1
|
Jomova K, Alomar SY, Nepovimova E, Kuca K, Valko M. Heavy metals: toxicity and human health effects. Arch Toxicol 2025; 99:153-209. [PMID: 39567405 PMCID: PMC11742009 DOI: 10.1007/s00204-024-03903-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Heavy metals are naturally occurring components of the Earth's crust and persistent environmental pollutants. Human exposure to heavy metals occurs via various pathways, including inhalation of air/dust particles, ingesting contaminated water or soil, or through the food chain. Their bioaccumulation may lead to diverse toxic effects affecting different body tissues and organ systems. The toxicity of heavy metals depends on the properties of the given metal, dose, route, duration of exposure (acute or chronic), and extent of bioaccumulation. The detrimental impacts of heavy metals on human health are largely linked to their capacity to interfere with antioxidant defense mechanisms, primarily through their interaction with intracellular glutathione (GSH) or sulfhydryl groups (R-SH) of antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and other enzyme systems. Although arsenic (As) is believed to bind directly to critical thiols, alternative hydrogen peroxide production processes have also been postulated. Heavy metals are known to interfere with signaling pathways and affect a variety of cellular processes, including cell growth, proliferation, survival, metabolism, and apoptosis. For example, cadmium can affect the BLC-2 family of proteins involved in mitochondrial death via the overexpression of antiapoptotic Bcl-2 and the suppression of proapoptotic (BAX, BAK) mechanisms, thus increasing the resistance of various cells to undergo malignant transformation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of antioxidant enzymes, the level of oxidative stress, and cellular resistance to oxidants and has been shown to act as a double-edged sword in response to arsenic-induced oxidative stress. Another mechanism of significant health threats and heavy metal (e.g., Pb) toxicity involves the substitution of essential metals (e.g., calcium (Ca), copper (Cu), and iron (Fe)) with structurally similar heavy metals (e.g., cadmium (Cd) and lead (Pb)) in the metal-binding sites of proteins. Displaced essential redox metals (copper, iron, manganese) from their natural metal-binding sites can catalyze the decomposition of hydrogen peroxide via the Fenton reaction and generate damaging ROS such as hydroxyl radicals, causing damage to lipids, proteins, and DNA. Conversely, some heavy metals, such as cadmium, can suppress the synthesis of nitric oxide radical (NO·), manifested by altered vasorelaxation and, consequently, blood pressure regulation. Pb-induced oxidative stress has been shown to be indirectly responsible for the depletion of nitric oxide due to its interaction with superoxide radical (O2·-), resulting in the formation of a potent biological oxidant, peroxynitrite (ONOO-). This review comprehensively discusses the mechanisms of heavy metal toxicity and their health effects. Aluminum (Al), cadmium (Cd), arsenic (As), mercury (Hg), lead (Pb), and chromium (Cr) and their roles in the development of gastrointestinal, pulmonary, kidney, reproductive, neurodegenerative (Alzheimer's and Parkinson's diseases), cardiovascular, and cancer (e.g. renal, lung, skin, stomach) diseases are discussed. A short account is devoted to the detoxification of heavy metals by chelation via the use of ethylenediaminetetraacetic acid (EDTA), dimercaprol (BAL), 2,3-dimercaptosuccinic acid (DMSA), 2,3-dimercapto-1-propane sulfonic acid (DMPS), and penicillamine chelators.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, 949 74, Nitra, Slovakia
| | - Suliman Y Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
2
|
Moreno F, Méndez L, Fernández I, Miralles-Pérez B, Giralt M, Romeu M, Ramos-Romero S, Torres JL, Medina I. Influence of the Degree of Unsaturation in Fish Oil Supplements on Oxidative Stress and Protein Carbonylation in the Cerebral Cortex and Cerebellum of Healthy Rats. Antioxidants (Basel) 2024; 13:1408. [PMID: 39594550 PMCID: PMC11591239 DOI: 10.3390/antiox13111408] [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: 09/30/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
ω-3 polyunsaturated fatty acids (PUFAs) are crucial for brain structure and function, especially docosahexaenoic acid (DHA). However, an excess of DHA may increase lipid peroxidation due to its high degree of unsaturation, particularly in tissues highly susceptible to oxidative stress, such as the brain. Therefore, this study evaluated the effects of 10 weeks of dietary supplementation with fish oil containing 80% DHA on oxidative stress and the modulation of the carbonylated proteome in both the cerebral cortex and cerebellum of male Sprague Dawley rats. The results were compared with those induced by oils with a lower degree of fat unsaturation (fish oil containing 25% DHA and 25% eicosapentaenoic acid, soybean oil containing 50% linoleic acid and coconut oil containing 90% saturated fat). The results demonstrated that fish oil containing 80% DHA significantly increased the ω3/ω6 ratio in both the cortex and cerebellum while stimulating antioxidant defense by enhancing the reduced glutathione amount and decreasing the carbonylation of specific proteins, mainly those involved in glycolysis and neurotransmission. The majority of sensitive proteins in both brain regions followed this carbonylation trend (in decreasing order): soybean > EPA/DHA 1:1 > coconut > 80% DHA. The results also indicated that the cerebellum is more responsive than the cortex to changes in the cellular redox environment induced by varying degrees of fat unsaturation. In conclusion, under healthy conditions, dietary supplementation with fish oils containing high DHA levels makes the brain more resilient to potential oxidative insults compared to oils with lower DHA content and a lower degree of fatty acid unsaturation.
Collapse
Affiliation(s)
- Francisco Moreno
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
- University of Vigo, Circunvalación ao Campus Universitario, 36310 Vigo, Spain
| | - Lucía Méndez
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| | - Ingrid Fernández
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| | - Bernat Miralles-Pérez
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Montserrat Giralt
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Marta Romeu
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.G.); (M.R.)
| | - Sara Ramos-Romero
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
- Nutrition & Food Safety Research Institute (INSA-UB), Maria de Maeztu Unit of Excellence, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain;
- Institute of Advanced Chemistry of Catalonia—Spanish National Research Council (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josep Lluís Torres
- Nutrition & Food Safety Research Institute (INSA-UB), Maria de Maeztu Unit of Excellence, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain;
- Institute of Advanced Chemistry of Catalonia—Spanish National Research Council (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Isabel Medina
- Institute of Marine Research—Spanish National Research Council (IIM-CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (F.M.); (I.F.); (B.M.-P.); (I.M.)
| |
Collapse
|
3
|
Das R, Karri R, Chalana A, Rai RK, Roy G. Uncovering the Role of Methylmercury on DNA Lesions at Cytotoxic Concentrations in Glutathione-Depleted Cells: Insights from Experimental and Computational Studies. Inorg Chem 2024; 63:10455-10465. [PMID: 38743433 DOI: 10.1021/acs.inorgchem.3c04579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Organomercurials (RHg+), especially methylmercury (MeHg+) and ethylmercury (EtHg+), are considered to be more neurotoxic than the inorganic counterpart (Hg2+). They cause massive DNA damage in cells, especially in neurons, where cellular glutathione (GSH) levels are significantly low. However, the mechanism by which RHg+ exerts massive DNA damage at cytotoxic concentrations in brain cells remains obscure. In this study, we investigated the effect of RHg+ on the structural and electronic properties of nucleosides and its effects on DNA damage. The direct interaction of RHg+ with the nucleoside significantly weakens N-glycosidic bonds, decreases the C-H bond energy of sugar moieties, and increases the electrophilicity of the C8-center of purine bases. As a consequence, RHg+-conjugated DNA molecules are extremely labile and highly sensitive to any nucleophiles/radicals present in GSH-depleted cells and, thus, undergo enhanced oxidative and unusual alkylative DNA damage. We also report a functional model of organomercurial lyase, which showed excellent cytoprotective effect against RHg+-induced cytotoxicity; this reverses the activity of glutathione reductase inhibited by MeHgCl and ceases oxidative and alkylating DNA damage. This intriguing finding provides new mechanistic insight into the mode of action of organomercurials in GSH-depleted cells and their adverse effects on individuals with neurodegenerative disorders associated with oxidative stress.
Collapse
Affiliation(s)
- Ranajit Das
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, UP 201314, India
| | - Ramesh Karri
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, UP 201314, India
- Ruhvenile Biomedical OPC Pvt. Ltd., New Delhi 110070, Delhi, India
| | - Ashish Chalana
- Centre for Development of Biomaterials, Department of Chemistry & Biochemistry, Sharda University, Greater Noida, UP 201306, India
| | - Rakesh Kumar Rai
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, AP 517619, India
| | - Gouriprasanna Roy
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, AP 517619, India
| |
Collapse
|
4
|
Abdelmoez WA. Evaluation of histological and ultrastructural changes provoked by prenatal tramadol on postnatal cortical cerebellar neuronal development in rats: possible implication of Ki67, GFAP and MicroRNA-7/P53 signalling trajectories. J Mol Histol 2024; 55:279-301. [PMID: 38639812 PMCID: PMC11102883 DOI: 10.1007/s10735-024-10189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Tramadol is a novel centrally acting analgesic. Despite, its implementation during pregnancy may impair neuronal survival and synaptic development in neonatal cerebella. The current investigation assessed the histological and ultrastructural alterations in postnatal cortical cerebellar neuronal development induced by prenatal tramadol. 30 offsprings were divided to control group I: fifteen pups born to mothers given saline from D10 till D21 of gestation. Tramadol-treated group II: fifteen pups born to mothers received tramadol HCL (50 mg/kg/day) from D10 till D21 of gestation. Pups were categorized into three subgroups (a, b, and c) and offered for sacrifice on the seventh, fourteenth and twenty-first post-natal days. Light microscopic examination revealed the overcrowding and signs of red degeneration affecting purkinje cell layer. Neurodegenerative signs of both purkinje and granule cell neurons were also confirmed by TEM in form of chromatin condensation, dilated Golgi channels, disrupted endoplasmic reticulum, marked infolding of the nuclear envelope and decrease in granule cell precursors. In addition, the astrocytic processes and terminal nerve axons appeared with different degrees of demyelination and decreased number of oligodendrocytes and degenerated mitochondria. Furthermore, group II exhibited an increase in P53 immune expression. The area percentage of apoptotic cells detected by TUNEL assay was significantly increased. Besides to the significant decrease of Ki67 immunoreactivity in the stem neuronal cell progenitors. Quantitative PCR results showed a significant decline in micro RNA7 gene expression in tramadol treated groups resulting in affection of multiple target genes in P53 signaling pathways, improper cortical size and defect in neuronal development.
Collapse
Affiliation(s)
- Walaa Adel Abdelmoez
- Department of Anatomy and Embryology, Faculty of Medicine, Ain-Shams University, Postal Code: 11591, Abbassia, Cairo, Egypt.
| |
Collapse
|
5
|
Environmentally relevant developmental methylmercury exposures alter neuronal differentiation in a human-induced pluripotent stem cell model. Food Chem Toxicol 2021; 152:112178. [PMID: 33831500 DOI: 10.1016/j.fct.2021.112178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Developmental methylmercury (MeHg) exposure selectively targets the cerebral and cerebellar cortices, as seen by disruption of cytoarchitecture and glutamatergic (GLUergic) neuron hypoplasia. To begin to understand the mechanisms of this loss of GLUergic neurons, we aimed to develop a model of developmental MeHg neurotoxicity in human-induced pluripotent stem cells differentiating into cortical GLUergic neurons. Three dosing paradigms at 0.1 μM and 1.0 μM MeHg, which span different stages of neurodevelopment and reflect toxicologically relevant accumulation levels seen in human studies and mammalian models, were established. With these exposure paradigms, no changes were seen in commonly studied endpoints of MeHg toxicity, including viability, proliferation, and glutathione levels. However, MeHg exposure induced changes in mitochondrial respiration and glycolysis and in markers of neuronal differentiation. Our novel data suggests that GLUergic neuron hypoplasia seen with MeHg toxicity may be due to the partial inhibition of neuronal differentiation, given the increased expression of the early dorsal forebrain marker FOXG1 and corresponding decrease in expression on neuronal markers MAP2 and DCX and the deep layer cortical neuronal marker TBR1. Future studies should examine the persistent and latent functional effects of this MeHg-induced disruption of neuronal differentiation as well as transcriptomic and metabolomic alterations that may mediate MeHg toxicity.
Collapse
|
6
|
THONGSAW A, SANANMUANG R, UDNAN Y, AMPIAH-BONNEY RJ, CHAIYASITH WC. Immobilized Activated Carbon as Sorbent in Solid Phase Extraction with Cold Vapor Atomic Absorption Spectrometry for the Preconcentration and Determination of Mercury Species in Water and Freshwater Fish Samples. ANAL SCI 2019; 35:1195-1202. [DOI: 10.2116/analsci.19p164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Arnon THONGSAW
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | - Ratana SANANMUANG
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | - Yuthapong UDNAN
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| | | | - Wipharat Chuachuad CHAIYASITH
- Department of Chemistry, Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University
| |
Collapse
|
7
|
Karri R, Chalana A, Kumar B, Jayadev SK, Roy G. Exploiting the κ 2 -Fashioned Coordination of [Se 2 ]-Donor Ligand L 3 Se for Facile Hg-C Bond Cleavage of Mercury Alkyls and Cytoprotection against Methylmercury-Induced Toxicity. Chemistry 2019; 25:12810-12819. [PMID: 31298434 DOI: 10.1002/chem.201902578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/09/2019] [Indexed: 11/10/2022]
Abstract
The Hg-C bond of MeHgCl, a ubiquitous environmental toxicant, is notoriously inert and exceedingly difficult to cleave. The cleavage of the Hg-C bond of MeHgCl at low temperature, therefore, is of significant importance for human health. Among various bis(imidazole)-2-selones Ln Se (n=1-4, or 6), the three-spacer L3 Se shows extraordinarily high reactivity in the degradation of various mercury alkyls including MeHgCl because of its unique ability to coordinate through κ2 -fashion, in which both the Se atoms simultaneously attack the Hg center of mercury alkyls for facile Hg-C bond cleavage. It has the highest softness (σ) parameter and the lowest HOMO(Ln Se)-LUMO(MeHgX) energy gap and, thus, L3 Se is the most reactive among Ln Se towards MeHgX (X=Cl or I). L3 Se is highly efficient, more than L1 Se, in restoring the activity of antioxidant enzyme glutathione reductase (GR) that is completely inhibited by MeHgCl; 80 % GR activity is recovered by L3 Se relative to 50 % by L1 Se. It shows an excellent cytoprotective effect in liver cells against MeHgCl-induced oxidative stress by protecting vital antioxidant enzymes from inhibition caused by MeHgCl and, thus, does not allow to increase the intracellular reactive oxygen species (ROS) levels. Furthermore, it protects the mitochondrial membrane potential (ΔΨm ) from perturbation by MeHgCl. Major Hg-responsive genes analyses demonstrate that L3 Se plays a significant role in MeHg+ detoxification in liver cells.
Collapse
Affiliation(s)
- Ramesh Karri
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Ashish Chalana
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Binayak Kumar
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Sri Krishna Jayadev
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| | - Gouriprasanna Roy
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NH91, Greater Noida, UP, 201314, India
| |
Collapse
|
8
|
Maciejczyk M, Żebrowska E, Chabowski A. Insulin Resistance and Oxidative Stress in the Brain: What's New? Int J Mol Sci 2019; 20:ijms20040874. [PMID: 30781611 PMCID: PMC6413037 DOI: 10.3390/ijms20040874] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
The latest studies have indicated a strong relationship between systemic insulin resistance (IR) and higher incidence of neurodegeneration, dementia, and mild cognitive impairment. Although some of these abnormalities could be explained by chronic hyperglycaemia, hyperinsulinemia, dyslipidaemia, and/or prolonged whole-body inflammation, the key role is attributed to the neuronal redox imbalance and oxidative damage. In this mini review, we provide a schematic overview of intracellular oxidative stress and mitochondrial abnormalities in the IR brain. We highlight important correlations found so far between brain oxidative stress, ceramide generation, β-amyloid accumulation, as well as neuronal apoptosis in the IR conditions.
Collapse
Affiliation(s)
- Mateusz Maciejczyk
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2c Str., 15-222 Bialystok, Poland.
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2c Str., 15-222 Bialystok, Poland.
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2c Str., 15-222 Bialystok, Poland.
| |
Collapse
|
9
|
Prince LM, Aschner M, Bowman AB. Human-induced pluripotent stems cells as a model to dissect the selective neurotoxicity of methylmercury. Biochim Biophys Acta Gen Subj 2019; 1863:129300. [PMID: 30742955 DOI: 10.1016/j.bbagen.2019.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/09/2019] [Accepted: 02/01/2019] [Indexed: 01/07/2023]
Abstract
Methylmercury (MeHg) is a potent neurotoxicant affecting both the developing and mature central nervous system (CNS) with apparent indiscriminate disruption of multiple homeostatic pathways. However, genetic and environmental modifiers contribute significant variability to neurotoxicity associated with human exposures. MeHg displays developmental stage and neural lineage selective neurotoxicity. To identify mechanistic-based neuroprotective strategies to mitigate human MeHg exposure risk, it will be critical to improve our understanding of the basis of MeHg neurotoxicity and of this selective neurotoxicity. Here, we propose that human-based pluripotent stem cell cellular approaches may enable mechanistic insight into genetic pathways that modify sensitivity of specific neural lineages to MeHg-induced neurotoxicity. Such studies are crucial for the development of novel disease modifying strategies impinging on MeHg exposure vulnerability.
Collapse
Affiliation(s)
- Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States.
| |
Collapse
|
10
|
Niño SA, Morales-Martínez A, Chi-Ahumada E, Carrizales L, Salgado-Delgado R, Pérez-Severiano F, Díaz-Cintra S, Jiménez-Capdeville ME, Zarazúa S. Arsenic Exposure Contributes to the Bioenergetic Damage in an Alzheimer's Disease Model. ACS Chem Neurosci 2019; 10:323-336. [PMID: 30141907 DOI: 10.1021/acschemneuro.8b00278] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Worldwide, every year there is an increase in the number of people exposed to inorganic arsenic (iAs) via drinking water. Human populations present impaired cognitive function as a result of prenatal and childhood iAs exposure, while studies in animal models demonstrate neurobehavioral deficits accompanied by neurotransmitter, protein, and enzyme alterations. Similar impairments have been observed in close association with Alzheimer's disease (AD). In order to determine whether iAs promotes the pathophysiological progress of AD, we used the 3xTgAD mouse model. Mice were exposed to iAs in drinking water from gestation until 6 months (As-3xTgAD group) and compared with control animals without arsenic (3xTgAD group). We investigated the behavior phenotype on a test battery (circadian rhythm, locomotor behavior, Morris water maze, and contextual fear conditioning). Adenosine triphosphate (ATP), reactive oxygen species, lipid peroxidation, and respiration rates of mitochondria were evaluated, antioxidant components were detected by immunoblots, and immunohistochemical studies were performed to reveal AD markers. As-3xTgAD displayed alterations in their circadian rhythm and exhibited longer freezing time and escape latencies compared to the control group. The bioenergetic profile revealed decreased ATP levels accompanied by the decline of complex I, and an oxidant state in the hippocampus. On the other hand, the cortex showed no changes of oxidant stress and complex I; however, the antioxidant response was increased. Higher immunopositivity to amyloid isoforms and to phosphorylated tau was observed in frontal cortex and hippocampus of exposed animals. In conclusion, mitochondrial dysfunction may be one of the triggering factors through which chronic iAs exposure exacerbates brain AD-like pathology.
Collapse
Affiliation(s)
- Sandra Aurora Niño
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| | - Adriana Morales-Martínez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Leticia Carrizales
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Roberto Salgado-Delgado
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava Martínez S/N, CP 78290 San Luis Potosí, SLP, Mexico
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, CP 76230 Juriquilla, Querétaro, México
| | - María E. Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Sergio Zarazúa
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| |
Collapse
|
11
|
Aboulhoda BE, Hassan SS. Effect of prenatal tramadol on postnatal cerebellar development: Role of oxidative stress. J Chem Neuroanat 2018; 94:102-118. [PMID: 30342117 DOI: 10.1016/j.jchemneu.2018.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/17/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM The adverse neurological effects of tramadol have recently raised attention. The literature pertaining to studying postnatal cerebellar changes induced by prenatal tramadol is very scanty, thus the current study has been designed to improve understanding of the cerebellar oxidative stress-related alterations associated with tramadol administration during pregnancy in this critical period of neuronal differentiation and synaptic development, thereby highlighting the importance of controlling prenatal prescription of opioids and optimizing care for opioid-dependent pregnant women and their infants. MATERIAL AND METHODS Twenty pregnant female rats of Sprague Dawley strains were used in the study. Their offspring were divided into two groups: group I (control group) offspring of mothers given saline; group II offspring of mothers given tramadol from the 10th day (D10) of gestation till D21. The pups were sacrificed on the 7th, 14th and 21st postnatal days. Cerebellar specimens were processed for histomorphometric, immunohistochemical and electron microscopic assessment and were evaluated for various oxidative stress parameters. RESULTS Tramadol administration during pregnancy caused profound structural abnormalities on the post-natal cerebellar cortex and was associated with oxidative stress evidenced by elevation of lipid peroxidation products and inhibition of antioxidant enzyme activities.
Collapse
Affiliation(s)
- Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Egypt.
| | - Sherif S Hassan
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Egypt; Department of Medical Education, California University of Sciences and Medicine, School of medicine, San Bernardino, 92408 CA, USA
| |
Collapse
|
12
|
Redox Balance, Antioxidant Defense, and Oxidative Damage in the Hypothalamus and Cerebral Cortex of Rats with High Fat Diet-Induced Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6940515. [PMID: 30271528 PMCID: PMC6146783 DOI: 10.1155/2018/6940515] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/09/2018] [Indexed: 01/07/2023]
Abstract
Oxidative stress is a key pathogenic factor in both neurogenerative and metabolic diseases. However, its contribution in the brain complications of insulin resistance is still not well understood. Therefore, the aim of this study was the evaluation of redox homeostasis and oxidative damage in the hypothalamus and cerebral cortex of insulin-resistant and control rats. 16 male Wistar rats were divided into two equal groups (n = 8): the control and high fat diet group (HFD). Prooxidant enzymes (xanthine oxidase and NADPH oxidase); enzymatic and nonenzymatic antioxidants [glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), superoxide dismutase-1 (SOD-1), and uric acid (UA)]; and oxidative damage products [advanced glycation end products (AGE), 4-hydroxynonenal (4-HNE), malondialdehyde (MDA), and 8-hydroxy-2′-deoxyguanosine (8-OHdG)] as well as the total antioxidant capacity (TAC), total oxidant status (TOS), oxidative stress index (OSI), and total ferric reducing ability of sample (FRAP) were evaluated in the hypothalamus and cerebral cortex as well as serum/plasma of HFD-fed and control rats. The activity of prooxidant enzymes was significantly increased in the cerebral cortex and hypothalamus of HFD-fed rats vs. control rats. Additionally, we have showed enhanced antioxidant efficiency in the hypothalamus (↑CAT, ↑UA, ↑TAC, and ↑FRAP) and cerebral cortex (↑GPx, ↑CAT, ↑SOD-1, ↑UA, ↑TAC, and ↑FRAP) of HFD-fed rats. All of the oxidative damage markers (AGE, 4-HNE, MDA, 8-OHdG, and OSI) were significantly increased in the cerebral cortex of insulin-resistant rats, while only 4-HNE and MDA were markedly higher in the hypothalamus of the HFD group. Summarizing, the results of our study indicate an adaptive brain response to the increased production of free radicals under insulin resistance conditions. Despite the increase in antioxidative defense systems, this mechanism does not protect both brain structures from oxidative damages. However, the cerebral cortex is more susceptible to oxidative stress caused by HFD.
Collapse
|
13
|
Antunes Dos Santos A, Ferrer B, Marques Gonçalves F, Tsatsakis AM, Renieri EA, Skalny AV, Farina M, Rocha JBT, Aschner M. Oxidative Stress in Methylmercury-Induced Cell Toxicity. TOXICS 2018; 6:toxics6030047. [PMID: 30096882 PMCID: PMC6161175 DOI: 10.3390/toxics6030047] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
Methylmercury (MeHg) is a hazardous environmental pollutant, which elicits significant toxicity in humans. The accumulation of MeHg through the daily consumption of large predatory fish poses potential health risks, and the central nervous system (CNS) is the primary target of toxicity. Despite well-described neurobehavioral effects (i.e., motor impairment), the mechanisms of MeHg-induced toxicity are not completely understood. However, several lines of evidence point out the oxidative stress as an important molecular mechanism in MeHg-induced intoxication. Indeed, MeHg is a soft electrophile that preferentially interacts with nucleophilic groups (mainly thiols and selenols) from proteins and low-molecular-weight molecules. Such interaction contributes to the occurrence of oxidative stress, which can produce damage by several interacting mechanisms, impairing the function of various molecules (i.e., proteins, lipids, and nucleic acids), potentially resulting in modulation of different cellular signal transduction pathways. This review summarizes the general aspects regarding the interaction between MeHg with regulators of the antioxidant response system that are rich in thiol and selenol groups such as glutathione (GSH), and the selenoenzymes thioredoxin reductase (TrxR) and glutathione peroxidase (Gpx). A particular attention is directed towards the role of the PI3K/Akt signaling pathway and the nuclear transcription factor NF-E2-related factor 2 (Nrf2) in MeHg-induced redox imbalance.
Collapse
Affiliation(s)
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Aristides M Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece.
| | - Elisavet A Renieri
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece.
| | - Anatoly V Skalny
- Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow 150000, Russia.
- Laboratory of Biotechnology and Applied Bioelementology, Yaroslavl State University, Yaroslavl 150014, Russia.
- All-Russian Research Institute of Medicinal and Aromatic Plants (VILAR), Moscow 150000, Russia.
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis 88040-900, Santa Catarina, Brazil.
| | - João B T Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria 97105-900, Rio Grande do Sul, Brazil.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
14
|
Wang Y, Qin Y, Guo T, Tang C, Liu L, Gao D. High Concentration of Glial Cell Line-Derived Neurotrophic Factor Protects Primary Astrocytes from Apoptosis. Dev Neurosci 2018; 40:134-144. [DOI: 10.1159/000487853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Studies have shown that astrocytes play an important role in a variety of biological processes, so damage to astrocytes can cause a series of related diseases. Glial cell line-derived neurotrophic factor (GDNF) has always been considered a protective factor for dopamine neurons. However, it remains unclear whether GDNF has a protective effect on glial cells, especially astrocytes. In this study, we put forward the hypothesis that a high concentration of GDNF in the microenvironment of astrocytes exerts an inhibitory effect on the apoptosis of astrocytes by DNA-damaging reagents. Methods: We isolated, purified, and identified primary astrocytes from neonate rats. Astrocytes were exposed to mitoxantrone (MTN, a DNA-damaging compound) for 24 h. The effects of MTN on astrocytes were tested by Hoechst 33342 staining, CCK-8 assay, and flow cytometry assay. One of the concentrations of MTN was applied to construct an apoptotic model of astrocytes. The astrocytes were then treated with GDNF together with a selected concentration of MTN for 24 h. The cell viability, cell nucleus morphology, and apoptosis ratio of the cells was assessed by Hoechst 33342 staining, CCK-8 assay, and flow cytometry assay, respectively. RNA sequencing (RNA-Seq), quantitative PCR analysis, and KEGG pathway mapping were performed to examine the genes involved in the procedure. Finally, Western blot analysis was applied to confirm the expression levels of the proteins of interest. Results: Hoechst 33342 staining revealed a one-tenth change in the percentage of Hoechst-positive cells after the addition of 500 ng/mL GDNF combined with 1,000 nM MTN for 24 h. The viability of the cells treated the same as described above was 1.4-fold that of the control group. Flow cytometry assays indicated that the apoptotic rates were 17.67, 8.67, and 4.34% for 0, 200, and 500 ng/mL GDNF, respectively. Birc2, Birc3, and Gadd45b were linked to the antiapoptotic process induced by GDNF in astrocytes. Western blot analysis confirmed the elevated expression of Birc2 and Gadd45b. Conclusions: Our studies revealed that GDNF has a noticeable antiapoptotic effect on gene-injured astrocytes. This may provide critical clues for the treatment of a series of diseases in which damaged astrocytes are involved.
