Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Behav Brain Res ; 465: 114969, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38548024

RESUMO

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.


Assuntos
Intoxicação por Manganês , Doença de Parkinson , Ratos , Animais , Manganês/toxicidade , Óleo de Gergelim/farmacologia , Doença de Parkinson/tratamento farmacológico , Estresse Oxidativo , Intoxicação por Manganês/tratamento farmacológico , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/patologia
2.
Neurotoxicology ; 95: 1-11, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36621467

RESUMO

Manganese (Mn) is an essential metal that serves as a cofactor for metalloenzymes important in moderating oxidative stress and the glutamate/glutamine cycle. Mn is typically obtained through the diet, but toxic overexposure can occur through other environmental or occupational exposure routes such as inhalation. Mn is known to accumulate in the brain following exposure and may contribute to the etiology of neurodegenerative disorders such as Alzheimer's disease (AD) even in the absence of acute neurotoxicity. In the present study, we used in vitro primary cell culture, ex vivo slice electrophysiology and in vivo behavioral approaches to determine if Mn-induced changes in glutamatergic signaling may be altered by genetic risk factors for AD neuropathology. Primary cortical astrocytes incubated with Mn exhibited early rapid clearance of glutamate compared to saline treated astrocytes but decreased clearance over longer time periods, with no effect of the AD genotype. Further, we found that in vivo exposure to a subcutaneous subacute, high dose of Mn as manganese chloride tetrahydrate (3 ×50 mg/kg MnCl2·4(H2O) over 7 days) resulted in increased expression of cortical GLAST protein regardless of genotype, with no changes in GLT-1. Hippocampal long-term potentiation was not altered in APP/PSEN1 mice at this age and neither was it disrupted following Mn exposure. Mn exposure did increase sensitivity to seizure onset following treatment with the excitatory agonist kainic acid, with differing responses between APP/PSEN1 and control mice. These results highlight the sensitivity of the glutamatergic system to Mn exposure. Experiments were performed in young adult APP/PSEN1 mice, prior to cognitive decline or accumulation of hallmark amyloid plaque pathology and following subacute exposure to Mn. The data support a role of Mn in pathophysiology of AD in early stages of the disease and support the need to better understand neurological consequences of Mn exposure in vulnerable populations.


Assuntos
Doença de Alzheimer , Intoxicação por Manganês , Animais , Camundongos , Manganês/toxicidade , Manganês/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Intoxicação por Manganês/metabolismo , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo
3.
Neurotherapeutics ; 17(4): 1878-1896, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32959271

RESUMO

The molecular mechanism of Alzheimer-like cognitive impairment induced by manganese (Mn) exposure has not yet been fully clarified, and there are currently no effective interventions to treat neurodegenerative lesions related to manganism. Protein phosphatase 2 A (PP2A) is a major tau phosphatase and was recently identified as a potential therapeutic target molecule for neurodegenerative diseases; its activity is directed by the methylation status of the catalytic C subunit. Methionine is an essential amino acid, and its downstream metabolite S-adenosylmethionine (SAM) participates in transmethylation pathways as a methyl donor. In this study, the neurotoxic mechanism of Mn and the protective effect of methionine were evaluated in Mn-exposed cell and rat models. We show that Mn-induced neurotoxicity is characterized by PP2Ac demethylation accompanied by abnormally decreased LCMT-1 and increased PME-1, which are associated with tau hyperphosphorylation and spatial learning and memory deficits, and that the poor availability of SAM in the hippocampus is likely to determine the loss of PP2Ac methylation. Importantly, maintenance of local SAM levels through continuous supplementation with exogenous methionine, or through specific inhibition of PP2Ac demethylation by ABL127 administration in vitro, can effectively prevent tau hyperphosphorylation to reduce cellular oxidative stress, apoptosis, damage to cell viability, and rat memory deficits in cell or animal Mn exposure models. In conclusion, our data suggest that SAM and PP2Ac methylation may be novel targets for the treatment of Mn poisoning and neurotoxic mechanism-related tauopathies.


Assuntos
Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Metionina/metabolismo , Proteína Fosfatase 2/metabolismo , Tauopatias/induzido quimicamente , Tauopatias/metabolismo , Animais , Linhagem Celular Tumoral , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Intoxicação por Manganês/patologia , Metilação/efeitos dos fármacos , Camundongos , Ratos , Ratos Sprague-Dawley , Tauopatias/patologia
4.
Arch Toxicol ; 94(10): 3409-3420, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32875357

RESUMO

Manganese (Mn) is a neurotoxicant that, due to its paramagnetic property, also functions as a magnetic resonance imaging (MRI) T1 contrast agent. Previous studies in Mn toxicity have shown that Mn accumulates in the brain, which may lead to parkinsonian symptoms. In this article, we trained support vector machines (SVM) using whole-brain R1 (R1 = 1/T1) maps from 57 welders and 32 controls to classify subjects based on their air Mn concentration ([Mn]Air), Mn brain accumulation (ExMnBrain), gross motor dysfunction (UPDRS), thalamic GABA concentration (GABAThal), and total years welding. R1 was highly predictive of [Mn]Air above a threshold of 0.20 mg/m3 with an accuracy of 88.8% and recall of 88.9%. R1 was also predictive of subjects with GABAThal having less than or equal to 2.6 mM with an accuracy of 82% and recall of 78.9%. Finally, we used an SVM to predict age as a method of verifying that the results could be attributed to Mn exposure. We found that R1 was predictive of age below 48 years of age with accuracies ranging between 75 and 82% with recall between 94.7% and 76.9% but was not predictive above 48 years of age. Together, this suggests that lower levels of exposure (< 0.20 mg/m3 and < 18 years of welding on the job) do not produce discernable signatures, whereas higher air exposures and subjects with more total years welding produce signatures in the brain that are readily identifiable using SVM.


Assuntos
Poluentes Ocupacionais do Ar/toxicidade , Encéfalo/metabolismo , Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Exposição Ocupacional , Adulto , Fatores Etários , Poluentes Ocupacionais do Ar/metabolismo , Química Encefálica , Humanos , Imageamento por Ressonância Magnética , Masculino , Manganês/metabolismo , Ferreiros , Pessoa de Meia-Idade , Modelos Biológicos , Transtornos dos Movimentos/diagnóstico , Transtornos dos Movimentos/metabolismo , Máquina de Vetores de Suporte , Tálamo/diagnóstico por imagem , Tálamo/metabolismo , Soldagem , Adulto Jovem , Ácido gama-Aminobutírico/análise
5.
J Biol Chem ; 295(46): 15662-15676, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32893191

RESUMO

Manganese (Mn)-induced neurotoxicity resembles Parkinson's disease (PD), but the mechanisms underpinning its effects remain unknown. Mn dysregulates astrocytic glutamate transporters, GLT-1 and GLAST, and dopaminergic function, including tyrosine hydroxylase (TH). Our previous in vitro studies have shown that Mn repressed GLAST and GLT-1 via activation of transcription factor Yin Yang 1 (YY1). Here, we investigated if in vivo astrocytic YY1 deletion mitigates Mn-induced dopaminergic neurotoxicity, attenuating Mn-induced reduction in GLAST/GLT-1 expression in murine substantia nigra (SN). AAV5-GFAP-Cre-GFP particles were infused into the SN of 8-week-old YY1 flox/flox mice to generate a region-specific astrocytic YY1 conditional knockout (cKO) mouse model. 3 weeks after adeno-associated viral (AAV) infusion, mice were exposed to 330 µg of Mn (MnCl2 30 mg/kg, intranasal instillation, daily) for 3 weeks. After Mn exposure, motor functions were determined in open-field and rotarod tests, followed by Western blotting, quantitative PCR, and immunohistochemistry to assess YY1, TH, GLAST, and GLT-1 levels. Infusion of AAV5-GFAP-Cre-GFP vectors into the SN resulted in region-specific astrocytic YY1 deletion and attenuation of Mn-induced impairment of motor functions, reduction of TH-expressing cells in SN, and TH mRNA/protein levels in midbrain/striatum. Astrocytic YY1 deletion also attenuated the Mn-induced decrease in GLAST/GLT-1 mRNA/protein levels in midbrain. Moreover, YY1 deletion abrogated its interaction with histone deacetylases in astrocytes. These results indicate that astrocytic YY1 plays a critical role in Mn-induced neurotoxicity in vivo, at least in part, by reducing astrocytic GLAST/GLT-1. Thus, YY1 might be a potential target for treatment of Mn toxicity and other neurological disorders associated with dysregulation of GLAST/GLT-1.


Assuntos
Intoxicação por Manganês/patologia , Substância Negra/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Cloretos/toxicidade , Regulação para Baixo/efeitos dos fármacos , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Feminino , Histona Desacetilases/metabolismo , Locomoção/efeitos dos fármacos , Masculino , Compostos de Manganês , Intoxicação por Manganês/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Fator de Transcrição YY1/genética
6.
Expert Rev Neurother ; 19(3): 243-260, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30759034

RESUMO

INTRODUCTION: Iron (Fe) and manganese (Mn) are essential nutrients for humans. They act as cofactors for a variety of enzymes. In the central nervous system (CNS), these two metals are involved in diverse neurological activities. Dyshomeostasis may interfere with the critical enzymatic activities, hence altering the neurophysiological status and resulting in neurological diseases. Areas covered: In this review, the authors cover the molecular mechanisms of Fe/Mn-induced toxicity and neurological diseases, as well as the diagnosis and potential treatment. Given that both Fe and Mn are abundant in the earth crust, nutritional deficiency is rare. In this review the authors focus on the neurological disorders associated with Mn and Fe overload. Expert commentary: Oxidative stress and mitochondrial dysfunction are the primary molecular mechanism that mediates Fe/Mn-induced neurotoxicity. Although increased Fe or Mn concentrations have been found in brain of patients, it remains controversial whether the elevated metal amounts are the primary cause or secondary consequence of neurological diseases. Currently, treatments are far from satisfactory, although chelation therapy can significantly decrease brain Fe and Mn levels. Studies to determine the primary cause and establish the molecular mechanism of toxicity may help to adapt more comprehensive and satisfactory treatments in the future.


Assuntos
Doenças do Sistema Nervoso Central/induzido quimicamente , Ferro/intoxicação , Intoxicação por Manganês/diagnóstico , Intoxicação por Manganês/tratamento farmacológico , Animais , Encéfalo/metabolismo , Doenças do Sistema Nervoso Central/diagnóstico , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/metabolismo , Humanos , Ferro/metabolismo , Intoxicação por Manganês/metabolismo , Estresse Oxidativo
7.
Neurotoxicology ; 64: 30-42, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28873337

RESUMO

Excessive occupational exposure to Manganese (Mn) has been associated with clinical symptoms resembling idiopathic Parkinson's disease (IPD), impairing cognitive and motor functions. Several studies point towards an involvement of the brain neurotransmitter system in Mn intoxication, which is hypothesized to be disturbed prior to onset of symptoms. Edited Magnetic Resonance Spectroscopy (MRS) offers the unique possibility to measure γ-amminobutyric acid (GABA) and other neurometabolites in vivo non-invasively in workers exposed to Mn. In addition, the property of Mn as Magnetic Resonance Imaging (MRI) contrast agent may be used to study Mn deposition in the human brain. In this study, using MRI, MRS, personal air sampling at the working place, work history questionnaires, and neurological assessment (UPDRS-III), the effects of chronic Mn exposure on the thalamic GABAergic system was studied in a group of welders (N=39) with exposure to Mn fumes in a typical occupational setting. Two subgroups of welders with different exposure levels (Low: N=26; mean air Mn=0.13±0.1mg/m3; High: N=13; mean air Mn=0.23±0.18mg/m3), as well as unexposed control workers (N=22, mean air Mn=0.002±0.001mg/m3) were recruited. The group of welders with higher exposure showed a significant increase of thalamic GABA levels by 45% (p<0.01, F(1,33)=9.55), as well as significantly worse performance in general motor function (p<0.01, F(1,33)=11.35). However, welders with lower exposure did not differ from the controls in GABA levels or motor performance. Further, in welders the thalamic GABA levels were best predicted by past-12-months exposure levels and were influenced by the Mn deposition in the substantia nigra and globus pallidus. Importantly, both thalamic GABA levels and motor function displayed a non-linear pattern of response to Mn exposure, suggesting a threshold effect.


Assuntos
Intoxicação por Manganês/diagnóstico por imagem , Exposição Ocupacional , Tálamo/metabolismo , Soldagem , Ácido gama-Aminobutírico/metabolismo , Adulto , Poluentes Ocupacionais do Ar/intoxicação , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Intoxicação por Manganês/metabolismo , Pessoa de Meia-Idade , Tálamo/diagnóstico por imagem
8.
Adv Neurobiol ; 16: 1-12, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28828603

RESUMO

Manganese (Mn) is an essential trace element, serving as a cofactor for several enzymes involved in various cellular and biochemical reactions in human body. However, chronic overexposure to Mn from occupational or environmental sources induces a neurological disorder, characterized by psychiatric, cognitive, and motor abnormalities, referred to as manganism. Mn-induced neurotoxicity is known to target astrocytes since these cells preferentially accumulate Mn. Astrocytes are the most abundant non-neuronal glial cells in the brain, and they play a critical role in maintaining the optimal glutamate levels to prevent excitotoxic death. The fine regulation of glutamate in the brain is accomplished by two major glutamate transporters - glutamate transporter-1 (GLT-1) and glutamate aspartate transporter (GLAST) that are predominantly expressed in astrocytes. Excitotoxic neuronal injury has been demonstrated as a critical mechanism involved in Mn neurotoxicity and implicated in the pathological signs of multiple neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Recent evidences also establish that Mn directly deregulates the expression and function of both astrocytic glutamate transporters by decreasing mRNA and protein levels of GLT-1 and GLAST. Herein, we will review the mechanisms of Mn-induced gene regulation of glutamate transporters at the transcriptional level and their role in Mn toxicity.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Manganês/metabolismo , Manganês/toxicidade , Síndromes Neurotóxicas/metabolismo , Animais , Astrócitos/metabolismo , Regulação da Expressão Gênica , Humanos , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/fisiopatologia , Síndromes Neurotóxicas/fisiopatologia
9.
BMC Pharmacol Toxicol ; 17(1): 57, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27814772

RESUMO

Manganese (Mn) is an essential heavy metal. However, Mn's nutritional aspects are paralleled by its role as a neurotoxicant upon excessive exposure. In this review, we covered recent advances in identifying mechanisms of Mn uptake and its molecular actions in the brain as well as promising neuroprotective strategies. The authors focused on reporting findings regarding Mn transport mechanisms, Mn effects on cholinergic system, behavioral alterations induced by Mn exposure and studies of neuroprotective strategies against Mn intoxication. We report that exposure to Mn may arise from environmental sources, occupational settings, food, total parenteral nutrition (TPN), methcathinone drug abuse or even genetic factors, such as mutation in the transporter SLC30A10. Accumulation of Mn occurs mainly in the basal ganglia and leads to a syndrome called manganism, whose symptoms of cognitive dysfunction and motor impairment resemble Parkinson's disease (PD). Various neurotransmitter systems may be impaired due to Mn, especially dopaminergic, but also cholinergic and GABAergic. Several proteins have been identified to transport Mn, including divalent metal tranporter-1 (DMT-1), SLC30A10, transferrin and ferroportin and allow its accumulation in the central nervous system. Parallel to identification of Mn neurotoxic properties, neuroprotective strategies have been reported, and these include endogenous antioxidants (for instance, vitamin E), plant extracts (complex mixtures containing polyphenols and non-characterized components), iron chelating agents, precursors of glutathione (GSH), and synthetic compounds that can experimentally afford protection against Mn-induced neurotoxicity.


Assuntos
Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/prevenção & controle , Manganês/toxicidade , Transtornos das Habilidades Motoras/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Animais , Encéfalo/metabolismo , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/metabolismo , Alimentos/efeitos adversos , Humanos , Manganês/metabolismo , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/prevenção & controle , Transtornos das Habilidades Motoras/induzido quimicamente , Transtornos das Habilidades Motoras/metabolismo , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/prevenção & controle
10.
Am J Clin Nutr ; 102(6): 1482-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26561627

RESUMO

BACKGROUND: Manganese, an essential metal for normal growth and development, is neurotoxic on excessive exposure. Standard trace element-supplemented neonatal parenteral nutrition (PN) has a high manganese content and bypasses normal gastrointestinal absorptive control mechanisms, which places infants at risk of manganese neurotoxicity. Magnetic resonance (MR) relaxometry demonstrating short T1 relaxation time (T1R) in the basal ganglia reflects excessive brain manganese accumulation. OBJECTIVE: This study tested the hypothesis that infants with greater parenteral manganese exposure have higher brain manganese accumulation, as measured by MR imaging, than do infants with lower parenteral manganese exposure. DESIGN: Infants exposed to parenteral manganese were enrolled in a prospective cohort study. Infants classified as having high manganese exposure received >75% of their nutrition in the preceding 4 wk as PN. All others were classified as having low exposure. Daily parenteral and enteral manganese intakes were calculated. Whole-blood manganese was measured by high-resolution inductively coupled plasma mass spectrometry. Brain MR relaxometry was interpreted by a masked reviewer. Linear regression models, adjusted for gestational age (GA) at birth, estimated the association of relaxometry indexes with total and parenteral manganese exposures. RESULTS: Seventy-three infants were enrolled. High-quality MR images were available for 58 infants, 39 with high and 19 with low manganese exposure. Four infants with a high exposure had blood manganese concentrations >30 µg/L. After controlling for GA, higher parenteral and total manganese intakes were associated with a lower T1R (P = 0.01) in the globus pallidus and putamen but were not associated with whole-blood manganese (range: 3.6-56.6 µg/L). Elevated conjugated bilirubin magnified the association between parenteral manganese and decreasing T1R. CONCLUSION: A short T1R for GA identifies infants at risk of increased brain manganese deposition associated with PN solutions commonly used to nourish critically ill infants. These trials were registered at clinicaltrials.gov as NCT00392977 and NCT00392730.


Assuntos
Gânglios da Base/metabolismo , Desenvolvimento Infantil , Fenômenos Fisiológicos da Nutrição do Lactente , Intoxicação por Manganês/diagnóstico , Manganês/metabolismo , Neurônios/metabolismo , Nutrição Parenteral/efeitos adversos , Bilirrubina/análogos & derivados , Bilirrubina/sangue , Estudos de Coortes , Feminino , Globo Pálido/metabolismo , Hospitais Pediátricos , Hospitais Universitários , Humanos , Lactente , Unidades de Terapia Intensiva Neonatal , Imageamento por Ressonância Magnética , Masculino , Manganês/sangue , Intoxicação por Manganês/sangue , Intoxicação por Manganês/etiologia , Intoxicação por Manganês/metabolismo , Neuroimagem , Projetos Piloto , Estudos Prospectivos , Putamen/metabolismo
11.
Neurochem Int ; 88: 53-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25128239

RESUMO

Astrocytes are the most abundant non-neuronal glial cells in the brain. Once relegated to a mere supportive role for neurons, contemporary dogmas ascribe multiple active roles for these cells in central nervous system (CNS) function, including maintenance of optimal glutamate levels in synapses. Regulation of glutamate levels in the synaptic cleft is crucial for preventing excitotoxic neuronal injury. Glutamate levels are regulated predominantly by two astrocytic glutamate transporters, glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST). Indeed, the dysregulation of these transporters has been linked to several neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD), as well as manganism, which is caused by overexposure to the trace metal, manganese (Mn). Although Mn is an essential trace element, its excessive accumulation in the brain as a result of chronic occupational or environmental exposures induces a neurological disorder referred to as manganism, which shares common pathological features with Parkinsonism. Mn decreases the expression and function of both GLAST and GLT-1. Astrocytes are commonly targeted by Mn, and thus reduction in astrocytic glutamate transporter function represents a critical mechanism of Mn-induced neurotoxicity. In this review, we will discuss the role of astrocytic glutamate transporters in neurodegenerative diseases and Mn-induced neurotoxicity.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Astrócitos/metabolismo , Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Fator de Transcrição YY1/fisiologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Astrócitos/efeitos dos fármacos , Humanos , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia
12.
Int Rev Neurobiol ; 110: 277-312, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24209443

RESUMO

Manganese (Mn) is an essential trace metal that is pivotal for normal cell function and metabolism. Its homeostasis is tightly regulated; however, the mechanisms of Mn homeostasis are poorly characterized. While a number of proteins such as the divalent metal transporter 1, the transferrin/transferrin receptor complex, the ZIP family metal transporters ZIP-8 and ZIP-14, the secretory pathway calcium ATPases SPCA1 and SPCA2, ATP13A2, and ferroportin have been suggested to play a role in Mn transport, the degree that each of them contributes to Mn homeostasis has still to be determined. The recent discovery of SLC30A10 as a crucial Mn transporter in humans has shed further light on our understanding of Mn transport across the cell. Although essential, Mn is toxic at high concentrations. Mn neurotoxicity has been attributed to impaired dopaminergic (DAergic), glutamatergic and GABAergic transmission, mitochondrial dysfunction, oxidative stress, and neuroinflammation. As a result of preferential accumulation of Mn in the DAergic cells of the basal ganglia, particularly the globus pallidus, Mn toxicity causes extrapyramidal motor dysfunction. Firstly described as "manganism" in miners during the nineteenth century, this movement disorder resembles Parkinson's disease characterized by hypokinesia and postural instability. To date, a variety of acquired causes of brain Mn accumulation can be distinguished from an autosomal recessively inherited disorder of Mn metabolism caused by mutations in the SLC30A10 gene. Both, acquired and inherited hypermanganesemia, lead to Mn deposition in the basal ganglia associated with pathognomonic magnetic resonance imaging appearances of hyperintense basal ganglia on T1-weighted images. Current treatment strategies for Mn toxicity combine chelation therapy to reduce the body Mn load and iron (Fe) supplementation to reduce Mn binding to proteins that interact with both Mn and Fe. This chapter summarizes our current understanding of Mn homeostasis and the mechanisms of Mn toxicity and highlights the clinical disorders associated with Mn neurotoxicity.


Assuntos
Encéfalo/metabolismo , Intoxicação por Manganês/metabolismo , Manganês/metabolismo , Animais , Homeostase/fisiologia , Humanos , Manganês/toxicidade
13.
Am J Hum Genet ; 90(3): 467-77, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22341971

RESUMO

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.


Assuntos
Proteínas de Transporte de Cátions/genética , Intoxicação por Manganês/genética , Proteínas de Membrana Transportadoras/genética , Doenças Metabólicas/genética , Transtornos Parkinsonianos/genética , Idoso , Sequência de Aminoácidos , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Mapeamento Cromossômico/métodos , Feminino , Mutação da Fase de Leitura/genética , Genes Recessivos , Predisposição Genética para Doença , Células Hep G2 , Homozigoto , Humanos , Imuno-Histoquímica/métodos , Fígado/metabolismo , Masculino , Manganês/metabolismo , Intoxicação por Manganês/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Doenças Metabólicas/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Alinhamento de Sequência/métodos , Células Tumorais Cultivadas , Transportador 8 de Zinco
14.
J Cereb Blood Flow Metab ; 31(12): 2324-33, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21712832

RESUMO

Manganese (Mn(2+))-enhanced magnetic resonance imaging studies of the neuronal pathways of the hypothalamus showed that information about the regulation of food intake and energy balance circulate through specific hypothalamic nuclei. The dehydration-induced anorexia (DIA) model demonstrated to be appropriate for studying the hypothalamus with Mn(2+)-enhanced magnetic resonance imaging. Manganese is involved in the normal functioning of a variety of physiological processes and is associated with enzymes contributing to neurotransmitter synthesis and metabolism. It also induces psychiatric and motor disturbances. The molecular mechanisms by which Mn(2+) produces alterations of the hypothalamic physiological processes are not well understood. (1)H-magnetic resonance spectroscopy measurements of the rodent hypothalamus are challenging due to the distant location of the hypothalamus resulting in limited measurement sensitivity. The present study proposed to investigate the effects of Mn(2+) on the neurochemical profile of the hypothalamus in normal, DIA, and overnight fasted female rats at 14.1 T. Results provide evidence that γ-aminobutyric acid has an essential role in the maintenance of energy homeostasis in the hypothalamus but is not condition specific. On the contrary, glutamine, glutamate, and taurine appear to respond more accurately to Mn(2+) exposure. An increase in glutamine levels could also be a characteristic response of the hypothalamus to DIA.


Assuntos
Cloretos/farmacologia , Hipotálamo/metabolismo , Compostos de Manganês/farmacologia , Aminoácidos/metabolismo , Animais , Anorexia/metabolismo , Desidratação/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Hipocampo/química , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Homeostase , Hipotálamo/química , Hipotálamo/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Intoxicação por Manganês/metabolismo , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
15.
Neurotoxicology ; 31(5): 572-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20188756

RESUMO

Chronic manganese (Mn) exposure produces a neurological syndrome with psychiatric, cognitive and parkinsonian features. Gene expression studies in the frontal cortex of Cynomolgus macaques exposed to different doses of Mn showed gene expression changes associated with cell cycle regulation, DNA repair, apoptosis, ubiquitin-proteasome system, protein folding, cholesterol homeostasis, axonal/vesicular transport and inflammation. Amyloid-beta (A-beta) precursor-like protein 1 (APLP1), a member of the amyloid precursor family, was the most highly up-regulated gene. Immunohistochemistry confirmed increased APLP1 expression and revealed the presence of A-beta diffuse plaques. Cortical neurons and white matter fibers from Mn-exposed animals exhibited accumulation of silver grains indicative of on-going degeneration. Cortical neurons also expressed nuclear hypertrophy, intracytoplasmic vacuoles, and apoptotis stigmata. The levels of p53 were increased in neurons and glial cells in Mn-exposed tissue. Analysis of another amyloidogenic protein, alpha-synuclein, also exhibited aggregation in the gray and white matter from Mn-exposed animals. In summary, chronic Mn exposure in non-human primates produces a cellular stress response leading to neurodegenerative changes, diffuse A-beta plaques and alpha-synuclein aggregation in the frontal cortex. These changes may help explain the cognitive and working memory deficits expressed by these animals.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Lobo Frontal/metabolismo , Intoxicação por Manganês , Manganês/toxicidade , Animais , Modelos Animais de Doenças , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/ultraestrutura , Macaca fascicularis , Intoxicação por Manganês/etiologia , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/patologia , Coloração pela Prata/métodos
16.
Neuromolecular Med ; 11(4): 252-66, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19657747

RESUMO

The purpose of this review is to highlight recent advances in the neuropathology associated with Mn exposures. We commence with a discussion on occupational manganism and clinical aspects of the disorder. This is followed by novel considerations on Mn transport (see also chapter by Yokel, this volume), advancing new hypotheses on the involvement of several transporters in Mn entry into the brain. This is followed by a brief description of the effects of Mn on neurotransmitter systems that are putative modulators of dopamine (DA) biology (the primary target of Mn neurotoxicity), as well as its effects on mitochondrial dysfunction and disruption of cellular energy metabolism. Next, we discuss inflammatory activation of glia in neuronal injury and how disruption of synaptic transmission and glial-neuronal communication may serve as underlying mechanisms of Mn-induced neurodegeneration commensurate with the cross-talk between glia and neurons. We conclude with a discussion on therapeutic aspects of Mn exposure. Emphasis is directed at treatment modalities and the utility of chelators in attenuating the neurodegenerative sequelae of exposure to Mn. For additional reading on several topics inherent to this review as well as others, the reader may wish to consult Aschner and Dorman (Toxicological Review 25:147-154, 2007) and Bowman et al. (Metals and neurodegeneration, 2009).


Assuntos
Intoxicação por Manganês/complicações , Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Exposição Ocupacional , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Animais , Transporte Biológico , Quelantes/uso terapêutico , Terapia por Quelação , Metabolismo Energético , Humanos , Manganês/metabolismo , Intoxicação por Manganês/tratamento farmacológico , Intoxicação por Manganês/patologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Mitocôndrias/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Doença de Parkinson Secundária/patologia , Ratos , Transmissão Sináptica/efeitos dos fármacos
17.
Neurotoxicology ; 30(2): 240-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19150464

RESUMO

Para-aminosalicylic acid (PAS), an FDA-approved anti-tuberculosis drug, has been used successfully in the treatment of severe manganese (Mn)-induced Parkinsonism in humans [Jiang Y-M, Mo X-A, Du FQ, Fu X, Zhu X-Y, Gao H-Y, et al. Effective treatment of manganese-induced occupational Parkinsonism with p-aminosalicylic acid: a case of 17-year follow-up study. J Occup Environ Med 2006;48:644-9]. This study was conducted to explore the capability of PAS in reducing Mn concentrations in body fluids and tissues of Mn-exposed animals. Sprague-Dawley rats received daily intraperitoneally (i.p.) injections of 6mg Mn/kg, 5 days/week for 4 weeks, followed by a daily subcutaneously (s.c.) dose of PAS (100 and 200mg/kg as the PAS-L and PAS-H group, respectively) for another 2, 3 or 6 weeks. Mn exposure significantly increased the concentrations of Mn in plasma, red blood cells (RBC), cerebrospinal fluid (CSF), brain and soft tissues. Following PAS-H treatment for 3 weeks, Mn levels in liver, heart, spleen and pancreas were significantly reduced by 25-33%, while 3 weeks of PAS-L treatment did not show any effect. Further therapy with PAS-H for 6 weeks reduced Mn levels in striatum, thalamus, choroid plexus, hippocampus and frontal cortex by 16-29% (p<0.05). Mn exposure greatly increased iron (Fe) and copper (Cu) concentrations in CSF, brain and liver. Treatment with PAS-H restored Fe and Cu levels comparable with control. These data suggest that PAS likely acts as a chelating agent to mobilize and remove tissue Mn. A high-dose and prolonged PAS treatment appears necessary for its therapeutic effectiveness.


Assuntos
Ácido Aminossalicílico/uso terapêutico , Terapia por Quelação/métodos , Intoxicação por Manganês/tratamento farmacológico , Manganês/toxicidade , Ácido Aminossalicílico/farmacologia , Animais , Masculino , Manganês/metabolismo , Intoxicação por Manganês/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
18.
Biol Pharm Bull ; 31(10): 1894-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18827350

RESUMO

Manganese (Mn) plays an important role in the etiology of several neurobehavioral disorders, but there is a lack of data regarding its specific effects on neurotransduction, especially dopaminergic neurotransduction. We investigated the relationship between motor deficits and alterations in the expression of tyrosine hydroxylase (TH) and dopamine D2-like receptors (DR), including the three dopaminergic subtypes, D2, D3, and D4, in low- and high-dose Mn-treated mice. After administration of Mn (intraperitoneal injections of 20 or 40 mg/kg MnCl(2).4H(2)O once per day for 5 d), motor activity and expression of TH and DR were examined in the striatum of the mouse brain. Mn treatment resulted in significant decrease in coordination and/or impaired motor learning after 5 d of treatment and this effect remained until 10 d after the end of Mn treatment. The expression of dopamine D2-like receptor D2 (DRD2), but not TH, DRD3, or DRD4, in the striatum was dose-dependent, and statistically significant increases were seen at the mRNA and protein levels. These findings indicate that Mn-induced motor deficits may be modulated in part by the expression of DRD2 in the striatum. In addition, our results suggest that the disturbance of dopaminergic neurotransmission mediated by DRD2 may be involved in the pathogenesis of Mn neurotoxicity.


Assuntos
Discinesia Induzida por Medicamentos/psicologia , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/psicologia , Neostriado/metabolismo , Receptores de Dopamina D2/biossíntese , Animais , Western Blotting , Densitometria , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Dopamina D3/biossíntese , Receptores de Dopamina D4/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina 3-Mono-Oxigenase/biossíntese
19.
Neuroscience ; 154(2): 848-60, 2008 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-18485605

RESUMO

In the present study, we examined whether exposing rats to a high-dose regimen of manganese chloride (Mn) during the postnatal period would depress presynaptic dopamine functioning and alter nonassociative and associative behaviors. To this end, rats were given oral supplements of Mn (750 microg/day) on postnatal days (PD) 1-21. On PD 90, dopamine transporter (DAT) immunoreactivity and [3H]dopamine uptake were assayed in the striatum and nucleus accumbens, while in vivo microdialysis was used to measure dopamine efflux in the same brain regions. The effects of postnatal Mn exposure on nigrostriatal functioning were evaluated by assessing rotorod performance and amphetamine-induced stereotypy in adulthood. In terms of associative processes, both cocaine-induced conditioned place preference (CPP) and sucrose-reinforced operant responding were examined. Results showed that postnatal Mn exposure caused persistent declines in DAT protein expression and [3H]dopamine uptake in the striatum and nucleus accumbens, as well as long-term reductions in striatal dopamine efflux. Rotorod performance did not differ according to exposure condition, however Mn-exposed rats did exhibit substantially more amphetamine-induced stereotypy than vehicle controls. Mn exposure did not alter performance on any aspect of the CPP task (preference, extinction, or reinstatement testing), nor did Mn affect progressive ratio responding (a measure of motivation). Interestingly, acquisition of a fixed ratio task was impaired in Mn-exposed rats, suggesting a deficit in procedural learning. In sum, these results indicate that postnatal Mn exposure causes persistent declines in various indices of presynaptic dopaminergic functioning. Mn-induced alterations in striatal functioning may have long-term impact on associative and nonassociative behavior.


Assuntos
Animais Recém-Nascidos/fisiologia , Aprendizagem por Associação/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/psicologia , Anfetamina/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Condicionamento Operante/efeitos dos fármacos , Interpretação Estatística de Dados , Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Feminino , Masculino , Microdiálise , Atividade Motora/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Comportamento Estereotipado/efeitos dos fármacos , Sacarose/farmacologia
20.
Toxicology ; 244(2-3): 249-56, 2008 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-18206288

RESUMO

The dopamine transporter has been shown to be the most relevant target site for the specificity of 1-methyl-4-phenylpyridinium ion (MPP+), a neurotoxin for dopaminergic neurons. In contrast, the mechanisms underlying the selective toxicity of manganese and rotenone, potentially toxic agents implicated in dopaminergic neuronal cell death, remain unknown. The aim of this study was to determine the cellular mechanisms of manganese or rotenone uptake in dopaminergic cells via the dopamine transporter. PC12 cells overexpressing the dopamine transporter, which were exposed to 10microM MPP+, showed extensive DNA fragmentation, a biochemical hallmark of apoptosis, whereas wild-type PC12 cells or vector-transfected PC12 cells, which were exposed to 5mM MPP+, did not show DNA fragmentation. In contrast, manganese and rotenone induced DNA fragmentation at slightly lower concentrations in PC12 cells overexpressing the dopamine transporter compared to control cells. Dopamine transporter inhibitors, such as mazindol, nomifensine, or GBR12909, inhibited MPP+-induced DNA fragmentation but did not affect manganese- and rotenone-induced DNA fragmentation in PC12 cells overexpressing the dopamine transporter. Finally, manganese accumulated to similar levels in PC12 cells overexpressing the dopamine transporter and control PC12 cells following incubation with manganese chloride. These results suggested that the dopamine transporter dose not confer cytotoxicity to manganese and rotenone.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/fisiologia , Intoxicação por Manganês/metabolismo , Manganês/toxicidade , Rotenona/toxicidade , Desacopladores/toxicidade , 1-Metil-4-fenilpiridínio/antagonistas & inibidores , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Western Blotting , Fragmentação do DNA/efeitos dos fármacos , DNA Complementar/biossíntese , DNA Complementar/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Humanos , Manganês/análise , Manganês/metabolismo , Células PC12 , Ratos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA