Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.105
1.
Int J Mol Sci ; 25(10)2024 May 14.
Article En | MEDLINE | ID: mdl-38791379

Manganese (Mn) is a heavy metal that can cause excessive Mn poisoning in plants, disrupting microstructural homeostasis and impairing growth and development. However, the specific response mechanisms of leaves to Mn poisoning have not been fully elucidated. This study revealed that Mn poisoning of soybean plants resulted in yellowing of old leaves. Physiological assessments of these old leaves revealed significant increases in the antioxidant enzymes activities (peroxidase (POD), superoxide dismutase (SOD), ascorbate peroxidase (APX), and catalase (CAT)) and elevated levels of malondialdehyde (MDA), proline, indoleacetic acid (IAA), and salicylic acid (SA), under 100 µM Mn toxicity. Conversely, the levels of abscisic acid (ABA), gibberellin 3 (GA3), and jasmonic acid (JA) significantly decreased. The Mn content in the affected leaves significantly increased, while the levels of Ca, Na, K, and Cu decreased. Transcriptome analysis revealed 2258 differentially expressed genes in the Mn-stressed leaves, 744 of which were upregulated and 1514 were downregulated; these genes included genes associated with ion transporters, hormone synthesis, and various enzymes. Quantitative RT-PCR (qRT-PCR) verification of fifteen genes confirmed altered gene expression in the Mn-stressed leaves. These findings suggest a complex gene regulatory mechanism under Mn toxicity and stress, providing a foundation for further exploration of Mn tolerance-related gene regulatory mechanisms in soybean leaves. Using the methods described above, this study will investigate the molecular mechanism of old soybean leaves' response to Mn poisoning, identify key genes that play regulatory roles in Mn toxicity stress, and lay the groundwork for cultivating high-quality soybean varieties with Mn toxicity tolerance traits.


Gene Expression Regulation, Plant , Glycine max , Manganese , Plant Leaves , Glycine max/drug effects , Glycine max/metabolism , Glycine max/genetics , Plant Leaves/drug effects , Plant Leaves/metabolism , Manganese/toxicity , Manganese/metabolism , Gene Expression Regulation, Plant/drug effects , Stress, Physiological/drug effects , Antioxidants/metabolism , Plant Proteins/metabolism , Plant Proteins/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Malondialdehyde/metabolism , Gene Expression Profiling
2.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732149

Manganese (Mn), a cofactor for various enzyme classes, is an essential trace metal for all organisms. However, overexposure to Mn causes neurotoxicity. Here, we evaluated the effects of exposure to Mn chloride (MnCl2) on viability, morphology, synapse function (based on neurogranin expression) and behavior of zebrafish larvae. MnCl2 exposure from 2.5 h post fertilization led to reduced survival (60%) at 5 days post fertilization. Phenotypical changes affected body length, eye and olfactory organ size, and visual background adaptation. This was accompanied by a decrease in both the fluorescence intensity of neurogranin immunostaining and expression levels of the neurogranin-encoding genes nrgna and nrgnb, suggesting the presence of synaptic alterations. Furthermore, overexposure to MnCl2 resulted in larvae exhibiting postural defects, reduction in motor activity and impaired preference for light environments. Following the removal of MnCl2 from the fish water, zebrafish larvae recovered their pigmentation pattern and normalized their locomotor behavior, indicating that some aspects of Mn neurotoxicity are reversible. In summary, our results demonstrate that Mn overexposure leads to pronounced morphological alterations, changes in neurogranin expression and behavioral impairments in zebrafish larvae.


Behavior, Animal , Larva , Manganese , Neurogranin , Zebrafish , Animals , Zebrafish/metabolism , Larva/drug effects , Behavior, Animal/drug effects , Neurogranin/metabolism , Neurogranin/genetics , Manganese/toxicity , Chlorides/toxicity , Manganese Compounds
3.
Int J Mol Sci ; 25(10)2024 May 13.
Article En | MEDLINE | ID: mdl-38791326

Chronic environmental exposure to toxic heavy metals, which often occurs as a mixture through occupational and industrial sources, has been implicated in various neurological disorders, including Parkinsonism. Vanadium pentoxide (V2O5) typically presents along with manganese (Mn), especially in welding rods and high-capacity batteries, including electric vehicle batteries; however, the neurotoxic effects of vanadium (V) and Mn co-exposure are largely unknown. In this study, we investigated the neurotoxic impact of MnCl2, V2O5, and MnCl2-V2O5 co-exposure in an animal model. C57BL/6 mice were intranasally administered either de-ionized water (vehicle), MnCl2 (252 µg) alone, V2O5 (182 µg) alone, or a mixture of MnCl2 (252 µg) and V2O5 (182 µg) three times a week for up to one month. Following exposure, we performed behavioral, neurochemical, and histological studies. Our results revealed dramatic decreases in olfactory bulb (OB) weight and levels of tyrosine hydroxylase, dopamine, and 3,4-dihydroxyphenylacetic acid in the treatment groups compared to the control group, with the Mn/V co-treatment group producing the most significant changes. Interestingly, increased levels of α-synuclein expression were observed in the substantia nigra (SN) of treated animals. Additionally, treatment groups exhibited locomotor deficits and olfactory dysfunction, with the co-treatment group producing the most severe deficits. The treatment groups exhibited increased levels of the oxidative stress marker 4-hydroxynonenal in the striatum and SN, as well as the upregulation of the pro-apoptotic protein PKCδ and accumulation of glomerular astroglia in the OB. The co-exposure of animals to Mn/V resulted in higher levels of these metals compared to other treatment groups. Taken together, our results suggest that co-exposure to Mn/V can adversely affect the olfactory and nigral systems. These results highlight the possible role of environmental metal mixtures in the etiology of Parkinsonism.


Manganese Compounds , Manganese , Mice, Inbred C57BL , Vanadium , Animals , Mice , Manganese/toxicity , Vanadium/toxicity , Male , Olfactory Bulb/metabolism , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Dopamine/metabolism , Vanadium Compounds , Oxidative Stress/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , alpha-Synuclein/metabolism , Chlorides/toxicity , Chlorides/metabolism , Tyrosine 3-Monooxygenase/metabolism , Aldehydes/metabolism , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Disease Models, Animal , 3,4-Dihydroxyphenylacetic Acid/metabolism
4.
Environ Sci Pollut Res Int ; 31(23): 34368-34380, 2024 May.
Article En | MEDLINE | ID: mdl-38703317

Manganese-based (Mn-based) nanomaterials (NMs) have great potential as alternatives to conventional Mn fertilizers. Yet, its environmental risks and effects on plant growth are not completely well understood. This study investigated the physiological effects of manganese dioxide (MnO2) and manganese tetroxide (Mn3O4) NMs on inter-root exposure (0-500 mg/L) of hydroponically grown rice. The results showed that on inter-root exposure, 50 mg/L Mn-based NMs promoted the uptake of mineral elements and enhanced the enzymatic activities of antioxidant systems (CAT and SOD) in rice, whereas 500 mg/L Mn3O4 NMs disrupted the mineral element homeostasis and led to phytotoxicity. The promotion effect of MnO2 NMs was better, firstly because MnO2 NMs treatment had lower Mn content in the plant than Mn3O4 NMs. In addition, MnO2 NMs are more transported and absorbed in the plant in ionic form, while Mn3O4 NMs exist in granular form. MnO2 NMs and Mn3O4 NMs both can be used as nano-fertilizers to improve the growth of rice by inter-root application, but the doses should be carefully selected.


Manganese , Oryza , Oryza/growth & development , Oryza/drug effects , Manganese/toxicity , Fertilizers , Nanostructures/toxicity , Manganese Compounds , Oxides , Plant Roots/drug effects , Plant Roots/growth & development
5.
Ecotoxicol Environ Saf ; 278: 116404, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38705038

Manganese (Mn) is an essential trace element for maintaining bodily functions. Excessive exposure to Mn can pose serious health risks to humans and animals, particularly to the nervous system. While Mn has been implicated as a neurotoxin, the exact mechanism of its toxicity remains unclear. Ferroptosis is a form of programmed cell death that results from iron-dependent lipid peroxidation. It plays a role in various physiological and pathological cellular processes and may be closely related to Mn-induced neurotoxicity. However, the mechanism of ferroptosis in Mn-induced neurotoxicity has not been thoroughly investigated. Therefore, this study aims to investigate the role and mechanism of ferroptosis in Mn-induced neurotoxicity. Using bioinformatics, we identified significant changes in genes associated with ferroptosis in Mn-exposed animal and cellular models. We then evaluated the role of ferroptosis in Mn-induced neurotoxicity at both the animal and cellular levels. Our findings suggest that Mn exposure causes weight loss and nervous system damage in mice. In vitro and in vivo experiments have shown that exposure to Mn increases malondialdehyde, reactive oxygen species, and ferrous iron, while decreasing glutathione and adenosine triphosphate. These findings suggest that Mn exposure leads to a significant increase in lipid peroxidation and disrupts iron metabolism, resulting in oxidative stress injury and ferroptosis. Furthermore, we assessed the expression levels of proteins and mRNAs related to ferroptosis, confirming its significant involvement in Mn-induced neurotoxicity.


