1
|
Ahmed G, Rahaman MS, Perez E, Khan KM. Associations of Environmental Exposure to Arsenic, Manganese, Lead, and Cadmium with Alzheimer's Disease: A Review of Recent Evidence from Mechanistic Studies. J Xenobiot 2025; 15:47. [PMID: 40278152 PMCID: PMC12029005 DOI: 10.3390/jox15020047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Numerous epidemiological studies indicate that populations exposed to environmental toxicants such as heavy metals have a higher likelihood of developing Alzheimer's disease (AD) compared to those unexposed, indicating a potential association between heavy metals exposure and AD. The aim of this review is to summarize contemporary mechanistic research exploring the associations of four important metals, arsenic (As), manganese (Mn), lead (Pb), and cadmium (Cd), with AD and possible pathways, processes, and molecular mechanisms on the basis of data from the most recent mechanistic studies. Primary research publications published during the last decade were identified via a search of the PubMed Database. A thorough literature search and final screening yielded 45 original research articles for this review. Of the 45 research articles, 6 pertain to As, 9 to Mn, 21 to Pb, and 9 to Cd exposures and AD pathobiology. Environmental exposure to these heavy metals induces a wide range of pathological processes that intersect with well-known mechanisms leading to AD, such as oxidative stress, mitochondrial dysfunction, protein aggregation, neuroinflammation, autophagy dysfunction, and tau hyperphosphorylation. While exposure to single metals shares some affected pathways, certain effects are unique to specific metals. For instance, Pb disrupts the blood-brain barrier (BBB) and mitochondrial functions and alters AD-related genes epigenetically. Cd triggers neuronal senescence via p53/p21/Rb. As disrupts nitric oxide (NO) signaling, cortical, and synaptic function. Mn causes glutamate excitotoxicity and dopamine neuron damage. Our review provides a deeper understanding of biological mechanisms showing how metals contribute to AD. Information regarding the potential metal-induced toxicity relevant to AD may help us develop effective therapeutic AD intervention, treatment, and prevention.
Collapse
Affiliation(s)
- Giasuddin Ahmed
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX 78041, USA;
| | - Md. Shiblur Rahaman
- Department of Public Health, College of Health Sciences, Sam Houston State University, Huntsville, TX 77341, USA;
- Department of Environmental Science and Disaster Management, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Enrique Perez
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX 78041, USA;
| | - Khalid M. Khan
- Department of Public Health, College of Health Sciences, Sam Houston State University, Huntsville, TX 77341, USA;
| |
Collapse
|
2
|
Xia Y, Li Z, Wang C, Zhang X, Li J, Zhou Q, Yang J, Chen Q, Meng X, Wang J. Dynamic alterations of locomotor activity and the microbiota in zebrafish larvae with low concentrations of lead exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:2042-2052. [PMID: 38051486 DOI: 10.1007/s11356-023-31279-w] [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: 06/22/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
Lead (Pb) is a ubiquitous heavy metal associated with developmental and behavioral disorders. The establishment of pioneer microbiota overlaps with the development of the brain during early life, and Pb-induced developmental neurotoxicity may be partially caused by early-life microbiota dysbiosis. This study investigated the locomotor activity and the microbiota in developing zebrafish at multiple developmental time points (five days post fertilization [5 dpf], 6 dpf, and 7 dpf) under exposure to low concentrations of lead (0.05 mg/L). Time-dependent reductions in the number of activities and the average movement distance of larvae compared to the control were observed following Pb exposure. Furthermore, Pb exposure significantly altered the composition of the gut microbiota of zebrafish larvae. At the phylum level, the abundance of Proteobacteria decreased from 5 to 7 dpf, while that of Actinobacteria increased in the control groups. At the class level, the proportion of Alphaproteobacteria decreased, while that of Actinobacteria increased in the control groups. Notably, all showed the opposite trend in Pb groups. A correlation analysis between indices of locomotor activity and microbial communities revealed genus-level features that were clearly linked to the neurobehavioral performance of zebrafish. Seven genera were significantly correlated with the two performance indicators of the locomotion analysis, namely Rhodococcus, Deinococcus, Bacillus, Bosea, Bradyrhizobium, Staphylococcus, and Rhizobium. Rhizobium was dominant in zebrafish and increased in the Pb groups in a time-dependent manner. In addition, the expression levels of bdnf, trkb1, trkb2, and p75ntr changed in zebrafish from 5 to 7 dpf under Pb exposure. Collectively, these results suggest that Pb-induced neurotoxicity could potentially be treated by targeting the gut microbiota.
Collapse
Affiliation(s)
- Yuan Xia
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Ziyi Li
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Chunyu Wang
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Xiaoshun Zhang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Junyi Li
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Yang
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Qingsong Chen
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Junyi Wang
- School of Public Health, Guangdong Pharmaceutical University, Jianghaidadao, Guangzhou, 283, Guangdong, China.
| |
Collapse
|
3
|
Parithathvi A, Choudhari N, Dsouza HS. Prenatal and early life lead exposure induced neurotoxicity. Hum Exp Toxicol 2024; 43:9603271241285523. [PMID: 39340316 DOI: 10.1177/09603271241285523] [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] [Indexed: 09/30/2024]
Abstract
Lead (Pb) has become a major environmental contaminant. There are several ways in which lead can enter the human body and cause toxic effects on human health. This review focuses on the impact of lead toxicity at prenatal and early life stages and its effect on neurodevelopment. Lead exposure to the developing foetus targets foetal neural stem cells. Hence, it has detrimental effects on developing neural and glial cells, adversely influencing cognition and behaviour. Lead has a profound influence on the movement of calcium ions (Ca2+), which can be attributed to most of the mechanisms by which lead affects neurodevelopment. There is no known safe threshold of lead exposure for children. Lead can affect foetal neurodevelopment leading to various neurological disorders, and neurotoxic effects on behavioural and cognitive outcomes. In this review, we discuss prenatal and early-life lead exposure, its mechanism, and consequences for neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease in later stages of life. This review further highlights the importance of lead exposure during pregnancy and lactation periods as well as early development of the child in understanding the extent of lead-induced neurological damage to the foetus/children and the associated future risks.
Collapse
Affiliation(s)
- Aluru Parithathvi
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Neha Choudhari
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Herman S Dsouza
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
4
|
Doroszkiewicz J, Farhan JA, Mroczko J, Winkel I, Perkowski M, Mroczko B. Common and Trace Metals in Alzheimer's and Parkinson's Diseases. Int J Mol Sci 2023; 24:15721. [PMID: 37958705 PMCID: PMC10649239 DOI: 10.3390/ijms242115721] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Trace elements and metals play critical roles in the normal functioning of the central nervous system (CNS), and their dysregulation has been implicated in neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). In a healthy CNS, zinc, copper, iron, and manganese play vital roles as enzyme cofactors, supporting neurotransmission, cellular metabolism, and antioxidant defense. Imbalances in these trace elements can lead to oxidative stress, protein aggregation, and mitochondrial dysfunction, thereby contributing to neurodegeneration. In AD, copper and zinc imbalances are associated with amyloid-beta and tau pathology, impacting cognitive function. PD involves the disruption of iron and manganese levels, leading to oxidative damage and neuronal loss. Toxic metals, like lead and cadmium, impair synaptic transmission and exacerbate neuroinflammation, impacting CNS health. The role of aluminum in AD neurofibrillary tangle formation has also been noted. Understanding the roles of these elements in CNS health and disease might offer potential therapeutic targets for neurodegenerative disorders. The Codex Alimentarius standards concerning the mentioned metals in foods may be one of the key legal contributions to safeguarding public health. Further research is needed to fully comprehend these complex mechanisms and develop effective interventions.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Jakub Ali Farhan
- Department of Public International Law and European Law, Faculty of Law, University of Bialystok, 15-089 Bialystok, Poland
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Scinawa, Poland
| | - Maciej Perkowski
- Department of Public International Law and European Law, Faculty of Law, University of Bialystok, 15-089 Bialystok, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
5
|
Shvachiy L, Amaro-Leal Â, Outeiro TF, Rocha I, Geraldes V. Intermittent Lead Exposure Induces Behavioral and Cardiovascular Alterations Associated with Neuroinflammation. Cells 2023; 12:cells12050818. [PMID: 36899953 PMCID: PMC10000953 DOI: 10.3390/cells12050818] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
The nervous system is the primary target for lead exposure and the developing brain appears to be especially susceptible, namely the hippocampus. The mechanisms of lead neurotoxicity remain unclear, but microgliosis and astrogliosis are potential candidates, leading to an inflammatory cascade and interrupting the pathways involved in hippocampal functions. Moreover, these molecular changes can be impactful as they may contribute to the pathophysiology of behavioral deficits and cardiovascular complications observed in chronic lead exposure. Nevertheless, the health effects and the underlying influence mechanism of intermittent lead exposure in the nervous and cardiovascular systems are still vague. Thus, we used a rat model of intermittent lead exposure to determine the systemic effects of lead and on microglial and astroglial activation in the hippocampal dentate gyrus throughout time. In this study, the intermittent group was exposed to lead from the fetal period until 12 weeks of age, no exposure (tap water) until 20 weeks, and a second exposure from 20 to 28 weeks of age. A control group (without lead exposure) matched in age and sex was used. At 12, 20 and 28 weeks of age, both groups were submitted to a physiological and behavioral evaluation. Behavioral tests were performed for the assessment of anxiety-like behavior and locomotor activity (open-field test), and memory (novel object recognition test). In the physiological evaluation, in an acute experiment, blood pressure, electrocardiogram, and heart and respiratory rates were recorded, and autonomic reflexes were evaluated. The expression of GFAP, Iba-1, NeuN and Synaptophysin in the hippocampal dentate gyrus was assessed. Intermittent lead exposure induced microgliosis and astrogliosis in the hippocampus of rats and changes in behavioral and cardiovascular function. We identified increases in GFAP and Iba1 markers together with presynaptic dysfunction in the hippocampus, concomitant with behavioral changes. This type of exposure produced significant long-term memory dysfunction. Regarding physiological changes, hypertension, tachypnea, baroreceptor reflex impairment and increased chemoreceptor reflex sensitivity were observed. In conclusion, the present study demonstrated the potential of lead intermittent exposure inducing reactive astrogliosis and microgliosis, along with a presynaptic loss that was accompanied by alterations of homeostatic mechanisms. This suggests that chronic neuroinflammation promoted by intermittent lead exposure since fetal period may increase the susceptibility to adverse events in individuals with pre-existing cardiovascular disease and/or in the elderly.
Collapse
Affiliation(s)
- Liana Shvachiy
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Ângela Amaro-Leal
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Max Planck Institute for Natural Science, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37073 Göttingen, Germany
| | - Isabel Rocha
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Vera Geraldes
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
- Correspondence: ; Tel.: +351-217999435
| |
Collapse
|
6
|
Islam F, Shohag S, Akhter S, Islam MR, Sultana S, Mitra S, Chandran D, Khandaker MU, Ashraf GM, Idris AM, Emran TB, Cavalu S. Exposure of metal toxicity in Alzheimer's disease: An extensive review. Front Pharmacol 2022; 13:903099. [PMID: 36105221 PMCID: PMC9465172 DOI: 10.3389/fphar.2022.903099] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Metals serve important roles in the human body, including the maintenance of cell structure and the regulation of gene expression, the antioxidant response, and neurotransmission. High metal uptake in the nervous system is harmful because it can cause oxidative stress, disrupt mitochondrial function, and impair the activity of various enzymes. Metal accumulation can cause lifelong deterioration, including severe neurological problems. There is a strong association between accidental metal exposure and various neurodegenerative disorders, including Alzheimer's disease (AD), the most common form of dementia that causes degeneration in the aged. Chronic exposure to various metals is a well-known environmental risk factor that has become more widespread due to the rapid pace at which human activities are releasing large amounts of metals into the environment. Consequently, humans are exposed to both biometals and heavy metals, affecting metal homeostasis at molecular and biological levels. This review highlights how these metals affect brain physiology and immunity and their roles in creating harmful proteins such as β-amyloid and tau in AD. In addition, we address findings that confirm the disruption of immune-related pathways as a significant toxicity mechanism through which metals may contribute to AD.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Shomaya Akhter
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sharifa Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Subang Jaya, Malaysia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
7
|
Rashno M, Sarkaki A, Ghaderi S, Khoshnam SE. Sesamin: Insights into its protective effects against lead-induced learning and memory deficits in rats. J Trace Elem Med Biol 2022; 72:126993. [PMID: 35550983 DOI: 10.1016/j.jtemb.2022.126993] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/03/2022] [Accepted: 05/03/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Lead (Pb) is one of the most hazardous pollutants that induce a wide spectrum of neurological changes such as learning and memory deficits. Sesamin, a phytonutrient of the lignan class, exhibits anti-inflammatory, anti-apoptotic, and neuroprotective properties. The present study was designed to investigate the effects of sesamin against Pb-induced learning and memory deficits, disruption of hippocampal theta and gamma rhythms, inflammatory response, inhibition of blood δ-aminolevulinic acid dehydratase (δ-ALA-D) activity, Pb accumulation, and neuronal loss in rats. METHODS Sesamin treatment (30 mg/kg/day; P.O.) was started simultaneously with Pb acetate exposure (500 ppm in standard drinking water) in rats, and they continued for eight consecutive weeks. RESULTS The results showed that chronic exposure to Pb disrupted the learning and memory functions in both passive-avoidance and water-maze tests, which was accompanied by increase in spectral theta power and theta/gamma ratio, and a decrease in spectral gamma power in the hippocampus. Additionally, Pb exposure resulted in an enhanced tumor necrosis factor-alpha (TNF-α) content, decreased interleukin-10 (IL-10) production, inhibited blood δ-ALA-D activity, increased Pb accumulation, and neuronal loss of rats. In contrast, sesamin treatment improved all the above-mentioned Pb-induced pathological changes. CONCLUSION This data suggests that sesamin could improve Pb-induced learning and memory deficits, possibly through amelioration of hippocampal theta and gamma rhythms, modulation of inflammatory status, restoration of the blood δ-ALA-D activity, reduction of Pb accumulation in the blood and the brain tissues, and prevention of neuronal loss.
Collapse
Affiliation(s)
| | - Alireza Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Tsamou M, Roggen EL. Building a Network of Adverse Outcome Pathways (AOPs) Incorporating the Tau-Driven AOP Toward Memory Loss (AOP429). J Alzheimers Dis Rep 2022; 6:271-296. [PMID: 35891639 PMCID: PMC9277675 DOI: 10.3233/adr-220015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/15/2022] [Indexed: 11/15/2022] Open
Abstract
The adverse outcome pathway (AOP) concept was first proposed as a tool for chemical hazard assessment facilitating the regulatory decision-making in toxicology and was more recently recommended during the BioMed21 workshops as a tool for the characterization of crucial endpoints in the human disease development. This AOP framework represents mechanistically based approaches using existing data, more realistic and relevant to human biological systems. In principle, AOPs are described by molecular initiating events (MIEs) which induce key events (KEs) leading to adverse outcomes (AOs). In addition to the individual AOPs, the network of AOPs has been also suggested to beneficially support the understanding and prediction of adverse effects in risk assessment. The AOP-based networks can capture the complexity of biological systems described by different AOPs, in which multiple AOs diverge from a single MIE or multiple MIEs trigger a cascade of KEs that converge to a single AO. Here, an AOP network incorporating a recently proposed tau-driven AOP toward memory loss (AOP429) related to sporadic (late-onset) Alzheimer’s disease is constructed. This proposed AOP network is an attempt to extract useful information for better comprehending the interactions among existing mechanistic data linked to memory loss as an early phase of sporadic Alzheimer’s disease pathology.
Collapse
Affiliation(s)
- Maria Tsamou
- ToxGenSolutions (TGS), Maastricht, The Netherlands
| | | |
Collapse
|
9
|
Gauvrit T, Benderradji H, Buée L, Blum D, Vieau D. Early-Life Environment Influence on Late-Onset Alzheimer's Disease. Front Cell Dev Biol 2022; 10:834661. [PMID: 35252195 PMCID: PMC8891536 DOI: 10.3389/fcell.2022.834661] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
With the expand of the population's average age, the incidence of neurodegenerative disorders has dramatically increased over the last decades. Alzheimer disease (AD) which is the most prevalent neurodegenerative disease is mostly sporadic and primarily characterized by cognitive deficits and neuropathological lesions such as amyloid -β (Aβ) plaques and neurofibrillary tangles composed of hyper- and/or abnormally phosphorylated Tau protein. AD is considered a complex disease that arises from the interaction between environmental and genetic factors, modulated by epigenetic mechanisms. Besides the well-described cognitive decline, AD patients also exhibit metabolic impairments. Metabolic and cognitive perturbations are indeed frequently observed in the Developmental Origin of Health and Diseases (DOHaD) field of research which proposes that environmental perturbations during the perinatal period determine the susceptibility to pathological conditions later in life. In this review, we explored the potential influence of early environmental exposure to risk factors (maternal stress, malnutrition, xenobiotics, chemical factors … ) and the involvement of epigenetic mechanisms on the programming of late-onset AD. Animal models indicate that offspring exposed to early-life stress during gestation and/or lactation increase both AD lesions, lead to defects in synaptic plasticity and finally to cognitive impairments. This long-lasting epigenetic programming could be modulated by factors such as nutriceuticals, epigenetic modifiers or psychosocial behaviour, offering thus future therapeutic opportunity to protect from AD development.
Collapse
Affiliation(s)
- Thibaut Gauvrit
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Hamza Benderradji
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
10
|
Gu H, Xu Y, Du N, Yu Y, Zheng W, Du Y. Pb Induces MCP-1 in the Choroid Plexus. BIOLOGY 2022; 11:308. [PMID: 35205174 PMCID: PMC8869661 DOI: 10.3390/biology11020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/06/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022]
Abstract
Lead (Pb) is an environmental element that has been implicated in the development of dementia and Alzheimer's disease (AD). Additionally, innate immune activation contributes to AD pathophysiology. However, the mechanisms involved remain poorly understood. The choroid plexus (CP) is not only the site of cerebrospinal fluid (CSF) production, but also an important location for communication between the circulation and the CSF. In this study, we investigated the involvement of the CP during Pb exposure by evaluating the expression of the monocyte chemoattractant protein-1 (MCP-1). MCP-1 is highly expressed in the CP compared to other CNS tissues. MCP-1 regulates macrophage infiltration and is upregulated in AD brains. Our study revealed that Pb exposure stimulated MCP-1 expression, along with a significantly increased macrophage infiltration into the CP. By using cultured Z310 rat CP cells, Pb exposure stimulated MCP-1 expression in a dose-related fashion and markedly activated both NF-κB and p38 MAP kinase. Interestingly, both SB 203580, a p38 inhibitor, and BAY 11-7082, an NF-κB p65 inhibitor, significantly blocked Pb-induced MCP-1 expression. However, SB203580 did not directly inhibit NF-κB p65 phosphorylation. In conclusion, Pb exposure stimulates MCP-1 expression via the p38 and NF-κB p65 pathways along with macrophage infiltration into the CP.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| | - Yundan Xu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Nicole Du
- Department of Pediatrics, Children’s National Hospital, Washington, DC 20010, USA;
| | - Yongqi Yu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| |
Collapse
|
11
|
Collins AE, Saleh TM, Kalisch BE. Naturally Occurring Antioxidant Therapy in Alzheimer's Disease. Antioxidants (Basel) 2022; 11:213. [PMID: 35204096 PMCID: PMC8868221 DOI: 10.3390/antiox11020213] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
It is estimated that the prevalence rate of Alzheimer's disease (AD) will double by the year 2040. Although currently available treatments help with symptom management, they do not prevent, delay the progression of, or cure the disease. Interestingly, a shared characteristic of AD and other neurodegenerative diseases and disorders is oxidative stress. Despite profound evidence supporting the role of oxidative stress in the pathogenesis and progression of AD, none of the currently available treatment options address oxidative stress. Recently, attention has been placed on the use of antioxidants to mitigate the effects of oxidative stress in the central nervous system. In preclinical studies utilizing cellular and animal models, natural antioxidants showed therapeutic promise when administered alone or in combination with other compounds. More recently, the concept of combination antioxidant therapy has been explored as a novel approach to preventing and treating neurodegenerative conditions that present with oxidative stress as a contributing factor. In this review, the relationship between oxidative stress and AD pathology and the neuroprotective role of natural antioxidants from natural sources are discussed. Additionally, the therapeutic potential of natural antioxidants as preventatives and/or treatment for AD is examined, with special attention paid to natural antioxidant combinations and conjugates that are currently being investigated in human clinical trials.
Collapse
Affiliation(s)
| | | | - Bettina E. Kalisch
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.E.C.); (T.M.S.)
| |
Collapse
|
12
|
Bandaru LJM, Ayyalasomayajula N, Murumulla L, Challa S. Mechanisms associated with the dysregulation of mitochondrial function due to lead exposure and possible implications on the development of Alzheimer's disease. Biometals 2022; 35:1-25. [PMID: 35048237 DOI: 10.1007/s10534-021-00360-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/09/2021] [Indexed: 01/17/2023]
Abstract
Lead (Pb) is a multimedia contaminant with various pathophysiological consequences, including cognitive decline and neural abnormalities. Recent findings have reported an association of Pb toxicity with Alzheimer's disease (AD). Studies have revealed that mitochondrial dysfunction is a pathological characteristic of AD. According to toxicology reports, Pb promotes mitochondrial oxidative stress by lowering complex III activity in the electron transport chain, boosting reactive oxygen species formation, and reducing the cell's antioxidant defence system. Here, we review recent advances in the role of mitochondria in Pb-induced AD pathology, as well as the mechanisms associated with the mitochondrial dysfunction, such as the depolarisation of the mitochondrial membrane potential, mitochondrial permeability transition pore opening; mitochondrial biogenesis, bioenergetics and mitochondrial dynamics alterations; and mitophagy and apoptosis. We also discuss possible therapeutic options for mitochondrial-targeted neurodegenerative disease (AD).
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Neelima Ayyalasomayajula
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
13
|
Tasin FR, Ahmed A, Halder D, Mandal C. On-going consequences of in utero exposure of Pb: An epigenetic perspective. J Appl Toxicol 2022; 42:1553-1569. [PMID: 35023172 DOI: 10.1002/jat.4287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 11/08/2022]
Abstract
Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hyper- and hypo-methylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration- and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as - impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.
Collapse
Affiliation(s)
- Fahim Rejanur Tasin
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Asif Ahmed
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Debasish Halder
- Rare Disease research center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
14
|
Ogbodo JO, Agbo CP, Njoku UO, Ogugofor MO, Egba SI, Ihim SA, Echezona AC, Brendan KC, Upaganlawar AB, Upasani CD. Alzheimer's Disease: Pathogenesis and Therapeutic Interventions. Curr Aging Sci 2022; 15:2-25. [PMID: 33653258 DOI: 10.2174/1874609814666210302085232] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/04/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common cause of dementia. Genetics, excessive exposure to environmental pollutants, as well as unhealthy lifestyle practices are often linked to the development of AD. No therapeutic approach has achieved complete success in treating AD; however, early detection and management with appropriate drugs are key to improving prognosis. INTERVENTIONS The pathogenesis of AD was extensively discussed in order to understand the reasons for the interventions suggested. The interventions reviewed include the use of different therapeutic agents and approaches, gene therapy, adherence to healthy dietary plans (Mediterranean diet, Okinawan diet and MIND diet), as well as the use of medicinal plants. The potential of nanotechnology as a multidisciplinary and interdisciplinary approach in the design of nano-formulations of AD drugs and the use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as theranostic tools for early detection of Alzheimer's disease were also discussed.
Collapse
Affiliation(s)
- John O Ogbodo
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Chinazom P Agbo
- Department of Pharmaceutics, University of Nigeria, Nsukka, Nigeria
| | - Ugochi O Njoku
- Department of Biochemistry, University of Nigeria, Nsukka, Nigeria
| | | | - Simeon I Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Nigeria
| | - Stella A Ihim
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | | | | | - Aman B Upaganlawar
- Department of Pharmacology, Sureshdada Shriman\'s College of Pharmacy, New Dehli, India
| | | |
Collapse
|
15
|
Basaly V, Hill J, Bihaqi SW, Marques E, Slitt AL, Zawia NH. Developmental Perfluorooctanesulfonic acid (PFOS) exposure as a potential risk factor for late-onset Alzheimer's disease in CD-1 mice and SH-SY5Y cells. Neurotoxicology 2021; 86:26-36. [PMID: 34224775 DOI: 10.1016/j.neuro.2021.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accounts for approximately 60-80% of dementia cases worldwide and is characterized by an accumulation of extracellular senile plaques composed of β-amyloid (Aβ) peptide and intracellular neurofibrillary tangles (NFTs) containing hyperphosphorylated tau protein. Sporadic or late-onset AD (LOAD) represents 95 % of the AD cases and its etiology does not appear to follow Mendelian laws of inheritance, thus, implicating the role of epigenetic programming and environmental factors. Apolipoprotein allele 4 (ApoE4), the only established genetic risk factor for LOAD, is suggested to accelerate the pathogenesis of AD by increasing tau hyperphosphorylation, inhibiting the clearance of amyloid-β (Aβ), and promoting Aβ aggregation. Perfluorooctanesulfonic acid (PFOS) is a persistent organic pollutant, with potential neurotoxic effects, that poses a major threat to the ecosystem and human health. By employing in vivo and in vitro models, the present study investigated PFOS as a potential risk factor for LOAD by assessing its impact on amyloidogenesis, tau pathology, and rodent behavior. Our behavioral analysis revealed that developmentally exposed male and female mice exhibited a strong trend of increased rearing and significantly increased distance traveled in the open field test. Biochemically, GSK3β and total ApoE were increased following developmental exposure, in vivo. Furthermore, in vitro, low concentrations of PFOS elevated protein levels of APP, tau, and its site-specific phosphorylation. Differentiated SH-SY5Y cells exposed to a series of PFOS concentrations, also, had elevated protein expression of GSK3β. These data suggest that total ApoE is inducible by environmental exposure to PFOS.
Collapse
Affiliation(s)
- Veronia Basaly
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Jaunetta Hill
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Syed Waseem Bihaqi
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Emily Marques
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Nasser H Zawia
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, 02881, USA; Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
16
|
Thawkar BS, Kaur G. Zebrafish as a Promising Tool for Modeling Neurotoxin-Induced Alzheimer's Disease. Neurotox Res 2021; 39:949-965. [PMID: 33687726 DOI: 10.1007/s12640-021-00343-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/17/2021] [Accepted: 02/21/2021] [Indexed: 12/01/2022]
Abstract
Drug discovery and development for Alzheimer's disease (AD) are complex and challenging due to the higher failure rate in the drug development process. The overproduction and deposition of Aβ senile plaque and intracellular neurofibrillary tangle (NFT) formation are well-recognized diagnostic hallmarks of AD. Numerous transgenic models of Alzheimer's disease have restrictions on cost-effectiveness and time in the preclinical setup. Zebrafish has emerged as an excellent complementary model for neurodegenerative research due to simpler organisms with robust, clearly visible behavior forms. Glutaminergic and cholinergic pathways responsible for learning and memory are present in zebrafish and actively participate in the transmission process. Therefore, it is imperative to study neurotoxic agents' mechanisms that induce dysfunction of memory, learning, and neurons in the zebrafish. This review illustrates the in-depth molecular mechanism of several neurotoxic agents such as okadaic acid, cigarette smoke extract, and metals to produce cognitive deficits or neurodegeneration similar to mammals. These updates would determine an ideal and effective neurotoxic agent for producing AD pathophysiology in the zebrafish brain for preclinical screening.
Collapse
Affiliation(s)
- Baban S Thawkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), 400056, Mumbai, India
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), 400056, Mumbai, India.
| |
Collapse
|
17
|
Finch CE, Morgan TE. Developmental Exposure to Air Pollution, Cigarettes, and Lead: Implications for Brain Aging. ACTA ACUST UNITED AC 2020. [DOI: 10.1146/annurev-devpsych-042320-044338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brain development is impaired by maternal exposure to airborne toxins from ambient air pollution, cigarette smoke, and lead. Shared postnatal consequences include gray matter deficits and abnormal behaviors as well as elevated blood pressure. These unexpectedly broad convergences have implications for later life brain health because these same airborne toxins accelerate brain aging. Gene-environment interactions are shown for ApoE alleles that influence the risk of Alzheimer disease. The multigenerational trace of these toxins extends before fertilization because egg cells are formed in the grandmaternal uterus. The lineage and sex-specific effects of grandmaternal exposure to lead and cigarettes indicate epigenetic processes of relevance to future generations from our current and recent exposure to airborne toxins.
Collapse
Affiliation(s)
- Caleb E. Finch
- Leonard Davis School of Gerontology and Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089-0191, USA;,
| | - Todd E. Morgan
- Leonard Davis School of Gerontology and Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089-0191, USA;,
| |
Collapse
|
18
|
Wu Z, Bai L, Tu R, Zhang L, Ba Y, Zhang H, Li X, Cheng X, Li W, Huang H. Disruption of synaptic expression pattern and age-related DNA oxidation in a neuronal model of lead-induced toxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 76:103350. [PMID: 32058320 DOI: 10.1016/j.etap.2020.103350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/18/2020] [Accepted: 02/03/2020] [Indexed: 06/10/2023]
Abstract
Lead (Pb) is recognized as a potent inducer of synaptic toxicity generally associated with reduced synaptic transmission and increased neuronal fiber excitability, becoming an environmental risk for neurodegenerative processes. Despite numerous toxicological studies on Pb have been directed to the developing brain, attention concerning long-term consequences of pubertal chronic Pb exposure on neuronal activity is still lacking. Thus, we exposed 4-week-old male mice to 0.2 % lead acetate solution for one month, then, conducted behavioral tests or extracted brain homogenate from mice prefrontal cortex (PFC) and hippocampus at the age of 4, 13 and 16-month-old respectively. Our results showed that treated mice exhibited an evident increase in latency to reach platform following pubertal Pb exposure and aging. The increase of 8-OHdG revealed evident neural DNA oxidative damage across time upon pubertal Pb exposure. In the hippocampus of lead exposed mice at three age nodes, the expression of brain-derived neurotrophic factor precursor (proBDNF) increased, while that of mature BDNF (mBDNF), cAMP-response element binding protein (CREB) and phosphorylated CREB (pCREB) decreased compared with the control group. Furthermore, the expression of BACE1 protein and tau phosphorylation level in PFC and hippocampus increased, APP mRNAs in PFC and prolonged induction of BACE1 in hippocampus. Our results show that chronic Pb exposure from pubertal stage onward can either initiate divergent synaptic-related gene expression patterns in adulthood or trigger time-course of neurodegenerative profile within the PFC or hippocampus, which can contribute consistent deficits of cognition across subsequent age-nodes.
Collapse
Affiliation(s)
- Zuntao Wu
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Lin Bai
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Runqi Tu
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Lijie Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Yue Ba
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Xing Li
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Xuemin Cheng
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Wenjie Li
- College of Public Health, Zhengzhou University, Zhengzhou, PR China
| | - Hui Huang
- College of Public Health, Zhengzhou University, Zhengzhou, PR China.
| |
Collapse
|
19
|
Meyer DN, Crofts EJ, Akemann C, Gurdziel K, Farr R, Baker BB, Weber D, Baker TR. Developmental exposure to Pb 2+ induces transgenerational changes to zebrafish brain transcriptome. CHEMOSPHERE 2020; 244:125527. [PMID: 31816550 PMCID: PMC7015790 DOI: 10.1016/j.chemosphere.2019.125527] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 05/24/2023]
Abstract
Lead (Pb2+) is a major public health hazard for urban children, with profound and well-characterized developmental and behavioral implications across the lifespan. The ability of early Pb2+ exposure to induce epigenetic changes is well-established, suggesting that Pb2+-induced neurobehavioral deficits may be heritable across generations. Understanding the long-term and multigenerational repercussions of lead exposure is crucial for clarifying both the genotypic alterations behind these behavioral outcomes and the potential mechanism of heritability. To study this, zebrafish (Danio rerio) embryos (<2 h post fertilization; EK strain) were exposed for 24 h to waterborne Pb2+ at a concentration of 10 μM. This exposed F0 generation was raised to adulthood and spawned to produce the F1 generation, which was subsequently spawned to produce the F2 generation. Previous avoidance conditioning studies determined that a 10 μM Pb2+ dose resulted in learning impairments persisting through the F2 generation. RNA was extracted from control- and 10 μM Pb2+-lineage F2 brains, (n = 10 for each group), sequenced, and transcript expression was quantified utilizing Quant-Seq. 648 genes were differentially expressed in the brains of F2 lead-lineage fish versus F2 control-lineage fish. Pathway analysis revealed altered genes in processes including synaptic function and plasticity, neurogenesis, endocrine homeostasis, and epigenetic modification, all of which are implicated in lead-induced neurobehavioral deficits and/or their inheritance. These data will inform future investigations to elucidate the mechanism of adult-onset and transgenerational health effects of developmental lead exposure.
Collapse
Affiliation(s)
- Danielle N Meyer
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Emily J Crofts
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Camille Akemann
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Katherine Gurdziel
- Applied Genome Technology Center, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Rebecca Farr
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Bridget B Baker
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA; Division of Laboratory Animal Resources, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Daniel Weber
- Children's Environmental Health Sciences Core Center, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Tracie R Baker
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
20
|
Owsianowska J, Kamińska MS, Bosiacki M, Chlubek D, Karakiewicz B, Jurczak A, Stanisławska M, Barczak K, Grochans E. Depression, changes in peripheral blood cell count, and changes in selected biochemical parameters related to lead concentration in whole blood (Pb-B) of women in the menopausal period. J Trace Elem Med Biol 2020; 61:126501. [PMID: 32289550 DOI: 10.1016/j.jtemb.2020.126501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/24/2020] [Accepted: 03/18/2020] [Indexed: 11/24/2022]
Abstract
THE AIM The aim of this study was to assess the severity of depression, vasomotor symptoms, changes in peripheral blood cell count, and selected biochemical parameters in relation to the concentration of lead in whole blood of women in the perimenopausal period. METHODS The study sample consisted of 233 women from the general population of the West Pomeranian Province (Poland) in age between 44-65 years. The intensity of menopausal symptoms was examined using the Blatt-Kupperman Index, and the severity of depression using the Beck Depression Inventory. The following biochemical data were evaluated: concentrations of glucose, triglycerides, HDL, C-reactive protein, glycated haemoglobin, cortisol, insulin, blood cell count, and lead concentration in whole blood (Pb-B). RESULTS A whole blood Pb concentration below 5 μg/dl was found in 55 subjects (23.61 %), in 142 women (60.94 %) it ranged from 5 to 10 μg/dl, while in 36 women (15.45 %) was higher than 10 μg/dl. There was a strong positive correlation between Pb concentration in the blood of the examined women and the severity of depressive symptoms (Rs=+0.60, p = 0.001). The lowest mean values for total leukocytes (5.07 ± 1.22 thousand/μl) and neutrophils (2.76 ± 0.86 thousand/μl) were found in women with Pb concentration above 10 μg/dl (p < 0.05). There was a significant negative correlation between the number of total leukocytes (r=-0.45, p = 0.002) and neutrophils (r=-0.50, p = 0.001) and blood Pb concentration. Analysis showed statistically significant differences in glucose concentration (p < 0.05) between groups. Blood glucose was higher in women with Pb-B <5 and between 5-10 μg/dl than in women with Pb-B >10 μg/dl. CONCLUSION Exposure to Pb may be a factor playing a significant role in the development of depressive symptoms in menopausal women. It may also be associated with glucose metabolism disorders and immunosuppression in women during this period of life.
Collapse
Affiliation(s)
- Joanna Owsianowska
- Department of Specialized Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Magdalena Sylwia Kamińska
- Subdepartment of Long-Term Care, Department of Social Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Mateusz Bosiacki
- Department of Functional Diagnostics and Physical Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 56 Żołnierska St., 71-210, Szczecin, Poland.
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wielkopolskich St., 70-111, Szczecin, Poland.
| | - Beata Karakiewicz
- Subdepartment of Social Medicine and Public Health, Department of Social Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Anna Jurczak
- Department of Specialized Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Marzanna Stanisławska
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, 72 Powstańców Wielkopolskich St., 70-111, Szczecin, Poland.
| | - Elżbieta Grochans
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210, Szczecin, Poland.
| |
Collapse
|
21
|
Bakulski KM, Seo YA, Hickman RC, Brandt D, Vadari HS, Hu H, KyunPark S. Heavy Metals Exposure and Alzheimer's Disease and Related Dementias. J Alzheimers Dis 2020; 76:1215-1242. [PMID: 32651318 PMCID: PMC7454042 DOI: 10.3233/jad-200282] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease and related dementias lack effective treatment or cures and are major public health challenges. Risk for Alzheimer's disease and related dementias is partially attributable to environmental factors. The heavy metals lead, cadmium, and manganese are widespread and persistent in our environments. Once persons are exposed to these metals, they are adept at entering cells and reaching the brain. Lead and cadmium are associated with numerous health outcomes even at low levels of exposure. Although manganese is an essential metal, deficiency or environmental exposure or high levels of the metal can be toxic. In cell and animal model systems, lead, cadmium, and manganese are well documented neurotoxicants that contribute to canonical Alzheimer's disease pathologies. Adult human epidemiologic studies have consistently shown lead, cadmium, and manganese are associated with impaired cognitive function and cognitive decline. No longitudinal human epidemiology study has assessed lead or manganese exposure on Alzheimer's disease specifically though two studies have reported a link between cadmium and Alzheimer's disease mortality. More longitudinal epidemiologic studies with high-quality time course exposure data and incident cases of Alzheimer's disease and related dementias are warranted to confirm and estimate the proportion of risk attributable to these exposures. Given the widespread and global exposure to lead, cadmium, and manganese, even small increases in the risks of Alzheimer's disease and related dementias would have a major population impact on the burden on disease. This article reviews the experimental and epidemiologic literature of the associations between lead, cadmium, and manganese on Alzheimer's disease and related dementias and makes recommendations of critical areas of future investment.
Collapse
Affiliation(s)
- Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Young Ah Seo
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ruby C. Hickman
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Brandt
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Harita S. Vadari
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Howard Hu
- School of Public Health, University of Washington, Seattle, WA, USA
| | - Sung KyunPark
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Early-life Pb exposure as a potential risk factor for Alzheimer’s disease: are there hazards for the Mexican population? J Biol Inorg Chem 2019; 24:1285-1303. [DOI: 10.1007/s00775-019-01739-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/25/2019] [Indexed: 12/30/2022]
|
23
|
Liu J, Liao G, Tu H, Huang Y, Peng T, Xu Y, Chen X, Huang Z, Zhang Y, Meng X, Zou F. A protective role of autophagy in Pb-induced developmental neurotoxicity in zebrafish. CHEMOSPHERE 2019; 235:1050-1058. [PMID: 31561294 DOI: 10.1016/j.chemosphere.2019.06.227] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 06/10/2023]
Abstract
Lead (Pb) is one of the most toxic heavy metals and has aroused widespread concern as it can cause severe impairments in the developing nervous system. Autophagy has been proposed as an injury factor in Pb-induced neurotoxicity. In this study, we used zebrafish embryo as a model, measured the general toxic effects of Pb, and investigated the effect of Pb exposure on autophagy, and its role in Pb-induced developmental neurotoxicity. Zebrafish embryos were exposed to Pb at concentrations of 0, 0.1, 1 or 10 μM until 4 days post-fertilization. Our data showed that exposure to 10 μM Pb significantly reduced survival rates and impaired locomotor activity. Uptake of Pb was enhanced as the concentration and duration of exposure increased. Inhibition of lysosomal degradation with bafilomycin A1 treatment abolished the suppression of Lc3-II protein expression by Pb. Furthermore, autophagosome formation was inhibited by Pb in the brain. In addition, mRNA expression of beclin1, one of the critical genes in autophagy, were decreased in Pb exposure groups at 72 h post-fertilization. Whole-mount in situ hybridization assay showed that beclin1 gene expression in the brain was reduced by Pb. Rapamycin, an autophagy inducer, partly resolved developmental neurotoxicity induced by Pb exposure. Our results suggest that autophagy plays a protective role in the developmental neurotoxicity of Pb in zebrafish embryos and larvae.
Collapse
Affiliation(s)
- Jiaxian Liu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Gengze Liao
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Tu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Huang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Chen
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhibin Huang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yiyue Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Masoud AM, Bihaqi SW, Alansi B, Dash M, Subaiea GM, Renehan WE, Zawia NH. Altered microRNA, mRNA, and Protein Expression of Neurodegeneration-Related Biomarkers and Their Transcriptional and Epigenetic Modifiers in a Human Tau Transgenic Mouse Model in Response to Developmental Lead Exposure. J Alzheimers Dis 2019; 63:273-282. [PMID: 29614648 DOI: 10.3233/jad-170824] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyloid deposits originating from the amyloid-β protein precursor (AβPP) and aggregates of the microtubule associated protein tau (MAPT) are the hallmarks of Alzheimer's disease (AD). Animal studies have demonstrated a link between early life exposure to lead (Pb) and latent overexpression of the AβPP and MAPT genes and their products via epigenetic reprogramming. The present study monitored APP gene and epigenetic mediators and transcription factors known to regulate it. Western blot analysis and quantitative polymerase chain reaction (qPCR) were used to study the mRNA, miRNA, and proteins levels of AβPP, specificity protein 1 (SP1; a transcriptional regulator of amyloid and tau pathway), and epigenetic intermediates namely: DNA methyltransferase (DNMT) 1, DNMT3a and Methyl- CpG protein binding 2 (MeCP2) in the cerebral cortex of transgenic mice (Knock-in for human MAPT). These transgenic mice were developmentally exposed to Pb and the impact on mRNA, miRNA, and protein levels was scrutinized on postnatal days (PND) 20 and 50. The data revealed a consistent inverse relationship between miRNA and protein levels for SP1 and AβPP both in the basal and exposed conditions, which may influence the levels of their corresponding proteins. On the other hand, the relationship between miRNA and protein levels was not correlative for DNMT1 and DNMT3a. MeCP2 miRNA protein levels corresponded only following environmental exposure. These results suggest that developmental exposure to Pb and subsequent AβPP protein levels may be controlled through transcriptional regulators and epigenetic mechanisms that mainly involve miRNA regulation.
Collapse
Affiliation(s)
- Anwar M Masoud
- Biochemical Technology Program, Faculty of Applied Science, Thamar University, Thamar, Yemen
| | - Syed W Bihaqi
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| | - Bothaina Alansi
- Department of Biomedical and Pharmaceutical Science, University of Rhode Island, Kingston RI, USA
| | - Miriam Dash
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston RI, USA
| | - Gehad M Subaiea
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - William E Renehan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| | - Nasser H Zawia
- Department of Biomedical and Pharmaceutical Science, University of Rhode Island, Kingston RI, USA.,Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston RI, USA.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| |
Collapse
|
25
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Abstract
Millions of Americans now entering midlife and old age were exposed to high levels of lead, a neurotoxin, as children. Evidence from animal-model and human observational studies suggest that childhood lead exposure may raise the risk of adult neurodegenerative disease, particularly dementia, through a variety of possible mechanisms including epigenetic modification, delayed cardiovascular and kidney disease, direct degenerative CNS injury from lead remobilized from bone, and lowered neural and cognitive reserve. Within the next ten years, the generation of children with the highest historical lead exposures, those born in the 1960s, 1970s, and 1980s, will begin to enter the age at which dementia symptoms tend to emerge. Many will also enter the age in which lead stored in the skeleton may be remobilized at greater rates, particularly for women entering menopause and men and women experiencing osteoporosis. Should childhood lead exposure prove pro-degenerative, the next twenty years will provide the last opportunities for possible early intervention to forestall greater degenerative disease burden across the aging lead-exposed population. More evidence is needed now to characterize the nature and magnitude of the degenerative risks facing adults exposed to lead as children and to identify interventions to limit long-term harm.
Collapse
Affiliation(s)
- Aaron Reuben
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| |
Collapse
|
27
|
Bihaqi SW. Early life exposure to lead (Pb) and changes in DNA methylation: relevance to Alzheimer’s disease. REVIEWS ON ENVIRONMENTAL HEALTH 2019; 34:187-195. [DOI: 10.1515/reveh-2018-0076] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 05/08/2023]
Abstract
Abstract
Recent advances in neuroepigenetics have revealed its essential role in governing body function and disease. Epigenetics regulates an array of mechanisms that are susceptible to undergoing alteration by intracellular or extracellular factors. DNA methylation, one of the most extensively studied epigenetic markers is involved in the regulation of gene expression and also plays a vital role in neuronal development. The epigenome is most vulnerable during early the embryonic stage and perturbation in DNA methylation during this period can result in a latent outcome which can persist during the entire lifespan. Accumulating evidence suggests that environmental insults during the developmental phase can impart changes in the DNA methylation landscape. Based on reports on human subjects and animal models this review will explore the evidence on how developmental exposure of the known environmental pollutant, lead (Pb), can induce changes in the DNA methylation of genes which later can induce development of neurodegenerative disorders like Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Syed Waseem Bihaqi
- George and Anne Ryan Institute for Neuroscience , University of Rhode Island , Avedisian Hall, Lab: 390, 7 Greenhouse Road , Kingston, RI 02881 , USA
| |
Collapse
|
28
|
Ouyang L, Zhang W, Du G, Liu H, Xie J, Gu J, Zhang S, Zhou F, Shao L, Feng C, Fan G. Lead exposure-induced cognitive impairment through RyR-modulating intracellular calcium signaling in aged rats. Toxicology 2019; 419:55-64. [PMID: 30905827 DOI: 10.1016/j.tox.2019.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/22/2019] [Accepted: 03/19/2019] [Indexed: 11/21/2022]
Abstract
Lead is widely distributed in the environment and has become a global public health issue. It is well known that lead exposure induces not only neurodevelopmental toxicity but also neurodegenerative diseases, with learning and memory impairment in the later stage. However, the molecular mechanisms remain elusive. The present study investigated the effects of early life and lifetime lead exposure on cognition and identified the molecular mechanisms involved in aged rats. The results herein demonstrated that the lead concentration in peripheral blood and brain tissues in aged rats was significantly increased in a lead dose-dependent manner. High-dose lead exposure caused cognitive functional impairment in aged rats, concomitant with a longer escape latency and a lower frequency of crossing the platform via Morris water maze testing compared to those in the control and low-dose lead exposure groups. Importantly, neuron functional defects were still observed even in early life lead exposure during the prenatal and weaning periods in aged rats. The neurotoxicity induced by lead exposure was morphologically evidenced by a recessed nuclear membrane, a swollen endoplasmic reticulum, and mitochondria in the neurons. Mechanistically, the exposure of aged rats to lead resulted in increasing free calcium concentration, reactive oxygen species, and apoptosis in the hippocampal neurons. Lead exposure increased RyR3 expression and decreased the levels of p-CaMKIIα/CaMKIIα and p-CREB/CREB in the hippocampus of aged rats. These findings indicated that early life lead exposure-induced cognition disorder was irreversible in aged rats. Lead-induced neurotoxicity might be related to the upregulation of RyR3 expression and high levels of intracellular free calcium with increasing lead concentration in injured neurons.
Collapse
Affiliation(s)
- Lu Ouyang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China; Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China
| | - Wei Zhang
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Guihua Du
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Haizhen Liu
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Jie Xie
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Junwang Gu
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Shuyun Zhang
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Fankun Zhou
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Lijian Shao
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Chang Feng
- Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China
| | - Guangqin Fan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China; Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006, China; Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
29
|
Huat TJ, Camats-Perna J, Newcombe EA, Valmas N, Kitazawa M, Medeiros R. Metal Toxicity Links to Alzheimer's Disease and Neuroinflammation. J Mol Biol 2019; 431:1843-1868. [PMID: 30664867 DOI: 10.1016/j.jmb.2019.01.018] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/11/2022]
Abstract
As the median age of the population increases, the number of individuals with Alzheimer's disease (AD) and the associated socio-economic burden are predicted to worsen. While aging and inherent genetic predisposition play major roles in the onset of AD, lifestyle, physical fitness, medical condition, and social environment have emerged as relevant disease modifiers. These environmental risk factors can play a key role in accelerating or decelerating disease onset and progression. Among known environmental risk factors, chronic exposure to various metals has become more common among the public as the aggressive pace of anthropogenic activities releases excess amount of metals into the environment. As a result, we are exposed not only to essential metals, such as iron, copper, zinc and manganese, but also to toxic metals including lead, aluminum, and cadmium, which perturb metal homeostasis at the cellular and organismal levels. Herein, we review how these metals affect brain physiology and immunity, as well as their roles in the accumulation of toxic AD proteinaceous species (i.e., β-amyloid and tau). We also discuss studies that validate the disruption of immune-related pathways as an important mechanism of toxicity by which metals can contribute to AD. Our goal is to increase the awareness of metals as players in the onset and progression of AD.
Collapse
Affiliation(s)
- Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, CA, USA
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
30
|
Epigenetic and Neurological Impairments Associated with Early Life Exposure to Persistent Organic Pollutants. Int J Genomics 2019; 2019:2085496. [PMID: 30733955 PMCID: PMC6348822 DOI: 10.1155/2019/2085496] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/14/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022] Open
Abstract
The incidence of neurodevelopmental and neurodegenerative diseases worldwide has dramatically increased over the last decades. Although the aetiology remains uncertain, evidence is now growing that exposure to persistent organic pollutants during sensitive neurodevelopmental periods such as early life may be a strong risk factor, predisposing the individual to disease development later in life. Epidemiological studies have associated environmentally persistent organic pollutant exposure to brain disorders including neuropathies, cognitive, motor, and sensory impairments; neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD); and neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS). In many ways, this expands the classical “Developmental Origins of Health and Disease” paradigm to include exposure to pollutants. This model has been refined over the years to give the current “three-hit” model that considers the individual's genetic factors as a first “hit.” It has an immediate interaction with the early-life exposome (including persistent organic pollutants) that can be considered to be a second “hit.” Together, these first two “hits” produce a quiescent or latent phenotype, most probably encoded in the epigenome, which has become susceptible to a third environmental “hit” in later life. It is only after the third “hit” that the increased risk of disease symptoms is crystallised. However, if the individual is exposed to a different environment in later life, they would be expected to remain healthy. In this review, we examine the effect of exposure to persistent organic pollutants and particulate matters in early life and the relationship to subsequent neurodevelopmental and neurodegenerative disorders. The roles of those environmental factors which may affect epigenetic DNA methylation and therefore influence normal neurodevelopment are then evaluated.
Collapse
|
31
|
Oxidative stress in the neurodegenerative brain following lifetime exposure to lead in rats: Changes in lifespan profiles. Toxicology 2019; 411:101-109. [DOI: 10.1016/j.tox.2018.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/26/2018] [Accepted: 11/12/2018] [Indexed: 12/27/2022]
|
32
|
Fathabadi B, Dehghanifiroozabadi M, Aaseth J, Sharifzadeh G, Nakhaee S, Rajabpour-Sanati A, Amirabadizadeh A, Mehrpour O. Comparison of Blood Lead Levels in Patients With Alzheimer's Disease and Healthy People. Am J Alzheimers Dis Other Demen 2018; 33:541-547. [PMID: 30134734 PMCID: PMC10852476 DOI: 10.1177/1533317518794032] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND It is argued that breakdown of β-amyloid in the brain causes deposition of senescent plaques and therefore Alzheimer's disease (AD). One of the influential factors for increasing level of this protein is exposure to lead. Our aim was to compare blood lead levels (BLLs) between patients with AD and healthy controls. METHODS This case-control study was performed on all patients with cognitive impairment who were referred to the Neurological Clinic of Birjand in 2016 to 2017. Patients were referred to the laboratory for measurement of their serum levels of lead. The controls and patients were matched by age and sex. RESULTS In the AD case group, the average BLL was 22.22 ± 28.57 μg/dL. Mann-Whitney U test showed that BLLs were significantly higher in the patients than in the controls. The unadjusted odds ratio for BLL among the patients was 1.05 (95% confidence interval: 1.01-1.09; P = .01) compared to the controls. CONCLUSION In the present study, BLL was associated with AD.
Collapse
Affiliation(s)
- Babak Fathabadi
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Dehghanifiroozabadi
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Department of Neurology, Birjand University of Medical Sciences, Complementary Alternative Medicine Research Center, Valiasr Hospital, Birjand, Iran
| | - Jan Aaseth
- Innlandet Hospital and Inland Norway University of Applied Sciences, Elverum, Norway
| | - Gholamreza Sharifzadeh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Nakhaee
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Ali Rajabpour-Sanati
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Amirabadizadeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Rocky Mountain Poison and Drug Center, Denver, CO, USA
| |
Collapse
|
33
|
Zhou CC, Gao ZY, Wang J, Wu MQ, Hu S, Chen F, Liu JX, Pan H, Yan CH. Lead exposure induces Alzheimers's disease (AD)-like pathology and disturbes cholesterol metabolism in the young rat brain. Toxicol Lett 2018; 296:173-183. [PMID: 29908845 DOI: 10.1016/j.toxlet.2018.06.1065] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Lead exposure has been evidenced as a risk factor for Alzheimer's disease (AD), mainly affecting the ageing. However, the early manifestation and mechanisms of AD-like pathology induced by lead exposure remains to be elucidated. Considering the fact that impaired cholesterol metabolism is associated with many neurodegenerative disorders including AD, in this study we focused on the role of cholesterol metabolism in lead induced premature AD-like pathology. We treated weaning rats with lead at different concentrations for 4 weeks. We found that developmental lead exposure increased amyloid-beta (Aβ) accumulation and amyloid plaque deposition in the cortex and hippocampus. Lead exposure increased amyloid precursor protein (APP) expression and activated the sterol regulatory element binding protein 2 (SREBP2)-beta secretase (BACE1) pathway. In addition, we found that lead exposure decreased cholesterol levels by upregulating the expression of liver X receptor-a (LXR-a) and ATP-binding cassette transporter protein family member A1 (ABCA1) and decreasing the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CR) and low density lipoprotein receptor (LDL-R) in young rat brain tissues. Taken together, our data demonstrated that developmental lead exposure induced early manifestation of AD-like pathology and disturbed cholesterol metabolism in young rat brains.
Collapse
Affiliation(s)
- Can-Can Zhou
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Zhen-Yan Gao
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Ju Wang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Mei-Qin Wu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Shuang Hu
- East China University of Science and Technology, Shanghai, 200237, PR China
| | - Fei Chen
- East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jun-Xia Liu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Hui Pan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China
| | - Chong-Huai Yan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| |
Collapse
|
34
|
Maloney B, Bayon BL, Zawia NH, Lahiri DK. Latent consequences of early-life lead (Pb) exposure and the future: Addressing the Pb crisis. Neurotoxicology 2018; 68:126-132. [PMID: 29981765 PMCID: PMC9873145 DOI: 10.1016/j.neuro.2018.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/29/2018] [Accepted: 06/30/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND The lead (Pb) exposure crisis in Flint, Michigan has passed from well-publicized event to a footnote, while its biological and social impact will linger for lifetimes. Interest in the "water crisis" has dropped to pre-event levels, which is neither appropriate nor safe. Flint's exposure was severe, but it was not unique. Problematic Pb levels have also been found in schools and daycares in 42 states in the USA. The enormity of Pb exposure via municipal water systems requires multiple responses. Herein, we focus on addressing a possible answer to long-term sequelae of Pb exposure. We propose "4R's" (remediation, renovation, reallocation, and research) against the Pb crisis that goes beyond a short-term fix. Remediation for affected individuals must continue to provide clean water and deal with both short and long-term effects of Pb exposure. Renovation of current water delivery systems, at both system-wide and individual site levels, is necessary. Reallocation of resources is needed to ensure these two responses occur and to get communities ready for potential sequelae of Pb exposure. Finally, properly focused research can track exposed individuals and illuminate latent (presumably epigenetic) results of Pb exposure and inform further resource reallocation. CONCLUSION Motivation to act by not only the general public but also by scientific and medical leaders must be maintained beyond initial news cycle spikes and an annual follow-up story. Environmental impact of Pb contamination of drinking water goes beyond one exposure incident in an impoverished and forgotten Michigan city. Population effects must be addressed long-term and nationwide.
Collapse
Affiliation(s)
- Bryan Maloney
- Department of Psychiatry, USA,Indiana Alzheimer Disesae Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Baindu L. Bayon
- Medical & Molecular Genetics, USA,Indiana Alzheimer Disesae Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Debomoy K. Lahiri
- Department of Psychiatry, USA,Medical & Molecular Genetics, USA,Indiana Alzheimer Disesae Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA,Corresponding author at: Department of Psychiatry, Neuroscience Research Building, Indiana University School of Medicine, 320 West 15th St., Indianapolis IN 46202, USA. (D.K. Lahiri)
| |
Collapse
|
35
|
Kelley KD, Checkoway H, Hall DA, Reich SG, Cunningham C, Litvan I. Traumatic Brain Injury and Firearm Use and Risk of Progressive Supranuclear Palsy Among Veterans. Front Neurol 2018; 9:474. [PMID: 29973911 PMCID: PMC6020251 DOI: 10.3389/fneur.2018.00474] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/31/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Progressive supranuclear palsy (PSP) is a tauopathy that has a multifactorial etiology. Numerous studies that have investigated lead exposure and traumatic brain injury (TBI) as risk factors for other tauopathies, such as Alzheimer's disease, but not for PSP. Objective: We sought to investigate the role of firearm usage, as a possible indicator of lead exposure, and TBI as risk factors for PSP in a population of military veterans. Methods: We included participants from a larger case-control study who reported previous military service. Our sample included 67 PSP cases and 68 controls. Participants were administered a questionnaire to characterize firearm use in the military and occurrence of TBI. Results: Cases were significantly less educated than controls. In unadjusted analyses, the proportion of PSP cases (80.6%) and controls (64.7%) who reported use of firearms as part of their military job was positively associated with PSP, odds ratio (OR) 2.2 (95% CI: 1-5.0). There were no significant case-control differences in mean service duration. There was only a weak association with history of TBI, OR 1.6 (95% CI: 0.8-3.4). In multivariate models, firearm usage (OR 3.7, 95% CI: 1.5, 9.8) remained significantly associated with PSP. Conclusions: Our findings show a positive association between firearm usage and PSP and an inverse association between education and PSP. The former suggests a possible etiologic role of lead. Further studies are needed to confirm the potential etiologic effects of metals on PSP. The study was registered in clinicaltrials.gov. ClinicalTrials.gov Identifier: NCT00431301.
Collapse
Affiliation(s)
- Kristen D. Kelley
- School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Harvey Checkoway
- Department of Family Medicine & Public Health, University of California, San Diego, La Jolla, CA, United States
| | - Deborah A. Hall
- Department of Neurology, Rush University Medical Center, Chicago, IL, United States
| | - Stephen G. Reich
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Chris Cunningham
- Clinical Trials Unit, University of Louisville, Louisville, KY, United States
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
36
|
Eid A, Bihaqi SW, Hemme C, Gaspar JM, Hart RP, Zawia NH. Histone acetylation maps in aged mice developmentally exposed to lead: epigenetic drift and Alzheimer-related genes. Epigenomics 2018; 10:573-583. [PMID: 29722544 DOI: 10.2217/epi-2017-0143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AIM Early life exposure to lead (Pb) has been shown to increase late life biomarkers involved in Alzheimer's disease (AD) pathology. Here, we tested the hypothesis that latent over expression of AD-related genes may be regulated through histone activation pathways. METHODS Chromatin immunoprecipitation sequencing was used to map the histone activation mark (H3K9Ac) to the mouse genome in developmentally Pb exposed mice on postnatal days 20, 270 and 700. RESULTS Exposure to Pb resulted in a global downregulation of H3K9Ac across the lifespan; except in genes associated with the Alzheimer pathway. DISCUSSION Early life exposure to Pb results in an epigenetic drift in H3K9Ac consistent with latent global gene repression. Alzheimer-related genes do not follow this trend.
Collapse
Affiliation(s)
- Aseel Eid
- Interdisciplinary Neurosciences Program, University of Rhode Island, Kingston, RI 02881, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| | - Syed Waseem Bihaqi
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| | - Christopher Hemme
- Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - John M Gaspar
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Ronald P Hart
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Nasser H Zawia
- Interdisciplinary Neurosciences Program, University of Rhode Island, Kingston, RI 02881, USA.,Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
37
|
Kim AC, Lim S, Kim YK. Metal Ion Effects on Aβ and Tau Aggregation. Int J Mol Sci 2018; 19:E128. [PMID: 29301328 PMCID: PMC5796077 DOI: 10.3390/ijms19010128] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/24/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023] Open
Abstract
Amyloid and tau aggregation are implicated in manifold neurodegenerative diseases and serve as two signature pathological hallmarks in Alzheimer's disease (AD). Though aging is considered as a prominent risk factor for AD pathogenesis, substantial evidence suggests that an imbalance of essential biometal ions in the body and exposure to certain metal ions in the environment can potentially induce alterations to AD pathology. Despite their physiological importance in various intracellular processes, biometal ions, when present in excessive or deficient amounts, can serve as a mediating factor for neurotoxicity. Recent studies have also demonstrated the contribution of metal ions found in the environment on mediating AD pathogenesis. In this regard, the neuropathological features associated with biometal ion dyshomeostasis and environmental metal ion exposure have prompted widespread interest by multiple research groups. In this review, we discuss and elaborate on findings from previous studies detailing the possible role of both endogenous and exogenous metal ions specifically on amyloid and tau pathology in AD.
Collapse
Affiliation(s)
- Anne Claire Kim
- Department of Neuroscience, Wellesley College, Wellesley, MA 02481, USA.
- Brain Science Institute, Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| | - Sungsu Lim
- Brain Science Institute, Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| | - Yun Kyung Kim
- Brain Science Institute, Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| |
Collapse
|
38
|
Wright K, Bihaqi SW, Lahouel A, Masoud A, Mushtaq F, Leso A, Eid A, Zawia NH. Importance of tau in cognitive decline as revealed by developmental exposure to lead. Toxicol Lett 2017; 284:63-69. [PMID: 29203278 DOI: 10.1016/j.toxlet.2017.11.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 12/21/2022]
Abstract
Previous reports by us have determined that developmental exposure to the heavy metal lead (Pb) resulted in cognitive impairment in aging wildtype mice, and a latent induction in biomarkers associated with both the tau and amyloid pathways. However, the relationship between these two pathways and their correlation to cognitive performance needs to be scrutinized. Here, we investigated the impact of developmental Pb (0.2%) exposure on the amyloid and tau pathways in a transgenic mouse model lacking the tau gene. Cognitive function, and levels of intermediates in the amyloid and tau pathways following postnatal Pb exposure were assessed on young adult and mature transgenic mice. No significant difference in behavioral performance, amyloid precursor protein (APP), or amyloid beta (Aβ) levels was observed in transgenic mice exposed to Pb. Regulators of the tau pathway were impacted by the absence of tau, but no additional change was imparted by Pb exposure. These results revealed that developmental Pb exposure does not cause cognitive decline or change the expression of the amyloid pathway in the absence of tau. The essentiality of tau to mediate cognitive decline by environmental perturbations needs further investigation.
Collapse
Affiliation(s)
- K Wright
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - S W Bihaqi
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Lahouel
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Masoud
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Biochemical Technology Program, Faculty of Applied Science, Thamar University, Yemen
| | - F Mushtaq
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Leso
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Eid
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - N H Zawia
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston Rhode Island, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA.
| |
Collapse
|
39
|
Wallin C, Sholts SB, Österlund N, Luo J, Jarvet J, Roos PM, Ilag L, Gräslund A, Wärmländer SKTS. Alzheimer's disease and cigarette smoke components: effects of nicotine, PAHs, and Cd(II), Cr(III), Pb(II), Pb(IV) ions on amyloid-β peptide aggregation. Sci Rep 2017; 7:14423. [PMID: 29089568 PMCID: PMC5663743 DOI: 10.1038/s41598-017-13759-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022] Open
Abstract
Cigarette smoking is a significant risk factor for Alzheimer's disease (AD), which is associated with extracellular brain deposits of amyloid plaques containing aggregated amyloid-β (Aβ) peptides. Aβ aggregation occurs via multiple pathways that can be influenced by various compounds. Here, we used AFM imaging and NMR, fluorescence, and mass spectrometry to monitor in vitro how Aβ aggregation is affected by the cigarette-related compounds nicotine, polycyclic aromatic hydrocarbons (PAHs) with one to five aromatic rings, and the metal ions Cd(II), Cr(III), Pb(II), and Pb(IV). All PAHs and metal ions modulated the Aβ aggregation process. Cd(II), Cr(III), and Pb(II) ions displayed general electrostatic interactions with Aβ, whereas Pb(IV) ions showed specific transient binding coordination to the N-terminal Aβ segment. Thus, Pb(IV) ions are especially prone to interact with Aβ and affect its aggregation. While Pb(IV) ions affected mainly Aβ dimer and trimer formation, hydrophobic toluene mainly affected formation of larger aggregates such as tetramers. The uncharged and hydrophilic nicotine molecule showed no direct interactions with Aβ, nor did it affect Aβ aggregation. Our Aβ interaction results suggest a molecular rationale for the higher AD prevalence among smokers, and indicate that certain forms of lead in particular may constitute an environmental risk factor for AD.
Collapse
Affiliation(s)
- Cecilia Wallin
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Sabrina B Sholts
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Nicklas Österlund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
- Department of Environmental Science and Analytical Chemistry, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jinghui Luo
- Chemical Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford Ox, 1 3TA, UK
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, Nobels väg 13, 171 77, Stockholm, Sweden
- Department of Clinical Physiology, Capio St.Göran Hospital, St.Göransplan 1, 112 19, Stockholm, Sweden
| | - Leopold Ilag
- Department of Environmental Science and Analytical Chemistry, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Sebastian K T S Wärmländer
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden.
| |
Collapse
|
40
|
Engstrom AK, Xia Z. Lead exposure in late adolescence through adulthood impairs short-term spatial memory and the neuronal differentiation of adult-born cells in C57BL/6 male mice. Neurosci Lett 2017; 661:108-113. [PMID: 28970130 DOI: 10.1016/j.neulet.2017.09.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/06/2017] [Accepted: 09/28/2017] [Indexed: 11/27/2022]
Abstract
Lead is a neurotoxicant of immense public health importance. Epidemiology studies suggest that heavy metal exposure may be associated with an increased risk of cognitive decline, yet few studies to date have assessed the effect of adult lead exposure on cognitive behavior in animal models. Here, we exposed 6-week-old male C57BL/6 mice to 0.2% lead acetate via drinking water for 12 weeks starting at 6 weeks of age and then assessed for deficits in hippocampus-dependent spatial memory and impairment of adult hippocampal neurogenesis. Lead did not cause locomotor deficits or anxiety in the open field test. However, we found that adult, subchronic lead exposure was sufficient to cause deficits in spatial short-term memory and these deficits persisted through at least 2 months post-lead exposure. Furthermore, we observed that lead-treated mice had fewer adult-born, mature neurons in the dentate gyrus of the hippocampus compared to control animals, suggesting that lead exposure during adolescence and adulthood may impair the neuronal differentiation of adult-born cells. These data suggest that adult lead exposure is sufficient to cause persistent deficits in spatial short-term memory and impair key processes in adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Anna K Engstrom
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA.
| | - Zhengui Xia
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
41
|
Glycogen metabolism in brain and neurons - astrocytes metabolic cooperation can be altered by pre- and neonatal lead (Pb) exposure. Toxicology 2017; 390:146-158. [PMID: 28916327 DOI: 10.1016/j.tox.2017.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 11/22/2022]
Abstract
Lead (Pb) is an environmental neurotoxin which particularly affects the developing brain but the molecular mechanism of its neurotoxicity still needs clarification. The aim of this paper was to examine whether pre- and neonatal exposure to Pb (concentration of Pb in rat offspring blood below the "threshold level") may affect the brain's energy metabolism in neurons and astrocytes via the amount of available glycogen. We investigated the glycogen concentration in the brain, as well as the expression of the key enzymes involved in glycogen metabolism in brain: glycogen synthase 1 (Gys1), glycogen phosphorylase (PYGM, an isoform active in astrocytes; and PYGB, an isoform active in neurons) and phosphorylase kinase β (PHKB). Moreover, the expression of connexin 43 (Cx43) was evaluated to analyze whether Pb poisoning during the early phase of life may affect the neuron-astrocytes' metabolic cooperation. This work shows for the first time that exposure to Pb in early life can impair brain energy metabolism by reducing the amount of glycogen and decreasing the rate of its metabolism. This reduction in brain glycogen level was accompanied by a decrease in Gys1 expression. We noted a reduction in the immunoreactivity and the gene expression of both PYGB and PYGM isoform, as well as an increase in the expression of PHKB in Pb-treated rats. Moreover, exposure to Pb induced decrease in connexin 43 immunoexpression in all the brain structures analyzed, both in astrocytes as well as in neurons. Our data suggests that exposure to Pb in the pre- and neonatal periods results in a decrease in the level of brain glycogen and a reduction in the rate of its metabolism, thereby reducing glucose availability, which as a further consequence may lead to the impairment of brain energy metabolism and the metabolic cooperation between neurons and astrocytes.
Collapse
|
42
|
Bihaqi SW, Eid A, Zawia NH. Lead exposure and tau hyperphosphorylation: An in vitro study. Neurotoxicology 2017; 62:218-223. [DOI: 10.1016/j.neuro.2017.07.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/20/2017] [Accepted: 07/22/2017] [Indexed: 10/19/2022]
|
43
|
Modgil S, Cameotra SS, Sharma VL, Anand A. Early Life Pb Exposure and its Effect on Later Life Retinal Degeneration. J Cell Biochem 2017; 118:3213-3224. [DOI: 10.1002/jcb.25968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/01/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Shweta Modgil
- Neuroscience Research LabDepartment of NeurologyPost Graduate Institute of Medical Education and ResearchChandigarhIndia
- Department of ZoologyPanjab UniversityChandigarhIndia
| | | | | | - Akshay Anand
- Neuroscience Research LabDepartment of NeurologyPost Graduate Institute of Medical Education and ResearchChandigarhIndia
| |
Collapse
|
44
|
Ferreira MCDF, Zucki F, Duarte JL, Iano FG, Ximenes VF, Buzalaf MAR, Oliveira RCD. Influence of iron on modulation of the antioxidant system in rat brains exposed to lead. ENVIRONMENTAL TOXICOLOGY 2017; 32:813-822. [PMID: 27170105 DOI: 10.1002/tox.22281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 06/05/2023]
Abstract
The objective of this study was to evaluate markers of oxidative stress in the brains of rats exposed to lead acetate (Pb(C2 H3 O2 )2 ), either associated or not associated with ferrous sulfate (FeSO4 ). A total of 36 weaning rats (Rattus norvegicus) were divided into 6 groups of six animals and exposed to lead acetate for six weeks. In the control group (control), the animals received deionized water. The Pb260 and Pb260 + Fe received 260 µM lead acetate, and the Pb1050 and Pb1050 + Fe received 1050 µM lead acetate. The Pb260 + Fe and Pb1050 + Fe were supplemented with 20 mg of ferrous sulfate/Kg body weight every 2 days. Group Fe received deionized water and ferrous sulfate. The rat brains were collected to analyze the enzymatic activity of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and the concentration of reduced glutathione (GSH), lipid peroxidation (TBARS), and total antioxidant substance (TAS) (DPPH• technique). The activity of SOD and GPx in the experimental groups decreased compared to the control, together with the concentration of GSH (p < 0.05). For CAT analysis, SOD tended to increase in concentration in the experimental groups without a concomitant exposure to FeSO4 , whereas GPx showed a slight tendency to increase in activity compared to the control. For TAS-DPPH• , there was a decrease in the experimental groups (p < 0.05). According to the results, SOD, GPx, and GSH were affected by lead acetate and exposure to ferrous sulfate changed this dynamic. However, further studies are needed to verify whether ferrous sulfate acts as a protectant against the toxic effects of lead. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 813-822, 2017.
Collapse
Affiliation(s)
- Maria Cecília de Freitas Ferreira
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Fernanda Zucki
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Josilene Luciene Duarte
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Flávia Godoy Iano
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Valdecir Farias Ximenes
- Department of Chemistry, Faculty of Sciences, São Paulo State University, Av. Engenheiro Luiz Edmundo Carrijo Coube, 1401 Bauru-SP 17033-360, Brazil
| | - Marília Afonso Rabelo Buzalaf
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| | - Rodrigo Cardoso de Oliveira
- Department of Biological Sciences/Biochemistry, Bauru School of Dentistry - University of São Paulo. Alameda Dr. Octavio Pinheiro Brisolla, 9-75 Bauru-SP, 17012-901, Brazil
| |
Collapse
|
45
|
|
46
|
Eid A, Zawia N. Consequences of lead exposure, and it’s emerging role as an epigenetic modifier in the aging brain. Neurotoxicology 2016; 56:254-261. [DOI: 10.1016/j.neuro.2016.04.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/14/2022]
|
47
|
Bellinger DC, Matthews-Bellinger JA, Kordas K. A developmental perspective on early-life exposure to neurotoxicants. ENVIRONMENT INTERNATIONAL 2016; 94:103-112. [PMID: 27235688 DOI: 10.1016/j.envint.2016.05.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/14/2016] [Accepted: 05/14/2016] [Indexed: 05/24/2023]
Abstract
BACKGROUND Studies of early-life neurotoxicant exposure have not been designed, analyzed, or interpreted in the context of a fully developmental perspective. OBJECTIVES The goal of this paper is to describe the key principles of a developmental perspective and to use examples from the literature to illustrate the relevance of these principles to early-life neurotoxicant exposures. METHODS Four principles are discussed: 1) the effects of early-life neurotoxicant exposure depend on a child's developmental context; 2) deficits caused by early-life exposure initiate developmental cascades that can lead to pathologies that differ from those observed initially; 3) early-life neurotoxicant exposure has intra-familial and intergenerational impacts; 4) the impacts of early-life neurotoxicant exposure influence a child's ability to respond to future insults. The first principle is supported by considerable evidence, but the other three have received much less attention. DISCUSSION Incorporating a developmental perspective in studies of early-life neurotoxicant exposures requires prospective collection of data on a larger array of covariates than usually considered, using analytical approaches that acknowledge the transactional processes between a child and the environment and the phenomenon of developmental cascades. CONCLUSION Consideration of early-life neurotoxicant exposure within a developmental perspective reveals that many issues remain to be explicated if we are to achieve a deep understanding of the societal health burden associated with early-life neurotoxicant exposures.
Collapse
Affiliation(s)
- David C Bellinger
- Department of Neurology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02112, USA; Department of Psychiatry, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - Julia A Matthews-Bellinger
- Department of Psychiatry, University of Massachusetts Medical School, Boston Psychoanalytic Society and Institute, 19 Fair Oaks Park, Needham, MA 02492, USA.
| | - Katarzyna Kordas
- Department of Epidemiology and Environmental Health, University at Buffalo, 270 Farber Hall, Buffalo, NY 14214, USA.
| |
Collapse
|
48
|
Developmental exposure to lead (Pb) alters the expression of the human tau gene and its products in a transgenic animal model. Neurotoxicology 2016; 55:154-159. [PMID: 27293183 DOI: 10.1016/j.neuro.2016.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/11/2016] [Accepted: 06/01/2016] [Indexed: 10/21/2022]
Abstract
Tauopathies are a class of neurodegenerative diseases associated with the pathological aggregation of the tau protein in the human brain. The best known of these illnesses is Alzheimer's disease (AD); a disease where the microtubule associated protein tau (MAPT) becomes hyperphosphorylated (lowering its binding affinity to microtubules) and aggregates within neurons in the form of neurofibrillary tangles (NFTs). In this paper we examine whether environmental factors play a significant role in tau pathogenesis. Our studies were conducted in a double mutant mouse model that expressed the human tau gene and lacked the gene for murine tau. The human tau mouse model was tested for the transgene's ability to respond to an environmental toxicant. Pups were developmentally exposed to lead (Pb) from postnatal day (PND) 1-20 with 0.2% Pb acetate. Mice were then sacrificed at PND 20, 30, 40 and 60. Protein and mRNA levels for tau and CDK5 as well as tau phosphorylation at Ser396 were determined. In addition, the potential role of miRNA in tau expression was investigated by measuring levels of miR-34c, a miRNA that targets the mRNA for human tau, at PND20 and 50. The expression of the human tau transgene was altered by developmental exposure to Pb. This exposure also altered the expression of miR-34c. Our findings are the first of their kind to test the responsiveness of the human tau gene to an environmental toxicant and to examine an epigenetic mechanism that may be involved in the regulation of this gene's expression.
Collapse
|
49
|
Masoud AM, Bihaqi SW, Machan JT, Zawia NH, Renehan WE. Early-Life Exposure to Lead (Pb) Alters the Expression of microRNA that Target Proteins Associated with Alzheimer’s Disease. J Alzheimers Dis 2016; 51:1257-64. [DOI: 10.3233/jad-151018] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Anwar M. Masoud
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Biochemical Technology Program, Faculty of Applied Science, Thamar University, Thamar, Yemen
| | - Syed W. Bihaqi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Jason T. Machan
- Lifespan Biostatistics Core and Departments of Orthopaedics and Surgery, Warren Alpert Medical School, Brown University, Providence RI, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - William E. Renehan
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
50
|
Perinatal exposure to lead (Pb) promotes Tau phosphorylation in the rat brain in a GSK-3β and CDK5 dependent manner: Relevance to neurological disorders. Toxicology 2016; 347-349:17-28. [PMID: 27012722 DOI: 10.1016/j.tox.2016.03.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/11/2016] [Accepted: 03/20/2016] [Indexed: 12/24/2022]
Abstract
Hyperphosphorylation of Tau is involved in the pathomechanism of neurological disorders such as Alzheimer's, Parkinson's diseases as well as Autism. Epidemiological data suggest the significance of early life exposure to lead (Pb) in etiology of disorders affecting brain function. However, the precise mechanisms by which Pb exerts neurotoxic effects are not fully elucidated. The purpose of this study was to evaluate the effect of perinatal exposure to low dose of Pb on the Tau pathology in the developing rat brain. Furthermore, the involvement of two major Tau-kinases: glycogen synthase kinase-3 beta (GSK-3β) and cyclin-dependent kinase 5 (CDK5) in Pb-induced Tau modification was evaluated. Pregnant female rats were divided into control and Pb-treated group. The control animals were maintained on drinking water while females from the Pb-treated group received 0.1% lead acetate (PbAc) in drinking water, starting from the first day of gestation until weaning of the offspring. During the feeding of pups, mothers from the Pb-treated group were still receiving PbAc. Pups of both groups were weaned at postnatal day 21 and then until postnatal day 28 received only drinking water. 28-day old pups were sacrificed and Tau mRNA and protein level as well as Tau phosphorylation were analyzed in forebrain cortex (FC), cerebellum (C) and hippocampus (H). Concomitantly, we examined the effect of Pb exposure on GSK-3β and CDK5 activation. Our data revealed that pre- and neonatal exposure to Pb (concentration of Pb in whole blood below 10μg/dL, considered safe for humans) caused significant increase in the phosphorylation of Tau at Ser396 and Ser199/202 with parallel rise in the level of total Tau protein in FC and C. Tau hyperphosphorylation in Pb-treated animals was accompanied by elevated activity of GSK-3β and CDK5. Western blot analysis revealed activation of GSK-3β in FC and C as well as CDK5 in C, via increased phosphorylation of Tyr-216 and calpain-dependent p25 formation, respectively. In conclusion, perinatal exposure to Pb up-regulates Tau protein level and induces Tau hyperphosphorylation in the rat brain cortex and cerebellum. We suggest that neurotoxic effect of Pb might be mediated, at least in part, by GSK-3β and CDK5-dependent Tau hyperphosphorylation, which may lead to the impairment of cytoskeleton stability and neuronal dysfunction.
Collapse
|