Collapse
|
15
|
Kosenko EA, Tikhonova LA, Alilova GA, Montoliu C, Barreto GE, Aliev G, Kaminsky YG. Portacaval shunting causes differential mitochondrial superoxide production in brain regions. Free Radic Biol Med 2017; 113:109-118. [PMID: 28964916 DOI: 10.1016/j.freeradbiomed.2017.09.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/08/2017] [Accepted: 09/25/2017] [Indexed: 01/23/2023]
Abstract
The portacaval shunting (PCS) prevents portal hypertension and recurrent bleeding of esophageal varices. On the other hand, it can induce chronic hyperammonemia and is considered to be the best model of mild hepatic encephalopathy (HE). Pathogenic mechanisms of HE and dysfunction of the brain in hyperammonemia are not fully elucidated, but it was originally suggested that the pathogenetic defect causes destruction of antioxidant defense which leads to an increase in the production of reactive oxygen species (ROS) and the occurrence of oxidative stress. In order to gain insight into the pathogenic mechanisms of HE in the brain tissue, we investigated the effects of PCS in rats on free radicals production and activity levels of antioxidant and prooxidant enzymes in mitochondria isolated from different brain areas. We found that O2·- production, activities of Mn-superoxide dismutase (Mn-SOD), glutathione peroxidase (GPx), glutathione reductase (GR), glutathione transferase (GT), nitric oxide synthase (NOS), and levels of carbonylated proteins differed between the four brain regions both in the amount and response to PCS. In PCS rats, Mn-SOD activity in the cerebellum was significantly decreased, and remained unchanged in the neocortex, hippocampus and striatum compared with that in sham-operated animals. Among the four brain regions in control rats, the levels of the carbonyl groups in mitochondrial proteins were maximal in the cerebellum. 4 weeks after PCS, the content of carbonylated proteins were higher only in mitochondria of this brain region. Under control conditions, O2·- production by submitochondrial particles in the cerebellum was significantly higher than in other brain regions, but was significantly increased in each brain region from PCS animals. Indeed, the production of O2·- by submitochondrial particles correlated with mitochondrial ammonia levels in the four brain regions of control and PCS-animals. These findings are the first to suggest that in vivo levels of ammonia in the brain directly affect the rate of mitochondrial O2·- production.
Collapse
Affiliation(s)
- Elena A Kosenko
- Institute of Theoretical and Experimental Biophysics, Pushchino, Russia.
| | | | - Gubidat A Alilova
- Institute of Theoretical and Experimental Biophysics, Pushchino, Russia
| | - Carmina Montoliu
- Fundación Investigación Hospital Clínico, Instituto Investigación Sanitaria-INCLIVA, Valencia, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Gjumrakch Aliev
- GALLY International Biomedical Research Institute Inc., 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; School of Health Science and Healthcare Administration, University of Atlanta, E. Johns Crossing, #175, Johns Creek, GA 30097, USA.
| | - Yury G Kaminsky
- Institute of Theoretical and Experimental Biophysics, Pushchino, Russia
| |
Collapse
|
16
|
Liu W, Xu Z, Yang T, Deng Y, Xu B, Feng S. Tea Polyphenols Protect Against Methylmercury-Induced Cell Injury in Rat Primary Cultured Astrocytes, Involvement of Oxidative Stress and Glutamate Uptake/Metabolism Disorders. Mol Neurobiol 2016; 53:2995-3009. [PMID: 25952541 DOI: 10.1007/s12035-015-9161-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/25/2015] [Indexed: 02/06/2023]
Abstract
Methylmercury (MeHg) is an extremely dangerous environmental contaminant, accumulating preferentially in CNS and causing a series of cytotoxic effects. However, the precise mechanisms are still incompletely understood. The current study explored the mechanisms that contribute to MeHg-induced cell injury focusing on the oxidative stress and Glu uptake/metabolism disorders in rat primary cultured astrocytes. Moreover, the neuroprotective effects of tea polyphenols (TP), a natural antioxidant, against MeHg cytotoxicity were also investigated. Astrocytes were exposed to 0, 2.5, 5, 10, and 20 μM MeHgCl for 6-30 h, or pretreated with 50, 100, 200, and 400 μM TP for 1-12 h; cell viability and LDH release were then determined. For further experiments, 50, 100, and 200 μM of TP pretreatment for 6 h followed by 10 μM MeHgCl for 24 h were performed for the examination of the responses of astrocytes, specifically addressing NPSH levels, ROS generation, ATPase activity, the expressions of Nrf2 pathway as well as Glu metabolism enzyme GS and Glu transporters (GLAST and GLT-1). Exposure of MeHg resulted in damages of astrocytes, which were shown by a loss of cell viability, and supported by high levels of LDH release, morphological changes, apoptosis rates, and NPSH depletion. In addition, astrocytes were sensitive to MeHg-mediated oxidative stress, a finding that is consistent with ROS overproduction; Nrf2 as well as its downstream genes HO-1 and γ-GCSh were markedly upregulated. Moreover, MeHg significantly inhibited GS activity, as well as expressions of GS, GLAST, and GLT-1. On the contrary, pretreatment with TP presented a concentration-dependent prevention against MeHg-mediated cytotoxic effects of astrocytes. In conclusion, the findings clearly indicated that MeHg aggravated oxidative stress and Glu uptake/metabolism dysfunction in astrocytes. TP possesses some abilities to prevent MeHg cytotoxicity through its antioxidative properties.
Collapse
Affiliation(s)
- Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China.
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China
| | - Shu Feng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning province, China
| |
Collapse
|
17
|
Abstract
Cerebellar disorders trigger the symptoms of movement problems, imbalance, incoordination, and frequent fall. Cerebellar disorders are shown in various CNS illnesses including a drinking disorder called alcoholism. Alcoholism is manifested as an inability to control drinking in spite of adverse consequences. Human and animal studies have shown that cerebellar symptoms persist even after complete abstinence from drinking. In particular, the abrupt termination (ethanol withdrawal) of long-term excessive ethanol consumption has shown to provoke a variety of neuronal and mitochondrial damage to the cerebellum. Upon ethanol withdrawal, excitatory neurotransmitter molecules such as glutamate are overly released in brain areas including cerebellum. This is particularly relevant to the cerebellar neuronal network as glutamate signals are projected to Purkinje neurons through granular cells that are the most populated neuronal type in CNS. This excitatory neuronal signal may be elevated by ethanol withdrawal stress, which promotes an increase in intracellular Ca(2+) level and a decrease in a Ca(2+)-binding protein, both of which result in the excessive entry of Ca(2+) to the mitochondria. Subsequently, mitochondria undergo a prolonged opening of mitochondrial permeability transition pore and the overproduction of harmful free radicals, impeding adenosine triphosphate (ATP)-generating function. This in turn provokes the leakage of mitochondrial molecule cytochrome c to the cytosol, which triggers a cascade of adverse cytosol reactions. Upstream to this pathway, cerebellum under the condition of ethanol withdrawal has shown aberrant gene modifications through altered DNA methylation, histone acetylation, or microRNA expression. Interplay between these events and molecules may result in functional damage to cerebellar mitochondria and consequent neuronal degeneration, thereby contributing to motoric deficit. Mitochondria-targeting research may help develop a powerful new therapy to manage cerebellar disorders associated with hyperexcitatory CNS disorders like ethanol withdrawal.
Collapse
Affiliation(s)
- Marianna E Jung
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107-2699, USA,
| |
Collapse
|
18
|
Wang H, Chen B, Zhu S, Yu X, He M, Hu B. Chip-Based Magnetic Solid-Phase Microextraction Online Coupled with MicroHPLC–ICPMS for the Determination of Mercury Species in Cells. Anal Chem 2015; 88:796-802. [DOI: 10.1021/acs.analchem.5b03130] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Han Wang
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| | - Beibei Chen
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| | - Siqi Zhu
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xiaoxiao Yu
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| | - Man He
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Key Laboratory of Analytical Chemistry
for Biology and Medicine, Ministry of Education, Department
of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
19
|
Mailloux RJ, Yumvihoze E, Chan HM. Superoxide anion radical (O2(-)) degrades methylmercury to inorganic mercury in human astrocytoma cell line (CCF-STTG1). Chem Biol Interact 2015; 239:46-55. [PMID: 26111762 DOI: 10.1016/j.cbi.2015.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 06/11/2015] [Accepted: 06/19/2015] [Indexed: 10/23/2022]
Abstract
Methylmercury (MeHg) is a global pollutant that is affecting the health of millions of people worldwide. However, the mechanism of MeHg toxicity still remains somewhat elusive and there is no treatment. It has been known for some time that MeHg can be progressively converted to inorganic mercury (iHg) in various tissues including the brain. Recent work has suggested that cleavage of the carbon-metal bond in MeHg in a biological environment is facilitated by reactive oxygen species (ROS). However, the oxyradical species that actually mediates this process has not been identified. Here, we provide evidence that superoxide anion radical (O2(-)) can convert MeHg to iHg. The calculated second-order rate constant for the degradation of 1μM MeHg by O2(-) generated by xanthine/xanthine oxidase was calculated to be 2×10(5)M(-1)s(-1). We were also able to show that this bioconversion can proceed in intact CCF-STTG1 human astrocytoma cells exposed to paraquat (PQ), a O2(-) generating viologen. Notably, exposure of cells to increasing amounts of PQ led to a dose dependent increase in both MeHg and iHg. Indeed, a 24h exposure to 500μM PQ induced a ∼13-fold and ∼18-fold increase in intracellular MeHg and iHg respectively. These effects were inhibited by superoxide dismutase mimetic MnTBAP. In addition, we also observed that a 24h exposure to a biologically relevant concentration of MeHg (1μM) did not induce cell death, oxidative stress, or even changes in cellular O2(-) and H2O2. However, co-exposure to PQ enhanced MeHg toxicity which was associated with a robust increase in cell death and oxidative stress. Collectively our results show that O2(-) can bioconvert MeHg to iHg in vitro and in intact cells exposed to conditions that simulate high intracellular O2(-) production. In addition, we show for the first time that O2(-) mediated degradation of MeHg to iHg enhances the toxicity of MeHg by facilitating an accumulation of both MeHg and iHg in the intracellular environment.
Collapse
Affiliation(s)
- Ryan J Mailloux
- University of Ottawa, Department of Biology, Center for Advanced Research in Environmental Genomics, Ottawa, ON K1N 6N5, Canada
| | - Emmanuel Yumvihoze
- University of Ottawa, Department of Biology, Center for Advanced Research in Environmental Genomics, Ottawa, ON K1N 6N5, Canada
| | - Hing Man Chan
- University of Ottawa, Department of Biology, Center for Advanced Research in Environmental Genomics, Ottawa, ON K1N 6N5, Canada.
| |
Collapse
|
20
|
Obiorah M, McCandlish E, Buckley B, DiCicco-Bloom E. Hippocampal developmental vulnerability to methylmercury extends into prepubescence. Front Neurosci 2015; 9:150. [PMID: 26029035 PMCID: PMC4429234 DOI: 10.3389/fnins.2015.00150] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 04/14/2015] [Indexed: 12/27/2022] Open
Abstract
The developing brain is sensitive to environmental toxicants such as methylmercury (MeHg), to which humans are exposed via contaminated seafood. Prenatal exposure in children is associated with learning, memory and IQ deficits, which can result from hippocampal dysfunction. To explore underlying mechanisms, we have used the postnatal day (P7) rat to model the third trimester of human gestation. We previously showed that a single low exposure (0.6 μg/gbw) that approaches human exposure reduced hippocampal neurogenesis in the dentate gyrus (DG) 24 h later, producing later proliferation and memory deficits in adolescence. Yet, the vulnerable stem cell population and period of developmental vulnerability remain undefined. In this study, we find that P7 exposure of stem cells has long-term consequences for adolescent neurogenesis. It reduced the number of mitotic S-phase cells (BrdU), especially those in the highly proliferative Tbr2+ population, and immature neurons (Doublecortin) in adolescence, suggesting partial depletion of the later stem cell pool. To define developmental vulnerability to MeHg in prepubescent (P14) and adolescent (P21) rats, we examined acute 24 h effects of MeHg exposure on mitosis and apoptosis. We found that low exposure did not adversely impact neurogenesis at either age, but that a higher exposure (5 μg/gbw) at P14 reduced the total number of neural stem cells (Sox2+) by 23% and BrdU+ cells by 26% in the DG hilus, suggesting that vulnerability diminishes with age. To determine whether these effects reflect changes in MeHg transfer across the blood brain barrier (BBB), we assessed Hg content in the hippocampus after peripheral injection and found that similar levels (~800 ng/gm) were obtained at 24 h at both P14 and P21, declining in parallel, suggesting that changes in vulnerability depend more on local tissue and cellular mechanisms. Together, we show that MeHg vulnerability declines with age, and that early exposure impairs later neurogenesis in older juveniles.
Collapse
Affiliation(s)
- Maryann Obiorah
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Brian Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey Piscataway, NJ, USA ; Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey New Brunswick, NJ, USA
| |
Collapse
|
21
|
Obiorah M, McCandlish E, Buckley B, DiCicco-Bloom E. Hippocampal developmental vulnerability to methylmercury extends into prepubescence. Front Neurosci 2015. [PMID: 26029035 DOI: 10.3389/fnins.2015.00150/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The developing brain is sensitive to environmental toxicants such as methylmercury (MeHg), to which humans are exposed via contaminated seafood. Prenatal exposure in children is associated with learning, memory and IQ deficits, which can result from hippocampal dysfunction. To explore underlying mechanisms, we have used the postnatal day (P7) rat to model the third trimester of human gestation. We previously showed that a single low exposure (0.6 μg/gbw) that approaches human exposure reduced hippocampal neurogenesis in the dentate gyrus (DG) 24 h later, producing later proliferation and memory deficits in adolescence. Yet, the vulnerable stem cell population and period of developmental vulnerability remain undefined. In this study, we find that P7 exposure of stem cells has long-term consequences for adolescent neurogenesis. It reduced the number of mitotic S-phase cells (BrdU), especially those in the highly proliferative Tbr2+ population, and immature neurons (Doublecortin) in adolescence, suggesting partial depletion of the later stem cell pool. To define developmental vulnerability to MeHg in prepubescent (P14) and adolescent (P21) rats, we examined acute 24 h effects of MeHg exposure on mitosis and apoptosis. We found that low exposure did not adversely impact neurogenesis at either age, but that a higher exposure (5 μg/gbw) at P14 reduced the total number of neural stem cells (Sox2+) by 23% and BrdU+ cells by 26% in the DG hilus, suggesting that vulnerability diminishes with age. To determine whether these effects reflect changes in MeHg transfer across the blood brain barrier (BBB), we assessed Hg content in the hippocampus after peripheral injection and found that similar levels (~800 ng/gm) were obtained at 24 h at both P14 and P21, declining in parallel, suggesting that changes in vulnerability depend more on local tissue and cellular mechanisms. Together, we show that MeHg vulnerability declines with age, and that early exposure impairs later neurogenesis in older juveniles.
Collapse
Affiliation(s)
- Maryann Obiorah
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Brian Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers The State University of New Jersey Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey Piscataway, NJ, USA ; Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers The State University of New Jersey New Brunswick, NJ, USA
| |
Collapse
|
22
|
Glutathione-Dependent Detoxification Processes in Astrocytes. Neurochem Res 2014; 40:2570-82. [PMID: 25428182 DOI: 10.1007/s11064-014-1481-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/10/2014] [Accepted: 11/15/2014] [Indexed: 01/17/2023]
Abstract
Astrocytes have a pivotal role in brain as partners of neurons in homeostatic and metabolic processes. Astrocytes also protect other types of brain cells against the toxicity of reactive oxygen species and are considered as first line of defence against the toxic potential of xenobiotics. A key component in many of the astrocytic detoxification processes is the tripeptide glutathione (GSH) which serves as electron donor in the GSH peroxidase-catalyzed reduction of peroxides. In addition, GSH is substrate in the detoxification of xenobiotics and endogenous compounds by GSH-S-transferases which generate GSH conjugates that are efficiently exported from the cells by multidrug resistance proteins. Moreover, GSH reacts with the reactive endogenous carbonyls methylglyoxal and formaldehyde to intermediates which are substrates of detoxifying enzymes. In this article we will review the current knowledge on the GSH metabolism of astrocytes with a special emphasis on GSH-dependent detoxification processes.
Collapse
|
23
|
Patel E, Reynolds M. Methylmercury impairs motor function in early development and induces oxidative stress in cerebellar granule cells. Toxicol Lett 2013; 222:265-72. [DOI: 10.1016/j.toxlet.2013.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/31/2013] [Accepted: 08/03/2013] [Indexed: 01/20/2023]
|
24
|
Cordero-Herrera I, Cuello S, Goya L, Madrid Y, Bravo L, Cámara C, Ramos S. Molecular mechanisms involved in the protective effect of selenocystine against methylmercury-induced cell death in human HepG2 cells. Food Chem Toxicol 2013; 59:554-63. [DOI: 10.1016/j.fct.2013.06.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 12/22/2022]
|
25
|
Low in situ expression of antioxidative enzymes in rat cerebellar granular cells susceptible to methylmercury. Arch Toxicol 2013; 88:109-13. [DOI: 10.1007/s00204-013-1089-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 06/20/2013] [Indexed: 10/26/2022]
|
26
|
Syversen T, Kaur P. The toxicology of mercury and its compounds. J Trace Elem Med Biol 2012; 26:215-26. [PMID: 22658719 DOI: 10.1016/j.jtemb.2012.02.004] [Citation(s) in RCA: 238] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/07/2012] [Indexed: 12/27/2022]
Abstract
A concentrated review on the toxicology of inorganic mercury together with an extensive review on the neurotoxicology of methylmercury is presented. The challenges of using inorganic mercury in dental amalgam are reviewed both regarding the occupational exposure and the possible health problems for the dental patients. The two remaining "mysteries" of methylmercury neurotoxicology are also being reviewed; the cellular selectivity and the delayed onset of symptoms. The relevant literature on these aspects has been discussed and some suggestions towards explaining these observations have been presented.
Collapse
Affiliation(s)
- Tore Syversen
- Norwegian University of Science and Technology, Department of Neuroscience, Trondheim, Norway.
| | | |
Collapse
|
27
|
Petroni D, Tsai J, Agrawal K, Mondal D, George W. Low-dose methylmercury-induced oxidative stress, cytotoxicity, and tau-hyperphosphorylation in human neuroblastoma (SH-SY5Y) cells. ENVIRONMENTAL TOXICOLOGY 2012; 27:549-555. [PMID: 21254321 DOI: 10.1002/tox.20672] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 10/12/2010] [Accepted: 10/15/2010] [Indexed: 05/28/2023]
Abstract
Acute neurotoxic effects of high-dose methylmercury (MeHg) in humans have been well documented in the scientific literature. However, low-dose effects are less well described. This study was designed to evaluate the effects of low-dose MeHg (<100 nM) on human brain cells in a tissue culture model. Neuroblastoma (NB) cells (SH-SY5Y) were used in the cell culture model to study low-dose effects of MeHg on cell growth, cell survival, reactive oxygen species (ROS), and the phosphorylation of tau protein, as a measure of potential markers of cellular events associated with tauopathies. When cells were incubated in culture with MeHg (50 and 100 nM), there were significant decreases in cell viability as well as significant increase in ROS generation as determined by fluorescent dye analysis (H(2)DCFDA). Furthermore, a concomitant decrease in glutathione levels to 25% of control was observed at both 50 and 100 nM MeHg. In addition, the level of phosphorylated tau was significantly increased after treatment at both 50 and 100 nM MeHg, compared with controls. Pretreatment of NB cells with the antioxidant, N-acetylcysteine (1.25 mM) and the calpain inhibitor, MDL-28170 (10 μM), significantly attenuated the effects of MeHg (50 and 100 nM) on cell viability as well as on tau phosphorylation. These results indicate that low-dose MeHg toxicity may be related to an induction of tau phosphorylation through an oxidative stress-dependent mechanism and that blockade of this pathway may attenuate the toxic effects of MeHg.
Collapse
Affiliation(s)
- Daniel Petroni
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
28
|
Riluzole-triggered GSH synthesis via activation of glutamate transporters to antagonize methylmercury-induced oxidative stress in rat cerebral cortex. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:534705. [PMID: 22966415 PMCID: PMC3432391 DOI: 10.1155/2012/534705] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/25/2012] [Accepted: 07/08/2012] [Indexed: 11/18/2022]
Abstract
OBJECTIVE This study was to evaluate the effect of riluzole on methylmercury- (MeHg-) induced oxidative stress, through promotion of glutathione (GSH) synthesis by activating of glutamate transporters (GluTs) in rat cerebral cortex. METHODS Eighty rats were randomly assigned to four groups, control group, riluzole alone group, MeHg alone group, and riluzole + MeHg group. The neurotoxicity of MeHg was observed by measuring mercury (Hg) absorption, pathological changes, and cell apoptosis of cortex. Oxidative stress was evaluated via determining reactive oxygen species (ROS), 8-hydroxy-2-deoxyguanosine (8-OHdG), malondialdehyde (MDAs), carbonyl, sulfydryl, and GSH in cortex. Glutamate (Glu) transport was studied by measuring Glu, glutamine (Gln), mRNA, and protein of glutamate/aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1). RESULT (1) MeHg induced Hg accumulation, pathological injury, and apoptosis of cortex; (2) MeHg increased ROS, 8-OHdG, MDA, and carbonyl, and inhibited sulfydryl and GSH; (3) MeHg elevated Glu, decreased Gln, and downregulated GLAST and GLT-1 mRNA expression and protein levels; (4) riluzole antagonized MeHg-induced downregulation of GLAST and GLT-1 function and expression, GSH depletion, oxidative stress, pathological injury, and apoptosis obviously. CONCLUSION Data indicate that MeHg administration induced oxidative stress in cortex and that riluzole could antagonize this situation through elevation of GSH synthesis by activating of GluTs.
Collapse
|
29
|
Falluel-Morel A, Lin L, Sokolowski K, McCandlish E, Buckley B, DiCicco-Bloom E. N-acetyl cysteine treatment reduces mercury-induced neurotoxicity in the developing rat hippocampus. J Neurosci Res 2012; 90:743-50. [PMID: 22420031 DOI: 10.1002/jnr.22819] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mercury is an environmental toxicant that can disrupt brain development. However, although progress has been made in defining its neurotoxic effects, we know far less about available therapies that can effectively protect the brain in exposed individuals. We previously developed an animal model in which we defined the sequence of events underlying neurotoxicity: Methylmercury (MeHg) injection in postnatal rat acutely induced inhibition of mitosis and stimulated apoptosis in the hippocampus, which later resulted in intermediate-term deficits in structure size and cell number. N-acetyl cysteine (NAC) is the N-acetyl derivative of L-cysteine used clinically for treatment of drug intoxication. Here, based on its known efficacy in promoting MeHg urinary excretion, we evaluated NAC for protective effects in the developing brain. In immature neurons and precursors, MeHg (3 μM) induced a >50% decrease in DNA synthesis at 24 hr, an effect that was completely blocked by NAC coincubation. In vivo, injection of MeHg (5 μg/g bw) into 7-day-old rats induced a 22% decrease in DNA synthesis in whole hippocampus and a fourfold increase in activated caspase-3-immunoreactive cells at 24 hr and reduced total cell numbers by 13% at 3 weeks. Treatment of MeHg-exposed rats with repeated injections of NAC abolished MeHg toxicity. NAC prevented the reduction in DNA synthesis and the marked increase in caspase-3 immunoreactivity. Moreover, the intermediate-term decrease in hippocampal cell number provoked by MeHg was fully blocked by NAC. Altogether these results suggest that MeHg toxicity in the perinatal brain can be ameliorated by using NAC, opening potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Anthony Falluel-Morel
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
30
|
Glutathione-mediated neuroprotection against methylmercury neurotoxicity in cortical culture is dependent on MRP1. Neurotoxicology 2012; 33:476-81. [DOI: 10.1016/j.neuro.2012.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 02/15/2012] [Accepted: 03/11/2012] [Indexed: 11/19/2022]
|
31
|
Differential protein expression of hepatic cells associated with MeHg exposure: deepening into the molecular mechanisms of toxicity. Anal Bioanal Chem 2012; 404:315-24. [DOI: 10.1007/s00216-012-6042-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 04/03/2012] [Accepted: 04/10/2012] [Indexed: 01/09/2023]
|
32
|
Caito S, Fretham S, Martinez-Finley E, Chakraborty S, Avila D, Chen P, Aschner M. Genome-Wide Analyses of Metal Responsive Genes in Caenorhabditis elegans. Front Genet 2012; 3:52. [PMID: 22514555 PMCID: PMC3322339 DOI: 10.3389/fgene.2012.00052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 03/20/2012] [Indexed: 11/22/2022] Open
Abstract
Metals are major contaminants that influence human health. Many metals have physiologic roles, but excessive levels can be harmful. Advances in technology have made toxicogenomic analyses possible to characterize the effects of metal exposure on the entire genome. Much of what is known about cellular responses to metals has come from mammalian systems; however the use of non-mammalian species is gaining wider attention. Caenorhabditis elegans is a small round worm whose genome has been fully sequenced and its development from egg to adult is well characterized. It is an attractive model for high throughput screens due to its short lifespan, ease of genetic mutability, low cost, and high homology with humans. Research performed in C. elegans has led to insights in apoptosis, gene expression, and neurodegeneration, all of which can be altered by metal exposure. Additionally, by using worms one can potentially study mechanisms that underline differential responses to metals in nematodes and humans, allowing for identification of novel pathways and therapeutic targets. In this review, toxicogenomic studies performed in C. elegans exposed to various metals will be discussed, highlighting how this non-mammalian system can be utilized to study cellular processes and pathways induced by metals. Recent work focusing on neurodegeneration in Parkinson’s disease will be discussed as an example of the usefulness of genetic screens in C. elegans and the novel findings that can be produced.
Collapse
Affiliation(s)
- Samuel Caito
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Cuello S, Ximénez-Embún P, Ruppen I, Schonthaler HB, Ashman K, Madrid Y, Luque-Garcia JL, Cámara C. Analysis of protein expression in developmental toxicity induced by MeHg in zebrafish. Analyst 2012; 137:5302-11. [DOI: 10.1039/c2an35913h] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Kaur P, Aschner M, Syversen T. Biochemical factors modulating cellular neurotoxicity of methylmercury. J Toxicol 2011; 2011:721987. [PMID: 21941541 PMCID: PMC3177097 DOI: 10.1155/2011/721987] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 06/28/2011] [Accepted: 07/13/2011] [Indexed: 11/30/2022] Open
Abstract
Methylmercury (MeHg), an environmental toxicant primarily found in fish and seafood, poses a dilemma to both consumers and regulatory authorities, given the nutritional benefits of fish consumption versus the possible adverse neurological damage. Several studies have shown that MeHg toxicity is influenced by a number of biochemical factors, such as glutathione (GSH), fatty acids, vitamins, and essential elements, but the cellular mechanisms underlying these complex interactions have not yet been fully elucidated. The objective of this paper is to outline the cellular response to dietary nutrients, as well as to describe the neurotoxic exposures to MeHg. In order to determine the cellular mechanism(s) of toxicity, the effect of pretreatment with biochemical factors (e.g., N-acetyl cysteine, (NAC); diethyl maleate, (DEM); docosahexaenoic acid, (DHA); selenomethionine, SeM; Trolox) and MeHg treatment on intercellular antioxidant status, MeHg content, and other endpoints was evaluated. This paper emphasizes that the protection against oxidative stress offered by these biochemical factors is among one of the major mechanisms responsible for conferring neuroprotection. It is therefore critical to ascertain the cellular mechanisms associated with various dietary nutrients as well as to determine the potential effects of neurotoxic exposures for accurately assessing the risks and benefits associated with fish consumption.
Collapse
Affiliation(s)
- Parvinder Kaur
- Department of Neuroscience, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Michael Aschner
- Departments of Pediatrics and Pharmacology and The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, B-3307 Medical Center North, 1162 21st Avenue, Nashville, TN 37232-2495, USA
| | - Tore Syversen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Olav Kyrres Gate 3, 7489 Trondheim, Norway
| |
Collapse
|
35
|
Masmoudi-Kouki O, Douiri S, Hamdi Y, Kaddour H, Bahdoudi S, Vaudry D, Basille M, Leprince J, Fournier A, Vaudry H, Tonon MC, Amri M. Pituitary adenylate cyclase-activating polypeptide protects astroglial cells against oxidative stress-induced apoptosis. J Neurochem 2011; 117:403-11. [PMID: 21244427 DOI: 10.1111/j.1471-4159.2011.07185.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oxidative stress, associated with a variety of disorders including neurodegenerative diseases, results from accumulation of reactive oxygen species (ROS). Oxidative stress is not only responsible for neuron apoptosis, but can also provoke astroglial cell death. Numerous studies indicate that pituitary adenylate cyclase-activating polypeptide (PACAP) promotes neuron survival, but nothing is known regarding the action of PACAP on astroglial cell survival. Thus, the purpose of the present study was to investigate the potential glioprotective effect of PACAP on H(2)O(2)-induced astrocyte death. Pre-treatment of cultured rat astrocytes with nanomolar concentrations of PACAP prevented cell death provoked by H(2)O(2) (300 μM), whereas vasoactive intestinal polypeptide was devoid of protective activity. The effect of PACAP on astroglial cell survival was abolished by the type 1 PACAP receptor antagonist, PACAP6-38. The protective action of PACAP was blocked by the protein kinase A inhibitor H89, the protein kinase C inhibitor chelerythrine and the mitogen-activated protein (MAP)-kinase kinase (MEK) inhibitor U0126. PACAP stimulated glutathione formation, and blocked H(2)O(2)-evoked ROS accumulation and glutathione content reduction. In addition, PACAP prevented the decrease of mitochondrial activity and caspase 3 activation induced by H(2)O(2). Taken together, these data indicate for the first time that PACAP, acting through type 1 PACAP receptor, exerts a potent protective effect against oxidative stress-induced astrocyte death. The anti-apoptotic activity of PACAP on astrocytes is mediated through the protein kinase A, protein kinase C and MAPK transduction pathways, and can be accounted for by inhibition of ROS-induced mitochondrial dysfunctions and caspase 3 activation.
Collapse
Affiliation(s)
- Olfa Masmoudi-Kouki
- Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, Tunis, Tunisia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sidoryk-Wegrzynowicz M, Wegrzynowicz M, Lee E, Bowman AB, Aschner M. Role of astrocytes in brain function and disease. Toxicol Pathol 2010; 39:115-23. [PMID: 21075920 DOI: 10.1177/0192623310385254] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astrocytes assume multiple roles in maintaining an optimally suited milieu for neuronal function. Select astrocytic functions include the maintenance of redox potential, the production of trophic factors, the regulation of neurotransmitter and ion concentrations, and the removal of toxins and debris from the cerebrospinal fluid (CSF). Impairments in these and other functions, as well as physiological reactions of astrocytes to injury, can trigger or exacerbate neuronal dysfunction. This review addresses select metabolic interactions between neurons and astrocytes and emphasizes the role of astrocytes in mediating and amplifying the progression of several neurodegenerative disorders, such as Parkinson's disease (PD), hepatic encephalopathy (HE), hyperammonemia (HA), Alzheimer's disease (AD), and ischemia.
Collapse
|
37
|
Zieminska E, Toczylowska B, Stafiej A, Lazarewicz JW. Low molecular weight thiols reduce thimerosal neurotoxicity in vitro: modulation by proteins. Toxicology 2010; 276:154-63. [PMID: 20696200 DOI: 10.1016/j.tox.2010.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/16/2010] [Accepted: 07/31/2010] [Indexed: 01/19/2023]
Abstract
Thimerosal (TH), an ethylmercury complex of thiosalicylic acid has been used as preservative in vaccines. In vitro neurotoxicity of TH at high nM concentrations has been reported. Although a number of toxicological experiments demonstrated high affinity of mercury to thiol groups of the extracellular amino acids and proteins that may decrease concentration of free TH in the organism, less is known about the role of interactions between proteins and amino acids in protection against TH neurotoxicity. In the present study we examined whether the presence of serum proteins and of l-cysteine (Cys), d,l-homocysteine (Hcy), N-acetyl cysteine (NAC), l-methionine (Met) and glutathione (GSH) in the incubation medium affects the TH-induced changes in the viability, the intracellular levels of calcium and zinc and mitochondrial membrane potential in primary cultures of rat cerebellar granule cells. The cells were exposed to 500 nM TH for 48 h or to 15-25 μM TH for 10 min. Our results demonstrated a decrease in the cells viability evoked by TH, which could be prevented partially by serum proteins, albumin or in a dose-dependent manner by 60, 120 or 600 μM Cys, Hcy, NAC and GSH, but not by Met. This neuroprotection was less pronounced in the presence of proteins. Incubation of neurons with TH also induced the rise in the intracellular calcium and zinc concentration and decrease in mitochondrial membrane potential, and these effects were abolished by all the sulfur containing compounds studied and administered at 600 μM concentration, except Met. The loss of the ethylmercury moiety from TH as a result of interaction with thiols studied was monitored by (1)H NMR spectroscopy. This extracellular process may be responsible for the neuroprotection seen in the cerebellar cell cultures, but also provides a molecular pathway for redistribution of TH-derived toxic ethylmercury in the organism. In conclusion, these results confirmed that proteins and sulfur-containing amino acids applied separately reduce TH neurotoxicity, while their combination modulates in more complex way neuronal survival in the presence of TH.
Collapse
Affiliation(s)
- E Zieminska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland.
| | | | | | | |
Collapse
|
38
|
Posser T, Dunkley PR, Dickson PW, Franco JL. Human neuroblastoma cells transfected with tyrosine hydroxylase gain increased resistance to methylmercury-induced cell death. Toxicol In Vitro 2010; 24:1498-503. [PMID: 20650311 DOI: 10.1016/j.tiv.2010.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 07/12/2010] [Accepted: 07/15/2010] [Indexed: 11/26/2022]
Abstract
In a previous study we demonstrated that human neuroblastoma SH-SY5Y cells transfected with human tyrosine hydroxylase isoform 1 (SH+TH cells) were substantially more resistant to cell death induced by pro-oxidants than wild type SH-SY5Y cells (SH cells). In the present communication we used methylmercury as a model of cell stress in order to test whether SH+TH cells would behave in a similar manner in response to this stressor. Incubation with methylmercury (0.1-3 microM) for 24h caused a significant reduction in cell viability and increased apoptotic markers in both cell types. However, the effects were significantly reduced in the SH+TH cells when compared to the SH cells. Activation of p38(MAPK) was also reduced in the SH+TH compared to the SH cells after methylmercury exposure. Since p38(MAPK) is known to participate in signal transduction pathways during cell stress, our data suggest that SH+TH cells develop an increased resistance to environmental stress caused by neurotoxins such as methylmercury. In conclusion our results show that insertion of the human TH gene in cells that originally do not express this protein leads to alterations in cell homeostasis and triggers defense mechanisms against pro-oxidative insults.
Collapse
Affiliation(s)
- Thaís Posser
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | | | | | | |
Collapse
|
39
|
Wang X, Michaelis EK. Selective neuronal vulnerability to oxidative stress in the brain. Front Aging Neurosci 2010; 2:12. [PMID: 20552050 PMCID: PMC2874397 DOI: 10.3389/fnagi.2010.00012] [Citation(s) in RCA: 443] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 03/11/2010] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress (OS), caused by the imbalance between the generation and detoxification of reactive oxygen and nitrogen species (ROS/RNS), plays an important role in brain aging, neurodegenerative diseases, and other related adverse conditions, such as ischemia. While ROS/RNS serve as signaling molecules at physiological levels, an excessive amount of these molecules leads to oxidative modification and, therefore, dysfunction of proteins, nucleic acids, and lipids. The response of neurons to this pervasive stress, however, is not uniform in the brain. While many brain neurons can cope with a rise in OS, there are select populations of neurons in the brain that are vulnerable. Because of their selective vulnerability, these neurons are usually the first to exhibit functional decline and cell death during normal aging, or in age-associated neurodegenerative diseases, such as Alzheimer's disease. Understanding the molecular and cellular mechanisms of selective neuronal vulnerability (SNV) to OS is important in the development of future intervention approaches to protect such vulnerable neurons from the stresses of the aging process and the pathological states that lead to neurodegeneration. In this review, the currently known molecular and cellular factors that contribute to SNV to OS are summarized. Included among the major underlying factors are high intrinsic OS, high demand for ROS/RNS-based signaling, low ATP production, mitochondrial dysfunction, and high inflammatory response in vulnerable neurons. The contribution to the selective vulnerability of neurons to OS by other intrinsic or extrinsic factors, such as deficient DNA damage repair, low calcium-buffering capacity, and glutamate excitotoxicity, are also discussed.
Collapse
Affiliation(s)
- Xinkun Wang
- Higuchi Biosciences Center, The University of Kansas Lawrence, KS, USA
| | | |
Collapse
|
40
|
Molecular mechanisms of methylmercury-induced cell death in human HepG2 cells. Food Chem Toxicol 2010; 48:1405-11. [PMID: 20226830 DOI: 10.1016/j.fct.2010.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 03/03/2010] [Accepted: 03/08/2010] [Indexed: 12/29/2022]
Abstract
Methylmercury (MeHg) has been suggested to exert cytotoxicity through multiple mechanisms, but the precise biochemical machinery has not been fully defined. This study was aimed at investigating the time-course (0-24h) effect of 2mg/L MeHg on cell death in human HepG2 cells. MeHg decreased cell viability in a time-dependent manner, which was concomitant with increased LDH leakage, reduced GSH levels, CAT activity and altered activity of the antioxidant enzymes GPx and GR at the longest times of incubation (16 and 24h). Activity of the detoxifying enzyme GST was also early enhanced (2h). Caspase-3 activity reached a maximum value at 8h and continued increased up to 24h. This feature was preceded by an enhancement in the caspase-9 activity (2h), whereas caspase-8 activity remained unchanged. MeHg early diminished Bcl-x(L)/Bcl-x(S) ratio and increased levels of the pro-apoptotic Bax and Bad. Moreover, MeHg-induced cytotoxicity was completely inhibited by the antioxidants (GSH and NAC) and notably by the mitochondrial complex I inhibitor rotenone, but not by the NADH oxidase inhibitor DPI. In summary, MeHg induced an oxidative stress responsible for apoptosis in HepG2 cells through direct activation of the caspase cascade and altered the cellular antioxidant and detoxificant enzymatic system to later provoke necrosis at later stages.
Collapse
|
41
|
The in vitro effects of selenomethionine on methylmercury-induced neurotoxicity. Toxicol In Vitro 2009; 23:378-85. [DOI: 10.1016/j.tiv.2008.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 11/14/2008] [Accepted: 12/19/2008] [Indexed: 11/23/2022]
|
42
|
Genomic and biochemical approaches in the discovery of mechanisms for selective neuronal vulnerability to oxidative stress. BMC Neurosci 2009; 10:12. [PMID: 19228403 PMCID: PMC2677396 DOI: 10.1186/1471-2202-10-12] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Accepted: 02/19/2009] [Indexed: 12/12/2022] Open
Abstract
Background Oxidative stress (OS) is an important factor in brain aging and neurodegenerative diseases. Certain neurons in different brain regions exhibit selective vulnerability to OS. Currently little is known about the underlying mechanisms of this selective neuronal vulnerability. The purpose of this study was to identify endogenous factors that predispose vulnerable neurons to OS by employing genomic and biochemical approaches. Results In this report, using in vitro neuronal cultures, ex vivo organotypic brain slice cultures and acute brain slice preparations, we established that cerebellar granule (CbG) and hippocampal CA1 neurons were significantly more sensitive to OS (induced by paraquat) than cerebral cortical and hippocampal CA3 neurons. To probe for intrinsic differences between in vivo vulnerable (CA1 and CbG) and resistant (CA3 and cerebral cortex) neurons under basal conditions, these neurons were collected by laser capture microdissection from freshly excised brain sections (no OS treatment), and then subjected to oligonucleotide microarray analysis. GeneChip-based transcriptomic analyses revealed that vulnerable neurons had higher expression of genes related to stress and immune response, and lower expression of energy generation and signal transduction genes in comparison with resistant neurons. Subsequent targeted biochemical analyses confirmed the lower energy levels (in the form of ATP) in primary CbG neurons compared with cortical neurons. Conclusion Low energy reserves and high intrinsic stress levels are two underlying factors for neuronal selective vulnerability to OS. These mechanisms can be targeted in the future for the protection of vulnerable neurons.
Collapse
|
43
|
Kaur P, Heggland I, Aschner M, Syversen T. Docosahexaenoic acid may act as a neuroprotector for methylmercury-induced neurotoxicity in primary neural cell cultures. Neurotoxicology 2008; 29:978-87. [DOI: 10.1016/j.neuro.2008.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 06/02/2008] [Accepted: 06/09/2008] [Indexed: 10/21/2022]
|
44
|
Kaur P, Schulz K, Heggland I, Aschner M, Syversen T. The use of fluorescence for detecting MeHg-induced ROS in cell cultures. Toxicol In Vitro 2008; 22:1392-8. [DOI: 10.1016/j.tiv.2008.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/23/2008] [Accepted: 01/31/2008] [Indexed: 11/16/2022]
|
45
|
Li Y, Shi W, Li Y, Zhou Y, Hu X, Song C, Ma H, Wang C, Li Y. Neuroprotective effects of chlorogenic acid against apoptosis of PC12 cells induced by methylmercury. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2008; 26:13-21. [PMID: 21783882 DOI: 10.1016/j.etap.2007.12.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2007] [Revised: 12/20/2007] [Accepted: 12/21/2007] [Indexed: 05/22/2023]
Abstract
Chlorogenic acid (CGA) widely exists in edible and medicinal plants. We aimed to evaluate the effect of CGA on the protection from apoptosis by methylmercury (MeHg) in PC12 cells. Cell viability was evaluated by MTT assay. Apoptosis was assayed by flow cytometry detection. Caspase-3 activity was measured by confocal microscopy. Intracellular GSH levels were determined by bicinchoninic acid protein assay. Intracellular reactive oxygen species (ROS) was assessed by means of chloromethyl-dihydrodichlorofluorescein diacetate. Glutathione peroxidase (GPx) activity was determined by UV. In order to elucidate the action of CGA, the protective effects of CGA were compared to Vit.E. CGA was effective at protecting PC12 cells against MeHg-induced damage in dose-dependent manner. CGA not only suppressed the generation of ROS, the decrease of activity in GPx and the decrease of GSH, but also attenuated caspase-3 activation in PC12 cells by MeHg. CGA eventually protected PC12 cells against MeHg-induced apoptosis. The results highlighted that CGA may exert neuroprotective effects through its antioxidant actions.
Collapse
Affiliation(s)
- Yongjin Li
- Department of Nutrition and Food Hygiene, Health Science Center, Peking University, Beijing 100083, PR China; School of Public Health, Jilin Medical College, Jilin 132013, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Carvalho CM, Chew EH, Hashemy SI, Lu J, Holmgren A. Inhibition of the Human Thioredoxin System. J Biol Chem 2008; 283:11913-23. [DOI: 10.1074/jbc.m710133200] [Citation(s) in RCA: 364] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
47
|
Soltan-Sharifi MS, Mojtahedzadeh M, Najafi A, Reza Khajavi M, Reza Rouini M, Moradi M, Mohammadirad A, Abdollahi M. Improvement by N-acetylcysteine of acute respiratory distress syndrome through increasing intracellular glutathione, and extracellular thiol molecules and anti-oxidant power: evidence for underlying toxicological mechanisms. Hum Exp Toxicol 2008; 26:697-703. [PMID: 17984140 DOI: 10.1177/0960327107083452] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In acute respiratory distress syndrome (ARDS), there is extensive overproduction of free radicals to the extent that endogenous anti-oxidants are overwhelmed, permitting oxidative cell damage. The present study examined the benefit of the anti-oxidant compound N-acetylcysteine (NAC) in the management of ARDS by measuring patient's intracellular glutathione (inside red blood cells) and extracellular (plasma) anti-oxidant defense biomarkers and outcome. Twenty-seven ARDS patients were recruited from the intensive care unit of a teaching Hospital and randomly divided into two groups. Both groups were managed similarly by regular treatments but 17 patients received NAC 150 mg/kg at the first day that followed by 50 mg/kg/day for three days and 10 patients did not receive NAC. Treatment by NAC increased extracellular total anti-oxidant power and total thiol molecules and also improved intracellular glutathione and the outcome of the patients. In conclusion, patients with ARDS are in a deficient oxidant-anti-oxidant balance that can get a significant benefit if supplemented with NAC.
Collapse
|
48
|
Kaur P, Schulz K, Aschner M, Syversen T. Role of Docosahexaenoic Acid in Modulating Methylmercury-Induced Neurotoxicity. Toxicol Sci 2007; 100:423-32. [PMID: 17728287 DOI: 10.1093/toxsci/kfm224] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The effect of docosahexaenoic acid (DHA) in modulating methylmercury (MeHg)-induced neurotoxicity was investigated in C6-glial and B35-neuronal cell lines. Gas chromatography measurements indicated increased DHA content in both the cell lines after 24 h supplementation. Mitochondrial activity evaluated by 3-(4, 5-dimethylthiazol-2-yl)-2, 5 diphenyltetrazolium bromide (MTT) reduction indicated that 10 microM MeHg treatment for 50 min led to a significant (p < 0.001) and similar decrease in MTT activity in both the cell lines. However, DHA pretreatment led to more pronounced depletion (p < 0.05) in the MTT activity in C6 cells as compared to B35 cells. The depletion of glutathione (GSH) content measured with the fluorescent indicator monochlorobimane was more apparent (p < 0.001) in C6 cells treated with DHA and MeHg. The amount of reactive oxygen species (ROS) detected with the fluorescent indicator -- chloromethyl derivative of dichloro dihydro fluorescein diacetate (CMH(2)DCFDA) -- indicated a fourfold increase in C6 cells (p < 0.001) as compared to twofold increase in B35 cells (p < 0.001) upon DHA and MeHg exposure. However, the cell-associated MeHg measurement using (14)C-labeled MeHg indicated a decrease (p < 0.05) in MeHg accumulation upon DHA exposure in both the cell lines. These findings provide experimental evidence that although pretreatment with DHA reduces cell-associated MeHg, it causes an increased ROS (p < 0.001) and GSH depletion (p < 0.05) in C6 cells.
Collapse
Affiliation(s)
- Parvinder Kaur
- Department of Neuroscience, Norwegian University of Science and Technology, N-7489, Trondheim, Norway.
| | | | | | | |
Collapse
|
49
|
Linares V, Sánchez DJ, Bellés M, Albina L, Gómez M, Domingo JL. Pro-oxidant effects in the brain of rats concurrently exposed to uranium and stress. Toxicology 2007; 236:82-91. [PMID: 17493736 DOI: 10.1016/j.tox.2007.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 03/31/2007] [Accepted: 04/02/2007] [Indexed: 11/23/2022]
Abstract
Metal toxicity may be associated with increased rates of reactive oxygen species (ROS) generation within the central nervous system (CNS). Although the kidney is the main target organ for uranium (U) toxicity, this metal can also accumulate in brain. In this study, we investigated the modifications on endogenous antioxidant capacity and oxidative damage in several areas of the brain of U-exposed rats. Eight groups of adult male rats received uranyl acetate dihydrate (UAD) in the drinking water at 0, 10, 20, and 40 mg/kg/day for 3 months. Animals in four groups were concurrently subjected to restraint stress during 2h/day throughout the study. At the end of the experimental period, cortex, hippocampus and cerebellum were removed and processed to examine the following stress markers: reduced glutathione (GSH), oxidized glutathione (GSSG), glutathione reductase (GR), glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT), thiobarbituric acid reactive substances (TBARS), as well as U concentrations. The results show that U significantly accumulated in hippocampus, cerebellum and cortex after 3 months of exposure. Moreover, UAD exposure promoted oxidative stress in these cerebral tissues. In cortex and cerebellum, TBARS levels were positively correlated with the U content, while in cerebellum GSSG and GSH levels were positively and negatively correlated, respectively, with U concentrations. In hippocampus, CAT and SOD activities were positively correlated with U concentration. The present results suggest that chronic oral exposure to UAD can cause progressive perturbations on physiological brain levels of oxidative stress markers. Although at the current UAD doses restraint scarcely showed additional adverse effects, its potential influence should not be underrated.
Collapse
Affiliation(s)
- Victoria Linares
- Laboratory of Toxicology and Environmental Health, Rovira i Virgili University, San Lorenzo 21, 43201 Reus, Spain
| | | | | | | | | | | |
Collapse
|