Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Sci Rep ; 11(1): 9832, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972601

ABSTRACT

We recently found that tumor necrosis factor-α (TNF-α) may be involved in neuronal cell death induced by methylmercury in the mouse brain. Here, we examined the cells involved in the induction of TNF-α expression by methylmercury in the mouse brain by in situ hybridization. TNF-α-expressing cells were found throughout the brain and were identified as microglia by immunostaining for ionized calcium binding adaptor molecule 1 (Iba1). Methylmercury induced TNF-α expression in mouse primary microglia and mouse microglial cell line BV2. Knockdown of apoptosis signal-regulating kinase 1 (ASK1), an inflammatory cytokine up-regulator that is responsible for reactive oxygen species (ROS), decreased methylmercury-induced TNF-α expression through decreased phosphorylation of p38 MAP kinase in BV2 cells. Suppression of methylmercury-induced reactive oxygen species (ROS) by antioxidant treatment largely abolished the induction of TNF-α expression and phosphorylation of p38 by methylmercury in BV2 cells. Finally, in mouse brain slices, the TNF-α antagonist (WP9QY) inhibited neuronal cell death induced by methylmercury, as did the p38 inhibitor SB203580 and liposomal clodronate (a microglia-depleting agent). These results indicate that methylmercury induces mitochondrial ROS that are involved in activation of the ASK1/p38 pathway in microglia and that this is associated with induction of TNF-α expression and neuronal cell death.


Subject(s)
Brain/pathology , Mercury Poisoning, Nervous System/pathology , Microglia/drug effects , Neurons/drug effects , Animals , Apoptosis/drug effects , Brain/cytology , Cell Line , Clodronic Acid/pharmacology , Disease Models, Animal , Environmental Pollutants/administration & dosage , Environmental Pollutants/toxicity , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mercury Poisoning, Nervous System/etiology , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/toxicity , Mice , Microglia/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/pathology , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Primary Cell Culture , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Toxicology ; 408: 62-69, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29981841

ABSTRACT

Mercury chloride (HgCl2), a neurotoxicant that cannot penetrate the blood-brain barrier (BBB). Although when the BBB are got damaged by neurodegenerative disorders, the absorbed HgCl2, mainly in form of Hg (II)-serum albumin adduct (Hg-HSA) in human plasma, can penetrate BBB and affect central nervous system (CNS) cells. Current study planned to evaluate the effect of Hg-HSA on the physiological function of N9 microglial cells. At low dosage (15 ng/mL) of Hg-HAS, the observed outcomes was: promoted cell propagation, Nitric Oxide (NO) and intracellular Ca2+ levels enhancement, suppressed the release of TNF-α and IL-1ß and inhibited cell proliferation. At high dosage (15 µg/mL) we observed decline in NO and intracellular Ca2+ levels, and increment in the release of TNF-α and IL-1ß. These biphasic effects are similar to hormesis, and the hormesis, in this case, was executed through ERK/MAPKs and JAK/STAT3 signaling pathways. Study of quantum chemistry revealed that Hg2+ could form stable coordination structures in both Asp249 and Cys34 sites of HSA. Although five-coordination structure in Asp249 site is more stable than four-coordination structure in Cys34 site but four-coordination structure is formed easily in-vivo in consideration of binding-site position in spatial structure of HSA.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hormesis , Janus Kinases/metabolism , Mercury Poisoning, Nervous System/etiology , Microglia/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Binding Sites , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mercury Poisoning, Nervous System/enzymology , Mercury Poisoning, Nervous System/pathology , Mice , Microglia/enzymology , Microglia/pathology , Molecular Dynamics Simulation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Protein Binding , Protein Conformation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Biochem Biophys Res Commun ; 502(4): 435-441, 2018 08 25.
Article in English | MEDLINE | ID: mdl-29856999

ABSTRACT

Methylmercury (MeHg) is the causative substance of Minamata disease, which is associated with various neurological disorders such as sensory disturbance and ataxia. It has been suggested low-level dietary intake of MeHg from MeHg-containing fish during gestation adversely affects the fetus. In our study, we investigated the toxicological effects of MeHg exposure on neuronal differentiation focusing on epigenetics. We used human fetal brain-derived immortalized cells (LUHMES cells) as a human neuronal differentiation model. Cell viability, neuronal, and catecholamine markers in LUHMES cells were assessed after exposure to MeHg (0-1000 nM) for 6 days (from day 2 to day 8 of neuronal differentiation). Cell viability on day 8 was not affected by exposure to 1 nM MeHg for 6 days. mRNA levels of AADC, DBH, TUJ1, and SYN1 also were unaffected by MeHg exposure. In contrast, levels of TH, the rate-limiting enzyme for dopamine synthesis, were significantly decreased after MeHg exposure. Acetylated histone H3, acetylated histone H3 lysine 9, and tri-methyl histone H3 lysine 9 levels at the TH gene promoter were not altered by MeHg exposure. However, tri-methylation of histone H3 lysine 27 levels, related to transcriptional repression, were significantly increased at the TH gene promotor after MeHg exposure. In summary, MeHg exposure during neuronal differentiation led to epigenetic changes that repressed TH gene expression. This study provides useful insights into the toxicological mechanisms underlying the effects of developmental MeHg exposure during neuronal differentiation.


Subject(s)
Epigenesis, Genetic/drug effects , Methylmercury Compounds/toxicity , Neurons/drug effects , Tyrosine 3-Monooxygenase/genetics , Animals , Cell Differentiation , Cell Line , Female , Fishes , Food Contamination , Humans , Mercury Poisoning, Nervous System/etiology , Mercury Poisoning, Nervous System/genetics , Mercury Poisoning, Nervous System/pathology , Methylation , Models, Neurological , Neurons/cytology , Neurons/enzymology , Pregnancy , Prenatal Exposure Delayed Effects , Promoter Regions, Genetic , Tyrosine 3-Monooxygenase/antagonists & inhibitors
4.
J Peripher Nerv Syst ; 23(1): 17-22, 2018 03.
Article in English | MEDLINE | ID: mdl-29168293

ABSTRACT

Mercury and its compounds possess strong neurotoxicity and patients with mercury poisoning often report pain and numbness in the distal extremities that conform to the "stocking-glove" pattern. However, no study has investigated whether damage to small nerve fibers is associated with mercury poisoning. The aims of the present study were to evaluate the effects of different doses of mercury chloride (HgCl2 ) on intraepidermal nerve fibers density (IENFD) and Langerhans cells (LCs) in the plantar skin of rats and to assess the possible relationship between changes in IENFD and sensory testing. Male Sprague-Dawley rats were divided into three experimental groups and administered HgCl2 solutions via gavage at three different doses (4.25, 8.5, and 17 mg/kg/day) for 21 days. Subsequently, behavioral tests and pathological changes in IENFD and LCs were assessed at three different time points (1, 2, and 3 weeks). Rats in all three HgCl2 groups exhibited varying degrees of weight and hair loss. Thermal hypersensitivity was evident in all the HgCl2 groups (for middle-2w subgroup, p < 0.05). Mechanical sensitivity tests revealed hyposensitivity in all the HgCl2 groups except the high-1w subgroup. Significant decreases in IENFD (for the high-1w, middle-1w, low-2w, and low-3w subgroups, p < 0.05) and significant increases in the density of LCs (except for the low-1w and high-2w subgroups, all p < 0.05) were found in all groups after HgCl2 exposure. An association analysis revealed a significant correlation between the decrease in IENFD and the increase in LCs densities (r = -0.573, p < 0.01). The present study demonstrated a decrease in IENFD and an increase in LCs density in the plantar skin of rats after HgCl2 poisoning, indicating that damage of the small nerve fibers occurs after mercury poisoning.


Subject(s)
Langerhans Cells/drug effects , Mercuric Chloride/toxicity , Mercury Poisoning, Nervous System/pathology , Nerve Fibers/drug effects , Skin/drug effects , Animals , Langerhans Cells/pathology , Male , Nerve Fibers/pathology , Rats , Rats, Sprague-Dawley , Skin/pathology
5.
Toxicol Lett ; 271: 66-73, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28267559

ABSTRACT

Methylmercury (MeHg) induces site-specific neurotoxicity in the adult brain. In this study, we investigated the site-specific expression of the signaling cascade related to neural activity in a mouse model of MeHg intoxication showing neurodegeneration only in the deep layer of the cerebral cortex, especially layer IV. We performed time course studies of c-fos and brain-derived neurotrophic factor (BDNF) expression levels which are proper markers of neural activity. We showed that upregulation of both markers preceded the neuronal degeneration in the cerebral cortex. Immunohistochemical analysis revealed the site-specific upregulation of c-fos in the deep layer of the cerebral cortex. Western blot analysis showed that c-fos and BDNF expression was associated with CREB phosphorylation, which was triggered by the activation of the p44/42 MAPK, p38 MAPK and PKA pathways. However, we did not detect any changes in the expression levels of c-fos and BDNF proteins and no signs of neuronal degeneration in the hippocampus and cerebellum, despite the fact that we could detect accumulation of MeHg in these two brain regions. These results suggested an intriguing possibility that MeHg-induced neuronal degeneration was caused by site-specific neural hyperactivity triggered by the activation of MAPK and PKA/CREB pathways followed by c-fos and BDNF upregulation.


Subject(s)
Cerebral Cortex/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mercury Poisoning, Nervous System/prevention & control , Methylmercury Compounds , Mitogen-Activated Protein Kinases/metabolism , Nerve Degeneration , Neurons/drug effects , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Enzyme Activation , Immunohistochemistry , Male , Mercury Poisoning, Nervous System/enzymology , Mercury Poisoning, Nervous System/pathology , Mercury Poisoning, Nervous System/physiopathology , Mice, Inbred ICR , Neurons/enzymology , Neurons/pathology , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Time Factors , Up-Regulation
6.
PLoS One ; 12(1): e0170623, 2017.
Article in English | MEDLINE | ID: mdl-28118383

ABSTRACT

Clinical manifestations of methylmercury (MeHg) intoxication include cerebellar ataxia, concentric constriction of visual fields, and sensory and auditory disturbances. The symptoms depend on the site of MeHg damage, such as the cerebellum and occipital lobes. However, the underlying mechanism of MeHg-induced tissue vulnerability remains to be elucidated. In the present study, we used a rat model of subacute MeHg intoxication to investigate possible MeHg-induced blood-brain barrier (BBB) damage. The model was established by exposing the rats to 20-ppm MeHg for up to 4 weeks; the rats exhibited severe cerebellar pathological changes, although there were no significant differences in mercury content among the different brain regions. BBB damage in the cerebellum after MeHg exposure was confirmed based on extravasation of endogenous immunoglobulin G (IgG) and decreased expression of rat endothelial cell antigen-1. Furthermore, expression of vascular endothelial growth factor (VEGF), a potent angiogenic growth factor, increased markedly in the cerebellum and mildly in the occipital lobe following MeHg exposure. VEGF expression was detected mainly in astrocytes of the BBB. Intravenous administration of anti-VEGF neutralizing antibody mildly reduced the rate of hind-limb crossing signs observed in MeHg-exposed rats. In conclusion, we demonstrated for the first time that MeHg induces BBB damage via upregulation of VEGF expression at the BBB in vivo. Further studies are required in order to determine whether treatment targeted at VEGF can ameliorate MeHg-induced toxicity.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cerebellar Ataxia/chemically induced , Cerebellum/drug effects , Mercury Poisoning, Nervous System/genetics , Methylmercury Compounds/toxicity , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Antibodies, Neutralizing/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Brain Chemistry , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/physiopathology , Cerebellum/metabolism , Cerebellum/pathology , Male , Mercury/analysis , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/pharmacology , Occipital Lobe/drug effects , Occipital Lobe/metabolism , Occipital Lobe/pathology , Rats , Rats, Wistar , Time Factors , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
7.
Brain Nerve ; 67(1): 31-8, 2015 Jan.
Article in Japanese | MEDLINE | ID: mdl-25585433

ABSTRACT

The Minamata disease was discovered in the Minamata region, Kumamoto Prefecture, Japan, in 1956. Symptoms of this disease included cerebellar ataxia, sensory disturbance, narrowing of the visual field, and hearing and speech disturbances. In 1965, similar conditions were identified in persons living around the Agano River area, Niigata Prefecture, Japan and accordingly termed as the Niigata Minamata disease or the second Minamata disease. Both the diseases have been attributed to poisoning with methyl mercury that was generated during the production of acetaldehyde using mercury as a catalyst. The discharged methyl mercury accumulated in fishes and shellfishes and caused poisoning on consumption. This review discusses the history, clinical presentation including atypical forms, and autopsy findings of the Niigata Minamata disease. In addition, it highlights the problems about criteria for official recognition and the therapeutic trial for this disease.


Subject(s)
Mercury Poisoning, Nervous System/diagnosis , Mercury Poisoning, Nervous System/therapy , Methylmercury Compounds/toxicity , Adult , Autopsy , Disease Progression , Humans , Japan , Male , Mercury Poisoning, Nervous System/pathology , Practice Guidelines as Topic
8.
Ukr Biochem J ; 86(1): 124-30, 2014.
Article in Ukrainian | MEDLINE | ID: mdl-24834726

ABSTRACT

Salts of heavy metals are excreted by the kidneys and, as pro-oxidants, stimulate the processes of free radical oxidation. Mercury ions are accumulated in the kidneys. So the study of the features of antioxidant enzymes adaptive response of different kidney layers in response to mercury dichloride is important. Catalase and glytathionperoxidase activity within rat kidneys 72 hours after mercury dichloride intoxication in the ratio of 5 ml per 1 kg of the animal weight was studied. It was important to reveal the influence of the mercury salts on rat kidney antioxidative system. Decreasing glytathionperoxidase activity in cortical and cerebral substances and renal papillae were accompanied by increased contents of oxidative modified proteins and lipids and morphological changes in renal tissue under salt and water loading after mercury dichloride poisoning. The results obtained evidence for the inhibition of antioxidative protection of enzymes in rat kidneys under the mercury dichloride effect.


Subject(s)
Kidney Cortex/enzymology , Kidney Glomerulus/enzymology , Kidney Medulla/enzymology , Mercuric Chloride/toxicity , Mercury Poisoning, Nervous System/enzymology , Animals , Antioxidants/metabolism , Body Weight/drug effects , Catalase/metabolism , Glutathione Peroxidase/metabolism , Histocytochemistry , Kidney Cortex/drug effects , Kidney Cortex/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Medulla/drug effects , Kidney Medulla/pathology , Male , Mercury Poisoning, Nervous System/pathology , Oxidation-Reduction , Rats , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Water-Electrolyte Balance
9.
Mol Cell Biochem ; 390(1-2): 1-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24623265

ABSTRACT

Interest in biochemistry of organoselenium compound has increased in the last decades, mainly due to their chemical and biological activities. Here, we investigated the protective effect of diphenyl diselenide (PhSe)2 (5 µmol/kg), in a mouse model of methylmercury (MeHg)-induced brain toxicity. Swiss male mice were divided into four experimental groups: control, (PhSe)2 (5 µmol/kg, subcutaneous administration), MeHg (40 mg/L, in tap water), and MeHg + (PhSe)2. After the treatment (21 days), the animals were killed and the cerebral cortex was analyzed. Electron microscopy indicated an enlarged and fused mitochondria leading to a reduced number of organelles, in the MeHg-exposed mice. Furthermore, cortical creatine kinase activity, a sensitive mitochondrial oxidative stress sensor, was almost abolished by MeHg. Subcutaneous (PhSe)2 co-treatment rescued from MeHg-induced mitochondrial alterations. (PhSe)2 also behaved as an enhancer of mitochondrial biogenesis, by increasing cortical mitochondria content in mouse-receiving (PhSe)2 alone. Mechanistically, (PhSe)2 (1 µM; 24 h) would trigger the cytoprotective Nrf-2 pathway for activating target genes, since astroglial cells exposed to the chalcogen showed increased content of hemeoxygenase type 1, a sensitive marker of the activation of this via. Thus, it is proposed that the (PhSe)2-neuroprotective effect might be linked to its mitoprotective activity.


Subject(s)
Benzene Derivatives/administration & dosage , Brain/metabolism , Heme Oxygenase-1/biosynthesis , Mitochondria/metabolism , Organoselenium Compounds/administration & dosage , Animals , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Male , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/toxicity , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects
10.
Neurol Sci ; 34(5): 663-9, 2013 May.
Article in English | MEDLINE | ID: mdl-22526770

ABSTRACT

The objective of this study is to elucidate the primary action of methylmercury chloride (MMC) intoxication on peripheral nervous system. We chronologically observed the pathological changes of sciatic nerve, dorsal root ganglion (DRG) neurons, ventral and dorsal roots in rats given 4 mg/kg/day of MMC on consecutive days and killed on days 11, 15, 18 and 21. On day 11, an initial axonal degeneration of type B neuron occurred, predominantly in the distal portions of sciatic nerve. The DRG type A neuron was infiltrated by MRF-1-positive macrophages on day 11. Electron microscopy also demonstrated degenerated mitochondria in type A neuron. On day 21, most of type A neurons seemed to have disappeared. However, type B neurons were well preserved. Immunoblotting with monoclonal antibodies, P0 and neurofilament, demonstrated that both of proteins significantly decreases from day 15. In conclusion, these results indicate that the primary action on type A neuron is the neuron body that consequently results in an anterograde degeneration of nerve fibers, while the type B neuron degeneration occurs in a dying-back process in this subacute model. These findings suggest that the mechanisms involved in the degeneration induced by MMC vary and may depend on certain intrinsic factors peculiar to these neurons.


Subject(s)
Ganglia, Spinal/pathology , Mercury Poisoning, Nervous System/etiology , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/toxicity , Nerve Degeneration/chemically induced , Sciatic Nerve/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Axons/pathology , Disease Models, Animal , Ganglia, Spinal/ultrastructure , In Situ Nick-End Labeling , Male , Microscopy, Electron, Transmission , Myelin P0 Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neurons/ultrastructure , Rats , Rats, Wistar , Time Factors
11.
Toxicol Sci ; 130(2): 373-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22903822

ABSTRACT

Methylmercury (MeHg) is an environmental pollutant that biomagnifies throughout the aquatic food chain, thus representing a toxicological concern for humans subsiding on fish for their dietary intake. Although the developing brain is considered the critical target organ of MeHg toxicity, recent evidence indicates that the cardiovascular system may be the most sensitive in adults. However, data on the mechanisms mediating MeHg-induced cardiovascular toxicity are scarce. Based on the close relationship between cardiovascular disease and dyslipidemia, this study was designed to investigate the effects of long-term MeHg exposure on plasma lipid levels in mice, as well as their underlying mechanisms and potential relationships to MeHg-induced neurotoxicity. Our major finding was that long-term MeHg exposure induced dyslipidemia in rodents. Specifically, Swiss and C57BL/6 mice treated for 21 days with a drinking solution of MeHg (40 mg/l, ad libitum) diluted in tap water showed increased total and non-HDL plasma cholesterol levels. MeHg-induced hypercholesterolemia was also observed in low-density lipoprotein receptor knockout (LDLr⁻/⁻) mice, indicating that this effect was not related to decreased LDLr-mediated cholesterol transport from blood to other tissues. Although the hepatic synthesis of cholesterol was unchanged, significant signs of nephrotoxicity (glomerular shrinkage, tubular vacuolization, and changed urea levels) were observed in MeHg-exposed mice, indicating that the involvement of nephropathy in MeHg-induced lipid dyshomeostasis may not be ruled out. Notably, Probucol (a lipid-lowering drug) prevented the development of hypercholesterolemia when coadministered with MeHg. Finally, hypercholesterolemic LDLr⁻/⁻ mice were more susceptible to MeHg-induced cerebellar glial activation, suggesting that hypercholesterolemia in itself may pose a risk factor in MeHg-induced neurotoxicity. Overall, based on the strong and graded positive association between total as well as LDL cholesterol and risk of cardiovascular diseases, our data support the concept of MeHg-induced cardiovascular toxicity.


Subject(s)
Cardiovascular Diseases/chemically induced , Hypercholesterolemia/chemically induced , Mercury Poisoning, Nervous System/etiology , Methylmercury Compounds/toxicity , Water Pollutants, Chemical/toxicity , Animals , Anticholesteremic Agents/pharmacology , Behavior, Animal/drug effects , Biomarkers/blood , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/physiopathology , Cholesterol/blood , Cholesterol, LDL/blood , Glutathione Peroxidase/metabolism , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Hypercholesterolemia/prevention & control , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Male , Mercury Poisoning, Nervous System/pathology , Mercury Poisoning, Nervous System/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Neuroglia/drug effects , Neuroglia/pathology , Probucol/pharmacology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Urea/blood
12.
Toxicol Sci ; 130(2): 383-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22918959

ABSTRACT

Methylmercury (MeHg) is an environmental contaminant with recognized neurotoxic effects, particularly to the developing nervous system. In the present study, we show that nanomolar concentrations of MeHg can induce long-lasting effects in neural stem cells (NSCs). We investigated short-term direct and long-term inherited effects of exposure to MeHg (2.5 or 5.0 nM) using primary cultures of rat embryonic cortical NSCs. We found that MeHg had no adverse effect on cell viability but reduced NSC proliferation and altered the expression of cell cycle regulators (p16 and p21) and senescence-associated markers. In addition, we demonstrated a decrease in global DNA methylation in the exposed cells, indicating that epigenetic changes may be involved in the mechanisms underlying the MeHg-induced effects. These changes were observed in cells directly exposed to MeHg (parent cells) and in their daughter cells cultured under MeHg-free conditions. In agreement with our in vitro data, a trend was found for decreased cell proliferation in the subgranular zone in the hippocampi of adult mice exposed to low doses of MeHg during the perinatal period. Interestingly, this impaired proliferation had a measurable impact on the total number of neurons in the hippocampal dentate gyrus. Importantly, this effect could be reversed by chronic antidepressant treatment. Our study provides novel evidence for programming effects induced by MeHg in NSCs and supports the idea that developmental exposure to low levels of MeHg may result in long-term consequences predisposing to neurodevelopmental disorders and/or neurodegeneration.


Subject(s)
Environmental Pollutants/toxicity , Epigenesis, Genetic/drug effects , Hippocampus/drug effects , Mercury Poisoning, Nervous System/etiology , Methylmercury Compounds/toxicity , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Animals , Antidepressive Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Environmental Pollutants/metabolism , Fluoxetine/pharmacology , Heredity , Hippocampus/metabolism , Hippocampus/pathology , Male , Mercury Poisoning, Nervous System/genetics , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/genetics , Rats , Rats, Sprague-Dawley , Time Factors
13.
Acta Neurobiol Exp (Wars) ; 72(2): 113-53, 2012.
Article in English | MEDLINE | ID: mdl-22810216

ABSTRACT

The purpose of this review is to examine the parallels between the effects mercury intoxication on the brain and the brain pathology found in autism spectrum disorder (ASD). This review finds evidence of many parallels between the two, including: (1) microtubule degeneration, specifically large, long-range axon degeneration with subsequent abortive axonal sprouting (short, thin axons); (2) dentritic overgrowth; (3) neuroinflammation; (4) microglial/astrocytic activation; (5) brain immune response activation; (6) elevated glial fibrillary acidic protein; (7) oxidative stress and lipid peroxidation; (8) decreased reduced glutathione levels and elevated oxidized glutathione; (9) mitochondrial dysfunction; (10) disruption in calcium homeostasis and signaling; (11) inhibition of glutamic acid decarboxylase (GAD) activity; (12) disruption of GABAergic and glutamatergic homeostasis; (13) inhibition of IGF-1 and methionine synthase activity; (14) impairment in methylation; (15) vascular endothelial cell dysfunction and pathological changes of the blood vessels; (16) decreased cerebral/cerebellar blood flow; (17) increased amyloid precursor protein; (18) loss of granule and Purkinje neurons in the cerebellum; (19) increased pro-inflammatory cytokine levels in the brain (TNF-α, IFN-γ, IL-1ß, IL-8); and (20) aberrant nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB). This review also discusses the ability of mercury to potentiate and work synergistically with other toxins and pathogens in a way that may contribute to the brain pathology in ASD. The evidence suggests that mercury may be either causal or contributory in the brain pathology in ASD, possibly working synergistically with other toxic compounds or pathogens to produce the brain pathology observed in those diagnosed with an ASD.


Subject(s)
Autistic Disorder/pathology , Brain/pathology , Mercury Poisoning, Nervous System/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Autistic Disorder/immunology , Autistic Disorder/psychology , Brain/immunology , Calcium/metabolism , Cerebellum/pathology , Cytokines/metabolism , Cytokines/physiology , Encephalitis/pathology , Endothelium, Vascular/pathology , Glutamic Acid/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Insulin-Like Growth Factor I/physiology , Lipid Peroxidation/drug effects , Mercury Poisoning, Nervous System/immunology , Mercury Poisoning, Nervous System/psychology , Microglia/drug effects , Microglia/pathology , Microtubules/pathology , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/pathology , NF-kappa B/metabolism , NF-kappa B/physiology , Neurites/pathology , Oxidative Stress/drug effects , gamma-Aminobutyric Acid/physiology
14.
Toxicology ; 300(3): 112-20, 2012 Oct 28.
Article in English | MEDLINE | ID: mdl-22722016

ABSTRACT

Methylmercury (MeHg) is one of the ubiquitous environmental toxicants, which can induce oxidative stress and an indirect excitotoxicity caused by altered glutamate (Glu) metabolism. However, little is known of the interaction between oxidative stress and Glu metabolism play in MeHg poisoning rats. We have investigated the neuroprotective role of MK-801, a non-competitive N-methyl-d-aspartate receptors (NMDAR) antagonist, against MeHg-induced neurotoxicity. Fifty rats were randomly divided into five groups of 10 animals in each group: control group, MK-801 control group, MeHg-treated group (4 and 12 µmol/kg) and MK-801 pre-treated group. Administration of MeHg at a dose of 12 µmol/kg for four weeks significantly increased in ROS and total Hg levels and that caused lipid, protein and DNA peroxidative damage in cerebral cortex. In addition, MeHg also reduced nonenzymic (reduced glutathione, GSH) and enzymic (glutathione peroxidase, GPx and superoxide dismutase, SOD) antioxidants and enhanced neurocyte apoptosis rate in cerebral cortex. MeHg-induced ROS production appears to inhibit the activity of the glutamine synthetase (GS), leading to Glu metabolism dysfunction. Pretreatment with MK-801 at a dose of 0.3 µmol/kg prevented the alterations of the activities of PAG and GS and oxidative stress. In addition, pretreatment with MK-801 significantly alleviated the neurocyte apoptosis rate and histopathological damage. In conclusion, the results suggested ROS formation resulting from MeHg- and Glu-induced oxidative stress contributed to neuronal injury. MK-801 possesses the ability to attenuate MeHg-induced neurotoxicity in the cerebral cortex through mechanisms involving its NMDA receptor binding properties and antioxidation.


Subject(s)
Cerebral Cortex/drug effects , Dizocilpine Maleate/therapeutic use , Environmental Pollutants/toxicity , Glutamic Acid/metabolism , Mercury Poisoning, Nervous System/prevention & control , Methylmercury Compounds/toxicity , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dizocilpine Maleate/administration & dosage , Dose-Response Relationship, Drug , Female , Male , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Neuroprotective Agents/administration & dosage , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
Neurotoxicology ; 33(3): 407-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22531226

ABSTRACT

The visual system is particularly sensitive to methylmercury (MeHg) exposure and, therefore, provides a useful model for investigating the fundamental mechanisms that direct toxic effects. During a period of 70 days, adult of a freshwater fish species Hoplias malabaricus were fed with fish prey previously labeled with two different doses of methylmercury (0.075 and 0.75 µgg(-1)) to determine the mercury distribution and morphological changes in the retina. Mercury deposits were found in the photoreceptor layer, in the inner plexiform layer and in the outer plexiform layer, demonstrating a dose-dependent bioaccumulation. The ultrastructure analysis of retina revealed a cellular deterioration in the photoreceptor layer, morphological changes in the inner and outer segments of rods, structural changes in the plasma membrane of rods and double cones, changes in the process of removal of membranous discs and a structural discontinuity. These results lead to the conclusion that methylmercury is able to cross the blood-retina barrier, accumulate in the cells and layers of retina and induce changes in photoreceptors of H. malabaricus even under subchronic exposure.


Subject(s)
Characiformes , Mercury Poisoning, Nervous System/etiology , Methylmercury Compounds/toxicity , Retina/drug effects , Water Pollutants, Chemical/toxicity , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/metabolism , Body Burden , Dose-Response Relationship, Drug , Food Chain , Food Contamination , Fresh Water , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/metabolism , Permeability , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Retina/metabolism , Retina/ultrastructure , Time Factors , Water Pollutants, Chemical/metabolism
17.
Neurotoxicology ; 33(3): 476-81, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22464990

ABSTRACT

Methylmercury (MeHg) exposure at high concentrations poses significant neurotoxic threat to humans worldwide. The present study investigated the mechanisms of glutathione-mediated attenuation of MeHg neurotoxicity in primary cortical culture. MeHg (5 µM) caused depletion of mono- and disulfide glutathione in neuronal, glial and mixed cultures. Supplementation with exogenous glutathione, specifically glutathione monoethyl ester (GSHME) protected against the MeHg induced neuronal death. MeHg caused increased reactive oxygen species (ROS) formation measured by dichlorodihydrofluorescein (DCF) fluorescence with an early increase at 30 min and a late increase at 6h. This oxidative stress was prevented by the presence of either GSHME or the free radical scavenger, trolox. While trolox was capable of quenching the ROS, it showed no neuroprotection. Exposure to MeHg at subtoxic concentrations (3 µM) caused an increase in system x(c)(-) mediated (14)C-cystine uptake that was blocked by the protein synthesis inhibitor, cycloheximide (CHX). Interestingly, blockade of the early ROS burst prevented the functional upregulation of system x(c)(-). Inhibition of multidrug resistance protein-1 (MRP1) potentiated MeHg neurotoxicity and increased cellular MeHg. Taken together, these data suggest glutathione offers neuroprotection against MeHg toxicity in a manner dependent on MRP1-mediated efflux.


Subject(s)
Cerebral Cortex/drug effects , Glutathione/analogs & derivatives , Mercury Poisoning, Nervous System/prevention & control , Methylmercury Compounds/toxicity , Multidrug Resistance-Associated Proteins/metabolism , Neuroprotective Agents/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cystine/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Female , Free Radical Scavengers/pharmacology , Glutathione/metabolism , Glutathione/pharmacology , Mercury Poisoning, Nervous System/etiology , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/metabolism , Mice , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neuroprotective Agents/metabolism , Oxidative Stress/drug effects , Protein Synthesis Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Time Factors
18.
Proc Natl Acad Sci U S A ; 109(13): 5057-62, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411806

ABSTRACT

In the brains of patients with fetal Minamata disease (FMD), which is caused by exposure to methylmercury (MeHg) during development, many neurons are hypoplastic, ectopic, and disoriented, indicating disrupted migration, maturation, and growth. MeHg affects a myriad of signaling molecules, but little is known about which signals are primary targets for MeHg-induced deficits in neuronal development. In this study, using a mouse model of FMD, we examined how MeHg affects the migration of cerebellar granule cells during early postnatal development. The cerebellum is one of the most susceptible brain regions to MeHg exposure, and profound loss of cerebellar granule cells is detected in the brains of patients with FMD. We show that MeHg inhibits granule cell migration by reducing the frequency of somal Ca(2+) spikes through alterations in Ca(2+), cAMP, and insulin-like growth factor 1 (IGF1) signaling. First, MeHg slows the speed of granule cell migration in a dose-dependent manner, independent of the mode of migration. Second, MeHg reduces the frequency of spontaneous Ca(2+) spikes in granule cell somata in a dose-dependent manner. Third, a unique in vivo live-imaging system for cell migration reveals that reducing the inhibitory effects of MeHg on somal Ca(2+) spike frequency by stimulating internal Ca(2+) release and Ca(2+) influxes, inhibiting cAMP activity, or activating IGF1 receptors ameliorates the inhibitory effects of MeHg on granule cell migration. These results suggest that alteration of Ca(2+) spike frequency and Ca(2+), cAMP, and IGF1 signaling could be potential therapeutic targets for infants with MeHg intoxication.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Movement , Fetal Diseases/pathology , Mercury Poisoning, Nervous System/pathology , Neurons/metabolism , Neurons/pathology , Adenine/pharmacology , Animals , Animals, Newborn , Caffeine/pharmacology , Calcium Signaling/drug effects , Cell Movement/drug effects , Cerebellum/drug effects , Cerebellum/embryology , Cerebellum/pathology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Disease Models, Animal , Female , Fetal Diseases/metabolism , Insulin-Like Growth Factor I/pharmacology , Male , Mercury Poisoning, Nervous System/metabolism , Methylmercury Compounds/toxicity , Mice , Neurons/drug effects , Signal Transduction/drug effects , Thionucleotides/pharmacology
19.
Toxicol Sci ; 125(1): 209-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21976372

ABSTRACT

Industrial pollution due to heavy metals such as mercury is a major concern for the environment and public health. Mercury, in particular methylmercury (MeHg), primarily affects brain development and neuronal activity, resulting in neurotoxic effects. Because chemokines can modulate brain functions and are involved in neuroinflammatory and neurodegenerative diseases, we tested the possibility that the neurotoxic effect of MeHg may interfere with the chemokine CCL2. We have used an original protocol in young mice using a MeHg-contaminated fish-based diet for 3 months relevant to human MeHg contamination. We observed that MeHg induced in the mice cortex a decrease in CCL2 concentrations, neuronal cell death, and microglial activation. Knock-out (KO) CCL2 mice fed with a vegetal control food already presented a decrease in cortical neuronal cell density in comparison with wild-type animals under similar diet conditions, suggesting that the presence of CCL2 is required for normal neuronal survival. Moreover, KO CCL2 mice showed a pronounced neuronal cell death in response to MeHg. Using in vitro experiments on pure rat cortical neurons in culture, we observed by blockade of the CCL2/CCR2 neurotransmission an increased neuronal cell death in response to MeHg neurotoxicity. Furthermore, we showed that sod genes are upregulated in brain of wild-type mice fed with MeHg in contrast to KO CCL2 mice and that CCL2 can blunt in vitro the decrease in glutathione levels induced by MeHg. These original findings demonstrate that CCL2 may act as a neuroprotective alarm system in brain deficits due to MeHg intoxication.


Subject(s)
Brain/drug effects , Chemokine CCL2/physiology , Environmental Pollutants/toxicity , Mercury Poisoning, Nervous System/etiology , Methylmercury Compounds/toxicity , Neurons/drug effects , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Cell Culture Techniques , Cell Death/drug effects , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Dose-Response Relationship, Drug , Environmental Pollutants/pharmacokinetics , Gene Expression/drug effects , Male , Mercury Poisoning, Nervous System/enzymology , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Superoxide Dismutase/genetics , Time Factors , Tissue Distribution
20.
Life Sci ; 89(15-16): 555-63, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21683713

ABSTRACT

Neurological disorders are common, costly, and can cause enduring disability. Although mostly unknown, a few environmental toxicants are recognized causes of neurological disorders and subclinical brain dysfunction. One of the best known neurotoxins is methylmercury (MeHg), a ubiquitous environmental toxicant that leads to long-lasting neurological and developmental deficits in animals and humans. In the aquatic environment, MeHg is accumulated in fish, which represent a major source of human exposure. Although several episodes of MeHg poisoning have contributed to the understanding of the clinical symptoms and histological changes elicited by this neurotoxicant in humans, experimental studies have been pivotal in elucidating the molecular mechanisms that mediate MeHg-induced neurotoxicity. The objective of this mini-review is to summarize data from experimental studies on molecular mechanisms of MeHg-induced neurotoxicity. While the full picture has yet to be unmasked, in vitro approaches based on cultured cells, isolated mitochondria and tissue slices, as well as in vivo studies based mainly on the use of rodents, point to impairment in intracellular calcium homeostasis, alteration of glutamate homeostasis and oxidative stress as important events in MeHg-induced neurotoxicity. The potential relationship among these events is discussed, with particular emphasis on the neurotoxic cycle triggered by MeHg-induced excitotoxicity and oxidative stress. The particular sensitivity of the developing brain to MeHg toxicity, the critical role of selenoproteins and the potential protective role of selenocompounds are also discussed. These concepts provide the biochemical bases to the understanding of MeHg neurotoxicity, contributing to the discovery of endogenous and exogenous molecules that counteract such toxicity and provide efficacious means for ablating this vicious cycle.


Subject(s)
Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/toxicity , Animals , Behavior/physiology , Behavior, Animal/physiology , Disease Models, Animal , Fishes , Humans , Mercury Poisoning, Nervous System/psychology , Mice , Oxidative Stress/drug effects , Rats , Selenium Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...