Ferroptosis , Iron Overload , Lipid Peroxidation , Manganese , Oxidation-Reduction , Ferroptosis/drug effects , Animals , Manganese/toxicity , Mice , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Neurotoxicity Syndromes/pathology , Male , Iron/toxicity , Iron/metabolism , Reactive Oxygen Species/metabolism , Humans
6.
Ecotoxicol Environ Saf ; 279: 116481, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38788562

Manganese (Mn) overexposure has been associated with the development of neurological damage reminiscent of Parkinson's disease, while the underlying mechanisms have yet to be fully characterized. This study aimed to investigate the mechanisms leading to injury in dopaminergic neurons induced by Mn and identify novel treatment approaches. In the in vivo and in vitro models, ICR mice and dopaminergic neuron-like PC12 cells were exposed to Mn, respectively. We treated them with anti-ferroptotic agents ferrostatin-1 (Fer-1), deferoxamine (DFO), HIF-1α activator dimethyloxalylglycine (DMOG) and inhibitor LW6. We also used p53-siRNA to verify the mechanism underlying Mn-induced neurotoxicity. Fe and Mn concentrations increased in ICR mice brains overexposed to Mn. Additionally, Mn-exposed mice exhibited movement impairment and encephalic pathological changes, with decreased HIF-1α, SLC7A11, and GPX4 proteins and increased p53 protein levels. Fer-1 exhibited protective effects against Mn-induced both behavioral and biochemical changes. Consistently, in vitro, Mn exposure caused ferroptosis-related changes and decreased HIF-1α levels, all ameliorated by Fer-1. Upregulation of HIF-1α by DMOG alleviated the Mn-associated ferroptosis, while LW6 exacerbated Mn-induced neurotoxicity through downregulating HIF-1α. p53 knock-down also rescued Mn-induced ferroptosis without altering HIF-1α protein expression. Mn overexposure resulted in ferroptosis in dopaminergic neurons, mediated through the HIF-1α/p53/SLC7A11 pathway.


Amino Acid Transport System y+ , Brain , Ferroptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Manganese , Mice, Inbred ICR , Tumor Suppressor Protein p53 , Animals , Ferroptosis/drug effects , PC12 Cells , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Manganese/toxicity , Brain/drug effects , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Rats , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Cyclohexylamines/pharmacology , Phenylenediamines/toxicity , Phenylenediamines/pharmacology , Deferoxamine/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Amino Acids, Dicarboxylic
7.
Ecotoxicol Environ Saf ; 279: 116496, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38816322

Microbially induced carbonate precipitation (MICP), as an eco-friendly and promising technology that can transform free metal ions into stable precipitation, has been extensively used in remediation of heavy metal contamination. However, its depressed efficiency of heavy metal elimination remains in question due to the inhibition effect of heavy metal toxicity on bacterial activity. In this work, an efficient, low-cost manganese (Mn) elimination strategy by coupling MICP with chitosan biopolymer as an additive with reduced treatment time was suggested, optimized, and implemented. The influences of chitosan at different concentrations (0.01, 0.05, 0.10, 0.15 and 0.30 %, w/v) on bacterial growth, enzyme activity, Mn removal efficiency and microstructure properties of the resulting precipitation were investigated. Results showed that Mn content was reduced by 94.5 % within 12 h with 0.15 % chitosan addition through adsorption and biomineralization as MnCO3 (at an initial Mn concentration of 3 mM), demonstrating a two-thirds decrease in remediation time compared to the chitosan-absent system, whereas maximum urease activity increased by ∼50 %. Microstructure analyses indicated that the mineralized precipitates were spherical-shaped MnCO3, and a smaller size and more uniform distribution of MnCO3 is obtained by the regulation of abundant amino and hydroxyl groups in chitosan. These results demonstrate that chitosan accelerates nucleation and tunes the growth of MnCO3 by providing nucleation sites for mineral formation and alleviating the toxicity of metal ions, which has the potential to upgrade MICP process in a sustainable and effective manner. This work provides a reference for further understanding of the biomineralization regulation mechanism, and gives a new perspective into the application of biopolymer-intensified strategies of MICP technology in heavy metal contamination.


Carbonates , Chitosan , Manganese , Chitosan/chemistry , Manganese/chemistry , Manganese/toxicity , Carbonates/chemistry , Adsorption , Biopolymers/chemistry , Chemical Precipitation , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/chemistry , Urease , Environmental Restoration and Remediation/methods , Biomineralization/drug effects , Biodegradation, Environmental
8.
Environ Int ; 187: 108672, 2024 May.
Article En | MEDLINE | ID: mdl-38648691

Manganese (Mn) is an essential micronutrient required for various biological processes but excess exposure to Mn can cause neurotoxicity. However, there are few reports regarding the toxicity effect of Mn on the kidney as well as the underlying molecule mechanism. Herein, in vivo experiments were adopted to assess the toxicity effects associated with Mn, and found that chronic Mn treatment induced the injury of glomerular podocytes but not renal tubule in rats. Genome-wide CRISPR/Cas9 knockout screen was then employed to explore the biotargets of the toxic effect of Mn on podocytes. Through functional analyses of the enriched candidate genes, NLRP10 was found to be significantly up-regulated and mediated Mn-induced podocyte apoptosis. Further mechanism investigation revealed that NLRP10 expression was regulated by demethylase AlkB homolog 5 (ALKBH5) in an m6A-dependent fashion upon Mn treatment. Moreover, Mn could directly bind to Metadherin (MTDH) and promoted its combination with ALKBH5 to promote NLRP10 expression and cell apoptosis. Finally, logistic regressions, restricted cubic spline regressions and uniform cubic B-spline were used to investigate the association between Mn exposure and the risk of chronic kidney disease (CKD). A U-shaped nonlinear relationship between CKD risk and plasma Mn level, and a positive linear relationship between CKD risk and urinary Mn levels was found in our case-control study. To sum up, our findings illustrated that m6A-dependent NLRP10 regulation is indispensable for podocyte apoptosis and nephrotoxicity induced by Mn, providing fresh insight into understanding the health risk of Mn and a novel target for preventing renal injury in Mn-intoxicated patients.


Manganese , Membrane Proteins , Podocytes , Podocytes/drug effects , Podocytes/metabolism , Animals , Rats , Membrane Proteins/metabolism , Membrane Proteins/genetics , Manganese/toxicity , Renal Insufficiency, Chronic/chemically induced , Humans , Male , Apoptosis/drug effects , Rats, Sprague-Dawley , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
9.
JCI Insight ; 9(10)2024 Apr 23.
Article En | MEDLINE | ID: mdl-38652538

Manganese is an essential yet potentially toxic metal. Initially reported in 2012, mutations in SLC30A10 are the first known inherited cause of manganese excess. SLC30A10 is an apical membrane protein that exports manganese from hepatocytes into bile and from enterocytes into the lumen of the gastrointestinal tract. SLC30A10 deficiency results in impaired gastrointestinal manganese excretion, leading to manganese excess, neurologic deficits, liver cirrhosis, polycythemia, and erythropoietin excess. Neurologic and liver disease are attributed to manganese toxicity. Polycythemia is attributed to erythropoietin excess. The goal of this study was to determine the basis of erythropoietin excess in SLC30A10 deficiency. Here, we demonstrate that transcription factors hypoxia-inducible factor 1a (Hif1a) and 2a (Hif2a), key mediators of the cellular response to hypoxia, are both upregulated in livers of Slc30a10-deficient mice. Hepatic Hif2a deficiency corrected erythropoietin expression and polycythemia and attenuated aberrant hepatic gene expression in Slc30a10-deficient mice, while hepatic Hif1a deficiency had no discernible impact. Hepatic Hif2a deficiency also attenuated manganese excess, though the underlying cause of this is not clear at this time. Overall, our results indicate that hepatic HIF2 is a key determinant of pathophysiology in SLC30A10 deficiency and expand our understanding of the contribution of HIFs to human disease.


Basic Helix-Loop-Helix Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Liver , Manganese , Polycythemia , Animals , Polycythemia/metabolism , Polycythemia/genetics , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Liver/metabolism , Manganese/metabolism , Manganese/toxicity , Manganese/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Humans , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Erythropoietin/metabolism , Erythropoietin/genetics , Mice, Knockout , Male , Hepatocytes/metabolism
10.
Ecotoxicol Environ Saf ; 277: 116365, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38657452

Microglia, the resident immune cells of the central nervous system (CNS), play a dual role in neurotoxicity by releasing the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome and brain-derived neurotrophic factor (BDNF) in response to environmental stress. Suppression of BDNF is implicated in learning and memory impairment induced by exposure to manganese (Mn) or lead (Pb) individually. Methyl CpG Binding Protein 2 (MeCp2) and its phosphorylation status are related to BDNF suppression. Protein phosphatase2A (PP2A), a member of the serine/threonine phosphatases family, dephosphorylates substrates based on the methylation state of its catalytic C subunit (PP2Ac). However, the specific impairment patterns and molecular mechanisms resulting from co-exposure to Mn and Pb remain unclear. Therefore, the purpose of this study was to explore the effects of Mn and Pb exposure, alone and in combination, on inducing neurotoxicity in the hippocampus of mice and BV2 cells, and to determine whether simultaneous exposure to both metals exacerbate their toxicity. Our findings reveal that co-exposure to Mn and Pb leads to severe learning and memory impairment in mice, which correlates with the accumulation of metals in the hippocampus and synergistic suppression of BDNF. This suppression is accompanied by up-regulation of the epigenetic repressor MeCp2 and its phosphorylation status, as well as demethylation of PP2Ac. Furthermore, inhibition of PP2Ac demethylation using ABL127, an inhibitor for its protein phosphatase methylesterase1 (PME1), or knockdown of MeCp2 via siRNA transfection in vitro effectively increases BDNF expression and mitigates BV2 cell damage induced by Mn and Pb co-exposure. We also observe abnormal activation of microglia characterized by enhanced release of the NLRP3 inflammasome, Casepase-1 and pro-inflammatory cytokines IL-1ß, in the hippocampus of mice and BV2 cells. In summary, our experiments demonstrate that simultaneous exposure to Mn and Pb results in more severe hippocampus-dependent learning and memory impairment, which is attributed to epigenetic suppression of BDNF mediated by PP2A regulation.


Brain-Derived Neurotrophic Factor , Epigenesis, Genetic , Hippocampus , Lead , Manganese , Memory Disorders , Animals , Brain-Derived Neurotrophic Factor/metabolism , Mice , Epigenesis, Genetic/drug effects , Manganese/toxicity , Lead/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Memory Disorders/chemically induced , Male , Mice, Inbred C57BL , Microglia/drug effects , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Protein Phosphatase 2/metabolism , Learning/drug effects
11.
Behav Brain Res ; 465: 114969, 2024 May 08.
Article En | MEDLINE | ID: mdl-38548024

Chronic exposure to manganese (Mn) results in motor dysfunction, biochemical and pathological alterations in the brain. Oxidative stress, inflammation, and dysfunction of dopaminergic and GABAergic systems stimulate activating transcription factor-6 (ATF-6) and protein kinase RNA-like ER kinase (PERK) leading to apoptosis. This study aimed to investigate the protective effect of sesame oil (SO) against Mn-induced neurotoxicity. Rats received 25 mg/kg MnCl2 and were concomitantly treated with 2.5, 5, or 8 ml/kg of SO for 5 weeks. Mn-induced motor dysfunction was indicated by significant decreases in the time taken by rats to fall during the rotarod test and in the number of movements observed during the open field test. Also, Mn resulted in neuronal degeneration as observed by histological staining. The striatal levels of lipid peroxides and reduced glutathione (oxidative stress markers), interleukin-6 and tumor necrosis factor-α (inflammatory markers) were significantly elevated. Mn significantly reduced the levels of dopamine and Bcl-2, while GABA, PERK, ATF-6, Bax, and caspase-3 were increased. Interestingly, all SO doses, especially at 8 ml/kg, significantly improved locomotor activity, biochemical deviations and reduced neuronal degeneration. In conclusion, SO may provide potential therapeutic benefits in enhancing motor performance and promoting neuronal survival in individuals highly exposed to Mn.


Manganese Poisoning , Parkinson Disease , Rats , Animals , Manganese/toxicity , Sesame Oil/pharmacology , Parkinson Disease/drug therapy , Oxidative Stress , Manganese Poisoning/drug therapy , Manganese Poisoning/metabolism , Manganese Poisoning/pathology
12.
Ecotoxicol Environ Saf ; 274: 116178, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38461577

BACKGROUND: The impact of heavy metals on liver function has been examined in numerous epidemiological studies. However, these findings lack consistency and longitudinal validation. METHODS: In this study, we conducted three follow-up surveys with 426 participants from Northeast China. Blood and urine samples were collected, along with questionnaire information. Urine samples were analyzed for concentrations of four metals (chromium [Cr], cadmium [Cd], lead [Pb], and manganese [Mn]), while blood samples were used to measure five liver function indicators (alanine aminotransferase [ALT], aspartate aminotransferase [AST], albumin [ALB], globulin [GLB], and total protein [TP]). We utilized a linear mixed-effects model (LME) to explore the association between individual heavy metal exposure and liver function. Joint effects of metal mixtures were investigated using quantile g-computation and Bayesian kernel machine regression (BKMR). Furthermore, we employed BKMR and Marginal Effect models to examine the interaction effects between metals on liver function. RESULTS: The LME results demonstrated a significant association between urinary heavy metals (Cr, Cd, Pb, and Mn) and liver function markers. BKMR results indicated positive associations between heavy metal mixtures and ALT, AST, and GLB, and negative associations with ALB and TP, which were consistent with the g-comp results. Synergistic effects were observed between Cd-Cr on ALT, Mn-Cr and Cr-Pb on ALB, while an antagonistic effect was found between Mn-Pb and Mn-Cd on ALB. Additionally, synergistic effects were observed between Mn-Cr on GLB and Cd-Cr on TP. Furthermore, a three-way antagonistic effect of Mn-Pb-Cr on ALB was identified. CONCLUSION: Exposure to heavy metals (Cr, Cd, Mn, Pb) is associated with liver function markers, potentially leading to liver damage. Moreover, there are joint and interaction effects among these metals, which warrant further investigation at both the population and mechanistic levels.


Cadmium , Metals, Heavy , Humans , Cadmium/toxicity , Bayes Theorem , Lead/pharmacology , Metals, Heavy/pharmacology , Manganese/toxicity , Chromium/pharmacology , Liver
13.
CNS Neurosci Ther ; 30(3): e14633, 2024 03.
Article En | MEDLINE | ID: mdl-38429921

AIMS: Excessive influx of manganese (Mn) into the brain across the blood-brain barrier induces neurodegeneration. CYP1B1 is involved in the metabolism of arachidonic acid (AA) that affects vascular homeostasis. We aimed to investigate the effect of brain CYP1B1 on Mn-induced neurotoxicity. METHOD: Brain Mn concentrations and α-synuclein accumulation were measured in wild-type and CYP1B1 knockout mice treated with MnCl2 (30 mg/kg) and biotin (0.2 g/kg) for 21 continuous days. Tight junctions and oxidative stress were analyzed in hCMEC/D3 and SH-SY5Y cells after the treatment with MnCl2 (200 µM) and CYP1B1-derived AA metabolites (HETEs and EETs). RESULTS: Mn exposure inhibited brain CYP1B1, and CYP1B1 deficiency increased brain Mn concentrations and accelerated α-synuclein deposition in the striatum. CYP1B1 deficiency disrupted the integrity of the blood-brain barrier (BBB) and increased the ratio of 3, 4-dihydroxyphenylacetic acid (DOPAC) to dopamine in the striatum. HETEs attenuated Mn-induced inhibition of tight junctions by activating PPARγ in endothelial cells. Additionally, EETs attenuated Mn-induced up-regulation of the KLF/MAO-B axis and down-regulation of NRF2 in neuronal cells. Biotin up-regulated brain CYP1B1 and reduced Mn-induced neurotoxicity in mice. CONCLUSIONS: Brain CYP1B1 plays a critical role in both cerebrovascular and dopamine homeostasis, which might serve as a novel therapeutic target for the prevention of Mn-induced neurotoxicity.


Blood-Brain Barrier , Cytochrome P-450 CYP1B1 , Neuroblastoma , Animals , Humans , Mice , alpha-Synuclein/metabolism , Biotin/metabolism , Blood-Brain Barrier/metabolism , Cytochrome P-450 CYP1B1/metabolism , Dopamine/metabolism , Endothelial Cells/metabolism , Manganese/toxicity , Oxidative Stress
14.
Neurotoxicology ; 102: 1-11, 2024 May.
Article En | MEDLINE | ID: mdl-38461971

Although overexposure to manganese (Mn) is known to cause neurotoxic damage, effective exposure markers for assessing Mn loading in Mn-exposed workers are lacking. Here, we construct a Mn-exposed rat model to perform correlation analysis between Mn-induced neurological damage and Mn levels in various biological samples. We combine this analysis with epidemiological investigation to assess whether Mn concentrations in red blood cells (MnRBCs) and urine (MnU) can be used as valid exposure markers. The results show that Mn exposure resulted in neurotoxic damage in rats and that MnRBCs correlated well with neurological damage, showing potential as a novel Mn exposure biomarker. These findings provide a basis for health monitoring of Mn-exposed workers and the development of more appropriate biological exposure limits.


Biomarkers , Erythrocytes , Manganese , Neurotoxicity Syndromes , Animals , Erythrocytes/drug effects , Erythrocytes/metabolism , Manganese/blood , Manganese/toxicity , Manganese/urine , Biomarkers/blood , Biomarkers/urine , Male , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/blood , Rats , Humans , Manganese Poisoning/blood , Rats, Sprague-Dawley , Occupational Exposure/adverse effects , Female
15.
Toxicol Appl Pharmacol ; 485: 116904, 2024 Apr.
Article En | MEDLINE | ID: mdl-38503349

Manganese (Mn)-induced pulmonary toxicity and the underlying molecular mechanisms remain largely enigmatic. Further, in recent years, microRNAs (miRNAs) have emerged as regulators of several pollutants-mediated toxicity. In this context, our study aimed at elucidating whether miRNAs are involved in manganese (II) chloride (MnCl2) (Mn2+)-induced cytotoxicity in lung epithelial cells. Growth inhibition of Mn2+ towards normal human bronchial epithelial (BEAS-2B) and adenocarcinomic human alveolar basal epithelial (A549) cells was analyzed by MTT assay following 24 or 48 h treatment. Reactive oxygen species (ROS) generation, mitochondrial membrane potential (ΔΨm), cell cycle arrest, and apoptosis were evaluated by flow cytometry. RT-qPCR and Western blot were performed to analyze the expression of cyclins, anti-oxidant genes, and miRNAs. We used small RNA sequencing to investigate Mn2+-induced changes in miRNA expression patterns. In both cell lines, Mn2+ treatment inhibited growth in a dose-dependent manner. Further, compared with vehicle-treated cells, Mn2+ (250 µM) treatment induced ROS generation, cell cycle arrest, apoptosis, and decreased ΔΨm as well as altered the expression of cyclins and anti-oxidant genes. Sequencing data revealed that totally 296 miRNAs were differentially expressed in Mn2+-treated cells. Among them, miR-221-3p was one of the topmost down-regulated miRNAs in Mn2+-treated cells. We further confirmed this association in A549 cells. In addition, transient transfection was performed to study gain-of-function experiments. Forced expression of miR-221-3p significantly improved cell viability and reduced Mn2+-induced cell cycle arrest and apoptosis in BEAS-2B cells. In conclusion, miR-221-3p may be the most likely target that accounts for the cytotoxicity of Mn2+-exposed lung epithelial cells.


Apoptosis , Epithelial Cells , Lung , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , A549 Cells , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Apoptosis/drug effects , Lung/drug effects , Lung/pathology , Lung/metabolism , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Cell Survival/drug effects , Manganese Compounds , Manganese/toxicity , Cell Line , Chlorides/toxicity , Cell Cycle Checkpoints/drug effects , Dose-Response Relationship, Drug
16.
Environ Toxicol Chem ; 43(4): 878-895, 2024 Apr.
Article En | MEDLINE | ID: mdl-38411291

Studies on the bioaccumulation and toxicity of contaminants in Crocodylians are scarce. We evaluated alterations in concentrations of the nondestructive biomarkers butyrylcholinesterase (BChE), glutathione-S-transferase (GST), superoxide dismutase (SOD), and reduced glutathione (GSH), together with bioaccumulation of the metals iron (Fe), copper (Cu), zinc (Zn), manganese (Mn), chronium (Cr), aluminium (Al), and lead (Pb) in Caiman latirostris captured in Tapacurá Reservoir (TR; São Lourenço da Mata, Pernambuco, Brasil), in urbanized areas of Pernambuco State (UA; Brasil) and from the AME Brasil caiman farm (AF; Marechal Deodoro, Alagoas, Brasil); the latter was used as a potential reference with low levels of contamination. For metal analysis, 500 µL of blood was digested in 65% HNO3 and 30% H2O2. The samples were analyzed by inductively coupled plasma-optical emission spectrometry. For analysis of biomarkers, an aliquot of blood was centrifuged to obtain plasma in which biochemical assays were performed. Blood concentrations of metals analyzed in animals from AF were lower compared with TR and UA, confirming that animals from the caiman farm could be used as references with low levels of contamination. Iron, Cu, Mn, Al, and Pb exceeded toxic levels for other vertebrates in animals from TR and UA. Butyrylcholinesterase activity showed significant reduction in adults from UA and TR compared with AF. An increase in the activity of GST and GSH, in adults of TR and UA in relation to AF, was verified. Superoxide dismutase activity showed a significant reduction in adults of TR in relation to AF, and the concentrations of Cu and Mn were negatively correlated with SOD activity. Animals from UA and TR showed greater concentrations of the analyzed metals compared with reference animals, and changes in biomarkers were seen, confirming the potential of these nondestructive chemical and biological parameters in blood of C. latirostris for biomonitoring of pollution. Environ Toxicol Chem 2024;43:878-895. © 2024 SETAC.


Alligators and Crocodiles , Metals, Heavy , Animals , Alligators and Crocodiles/metabolism , Butyrylcholinesterase , Bioaccumulation , Hydrogen Peroxide , Lead , Manganese/toxicity , Superoxide Dismutase/metabolism , Iron , Biomarkers , Metals, Heavy/analysis
17.
Neurotoxicol Teratol ; 102: 107330, 2024.
Article En | MEDLINE | ID: mdl-38307398

Epidemiological studies have reported associations between elevated manganese (Mn) exposure and poorer psychomotor performance in children. Our studies in adult male rats have established that this relationship is causal and that prolonged methylphenidate (MPH) treatment is efficacious in treating this area of dysfunction. However, it is unclear if sensitivity to these Mn deficits differs between females and males, and whether existing pharmacological therapies are efficacious in improving sensorimotor dysfunction in females. To address these questions, we used our rat model of childhood environmental Mn exposure and the Montoya staircase test to determine whether 1) there are sex differences in the lasting sensorimotor dysfunction caused by developmental Mn exposure, and 2) MPH treatment is efficacious in ameliorating the sensorimotor deficits in females. Female and male neonates were treated orally with Mn (50 mg Mn/kg/d) from postnatal day 1 to 21 and evaluated for skilled forelimb sensorimotor performance as adults. Subsequently, the efficacy of acute oral MPH treatment (doses of 0, 0.5, and 3.0 mg MPH/kg/d) was assessed in females using a within-subject MPH treatment design. Developmental postnatal Mn exposure produced lasting sensorimotor reaching and grasping deficits that were milder in females than in males. Acute MPH treatment of Mn-exposed females with the 0.5 mg/kg/d dose attenuated the reaching dysfunction without alleviating grasping dysfunction. These findings show sex-based variations in sensitivity to the sensorimotor impairment caused by developmental Mn exposure, and they are consistent with prior studies showing less vulnerability of females to Mn-induced dysfunction in other functional domains, possibly due to the protective effects of estrogen. Given our previous work showing the efficacy of MPH treatment to alleviate Mn-induced inattention, impulsiveness, and sensorimotor dysfunctions in adult male rats, they also highlight the need for further research into sex-based differences in cognitive and behavioral areas of brain function, and the efficacy of therapeutics in treating behavioral dysfunction in females. Supported by NIEHS R01ES028369.


Central Nervous System Stimulants , Methylphenidate , Humans , Child , Rats , Animals , Male , Female , Methylphenidate/pharmacology , Manganese/toxicity , Psychomotor Performance
18.
Neurotoxicol Teratol ; 102: 107337, 2024.
Article En | MEDLINE | ID: mdl-38423398

Studies in children have reported associations between elevated manganese (Mn) exposure and ADHD-related symptoms of inattention, impulsivity/hyperactivity, and psychomotor impairment. Maternal choline supplementation (MCS) during pregnancy/lactation may hold promise as a protective strategy because it has been shown to lessen cognitive dysfunction caused by numerous early insults. Our objectives were to determine whether (1) developmental Mn exposure alters behavioral reactivity/emotion regulation, in addition to impairing learning, attention, impulse control, and sensorimotor function, and (2) MCS protects against these Mn-induced impairments. Pregnant Long-Evans rats were given standard diet, or a diet supplemented with additional choline throughout gestation and lactation (GD 3 - PND 21). Male offspring were exposed orally to 0 or 50 mg Mn/kg/day over PND 1-21. In adulthood, animals were tested in a series of learning, attention, impulse control, and sensorimotor tasks. Mn exposure caused lasting dysfunction in attention, reactivity to errors and reward omission, learning, and sensorimotor function, recapitulating the constellation of symptoms seen in ADHD children. MCS lessened Mn-induced attentional dysfunction and partially normalized reactivity to committing an error or not receiving an expected reward but provided no protection against Mn-induced learning or sensorimotor dysfunction. In the absence of Mn exposure, MCS produces lasting offspring benefits in learning, attention, and reactivity to errors. To conclude, developmental Mn exposure produces a constellation of deficits consistent with ADHD symptomology, and MCS offered some protection against the adverse Mn effects, adding to the evidence that maternal choline supplementation is neuroprotective for offspring and improves offspring cognitive functioning.


Attention Deficit Disorder with Hyperactivity , Manganese , Humans , Animals , Rats , Female , Pregnancy , Child , Male , Manganese/toxicity , Rodentia , Attention Deficit Disorder with Hyperactivity/chemically induced , Attention Deficit Disorder with Hyperactivity/prevention & control , Rats, Long-Evans , Dietary Supplements , Choline
19.
Ecotoxicol Environ Saf ; 273: 116155, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38417317

Excessive exposure to manganese in the environment or workplace is strongly linked to neurodegeneration and cognitive impairment, but the precise pathogenic mechanism and preventive measures are still not fully understood. The study aimed to investigate manganese -induced oxidative damage in the nervous system from an epigenetic perspective, focusing on the H3K36ac-dependent antioxidant pathway. Additionally, it sought to examine the potential of curcumin in preventing manganese-induced oxidative damage. Histopathology and transmission electron microscopy revealed that apoptosis and necrosis of neurons and mitochondrial ultrastructure damage were observed in the striatum of manganese-exposed rats. manganese suppressed the expression of mitochondrial antioxidant genes, leading to oxidative damage in the rats' striatum and SH-SY5Y cells. With higher doses of manganese, levels of histone acetyltransferase lysine acetyltransferase 2 A (KAT2A) expression and H3K36ac level decreased. ChIP-qPCR confirmed that H3K36ac enrichment in the promoter regions of antioxidant genes SOD2, PRDX3, and TXN2 was reduced in SH-SY5Y cells after manganese exposure, leading to decreased expression of these genes. Overexpression of KAT2A confirms that it attenuates manganese-induced mitochondrial oxidative damage by regulating H3K36ac levels, which in turn controls the expression of antioxidant genes SOD2, PRDX3, and TXN2 in the manganese-exposed cell model. Furthermore, curcumin might control H3K36ac levels by influencing KAT2A expression, boosting antioxidant genes expression, and reducing manganese-induced mitochondrial oxidative damage. In conclusion, the regulation of mitochondrial oxidative stress by histone acetylation may be an important mechanism of manganese-induced neurotoxicity. This regulation could be achieved by reducing the level of H3K36ac near the promoter region of mitochondrial-associated antioxidant genes via KAT2A. Curcumin mitigates manganese-induced oxidative damage in mitochondria and plays a crucial protective role in manganese-induced oxidative injury in the nervous system.


Curcumin , Neuroblastoma , Humans , Rats , Animals , Manganese/toxicity , Manganese/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Curcumin/pharmacology , Neuroblastoma/metabolism , Oxidative Stress , Mitochondria/metabolism , Histones/metabolism , Apoptosis , Neurons/metabolism , Histone Acetyltransferases/metabolism
20.
Sci Rep ; 14(1): 3121, 2024 02 07.
Article En | MEDLINE | ID: mdl-38326390

A response to manganese nanoparticles was studied in seedlings of two wheat cultivars and a model system of plant cell membranes. Nanoparticles at concentrations of 125 and 250 mg/ml were applied foliar. The application of NPs enhanced the content of Mn in plant cells, indicating its penetration through the leaf surface. The stressful effect in the plant cells was estimated based on changes in the activity of antioxidant enzymes, content of chlorophylls and starch. MnNPs evoked no significant changes in the leaf morphology, however, an increase in enzyme activity, starch accumulation, and a decrease in chlorophyll synthesis indicated the stress occurrence. Moreover, a rise in the electrokinetic potential of the chloroplast membrane surface and the reconstruction of their hydrophobic parts toward an increase in fatty acid saturation was found.


Manganese , Nanoparticles , Manganese/toxicity , Manganese/metabolism , Seedlings/metabolism , Triticum/metabolism , Oxidative Stress , Antioxidants/metabolism , Chlorophyll/metabolism , Nanoparticles/toxicity , Nanoparticles/chemistry , Cell Membrane/metabolism , Starch/metabolism
...