Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Glia ; 70(10): 1886-1901, 2022 10.
Article in English | MEDLINE | ID: mdl-35638297

ABSTRACT

Chronic manganese (Mn) overexposure causes a neurological disorder, referred to as manganism, exhibiting symptoms similar to parkinsonism. Dysfunction of the repressor element-1 silencing transcription factor (REST) is associated with various neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and Mn-induced neurotoxicity, but its cellular and molecular mechanisms have yet to be fully characterized. Although neuronal REST is known to be neuroprotective, the role of astrocytic REST in neuroprotection remains to be established. We investigated if astrocytic REST in the striatal region of the mouse brain where Mn preferentially accumulates plays a role in Mn-induced neurotoxicity. Striatal astrocytic REST was deleted by infusion of adeno-associated viral vectors containing sequences of the glial fibrillary acidic protein promoter-driven Cre recombinase into the striatum of RESTflox/flox mice for 3 weeks, followed by Mn exposure (30 mg/kg, daily, intranasally) for another 3 weeks. Striatal astrocytic REST deletion exacerbated Mn-induced impairment of locomotor activity and cognitive function with further decrease in Mn-reduced protein levels of tyrosine hydroxylase and glutamate transporter 1 (GLT-1) in the striatum. Astrocytic REST deletion also exacerbated the Mn-induced proinflammatory mediator COX-2, as well as cytokines such as TNF-α, IL-1ß, and IL-6, in the striatum. Mn-induced detrimental astrocytic products such as proinflammatory cytokines on neuronal toxicity were attenuated by astrocytic REST overexpression, but exacerbated by REST inhibition in an in vitro model using primary human astrocytes and Lund human mesencephalic (LUHMES) neuronal culture. These findings indicate that astrocytic REST plays a critical role against Mn-induced neurotoxicity by modulating astrocytic proinflammatory factors and GLT-1.


Subject(s)
Astrocytes , Manganese Poisoning , Repressor Proteins , Animals , Astrocytes/metabolism , Gene Deletion , Humans , Manganese/toxicity , Manganese Poisoning/genetics , Mice , Repressor Proteins/genetics
2.
Neurobiol Dis ; 158: 105467, 2021 10.
Article in English | MEDLINE | ID: mdl-34358615

ABSTRACT

Inherited autosomal recessive mutations of the manganese (Mn) transporter gene SLC39A14 in humans, results in elevated blood and brain Mn concentrations and childhood-onset dystonia-parkinsonism. The pathophysiology of this disease is unknown, but the nigrostriatal dopaminergic system of the basal ganglia has been implicated. Here, we describe pathophysiological studies in Slc39a14-knockout (KO) mice as a preclinical model of dystonia-parkinsonism in SLC39A14 mutation carriers. Blood and brain metal concentrations in Slc39a14-KO mice exhibited a pattern similar to the human disease with highly elevated Mn concentrations. We observed an early-onset backward-walking behavior at postnatal day (PN) 21 which was also noted in PN60 Slc39a14-KO mice as well as dystonia-like movements. Locomotor activity and motor coordination were also impaired in Slc39a14-KO relative to wildtype (WT) mice. From a neurochemical perspective, striatal dopamine (DA) and metabolite concentrations and their ratio in Slc39a14-KO mice did not differ from WT. Striatal tyrosine hydroxylase (TH) immunohistochemistry did not change in Slc39a14-KO mice relative to WT. Unbiased stereological cell quantification of TH-positive and Nissl-stained estimated neuron number, neuron density, and soma volume in the substantia nigra pars compacta (SNc) was the same in Slc39a14-KO mice as in WT. However, we measured a marked inhibition (85-90%) of potassium-stimulated DA release in the striatum of Slc39a14-KO mice relative to WT. Our findings indicate that the dystonia-parkinsonism observed in this genetic animal model of the human disease is associated with a dysfunctional but structurally intact nigrostriatal dopaminergic system. The presynaptic deficit in DA release is unlikely to explain the totality of the behavioral phenotype and points to the involvement of other neuronal systems and brain regions in the pathophysiology of the disease.


Subject(s)
Behavior, Animal , Cation Transport Proteins/genetics , Dystonia/chemically induced , Manganese Poisoning/metabolism , Manganese Poisoning/psychology , Parkinson Disease, Secondary/chemically induced , Animals , Brain/metabolism , Dopamine/metabolism , Dystonia/genetics , Female , Male , Manganese Poisoning/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Neostriatum/metabolism , Parkinson Disease, Secondary/genetics , Psychomotor Performance , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
3.
Int J Mol Sci ; 21(9)2020 May 07.
Article in English | MEDLINE | ID: mdl-32392784

ABSTRACT

As an essential nutrient, manganese is required for the regulation of numerous cellular processes, including cell growth, neuronal health, immune cell function, and antioxidant defense. However, excess manganese in the body is toxic and produces symptoms of neurological and behavioral defects, clinically known as manganism. Therefore, manganese balance needs to be tightly controlled. In the past eight years, mutations of genes encoding metal transporters ZIP8 (SLC39A8), ZIP14 (SLC39A14), and ZnT10 (SLC30A10) have been identified to cause dysregulated manganese homeostasis in humans, highlighting the critical roles of these genes in manganese metabolism. This review focuses on the most recent advances in the understanding of physiological functions of these three identified manganese transporters and summarizes the molecular mechanisms underlying how the loss of functions in these genes leads to impaired manganese homeostasis and human diseases.


Subject(s)
Cation Transport Proteins/metabolism , Manganese/metabolism , Animals , Cation Transport Proteins/genetics , Homeostasis , Humans , Intestinal Absorption , Manganese/deficiency , Manganese Poisoning/genetics , Mutation
4.
J Neurosci ; 37(25): 5996-6006, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28536273

ABSTRACT

Mutations in human ZIP14 have been linked to symptoms of the early onset of Parkinsonism and Dystonia. This phenotype is likely related to excess manganese accumulation in the CNS. The metal transporter ZIP14 (SLC39A14) is viewed primarily as a zinc transporter that is inducible via proinflammatory stimuli. In vitro evidence shows that ZIP14 can also transport manganese. To examine a role for ZIP14 in manganese homeostasis, we used Zip14 knock-out (KO) male and female mice to conduct comparative metabolic, imaging, and functional studies. Manganese accumulation was fourfold to fivefold higher in brains of Zip14 KO mice compared with young adult wild-type mice. There was less accumulation of subcutaneously administered 54Mn in the liver, gallbladder, and gastrointestinal tract of the KO mice, suggesting that manganese elimination is impaired with Zip14 ablation. Impaired elimination creates the opportunity for atypical manganese accumulation in tissues, including the brain. The intensity of MR images from brains of the Zip14 KO mice is indicative of major manganese accumulation. In agreement with excessive manganese accumulation was the impaired motor function observed in the Zip14 KO mice. These results also demonstrate that ZIP14 is not essential for manganese uptake by the brain. Nevertheless, the upregulation of signatures of brain injury observed in the Zip14 KO mice demonstrates that normal ZIP14 function is an essential factor required to prevent manganese-linked neurodegeneration.SIGNIFICANCE STATEMENT Manganese is an essential micronutrient. When acquired in excess, manganese accumulates in tissues of the CNS and is associated with neurodegenerative disease, particularly Parkinson-like syndrome and dystonia. Some members of the ZIP metal transporter family transport manganese. Using mutant mice deficient in the ZIP14 metal transporter, we have discovered that ZIP14 is essential for manganese elimination via the gastrointestinal tract, and a lack of ZIP14 results in manganese accumulation in critical tissues such as the brain, as measured by MRI, and produces signatures of brain injury and impaired motor function. Humans with altered ZIP14 function would lack this gatekeeper function of ZIP14 and therefore would be prone to manganese-related neurological diseases.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese/metabolism , Motor Activity/genetics , Animals , Brain Chemistry/genetics , Female , Gastrointestinal Motility/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution , Zinc/metabolism , Zinc/pharmacology
5.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 36(12): 930-934, 2018 Dec 20.
Article in Zh | MEDLINE | ID: mdl-30812084

ABSTRACT

Objective: To investigate the functional classification of differentially expressed genes in manganese-poisoned rats and related metabolic pathways, and to provide a reference for the study of the mechanism of manganese poisoning and gene regulation in the prevention and treatment of manganese poisoning. Methods: Six healthy specific pathogen-free male Sprague-Dawley rats were randomly divided into control group and experimental group according to body weight, with 3 rats in each group. Rats in the experimental group were injected intraperitoneally with MnCl(2)·4H(2)O (25 mg/kg) at 0.2 ml/100 g once every 48 h, and the control group was injected with phosphate-buffered saline at the same dose. After one month of exposure, the rats were anesthetized and then sacrificed by cardiac puncture blood collection. The striatum was isolated on ice, and RNA was extracted to establish a DNA data library. Whole genome sequencing was used to identify the differentially expressed genes in the rats with manganese poisoning. Gene Ontology functional enrichment analysis and pathway enrichment analysis were performed to investigate the possible metabolic pathways in which the differentially expressed genes may participate. Results: A total of 18439 genes were detected in the striatum of rats, and 17 differentially expressed genes were screened out. Among them, 10 genes were up-regulated, and 7 genes were down-regulated. According to gene function analysis, 164 functional branches and 26 metabolic pathways with high gene enrichment were screened out. The genes were enriched in synaptic signaling, signal transduction, etc., especially behavioral function. The metabolic pathways with high gene enrichment were endocytosis pathway, PI3K-Akt pathway, and neuroactive ligand-receptor interaction pathway, in which the PI3K-Akt pathway had enrichment of the same differentially expressed gene (29 517) as the FoxO signaling pathway and mTOR signaling pathway, and the neuroactive ligand-receptor interaction pathway had enrichment of the same differentially expressed gene (24 415) as the glutamatergic synaptic pathway. Conclusion: The differentially expressed genes in manganese-poisoned rats may influence the susceptibility to manganese poisoning through the PI3K-Akt pathway, mTOR metabolic pathway, or FoxO metabolic pathway, and may be involved in behavioral changes.


Subject(s)
Gene Expression/physiology , Manganese Poisoning/genetics , Metabolic Networks and Pathways/genetics , Animals , Male , Phosphatidylinositol 3-Kinases , Random Allocation , Rats , Rats, Sprague-Dawley
6.
Toxicol Appl Pharmacol ; 336: 94-100, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29054681

ABSTRACT

Manganese neurotoxicity is characterized by Parkinson-like symptoms with degeneration of dopaminergic neurons in the basal ganglia as the principal pathological feature. Manganese neurotoxicity studies may contribute to a good understanding of the mechanism of Parkinson's disease (PD). In this study, we first confirmed that MnCl2 can promote the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) protein in the nucleus or cytoplasm while increasing the binding activity of Nrf2 and antioxidant response elements, further promoting the expression of downstream target gene heme oxygenase 1 (HO-1) and leading to increase levels of reactive oxygen species (ROS) and reduce the levels of reduced glutathione (GSH). Second, we investigated the role of histone acetylation in the activation of Nrf2/HO-1 pathway by manganese chloride in rat adrenal pheochromocytoma (PC12) cells. Histone acetyltransferase inhibitor (anacardic acid) and histone deacetylase inhibitor (trichostatin A, TSA) were used as pretreatment reagents to adjust the level of histone acetylation. Here, we show that downregulation of histone acetylation can inhibit Mn-induced Nrf2 nuclear translocation and further inhibits the Mn-activated Nrf2/HO-1 pathway. This downregulation also promotes manganese-induced increase of ROS and decrease of GSH in neurons. These results suggest that the downregulation of histone acetylation may play an important role in the neurotoxicity caused by manganese and that TSA may provide new ideas and targets in treating manganese-induced Parkinson's syndrome and PD.


Subject(s)
Chlorides/toxicity , Heme Oxygenase (Decyclizing)/metabolism , Histones/metabolism , Manganese Poisoning/etiology , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Acetylation , Active Transport, Cell Nucleus , Anacardic Acids/pharmacology , Animals , Glutathione/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Manganese Compounds , Manganese Poisoning/drug therapy , Manganese Poisoning/enzymology , Manganese Poisoning/genetics , NF-E2-Related Factor 2/genetics , Neurons/enzymology , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
7.
Am J Hum Genet ; 90(3): 457-66, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22341972

ABSTRACT

Environmental manganese (Mn) toxicity causes an extrapyramidal, parkinsonian-type movement disorder with characteristic magnetic resonance images of Mn accumulation in the basal ganglia. We have recently reported a suspected autosomal recessively inherited syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia in cases without environmental Mn exposure. Whole-genome mapping of two consanguineous families identified SLC30A10 as the affected gene in this inherited type of hypermanganesemia. This gene was subsequently sequenced in eight families, and homozygous sequence changes were identified in all affected individuals. The function of the wild-type protein and the effect of sequence changes were studied in the manganese-sensitive yeast strain Δpmr1. Expressing human wild-type SLC30A10 in the Δpmr1 yeast strain rescued growth in high Mn conditions, confirming its role in Mn transport. The presence of missense (c.266T>C [p.Leu89Pro]) and nonsense (c.585del [p.Thr196Profs(∗)17]) mutations in SLC30A10 failed to restore Mn resistance. Previously, SLC30A10 had been presumed to be a zinc transporter. However, this work has confirmed that SLC30A10 functions as a Mn transporter in humans that, when defective, causes Mn accumulation in liver and brain. This is an important step toward understanding Mn transport and its role in neurodegenerative processes.


Subject(s)
Cation Transport Proteins/genetics , Codon, Nonsense , Manganese Poisoning/genetics , Manganese/metabolism , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mutation, Missense , Adolescent , Adult , Amino Acid Sequence , Brain/metabolism , Cation Transport Proteins/metabolism , Child , Child, Preschool , Chromosome Mapping/methods , Female , Genetic Predisposition to Disease , Humans , Liver/metabolism , Male , Manganese Poisoning/metabolism , Molecular Sequence Data , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Analysis, DNA , Young Adult , Zinc Transporter 8
8.
Am J Hum Genet ; 90(3): 467-77, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22341971

ABSTRACT

Manganese is essential for several metabolic pathways but becomes toxic in excessive amounts. Manganese levels in the body are therefore tightly regulated, but the responsible protein(s) remain incompletely known. We studied two consanguineous families with neurologic disorders including juvenile-onset dystonia, adult-onset parkinsonism, severe hypermanganesemia, polycythemia, and chronic hepatic disease, including steatosis and cirrhosis. We localized the genetic defect by homozygosity mapping and then identified two different homozygous frameshift SLC30A10 mutations, segregating with disease. SLC30A10 is highly expressed in the liver and brain, including in the basal ganglia. Its encoded protein belongs to a large family of membrane transporters, mediating the efflux of divalent cations from the cytosol. We show the localization of SLC30A10 in normal human liver and nervous system, and its depletion in liver from one affected individual. Our in silico analyses suggest that SLC30A10 possesses substrate specificity different from its closest (zinc-transporting) homologs. We also show that the expression of SLC30A10 and the levels of the encoded protein are markedly induced by manganese in vitro. The phenotype associated with SLC30A10 mutations is broad, including neurologic, hepatic, and hematologic disturbances. Intrafamilial phenotypic variability is also present. Chelation therapy can normalize the manganesemia, leading to marked clinical improvements. In conclusion, we show that SLC30A10 mutations cause a treatable recessive disease with pleomorphic phenotype, and provide compelling evidence that SLC30A10 plays a pivotal role in manganese transport. This work has broad implications for understanding of the manganese biology and pathophysiology in multiple human organs.


Subject(s)
Cation Transport Proteins/genetics , Manganese Poisoning/genetics , Membrane Transport Proteins/genetics , Metabolic Diseases/genetics , Parkinsonian Disorders/genetics , Aged , Amino Acid Sequence , Brain/metabolism , Cation Transport Proteins/metabolism , Chromosome Mapping/methods , Female , Frameshift Mutation/genetics , Genes, Recessive , Genetic Predisposition to Disease , Hep G2 Cells , Homozygote , Humans , Immunohistochemistry/methods , Liver/metabolism , Male , Manganese/metabolism , Manganese Poisoning/metabolism , Membrane Transport Proteins/metabolism , Metabolic Diseases/metabolism , Middle Aged , Molecular Sequence Data , Phenotype , Sequence Alignment/methods , Tumor Cells, Cultured , Zinc Transporter 8
9.
Article in Zh | MEDLINE | ID: mdl-24370358

ABSTRACT

OBJECTIVE: To investigate the relationship between mRNA expression of manganese superoxide dismutase (MnSOD) and manganese neurotoxicity. METHODS: Thirty-one patients with occupational chronic manganese poisoning (case group), as well as 31 controls exposed to the same condition (control group), were included in the study. Whole blood RNA was extracted, and the mRNA expression of MnSOD was measured by RT-PCR; the two groups were compared in terms of the mRNA expression of MnSOD. PC12 cells were treated with 0, 100, 200, 400, 600, 800, and 1000 ümol/L MnCl2 for l, 2, 3, and 4 d; the cell viability was determined by MTT assay, and the mRNA expression of MnSOD was measured by RT-PCR. RESULTS: The case group had significantly lower mRNA expression of MnSOD than the control group (0.390 ± 0.080 vs 0.582 ± 0.219, P < 0.05). MnCl2 had a toxic effect on PC12 cells; the concentration of MnCl2 was positively correlated with the toxic effect but negatively correlated with the mRNA expression of MnSOD. CONCLUSION: MnSOD mRNA may be involved in the manganese-induced damage of nerve cells. It is hypothesized that high mRNA expression of MnSOD may play an inhibitory effect on manganese neurotoxicity.


Subject(s)
Manganese Poisoning/genetics , Neurotoxicity Syndromes/genetics , Superoxide Dismutase/genetics , Adult , Animals , Female , Gene Expression , Humans , Male , Middle Aged , PC12 Cells , RNA, Messenger/genetics , Rats
10.
Article in Zh | MEDLINE | ID: mdl-21619820

ABSTRACT

OBJECTIVE: To study the relationship between polymorphisms of MnSOD and the susceptibility of chronic poisoning exposed to manganism occupationally. METHODS: In a study of case-control, genotypes were determined by PCR-RFLP in 164 patients with chronic occupational mangamism poisoning and 328 controls with age- and sex-matched for MnSOD 9Ala-Val. RESULTS: There was a significant difference in the frequency of MnSOD 9Ala-Val at V locus mutant allele between cases and controls (χ(2) = 15.225, P < 0.01, 95%CI = 1.43 ∼ 3.00). Individuals with the genotype VV had a 1.30 of risk increase of occupational chronic manganism poisoning compared with the the genotype AV or AA (OR = 2.30, 95%CI = 1.52 ∼ 3.49, P < 0.05). CONCLUSION: The MnSOD polymorphisms may be related with the susceptibility to chronic occupational manganism poisoning, the risk of chronic occupational manganism poisoning increases in carriers with genotype VV at MnSOD 9Ala-Val locus.


Subject(s)
Genetic Predisposition to Disease , Manganese Poisoning/genetics , Occupational Diseases/genetics , Superoxide Dismutase/genetics , Adult , Case-Control Studies , Female , Genotype , Humans , Male , Middle Aged , Occupational Exposure , Polymorphism, Single Nucleotide
11.
J Biol Chem ; 284(51): 35758-68, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19801673

ABSTRACT

Parkinson disease (PD) and manganism are characterized by motor deficits and a loss of dopamine (DA) neurons in the substantia nigra pars compacta. Epidemiological studies indicate significant correlations between manganese exposure and the propensity to develop PD. The vertebrate divalent metal transporter-1 (DMT-1) contributes to maintaining cellular Mn(2+) homeostasis and has recently been implicated in Fe(2+)-mediated neurodegeneration in PD. In this study we describe a novel model for manganism that incorporates the genetically tractable nematode Caenorhabditis elegans. We show that a brief exposure to Mn(2+) increases reactive oxygen species and glutathione production, decreases oxygen consumption and head mitochondria membrane potential, and confers DA neuronal death. DA neurodegeneration is partially dependent on a putative homologue to DMT-1, SMF-1, as genetic knockdown or deletion partially inhibits the neuronal death. Mn(2+) also amplifies the DA neurotoxicity of the PD-associated protein alpha-synuclein. Furthermore, both SMF-1 and SMF-2 are expressed in DA neurons and contribute to PD-associated neurotoxicant-induced DA neuron death. These studies describe a C. elegans model for manganism and show that DMT-1 homologues contribute to Mn(2+)- and PD-associated DA neuron vulnerability.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cation Transport Proteins/metabolism , Dopamine/metabolism , Manganese Poisoning/metabolism , Manganese/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cation Transport Proteins/genetics , Cell Death , Disease Models, Animal , Dopamine/genetics , Humans , Iron/metabolism , Manganese/toxicity , Manganese Poisoning/genetics , Parkinson Disease/genetics , Reactive Oxygen Species/metabolism , Sequence Homology, Amino Acid
12.
Neurotoxicology ; 80: 71-75, 2020 09.
Article in English | MEDLINE | ID: mdl-32621835

ABSTRACT

Environmental and occupational metal exposure poses serious global concerns. Metal exposure have severally been associated with neurotoxicity and brain damage. Furthermore, receptor for advanced glycation end products (RAGE) is also implicated in neurological disorders, particularly those with altered glucose metabolism. Here, we examine potential compounding effect of metal exposure and RAGE expression on dopamine (DA) and serotonin (SER) neurons in C. elegans. In addition, we evaluate the effect of RAGE expression on DA and SER neurons in hyperglycemic conditions. Newly generated RAGE-expressing C. elegans tagged with green fluorescent proteins (GFP) in DAergic and SERergic neurons were treated with cadmium (Cd) or manganese (Mn). Additionally, the RAGE-expressing worms were also exposed to high glucose conditions. Results showed metals induced neurodegeneration both in the presence and absence of RAGE expression, but the manner of degeneration differed between Cd and Mn treated nematodes. Furthermore, RAGE-expressing worms showed significant neurodegeneration in both DAergic and SERergic neurons. Our results indicate co-occurrence of metal exposure and RAGE expression can induce neurodegeneration. Additionally, we show that RAGE expression can exacerbate hyperglycemic induced neurodegeneration.


Subject(s)
Cadmium Poisoning/metabolism , Caenorhabditis elegans/metabolism , Dopaminergic Neurons/metabolism , Manganese Poisoning/metabolism , Nerve Degeneration , Receptor for Advanced Glycation End Products/metabolism , Serotonergic Neurons/metabolism , Animals , Animals, Genetically Modified , Cadmium Chloride , Cadmium Poisoning/etiology , Cadmium Poisoning/genetics , Cadmium Poisoning/pathology , Caenorhabditis elegans/genetics , Chlorides , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Glucose/toxicity , Manganese Compounds , Manganese Poisoning/etiology , Manganese Poisoning/genetics , Manganese Poisoning/pathology , Receptor for Advanced Glycation End Products/genetics , Serotonergic Neurons/drug effects , Serotonergic Neurons/pathology
13.
Environ Toxicol Pharmacol ; 64: 60-69, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30300793

ABSTRACT

Manganese poisoning is a common occupational disease, studies have found that the susceptibility to manganese poisoning differs in individuals. We adopted genome-wide sequencing methods to screen for susceptibility genes involved in gene-mediated metabolic pathways from the perspective of manganese poisoning. We identified 18,439 genes in this study, including 14,272 known genes and 4398 new genes. We then selected 17 differential genes using p values, of which 7 genes were down-regulated and 10 genes were up-regulated. Possible interaction genes for each differential gene were selected according to the String database. Sgk1, HCRTr1, HspB1, Rem2, Oprd1, ATF5, and TRHr identified in this study may be involved in oxidative stress mechanisms, dopamine (DA) synthesis, and neuronal survival during apoptosis and may affect susceptibility to manganese poisoning.


Subject(s)
Corpus Striatum/metabolism , Genetic Predisposition to Disease , Manganese Poisoning/genetics , Animals , Epistasis, Genetic , Male , Rats, Sprague-Dawley , Transcriptome
14.
Adv Neurobiol ; 18: 35-49, 2017.
Article in English | MEDLINE | ID: mdl-28889262

ABSTRACT

While the neurotoxic effects of manganese were recognized in 1837, the first genetic disorder of manganese metabolism was described only in 2012 when homozygous mutations in SLC30A10 were reported to cause manganese-induced neurotoxicity. Two other genetic disorders of manganese metabolism have now been described - mutations in SLC39A14 cause manganese toxicity, while mutations in SLC39A8 cause manganese and zinc deficiency. Study of rare genetic disorders often provides unique insights into disease pathobiology, and the discoveries of these three inherited disorders of manganese metabolism are already transforming our understanding of manganese homeostasis, detoxification, and neurotoxicity. Here, we review the mechanisms by which mutations in SLC30A10, SLC39A14, and SLC39A8 impact manganese homeostasis to cause human disease.


Subject(s)
Deficiency Diseases/metabolism , Manganese Poisoning/metabolism , Manganese/metabolism , Metal Metabolism, Inborn Errors/metabolism , Cation Transport Proteins/genetics , Deficiency Diseases/genetics , Deficiency Diseases/psychology , Humans , Manganese/deficiency , Manganese Poisoning/genetics , Manganese Poisoning/psychology , Metal Metabolism, Inborn Errors/genetics , Metal Metabolism, Inborn Errors/psychology , Zinc/deficiency , Zinc Transporter 8/genetics
15.
Neurochem Int ; 49(1): 62-71, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16488514

ABSTRACT

Redox-active metals are of paramount importance for biological functions. Their impact and cellular activities participate in the physiological and pathophysiological processes of the central nervous system (CNS), including inflammatory responses. Manganese is an essential trace element and it is required for normal biological activities and ubiquitous enzymatic reactions. However, excessive chronic exposure to manganese results in neurobehavioral deficits. Recent evidence suggests that manganese neurotoxicity involves activation of microglia or astrocytes, representative CNS immune cells. In this study, we assessed the molecular basis of the effects of manganese on the modulation of pro-inflammatory cytokines and nitric oxide (NO) production in primary rat cortical glial cells. Cultured glial cells consisted of 85% of astrocytes and 15% of microglia. Within the assayed concentrations, manganese was unable to induce tumor necrosis factor alpha (TNF-alpha) and inducible nitric oxide synthase (iNOS) expression, whereas it potentiated iNOS and TNF-alpha gene expression by lipopolysaccharide/interferon-gamma-activated glial cells. The enhancement was accompanied by elevation of free manganese, generation of oxidative stress, activation of mitogen-activated protein kinases, and increased NF-kappaB and AP-1 binding activities. The potentiated degradation of inhibitory molecule IkappaB-alpha was one of underlying mechanisms for the increased activation of NF-kappaB by manganese. However, manganese decreased iNOS enzymatic activity possibly through the depletion of cofactor since exogenous tetrahydrobiopterin reversed manganese's action. These data indicate that manganese could modulate glial inflammation through variable strategies.


Subject(s)
Encephalitis/chemically induced , Encephalitis/genetics , Gliosis/chemically induced , Gliosis/genetics , Manganese Poisoning/genetics , Manganese/toxicity , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Biopterins/analogs & derivatives , Biopterins/metabolism , Biopterins/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cytokines/genetics , Cytokines/metabolism , Cytokines/pharmacology , Encephalitis/physiopathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gliosis/physiopathology , I-kappa B Proteins/metabolism , Manganese Poisoning/metabolism , Manganese Poisoning/physiopathology , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Oxidative Stress/drug effects , Oxidative Stress/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Up-Regulation/genetics
16.
J Toxicol Sci ; 41(5): 573-81, 2016.
Article in English | MEDLINE | ID: mdl-27665767

ABSTRACT

Sodium para-aminosalicylate (PAS-Na) was first applied successfully in clinical treatment of two manganism patients with good prognosis. However, the mechanism of how PAS-Na protects against Mn-induced neurotoxicity is still elusive. The current study was conducted to explore the effects of PAS-Na on Mn-induced basal ganglia astrocyte injury, and the involvement of amino acid neurotransmitter in vitro. Basal ganglia astrocytes were exposed to 500 µM manganese chloride (MnCl2) for 24 hr, following by 50, 150, or 450 µM PAS-Na treatment for another 24 hr. MnCl2 significantly decreased viability of astrocytes and induced DNA damages via increasing the percentage of tail DNA and Olive tail moment of DNA. Moreover, Mn interrupted amino acid neurotransmitters by decreasing Gln levels and increasing Glu, Gly levels. In contrast, PAS-Na treatment reversed the aforementioned Mn-induced toxic effects on basal ganglia astrocytes. Taken together, our results demonstrated that excessive Mn exposure may induce toxic effects on basal ganglia astrocytes, while PAS-Na could protect basal ganglia astrocytes from Mn-induced neurotoxicity.


Subject(s)
Aminosalicylic Acid/pharmacology , Astrocytes/drug effects , Basal Ganglia/drug effects , Chlorides/toxicity , DNA Damage/drug effects , Glutamic Acid/metabolism , Glutamine/metabolism , Glycine/metabolism , Manganese Poisoning/prevention & control , Protective Agents/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Basal Ganglia/metabolism , Basal Ganglia/pathology , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Manganese Compounds , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese Poisoning/pathology , Rats, Sprague-Dawley
17.
Toxicology ; 328: 168-78, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25549921

ABSTRACT

Welding fumes (WF) are a complex mixture of toxic metals and gases, inhalation of which can lead to adverse health effects among welders. The presence of manganese (Mn) in welding electrodes is cause for concern about the potential development of Parkinson's disease (PD)-like neurological disorder. Consequently, from an occupational safety perspective, there is a critical need to prevent adverse exposures to WF. As the fume generation rate and physicochemical characteristics of welding aerosols are influenced by welding process parameters like voltage, current or shielding gas, we sought to determine if changing such parameters can alter the fume profile and consequently its neurotoxic potential. Specifically, we evaluated the influence of voltage on fume composition and neurotoxic outcome. Rats were exposed by whole-body inhalation (40 mg/m(3); 3h/day × 5 d/week × 2 weeks) to fumes generated by gas-metal arc welding using stainless steel electrodes (GMA-SS) at standard/regular voltage (25 V; RVSS) or high voltage (30 V; HVSS). Fumes generated under these conditions exhibited similar particulate morphology, appearing as chain-like aggregates; however, HVSS fumes comprised of a larger fraction of ultrafine particulates that are generally considered to be more toxic than their fine counterparts. Paradoxically, exposure to HVSS fumes did not elicit dopaminergic neurotoxicity, as monitored by the expression of dopaminergic and PD-related markers. We show that the lack of neurotoxicity is due to reduced solubility of Mn in HVSS fumes. Our findings show promise for process control procedures in developing prevention strategies for Mn-related neurotoxicity during welding; however, it warrants additional investigations to determine if such modifications can be suitably adapted at the workplace to avert or reduce adverse neurological risks.


Subject(s)
Air Pollutants, Occupational/toxicity , Brain/drug effects , Inhalation Exposure/prevention & control , Manganese Poisoning/prevention & control , Manganese/toxicity , Parkinson Disease, Secondary/prevention & control , Welding/methods , Aerosols , Air Pollutants, Occupational/chemistry , Animals , Body Burden , Brain/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Equipment Design , Gene Expression Regulation/drug effects , Humans , Inhalation Exposure/adverse effects , Male , Manganese/chemistry , Manganese Poisoning/etiology , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Parkinson Disease, Secondary/etiology , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/metabolism , Particle Size , Rats, Sprague-Dawley , Risk Assessment , Solubility , Time Factors , Welding/instrumentation
18.
Toxicol Lett ; 235(1): 17-27, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25791630

ABSTRACT

Overexposure to manganese (Mn) has been known to induce neuronal death and neurodegenerative symptoms. However, the precise mechanisms underlying Mn neurotoxicity remain incompletely understood. In the present study, we established a Mn-exposed rat model and found that downregulation of wild type p53-induced phosphatase 1 (Wip1) might contribute to p53 activation and resultant neuronal apoptosis following Mn exposure. Western blot and immunohistochemical analyses revealed that the expression of Wip1 was markedly decreased following Mn exposure. In addition, immunofluorescence assay demonstrated that Mn exposure led to significant reduction in the number of Wip1-positive neurons. Accordingly, the expression of Mdm2 was progressively decreased, which was accompanied with markedly increased expression of p53, as well as the ratio of Bax/Bcl-xl. Furthermore, we showed that Mn exposure decreased the viability and induced apparent apoptosis in NFG-differentiated neuron-like PC12 cells. Importantly, the expression of Wip1 decreased progressively, whereas the level of cellular p53 and the ratio of Bax/Bcl-xl were elevated, which resembled the expression of the proteins in animal model studies. Depletion of p53 significantly ameliorated Mn-mediated cytotoxic effect in PC12 cells. In addition, ectopic expression of Wip1 attenuated Mn-induced p53 signaling as well as apoptosis in PC12 cells. Finally, we observed that depletion of Wip1 augmented Mn-induced apoptosis in PC12 cells. Collectively, these findings suggest that downregulated Wip1 expression plays an important role in Mn-induced neuronal death in the brain striatum via the modulation of p53 signaling.


Subject(s)
Apoptosis , Basal Ganglia/enzymology , Manganese Poisoning/enzymology , Neurons/enzymology , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Basal Ganglia/pathology , Chlorides , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Manganese Compounds , Manganese Poisoning/etiology , Manganese Poisoning/genetics , Manganese Poisoning/pathology , Nerve Degeneration , Neurons/drug effects , Neurons/pathology , PC12 Cells , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
19.
Toxicol Sci ; 143(2): 454-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25416158

ABSTRACT

The pathological role of α-synuclein (α-Syn) aggregation in neurodegeneration is well recognized, but the physiological function of normal α-Syn remains unknown. As α-Syn protein contains multiple divalent metal binding sites, herein we conducted a comprehensive characterization of the role of α-Syn in manganese-induced dopaminergic neurotoxicity. We established transgenic N27 dopaminergic neuronal cells by stably expressing human wild-type α-Syn at normal physiological levels. α-Syn-expressing dopaminergic cells significantly attenuated Mn-induced neurotoxicity for 24-h exposures relative to vector control cells. To further explore cellular mechanisms, we studied the mitochondria-dependent apoptotic pathway. Analysis of a key mitochondrial apoptotic initiator, cytochrome c, revealed that α-Syn significantly reduces the Mn-induced cytochrome c release into cytosol. The downstream caspase cascade, involving caspase-9 and caspase-3 activation, during Mn exposure was also largely attenuated in Mn-treated α-Syn cells in a time-dependent manner. α-Syn cells also showed a dramatic reduction in the Mn-induced proteolytic activation of the pro-apoptotic kinase PKCδ. The generation of Mn-induced reactive oxygen species (ROS) did not differ between α-Syn and vector control cells, indicating that α-Syn exerts its protective effect independent of altering ROS generation. Inductively coupled plasma-mass spectrometry (ICP-MS) revealed no significant differences in intracellular Mn levels between treated vector and α-Syn cells. Notably, the expression of wild-type α-Syn in primary mesencephalic cells also rescued cells from Mn-induced neurotoxicity. However, prolonged exposure to Mn promoted protein aggregation in α-Syn-expressing cells. Collectively, these results demonstrate that wild-type α-Syn exhibits neuroprotective effects against Mn-induced neurotoxicity during the early stages of exposure in a dopaminergic neuronal model of PD.


Subject(s)
Chlorides/toxicity , Dopaminergic Neurons/drug effects , Manganese Poisoning/genetics , Models, Neurological , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites , Blotting, Western , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Chlorides/metabolism , DNA Fragmentation/drug effects , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Manganese Compounds/metabolism , Manganese Poisoning/complications , Manganese Poisoning/pathology , Manganese Poisoning/prevention & control , Mesencephalon/drug effects , Mesencephalon/metabolism , Mesencephalon/pathology , Parkinson Disease/etiology , Parkinson Disease/pathology , Parkinson Disease/prevention & control , Protein Binding , Rats , Reactive Oxygen Species/metabolism , Spectrophotometry, Atomic , Transfection , alpha-Synuclein/metabolism
20.
Toxicology ; 331: 24-34, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25698507

ABSTRACT

We previously found persistent aberration of hippocampal adult neurogenesis, along with brain manganese (Mn) accumulation, in mouse offspring after developmental exposure to 800-ppm dietary Mn. Reduction of parvalbumin (Pvalb)(+) γ-aminobutyric acid (GABA)-ergic interneurons in the hilus of the dentate gyrus along with promoter region hypermethylation are thought to be responsible for this aberrant neurogenesis. The present study was conducted to examine the relationship between the induction of aberrant neurogenesis and brain Mn accumulation after oral Mn exposure as well as the responsible mechanism in young adult animals. We used two groups of mice with 28- or 56-day exposure periods to oral MnCl2·xH2O at 800 ppm as Mn, a dose sufficient to lead to aberrant neurogenesis after developmental exposure. A third group of mice received intravenous injections of Mn at 5-mg/kg body weight once weekly for 28 days. The 28-day oral Mn exposure did not cause aberrations in neurogenesis. In contrast, 56-day oral exposure caused aberrations in neurogenesis suggestive of reductions in type 2b and type 3 progenitor cells and immature granule cells in the dentate subgranular zone. Brain Mn accumulation in 56-day exposed cases, as well as in directly Mn-injected cases occurred in parallel with reduction of Pvalb(+) GABAergic interneurons in the dentate hilus, suggesting that this may be responsible for aberrant neurogenesis. For reduction of Pvalb(+) interneurons, suppression of brain-derived neurotrophic factor-mediated signaling of mature granule cells may occur via suppression of c-Fos-mediated neuronal plasticity due to direct Mn-toxicity rather than promoter region hypermethylation of Pvalb.


Subject(s)
Chlorides/toxicity , GABAergic Neurons/drug effects , Hippocampus/drug effects , Interneurons/drug effects , Manganese Poisoning/etiology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Administration, Oral , Animals , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Chlorides/administration & dosage , Chlorides/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Interneurons/metabolism , Interneurons/pathology , Manganese Compounds/administration & dosage , Manganese Compounds/metabolism , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese Poisoning/pathology , Manganese Poisoning/physiopathology , Mice, Inbred ICR , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neuronal Plasticity/drug effects , Parvalbumins/genetics , Parvalbumins/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Time Factors , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL