Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.370
Filter
1.
Sci Total Environ ; 943: 173574, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38823721

ABSTRACT

Mercury is a hazardous heavy metal that is distributed worldwide in aquatic ecosystems. Methylmercury (MeHg) poses significant toxicity risks to aquatic organisms, primarily through bioaccumulation and biomagnification, due to its strong affinity for protein thiol groups, which results in negative effects even at low concentrations. MeHg exposure can cause various physiological changes, oxidative stress, neurotoxicity, metabolic disorders, genetic damage, and immunotoxicity. To assess the risks of MeHg contamination in actual aquatic ecosystems, it is important to understand how MeHg interacts with environmental factors such as temperature, pH, dissolved organic matter, salinity, and other pollutants such as microplastics and organic compounds. Complex environmental conditions can cause potential toxicity, such as synergistic, antagonistic, and unchanged effects, of MeHg in aquatic organisms. This review focuses on demonstrating the toxic effects of single MeHg exposure and the interactive relationships between MeHg and surrounding environmental factors or pollutants on aquatic organisms. Our review also recommends further research on biological and molecular responses in aquatic organisms to better understand the potential toxicity of combinational exposure.


Subject(s)
Aquatic Organisms , Methylmercury Compounds , Water Pollutants, Chemical , Methylmercury Compounds/toxicity , Aquatic Organisms/drug effects , Water Pollutants, Chemical/toxicity , Animals , Environmental Monitoring
2.
Sci Total Environ ; 943: 173577, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38852866

ABSTRACT

Mercury is a well-known neurotoxicant for humans and wildlife. The epidemic of mercury poisoning in Japan has clearly demonstrated that chronic exposure to methylmercury (MeHg) results in serious neurological damage to the cerebral and cerebellar cortex, leading to the dysfunction of the central nervous system (CNS), especially in infants exposed to MeHg in utero. The occurrences of poisoning have caused a wide public concern regarding the health risk emanating from MeHg exposure; particularly those eating large amounts of fish may experience the low-level and long-term exposure. There is growing evidence that MeHg at environmentally relevant concentrations can affect the health of biota in the ecosystem. Although extensive in vivo and in vitro studies have demonstrated that the disruption of redox homeostasis and microtube assembly is mainly responsible for mercurial toxicity leading to adverse health outcomes, it is still unclear whether we could quantitively determine the occurrence of interaction between mercurial and thiols and/or selenols groups of proteins linked directly to outcomes, especially at very low levels of exposure. Furthermore, intracellular calcium homeostasis, cytoskeleton, mitochondrial function, oxidative stress, neurotransmitter release, and DNA methylation may be the targets of mercury compounds; however, the primary targets associated with the adverse outcomes remain to be elucidated. Considering these knowledge gaps, in this article, we conducted a comprehensive review of mercurial toxicity, focusing mainly on the mechanism, and genes/proteins expression. We speculated that comprehensive analyses of transcriptomics, proteomics, and metabolomics could enhance interpretation of "omics" profiles, which may reveal specific biomarkers obviously correlated with specific pathways that mediate selective neurotoxicity.


Subject(s)
Methylmercury Compounds , Humans , Methylmercury Compounds/toxicity , Gene Expression Regulation/drug effects , Mercury/toxicity , Animals , Oxidative Stress
3.
J Hazard Mater ; 473: 134699, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38795488

ABSTRACT

Identifying metabolism and detoxification mechanisms of Hg in biota has important implications for biomonitoring, ecotoxicology, and food safety. Compared to marine mammals and waterbirds, detoxification of MeHg in fish is understudied. Here, we investigated Hg detoxification in Atlantic bluefin tuna Thunnus thynnus using organ-specific Hg and Se speciation data, stable Hg isotope signatures, and Hg and Se particle measurements in multiple tissues. Our results provide evidence for in vivo demethylation and biomineralization of HgSe particles, particularly in spleen and kidney. We observed a maximum range of 1.83‰ for δ202Hg between spleen and lean muscle, whereas Δ199Hg values were similar across all tissues. Mean percent methylmercury ranged from 8% in spleen to 90% in lean muscle. The particulate masses of Hg and Se were higher in spleen and kidney (Hg: 61% and 59%, Se: 12% and 6%, respectively) compared to muscle (Hg: 2%, Se: 0.05%). Our data supports the hypothesis of an organ-specific, two-step detoxification of methylmercury in wild marine fish, consisting of demethylation and biomineralization, like reported for waterbirds. While mass dependent fractionation signatures were highly organ specific, stable mass independent fractionation signatures across all tissues make them potential candidates for source apportionment studies of Hg using ABFT.


Subject(s)
Mercury Isotopes , Methylmercury Compounds , Tuna , Water Pollutants, Chemical , Animals , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Tuna/metabolism , Mercury Isotopes/metabolism , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/analysis , Kidney/metabolism , Spleen/metabolism , Inactivation, Metabolic , Mercury/metabolism , Mercury/analysis , Environmental Monitoring/methods , Muscles/metabolism , Muscles/chemistry , Selenium/metabolism , Selenium/analysis
4.
J Toxicol Sci ; 49(5): 241-248, 2024.
Article in English | MEDLINE | ID: mdl-38692911

ABSTRACT

Methylmercury is an environmental polluting organometallic compound that exhibits neurotoxicity, as observed in Minamata disease patients. Methylmercury damages peripheral nerves in Minamata patients, causing more damage to sensory nerves than motor nerves. Peripheral nerves are composed of three cell types: dorsal root ganglion (DRG) cells, anterior horn cells (AHCs), and Schwann cells. In this study, we compared cultured these three cell types derived from the rat for susceptibility to methylmercury cytotoxicity, intracellular accumulation of mercury, expression of L-type amino acid transporter 1 (LAT1), which transports methylmercury into cells, and expression of multidrug resistance-associated protein 2 (MRP2), which transports methylmercury-glutathione conjugates into the extracellular space. Of the cells examined, we found that DRG cells were the most susceptible to methylmercury with markedly higher intracellular accumulation of mercury. The constitutive level of LAT1 was higher and that of MRP2 lower in DRG cells compared with those in AHC and Schwann cells. Additionally, decreased cell viability caused by methylmercury was significantly reduced by either the LAT1 inhibitor, JPH203, or siRNA-mediated knockdown of LAT1. On the other hand, an MRP2 inhibitor, MK571, significantly intensified the decrease in the cell viability caused by methylmercury. Our results provide a cellular basis for sensory neve predominant injury in the peripheral nerves of Minamata disease patients.


Subject(s)
ATP-Binding Cassette Transporters , Cell Survival , Ganglia, Spinal , Methylmercury Compounds , Schwann Cells , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Methylmercury Compounds/toxicity , Schwann Cells/drug effects , Schwann Cells/metabolism , Cell Survival/drug effects , Cells, Cultured , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Peripheral Nerves/metabolism , Peripheral Nerves/drug effects , Male , Rats , Multidrug Resistance-Associated Protein 2
5.
Chemosphere ; 358: 141839, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636911

ABSTRACT

Mediterranean marine biota suffers from various anthropogenic threats. Among them, pollutants such as mercury (Hg) represent important environmental issues that are exacerbated by bioaccumulation and bioamplification along food webs via its organic form, monomethylmercury (MMHg). To date, very little is known regarding the impact of mercury on Porifera and the few available studies have been exclusively focused on Demospongiae. This work studies the effect of MMHgCl at different biological levels of Oscarella lobularis (Porifera, Homoscleromorpha). Bioaccumulation assays show that MMHgCl significantly accumulated in sponge tissues after a 96-h exposure to 0.1 µg L-1. Toxicity assays (LC5096h) show a sensibility that depends on life-stage (adult vs bud). Additionally, we show that the exposure to 1 µg L-1 MMHgCl negatively impacts the epithelial integrity and the regeneration process in buds, as shown by the loss of cell-cell contacts and the alteration of osculum morphogenesis. For the first time in a sponge, a whole set of genes classically involved in metal detoxification and in antioxidant response were identified. Significant changes in catalase, superoxide dismutase and nuclear factor (erythroid-derived 2)-like 2 expressions in exposed juveniles were measured. Such an integrative approach from the physiological to the molecular scales on a non-model organism expands our knowledge concerning sensitivity and toxicity mechanisms induced by MMHg in Porifera, raising new questions regarding the possible defences used by marine sponges.


Subject(s)
Methylmercury Compounds , Porifera , Water Pollutants, Chemical , Animals , Methylmercury Compounds/toxicity , Water Pollutants, Chemical/toxicity , Bioaccumulation , Catalase/metabolism , Superoxide Dismutase/metabolism
6.
Environ Toxicol Chem ; 43(6): 1195-1241, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38682592

ABSTRACT

Birds are used as bioindicators of environmental mercury (Hg) contamination, and toxicity reference values are needed for injury assessments. We conducted a comprehensive review, summarized data from 168 studies, performed a series of Bayesian hierarchical meta-analyses, and developed new toxicity reference values for the effects of methylmercury (MeHg) on birds using a benchmark dose analysis framework. Lethal and sublethal effects of MeHg on birds were categorized into nine biologically relevant endpoint categories and three age classes. Effective Hg concentrations where there was a 10% reduction (EC10) in the production of juvenile offspring (0.55 µg/g wet wt adult blood-equivalent Hg concentrations, 80% credible interval: [0.33, 0.85]), histology endpoints (0.49 [0.15, 0.96] and 0.61 [0.09, 2.48]), and biochemical markers (0.77 [<0.25, 2.12] and 0.57 [0.35, 0.92]) were substantially lower than those for survival (2.97 [2.10, 4.73] and 5.24 [3.30, 9.55]) and behavior (6.23 [1.84, >13.42] and 3.11 [2.10, 4.64]) of juveniles and adults, respectively. Within the egg age class, survival was the most sensitive endpoint (EC10 = 2.02 µg/g wet wt adult blood-equivalent Hg concentrations [1.39, 2.94] or 1.17 µg/g fresh wet wt egg-equivalent Hg concentrations [0.80, 1.70]). Body morphology was not particularly sensitive to Hg. We developed toxicity reference values using a combined survival and reproduction endpoints category for juveniles, because juveniles were more sensitive to Hg toxicity than eggs or adults. Adult blood-equivalent Hg concentrations (µg/g wet wt) and egg-equivalent Hg concentrations (µg/g fresh wet wt) caused low injury to birds (EC1) at 0.09 [0.04, 0.17] and 0.04 [0.01, 0.08], moderate injury (EC5) at 0.6 [0.37, 0.84] and 0.3 [0.17, 0.44], high injury (EC10) at 1.3 [0.94, 1.89] and 0.7 [0.49, 1.02], and severe injury (EC20) at 3.2 [2.24, 4.78] and 1.8 [1.28, 2.79], respectively. Maternal dietary Hg (µg/g dry wt) caused low injury to juveniles at 0.16 [0.05, 0.38], moderate injury at 0.6 [0.29, 1.03], high injury at 1.1 [0.63, 1.87], and severe injury at 2.4 [1.42, 4.13]. We found few substantial differences in Hg toxicity among avian taxonomic orders, including for controlled laboratory studies that injected Hg into eggs. Our results can be used to quantify injury to birds caused by Hg pollution. Environ Toxicol Chem 2024;43:1195-1241. Published 2024. This article is a U.S. Government work and is in the public domain in the USA. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.


Subject(s)
Birds , Environmental Pollutants , Methylmercury Compounds , Animals , Methylmercury Compounds/toxicity , Environmental Pollutants/toxicity , Reference Values , Diet , Bayes Theorem
7.
Ecotoxicol Environ Saf ; 278: 116360, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38678690

ABSTRACT

Methylmercury (MeHg) is a neurotoxin associated with foetal neurodevelopmental and adult cognitive deficits. Neurons are highly dependent on the tricarboxylic acid cycle and oxidative phosphorylation to produce ATP and meet their high energy demands. Therefore, mitochondrial quality control (MQC) is critical for neuronal homeostasis. While existing studies have generated a wealth of data on the toxicity of MeHg, the complex cascades and molecular pathways governing the mitochondrial network remain to be elucidated. Here, 0.6, 1.2 and 2.4 mg/kg body weight of MeHg were administered intragastrically to pregnant Sprague Dawley rats to model maternal MeHg exposure. The results of the in vivo study revealed that MeHg-treated rats tended to perform more directionless repetitive strategies in the Morris Water Maze and fewer target-orientation strategies than control offspring. Moreover, pathological injury and synaptic toxicity were observed in the hippocampus. Transmission electron microscopy (TEM) demonstrated that the autophagosomes encapsulated damaged mitochondria, while showing a typical mitochondrial fission phenotype, which was supported by the activation of PINK1-dependent key regulators of mitophagy. Moreover, there was upregulation of DRP1 and FIS1. Additionally, MeHg compensation promoted mitochondrial biogenesis, as evidenced by the activation of the mitochondrial PGC1-α-NRF1-TFAM signalling pathway. Notably, SIRT3/AMPK was activated by MeHg, and the expression and activity of p-AMPK, p-LKB1 and SIRT3 were consistently coordinated. Collectively, these findings provide new insights into the potential molecular mechanisms regulating MeHg-induced cognitive deficits through SIRT3/AMPK MQC network coordination.


Subject(s)
Cognitive Dysfunction , Methylmercury Compounds , Mitochondria , Rats, Sprague-Dawley , Methylmercury Compounds/toxicity , Animals , Mitochondria/drug effects , Rats , Female , Cognitive Dysfunction/chemically induced , Pregnancy , Hippocampus/drug effects , Hippocampus/pathology , Maternal Exposure , Prenatal Exposure Delayed Effects/chemically induced
8.
Environ Int ; 186: 108631, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38588609

ABSTRACT

Methylmercury (MeHg) is a global environmental pollutant with neurotoxicity, which can easily crosses the blood-brain barrier and cause irreversible damage to the human central nervous system (CNS). CNS inflammation and autophagy are known to be involved in the pathology of neurodegenerative diseases. Meanwhile, MeHg has the potential to induce microglia-mediated neuroinflammation as well as autophagy. This study aims to further explore the exact molecular mechanism of MeHg neurotoxicity. We conducted in vitro studies using BV2 microglial cell from the central nervous system of mice. The role of inflammation and autophagy in the damage of BV2 cells induced by MeHg was determined by detecting cell viability, cell morphology and structure, reactive oxygen species (ROS), antioxidant function, inflammatory factors, autophagosomes, inflammation and autophagy-related proteins. We further investigated the relationship between the inflammatory response and autophagy induced by MeHg by inhibiting them separately. The results indicated that MeHg could invade cells, change cell structure, activate NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and autophagosome, release a large amount of inflammatory factors and trigger the inflammatory response and autophagy. It was also found that MeHg could disrupt the antioxidant function of cells. In addition, the inhibition of NLRP3 inflammasome alleviated both cellular inflammation and autophagy, while inhibition of autophagy increased cellular inflammation. Our current research suggests that MeHg might induce BV2 cytotoxicity through inflammatory response and autophagy, which may be mediated by the NLRP3 inflammasome activated by oxidative stress.


Subject(s)
Autophagy , Inflammasomes , Inflammation , Methylmercury Compounds , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Methylmercury Compounds/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Microglia/drug effects , Microglia/metabolism , Autophagy/drug effects , Mice , Inflammasomes/metabolism , Animals , Inflammation/chemically induced , Reactive Oxygen Species/metabolism , Cell Line , Cell Survival/drug effects
9.
Int J Mol Sci ; 25(7)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38612696

ABSTRACT

Methylmercury is a known environmental pollutant that exhibits severe neurotoxic effects. However, the mechanism by which methylmercury causes neurotoxicity remains unclear. To date, we have found that oxidative stress-induced growth inhibitor 1 (OSGIN1), which is induced by oxidative stress and DNA damage, is also induced by methylmercury. Therefore, in this study, we investigated the relationship between methylmercury toxicity and the induction of OSGIN1 expression using C17.2 cells, which are mouse brain neural stem cells. Methylmercury increased both OSGIN1 mRNA and protein levels in a time- and concentration-dependent manner. Moreover, these increases were almost entirely canceled out by pretreatment with actinomycin D, a transcription inhibitor. Furthermore, similar results were obtained from cells in which expression of the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) was suppressed, indicating that methylmercury induces OSGIN1 expression via NRF2. Methylmercury causes neuronal cell death by inducing apoptosis. Therefore, we next investigated the role of OSGIN1 in methylmercury-induced neuronal cell death using the activation of caspase-3, which is involved in apoptosis induction, as an indicator. As a result, the increase in cleaved caspase-3 (activated form) induced by methylmercury exposure was decreased by suppressing OSGIN1, and the overexpression of OSGIN1 further promoted the increase in cleaved caspase-3 caused by methylmercury. These results suggest, for the first time, that OSGIN1 is a novel factor involved in methylmercury toxicity, and methylmercury induces apoptosis in C17.2 cells through the induction of OSGIN1 expression by NRF2.


Subject(s)
Methylmercury Compounds , Neural Stem Cells , Neurotoxicity Syndromes , Animals , Mice , Caspase 3/genetics , Methylmercury Compounds/toxicity , NF-E2-Related Factor 2/genetics , Apoptosis
10.
Food Chem Toxicol ; 188: 114684, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663761

ABSTRACT

Exposure to mercury and its organic form methylmercury (MeHg), is of great concern for the developing nervous system. Despite available literature on MeHg neurotoxicity, there is still uncertainty about its mechanisms of action and the doses that trigger developmental effects. Our study combines two alternative methodologies, the human neural stem cells (NSC) and the zebrafish (ZF) embryo, to address the neurotoxic effects of early exposure to nanomolar concentrations of MeHg. Our results show linear or nonmonotonic (hormetic) responses depending on studied parameters. In ZF, we observed a hormetic response in locomotion and larval rotation, but a concentration-dependent response for sensory organ size and habituation. We also observed a possible delayed response as MeHg had greater effects on larval activity at 5 days than at 24 h. In NSC cells, some parameters show a clear dose dependence, such as increased apoptosis and differentiation to glial cells or decreased neuronal precursors; while others show a hormetic response: neuronal differentiation or cell proliferation. This study shows that the ZF model was more susceptible than NSC to MeHg neurotoxicity. The combination of different models has improved the understanding of the underlying mechanisms of toxicity and possible compensatory mechanisms at the cellular and organismal level.


Subject(s)
Embryo, Nonmammalian , Methylmercury Compounds , Neural Stem Cells , Zebrafish , Methylmercury Compounds/toxicity , Zebrafish/embryology , Animals , Neural Stem Cells/drug effects , Humans , Embryo, Nonmammalian/drug effects , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Apoptosis/drug effects , Cell Proliferation/drug effects
11.
Arch Toxicol ; 98(6): 1645-1658, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38546836

ABSTRACT

A number of environmental toxicants are noted for their activity that leads to declined motor function. However, the role of muscle as a proximal toxicity target organ for environmental agents has received considerably less attention than the toxicity targets in the nervous system. Nonetheless, the effects of conventional neurotoxicants on processes of myogenesis and muscle maintenance are beginning to resolve a concerted role of muscle as a susceptible toxicity target. A large body of evidence from epidemiological, animal, and in vitro studies has established that methylmercury (MeHg) is a potent developmental toxicant, with the nervous system being a preferred target. Despite its well-recognized status as a neurotoxicant, there is accumulating evidence that MeHg also targets muscle and neuromuscular development as well as contributes to the etiology of motor defects with prenatal MeHg exposure. Here, we summarize evidence for targets of MeHg in the morphogenesis and maintenance of skeletal muscle that reveal effects on MeHg distribution, myogenesis, myotube formation, myotendinous junction formation, neuromuscular junction formation, and satellite cell-mediated muscle repair. We briefly recapitulate the molecular and cellular mechanisms of skeletal muscle development and highlight the pragmatic role of alternative model organisms, Drosophila and zebrafish, in delineating the molecular underpinnings of muscle development and MeHg-mediated myotoxicity. Finally, we discuss how toxicity targets in muscle development may inform the developmental origins of health and disease theory to explain the etiology of environmentally induced adult motor deficits and accelerated decline in muscle fitness with aging.


Subject(s)
Environmental Exposure , Environmental Pollutants , Methylmercury Compounds , Muscle Development , Muscle, Skeletal , Methylmercury Compounds/toxicity , Animals , Muscle Development/drug effects , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Environmental Pollutants/toxicity , Environmental Exposure/adverse effects , Neuromuscular Junction/drug effects
12.
Mar Pollut Bull ; 201: 116287, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38547612

ABSTRACT

Specimens of the Mediterranean sea anemone Anemonia viridis were exposed to methylmercury (MeHg) and bacterial infection to study their immune responses to a well-known toxic pollutant. Anemones were housed in laboratory conditions and divided into five experimental groups: 1. control (no microinjection); 2. filtered seawater + buffer injection; 3. filtered seawater + Escherichia coli injection; 4. MeHg + buffer injection; 5. MeHg + E. coli injection. Data showed an increase in antioxidant enzyme production compared to the constitutive condition, while methylmercury inhibited lysozyme production. The buffer inoculation had no statistically significant effects on the animals. In addition, electrophoretic and protease analyses revealed differences in the type of proteins produced, as well as a modulation of proteases depending on the treatment. The study demonstrated the immunomodulatory effect of the organic pollutant on A. viridis, validating its use as a model organism for marine coastal biomonitoring programmes and multiple stress studies.


Subject(s)
Bacterial Infections , Environmental Pollutants , Methylmercury Compounds , Sea Anemones , Animals , Methylmercury Compounds/toxicity , Methylmercury Compounds/metabolism , Sea Anemones/physiology , Escherichia coli , Environmental Pollutants/metabolism
13.
Food Chem Toxicol ; 187: 114598, 2024 May.
Article in English | MEDLINE | ID: mdl-38493981

ABSTRACT

Seafood products accumulate methylmercury throughout the food chain and are the main source of methylmercury exposure. Methylmercury may trigger a number of adverse health effects, such as neurodevelopmental or nephrotoxic effects, the risk of which cannot be ruled out for the French high consumers of seafood. The characterisation of methylmercury-related risks is generally based on short-term dietary exposure without considering changes in consumption and exposure over the lifetime. Additionally, focusing on short-term dietary exposure, the fate of methylmercury (especially its accumulation) in the organism is not considered. The present study proposes a methodology basing risk characterization on estimates of body burden over a lifetime. First, trajectories of dietary exposures throughout lifetime were constructed based on the actual concentrations of total diet studies for a fictive representative French population, taking into account the social, economic and demographic parameters of individuals. Next, the fate of methylmercury in the body was estimated, based on these trajectories, using a specific physiologically-based kinetic (PBK) model that generated a representative pool of body burden trajectories. Simulated hair mercury concentrations were closed to previously reported French representative human biomonitoring data. Results showed that at certain stages of life, concentrations of methylmercury in hair were higher than the human biomonitoring guidance value set at 2.5 µg/g of hair by JECFA. This study showed the added value, in the case of substances accumulating in the body, of estimating dietary exposure over a lifetime and using exposure biomarkers estimated by a PBK model characterize the risk.


Subject(s)
Mercury , Methylmercury Compounds , Humans , Methylmercury Compounds/toxicity , Methylmercury Compounds/analysis , Seafood/analysis , Food Contamination/analysis , Diet , Dietary Exposure , Mercury/analysis
14.
Sci Total Environ ; 923: 171398, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38442753

ABSTRACT

Methylmercury (MeHg), as a global environmental pollutant, is of concern globally due to its neurodevelopmental toxicity. Mitochondria-associated membranes (MAMs) are highly dynamic sites of endoplasmic reticulum (ER)-haemocyte contact. MAMs are closely associated with the pathophysiology of neurological disorders due to their role in the transfer of calcium ions (Ca2+) between mitochondria and the ER. However, the molecular mechanisms that control these interactions in MeHg-induced neurotoxicity have not yet been characterized. In the current study, MeHg caused increases in the levels of both cytosolic and mitochondrial Ca2+ in PC12 cells and promoted MAMs formation in both in vivo and in vitro experiments. Of note, MeHg perturbed mitochondrial dynamics, promoting a shift toward a fission phenotype, and this was supported by the dysregulation of fission regulators. Interestingly, the MeHg-induced promotion of MAMs formation and increase in Ca2+ levels were effectively attenuated by the inhibition of mitochondrial fission using Mdivi-1, a DRP1 inhibitor. Furthermore, MeHg triggered the AMPK pathway, and most of the aforementioned changes were significantly rescued by Compound C. Mechanistic investigations revealed a reciprocal relationship between AMPK- and Ca2+-mediated mitochondrial fission. The specific inhibitor of Ca2+ uniporter, ruthenium-red (RuR), effectively abolished the feedback regulation of mitochondrial dynamics and MAMs formation mediated by AMPK in response to MeHg-induced Ca2+ overload. This study reveals a novel role of AMPK-DRP1-mediated mitochondrial fragmentation in the coupling of ER-mitochondrial calcium microdomains in MeHg-induced neurotoxicity. The findings provide valuable insights for the development of strategies to regulate mitochondrial imbalances in neurological diseases.


Subject(s)
Calcium , Methylmercury Compounds , Rats , Animals , Calcium/metabolism , Mitochondrial Dynamics , Methylmercury Compounds/toxicity , Methylmercury Compounds/metabolism , AMP-Activated Protein Kinases/metabolism , Mitochondria , Endoplasmic Reticulum/metabolism , Homeostasis
15.
Aquat Toxicol ; 270: 106896, 2024 May.
Article in English | MEDLINE | ID: mdl-38490093

ABSTRACT

Inorganic mercury (IHg) is hazardous to marine organisms especially resulting in neurotoxicity, bivalves are sensitive to pollutants as "ocean sentinel", but data on the neurotoxicity of IHg in bivalves are sparse. So we chosed M. chinensis philippi with typical neural structures in bivalves to investigate the neurotoxicity of IHg, which could be helpful to understand the specificity of neural regulation and the response characteristics of bivalves. After acute exposed to IHg (HgCl2) for 24 h, the metabolites of ganglion tissues in M. chinensis philippi were evaluated using 1H-nuclear magnetic resonance based metabolomics; Ca2+, neurotransmitters (nitric oxide, glutamate, acetylcholine) and related enzymes (calcineurin, nitric oxide synthase and acetylcholinesterase) were measured using biochemical detection. Compared to the control group, the levels of the nitric oxide (81.04 ± 12.84 µmol/g prot) and acetylcholine (30.93 ± 12.57 µg/mg prot) in M. chinensis philippi of IHg-treated were decreased, while glutamate (2.11 ± 0.61 mmol/L) increased significantly; the activity of nitric oxide synthase (679.34 ± 135.33 U/mg prot) was increased, while acetylcholinesterase (1.39 ± 0.44 U/mg prot) decreased significantly, and the activity of calcineurin (0.52 ± 0.02 U/mg prot) had a statistically insignificant increasing tendency. The concentration of Ca2+ (0.92 ± 0.46 mmol/g prot) in the IHg-treated group was significantly higher than that in the control group. OPLS-DA was performed to reveal the difference in metabolites between the control and IHg-challenged groups, the metabolites of glucose, glutamine, inosine, succinate, glutamate, homarine, and alanine were sensitive to IHg, subsequently metabolic pathways that were affected including glucose metabolism, glutamine metabolism, nucleotide metabolism, Krebs cycle, amino acid metabolism and osmotic regulation. In our study, IHg interfered with metabolites in M. chinensis philippi, thus the corresponding metabolic pathways were changed, which influenced the neurotransmitters subsequently. Furthermore, Ca2+overload affected the synthesis or degradation of the neurotransmitters, and then the altered neurotransmitters involved in changes in metabolic pathways again. Overall, we hypothesized that the neurotoxic effects of IHg on bivalve were in close contact with metabolism, neurotransmitters, related enzymes and Ca2+, which could be effective neurotoxic biomarkers for marine environmental quality assessment, and also provide effective data for the study of the regulatory mechanism of the nervous system in response to IHg in bivalves.


Subject(s)
Bivalvia , Mercury , Methylmercury Compounds , Water Pollutants, Chemical , Animals , Mercury/toxicity , Mercury/metabolism , Acetylcholinesterase , Nitric Oxide , Acetylcholine , Calcineurin , Glutamine , Water Pollutants, Chemical/toxicity , Bivalvia/metabolism , Glutamates , Neurotransmitter Agents , Nitric Oxide Synthase , Methylmercury Compounds/toxicity
16.
Arch Toxicol ; 98(4): 1163-1175, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367039

ABSTRACT

Methylmercury (MeHg) is a well-known environmental neurotoxicant that causes severe brain disorders such as Minamata disease. Although some patients with Minamata disease develop olfactory dysfunction, the underlying pathomechanism is largely unknown. We examined the effects of MeHg on the olfactory system using a model of MeHg poisoning in which mice were administered 30 ppm MeHg in drinking water for 8 weeks. Mice exposed to MeHg displayed significant mercury accumulation in the olfactory pathway, including the nasal mucosa, olfactory bulb, and olfactory cortex. The olfactory epithelium was partially atrophied, and olfactory sensory neurons were diminished. The olfactory bulb exhibited an increase in apoptotic cells, hypertrophic astrocytes, and amoeboid microglia, mainly in the granular cell layer. Neuronal cell death was observed in the olfactory cortex, particularly in the ventral tenia tecta. Neuronal cell death was also remarkable in higher-order areas such as the orbitofrontal cortex. Correlation analysis showed that neuronal loss in the olfactory cortex was strongly correlated with the plasma mercury concentration. Our results indicate that MeHg is an olfactory toxicant that damages the central regions involved in odor perception. The model described herein is useful for analyzing the mechanisms and treatments of olfactory dysfunction in MeHg-intoxicated patients.


Subject(s)
Mercury Poisoning, Nervous System , Mercury , Methylmercury Compounds , Olfaction Disorders , Humans , Mice , Animals , Methylmercury Compounds/toxicity , Microglia/pathology , Olfaction Disorders/chemically induced , Olfaction Disorders/complications
17.
Environ Geochem Health ; 46(3): 83, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38367093

ABSTRACT

To investigate the influence of mercury (Hg) mining/smelting on the surrounding soil environment, ninety soil samples were collected around Hg mining/smelting areas in Tongren city, Guizhou Province, Southwest China. The total mercury (THg), methylmercury (MeHg), bioavailability and fractions of Hg in the soil and their potential risk were evaluated. The results showed that Hg mining/smelting significantly increased the soil pH and decreased the soil organic matter content (p < 0.05). The THg content in the surrounding soil was much higher than that at the control site, with almost all the samples exceeding the national standard in China (3.4 mg/kg, GB15618-2018). Similarly, the concentrations of MeHg (0.09-2.74 µg/kg) and bioavailable Hg (0.64-62.94 µg/kg) in these soil samples were also significantly higher than those in the control site. However, the MeHg/THg ratio was significantly lower in mining/smelting influenced soils (0.01-0.68%) than in control soils (0.60-3.72%). Fraction analysis revealed that residual (RES-Hg) and organic matter-bounded (OM-Hg) Hg accounted for more than 50% of the THg. Ecological risk assessment revealed that the potential ecological risk for most of the Hg mining/smelting-influenced soils (30.16 ≤ Er ≤ 2280.02) were higher than those at the control site (15.12 ≤ Er ≤ 27.1). In addition, these Hg mining/smelting-influenced soils posed acceptable noncarcinogenic risks to adults (except for two soil samples), with hazard indices (HIs) ranging from 0.04 to 1.11 and a mean HI of 0.44. However, children suffer serious noncarcinogenic risks, with HIs ranging from 0.34 to 7.43 and a mean HI of 3.10.


Subject(s)
Mercury , Methylmercury Compounds , Soil Pollutants , Child , Humans , Mercury/analysis , Soil/chemistry , Soil Pollutants/toxicity , Soil Pollutants/analysis , Environmental Monitoring/methods , Methylmercury Compounds/toxicity , Methylmercury Compounds/analysis , China , Mining , Risk Assessment
18.
Sci Total Environ ; 920: 170939, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38365040

ABSTRACT

Mercury is a highly toxic metal widely used in human activities worldwide, therefore considered a global public health problem. Many cases of mercury intoxication have occurred in history and represent a huge challenge nowadays. Of particular importance is its methylated form, methylmercury (MeHg). This mercurial species induces damage to several organs in the human body, especially to the central nervous system. Neurological impairments such as executive, memory, motor and visual deficits are associated with MeHg neurotoxicity. Molecular mechanisms involved in MeHg-induced neurotoxicity include excitotoxicity due to glutamatergic imbalance, disturbance in calcium homeostasis and oxidative balance, failure in synaptic support, and inflammatory response. Although neurons are largely affected by MeHg intoxication, they only represent half of the brain cells. Glial cells represent roughly 50 % of the brain cells and are key elements in the functioning of the central nervous system. Particularly, astrocytes and microglia are deeply involved in MeHg-induced neurotoxicity, resulting in distinct neurological outcomes depending on the context. In this review, we discuss the main findings on astroglial and microglial involvement as mediators of neuroprotective and neurotoxic responses to MeHg intoxication. The literature shows that these responses depend on chemical and morphophysiological features, thus, we present some insights for future investigations, considering the particularities of the context, including time and dose of exposure, brain region, and species of study.


Subject(s)
Mercury , Methylmercury Compounds , Humans , Methylmercury Compounds/toxicity , Brain , Oxidation-Reduction , Neurons , Oxidative Stress
19.
Environ Toxicol ; 39(5): 3149-3159, 2024 May.
Article in English | MEDLINE | ID: mdl-38323385

ABSTRACT

Methylmercury (MeHg) causes selective neuronal damage to cerebrocortical neurons (CCNs) in the central nervous system, but not to hippocampal neurons (HiNs), which are highly vulnerable to neurodegenerative diseases. In our previous study using cultured rat neurons, we performed a comprehensive gene expression analysis and found that the brain-derived neurotrophic factor (BDNF), a neurotrophin (NT), was specifically expressed in HiNs. Therefore, to elucidate the causal factors of MeHg toxicity resistance in HiNs, we conducted a comparative study of the protein expression and function of several NTs, including BDNF, using CCNs showing vulnerability to MeHg toxicity and HiNs showing resistance. BDNF was specifically expressed in HiNs, whereas nerve growth factor was barely detectable in either neuron type. In addition, other NTs, NT3 and NT4/5, were expressed in small but nearly equal amounts in both neuron types. Furthermore, among the various pathways involved in MeHg neurotoxicity, the p44/42 MAPK pathway was specifically activated in HiNs, even without MeHg treatment. siRNAs were used to reduce NTs in both neuron types. Only a specific reduction in BDNF attenuated the resistance to MeHg toxicity and p44/42 MAPK activation in HiNs. In addition, the external addition of BDNF and NT4/5, which act on the same tyrosine receptor kinase (Trk), TrkB, suppressed MeHg neurotoxicity in both neuron types. These results suggest that BDNF, expressed specifically in HiNs, is involved in the resistance to MeHg neurotoxicity via TrkB. Additionally, the activation of the p44/42 MAPK pathway may contribute to the inhibitory effect of BDNF on MeHg neurotoxicity.


Subject(s)
Methylmercury Compounds , Neurotoxicity Syndromes , Rats , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Methylmercury Compounds/toxicity , Neurons , Neurotoxicity Syndromes/metabolism , Hippocampus/metabolism
20.
Brain Res ; 1827: 148761, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38211826

ABSTRACT

As an extremely dangerous environmental contaminant, methylmercury (MeHg) results in detrimental health effects in human brain nervous system, one of its main targets. However, as a developmental toxicant, the brain of offspring is vulnerable to MeHg during pregnancy and lactation exposure. Unfortunately, mechanisms of neurodevelopmental injuries induced by MeHg have not been fully elucidated. N-acetylcysteine (NAC) has been used for several decades as an antioxidant to antagonize oxidative stress. However, the molecular mechanisms of NAC alleviating MeHg-induced neurodevelopmental toxicity are not clear. Here, for evaluation of the dose-dependent effects of MeHg exposure on neurodevelopmental injuries of offspring, and the possible protective effects of NAC, the pregnant female mice were exposed to MeHg (4, 8, 12 mg/L, respectively) and NAC (50, 100, 150 mg/kg, respectively) from gestational day 1 (GD1) to postnatal day 21 (PND21). Our results indicated that administering MeHg caused behavioral impairment and neuronal injuries in the cerebral cortex of newborn mice. MeHg dose-dependently caused reactive oxygen species (ROS) overproduction and oxidative stress aggravation, together with expression of Nrf2, HO-1, Notch1, and p21 up-regulation, and CDK2 inhibition. NAC treatment dose-dependently antagonized MeHg-induced oxidative stress that may contribute to alleviating neurobehavioral and neurodevelopmental impairments. These results give insight into that NAC can protect against MeHg-induced neurodevelopmental toxicity by its antioxidation capacity.


Subject(s)
Acetylcysteine , Methylmercury Compounds , Humans , Pregnancy , Female , Animals , Mice , Acetylcysteine/pharmacology , Methylmercury Compounds/toxicity , Lactation , Antioxidants/pharmacology , Brain
SELECTION OF CITATIONS
SEARCH DETAIL
...