, F1 = 96.8%) for the discovery sample and AUC = 0.90 [0.70-1.00] (Accuracy = 90.0%, Precision = 90.0%, Sensitivity = 90.0%, Specificity = 90.0%, F1 = 90.0%) in the replication sample. Conclusion Analysis of bilateral regional CTX data derived from T1-weighted MPRAGE images by ANN analysis demonstrated excellent accuracy in distinguishing WTC responders with early-onset CI.
Collapse
Affiliation(s)
- Sean A.P. Clouston
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Minos Kritikos
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yael Deri
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Megan Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alison C. Pellecchia
- Stony Brook World Trade Center Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Stephanie Santiago-Michels
- Stony Brook World Trade Center Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Melissa A. Carr
- Stony Brook World Trade Center Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sam Gandy
- Center for Cognitive Health and NFL Neurological Care, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry and Mount Sinai Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mary Sano
- Department of Psychiatry and Mount Sinai Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Evelyn J. Bromet
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| | - Roberto G. Lucchini
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Benjamin J. Luft
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
42
|
Iaccarino L, La Joie R, Lesman-Segev OH, Lee E, Hanna L, Allen IE, Hillner BE, Siegel BA, Whitmer RA, Carrillo MC, Gatsonis C, Rabinovici GD. Association Between Ambient Air Pollution and Amyloid Positron Emission Tomography Positivity in Older Adults With Cognitive Impairment. JAMA Neurol 2021; 78:197-207. [PMID: 33252608 DOI: 10.1001/jamaneurol.2020.3962] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Importance Amyloid-β (Aβ) deposition is a feature of Alzheimer disease (AD) and may be promoted by exogenous factors, such as ambient air quality. Objective To examine the association between the likelihood of amyloid positron emission tomography (PET) scan positivity and ambient air quality in individuals with cognitive impairment. Design, Setting, and Participants This cross-sectional study used data from the Imaging Dementia-Evidence for Amyloid Scanning Study, which included more than 18 000 US participants with cognitive impairment who received an amyloid PET scan with 1 of 3 Aβ tracers (fluorine 18 [18F]-labeled florbetapir, 18F-labeled florbetaben, or 18F-labeled flutemetamol) between February 16, 2016, and January 10, 2018. A sample of older adults with mild cognitive impairment (MCI) or dementia was selected. Exposures Air pollution was estimated at the patient residence using predicted fine particulate matter (PM2.5) and ground-level ozone (O3) concentrations from the Environmental Protection Agency Downscaler model. Air quality was estimated at 2002 to 2003 (early, or approximately 14 [range, 13-15] years before amyloid PET scan) and 2015 to 2016 (late, or approximately 1 [range, 0-2] years before amyloid PET scan). Main Outcomes and Measures Primary outcome measure was the association between air pollution and the likelihood of amyloid PET scan positivity, which was measured as odds ratios (ORs) and marginal effects, adjusting for demographic, lifestyle, and socioeconomic factors and medical comorbidities, including respiratory, cardiovascular, cerebrovascular, psychiatric, and neurological conditions. Results The data set included 18 178 patients, of which 10 991 (60.5%) had MCI and 7187 (39.5%) had dementia (mean [SD] age, 75.8 [6.3] years; 9333 women [51.3%]). Living in areas with higher estimated biennial PM2.5 concentrations in 2002 to 2003 was associated with a higher likelihood of amyloid PET scan positivity (adjusted OR, 1.10; 95% CI, 1.05-1.15; z score = 3.93; false discovery rate [FDR]-corrected P < .001; per 4-μg/m3 increments). Results were similar for 2015 to 2016 data (OR, 1.15; 95% CI, 1.05-1.26, z score = 3.14; FDR-corrected P = .003). An average marginal effect (AME) of +0.5% (SE = 0.1%; z score, 3.93; 95% CI, 0.3%-0.7%; FDR-corrected P < .001) probability of amyloid PET scan positivity for each 1-μg/m3 increase in PM2.5 was observed for 2002 to 2003, whereas an AME of +0.8% (SE = 0.2%; z score = 3.15; 95% CI, 0.3%-1.2%; FDR-corrected P = .002) probability was observed for 2015 to 2016. Post hoc analyses showed no effect modification by sex (2002-2003: interaction term β = 1.01 [95% CI, 0.99-1.04; z score = 1.13; FDR-corrected P = .56]; 2015-2016: β = 1.02 [95% CI, 0.98-1.07; z score = 0.91; FDR-corrected P = .56]) or clinical stage (2002-2003: interaction term β = 1.01 [95% CI, 0.99-1.03; z score = 0.77; FDR-corrected P = .58]; 2015-2016: β = 1.03; 95% CI, 0.99-1.08; z score = 1.46; FDR-corrected P = .47]). Exposure to higher O3 concentrations was not associated with amyloid PET scan positivity in both time windows. Conclusions and Relevance This study found that higher PM2.5 concentrations appeared to be associated with brain Aβ plaques. These findings suggest the need to consider airborne toxic pollutants associated with Aβ pathology in public health policy decisions and to inform individual lifetime risk of developing AD and dementia.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Orit H Lesman-Segev
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco.,Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Eunice Lee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco
| | - Lucy Hanna
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco
| | - Bruce E Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond
| | - Barry A Siegel
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Rachel A Whitmer
- Division of Research, Kaiser Permanente, Oakland, California.,Department of Public Health Sciences, University of California, Davis, Davis
| | - Maria C Carrillo
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, Illinois
| | - Constantine Gatsonis
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island.,Department of Biostatistics, Brown University School of Public Health, Providence, Rhode Island
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco.,Associate Editor, JAMA Neurology
| |
Collapse
|
43
|
Shabani S. A mechanistic view on the neurotoxic effects of air pollution on central nervous system: risk for autism and neurodegenerative diseases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:6349-6373. [PMID: 33398761 DOI: 10.1007/s11356-020-11620-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
Many reports have shown a strong association between exposure to neurotoxic air pollutants like heavy metal and particulate matter (PM) as an active participant and neurological disorders. While the effects of these toxic pollutants on cardiopulmonary morbidity have principally been studied, growing evidence has shown that exposure to polluted air is associated with memory impairment, communication deficits, and anxiety/depression among all ages. So, these toxic pollutants in the environment increase the risk of neurodegenerative disease, ischemia, and autism spectrum disorders (ASD). The precise mechanisms in which air pollutants lead to communicative inability, social inability, and declined cognition have remained unknown. Various animal model studies show that amyloid precursor protein (APP), processing, oxidant/antioxidant balance, and inflammation pathways change following the exposure to constituents of polluted air. In the present review study, we collect the probable molecular mechanisms of deleterious CNS effects in response to various air pollutants.
Collapse
Affiliation(s)
- Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
44
|
Petkus AJ, Younan D, Wang X, Beavers DP, Espeland MA, Gatz M, Gruenewald T, Kaufman JD, Chui HC, Millstein J, Rapp SR, Manson JE, Resnick SM, Wellenius GA, Whitsel EA, Widaman K, Chen JC. Associations Between Air Pollution Exposure and Empirically Derived Profiles of Cognitive Performance in Older Women. J Alzheimers Dis 2021; 84:1691-1707. [PMID: 34744078 PMCID: PMC9057084 DOI: 10.3233/jad-210518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Elucidating associations between exposures to ambient air pollutants and profiles of cognitive performance may provide insight into neurotoxic effects on the aging brain. OBJECTIVE We examined associations between empirically derived profiles of cognitive performance and residential concentrations of particulate matter of aerodynamic diameter < 2.5 (PM2.5) and nitrogen dioxide (NO2) in older women. METHOD Women (N = 2,142) from the Women's Health Initiative Study of Cognitive Aging completed a neuropsychological assessment measuring attention, visuospatial, language, and episodic memory abilities. Average yearly concentrations of PM2.5 and NO2 were estimated at the participant's addresses for the 3 years prior to the assessment. Latent profile structural equation models identified subgroups of women exhibiting similar profiles across tests. Multinomial regressions examined associations between exposures and latent profile classification, controlling for covariates. RESULT Five latent profiles were identified: low performance across multiple domains (poor multi-domain; n = 282;13%), relatively poor verbal episodic memory (poor memory; n = 216; 10%), average performance across all domains (average multi-domain; n = 974; 45%), superior memory (n = 381; 18%), and superior attention (n = 332; 15%). Using women with average cognitive ability as the referent, higher PM2.5 (per interquartile range [IQR] = 3.64μg/m3) was associated with greater odds of being classified in the poor memory (OR = 1.29; 95% Confidence Interval [CI] = 1.10-1.52) or superior attention (OR = 1.30; 95% CI = 1.10-1.53) profiles. NO2 (per IQR = 9.86 ppb) was associated with higher odds of being classified in the poor memory (OR = 1.38; 95% CI = 1.17-1.63) and lower odds of being classified with superior memory (OR = 0.81; 95% CI = 0.67-0.97). CONCLUSION Exposure to PM2.5 and NO2 are associated with patterns of cognitive performance characterized by worse verbal episodic memory relative to performance in other domains.
Collapse
Affiliation(s)
- Andrew J Petkus
- University of Southern California, Department of Neurology, Los Angeles, CA, USA
| | - Diana Younan
- University of Southern California, Department of Population and Public Health Sciences, Los Angeles, CA, USA
| | - Xinhui Wang
- University of Southern California, Department of Neurology, Los Angeles, CA, USA
| | - Daniel P Beavers
- Wake Forest School of Medicine, Department of Biostatistics, Winston-Salem, NC, USA
| | - Mark A Espeland
- Wake Forest School of Medicine, Department of Biostatistics, Winston-Salem, NC, USA
| | - Margaret Gatz
- University of Southern California, Center for Economic and Social Research, Los Angeles, CA, USA
| | - Tara Gruenewald
- Chapman University, Department of Psychology, Orange, CA, USA
| | - Joel D Kaufman
- University of Washington, Department of Environmental and Occupational Health Sciences, Seattle, WA, USA
| | - Helena C Chui
- University of Southern California, Department of Neurology, Los Angeles, CA, USA
| | - Joshua Millstein
- University of Southern California, Department of Population and Public Health Sciences, Los Angeles, CA, USA
| | - Stephen R Rapp
- Wake Forest School of Medicine, Department of Psychiatry and Behavioral Medicine, Winston-Salem, NC, USA
| | - JoAnn E Manson
- Harvard Medical School, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Susan M Resnick
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD, USA
| | - Gregory A Wellenius
- Boston University, Boston, Department of Environmental Health, Boston, MA, USA
| | - Eric A Whitsel
- University of North Carolina, Departments of Epidemiology and Medicine, Chapel Hill, NC, USA
| | - Keith Widaman
- University of California, Riverside, Graduate School of Education, Riverside, CA, USA
| | - Jiu-Chiuan Chen
- University of Southern California, Department of Neurology, Los Angeles, CA, USA
- University of Southern California, Department of Population and Public Health Sciences, Los Angeles, CA, USA
| |
Collapse
|
45
|
Shaffer RM, Li G, Adar SD, Dirk Keene C, Latimer CS, Crane PK, Larson EB, Kaufman JD, Carone M, Sheppard L. Fine Particulate Matter and Markers of Alzheimer's Disease Neuropathology at Autopsy in a Community-Based Cohort. J Alzheimers Dis 2021; 79:1761-1773. [PMID: 33459717 PMCID: PMC8061707 DOI: 10.3233/jad-201005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Evidence links fine particulate matter (PM2.5) to Alzheimer's disease (AD), but no community-based prospective cohort studies in older adults have evaluated the association between long-term exposure to PM2.5 and markers of AD neuropathology at autopsy. OBJECTIVE Using a well-established autopsy cohort and new spatiotemporal predictions of air pollution, we evaluated associations of 10-year PM2.5 exposure prior to death with Braak stage, Consortium to Establish a Registry for AD (CERAD) score, and combined AD neuropathologic change (ABC score). METHODS We used autopsy specimens (N = 832) from the Adult Changes in Thought (ACT) study, with enrollment ongoing since 1994. We assigned long-term exposure at residential address based on two-week average concentrations from a newly developed spatiotemporal model. To account for potential selection bias, we conducted inverse probability weighting. Adjusting for covariates with tiered models, we performed ordinal regression for Braak and CERAD and logistic regression for dichotomized ABC score. RESULTS 10-year average (SD) PM2.5 from death across the autopsy cohort was 8.2 (1.9) μg/m3. Average age (SD) at death was 89 (7) years. Each 1μg/m3 increase in 10-year average PM2.5 prior to death was associated with a suggestive increase in the odds of worse neuropathology as indicated by CERAD score (OR: 1.35 (0.90, 1.90)) but a suggestive decreased odds of neuropathology as defined by the ABC score (OR: 0.79 (0.49, 1.19)). There was no association with Braak stage (OR: 0.99 (0.64, 1.47)). CONCLUSION We report inconclusive associations between PM2.5 and AD neuropathology at autopsy among a cohort where 94% of individuals experienced 10-year exposures below the current EPA standard. Prior studies of AD risk factors and AD neuropathology are similarly inconclusive, suggesting alternative mechanistic pathways for disease or residual confounding.
Collapse
Affiliation(s)
- Rachel M. Shaffer
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
| | - Ge Li
- VA Northwest Network Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Sara D. Adar
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - C. Dirk Keene
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Caitlin S. Latimer
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Paul K. Crane
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Eric B. Larson
- School of Medicine, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Joel D. Kaufman
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
- Departments of Medicine and Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Lianne Sheppard
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| |
Collapse
|
46
|
Finch CE, Morgan TE. Developmental Exposure to Air Pollution, Cigarettes, and Lead: Implications for Brain Aging. ACTA ACUST UNITED AC 2020. [DOI: 10.1146/annurev-devpsych-042320-044338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brain development is impaired by maternal exposure to airborne toxins from ambient air pollution, cigarette smoke, and lead. Shared postnatal consequences include gray matter deficits and abnormal behaviors as well as elevated blood pressure. These unexpectedly broad convergences have implications for later life brain health because these same airborne toxins accelerate brain aging. Gene-environment interactions are shown for ApoE alleles that influence the risk of Alzheimer disease. The multigenerational trace of these toxins extends before fertilization because egg cells are formed in the grandmaternal uterus. The lineage and sex-specific effects of grandmaternal exposure to lead and cigarettes indicate epigenetic processes of relevance to future generations from our current and recent exposure to airborne toxins.
Collapse
Affiliation(s)
- Caleb E. Finch
- Leonard Davis School of Gerontology and Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089-0191, USA;,
| | - Todd E. Morgan
- Leonard Davis School of Gerontology and Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089-0191, USA;,
| |
Collapse
|
47
|
Armstrong TD, Suwannasual U, Kennedy CL, Thasma A, Schneider LJ, Phillippi D, Lund AK. Exposure to Traffic-Generated Pollutants Exacerbates the Expression of Factors Associated with the Pathophysiology of Alzheimer’s Disease in Aged C57BL/6 Wild-Type Mice. J Alzheimers Dis 2020; 78:1453-1471. [DOI: 10.3233/jad-200929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Multiple studies report a strong correlation between traffic-generated air pollution-exposure and detrimental outcomes in the central nervous system (CNS), including Alzheimer’s disease (AD). Incidence of AD is rapidly increasing and, worldwide, many live in regions where pollutants exceed regulatory standards. Thus, it is imperative to identify environmental pollutants that contribute to AD, and the mechanisms involved. Objective: We investigated the effects of mixed gasoline and diesel engine emissions (MVE) on the expression of factors involved in progression of AD in the hippocampus and cerebrum in a young versus aged mouse model. Methods: Young (2 months old) and aged (18 months old) male C57BL/6 mice were exposed to either MVE (300μg/m3 PM) or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-qPCR were used to quantify oxidative stress (8-OHdG) and expression of amyloid-β protein precursor (AβPP), β secretase (BACE1), amyloid-β (Aβ), aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) 1B1, angiotensin-converting enzyme (ACE1), and angiotensin II type 1 (AT1) receptor in the cerebrum and hippocampus, in addition to cerebral microvascular tight junction (TJ) protein expression. Results: We observed age-related increases in oxidative stress, AhR, CYP1B1, Aβ, BACE1, and AT1 receptor in the CA1 region of the hippocampus, and elevation of cerebral AβPP, AhR, and CYP1B1 mRNA, associated with decreased cerebral microvascular TJ protein claudin-5. MVE-exposure resulted in further promotion of oxidative stress, and significant increases in AhR, CYP1B1, BACE1, ACE1, and Aβ, compared to the young and aged FA-exposed mice. Conclusion: Such findings suggest that MVE-exposure exacerbates the expression of factors in the CNS associated with AD pathogenesis in aged populations.
Collapse
Affiliation(s)
- Tyler D. Armstrong
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Usa Suwannasual
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Conner L. Kennedy
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Akshaykumar Thasma
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Leah J. Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Amie K. Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| |
Collapse
|
48
|
Green EH, Kikis EA. Determining the effects of nanoparticulate air pollution on proteostasis in Caenorhabditis elegans. PLoS One 2020; 15:e0243419. [PMID: 33270781 PMCID: PMC7714337 DOI: 10.1371/journal.pone.0243419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/20/2020] [Indexed: 11/24/2022] Open
Abstract
The proteostasis network comprises the biochemical pathways that together maintain and regulate proper protein synthesis, transport, folding, and degradation. Many neurodegenerative diseases are characterized by a failure of the proteostasis network to sustain the health of the proteome, resulting in protein misfolding, aggregation, and, often, neurotoxicity. Although important advances have been made in recent years to identify genetic risk factors for neurodegenerative diseases, we still know relatively little about environmental risk factors such as air pollution. Exposure to nano-sized particulate air pollution, referred to herein as nanoparticulate matter (nPM), has been shown to trigger the accumulation of misfolded and oligomerized amyloid beta in mice. This suggests that the ability to maintain proteostasis is likely compromised in Alzheimer 's disease (AD) pathogenesis upon exposure to nPM. We aim to determine whether this aspect of the environment interacts with proteostasis network machinery to trigger protein misfolding. This could at least partially explain how air pollution exacerbates the symptoms of neurodegenerative diseases of aging, such as AD. We hypothesize that nPM challenges the buffering capacity of the proteostasis network by reducing the efficiency of folding for metastable proteins, thereby disrupting what has proven to be a very delicate proteostasis balance. We will test this hypothesis using C. elegans as our model system. Specifically, we will determine the impact of particulate air pollution on the aggregation and toxicity of disease-associated reporters of proteostasis and on transcriptional responses to stress.
Collapse
Affiliation(s)
- Emily H. Green
- Biology Department, The University of the South, Sewanee, TN, United States of America
| | - Elise A. Kikis
- Biology Department, The University of the South, Sewanee, TN, United States of America
- * E-mail:
| |
Collapse
|
49
|
Greve HJ, Mumaw CL, Messenger EJ, Kodavanti PRS, Royland JL, Kodavanti UP, Block ML. Diesel exhaust impairs TREM2 to dysregulate neuroinflammation. J Neuroinflammation 2020; 17:351. [PMID: 33222683 PMCID: PMC7682066 DOI: 10.1186/s12974-020-02017-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/29/2020] [Indexed: 01/07/2023] Open
Abstract
Background Air pollution has been linked to neurodegenerative diseases, including Alzheimer’s disease (AD), and the underlying neuroimmune mechanisms remain poorly understood. TREM2 is a myeloid cell membrane receptor that is a key regulator of disease-associated microglia (DAM) cells, where loss-of-function TREM2 mutations are associated with an increased risk of AD. At present, the basic function of TREM2 in neuroinflammation is a point of controversy. Further, the impact of air pollution on TREM2 and the DAM phenotype is largely unknown. Using diesel exhaust (DE) as a model of urban air pollution exposure, we sought to address its impact on TREM2 expression, the DAM phenotype, the association of microglia with the neurovasculature, and the role of TREM2 in DE-induced neuroinflammation. Methods WYK rats were exposed for 4 weeks to DE (0, 50, 150, 500 μg/m3) by inhalation. DE particles (DEP) were administered intratracheally once (600 μg/mouse) or 8 times (100 μg/mouse) across 28 days to male mice (Trem2+/+, Trem2−/−, PHOX+/+, and PHOX−/−). Results Rats exposed to DE exhibited inverted-U patterns of Trem2 mRNA expression in the hippocampus and frontal cortex, while TREM2 protein was globally diminished, indicating impaired TREM2 expression. Analysis of DAM markers Cx3Cr1, Lyz2, and Lpl in the frontal cortex and hippocampus showed inverted-U patterns of expression as well, supporting dysregulation of the DAM phenotype. Further, microglial-vessel association decreased with DE inhalation in a dose-dependent manner. Mechanistically, intratracheal administration of DEP increased Tnf (TNFα), Ncf1 (p47PHOX), and Ncf2 (p67PHOX) mRNA expression in only Trem2+/+ mice, where Il1b (IL-1β) expression was elevated in only Trem2−/− mice, emphasizing an important role for TREM2 in DEP-induced neuroinflammation. Conclusions Collectively, these findings reveal a novel role for TREM2 in how air pollution regulates neuroinflammation and provides much needed insight into the potential mechanisms linking urban air pollution to AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02017-7.
Collapse
Affiliation(s)
- Hendrik J Greve
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB 214D, Indianapolis, IN, 46202, USA
| | - Christen L Mumaw
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB 214D, Indianapolis, IN, 46202, USA
| | - Evan J Messenger
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB 214D, Indianapolis, IN, 46202, USA
| | - Prasada R S Kodavanti
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Joyce L Royland
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Urmila P Kodavanti
- Cardiopulmonary and Immunotoxicology Branch, Public Health and Integrated Toxicology Division, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Michelle L Block
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB 214D, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Daiber A, Kuntic M, Hahad O, Delogu LG, Rohrbach S, Di Lisa F, Schulz R, Münzel T. Effects of air pollution particles (ultrafine and fine particulate matter) on mitochondrial function and oxidative stress - Implications for cardiovascular and neurodegenerative diseases. Arch Biochem Biophys 2020; 696:108662. [PMID: 33159890 DOI: 10.1016/j.abb.2020.108662] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Environmental pollution is a major cause of global mortality and burden of disease. All chemical pollution forms together may be responsible for up to 12 million annual excess deaths as estimated by the Lancet Commission on pollution and health as well as the World Health Organization. Ambient air pollution by particulate matter (PM) and ozone was found to be associated with an all-cause mortality rate of up to 9 million in the year 2015, with the majority being of cerebro- and cardiovascular nature (e.g. stroke and ischemic heart disease). Recent evidence suggests that exposure to airborne particles and gases contributes to and accelerates neurodegenerative diseases. Especially, airborne toxic particles contribute to these adverse health effects. Whereas it is well established that air pollution in the form of PM may lead to dysregulation of neurohormonal stress pathways and may trigger inflammation as well as oxidative stress, leading to secondary damage of cardiovascular structures, the mechanistic impact of PM-induced mitochondrial damage and dysfunction is not well established. With the present review we will discuss similarities between mitochondrial damage and dysfunction observed in the development and progression of cardiovascular disease and neurodegeneration as well as those adverse mitochondrial pathomechanisms induced by airborne PM.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg University, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Marin Kuntic
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg University, Germany
| | - Omar Hahad
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg University, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Lucia G Delogu
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Susanne Rohrbach
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg University, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|