1
|
Zhang L, Yao T, Luo J, Yi H, Han X, Pan W, Xue Q, Liu X, Fu J, Zhang A. ChemNTP: Advanced Prediction of Neurotoxicity Targets for Environmental Chemicals Using a Siamese Neural Network. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22646-22656. [PMID: 39661815 DOI: 10.1021/acs.est.4c10081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Environmental chemicals can enter the human body through various exposure pathways, potentially leading to neurotoxic effects that pose significant health risks. Many such chemicals have been identified as neurotoxic, but the molecular mechanisms underlying their toxicity, including specific binding targets, remain unclear. To address this, we developed ChemNTP, a predictive model for identifying neurotoxicity targets of environmental chemicals. ChemNTP integrates a comprehensive representation of chemical structures and biological targets, improving upon traditional methods that are limited to single targets and mechanisms. By leveraging these structural representations, ChemNTP enables rapid screening across 199 potential neurotoxic targets or key molecular initiating events (MIEs). The model demonstrates robust predictive performance, achieving an area under the receiver operating characteristic curve (AUCROC) of 0.923 on the test set. Additionally, ChemNTP's attention mechanism highlights critical residues in binding targets and key functional groups or atoms in molecules, offering insights into the structural basis of interactions. Experimental validation through in vitro enzyme activity assays and molecular docking confirmed the binding of eight polybrominated diphenyl ethers (PBDEs) to acetylcholinesterase (AChE). We also provide a user-friendly software interface to facilitate the rapid identification of neurotoxicity targets for emerging environmental pollutants, with potential applications in studying MIEs for more types of toxicity.
Collapse
Affiliation(s)
- Lingjing Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Tingji Yao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Jiaqi Luo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Hang Yi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Xiaoxiao Han
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Wenxiao Pan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Qiao Xue
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Xian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Jianjie Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| |
Collapse
|
2
|
Cui J, Xiao S, Cao Y, Zhang Y, Yang J, Zheng L, Zhao F, Liu X, Liu D, Zhou Z, Wang P. Organophosphate Insecticide Malathion Induces Alzheimer's Disease-Like Cognitive Impairment in Mice: Evidence of the Microbiota-Gut-Brain Axis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:21966-21977. [PMID: 39545844 DOI: 10.1021/acs.est.4c07427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Evidence suggests that exposure to organophosphate pesticides increases the risk of neurodegenerative diseases, but the mechanisms remain unclear. This study investigated the effects of malathion on Alzheimer's disease (AD)-like symptoms at environmentally relevant concentrations using wild-type (WT) and APP/PS1 transgenic mouse models. Results showed that malathion exposure induced AD-like cognitive impairment, amyloid-β (Aβ) accumulation, and neuroinflammation in WT mice, with worsened symptoms in APP/PS1 mice. Mechanistic studies revealed that malathion induced AD-like gut microbiota dysbiosis (reduced Lactobacillus and Akkermansia, and increased Dubosiella), causing gut barrier impairment and tryptophan metabolism disruptions. This resulted in a significant increase in indole derivatives and activation of the colonic aryl hydrocarbon receptor (AhR), promoting the kynurenine (KYN) pathway while inhibiting the serotonin (5-HT) pathway. Increased neurotoxic KYN metabolites (3-hydroxykynurenine and quinolinic acid) triggered gut and systemic inflammation, upregulating hippocampal IL-6 and IL-1β mRNA levels and thereby causing neuroinflammation. Gut tryptophan metabolism disruptions caused hippocampal neurotransmitter imbalances, reducing the levels of 5-HT and its derivatives. These effects promoted AD progression in both WT and APP/PS1 mice. This study highlights the crucial role of the microbiota-gut-brain axis in AD-like cognitive impairment induced by malathion exposure, providing insights into the neurodegenerative disease risks posed by organophosphate pesticides.
Collapse
Affiliation(s)
- Jingna Cui
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Shouchun Xiao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yue Cao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yaru Zhang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Jiaxing Yang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Li Zheng
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Fanrong Zhao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Xueke Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Donghui Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Peng Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| |
Collapse
|
3
|
Khandayataray P, Murthy MK. Dietary interventions in mitigating the impact of environmental pollutants on Alzheimer's disease - A review. Neuroscience 2024; 563:148-166. [PMID: 39542342 DOI: 10.1016/j.neuroscience.2024.11.020] [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: 08/25/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Numerous studies linking environmental pollutants to oxidative stress, inflammation, and neurotoxicity have assigned pollutants to several neurodegenerative disorders, including Alzheimer's disease (AD). Heavy metals, pesticides, air pollutants, and endocrine disruptor chemicals have been shown to play important roles in AD development, with some traditional functions in amyloid-β formation, tau kinase action, and neuronal degeneration. However, pharmacological management and supplementation have resulted in limited improvement. This raises the interesting possibility that activities usually considered preventive, including diet, exercise, or mental activity, might be more similar to treatment or therapy for AD. This review focuses on the effects of diet on the effects of environmental pollutants on AD. One of the primary issues addressed in this review is a group of specific diets, including the Mediterranean diet (MeDi), Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH intervention for Neurodegenerative Delay (MIND), which prevent exposure to these toxins. Such diets have been proven to decrease oxidative stress and inflammation, which are unfavorable for neuronal growth. Furthermore, they contribute to positive changes in the composition of the human gut microbiota and thus encourage interactions in the Gut-Brain Axis, reducing inflammation caused by pollutants. This review emphasizes a multi-professional approach with reference to nutritional activities that would lower the neurotoxic load in populations with a high level of exposure to pollutants. Future studies focusing on diet and environment association plans may help identify preventive measures aimed at enhancing current disease deceleration.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
4
|
Zhang H, Zhang C, Wang Q, Fu W, Xing W, Jin P, Wu H, Bu Y, Xu D, Xu D. PFOS sub-chronic exposure selectively activates Aβ clearance pathway to improve the cognitive ability of AD mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125031. [PMID: 39454812 DOI: 10.1016/j.envpol.2024.125031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024]
Abstract
Perfluorooctane sulfonate (PFOS), an emerging persistent organic pollutant, has been controversial in its impact on cognitive functions. Our previous research has confirmed that the sub-chronic PFOS exposure leads to neuronal apoptosis in the cerebral cortex, impairing cognitive functions in normal mice. However, our current study presents a surprising finding: sub-chronic exposure to PFOS effectively reduces cognitive impairments in Alzheimer's disease (AD) mice and significantly retards the disease's progression. Our results indicate that PFOS exposure upregulates the expression level of insulin-degrading enzyme (IDE) in the prefrontal cortex (PFC) of AD mice, thereby selectively enhancing the amyloid-beta (Aβ) clearance pathway without affecting the Aβ production. Moreover, PFOS exposure inhibits microglial proliferation and reduces inflammatory cytokines levels in the PFC of AD mice, providing further supporting for the pivotal role of IDE in attenuating AD progression under PFOS exposure. Collectively, our study is the first to demonstrate that sub-chronic PFOS exposure can alleviates cognitive impairments in AD pathology, with the IDE-mediated Aβ clearance pathway potentially playing a critical role.
Collapse
Affiliation(s)
- Haijing Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Chao Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Qin Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Wenliang Fu
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Weiwei Xing
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Peng Jin
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Haowei Wu
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Yuanjing Bu
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China
| | - Dongqun Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China.
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, Beijing, 100039, China.
| |
Collapse
|
5
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [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: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
6
|
Song Z, Lynch K, Parker-Allotey NA, Bennett EE, Xu X, Whitsel EA, Smith R, Stewart JD, Park ES, Ying Q, Power MC. Association of midlife air pollution exposures and residential road proximity with incident dementia: The Atherosclerosis Risk in Communities (ARIC) study. ENVIRONMENTAL RESEARCH 2024; 258:119425. [PMID: 38879108 PMCID: PMC11323165 DOI: 10.1016/j.envres.2024.119425] [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: 03/22/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Increasing evidence links higher air pollution exposures to increased risk of cognitive impairment. While midlife risk factors are often most strongly linked to dementia risk, few studies have considered associations between midlife roadway proximity or ambient air pollution exposure and incident dementia decades later, in late life. OBJECTIVES Our objective was to determine if midlife exposures to ambient air pollution or roadway proximity are associated with increased risk of dementia in the Atherosclerosis Risk in Communities (ARIC) study over up to 29 years of follow-up. METHODS Our eligible sample included Black and White ARIC participants without dementia at Visit 2 (1990-1992). Participants were followed through Visit 7 (2018-2019), with dementia status and onset date defined based on formal dementia ascertainment at study visits, informant interviews, and surveillance efforts. We used adjusted Weibull survival models to assess the associations of midlife ambient air pollution and road proximity with incident dementia. RESULTS The median age at baseline (1990-1992, Visit 2) of the 12,700 eligible ARIC participants was 57.0 years; 56.0% were female, 24.2% were Black, and 78.9% had at least a high school education. Over up to 29 years of follow-up, 2511 (19.8%) persons developed dementia. No associations were found between ambient air pollutants and proximity to major roadways with risk of incident dementia. In exploratory analyses, living closer to roadways in midlife increased dementia risk in individuals younger at baseline and those without midlife hypertension, and there was evidence of increased risk of dementia with increased midlife exposure to NOx, several PM2.5 components, and trace metals among those with diabetes in midlife. CONCLUSIONS Midlife exposure to ambient air pollution and midlife roadway proximity was not associated with dementia risk over decades of follow-up. Further investigation to explore potential for greater susceptibility among specific subgroups identified here is needed.
Collapse
Affiliation(s)
- Ziwei Song
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Katie Lynch
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Naa Adoley Parker-Allotey
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Erin E Bennett
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Xiaohui Xu
- School of Public Health, Texas A&M Health Science Center, College Station, TX, United States
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States; Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Richard Smith
- Department of Statistics and Operations Research, College of Arts and Sciences, University of North Carolina, Chapel Hill, NC, United States; Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States
| | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States
| | - Eun Sug Park
- Texas A&M Transportation Institute, Texas A&M University System, College Station, TX, United States
| | - Qi Ying
- Zachry Department of Civil Engineering, Texas A&M University, College Station, TX, 77843, United States
| | - Melinda C Power
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States.
| |
Collapse
|
7
|
Phillips MCL, Picard M. Neurodegenerative disorders, metabolic icebergs, and mitohormesis. Transl Neurodegener 2024; 13:46. [PMID: 39242576 PMCID: PMC11378521 DOI: 10.1186/s40035-024-00435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/25/2024] [Indexed: 09/09/2024] Open
Abstract
Neurodegenerative disorders are typically "split" based on their hallmark clinical, anatomical, and pathological features, but they can also be "lumped" by a shared feature of impaired mitochondrial biology. This leads us to present a scientific framework that conceptualizes Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD) as "metabolic icebergs" comprised of a tip, a bulk, and a base. The visible tip conveys the hallmark neurological symptoms, neurodegenerative regions, and neuronal protein aggregates for each disorder. The hidden bulk depicts impaired mitochondrial biology throughout the body, which is multifaceted and may be subdivided into impaired cellular metabolism, cell-specific mitotypes, and mitochondrial behaviours, functions, activities, and features. The underlying base encompasses environmental factors, especially modern industrial toxins, dietary lifestyles, and cognitive, physical, and psychosocial behaviours, but also accommodates genetic factors specific to familial forms of AD, PD, and ALS, as well as HD. Over years or decades, chronic exposure to a particular suite of environmental and genetic factors at the base elicits a trajectory of impaired mitochondrial biology that maximally impacts particular subsets of mitotypes in the bulk, which eventually surfaces as the hallmark features of a particular neurodegenerative disorder at the tip. We propose that impaired mitochondrial biology can be repaired and recalibrated by activating "mitohormesis", which is optimally achieved using strategies that facilitate a balanced oscillation between mitochondrial stressor and recovery phases. Sustainably harnessing mitohormesis may constitute a potent preventative and therapeutic measure for people at risk of, or suffering with, neurodegenerative disorders.
Collapse
Affiliation(s)
- Matthew C L Phillips
- Department of Neurology, Waikato Hospital, Hamilton, 3204, New Zealand.
- Department of Medicine, University of Auckland, Auckland, 1142, New Zealand.
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
8
|
Lim L. Modifying Alzheimer's disease pathophysiology with photobiomodulation: model, evidence, and future with EEG-guided intervention. Front Neurol 2024; 15:1407785. [PMID: 39246604 PMCID: PMC11377238 DOI: 10.3389/fneur.2024.1407785] [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: 03/27/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
This manuscript outlines a model of Alzheimer's Disease (AD) pathophysiology in progressive layers, from its genesis to the development of biomarkers and then to symptom expression. Genetic predispositions are the major factor that leads to mitochondrial dysfunction and subsequent amyloid and tau protein accumulation, which have been identified as hallmarks of AD. Extending beyond these accumulations, we explore a broader spectrum of pathophysiological aspects, including the blood-brain barrier, blood flow, vascular health, gut-brain microbiodata, glymphatic flow, metabolic syndrome, energy deficit, oxidative stress, calcium overload, inflammation, neuronal and synaptic loss, brain matter atrophy, and reduced growth factors. Photobiomodulation (PBM), which delivers near-infrared light to selected brain regions using portable devices, is introduced as a therapeutic approach. PBM has the potential to address each of these pathophysiological aspects, with data provided by various studies. They provide mechanistic support for largely small published clinical studies that demonstrate improvements in memory and cognition. They inform of PBM's potential to treat AD pending validation by large randomized controlled studies. The presentation of brain network and waveform changes on electroencephalography (EEG) provide the opportunity to use these data as a guide for the application of various PBM parameters to improve outcomes. These parameters include wavelength, power density, treatment duration, LED positioning, and pulse frequency. Pulsing at specific frequencies has been found to influence the expression of waveforms and modifications of brain networks. The expression stems from the modulation of cellular and protein structures as revealed in recent studies. These findings provide an EEG-based guide for the use of artificial intelligence to personalize AD treatment through EEG data feedback.
Collapse
Affiliation(s)
- Lew Lim
- Vielight Inc., Toronto, ON, Canada
| |
Collapse
|
9
|
Islam MR, Rabbi MA, Hossain T, Sultana S, Uddin S. Mechanistic Approach to Immunity and Immunotherapy of Alzheimer's Disease: A Review. ACS Chem Neurosci 2024. [PMID: 39173186 DOI: 10.1021/acschemneuro.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative condition characterized by progressive cognitive decline and memory loss, affecting millions of people worldwide. Traditional treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, offer limited symptomatic relief without addressing the underlying disease mechanisms. These limitations have driven the development of more potent and effective therapies. Recent advances in immunotherapy present promising avenues for AD treatment. Immunotherapy strategies, including both active and passive approaches, harness the immune system to target and mitigate AD-related pathology. Active immunotherapy stimulates the patient's immune response to produce antibodies against AD-specific antigens, while passive immunotherapy involves administering preformed antibodies or immune cells that specifically target amyloid-β (Aβ) or tau proteins. Monoclonal antibodies, such as aducanumab and lecanemab, have shown potential in reducing Aβ plaques and slowing cognitive decline in clinical trials, despite challenges related to adverse immune responses and the need for precise targeting. This comprehensive review explores the role of the immune system in AD, evaluates the current successes and limitations of immunotherapeutic approaches, and discusses future directions for enhancing the treatment efficacy.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md Afser Rabbi
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tanbir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Sadia Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Shihab Uddin
- Department of Bioengineering, King Fahad University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Bio Systems and Machines, King Fahad University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| |
Collapse
|
10
|
Wei S, Xu T, Sang N, Yue H, Chen Y, Jiang T, Jiang T, Yin D. Mixed Metal Components in PM 2.5 Contribute to Chemokine Receptor CCR5-Mediated Neuroinflammation and Neuropathological Changes in the Mouse Olfactory Bulb. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:4914-4925. [PMID: 38436231 DOI: 10.1021/acs.est.3c08506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Particulate matter, especially PM2.5, can invade the central nervous system (CNS) via the olfactory pathway to induce neurotoxicity. The olfactory bulb (OB) is the key component integrating immunoprotection and olfaction processing and is necessarily involved in the relevant CNS health outcomes. Here we show that a microglial chemokine receptor, CCR5, is the target of environmentally relevant PM2.5 in the OB to trigger neuroinflammation and then neuropathological injuries. Mechanistically, PM2.5-induced CCR5 upregulation results in the pro-inflammatory paradigm of microglial activation, which subsequently activates TLR4-NF-κB neuroinflammation signaling and induces neuropathological changes that are closely related to neurodegenerative disorders (e.g., Aβ deposition and disruption of the blood-brain barrier). We specifically highlight that manganese and lead in PM2.5 are the main contributors to CCR5-mediated microglial activation and neuroinflammation in synergy with aluminum. Our results uncover a possible pathway of PM2.5-induced neuroinflammation and identify the principal neurotoxic components, which can provide new insight into efficiently diminishing the adverse health effects of PM2.5.
Collapse
Affiliation(s)
- Sheng Wei
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Ting Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
- Department of Key Laboratory, Changshu No.2 People's Hospital, Changshu 215500, China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Yawen Chen
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
- Postdoctoral Research Station of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Tao Jiang
- Lyon Neuroscience Research Center (CRNL), Sensory Neuro-Ethology Team, 59 Bd Pinel, Bron 69500, France
| | - Tingwang Jiang
- Department of Key Laboratory, Changshu No.2 People's Hospital, Changshu 215500, China
| | - Daqiang Yin
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
- Department of Key Laboratory, Changshu No.2 People's Hospital, Changshu 215500, China
| |
Collapse
|
11
|
Yadav B, Kaur S, Yadav A, Verma H, Kar S, Sahu BK, Pati KR, Sarkar B, Dhiman M, Mantha AK. Implications of organophosphate pesticides on brain cells and their contribution toward progression of Alzheimer's disease. J Biochem Mol Toxicol 2024; 38:e23660. [PMID: 38356323 DOI: 10.1002/jbt.23660] [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: 09/12/2023] [Revised: 01/04/2024] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
The most widespread neurodegenerative disorder, Alzheimer's disease (AD) is marked by severe behavioral abnormalities, cognitive and functional impairments. It is inextricably linked with the deposition of amyloid β (Aβ) plaques and tau protein in the brain. Loss of white matter, neurons, synapses, and reactive microgliosis are also frequently observed in patients of AD. Although the causative mechanisms behind the neuropathological alterations in AD are not fully understood, they are likely influenced by hereditary and environmental factors. The etiology and pathogenesis of AD are significantly influenced by the cells of the central nervous system, namely, glial cells and neurons, which are directly engaged in the transmission of electrical signals and the processing of information. Emerging evidence suggests that exposure to organophosphate pesticides (OPPs) can trigger inflammatory responses in glial cells, leading to various cascades of events that contribute to neuroinflammation, neuronal damage, and ultimately, AD pathogenesis. Furthermore, there are striking similarities between the biomarkers associated with AD and OPPs, including neuroinflammation, oxidative stress, dysregulation of microRNA, and accumulation of toxic protein aggregates, such as amyloid β. These shared markers suggest a potential mechanistic link between OPP exposure and AD pathology. In this review, we attempt to address the role of OPPs on altered cell physiology of the brain cells leading to neuroinflammation, mitochondrial dysfunction, and oxidative stress linked with AD pathogenesis.
Collapse
Affiliation(s)
- Bharti Yadav
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Anuradha Yadav
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Swastitapa Kar
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Binit Kumar Sahu
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Kumari Riya Pati
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Bibekanada Sarkar
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
12
|
Petit P, Gondard E, Gandon G, Moreaud O, Sauvée M, Bonneterre V. Agricultural activities and risk of Alzheimer's disease: the TRACTOR project, a nationwide retrospective cohort study. Eur J Epidemiol 2024; 39:271-287. [PMID: 38195954 PMCID: PMC10995077 DOI: 10.1007/s10654-023-01079-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 11/02/2023] [Indexed: 01/11/2024]
Abstract
Data regarding Alzheimer's disease (AD) occurrence in farming populations is lacking. This study aimed to investigate whether, among the entire French farm manager (FM) workforce, certain agricultural activities are more strongly associated with AD than others, using nationwide data from the TRACTOR (Tracking and monitoring occupational risks in agriculture) project. Administrative health insurance data (digital electronic health/medical records and insurance claims) for the entire French agricultural workforce, over the period 2002-2016, on the entire mainland France were used to estimate the risk of AD for 26 agricultural activities with Cox proportional hazards model. For each analysis (one for each activity), the exposed group included all FMs that performed the activity of interest (e.g. crop farming), while the reference group included all FMs who did not carry out the activity of interest (e.g. FMs that never farmed crops between 2002 and 2016). There were 5067 cases among 1,036,069 FMs who worked at least one year between 2002 and 2016. Analyses showed higher risks of AD for crop farming (hazard ratio (HR) = 3.72 [3.47-3.98]), viticulture (HR = 1.29 [1.18-1.42]), and fruit arboriculture (HR = 1.36 [1.15-1.62]). By contrast, lower risks of AD were found for several animal farming types, in particular for poultry and rabbit farming (HR = 0.29 [0.20-0.44]), ovine and caprine farming (HR = 0.50 [0.41-0.61]), mixed dairy and cow farming (HR = 0.46 [0.37-0.57]), dairy farming (HR = 0.67 [0.61-0.73]), and pig farming (HR = 0.30 [0.18-0.52]). This study shed some light on the association between a wide range of agricultural activities and AD in the entire French FMs population.
Collapse
Affiliation(s)
- Pascal Petit
- CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, Univ. Grenoble Alpes, 38000, Grenoble, France.
- Centre Régional de Pathologies Professionnelles et Environnementales, CHU Grenoble Alpes, 38000, Grenoble, France.
- AGEIS, Univ. Grenoble Alpes, 38000, Grenoble, France.
| | - Elise Gondard
- CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, Univ. Grenoble Alpes, 38000, Grenoble, France
| | - Gérald Gandon
- Centre Régional de Pathologies Professionnelles et Environnementales, CHU Grenoble Alpes, 38000, Grenoble, France
| | - Olivier Moreaud
- Centre Mémoire de Ressources et de Recherche, CHU Grenoble Alpes, 38000, Grenoble, France
- Laboratoire de Psychologie et Neurocognition, UMR 5105, CNRS, LPNC, Univ. Grenoble Alpes, Univ. Savoie Mont Blanc, 38000, Grenoble, France
| | - Mathilde Sauvée
- Centre Mémoire de Ressources et de Recherche, CHU Grenoble Alpes, 38000, Grenoble, France
- Laboratoire de Psychologie et Neurocognition, UMR 5105, CNRS, LPNC, Univ. Grenoble Alpes, Univ. Savoie Mont Blanc, 38000, Grenoble, France
| | - Vincent Bonneterre
- CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, Univ. Grenoble Alpes, 38000, Grenoble, France
- Centre Régional de Pathologies Professionnelles et Environnementales, CHU Grenoble Alpes, 38000, Grenoble, France
| |
Collapse
|
13
|
Wan C, Ma H, Liu J, Liu F, Liu J, Dong G, Zeng X, Li D, Yu Z, Wang X, Li J, Zhang G. Quantitative relationships of FAM50B and PTCHD3 methylation with reduced intelligence quotients in school aged children exposed to lead: Evidence from epidemiological and in vitro studies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:167976. [PMID: 37866607 DOI: 10.1016/j.scitotenv.2023.167976] [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: 06/26/2023] [Revised: 09/22/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
At present, the application of DNA methylation (DNAm) biomarkers in environmental health risk assessment (EHRA) is more challenging due to the unclearly quantitative relationship between them. We aimed to explore the role of FAM50B and PTCHD3 at the level of signaling pathways, and establish the quantitative relationship between them and children's intelligence quotients (IQs). DNAm of target regions was measured in multiple cell models and was compared with the human population data. Then the dose-response relationships of lead exposure with neurotoxicity and DNAm were established by benchmark dose (BMD) model, followed by potential signaling pathway screening. Results showed that there was a quantitative linear relationship between children's IQs and FAM50B/PTCHD3 DNAm (DNAm between 51.40 % - 78.78 % and 31.41 % - 74.19 % for FAM50B and PTCHD3, respectively), and this relationship was more significant when children's IQs > 90. The receiver operating characteristic (ROC) and calibration curves showed that FAM50B/PTCHD3 DNAm had a satisfying accuracy and consistency in predicting children's IQs, which was confirmed by sensitivity analysis of gender and CpG site grouping data. In cell experiments, there was also a quantitative linear relationship between FAM50B DNAm and reactive oxygen species (ROS) production, which was mediated by PI3K-AKT signaling pathway. In addition, the lead BMD of ROS was close to that of FAM50B DNAm, suggesting that FAM50B DNAm was a suitable biomarker for the risk assessments of adverse outcomes induced by lead. Taken collectively, these results suggest that FAM50B/PTCHD3 can be applied to EHRA and the prevention/intervention of adverse effects of lead on children's IQs.
Collapse
Affiliation(s)
- Cong Wan
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Huimin Ma
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China.
| | - Jiahong Liu
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Liu
- School of Business Administration, South China University of Technology, Guangzhou 510641, China
| | - Jing Liu
- Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Guanghui Dong
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaowen Zeng
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Xinming Wang
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Jun Li
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Gan Zhang
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| |
Collapse
|
14
|
Bjørklund G, Tippairote T, Hangan T, Chirumbolo S, Peana M. Early-Life Lead Exposure: Risks and Neurotoxic Consequences. Curr Med Chem 2024; 31:1620-1633. [PMID: 37031386 DOI: 10.2174/0929867330666230409135310] [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: 08/14/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Lead (Pb) does not have any biological function in a human, and it is likely no safe level of Pb in the human body. The Pb exposure impacts are a global concern for their potential neurotoxic consequences. Despite decreasing both the environmental Pb levels and the average blood Pb levels in the survey populations, the lifetime redistribution from the tissues-stored Pb still poses neurotoxic risks from the low-level exposure in later life. The growing fetus and children hold their innate high-susceptible to these Pb-induced neurodevelopmental and neurobehavioral effects. OBJECTIVE This article aims to evaluate cumulative studies and insights on the topic of Pb neurotoxicology while assessing the emerging trends in the field. RESULTS The Pb-induced neurochemical and neuro-immunological mechanisms are likely responsible for the high-level Pb exposure with the neurodevelopmental and neurobehavioral impacts at the initial stages. Early-life Pb exposure can still produce neurodegenerative consequences in later life due to the altered epigenetic imprints and the ongoing endogenous Pb exposure. Several mechanisms contribute to the Pb-induced neurotoxic impacts, including the direct neurochemical effects, the induction of oxidative stress and inflammation through immunologic activations, and epigenetic alterations. Furthermore, the individual nutritional status, such as macro-, micro-, or antioxidant nutrients, can significantly influence the neurotoxic impacts even at low-level exposure to Pb. CONCLUSION The prevention of early-life Pb exposure is, therefore, the critical determinant for alleviating various Pb-induced neurotoxic impacts across the different age groups.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, Mo i Rana, 8610, Norway
| | - Torsak Tippairote
- Department of Nutritional and Environmental Medicine, HP Medical Center, Bangkok 10540, Thailand
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, 900470, Romania
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
- CONEM Scientific Secretary, Strada Le Grazie 9, 37134, Verona, Italy
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, Sassari, 07100, Italy
| |
Collapse
|
15
|
Sande R, Godad A, Doshi G. Zebrafish Experimental Animal Models for AD: A Comprehensive Review. Curr Rev Clin Exp Pharmacol 2024; 19:295-311. [PMID: 38284707 DOI: 10.2174/0127724328279684240104094257] [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: 08/29/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 01/30/2024]
Abstract
AD disease (AD) is a multifaceted and intricate neurodegenerative disorder characterized by intracellular neurofibrillary tangle (NFT) formation and the excessive production and deposition of Aβ senile plaques. While transgenic AD models have been found instrumental in unravelling AD pathogenesis, they involve cost and time constraints during the preclinical phase. Zebrafish, owing to their simplicity, well-defined behavioural patterns, and relevance to neurodegenerative research, have emerged as a promising complementary model. Zebrafish possess glutaminergic and cholinergic pathways implicated in learning and memory, actively contributing to our understanding of neural transmission processes. This review sheds light on the molecular mechanisms by which various neurotoxic agents, including okadaic acid (OKA), cigarette smoke extract, metals, and transgenic zebrafish models with genetic similarities to AD patients, induce cognitive impairments and neuronal degeneration in mammalian systems. These insights may facilitate the identification of effective neurotoxic agents for replicating AD pathogenesis in the zebrafish brain. In this comprehensive review, the pivotal role of zebrafish models in advancing our comprehension of AD is emphasized. These models hold immense potential for shaping future research directions and clinical interventions, ultimately contributing to the development of novel AD therapies.
Collapse
Affiliation(s)
- Ruksar Sande
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Angel Godad
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| |
Collapse
|
16
|
Wang H, Bullert AJ, Li X, Stevens H, Klingelhutz AJ, Ankrum JA, Adamcakova-Dodd A, Thorne PS, Lehmler HJ. Use of a polymeric implant system to assess the neurotoxicity of subacute exposure to 2,2',5,5'-tetrachlorobiphenyl-4-ol, a human metabolite of PCB 52, in male adolescent rats. Toxicology 2023; 500:153677. [PMID: 37995827 PMCID: PMC10757425 DOI: 10.1016/j.tox.2023.153677] [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: 09/05/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic pollutants (POPs) that ubiquitously exist in the environment. PCB exposure has been linked to cancer and multi-system toxicity, including endocrine disruption, immune inhibition, and reproductive and neurotoxicity. 2,2',5,5'-Tetrachlorobiphenyl (PCB 52) is one of the most frequently detected congeners in the environment and human blood. The hydroxylated metabolites of PCB 52 may also be neurotoxic, especially for children whose brains are still developing. However, it is challenging to discern the contribution of these metabolites to PCB neurotoxicity because the metabolism of PCB is species-dependent. In this study, we evaluated the subacute neurotoxicity of a human-relevant metabolite, 2,2',5,5'-tetrachlorobiphenyl-4-ol (4-52), on male adolescent Sprague Dawley rats, via a novel polymeric implant drug delivery system grafted subcutaneously, at total loading concentrations ranging from 0%, 1%, 5%, and 10% of the implant (w/w) for 28 days. Y-maze, hole board test, open field test, and elevated plus maze were performed on exposure days 24-28 to assess their locomotor activity, and exploratory and anxiety-like behavior. 4-52 and other possible hydroxylated metabolites in serum and vital tissues were quantified using gas chromatography with tandem mass spectrometry (GC-MS/MS). Our results demonstrate the sustained release of 4-52 from the polymeric implants into the systemic circulation in serum and tissues. Dihydroxylated and dechlorinated metabolites were detected in serum and tissues, depending on the dose and tissue type. No statistically significant changes were observed in the neurobehavioral tasks across all exposure groups. The results demonstrate that subcutaneous polymeric implants provide a straightforward method to expose rats to phenolic PCB metabolites to study neurotoxic outcomes, e.g., in memory, anxiety, and exploratory behaviors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Amanda J Bullert
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA
| | - Xueshu Li
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Hanna Stevens
- Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Department of Psychiatry, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA
| | | | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, the University of Iowa, Iowa City, IA, USA
| | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA
| | - Peter S Thorne
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, the University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Human Toxicology, the University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
17
|
Zuin M, Rosta V, Trentini A, Bosi C, Zuliani G, Cervellati C. Paraoxonase 1 activity in patients with Alzheimer disease: Systematic review and meta-analysis. Chem Biol Interact 2023; 382:110601. [PMID: 37330180 DOI: 10.1016/j.cbi.2023.110601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023]
Abstract
Cumulating evidence links environmental toxicants, such as organophosphate (OP) pesticides, to the pathogenesis of Alzheimer's disease (AD). The calcium-dependent Paraoxonase 1 (PON1) can neutralize these toxicants with good catalytic efficiency, thus protecting from OP-induced biological damage. Although different previous studies have already partially described an association between PON1 activity and AD, this intriguing relationship has not yet been comprehensively examined. To fill this gap, we performed a meta-analysis of existing data comparing the PON1 arylesterase activity in AD and healthy subjects from the general population. Data were obtained by searching MEDLINE, Embase and CENTRAL, Google Scholar, and SCOPUS electronic databases for all studies published at any time up to February 2023, reporting and comparing the PON1- paraoxonase activity between AD patients and controls. Seven studies, based on 615 subjects (281 AD and 356 controls) met the inclusion criteria and were included into the final analysis. A random effect model revealed that PON1 arylesterase activity was significantly lower in the AD group compared to controls, exhibiting low level of heterogeneity (SMD = - 1.62, 95% CI = -2.65 to -0.58, p = 0.0021, I2 = 12%). These findings suggest that PON1 activity might be reduced in AD reflecting a major susceptibility to OPs neurotoxicity. Further studies should be conducted to definitely ascertain this link and to establish the cause-effect relationship between PON1 reduction and AD onset.
Collapse
Affiliation(s)
- Marco Zuin
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Valentina Rosta
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Alessandro Trentini
- Department of Environmental and Prevention Sciences, University of Ferrara, Italy
| | - Cristina Bosi
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy; Medical Department, University Hospital of Ferrara Arcispedale Sant'Anna, Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
18
|
Torres-Sánchez ED, Ortiz GG, Reyes-Uribe E, Torres-Jasso JH, Salazar-Flores J. Effect of pesticides on phosphorylation of tau protein, and its influence on Alzheimer's disease. World J Clin Cases 2023; 11:5628-5642. [PMID: 37727721 PMCID: PMC10506003 DOI: 10.12998/wjcc.v11.i24.5628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive and neurodegenerative illness which results in alterations in cognitive development. It is characterized by loss/dysfunction of cholinergic neurons, and formation of amyloid plaques, and formation of neurofibrillary tangles, among other changes, due to hyperphosphorylation of tau-protein. Exposure to pesticides in humans occurs frequently due to contact with contaminated food, water, or particles. Organochlorines, organophosphates, carbamates, pyrethroids and neonicotinoids are associated with the most diagnosed incidents of severe cognitive impairment. The aim of this study was to determine the effects of these pesticides on the phosphorylation of tau protein, and its cognitive implications in the development of AD. It was found that exposure to pesticides increased the phosphorylation of tau protein at sites Ser198, Ser199, Ser202, Thr205, Ser396 and Ser404. Contact with these chemicals altered the enzymatic activities of cyclin-dependent kinase 5 and glycogen synthase kinase 3 beta, and protein phosphatase-2A. Moreover, it altered the expression of the microtubule associated protein tau gene, and changed levels of intracellular calcium. These changes affected tau protein phosphorylation and neuroinflammation, and also increased oxidative stress. In addition, the exposed subjects had poor level of performance in tests that involved evaluation of novelty, as test on verbal, non-verbal, spatial memory, attention, and problem-solving skills.
Collapse
Affiliation(s)
- Erandis D Torres-Sánchez
- Department of Medical and Life Sciences, University Center of la Cienega, University of Guadalajara, Ocotlan 47820, Jalisco, Mexico
| | - Genaro G Ortiz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in Medicine Hospital Civil, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Emmanuel Reyes-Uribe
- Department of Medical and Life Sciences, University Center of la Cienega, University of Guadalajara, Ocotlan 47820, Jalisco, Mexico
| | - Juan H Torres-Jasso
- Department of Biological Sciences, CUCOSTA, University of Guadalajara, Puerto Vallarta 48280, Jalisco, Mexico
| | - Joel Salazar-Flores
- Department of Medical and Life Sciences, University Center of la Cienega, University of Guadalajara, Ocotlan 47820, Jalisco, Mexico
| |
Collapse
|
19
|
Kolińska J, Grzelakowska A, Szala M, Podsiadły R. Comparison of Reactive Sites in 2(1 H)-Quinolone Derivatives for the Detection of Biologically Important Sulfur Compounds. Molecules 2023; 28:5965. [PMID: 37630217 PMCID: PMC10459984 DOI: 10.3390/molecules28165965] [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: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Novel fluorescent probes based on 2(1H)-quinolone skeleton containing a malonate group (Q1-Q3) were synthesized and proposed for biothiols detection. Their chemical reactivity toward thiols was compared to the reactivity of derivative having a dicyanovinyl group (Q4) as a reactive site. The detailed photophysical properties of these compounds were assessed through the determination of absorption and fluorescence spectra, fluorescence quantum yield, and fluorescence lifetime. In the presence of biothiols, an increase in the fluorescence intensity of compounds Q1-Q3 and a hypsochromic shift in their emission bands were observed. In contrast, the compound with the dicyanovinyl group (Q4) in the presence of biothiols and cyanide ion showed the quenching of fluorescence, while a fluorescence "turn on" effect was observed toward reactive sulfur species.
Collapse
Affiliation(s)
- Jolanta Kolińska
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, 90-537 Lodz, Poland; (A.G.); (M.S.); (R.P.)
| | | | | | | |
Collapse
|
20
|
Moyano P, Sola E, Naval MV, Guerra-Menéndez L, Fernández MDLC, del Pino J. Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools. Pharmaceutics 2023; 15:2048. [PMID: 37631262 PMCID: PMC10458078 DOI: 10.3390/pharmaceutics15082048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Environmental pollutants' (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins' homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Maria De la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
21
|
Chang YT, Jung CR, Chang YC, Chuang BR, Chen ML, Hwang BF. Prenatal and postnatal exposure to PM 2 .5 and the risk of tic disorders. Paediatr Perinat Epidemiol 2023; 37:191-200. [PMID: 36562434 DOI: 10.1111/ppe.12943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Tic disorders are common neurodevelopmental disorders during childhood. Whether prenatal and postnatal exposure to particulate matter with an aerodynamic diameter less than 2.5 μm (PM2.5 ) plays a role in the development of tic disorders remains unexplored. OBJECTIVES To investigate the association of exposure between PM2.5 during the pregnancy and infancy periods and the risk of tic disorders. METHODS This birth cohort study recruited singleton live births at term gestations in central Taiwan from the Taiwan Maternal and Child Health Database between 2004 and 2012 and followed up to the end of 2017. New cases of tic disorders were defined using the ICD-9-CM (307.2) and ICD-10-CM (F95), which include all tic spectrum disorders. We assigned daily PM2.5 concentrations derived from a satellite-based model to individuals based on maternal residential addresses at delivery. We fit Cox proportional hazard model and distributed lag non-linear model to estimate the associations between PM2.5 and tic disorders, with hazard ratio (HR) with 95% confidence interval (CI) as the effect measure. RESULTS Of the 309,376 singleton live births at term gestations, we identified 5902 (1.9%) tic disorder cases. The HR of tic disorders was positively associated with a 10 μg/m3 increase in PM2.5 : during pregnancy HR 1.09, 95% CI 1.04, 1.15 and during infancy HR 1.12, 95% CI 1.06, 1.18. The vulnerable time window for infants with increased risk of tic disorders was 6-52 weeks after birth. We observed a nonlinear relationship between PM2.5 and the risk of tic disorders, with exposure to PM2.5 between 16 and 64 μg/m3 being associated with the risk of tic disorders. The association was restricted to Tourette's disorder group. Infant sex did not modify these associations. CONCLUSIONS Infants delivered at term and exposed to PM2.5 are associated with an increased risk of tic disorders (6-52 weeks). Further studies are needed to confirm these associations.
Collapse
Affiliation(s)
- Yu-Tzu Chang
- Division of Pediatric Neurology, China Medical University Children's Hospital, Taichung, Taiwan R.O.C
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan R.O.C
| | - Chau-Ren Jung
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan R.O.C
- Japan Environment and Children's Study Programme Office, National Institute for Environmental Studies, Tsukuba, Japan
| | - Ya-Chu Chang
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan R.O.C
| | - Bao-Ru Chuang
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan R.O.C
| | - Mei-Ling Chen
- College of Human Science and Social Innovation, HungKuang University, Taichung, Taiwan R.O.C
| | - Bing-Fang Hwang
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan R.O.C
- Department of Occupational Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan R.O.C
| |
Collapse
|
22
|
Eiser AR, Fulop T. Alzheimer's Disease Is a Multi-Organ Disorder: It May Already Be Preventable. J Alzheimers Dis 2023; 91:1277-1281. [PMID: 36617785 DOI: 10.3233/jad-221078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this commentary, we offer an overview of the several environmental and metabolic factors that have been identified as contributing to the development of Alzheimer's disease (AD). Many of these factors involve extracranial organ systems including immune system dysfunction accompanied by neuroinflammation (inflammaging), gastrointestinal dysbiosis, insulin resistance, and hepatic dysfunction. A variety of microbial factors including mouth flora, viruses, and fungi appear to play a significant role. There is a role for the colonic microbiome becoming dysbiotic and producing toxic metabolites. Declining hepatic function contributes diminished neuronal precursors and reduces toxin elimination. Environmental toxins especially metals play an important role in impairing the blood-brain barrier and acting synergistically with biotoxins and other toxic chemicals. Prevention and treatment of AD appears to require measuring several of these biomarkers and implementing corrective actions regarding such toxicants and correcting metabolic dysfunction at early or preclinical stages of this disorder.
Collapse
Affiliation(s)
- Arnold R Eiser
- Penn Center for Public Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Environmental Neuroscience Group, Center for Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamas Fulop
- Center on Aging, University of Sherbrooke, Quebec, Canada
| |
Collapse
|
23
|
Li J, Qi L, Chen Y, Lv H, Bi H. Bioinformatics analysis of the potential mechanisms of Alzheimer's disease induced by exposure to combined triazine herbicides. Ann Hum Biol 2023; 50:442-451. [PMID: 37819172 DOI: 10.1080/03014460.2023.2259242] [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: 03/07/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND The development of Alzheimer's disease (AD) is promoted by a combination of genetic and environmental factors. Notably, combined exposure to triazine herbicides atrazine (ATR), simazine (SIM), and propazine (PRO) may promote the development of AD, but the mechanism is unknown. AIM To study the molecular mechanism of AD induced by triazine herbicides. METHODS Differentially expressed genes (DEGs) of AD patients and controls were identified. The intersectional targets of ATR, SIM, and PRO for possible associations with AD were screened through network pharmacology and used for gene ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analysis. The binding potentials between the core targets and herbicides were validated by molecular docking and molecular dynamics. RESULTS A total of 1,062 DEGs were screened between the AD patients and controls, which identified 148 intersectional targets of herbicides causing AD that were screened by network pharmacology analysis. GO and KEGG enrichment analysis revealed that cell cycling and cellular senescence were important signalling pathways. Finally, the core targets EGFR, FN1, and TYMS were screened and validated by molecular docking and molecular dynamics. CONCLUSION Our results suggest that combined exposure to triazine herbicides might promote the development of AD, thereby providing new insights for the prevention of AD.
Collapse
Affiliation(s)
- Jianan Li
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ling Qi
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Yuxin Chen
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Haoming Lv
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Haoran Bi
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
24
|
Bellavite P. Neuroprotective Potentials of Flavonoids: Experimental Studies and Mechanisms of Action. Antioxidants (Basel) 2023; 12:antiox12020280. [PMID: 36829840 PMCID: PMC9951959 DOI: 10.3390/antiox12020280] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Neurological and neurodegenerative diseases, particularly those related to aging, are on the rise, but drug therapies are rarely curative. Functional disorders and the organic degeneration of nervous tissue often have complex causes, in which phenomena of oxidative stress, inflammation and cytotoxicity are intertwined. For these reasons, the search for natural substances that can slow down or counteract these pathologies has increased rapidly over the last two decades. In this paper, studies on the neuroprotective effects of flavonoids (especially the two most widely used, hesperidin and quercetin) on animal models of depression, neurotoxicity, Alzheimer's disease (AD) and Parkinson's disease are reviewed. The literature on these topics amounts to a few hundred publications on in vitro and in vivo models (notably in rodents) and provides us with a very detailed picture of the action mechanisms and targets of these substances. These include the decrease in enzymes that produce reactive oxygen and ferroptosis, the inhibition of mono-amine oxidases, the stimulation of the Nrf2/ARE system, the induction of brain-derived neurotrophic factor production and, in the case of AD, the prevention of amyloid-beta aggregation. The inhibition of neuroinflammatory processes has been documented as a decrease in cytokine formation (mainly TNF-alpha and IL-1beta) by microglia and astrocytes, by modulating a number of regulatory proteins such as Nf-kB and NLRP3/inflammasome. Although clinical trials on humans are still scarce, preclinical studies allow us to consider hesperidin, quercetin, and other flavonoids as very interesting and safe dietary molecules to be further investigated as complementary treatments in order to prevent neurodegenerative diseases or to moderate their deleterious effects.
Collapse
|
25
|
Rao RV, Subramaniam KG, Gregory J, Bredesen AL, Coward C, Okada S, Kelly L, Bredesen DE. Rationale for a Multi-Factorial Approach for the Reversal of Cognitive Decline in Alzheimer's Disease and MCI: A Review. Int J Mol Sci 2023; 24:ijms24021659. [PMID: 36675177 PMCID: PMC9865291 DOI: 10.3390/ijms24021659] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, progressive, neurodegenerative disease typically characterized by memory loss, personality changes, and a decline in overall cognitive function. Usually manifesting in individuals over the age of 60, this is the most prevalent type of dementia and remains the fifth leading cause of death among Americans aged 65 and older. While the development of effective treatment and prevention for AD is a major healthcare goal, unfortunately, therapeutic approaches to date have yet to find a treatment plan that produces long-term cognitive improvement. Drugs that may be able to slow down the progression rate of AD are being introduced to the market; however, there has been no previous solution for preventing or reversing the disease-associated cognitive decline. Recent studies have identified several factors that contribute to the progression and severity of the disease: diet, lifestyle, stress, sleep, nutrient deficiencies, mental health, socialization, and toxins. Thus, increasing evidence supports dietary and other lifestyle changes as potentially effective ways to prevent, slow, or reverse AD progression. Studies also have demonstrated that a personalized, multi-therapeutic approach is needed to improve metabolic abnormalities and AD-associated cognitive decline. These studies suggest the effects of abnormalities, such as insulin resistance, chronic inflammation, hypovitaminosis D, hormonal deficiencies, and hyperhomocysteinemia, in the AD process. Therefore a personalized, multi-therapeutic program based on an individual's genetics and biochemistry may be preferable over a single-drug/mono-therapeutic approach. This article reviews these multi-therapeutic strategies that identify and attenuate all the risk factors specific to each affected individual. This article systematically reviews studies that have incorporated multiple strategies that target numerous factors simultaneously to reverse or treat cognitive decline. We included high-quality clinical trials and observational studies that focused on the cognitive effects of programs comprising lifestyle, physical, and mental activity, as well as nutritional aspects. Articles from PubMed Central, Scopus, and Google Scholar databases were collected, and abstracts were reviewed for relevance to the subject matter. Epidemiological, pathological, toxicological, genetic, and biochemical studies have all concluded that AD represents a complex network insufficiency. The research studies explored in this manuscript confirm the need for a multifactorial approach to target the various risk factors of AD. A single-drug approach may delay the progression of memory loss but, to date, has not prevented or reversed it. Diet, physical activity, sleep, stress, and environment all contribute to the progression of the disease, and, therefore, a multi-factorial optimization of network support and function offers a rational therapeutic strategy. Thus, a multi-therapeutic program that simultaneously targets multiple factors underlying the AD network may be more effective than a mono-therapeutic approach.
Collapse
Affiliation(s)
- Rammohan V. Rao
- Apollo Health, Burlingame, CA 94011, USA
- Correspondence: (R.V.R.); (D.E.B.)
| | | | | | | | | | - Sho Okada
- Apollo Health, Burlingame, CA 94011, USA
| | | | - Dale E. Bredesen
- Apollo Health, Burlingame, CA 94011, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90024, USA
- Correspondence: (R.V.R.); (D.E.B.)
| |
Collapse
|
26
|
Hossain MM, Belkadi A, Zhou X, DiCicco-Bloom E. Exposure to deltamethrin at the NOAEL causes ER stress and disruption of hippocampal neurogenesis in adult mice. Neurotoxicology 2022; 93:233-243. [DOI: 10.1016/j.neuro.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
|
27
|
Abdel Ghfar SS, Ali ME, Momenah MA, Al-Saeed FA, Al-Doaiss AA, Mostafa YS, Ahmed AE, Abdelrahman M. Effect of Allium sativum and Nigella sativa on alleviating aluminum toxicity state in the albino rats. Front Vet Sci 2022; 9:1042640. [PMID: 36524230 PMCID: PMC9745150 DOI: 10.3389/fvets.2022.1042640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/25/2022] [Indexed: 12/02/2022] Open
Abstract
The study objective was to evaluate Allium sativum's potential and Nigella Sativa's combination's potential to reduce aluminum toxicity and return to the normal state. In the present study, a hundred albino rats were randomly divided into five equal groups. The first group was used as a control group; the other four groups were exposed to aluminum 1,600 ppm. The second exposed to aluminum only; the third and fourth groups were treated with Allium sativum 5% and Nigella sativa 5%, respectively, while the fifth group was treated with a mix of Allium sativum 2.5% and Nigella sativa 2.5% for 8th weeks. After 8 weeks, the aluminum administration was stopped, and the second group was divided into three groups. The groups were treated with Allium sativum 5% and Nigella sativa 5%, and a mix of Allium sativum 2.5% and Nigella sativa 2.5%, respectively. The first group was the control group (continued from the first experiment). Garlic and Nigella sativa were crushed and added to feed while receiving aluminum chloride daily at a dose of 1.6 ml/l was added to the drinking water. Histopathological changes in the liver, kidney, and testes were investigated after 8 and 16 weeks, and blood samples were collected after 4, 8, and 16 weeks for biochemical blood parameters. The results showed that the histopathological examination of the liver, kidney, and testes showed signs of congestion in blood vessels after aluminum exposure. Meanwhile, the treatment with Allium sativum or Nigella sativum or the mixture between them had positive effects on evading the harmful effects of aluminum in the liver, Kidney, and testes tissues. In addition, there were protective effects for Allium sativum and Nigella sativa against aluminum on serum creatinine, urea, ALT, and AST concentrations. The present study concluded that supplementation with Allium sativum or Nigella sativa or their combination could reduce aluminum toxicity and return the liver, kidney, and testes to normal.
Collapse
Affiliation(s)
| | - Montaser Elsayed Ali
- Department of Animal Productions, Faculty of Agriculture, Al-Azhar University, Assiut, Egypt
| | - Maha Abdullah Momenah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fatimah A. Al-Saeed
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Amin A. Al-Doaiss
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Yasser Sabry Mostafa
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Theriogenology, Obstetrics, and Artificial Insemination, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Mohamed Abdelrahman
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Animal Production Department, Faculty of Agriculture, Assiut University, Asyut, Egypt
| |
Collapse
|
28
|
Chang YC, Chen WT, Su SH, Jung CR, Hwang BF. PM 2.5 exposure and incident attention-deficit/hyperactivity disorder during the prenatal and postnatal periods: A birth cohort study. ENVIRONMENTAL RESEARCH 2022; 214:113769. [PMID: 35777438 DOI: 10.1016/j.envres.2022.113769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 05/14/2023]
Abstract
Only a few studies have assessed the effects of fine particulate matter (PM2.5) exposure during the prenatal and postnatal periods on the development of attention-deficit/hyperactivity disorder (ADHD). We investigated the association of exposure to PM2.5 during pregnancy and early life with ADHD. This birth cohort consisted of 425,736 singleton live-term births between 2004 and 2015 in Taiwan. Daily PM2.5 concentrations were derived from a 1-km satellite-based estimation model. A time-dependent Cox model was used to assess the effects of PM2.5 on ADHD during the first, second, and third trimesters and from age 1-5 years after birth. The distributed lag nonlinear model was utilized to explore the dose-response relationship. Total 9,294 children were diagnosed with ADHD during the study period. The hazard ratio (HR) of ADHD was significantly associated with a 10 μg/m3 increase in PM2.5 during the first trimester (HR = 1.26; 95% confidence interval [CI]: 1.13-1.40) and increased at PM2.5 over 16 μg/m3. For postnatal periods, the HR of ADHD was significantly associated with increased PM2.5 at the first to third year of life (HR ranged between 1.40 and 1.87). According to the dose-response relationship of exposure to PM2.5 at the third year of life, the HR of ADHD was significantly associated with PM2.5 above 16 μg/m3 and sharply increased as PM2.5 >50 μg/m3. We did not observe a significant modification of sex on the relation between PM2.5 and ADHD. Exposure of pregnant women to PM2.5 above 16 μg/m3 from conception to the early life of their children may increase the risk of ADHD. The government should improve the criteria for air quality control and meet the WHO air quality guidelines to protect pregnant women and children from developing ADHD in the future.
Collapse
Affiliation(s)
- Ya-Chu Chang
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Wei-Ting Chen
- Department of Atmospheric Sciences, National Taiwan University, Taipei, Taiwan
| | - Shih-Hao Su
- Department of Atmospheric Sciences, Chinese Culture University, Taipei, Taiwan
| | - Chau-Ren Jung
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan; Japan Environment and Children's Study Programme Office, Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, Japan.
| | - Bing-Fang Hwang
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan; Department of Occupational Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
29
|
Sehar U, Rawat P, Reddy AP, Kopel J, Reddy PH. Amyloid Beta in Aging and Alzheimer's Disease. Int J Mol Sci 2022; 23:12924. [PMID: 36361714 PMCID: PMC9655207 DOI: 10.3390/ijms232112924] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 12/06/2022] Open
Abstract
Alzheimer's disease (AD), is a progressive neurodegenerative disease that affects behavior, thinking, learning, and memory in elderly individuals. AD occurs in two forms, early onset familial and late-onset sporadic; genetic mutations in PS1, PS2, and APP genes cause early onset familial AD, and a combination of lifestyle, environment and genetic factors causes the late-onset sporadic form of the disease. However, accelerated disease progression is noticed in patients with familial AD. Disease-causing pathological changes are synaptic damage, and mitochondrial structural and functional changes, in addition to increased production and accumulation of phosphorylated tau (p-tau), and amyloid beta (Aβ) in the affected brain regions in AD patients. Aβ is a peptide derived from amyloid precursor protein (APP) by proteolytic cleavage of beta and gamma secretases. APP is a glycoprotein that plays a significant role in maintaining neuronal homeostasis like signaling, neuronal development, and intracellular transport. Aβ is reported to have both protective and toxic effects in neurons. The purpose of our article is to summarize recent developments of Aβ and its association with synapses, mitochondria, microglia, astrocytes, and its interaction with p-tau. Our article also covers the therapeutic strategies that reduce Aβ toxicities in disease progression and discusses the reasons for the failures of Aβ therapeutics.
Collapse
Affiliation(s)
- Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Priyanka Rawat
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P. Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Jonathan Kopel
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
30
|
Liu S, Wang Z, Xiang Q, Wu B, Lv W, Xu S. A comparative study in healthy and diabetic mice followed the exposure of polystyrene microplastics: Differential lipid metabolism and inflammation reaction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114031. [PMID: 36087466 DOI: 10.1016/j.ecoenv.2022.114031] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/08/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
Human exposure to microplastics (MPs) continues to occur due to ingestion of contaminated food, water and air. Intake of MPs can pose potential health risks by interfering with the production and circulation of nutrients, leading to physiological stress (such as immune responses and metabolic abnormalities). Toxicity data of MPs based on healthy individuals may not be applicable to large populations of patients with chronic diseases represented by diabetes. Therefore, in this study, the response of diabetic mice was compared with that of healthy mice after exposure to polystyrene microplastics (PS-MPs), and interesting differences were observed. PS-MPs exposure significantly increased liver tissue damage, abnormal lipid metabolism, inflammatory effect, liver metabolic disorder and changes of intestinal microbial composition in diabetic mice. Moreover, PS-MPs overstated abnormal lipid metabolism in diabetic mice. The difference between the increased inflammation after exposure to PS-MPs in healthy and diabetic mice involves that the former is mainly modulated by gut microbes, while diabetic mice seem to be more susceptible to lipid metabolism disturbances. In addition, the size effect of MPs was also observed in diabetic mice. These results suggested that individuals with chronic diseases may be more sensitive to pollution due to altered homeostasis, and therefore disease status should be fully considered when assessing the health risk of pollutants.
Collapse
Affiliation(s)
- Su Liu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China; School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhizhi Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Qi Xiang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Wang Lv
- School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Shimin Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
31
|
Song X, Ding Q, Wei W, Pu Y. LP-05 A Novel Targeted Prussian Blue-Based Nanomaterial for Inhibiting Oxidative Stress and Aβ Aggregation of Alzheimer’s Disease. Toxicol Lett 2022. [DOI: 10.1016/j.toxlet.2022.07.749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Şanlı BA, Whittaker KJ, Motsi GK, Shen E, Julian TH, Cooper-Knock J. Unbiased metabolome screen links serum urate to risk of Alzheimer's disease. Neurobiol Aging 2022; 120:167-176. [DOI: 10.1016/j.neurobiolaging.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
|
33
|
James LM, Georgopoulos AP. High Correlations Among Worldwide Prevalences of Dementias, Parkinson's Disease, Multiple Sclerosis, and Motor Neuron Diseases Indicate Common Causative Factors. Neurosci Insights 2022; 17:26331055221117598. [PMID: 35965966 PMCID: PMC9364200 DOI: 10.1177/26331055221117598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Dementia, Parkinson's disease, multiple sclerosis, and motor neuron diseases cause significant disability and mortality worldwide. Although the etiology of these diseases is unknown, highly correlated disease prevalences would indicate the involvement of common etiologic factors. Here we used published epidemiological data in 195 countries worldwide to investigate the possible intercorrelations among the prevalences of these diseases. All analyses were carried out using nonparametric statistics on rank-transformed data to assure the robustness of the results. We found that all 6 pairwise correlations among the prevalences of the 4 diseases were very high (>.9, P < .001). A factor analysis (FA) yielded only a single component which comprised all 4 disease prevalences and explained 96.3% of the variance. These findings indicate common etiologic factor(s). Next, we quantified the contribution of 3 country-specific factors (population size, life expectancy, latitude) to the common grouping of prevalences by estimating the reduction in total FA variance explained when the effect of these factors was eliminated by using the prevalence residuals from a linear regression where theses factor were covariates. FA of these residuals yielded again only a single component comprising all 4 diseases which explained 71.5% of the variance, indicating that the combined contribution of population size, life expectancy and latitude accounted for 96.3% - 71.5% = 24.8% of the FA variance explained. The fact that the 3 country-specific factors above accounted for only 24.8% of the FA variance explained by the original (ranked) disease prevalences, in the presence still of a single grouping factor, strongly indicates the operation of other unknown factors jointly contributing to the pathogenesis of the 4 diseases. We discuss various possible factors involved, with an emphasis on biologic pathogens (viruses, bacteria) which have been implicated in the pathogenesis of these diseases in previous studies.
Collapse
Affiliation(s)
- Lisa M James
- Brain Sciences Center, Department of
Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University
of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of
Minnesota Medical School, Minneapolis, MN, USA
- Center for Cognitive Sciences,
University of Minnesota, Minneapolis, MN, USA
| | - Apostolos P Georgopoulos
- Brain Sciences Center, Department of
Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University
of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of
Minnesota Medical School, Minneapolis, MN, USA
- Center for Cognitive Sciences,
University of Minnesota, Minneapolis, MN, USA
- Department of Neurology, University of
Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
34
|
Costantino AR, Charbe N, Duarte Y, Gutiérrez M, Giordano A, Prasher P, Dua K, Mandolesi S, Zacconi FC. Toward the cholinesterase inhibition potential of TADDOL derivatives: Seminal biological and computational studies. Arch Pharm (Weinheim) 2022; 355:e2200142. [PMID: 35892245 DOI: 10.1002/ardp.202200142] [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: 03/18/2022] [Revised: 06/03/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease (AD) is a degenerative neurological disease characterized by gradual loss of cognitive skills and memory. The exact pathogenesis involved still remains unrevealed, but several studies indicate the involvement of an array of different enzymes, underlining the multifactorial character of the disease. Inhibition of these enzymes is therefore a powerful approach in the development of AD treatments, with promising candidates, including acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), and monoamine oxidase. Interestingly, AChE is the target of a major pesticide family (organophosphates), with several reports indicating an intersection between the pesticide's activity and AD. In this study, various TADDOL derivatives were synthesized and their in vitro activities as AChE/BuChE inhibitors as well as their antioxidant activities were studied. Molecular modeling studies revealed the capability of TADDOL derivatives to bind to AChE and induce inhibition, especially compounds 2b and 3c furnishing IC50 values of 36.78 ± 8.97 and 59.23 ± 5.31 µM, respectively. Experimental biological activities and molecular modeling studies clearly demonstrate that TADDOL derivatives with specific stereochemistry have an interesting potential for the design of potent AChE inhibitors. The encouraging results for compounds 2b and 3c indicate them as promising scaffolds for selective and potent AChE inhibitors.
Collapse
Affiliation(s)
- Andrea R Costantino
- INQUISUR, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Nitin Charbe
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, Texas, USA
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Margarita Gutiérrez
- Organic Synthesis Laboratory and Biological Activity (LSO-Act-Bio), Institute of Chemistry and Natural Resources, Universidad de Talca, Talca, Chile
| | - Ady Giordano
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, Uttarakhand, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, New South Wales, Australia.,Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, New South Wales, Australia.,Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sandra Mandolesi
- INQUISUR, Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Flavia C Zacconi
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,The Research Center for Nanotechnology and Advanced Materials, CIEN-UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
35
|
Ribeiro LW, Pietri M, Ardila-Osorio H, Baudry A, Boudet-Devaud F, Bizingre C, Arellano-Anaya ZE, Haeberlé AM, Gadot N, Boland S, Devineau S, Bailly Y, Kellermann O, Bencsik A, Schneider B. Titanium dioxide and carbon black nanoparticles disrupt neuronal homeostasis via excessive activation of cellular prion protein signaling. Part Fibre Toxicol 2022; 19:48. [PMID: 35840975 PMCID: PMC9284759 DOI: 10.1186/s12989-022-00490-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background Epidemiological emerging evidence shows that human exposure to some nanosized materials present in the environment would contribute to the onset and/or progression of Alzheimer’s disease (AD). The cellular and molecular mechanisms whereby nanoparticles would exert some adverse effects towards neurons and take part in AD pathology are nevertheless unknown. Results Here, we provide the prime evidence that titanium dioxide (TiO2) and carbon black (CB) nanoparticles (NPs) bind the cellular form of the prion protein (PrPC), a plasma membrane protein well known for its implication in prion diseases and prion-like diseases, such as AD. The interaction between TiO2- or CB-NPs and PrPC at the surface of neuronal cells grown in culture corrupts PrPC signaling function. This triggers PrPC-dependent activation of NADPH oxidase and subsequent production of reactive oxygen species (ROS) that alters redox equilibrium. Through PrPC interaction, NPs also promote the activation of 3-phosphoinositide-dependent kinase 1 (PDK1), which in turn provokes the internalization of the neuroprotective TACE α-secretase. This diverts TACE cleavage activity away from (i) TNFα receptors (TNFR), whose accumulation at the plasma membrane augments the vulnerability of NP-exposed neuronal cells to TNFα -associated inflammation, and (ii) the amyloid precursor protein APP, leading to overproduction of neurotoxic amyloid Aβ40/42 peptides. The silencing of PrPC or the pharmacological inhibition of PDK1 protects neuronal cells from TiO2- and CB-NPs effects regarding ROS production, TNFα hypersensitivity, and Aβ rise. Finally, we show that dysregulation of the PrPC-PDK1-TACE pathway likely occurs in the brain of mice injected with TiO2-NPs by the intra-cerebro-ventricular route as we monitor a rise of TNFR at the cell surface of several groups of neurons located in distinct brain areas. Conclusion Our in vitro and in vivo study thus posits for the first time normal cellular prion protein PrPC as being a neuronal receptor of TiO2- and CB-NPs and identifies PrPC-coupled signaling pathways by which those nanoparticles alter redox equilibrium, augment the intrinsic sensitivity of neurons to neuroinflammation, and provoke a rise of Aβ peptides. By identifying signaling cascades dysregulated by TiO2- and CB-NPs in neurons, our data shed light on how human exposure to some NPs might be related to AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00490-x.
Collapse
Affiliation(s)
- Luiz W Ribeiro
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Mathéa Pietri
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Hector Ardila-Osorio
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Anne Baudry
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - François Boudet-Devaud
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Chloé Bizingre
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Zaira E Arellano-Anaya
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Anne-Marie Haeberlé
- Institut Des Neurosciences Cellulaires Et Intégratives, CNRS UPR 3212, Université de Strasbourg, 67084, Strasbourg, France
| | - Nicolas Gadot
- Plateforme Anatomopathologie Recherche, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, Université de Lyon, 69373, Lyon, France
| | - Sonja Boland
- CNRS UMR 8251, Unité de Biologie Fonctionnelle Et Adaptative, Université Paris Cité, 75013, Paris, France
| | - Stéphanie Devineau
- CNRS UMR 8251, Unité de Biologie Fonctionnelle Et Adaptative, Université Paris Cité, 75013, Paris, France
| | - Yannick Bailly
- Institut Des Neurosciences Cellulaires Et Intégratives, CNRS UPR 3212, Université de Strasbourg, 67084, Strasbourg, France
| | - Odile Kellermann
- INSERM, UMR-S 1124, 75006, Paris, France.,UMR-S 1124, Université Paris Cité, 75006, Paris, France
| | - Anna Bencsik
- ANSES Laboratoire de Lyon, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université Claude Bernard Lyon 1, 69364, Lyon, France
| | - Benoit Schneider
- INSERM, UMR-S 1124, 75006, Paris, France. .,UMR-S 1124, Université Paris Cité, 75006, Paris, France.
| |
Collapse
|
36
|
Luo JJ, Wallace W, Kusiak JW. A tough trek in the development of an anti-amyloid therapy for Alzheimer's disease: Do we see hope in the distance? J Neurol Sci 2022; 438:120294. [DOI: 10.1016/j.jns.2022.120294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/27/2022] [Accepted: 05/18/2022] [Indexed: 12/17/2022]
|
37
|
Recent trends of natural based therapeutics for mitochondria targeting in Alzheimer’s disease. Mitochondrion 2022; 64:112-124. [DOI: 10.1016/j.mito.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
|
38
|
Soares MV, Mesadri J, Gonçalves DF, Cordeiro LM, Franzen da Silva A, Obetine Baptista FB, Wagner R, Dalla Corte CL, Soares FAA, Ávila DS. Neurotoxicity induced by toluene: In silico and in vivo evidences of mitochondrial dysfunction and dopaminergic neurodegeneration. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 298:118856. [PMID: 35033616 DOI: 10.1016/j.envpol.2022.118856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/21/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
Toluene is an air pollutant widely used as an organic solvent in industrial production and emitted by fossil fuel combustion, in addition to being used as a drug of abuse. Its toxic effects in the central nervous system have not been well established, and how and which neurons are affected remains unknown. Hence, this study aimed to fill this gap by investigating three central questions: 1) How does toluene induce neurotoxicity? 2) Which neurons are affected? And 3) What are the long-term effects induced by airborne exposure to toluene? To this end, a Caenorhabditis elegans model was employed, in which worms at the fourth larval stage were exposed to toluene in the air for 24 h in a vapor chamber to simulate four exposure scenarios. After the concentration-response curve analysis, we chose scenarios 3 (E3: 792 ppm) and 4 (E4: 1094 ppm) for the following experiments. The assays were performed 1, 48, or 96 h after removal from the exposure environments, and an irreversible reduction in neuron fluorescence and morphologic alterations were observed in different neurons of exposed worms, particularly in the dopaminergic neurons. Moreover, a significant impairment in a dopaminergic-dependent behavior was also associated with negative effects in healthspan endpoints, and we also noted that mitochondria may be involved in toluene-induced neurotoxicity since lower adenosine 5'-triphosphate (ATP) levels and mitochondrial viability were observed. In addition, a reduction of electron transport chain activity was evidenced using ex vivo protocols, which were reinforced by in silico and in vitro analysis, demonstrating toluene action in the mitochondrial complexes. Based on these findings model, it is plausible that toluene neurotoxicity can be initiated by complex I inhibition, triggering a mitochondrial dysfunction that may lead to irreversible dopaminergic neuronal death, thus impairing neurobehavioral signaling.
Collapse
Affiliation(s)
- Marcell Valandro Soares
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil; Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Juliana Mesadri
- Departamento: Tecnologia e Ciência dos Alimentos, Centro de Ciência Rurais, Programa de Pós-graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria, RS, Brazil
| | - Débora Farina Gonçalves
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Larissa Marafiga Cordeiro
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Aline Franzen da Silva
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Fabiane Bicca Obetine Baptista
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Roger Wagner
- Departamento: Tecnologia e Ciência dos Alimentos, Centro de Ciência Rurais, Programa de Pós-graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria, RS, Brazil
| | - Cristiane Lenz Dalla Corte
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Daiana Silva Ávila
- Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil.
| |
Collapse
|
39
|
Chen D, Li J, Zhao Y, Wu Y. Human Exposure of Fipronil Insecticide and the Associated Health Risk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:63-71. [PMID: 34971309 DOI: 10.1021/acs.jafc.1c05694] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fipronil, as an emerging phenylpyrazole insecticide, is ubiquitous in the environment and food due to its broad spectrum and persistent characteristics, but the research on pathways of human exposure to fipronil and the associated health risk is relatively unclear. In this regard, we summarize potential human exposures to fipronil through ingestion and inhalation, as well as results of human biomonitoring studies. This scientific information will contribute to future assessment of fipronil exposure and subsequent characterization of human health risks. Additionally, this Perspective highlights the lack of epidemiological studies and total diet studies for the general population on fipronil.
Collapse
Affiliation(s)
- Dawei Chen
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Jingguang Li
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Yunfeng Zhao
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| |
Collapse
|
40
|
Folarin OR, Olopade FE, Olopade JO. Essential Metals in the Brain and the Application of Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry for their Detection. Niger J Physiol Sci 2021; 36:123-147. [PMID: 35947740 DOI: 10.54548/njps.v36i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 06/15/2023]
Abstract
Metals are natural component of the ecosystem present throughout the layers of atmosphere; their abundant expression in the brain indicates their importance in the central nervous system (CNS). Within the brain tissue, their distribution is highly compartmentalized, the pattern of which is determined by their primary roles. Bio-imaging of the brain to reveal spatial distribution of metals within specific regions has provided a unique understanding of brain biochemistry and architecture, linking both the structures and the functions through several metal mediated activities. Bioavailability of essential trace metal is needed for normal brain function. However, disrupted metal homeostasis can influence several biochemical pathways in different fields of metabolism and cause characteristic neurological disorders with a typical disease process usually linked with aberrant metal accumulations. In this review we give a brief overview of roles of key essential metals (Iron, Copper and Zinc) including their molecular mechanisms and bio-distribution in the brain as well as their possible involvement in the pathogenesis of related neurodegenerative diseases. In addition, we also reviewed recent applications of Laser Ablation Inductively Couple Plasma Mass Spectrophotometry (LA-ICP-MS) in the detection of both toxic and essential metal dyshomeostasis in neuroscience research and other related brain diseases.
Collapse
|
41
|
Lutz A, Chételat G, Collette F, Klimecki OM, Marchant NL, Gonneaud J. The protective effect of mindfulness and compassion meditation practices on ageing: Hypotheses, models and experimental implementation. Ageing Res Rev 2021; 72:101495. [PMID: 34718153 DOI: 10.1016/j.arr.2021.101495] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/09/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Alzheimer's disease (AD) represents a major health and societal issue; there is no treatment to date and the pathophysiological mechanisms underlying this disease are not well understood. Yet, there is hope that AD risk factors and thus the number of AD cases can be significantly reduced by prevention measures based on lifestyle modifications as targeted by non-pharmacological preventive interventions. So far, these interventions have rarely targeted the psycho-affective risk factors related to depression, stress, anxiety, and feeling of loneliness, which are all prevalent in ageing. This paper presents the hypothesis that the regular practice of mindfulness meditation (MM) and loving-kindness and compassion meditation (LKCM) in the ageing population constitutes a lifestyle that is protective against AD. In this model, these practices can promote cognition, mental health, and well-being by strengthening attention control, metacognitive monitoring, emotion regulation and pro-social capacities. Training these capacities could reduce the risk of AD by upregulating beneficial age-related factors such as cognitive reserve, and down-regulating detrimental age-related factors, such as stress, or depression. As an illustration, we present the Medit-Ageing study (public name Silver Santé Study), an on-going European project that assesses the impact and mechanisms of non-pharmacological interventions including meditation, in the ageing population.
Collapse
|
42
|
Hussien HM, Ghareeb DA, Ahmed HEA, Hafez HS, Saleh SR. Pharmacological implications of ipriflavone against environmental metal-induced neurodegeneration and dementia in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:65349-65362. [PMID: 34235690 DOI: 10.1007/s11356-021-15193-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
Long-term exposure to environmental neurotoxic metals is implicated in the induction of dementia and cognitive decline. The present study aims to illustrate the therapeutic role of ipriflavone as a synthetic isoflavone against environmental metal-induced cognitive impairment in rats. Dementia was induced by a mixture of aluminum, cadmium, and fluoride for 90 days followed by ipriflavone for a further 30 days. Metal-treated animals exhibited abnormal behaviors in the Morris water maze task. Neuropathological biomarkers including oxidative stress (TBARS, NO, SOD, GPX, GST, and GSH), inflammation (TNF- α, IL-6, and IL-1β), neurotransmission (AChE and MAO), and insulin resistance (insulin, insulin receptor, and insulin-degrading enzyme) were altered, which consequently elevated the level of amyloid-β42 and tau protein in the hippocampus tissues inducing neuronal injury. Ipriflavone significantly (P < 0.05) ameliorated the neurobehavioral abnormalities and the cognitive dysfunction biomarkers via antioxidant/anti-inflammatory mechanism. Moreover, ipriflavone downregulated the mRNA expression level of amyloid precursor protein and tau protein, preventing amyloid plaques and neurofibrillary tangle aggregation at P < 0.05. A molecular docking study revealed that ipriflavone has a potent binding affinity towards AChE more than donepezil and acts as a strong AChE inhibitor. Our data concluded that the therapeutic potential of ipriflavone against dementia could provide a new strategy in AD treatment.
Collapse
Affiliation(s)
- Hend M Hussien
- Department of Pharmacology and Therapeutics Department, Faculty of Pharmacy, Pharos University, Canal El Mahmoudia Street, Smouha, Sidi Gaber, P.O. Box 37, Alexandria, Egypt.
| | - Doaa A Ghareeb
- Biological Screening and Preclinical Trial Laboratory, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center of Excellency for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industries Development Centre (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, Egypt
| | - Hany E A Ahmed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Hani S Hafez
- Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Samar R Saleh
- Biological Screening and Preclinical Trial Laboratory, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center of Excellency for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industries Development Centre (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, Egypt
| |
Collapse
|
43
|
Nisa FY, Rahman MA, Hossen MA, Khan MF, Khan MAN, Majid M, Sultana F, Haque MA. Role of neurotoxicants in the pathogenesis of Alzheimer's disease: a mechanistic insight. Ann Med 2021; 53:1476-1501. [PMID: 34433343 PMCID: PMC8405119 DOI: 10.1080/07853890.2021.1966088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most conspicuous chronic neurodegenerative syndrome, which has become a significant challenge for the global healthcare system. Multiple studies have corroborated a clear association of neurotoxicants with AD pathogenicity, such as Amyloid beta (Aβ) proteins and neurofibrillary tangles (NFTs), signalling pathway modifications, cellular stress, cognitive dysfunctions, neuronal apoptosis, neuroinflammation, epigenetic modification, and so on. This review, therefore, aimed to address several essential mechanisms and signalling cascades, including Wnt (wingless and int.) signalling pathway, autophagy, mammalian target of rapamycin (mTOR), protein kinase C (PKC) signalling cascades, cellular redox status, energy metabolism, glutamatergic neurotransmissions, immune cell stimulations (e.g. microglia, astrocytes) as well as an amyloid precursor protein (APP), presenilin-1 (PSEN1), presenilin-2 (PSEN2) and other AD-related gene expressions that have been pretentious and modulated by the various neurotoxicants. This review concluded that neurotoxicants play a momentous role in developing AD through modulating various signalling cascades. Nevertheless, comprehension of this risk agent-induced neurotoxicity is far too little. More in-depth epidemiological and systematic investigations are needed to understand the potential mechanisms better to address these neurotoxicants and improve approaches to their risk exposure that aid in AD pathogenesis.Key messagesInevitable cascade mechanisms of how Alzheimer's Disease-related (AD-related) gene expressions are modulated by neurotoxicants have been discussed.Involvement of the neurotoxicants-induced pathways caused an extended risk of AD is explicited.Integration of cell culture, animals and population-based analysis on the clinical severity of AD is addressed.
Collapse
Affiliation(s)
- Fatema Yasmin Nisa
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Atiar Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Amjad Hossen
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad Forhad Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Md. Asif Nadim Khan
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Liu C, Li Y, Luo Y, Zhang Y, Zhou T, Deng J. Lab-on-a-ZnO-Submicron-Particle Sensor Array for Monitoring AD upon Cd 2+ Exposure with CSF Tau441% as an Effective Hallmark. Anal Chem 2021; 93:15005-15014. [PMID: 34738809 DOI: 10.1021/acs.analchem.1c02570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this study, based on the posttreatment strategy, blue-color-emissive ZnO submicron particles (B-ZnO SMPs) and red-color-emissive ZnO submicron particles (R-ZnO SMPs) were obtained from rationally designed Zn-infinite coordination polymer (ICP) precursors. After modification of thiol-containing aptamers, diverse spectral changes in the ultraviolet and visible regions of B- and R-ZnO SMPs toward different tau species were explored to construct a lab-on-a-ZnO-submicron-particle sensor array. Assisted by principal component analysis (PCA), the unique fingerprints of the sensor array enabled the simultaneous differentiation and quantitative detection of different tau species (tau381, tau410, and tau441) for the first time. Furthermore, the dynamic changes of tau441% (the ratio of the two most reported representative 4R isoform (full-length tau441) and 3R isoform (tau381)) in cerebrospinal fluid (CSF) during the Alzheimer's disease (AD) onset of Cd2+-exposed rats could also be monitored by the lab-on-a-ZnO-submicron-particle sensor array, which was supposed to be an effective hallmark and highly correlated with the formation of neurofibrillary tangles (NFTs). This study not only provides a further insight into the involvement of subchronic Cd2+ exposure in the tau etiology of AD but also offers more comprehensive and effective information about the asymptomatic stage of AD upon environmental risk, which has potential applications in the early diagnosis and therapy.
Collapse
Affiliation(s)
- Chang Liu
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| | - Yuanting Li
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| | - Yuxin Luo
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| | - Ying Zhang
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| | - Tianshu Zhou
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| | - Jingjing Deng
- School of Ecological and Environmental Sciences, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Shanghai Key Lab for Urban Ecological Process and Eco-Restoration, East China Normal University, Shanghai 200241, China.,Institute of Eco-Chongming, 3663 Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
45
|
Samieri C, Yassine HN, Melo van Lent D, Lefèvre-Arbogast S, van de Rest O, Bowman GL, Scarmeas N. Personalized nutrition for dementia prevention. Alzheimers Dement 2021; 18:1424-1437. [PMID: 34757699 DOI: 10.1002/alz.12486] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
The role of nutrition has been investigated for decades under the assumption of one-size-fits-all. Yet there is heterogeneity in metabolic and neurobiological responses to diet. Thus a more personalized approach may better fit biological reality and have increased efficacy to prevent dementia. Personalized nutrition builds on the food exposome, defined as the history of diet-related exposures over the lifetime, and on its interactions with the genome and other biological characteristics (eg, metabolism, the microbiome) to shape health. We review current advances of personalized nutrition in dementia research. We discuss key questions, success milestones, and future roadmap from observational epidemiology to clinical studies through basic science. A personalized nutrition approach based on the best prescription for the most appropriate target population in the most relevant time-window has the potential to strengthen dementia-prevention efforts.
Collapse
Affiliation(s)
- Cécilia Samieri
- Univ. Bordeaux, ISPED, Inserm, Bordeaux Population Health Research Center, Bordeaux, France
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine USC, Los Angeles, California, USA.,Department of Neurology, Keck School of Medicine USC, Los Angeles, California, USA
| | - Debora Melo van Lent
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Ondine van de Rest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Gene L Bowman
- Department of Neurology and Layton Aging and Alzheimer's Disease Center, Oregon Health and Science University, Portland, Oregon, USA.,Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
46
|
Shea TB. Improvement of cognitive performance by a nutraceutical formulation: Underlying mechanisms revealed by laboratory studies. Free Radic Biol Med 2021; 174:281-304. [PMID: 34352370 DOI: 10.1016/j.freeradbiomed.2021.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/28/2022]
Abstract
Cognitive decline, decrease in neuronal function and neuronal loss that accompany normal aging and dementia are the result of multiple mechanisms, many of which involve oxidative stress. Herein, we review these various mechanisms and identify pharmacological and non-pharmacological approaches, including modification of diet, that may reduce the risk and progression of cognitive decline. The optimal degree of neuronal protection is derived by combinations of, rather than individual, compounds. Compounds that provide antioxidant protection are particularly effective at delaying or improving cognitive performance in the early stages of Mild Cognitive Impairment and Alzheimer's disease. Laboratory studies confirm alleviation of oxidative damage in brain tissue. Lifestyle modifications show a degree of efficacy and may augment pharmacological approaches. Unfortunately, oxidative damage and resultant accumulation of biomarkers of neuronal damage can precede cognitive decline by years to decades. This underscores the importance of optimization of dietary enrichment, antioxidant supplementation and other lifestyle modifications during aging even for individuals who are cognitively intact.
Collapse
Affiliation(s)
- Thomas B Shea
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA.
| |
Collapse
|
47
|
Sarrouilhe D, Defamie N, Mesnil M. Is the Exposome Involved in Brain Disorders through the Serotoninergic System? Biomedicines 2021; 9:1351. [PMID: 34680468 PMCID: PMC8533279 DOI: 10.3390/biomedicines9101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 11/24/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a biogenic monoamine acting as a neurotransmitter in the central nervous system (CNS), local mediator in the gut, and vasoactive agent in the blood. It has been linked to a variety of CNS functions and is implicated in many CNS and psychiatric disorders. The high comorbidity between some neuropathies can be partially understood by the fact that these diseases share a common etiology involving the serotoninergic system. In addition to its well-known functions, serotonin has been shown to be a mitogenic factor for a wide range of normal and tumor cells, including glioma cells, in vitro. The developing CNS of fetus and newborn is particularly susceptible to the deleterious effects of neurotoxic substances in our environment, and perinatal exposure could result in the later development of diseases, a hypothesis known as the developmental origin of health and disease. Some of these substances affect the serotoninergic system and could therefore be the source of a silent pandemic of neurodevelopmental toxicity. This review presents the available data that are contributing to the appreciation of the effects of the exposome on the serotoninergic system and their potential link with brain pathologies (neurodevelopmental, neurodegenerative, neurobehavioral disorders, and glioblastoma).
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculté de Médecine et Pharmacie, 6 Rue de la Milétrie, Bât D1, TSA 51115, CEDEX 09, 86073 Poitiers, France
| | - Norah Defamie
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| | - Marc Mesnil
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| |
Collapse
|
48
|
Dragić M, Mitrović N, Adžić M, Nedeljković N, Grković I. Microglial- and Astrocyte-Specific Expression of Purinergic Signaling Components and Inflammatory Mediators in the Rat Hippocampus During Trimethyltin-Induced Neurodegeneration. ASN Neuro 2021; 13:17590914211044882. [PMID: 34569324 PMCID: PMC8495514 DOI: 10.1177/17590914211044882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study examined the involvement of purinergic signaling components in
the rat model of hippocampal degeneration induced by trimethyltin (TMT)
intoxication (8 mg/kg, single intraperitoneal injection), which results in
behavioral and neurological dysfunction similar to neurodegenerative disorders.
We investigated spatial and temporal patterns of ecto-nucleoside triphosphate
diphosphohydrolase 1 (NTPDase1/CD39) and ecto-5′ nucleotidase (eN/CD73)
activity, their cell-specific localization, and analyzed gene expression pattern
and/or cellular localization of purinoreceptors and proinflammatory mediators
associated with reactive glial cells. Our study demonstrated that all Iba1+
cells at the injured area, irrespective of their morphology, upregulated
NTPDase1/CD39, while induction of eN/CD73 has been observed at amoeboid Iba1+
cells localized within the hippocampal neuronal layers with pronounced cell
death. Marked induction of P2Y12R, P2Y6R, and
P2X4-messenger RNA at the early stage of TMT-induced
neurodegeneration might reflect the functional properties, migration, and
chemotaxis of microglia, while induction of P2X7R at amoeboid cells
probably modulates their phagocytic role. Reactive astrocytes expressed
adenosine A1, A2A, and P2Y1 receptors, revealed
induction of complement component C3, inducible nitric oxide synthase, nuclear
factor-kB, and proinflammatory cytokines at the late stage of TMT-induced
neurodegeneration. An increased set of purinergic system components on activated
microglia (NTPDase1/CD39, eN/CD73, and P2X7) and astrocytes
(A1R, A2AR, and P2Y1), and loss of
homeostatic glial and neuronal purinergic pathways (P2Y12 and
A1R) may shift purinergic signaling balance toward excitotoxicity
and inflammation, thus favoring progression of pathological events. These
findings may contribute to a better understanding of the involvement of
purinergic signaling components in the progression of neurodegenerative
disorders that could be target molecules for the development of novel
therapies.
Collapse
Affiliation(s)
- Milorad Dragić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences-National Institute of thе Republic of Serbia, 89101University of Belgrade, Belgrade, Serbia
| | - Marija Adžić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Center for Laser Microscopy, 98829Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nadežda Nedeljković
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ivana Grković
- Department of Molecular Biology and Endocrinology, VINČA Institute of Nuclear Sciences-National Institute of thе Republic of Serbia, 89101University of Belgrade, Belgrade, Serbia
| |
Collapse
|
49
|
Fu P, Yung KKL. Air Pollution and Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2021; 77:701-714. [PMID: 32741830 DOI: 10.3233/jad-200483] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ambient air pollution has been associated with Alzheimer's disease (AD) in the elderly. However, its effects on AD have not been meta-analyzed comprehensively. OBJECTIVE We conducted a systematic review and meta-analysis to assess the associations between air pollution and AD incidence. METHODS We searched PubMed and Web of Science for indexed publications up to March 2020. Odds risk (OR) and confidence intervals (CI) were estimated for particulate matter (PM)10 (PM10), PM2.5, ozone (O3), nitrogen dioxide (NO2), sulfur dioxide (SO2), and carbon monoxide (CO). The subgroup analysis was conducted based on the pollution levels. RESULTS Nine studies were included in the meta-analysis and review. The OR per 10μg/m3 increase of PM2.5 was 1.95 (95% CI: 0.88-4.30). The corresponding values per 10μg/m3 increment of other pollutants were 1.03 (95% CI: 0.68-1.57) for O3, 1.00 (95% CI: 0.89-1.13) for NO2, and 0.95 (95% CI: 0.91-0.99) for PM10 (only one study), respectively. Overall OR of the five air pollutants above with AD was 1.32 (95% CI: 1.09-1.61), suggesting a positive association between ambient air pollution and AD incidence. The sub-analysis indicated that the OR (2.20) in heavily polluted regions was notably higher than that in lightly polluted regions (1.06). Although AD risk rate data related to SO2 or CO exposure are still limited, the epidemiologic and toxicological evidence indicated that higher concentration of SO2 or CO exposure increased risks of dementia, implying that SO2 or CO might have a potential impact on AD. CONCLUSION Air pollution exposure may exacerbate AD development.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ken Kin Lam Yung
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.,Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
50
|
Tsamou M, Pistollato F, Roggen EL. A Tau-Driven Adverse Outcome Pathway Blueprint Toward Memory Loss in Sporadic (Late-Onset) Alzheimer's Disease with Plausible Molecular Initiating Event Plug-Ins for Environmental Neurotoxicants. J Alzheimers Dis 2021; 81:459-485. [PMID: 33843671 DOI: 10.3233/jad-201418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The worldwide prevalence of sporadic (late-onset) Alzheimer's disease (sAD) is dramatically increasing. Aging and genetics are important risk factors, but systemic and environmental factors contribute to this risk in a still poorly understood way. Within the frame of BioMed21, the Adverse Outcome Pathway (AOP) concept for toxicology was recommended as a tool for enhancing human disease research and accelerating translation of data into human applications. Its potential to capture biological knowledge and to increase mechanistic understanding about human diseases has been substantiated since. In pursuit of the tau-cascade hypothesis, a tau-driven AOP blueprint toward the adverse outcome of memory loss is proposed. Sequences of key events and plausible key event relationships, triggered by the bidirectional relationship between brain cholesterol and glucose dysmetabolism, and contributing to memory loss are captured. To portray how environmental factors may contribute to sAD progression, information on chemicals and drugs, that experimentally or epidemiologically associate with the risk of AD and mechanistically link to sAD progression, are mapped on this AOP. The evidence suggests that chemicals may accelerate disease progression by plugging into sAD relevant processes. The proposed AOP is a simplified framework of key events and plausible key event relationships representing one specific aspect of sAD pathology, and an attempt to portray chemical interference. Other sAD-related AOPs (e.g., Aβ-driven AOP) and a better understanding of the impact of aging and genetic polymorphism are needed to further expand our mechanistic understanding of early AD pathology and the potential impact of environmental and systemic risk factors.
Collapse
|