Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Glia ; 70(10): 1886-1901, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35638297

RESUMEN

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.


Asunto(s)
Astrocitos , Intoxicación por Manganeso , Proteínas Represoras , Animales , Astrocitos/metabolismo , Eliminación de Gen , Humanos , Manganeso/toxicidad , Intoxicación por Manganeso/genética , Ratones , Proteínas Represoras/genética
2.
Neurobiol Dis ; 158: 105467, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34358615

RESUMEN

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.


Asunto(s)
Conducta Animal , Proteínas de Transporte de Catión/genética , Distonía/inducido químicamente , Intoxicación por Manganeso/metabolismo , Intoxicación por Manganeso/psicología , Enfermedad de Parkinson Secundaria/inducido químicamente , Animales , Encéfalo/metabolismo , Dopamina/metabolismo , Distonía/genética , Femenino , Masculino , Intoxicación por Manganeso/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Neostriado/metabolismo , Enfermedad de Parkinson Secundaria/genética , Desempeño Psicomotor , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
3.
Neurotoxicology ; 80: 71-75, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32621835

RESUMEN

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.


Asunto(s)
Intoxicación por Cadmio/metabolismo , Caenorhabditis elegans/metabolismo , Neuronas Dopaminérgicas/metabolismo , Intoxicación por Manganeso/metabolismo , Degeneración Nerviosa , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Neuronas Serotoninérgicas/metabolismo , Animales , Animales Modificados Genéticamente , Cloruro de Cadmio , Intoxicación por Cadmio/etiología , Intoxicación por Cadmio/genética , Intoxicación por Cadmio/patología , Caenorhabditis elegans/genética , Cloruros , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Glucosa/toxicidad , Compuestos de Manganeso , Intoxicación por Manganeso/etiología , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/patología , Receptor para Productos Finales de Glicación Avanzada/genética , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/patología
4.
Int J Mol Sci ; 21(9)2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32392784

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Manganeso/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Homeostasis , Humanos , Absorción Intestinal , Manganeso/deficiencia , Intoxicación por Manganeso/genética , Mutación
5.
Environ Toxicol Pharmacol ; 64: 60-69, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30300793

RESUMEN

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.


Asunto(s)
Cuerpo Estriado/metabolismo , Predisposición Genética a la Enfermedad , Intoxicación por Manganeso/genética , Animales , Epistasis Genética , Masculino , Ratas Sprague-Dawley , Transcriptoma
6.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 36(12): 930-934, 2018 Dec 20.
Artículo en Chino | MEDLINE | ID: mdl-30812084

RESUMEN

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.


Asunto(s)
Expresión Génica/fisiología , Intoxicación por Manganeso/genética , Redes y Vías Metabólicas/genética , Animales , Masculino , Fosfatidilinositol 3-Quinasas , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
7.
Toxicol Appl Pharmacol ; 336: 94-100, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29054681

RESUMEN

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.


Asunto(s)
Cloruros/toxicidad , Hemo Oxigenasa (Desciclizante)/metabolismo , Histonas/metabolismo , Intoxicación por Manganeso/etiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Acetilación , Transporte Activo de Núcleo Celular , Ácidos Anacárdicos/farmacología , Animales , Glutatión/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Compuestos de Manganeso , Intoxicación por Manganeso/tratamiento farmacológico , Intoxicación por Manganeso/enzimología , Intoxicación por Manganeso/genética , Factor 2 Relacionado con NF-E2/genética , Neuronas/enzimología , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Adv Neurobiol ; 18: 35-49, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28889262

RESUMEN

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.


Asunto(s)
Enfermedades Carenciales/metabolismo , Intoxicación por Manganeso/metabolismo , Manganeso/metabolismo , Errores Innatos del Metabolismo de los Metales/metabolismo , Proteínas de Transporte de Catión/genética , Enfermedades Carenciales/genética , Enfermedades Carenciales/psicología , Humanos , Manganeso/deficiencia , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/psicología , Errores Innatos del Metabolismo de los Metales/genética , Errores Innatos del Metabolismo de los Metales/psicología , Zinc/deficiencia , Transportador 8 de Zinc/genética
9.
J Neurosci ; 37(25): 5996-6006, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28536273

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/metabolismo , Manganeso/metabolismo , Actividad Motora/genética , Animales , Química Encefálica/genética , Femenino , Motilidad Gastrointestinal/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Tisular , Zinc/metabolismo , Zinc/farmacología
10.
J Toxicol Sci ; 41(5): 573-81, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27665767

RESUMEN

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.


Asunto(s)
Ácido Aminosalicílico/farmacología , Astrocitos/efectos de los fármacos , Ganglios Basales/efectos de los fármacos , Cloruros/toxicidad , Daño del ADN/efectos de los fármacos , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Glicina/metabolismo , Intoxicación por Manganeso/prevención & control , Sustancias Protectoras/farmacología , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Astrocitos/patología , Ganglios Basales/metabolismo , Ganglios Basales/patología , Células Cultivadas , Citoprotección , Relación Dosis-Respuesta a Droga , Compuestos de Manganeso , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/metabolismo , Intoxicación por Manganeso/patología , Ratas Sprague-Dawley
11.
Toxicol Lett ; 235(1): 17-27, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25791630

RESUMEN

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.


Asunto(s)
Apoptosis , Ganglios Basales/enzimología , Intoxicación por Manganeso/enzimología , Neuronas/enzimología , Fosfoproteínas Fosfatasas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ganglios Basales/patología , Cloruros , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Compuestos de Manganeso , Intoxicación por Manganeso/etiología , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/patología , Degeneración Nerviosa , Neuronas/efectos de los fármacos , Neuronas/patología , Células PC12 , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2C , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transfección , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
12.
Toxicology ; 331: 24-34, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25698507

RESUMEN

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.


Asunto(s)
Cloruros/toxicidad , Neuronas GABAérgicas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Interneuronas/efectos de los fármacos , Intoxicación por Manganeso/etiología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Administración Oral , Animales , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cloruros/administración & dosificación , Cloruros/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Interneuronas/metabolismo , Interneuronas/patología , Compuestos de Manganeso/administración & dosificación , Compuestos de Manganeso/metabolismo , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/metabolismo , Intoxicación por Manganeso/patología , Intoxicación por Manganeso/fisiopatología , Ratones Endogámicos ICR , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Plasticidad Neuronal/efectos de los fármacos , Parvalbúminas/genética , Parvalbúminas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Tiempo , Ácido gamma-Aminobutírico/metabolismo
13.
Toxicology ; 328: 168-78, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25549921

RESUMEN

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.


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Encéfalo/efectos de los fármacos , Exposición por Inhalación/prevención & control , Intoxicación por Manganeso/prevención & control , Manganeso/toxicidad , Enfermedad de Parkinson Secundaria/prevención & control , Soldadura/métodos , Aerosoles , Contaminantes Ocupacionales del Aire/química , Animales , Carga Corporal (Radioterapia) , Encéfalo/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Diseño de Equipo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Exposición por Inhalación/efectos adversos , Masculino , Manganeso/química , Intoxicación por Manganeso/etiología , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/metabolismo , Enfermedad de Parkinson Secundaria/etiología , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/metabolismo , Tamaño de la Partícula , Ratas Sprague-Dawley , Medición de Riesgo , Solubilidad , Factores de Tiempo , Soldadura/instrumentación
14.
Toxicol Sci ; 143(2): 454-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25416158

RESUMEN

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.


Asunto(s)
Cloruros/toxicidad , Neuronas Dopaminérgicas/efectos de los fármacos , Intoxicación por Manganeso/genética , Modelos Neurológicos , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Sitios de Unión , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cloruros/metabolismo , Fragmentación del ADN/efectos de los fármacos , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Compuestos de Manganeso/metabolismo , Intoxicación por Manganeso/complicaciones , Intoxicación por Manganeso/patología , Intoxicación por Manganeso/prevención & control , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/prevención & control , Unión Proteica , Ratas , Especies Reactivas de Oxígeno/metabolismo , Espectrofotometría Atómica , Transfección , alfa-Sinucleína/metabolismo
15.
J Neurol Sci ; 341(1-2): 150-2, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24746291

RESUMEN

Manganese (Mn) toxicity causes an extrapyramidal, parkinsonian-type movement disorder with characteristic magnetic resonance images of Mn accumulation in the basal ganglia. This letter highlights the neurological manifestations and neuroimaging features of inherited manganism (IMn), an unusual and treatable inborn error of Mn homeostasis. Early-onset dystonia with "cock-walk" gait and hyperintense signal in basal ganglia, associated to polycythemia, chronic liver disease and hypermanganesemia, promptly suggest IMn, and a genetic evaluation should be performed.


Asunto(s)
Trastornos Neurológicos de la Marcha , Intoxicación por Manganeso , Neuroimagen , Proteínas de Transporte de Catión/genética , Niño , Femenino , Trastornos Neurológicos de la Marcha/complicaciones , Trastornos Neurológicos de la Marcha/genética , Trastornos Neurológicos de la Marcha/patología , Humanos , Intoxicación por Manganeso/complicaciones , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/patología , Transportador 8 de Zinc
16.
Artículo en Chino | MEDLINE | ID: mdl-24370358

RESUMEN

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.


Asunto(s)
Intoxicación por Manganeso/genética , Síndromes de Neurotoxicidad/genética , Superóxido Dismutasa/genética , Adulto , Animales , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Células PC12 , ARN Mensajero/genética , Ratas
17.
Toxicol Sci ; 136(1): 154-65, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23976782

RESUMEN

We have shown that maternal manganese (Mn) exposure caused sustained disruption of hippocampal neurogenesis of mouse offspring. To clarify the effects of maternal Mn exposure on epigenetic gene regulation contributing to the sustained disruption of hippocampal neurogenesis, we treated pregnant ICR mice with MnCl2 in diet from gestational day 10 through day 21 after delivery on weaning and searched epigenetically downregulated genes by global promoter methylation analysis in the hippocampal dentate gyrus of male offspring on postnatal day (PND) 21 and PND 77. By CpG promoter microarray analysis on PND 21 following 800-ppm Mn exposure, sustained promoter hypermethylation and transcript downregulation through PND 77 were confirmed with Mid1, Atp1a3, and Nr2f1, whereas Pvalb showed a transient hypermethylation only on weaning. The numbers of Pvalb⁺ and ATP1a3⁺ neurons suggestive of γ-aminobutyric acid (GABA)ergic interneurons, Mid1⁺ cells suggestive of late-stage granule cell lineage and GABAergic interneurons, and COUP-TF1⁺ cells suggestive of early-stage granule cell lineage were all reduced on PND 21, and reductions were sustained on PND 77 except for no change in Pvalb⁺ cells. Mid1⁺ cells showed asymmetric distribution with right-side predominance, and Mn exposure abolished it by promoter hypermethylation of the right side. These findings indicate epigenetic mechanisms as mediators, through which Mn exposure modulates neurogenesis involving both granule cell lineage and GABAergic interneurons with long-lasting and stable repercussions. Disruption of asymmetric cellular distribution of Mid1 suggests that higher brain functions specialized in the left or right side of the brain were affected.


Asunto(s)
Cloruros/toxicidad , Giro Dentado/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Intoxicación por Manganeso/etiología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP I/metabolismo , Islas de CpG , Metilación de ADN/efectos de los fármacos , Giro Dentado/crecimiento & desarrollo , Giro Dentado/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Masculino , Compuestos de Manganeso , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/fisiopatología , Exposición Materna , Ratones , Ratones Endogámicos ICR , Neurogénesis/genética , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Parvalbúminas/genética , Parvalbúminas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Regiones Promotoras Genéticas , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ubiquitina-Proteína Ligasas , Destete
19.
Neurotoxicology ; 35: 121-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23313730

RESUMEN

Chronic exposure to Mn results in the development of a neurological disorder known as manganism characterized by neurological deficits resembling that seen in Parkinsonism. Although dopaminergic neurons within the nigrostriatal pathway appear intact, Mn-induced irregularities in DA transmission have been observed including decreased amphetamine-induced DA release and loss of the dopamine transporter (DAT). Results of studies to evaluate the effect of Mn and DA on cell viability in control and DAT-transfected HEK cells reveal that Mn is equally toxic to both cell lines whereas DA was only toxic to cells containing DAT. DA toxicity was saturable suggesting that transport may be rate limiting. When Mn and DA were added simultaneously to the media, cell toxicity was similar to that produced by Mn alone suggesting that Mn may suppress DA uptake in the DAT containing cells. Preincubation of DA prior to the addition of Mn resulted in cell death which was essentially additive with that produced independently by the two agents. Mn was also shown to decrease DA uptake and amphetamine-induced DA efflux in DAT containing cells. Time-lapsed confocal microscopy indicates that Mn can promote trafficking of cell surface DAT into intracellular compartments which may account for the decrease in DA uptake and DA efflux in these cells. Mn-induced internalization of DAT may provide an explanation for disruption in DA transmission previously reported in the striatum.


Asunto(s)
Cloruros/toxicidad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/efectos de los fármacos , Dopamina/toxicidad , Intoxicación por Manganeso/etiología , Anfetamina/farmacología , Supervivencia Celular/efectos de los fármacos , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Compuestos de Manganeso , Intoxicación por Manganeso/genética , Intoxicación por Manganeso/metabolismo , Intoxicación por Manganeso/patología , Potenciales de la Membrana , Microscopía Confocal , Transporte de Proteínas , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Imagen de Lapso de Tiempo , Transfección
20.
Neurotoxicology ; 33(6): 1443-1449, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23099318

RESUMEN

Manganese (Mn) is an environmental risk factor for Parkinson's disease (PD). Recessive inheritance of PARK2 mutations is strongly associated with early onset PD (EOPD). It is widely assumed that the influence of PD environmental risk factors may be enhanced by the presence of PD genetic risk factors in the genetic background of individuals. However, such interactions may be difficult to predict owing to the complexities of genetic and environmental interactions. Here we examine the potential of human induced pluripotent stem (iPS) cell-derived early neural progenitor cells (NPCs) to model differences in Mn neurotoxicity between a control subject (CA) with no known PD genetic risk factors and a subject (SM) with biallelic loss-of-function mutations in PARK2 and family history of PD but no evidence of PD by neurological exam. Human iPS cells were generated from primary dermal fibroblasts of both subjects. We assessed several outcome measures associated with Mn toxicity and PD. No difference in sensitivity to Mn cytotoxicity or mitochondrial fragmentation was observed between SM and CA NPCs. However, we found that Mn exposure was associated with significantly higher reactive oxygen species (ROS) generation in SM compared to CA NPCs despite significantly less intracellular Mn accumulation. Thus, this report offers the first example of human subject-specific differences in PD-relevant environmental health related phenotypes that are consistent with pathogenic interactions between known genetic and environmental risk factors for PD.


Asunto(s)
Cloruros/toxicidad , Contaminantes Ambientales/toxicidad , Interacción Gen-Ambiente , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Intoxicación por Manganeso/genética , Mutación , Células-Madre Neurales/efectos de los fármacos , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson/genética , Ubiquitina-Proteína Ligasas/genética , Técnicas Biosensibles , Estudios de Casos y Controles , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cloruros/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Compuestos de Manganeso/metabolismo , Intoxicación por Manganeso/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Examen Neurológico , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson Secundaria/diagnóstico , Enfermedad de Parkinson Secundaria/metabolismo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA