1
|
Dietary Selenomethionine Reduce Mercury Tissue Levels and Modulate Methylmercury Induced Proteomic and Transcriptomic Alterations in Hippocampi of Adolescent BALB/c Mice. Int J Mol Sci 2022; 23:ijms232012242. [PMID: 36293098 PMCID: PMC9603801 DOI: 10.3390/ijms232012242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
Methylmercury (MeHg) is a well-known environmental contaminant, particularly harmful to the developing brain. The main human dietary exposure to MeHg occurs through seafood consumption. However, seafood also contains several nutrients, including selenium, which has been shown to interact with MeHg and potentially ameliorate its toxicity. The aim of this study was to investigate the combined effects of selenium (as selenomethionine; SeMet) and MeHg on mercury accumulation in tissues and the effects concomitant dietary exposure of these compounds exert on the hippocampal proteome and transcriptome in mice. Adolescent male BALB/c mice were exposed to SeMet and two different doses of MeHg through their diet for 11 weeks. Organs, including the brain, were sampled for mercury analyses. Hippocampi were collected and analyzed using proteomics and transcriptomics followed by multi-omics bioinformatics data analysis. The dietary presence of SeMet reduced the amount of mercury in several organs, including the brain. Proteomic and RNA-seq analyses showed that both protein and RNA expression patterns were inversely regulated in mice receiving SeMet together with MeHg compared to MeHg alone. Several pathways, proteins and RNA transcripts involved in conditions such as immune responses and inflammation, oxidative stress, cell plasticity and Alzheimer’s disease were affected inversely by SeMet and MeHg, indicating that SeMet can ameliorate several toxic effects of MeHg in mice.
Collapse
|
2
|
Rodríguez-Viso P, Domene A, Vélez D, Devesa V, Monedero V, Zúñiga M. Mercury toxic effects on the intestinal mucosa assayed on a bicameral in vitro model: Possible role of inflammatory response and oxidative stress. Food Chem Toxicol 2022; 166:113224. [PMID: 35700822 DOI: 10.1016/j.fct.2022.113224] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022]
Abstract
Exposure to mercury (Hg) mostly occurs through diet, where it is mainly found as inorganic Hg [Hg(II)] or methylmercury (MeHg). In vivo studies have linked its exposure with neurological and renal diseases, however, its toxic effects upon the gastrointestinal tract are largely unknown. In order to evaluate the effect of Hg on intestinal mucosa, a bicameral system was employed with co-cultures of Caco-2 and HT29-MTX intestinal epithelial cells and THP-1 macrophages. Cells were exposed to Hg(II) and MeHg (0.1, 0.5, 1 mg/L) during 11 days. The results evidenced a greater pro-inflammatory response in cells exposed to Hg with increments of IL-8 (15-126%) and IL-1β release (39-63%), mainly induced by macrophages which switched to a M1 phenotype. A pro-oxidant response was also observed in both cell types with an increase in ROS/RNS levels (44-140%) and stress proteins expression. Intestinal cells treated with Hg displayed structural abnormalities, hypersecretion of mucus and defective tight junctions. An increased paracellular permeability (123-170%) at the highest concentrations of Hg(II) and MeHg and decreased capacity to restore injuries in the cell monolayer were also observed. All these toxic effects were governed by various inflammatory signalling pathways (p38 MAPK, JNK and NF-κB).
Collapse
Affiliation(s)
- Pilar Rodríguez-Viso
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | - Adrián Domene
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | - Dinoraz Vélez
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | - Vicenta Devesa
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | - Vicente Monedero
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| | - Manuel Zúñiga
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Agustín Escardino 7, 46980, Paterna, Valencia, Spain.
| |
Collapse
|
3
|
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: 15] [Impact Index Per Article: 7.5] [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
|
4
|
Determination of stability constants of mercury(II) by garlic organosulfur ligands with differential pulse voltammetry. J APPL ELECTROCHEM 2021. [DOI: 10.1007/s10800-021-01577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
5
|
Azar J, Yousef MH, El-Fawal HAN, Abdelnaser A. Mercury and Alzheimer's disease: a look at the links and evidence. Metab Brain Dis 2021; 36:361-374. [PMID: 33411216 DOI: 10.1007/s11011-020-00649-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/20/2020] [Indexed: 12/01/2022]
Abstract
This review paper investigates a specific environmental-disease interaction between mercury exposure and Alzheimer's disease hallmarks. Alzheimer's disease is a neurodegenerative disorder affecting predominantly the memory of the affected individual. It prevails mostly in the elderly, rendering many factors as possible causative agents, which potentially contribute to the disease pathogenicity cumulatively. Alzheimer's disease affects nearly 50 million people worldwide and is considered one the most devastating diseases not only for the patient, but also for their families and caregivers. Mercury is a common environmental toxin, found in the atmosphere mostly due to human activity, such as coal burning for heating and cooking. Natural release of mercury into the atmosphere occurs by volcanic eruptions, in the form of vapor, or weathering rocks. The most toxic form of mercury to humans is methylmercury, to which humans are exposed to by ingestion of fish. Methylmercury was found to exert its toxic effects on different parts of the human body, with predominance on the brain. There is no safe concentration for mercury in the atmosphere, even trace amounts can elicit harm to humans in the long term. Mercury's effect on Alzheimer's disease hallmarks formation, extracellular senile plaques and intracellular neurofibrillary tangles, has been widely studied. This review demonstrates the involvement of mercury, in its different forms, in the pathway of amyloid beta deposition and tau tangles formation. It aims to understand the link between mercury exposure and Alzheimer's disease so that, in the future, prevention strategies can be applied to halt the progression of this disease.
Collapse
Affiliation(s)
- Jihan Azar
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt
| | - Mohamed H Yousef
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| | - Hassan A N El-Fawal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt.
| |
Collapse
|
6
|
S-methyl-L-cysteine Protects against Antimycin A-induced Mitochondrial Dysfunction in Neural Cells via Mimicking Endogenous Methionine-centered Redox Cycle. Curr Med Sci 2020; 40:422-433. [PMID: 32681247 DOI: 10.1007/s11596-020-2196-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/30/2020] [Indexed: 12/26/2022]
Abstract
Mitochondrial superoxide overproduction is believed to be responsible for the neurotoxicity associated with neurodegeneration. Mitochondria-targeted antioxidants, such as MitoQ, have emerged as potentially effective antioxidant therapies. Methionine sulfoxide reductase A (MsrA) is a key mitochondrial-localized endogenous antioxidative enzyme and it can scavenge oxidizing species by catalyzing the methionine (Met)-centered redox cycle (MCRC). In this study, we observed that the natural L-Met acted as a good scavenger for antimycin A-induced mitochondrial superoxide overproduction in PC12 cells. This antioxidation was largely dependent on the Met oxidase activity of MsrA. S-methyl-L-cysteine (SMLC), a natural analogue of Met that is abundantly found in garlic and cabbage, could activate the Met oxidase activity of MsrA to scavenge free radicals. Furthermore, SMLC protected against antimycin A-induced mitochondrial membrane depolarization and alleviated 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity. Thus, our data highlighted the possibility for SMLC supplement in the detoxication of mitochondrial damage by activating the Met oxidase activity of MsrA.
Collapse
|
7
|
Wang X, Zhang R, Lin Y, Shi P. Inhibition of NF-κB might enhance the protective role of roflupram on SH-SY5Y cells under amyloid β stimulation via PI3K/AKT/mTOR signaling pathway. Int J Neurosci 2020; 131:864-874. [PMID: 32314929 DOI: 10.1080/00207454.2020.1759588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disease and mostly endanger the health of people older than 65 years. Accumulation of beta amyloid protein (Aβ) is the main characteristic of AD. Roflupram (ROF) could improve the behavior of AD in a mouse model. In this study, we first detected the increased concentration of molecules related to inflammatory response in serum sample of patients with AD. Next, a cell model of nuclear factor kappa B (NF-κB) inhibition and NF-κB overexpression was established in SH-SY5Y cells, Aβ was used to simulate the toxicity to cells. ROF treatment decreased expression of apoptosis-related molecules via inhibition of PI3K/AKT/mTOR signaling pathway, decreased expression of pro-inflammatory factors, and increased expression of key enzymes in the tricarboxylic acid (TCA) cycle was observed in SH-SY5Y cells after ROF treatment. Inhibition of NF-κB could enlarge these trends whereas overexpression of NF-κB could reduce these trends.
Collapse
Affiliation(s)
- Xinqiang Wang
- Neurology Department, Liaocheng Second People's Hospital, Liaocheng, China.,Neurology Department, The Second Hospital of Affiliated to Shandong First Medical University,Shandong, China
| | - Rui Zhang
- Neurology Department, Liaocheng People's Hospital, Liaocheng, China
| | - Yongquan Lin
- Emergency Department, Yidu Central Hospital of Weifang, Weifang, China
| | - Peng Shi
- No. 2 Department of Neurology, Yan Tai Yeda Hospital, Yantai, China
| |
Collapse
|
8
|
Maniero MÁ, Wuilloud RG, Callegari EA, Smichowski PN, Fanelli MA. Metalloproteomics analysis in human mammary cell lines treated with inorganic mercury. J Trace Elem Med Biol 2020; 58:126441. [PMID: 31812871 PMCID: PMC8061084 DOI: 10.1016/j.jtemb.2019.126441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 11/23/2022]
Abstract
The interest in inorganic Hg toxicity and carcinogenicity has been pointed to target organs such as kidney, brain or placenta, but only a few studies have focused on the mammary gland. In this work, analytical combination techniques (SDS-PAGE followed by CV-AFS, and nanoUPLC-ESI-MS/MS) were used to determine proteins that could bind Hg in three human mammary cell lines. Two of them were tumorigenic (MCF-7 and MDA-MB-231) and the other one was the non-tumorigenic cell line (MCF-10A). There are no studies that provide this kind of information in breast cell lines with IHg treatment. Previously, we described the viability, uptake and the subcellular distribution of Hg in human breast cells and analysis of RNA-seq about the genes that encode proteins which are related to cytotoxicity of Hg. This work provides important protein candidates for further studies of Hg toxicity in the mammary gland, thus expanding our understanding of how environmental contaminants might affect tumor progression and contribute with future therapeutic methods.
Collapse
Affiliation(s)
- Mariángeles Ávila Maniero
- Laboratorio de Química Analítica para Investigación y Desarrollo (QUIANID), Instituto Interdisciplinario de Ciencias Básicas, Universidad Nacional de Cuyo, CONICET, Facultad de Ciencias Exactas y Naturales, Padre J. Contreras 1300, 5500, Mendoza, Argentina; Facultad de Farmacia y Bioquímica, Universidad Juan Agustín Maza, Lateral Sur del Acceso Este 2245, M5519, Guaymallén, Mendoza, Argentina
| | - Rodolfo G Wuilloud
- Laboratorio de Química Analítica para Investigación y Desarrollo (QUIANID), Instituto Interdisciplinario de Ciencias Básicas, Universidad Nacional de Cuyo, CONICET, Facultad de Ciencias Exactas y Naturales, Padre J. Contreras 1300, 5500, Mendoza, Argentina.
| | - Eduardo A Callegari
- BRIN-USDS SOM Proteomics Facility, University of South Dakota, 414 E Clark St, Vermillion, SD, 57069, USA
| | - Patricia N Smichowski
- Comisión Nacional de Energía Atómica, Gerencia Química, CONICET, Av. Gral. Paz 1499, B1650 Villa Maipú, Buenos Aires, Argentina
| | - Mariel A Fanelli
- Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU-CONICET), Av. Dr. Adrian Ruiz Leal, Mendoza, Argentina
| |
Collapse
|
9
|
Ehnert-Russo SL, Gelsleichter J. Mercury Accumulation and Effects in the Brain of the Atlantic Sharpnose Shark (Rhizoprionodon terraenovae). ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2020; 78:267-283. [PMID: 31760438 DOI: 10.1007/s00244-019-00691-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
Few published studies have examined whether the elevated concentrations of the nonessential toxic metal mercury (Hg) often observed in shark muscle also occur in the shark brain or whether Hg accumulation affects shark neurophysiology. Therefore, this study examined accumulation and distribution of Hg in the shark brain, as well as effects of Hg on oxidative stress in the shark central nervous system, with particular focus on the Atlantic sharpnose shark (Rhizoprionodon terraenovae). Sharks were collected along the southeastern U.S. coast throughout most of this species' U.S. geographical range. Total Hg (THg) concentrations were measured in and compared between shark muscle and brain, whereas known biomarkers of Hg-induced neurological effects, including glutathione depletion, lipid peroxidation, and concentrations of a protein marker of glial cell damage (S100b), were measured in shark cerebrospinal fluid. Brain THg concentrations were correlated with muscle THg levels but were significantly lower and did not exceed most published thresholds for neurological effects, suggesting limited potential for detrimental responses. Biomarker concentrations supported this premise, because these data were not correlated with brain THg levels. Hg speciation also was examined. Unlike muscle, methylmercury (MeHg) did not comprise a high percentage of THg in the brain, suggesting that differential uptake or loss of organic and inorganic Hg and/or demethylation of MeHg may occur in this organ. Although Hg accumulation in the shark brain generally fell below toxicity thresholds, higher THg levels were measured in the shark forebrain compared with the midbrain and hindbrain. Therefore, there is potential for selective effects on certain aspects of shark neurophysiology if brain Hg accumulation is increased.
Collapse
Affiliation(s)
- S L Ehnert-Russo
- University of North Florida, 1 UNF Dr, Jacksonville, FL, 32224, USA
| | - J Gelsleichter
- University of North Florida, 1 UNF Dr, Jacksonville, FL, 32224, USA.
| |
Collapse
|
10
|
Li W, Wang WX. In vivo oral bioavailability of fish mercury and comparison with in vitro bioaccessibility. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 683:648-658. [PMID: 31150885 DOI: 10.1016/j.scitotenv.2019.05.290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/17/2019] [Accepted: 05/20/2019] [Indexed: 06/09/2023]
Abstract
Fish consumption is considered to be a major human exposure route for mercury (Hg), but assessing the actual Hg bioavailability from consumed fish is challenging. In this study, we conducted both in vivo bioavailability (using a mouse model) and in vitro bioaccessibility (using various gastrointestinal digestion methods) assessments of Hg from consumed fish. Lyophilized fish muscles which already absorbed Hg through natural incorporation were introduced to mice by active feeding. Assimilation efficiency (AE) was measured as a short-term kinetic parameter, while a 7-day accumulation of Hg in mice blood, liver and kidney was determined. The AEs of Hg in mice ranged between 82 and 96% and showed a positive relationship with MeHg in fish independent of the fish species. For long-term bioavailability tests in which the Hg retention in organs was measured after a 7-day exposure, most Hg was found to be accumulated in liver and kidney, resulting in a strong correlation between Hg dosage and accumulation in mice organs. The long-term absolute bioavailability of mice was comparable between the liver and kidney, but much lower in the blood. The calculated absolute total Hg bioavailability ranged between 38% and 99% and decreased as the Hg dosage increased. Results of bioaccessibility tests varied considerably among different methods, illustrating that there were limitations for the in vitro bioaccessibility assay to predict the digestive dynamics of Hg in mammalian gastrointestinal tract. Our study strongly demonstrated the expediency of direct determination of Hg bioavailability, but more bioaccessibility assessments should be explored and optimized as an alternative to traditional animal experimentation.
Collapse
Affiliation(s)
- Wanze Li
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen 518057, Department of Ocean Science, Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Wen-Xiong Wang
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen 518057, Department of Ocean Science, Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
11
|
Takanezawa Y, Nakamura R, Matsuda H, Yagi T, Egawa Z, Sone Y, Uraguchi S, Adachi T, Kiyono M. Intracellular Demethylation of Methylmercury to Inorganic Mercury by Organomercurial Lyase (MerB) Strengthens Cytotoxicity. Toxicol Sci 2019; 170:438-451. [DOI: 10.1093/toxsci/kfz094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Abstract
Some methylmercury (MeHg) is converted to inorganic mercury (Hg2+) after incorporation into human and animal tissues, where it can remain for a long time. To determine the overall toxicity of MeHg in tissues, studies should evaluate low concentrations of Hg2+. Although demethylation is involved, the participating enzymes or underlying mechanisms are unknown; in addition, the low cell membrane permeability of Hg2+ makes these analyses challenging. We established model cell lines to assess toxicities of low concentrations of Hg2+ using bacterial organomercury lyase (MerB). We engineered MerB-expressing HEK293 and HeLa cell lines that catalyze MeHg demethylation. These cells were significantly more sensitive to MeHg exposure compared to the parental cells. MeHg treatment remarkably induced metallothioneins (MTs) and hemeoxygenase-1 (HMOX-1) mRNAs and modest expression of superoxide dismutase 1, whereas catalase and glutathione peroxidase 1 mRNAs were not up-regulated. merB knockdown using small interfering RNA supported the induction of MT and HMOX-1 mRNA by MerB enzymatic activity. Pretreatment with Trolox, a water-soluble vitamin E analog, did not inhibit MeHg-induced elevation of MT-Ix and HMOX-1 mRNAs in MerB-expressing cells, suggesting that Hg2+ works independently of reactive oxygen species generation. Similar results were obtained in cells expressing MerB, suggesting that high MTs and HMOX-1 induction and cytotoxicity are common cellular responses to low intracellular Hg2+ concentrations. This is the first study to establish cell lines that demethylate intracellular MeHg to Hg2+ using bacterial MerB for overcoming the low membrane permeability of Hg2+ and exploring the intracellular responses and toxicities of low Hg2+ concentrations.
Collapse
Affiliation(s)
- Yasukazu Takanezawa
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Ryosuke Nakamura
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Haruki Matsuda
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Tomomi Yagi
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Zen Egawa
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Yuka Sone
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Shimpei Uraguchi
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| | - Tatsumi Adachi
- Faculty of Pharmacy, Chiba Institute of Science, Choshi, Chiba 288-0025, Japan
| | - Masako Kiyono
- Department of Public Health, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641
| |
Collapse
|
12
|
Insights into the Potential Role of Mercury in Alzheimer's Disease. J Mol Neurosci 2019; 67:511-533. [PMID: 30877448 DOI: 10.1007/s12031-019-01274-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022]
Abstract
Mercury (Hg), which is a non-essential element, is considered a highly toxic pollutant for biological systems even when present at trace levels. Elevated Hg exposure with the growing release of atmospheric pollutant Hg and rising accumulations of mono-methylmercury (highly neurotoxic) in seafood products have increased its toxic potential for humans. This review aims to highlight the potential relationship between Hg exposure and Alzheimer's disease (AD), based on the existing literature in the field. Recent reports have hypothesized that Hg exposure could increase the potential risk of developing AD. Also, AD is known as a complex neurological disorder with increased amounts of both extracellular neuritic plaques and intracellular neurofibrillary tangles, which may also be related to lifestyle and genetic variables. Research reports on AD and relationships between Hg and AD indicate that neurotransmitters such as serotonin, acetylcholine, dopamine, norepinephrine, and glutamate are dysregulated in patients with AD. Many researchers have suggested that AD patients should be evaluated for Hg exposure and toxicity. Some authors suggest further exploration of the Hg concentrations in AD patients. Dysfunctional signaling pathways in AD and Hg exposure appear to be interlinked with some driving factors such as arachidonic acid, homocysteine, dehydroepiandrosterone (DHEA) sulfate, hydrogen peroxide, glucosamine glycans, glutathione, acetyl-L carnitine, melatonin, and HDL. This evidence suggests the need for a better understanding of the relationship between AD and Hg exposure, and potential mechanisms underlying the effects of Hg exposure on regional brain functions. Also, further studies evaluating brain functions are needed to explore the long-term effects of subclinical and untreated Hg toxicity on the brain function of AD patients.
Collapse
|
13
|
Barai P, Raval N, Acharya S, Acharya N. Neuroprotective effects of Bergenia ciliata on NMDA induced injury in SH-SY5Y cells and attenuation of cognitive deficits in scopolamine induced amnesia in rats. Biomed Pharmacother 2018; 108:374-390. [PMID: 30227331 DOI: 10.1016/j.biopha.2018.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023] Open
Abstract
Bergenia ciliata (Haw) Sternb. possess immunomodulatory, anti-inflammatory, antioxidant, anti-urolithiatic, wound healing, anti-malarial, anti-diabetic and anti-cancer properties. Moreover, the methanolic extracts of the rhizomes of the plant were found to demonstrate beneficial neuroprotective effects in the intracerebroventricular streptozotocin-induced model in rats. Thus, the present study was undertaken to further explore the neuroprotective potential of the aqueous (BA) and methanolic extracts (BM) of B. ciliata through various in-vitro and in-vivo studies. Both the extracts at all tested concentrations i.e. 50-50,000 ng/mL did not cause any significant reduction of cell viability of SH-SY5Y cells when tested for 48 h when assessed through MTT and resazurin metabolism- based cell viability assays. The pre-treatment with the extracts could confer significant (p < 0.001) and dose-dependent protective effects against NMDA induced injury in SH-SY5Y cells. BM [IC50: 5.7 and 5.19 μg/mL for acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) respectively] led to more potent inhibition of both the enzymes as compared to BA (IC50: 227.12 and 23.25 μg/mL for AChE and BuChE respectively). BM also proved to be a 1.85-fold better scavenger of the DPPH free radicals as compared to BA. Thus, BM was taken further for the evaluation of the beneficial effects of 14-day pre-treatment in rats in the scopolamine (2 mg/kg, i.p.) induced amnesia model at 125, 250 and 500 mg/kg, p.o. BM pre-treatment at 250 and 500 mg/kg could significantly ameliorate the cognitive impairment (p < 0.001), inhibit AChE (p < 0.001) and BuChE (p < 0.05) activity, restore GSH levels (p < 0.05) in serum and brain homogenates and recover the morphology of hippocampal neurons back to normal. Moreover, the BM administration at 500 mg/kg also showed beneficial effects through the significant (p < 0.05) reduction of Aβ1-42, phosphorylated tau (p-tau) and GSK-3β immunoreactivity in the brain homogenates of the intracerebroventricularly streptozotocin (ICV STZ) injected rats as observed from the results of the ELISA assays. The outcomes of the study unveiled that BM exerts its beneficial effects through prevention of NMDA induced excitotoxic cell death, dual cholinesterase inhibition, antioxidant activity coupled with the reduction of the immunoreactivity for the Aβ1-42, p-tau and GSK-3β indicating its potential to be screened further for various other models to determine the exact mechanism of action.
Collapse
Affiliation(s)
- Priyal Barai
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, 382481, Gujarat, India
| | - Nisith Raval
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, 382481, Gujarat, India
| | - Sanjeev Acharya
- SSR College of Pharmacy, Sayli, Silvassa, 306230, U. T. of D&NH, India
| | - Niyati Acharya
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
14
|
Barai P, Raval N, Acharya S, Borisa A, Bhatt H, Acharya N. Neuroprotective effects of bergenin in Alzheimer's disease: Investigation through molecular docking, in vitro and in vivo studies. Behav Brain Res 2018; 356:18-40. [PMID: 30118774 DOI: 10.1016/j.bbr.2018.08.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/20/2018] [Accepted: 08/11/2018] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is an enervating and chronic progressive neurodegenerative disorder, occurring frequently in the elderly and adversely affecting intellectual capabilities and the cognitive processes. Bergenin possesses efficacious antioxidant, antiulcerogenic, anti-HIV, hepatoprotective, neuroprotective, anti-inflammatory and immunomodulatory activity along with antinociceptive effect and wound healing properties. Previous studies have shown that bergenin has in vitro bovine adrenal tyrosine hydroxylase inhibitory activity, mushroom tyrosinase inhibitory activities, β-secretase (BACE-1) enzyme inhibitory activity and prevented neuronal death in the primary culture of rat cortical neurons. Protein tyrosine phosphatase-1B (PTP1B) is an intriguing target for anticancer and antidiabetic drugs and has recently been implicated to act as a positive regulator of neuroinflammation. Bergenin is also found to inhibit human protein tyrosine phosphatase-1B (hPTP1B) in vitro. Thus, bergenin was screened by molecular docking study using GOLD suite (version 5.2), CCDC for predicting its activity against targets of AD management like acetylcholinesterase (AChE) (1B41), butyrylcholinesterase (BuChE) (1P0I), Tau protein kinase 1 (GSK-3β) (1J1B), BACE-1 (1FKN) wherein the GOLD score and fitness of bergenin were comparable to those of standard drugs like donepezil, galanthamine, physostigmine, etc. Bergenin demonstrated dose-dependent inhibition of both AChE and BuChE in vitro and found to be safe up to 50 μM when screened in vitro on SH-SY5Y cell lines by cytotoxicity studies using MTT and Alamar blue assays. It also led to dose-dependent prevention of NMDA induced toxicity in these cells. Pretreatment with bergenin (14 days) in rats at three dose levels (20, 40 and 80 mg/kg; p.o.) significantly (p < 0.01) and dose-dependently alleviated amnesia induced by scopolamine (2 mg/kg, i.p.). The therapeutic effect of bergenin supplementation for 28 days, at three dose levels, was also evaluated in streptozotocin (3 mg/kg, ICV, unilateral) induced AD model in Wistar rats using Morris water maze and Y maze on 7th, 14th, 21st and 28th days. STZ caused significant (p < 0.001) cognitive impairment and cholinergic deficit and increased oxidative stress in rats. Bergenin could significantly ameliorate STZ induced behavioral deficits, inhibit the AChE and BuChE activity in parallel with an increase in the diminished GSH levels in a dose-dependent fashion. The histopathological investigations were also supportive of this datum. The bergenin treatment at 80 mg/kg led to significant (p < 0.05) abatement of the raised Aβ-1-42 levels and alleviated the perturbed p- tau levels leading to significantly low (p < 0.01) levels of p-tau in brain homogenates of rats as compared to ICV STZ injected rats. In conclusion, the observed effects might be attributed to the cholinesterase inhibitory activity of bergenin coupled with its antioxidant effect, anti-inflammatory activity and reduction of Aβ-1-42 and p-tau levels which could have collectively helped in the attenuation of cognitive deficits. The current findings of the study are indicative of the promising preventive and ameliorative potential of bergenin in the management of AD through multiple targets.
Collapse
Affiliation(s)
- Priyal Barai
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, Gujarat, 382481, India
| | - Nisith Raval
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, Gujarat, 382481, India
| | - Sanjeev Acharya
- SSR College of Pharmacy, Sayli, Silvassa - 306230, U. T. of D&NH, India
| | - Ankit Borisa
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, Gujarat, 382481, India
| | - Hardik Bhatt
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, Gujarat, 382481, India
| | - Niyati Acharya
- Institute of Pharmacy, Nirma University, S. G. Highway, Ahmedabad, Gujarat, 382481, India.
| |
Collapse
|
15
|
|
16
|
Aragão WAB, da Costa NMM, Fagundes NCF, Silva MCF, Alves-Junior SM, Pinheiro JJV, Amado LL, Crespo-López ME, Maia CSF, Lima RR. Chronic exposure to inorganic mercury induces biochemical and morphological changes in the salivary glands of rats. Metallomics 2018; 9:1271-1278. [PMID: 28795724 DOI: 10.1039/c7mt00123a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mercury exposure is considered to be a public health problem due to the generation of toxic effects on human health as a result of environmental and occupational conditions. The inorganic form of mercury (HgCl2) can cause several biological changes in cells and tissues through its cumulative toxic potential, but little has been experimentally proven about the effects of inorganic mercury on salivary glands, an important modulator organ of oral health. This study analyzes the effects of prolonged low dose exposure to HgCl2 on the salivary glands of rats. Adult animals received a dose of 0.375 mg kg-1 day-1 over a period of 45 days. The parotid and submandibular glands were collected for analysis of the mercury levels and evaluation of oxidative stress, histological parameters and immunomodulation for metallothionein I and II (MT-I/II). In this investigation, biochemical and tissue changes in the salivary glands were verified due to the mercury levels, causing reduction in antioxidant capacity against peroxyl radicals, with consequent cellular lipid peroxidation and an increase in nitrite levels, volumetric changes and cytoskeletal damage in the submandibular glands, with less severe damage to the parotid glands. The results also have shown the occurrence of a cytoprotection mechanism due to increased MT-I/II expression, but not enough to avoid the morphology and oxidative damage. This evidence highlights, for the first time, that inorganic mercury is able to alter the morphology and oxidative biochemistry in salivary glands when exposed for a long time in low doses.
Collapse
Affiliation(s)
- W A B Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Street Augusto Corrêa 1, Guamá, 66075-900, Belém, Pará, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Polverino A, Grimaldi M, Sorrentino P, Jacini F, D'Ursi AM, Sorrentino G. Effects of Acetylcholine on β-Amyloid-Induced cPLA2 Activation in the TB Neuroectodermal Cell Line: Implications for the Pathogenesis of Alzheimer's Disease. Cell Mol Neurobiol 2018; 38:817-826. [PMID: 28993924 DOI: 10.1007/s10571-017-0555-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
The role of β-amyloid (Aβ) in the pathogenesis of Alzheimer's disease (AD) is still considered crucial. The state of Aβ aggregation is critical in promoting neuronal loss and neuronal function impairment. Recently, we demonstrated that Acetylcholine (ACh) is neuroprotective against the toxic effects of Aβ in the cholinergic LAN-2 cells. In biophysical experiments, ACh promotes the soluble Aβ peptide conformation rather than the aggregation-prone β-sheet conformation. In order to better understand the biological role of ACh in AD, we studied the effect of Aβ on the phosphorylation of the cytosolic phospholipase A2 (cPLA2) in the TB neuroectodermal cell line, which differentiates toward a neuronal phenotype when cultured in the presence of retinoic acid (RA). We chose the phosphorylated form of cPLA2 (Ser505, Phospho-cPLA2) as a biomarker to test the influence of ACh on the effects of Aβ in both undifferentiated and RA-differentiated TB cells. Our results show that TB cells are responsive to Aβ. Moreover, in undifferentiated cells 1 h treatment with Aβ induces a 2.5-fold increase of the Phospho-cPLA2 level compared to the control after 24 h in vitro, while no significant difference is observed between Aβ-treated and non-treated cells after 4 and 7 days in vitro. The RA-differentiated cells are not sensitive to Aβ. In TB cell line ACh is able to blunt the effects of Aβ. The ability of ACh to protect non-cholinergic cells against Aβ reinforces the hypothesis that, in addition to its role in cholinergic transmission, ACh could also act as a neuroprotective agent.
Collapse
Affiliation(s)
- Arianna Polverino
- Department of Motor Sciences and Wellness, University of Naples Parthenope, Via Medina, 40, 80133, Naples, NA, Italy
- Institute of Diagnosis and Treatment Hermitage, Via Cupa delle Tozzole, 2, 80131, Naples, NA, Italy
| | - Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, Salerno, SA, Italy
| | - Pierpaolo Sorrentino
- Department of Engineering, University of Naples Parthenope, Centro Direzionale di Napoli, isola C4, 80143, Naples, NA, Italy
| | - Francesca Jacini
- Department of Motor Sciences and Wellness, University of Naples Parthenope, Via Medina, 40, 80133, Naples, NA, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, Salerno, SA, Italy
| | - Giuseppe Sorrentino
- Department of Motor Sciences and Wellness, University of Naples Parthenope, Via Medina, 40, 80133, Naples, NA, Italy.
- Institute of Diagnosis and Treatment Hermitage, Via Cupa delle Tozzole, 2, 80131, Naples, NA, Italy.
| |
Collapse
|
18
|
Bautista E, Zarco N, Aguirre-Pineda N, Lara-Lozano M, Vergara P, González-Barrios JA, Aguilar-Roblero R, Segovia J. Expression of Gas1 in Mouse Brain: Release and Role in Neuronal Differentiation. Cell Mol Neurobiol 2018; 38:841-859. [PMID: 29110208 DOI: 10.1007/s10571-017-0559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/14/2017] [Indexed: 10/18/2022]
Abstract
Growth arrest-specific 1 (Gas1) is a pleiotropic protein that induces apoptosis of tumor cells and has important roles during development. Recently, the presence of two forms of Gas1 was reported: one attached to the cell membrane by a GPI anchor; and a soluble extracellular form shed by cells. Previously, we showed that Gas1 is expressed in different areas of the adult mouse CNS. Here, we report the levels of Gas1 mRNA protein in different regions and analyzed its expressions in glutamatergic, GABAergic, and dopaminergic neurons. We found that Gas1 is expressed in GABAergic and glutamatergic neurons in the Purkinje-molecular layer of the cerebellum, hippocampus, thalamus, and fastigial nucleus, as well as in dopaminergic neurons of the substantia nigra. In all cases, Gas1 was found in the cell bodies, but not in the neuropil. The Purkinje and the molecular layers show the highest levels of Gas1, whereas the granule cell layer has low levels. Moreover, we detected the expression and release of Gas1 from primary cultures of Purkinje cells and from hippocampal neurons as well as from neuronal cell lines, but not from cerebellar granular cells. In addition, using SH-SY5Y cells differentiated with retinoic acid as a neuronal model, we found that extracellular Gas1 promotes neurite outgrowth, increases the levels of tyrosine hydroxylase, and stimulates the inhibition of GSK3β. These findings demonstrate that Gas1 is expressed and released by neurons and promotes differentiation, suggesting an important role for Gas1 in cellular signaling in the CNS.
Collapse
Affiliation(s)
- Elizabeth Bautista
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico
| | - Natanael Zarco
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico
| | - Nicolás Aguirre-Pineda
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico
| | - Manuel Lara-Lozano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico
- Laboratorio de Medicina Genómica, Hospital Regional 1 de Octubre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Avenida Instituto Politécnico Nacional N° 1669, Gustavo A. Madero, Col. Magdalena de las Salinas, Del. Gustavo A. Madero, 07760, Mexico, DF, Mexico
| | - Paula Vergara
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico
| | - Juan Antonio González-Barrios
- Laboratorio de Medicina Genómica, Hospital Regional 1 de Octubre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Avenida Instituto Politécnico Nacional N° 1669, Gustavo A. Madero, Col. Magdalena de las Salinas, Del. Gustavo A. Madero, 07760, Mexico, DF, Mexico
| | - Raúl Aguilar-Roblero
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Coyoacan, 04510, Mexico, DF, Mexico
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico, DF, Mexico.
| |
Collapse
|
19
|
Hippocampal Dysfunction Provoked by Mercury Chloride Exposure: Evaluation of Cognitive Impairment, Oxidative Stress, Tissue Injury and Nature of Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7878050. [PMID: 29849915 PMCID: PMC5914100 DOI: 10.1155/2018/7878050] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/31/2018] [Accepted: 02/21/2018] [Indexed: 01/03/2023]
Abstract
Mercury (Hg) is a highly toxic metal, which can be found in its inorganic form in the environment. This form presents lower liposolubility and lower absorption in the body. In order to elucidate the possible toxicity of inorganic Hg in the hippocampus, we investigated the potential of low doses of mercury chloride (HgCl2) to promote hippocampal dysfunction by employing a chronic exposure model. For this, 56 rats were exposed to HgCl2 (0.375 mg/kg/day) via the oral route for 45 days. After the exposure period, the animals were submitted to the cognitive test of fear memory. The hippocampus was collected for the measurement of total Hg levels, analysis of oxidative stress, and evaluation of cytotoxicity, apoptosis, and tissue injury. It was observed that chronic exposure to inorganic Hg promotes an increase in mercury levels in this region and damage to short- and long-term memory. Furthermore, we found that this exposure model provoked oxidative stress, which led to cytotoxicity and cell death by apoptosis, affecting astrocytes and neurons in the hippocampus. Our study demonstrated that inorganic Hg, even with its low liposolubility, is able to produce deleterious effects in the central nervous system, resulting in cognitive impairment and hippocampal damage when administered for a long time at low doses in rats.
Collapse
|
20
|
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: 109] [Impact Index Per Article: 18.2] [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
|