Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Arch Toxicol ; 97(7): 1887-1897, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37193757

RESUMO

We previously found that methylmercury induces expression of oncostatin M (OSM), which is released extracellularly and binds to tumor necrosis factor receptor 3 (TNFR3), possibly enhancing its own toxicity. However, the mechanism by which methylmercury causes OSM to bind to TNFR3 rather than to its known receptors, OSM receptor and LIFR, is unknown. In this study, we aimed to elucidate the effect of methylmercury modification of cysteine residues in OSM on binding to TNFR3. Immunostaining of TNFR3-V5-expressing cells suggested that methylmercury promoted binding of OSM to TNFR3 on the cell membrane. In an in vitro binding assay, OSM directly bound to the extracellular domain of TNFR3, and this binding was promoted by methylmercury. Additionally, the formation of a disulfide bond in the OSM molecule was essential for the binding of both proteins, and LC/MS analysis revealed that methylmercury directly modified the 105th cysteine residue (Cys105) in OSM. Next, mutant OSM, in which Cys105 was replaced by serine or methionine, increased the binding to TNFR3, and a similar effect was observed in immunoprecipitation using cultured cells. Furthermore, cell proliferation was inhibited by treatment with Cys105 mutant OSMs compared with wildtype OSM, and this effect was cancelled by TNFR3 knockdown. In conclusion, we revealed a novel mechanism of methylmercury toxicity, in which methylmercury directly modifies Cys105 in OSM, thereby inhibiting cell proliferation via promoting binding to TNFR3. This indicates a chemical disruption in the interaction between the ligand and the receptor is a part of methylmercury toxicity.


Assuntos
Cisteína , Compostos de Metilmercúrio , Oncostatina M/química , Oncostatina M/metabolismo , Compostos de Metilmercúrio/toxicidade , Receptores do Fator de Necrose Tumoral , Proliferação de Células
2.
Environ Health Prev Med ; 25(1): 30, 2020 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-32680455

RESUMO

Methylmercury is an environmental pollutant that causes neurotoxicity. Recent studies have reported that the ubiquitin-proteasome system is involved in defense against methylmercury toxicity through the degradation of proteins synthesizing the pyruvate. Mitochondrial accumulation of pyruvate can enhance methylmercury toxicity. In addition, methylmercury exposure induces several immune-related chemokines, specifically in the brain, and may cause neurotoxicity. This summary highlights several molecular mechanisms of methylmercury-induced neurotoxicity.


Assuntos
Quimiocinas/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Neurotoxinas/toxicidade , Proteólise/efeitos dos fármacos , Animais , Quimiocinas/metabolismo , Humanos , Camundongos , Ratos , Saccharomyces cerevisiae/efeitos dos fármacos
3.
Genes Cells ; 22(2): 160-173, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28097745

RESUMO

The structural protein Core of hepatitis C virus (HCV), a cytosolic protein, induces endoplasmic reticulum (ER) stress and unfolded protein response (UPR) in hepatocytes, and is responsible for the pathogenesis of persistent HCV infection. Using yeast as a model system, we evaluated mechanisms underlying Core-induced interference of ER homeostasis and UPR, and found that UPR is induced by the immature Core (aa 1-191, Core191) but not by the mature Core (aa 1-177, Core177). Interestingly, Core191 inhibits both ERAD-L, a degradation system responsible for misfolded/unfolded proteins in the ER lumen, and ERAD-M, a degradation system responsible for proteins carrying a misfolded/unfolded region in the ER membrane. In contrast, Core177 inhibits ERAD-M but not ERAD-L. In addition, requirement of an unfolded protein sensor in the ER lumen suggested that inhibition of ERAD-L is probably responsible for Core191-dependent UPR activation. These results implicate inadequate maturation of Core as a trigger for induction of ER stress and UPR.


Assuntos
Degradação Associada com o Retículo Endoplasmático/fisiologia , Hepacivirus/metabolismo , Saccharomyces cerevisiae/virologia , Resposta a Proteínas não Dobradas/fisiologia , Proteínas do Core Viral/metabolismo , Animais , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Humanos , Proteínas de Membrana/metabolismo , Dobramento de Proteína , Saccharomyces cerevisiae/metabolismo
4.
Toxicol Appl Pharmacol ; 356: 120-126, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30081057

RESUMO

We previously reported significantly increased level of putrescine, a polyamine, in the brains of mice administered methylmercury. Moreover, addition of putrescine to culture medium reduced methylmercury toxicity in C17.2 mouse neural stem cells. In this study, the role of ornithine decarboxylase (ODC), an enzyme involved in putrescine synthesis, in response to methylmercury toxicity was investigated. Methylmercury increased ODC activity in mouse cerebrum and cerebellum, but this increase was hardly observed in the kidney and liver, where methylmercury accumulated at a high concentration. In the cerebrum and cerebellum, increased putrescine was observed with methylmercury administration. Methylmercury increased ODC activity in C17.2 cells, but this was almost completely abolished in the presence of an ODC inhibitor. Methylmercury also increased the level of ODC protein in mouse brain and C17.2 cells. In addition, C17.2 cells pretreated with ODC inhibitor showed higher methylmercury sensitivity than control cells. These results suggest that the increased ODC activity by methylmercury is involved in the increase in putrescine level, and ODC plays an important role in the reduction of methylmercury toxicity. This is the first study to provide evidence that increased ODC activity may be a protective response against methylmercury-induced neurotoxicity.


Assuntos
Ativação Enzimática/efeitos dos fármacos , Intoxicação por Mercúrio/metabolismo , Intoxicação por Mercúrio/prevenção & controle , Compostos de Metilmercúrio/toxicidade , Ornitina Descarboxilase/efeitos dos fármacos , Putrescina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Linhagem Celular , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Mercúrio/farmacocinética , Camundongos , Células-Tronco Neurais , Inibidores da Ornitina Descarboxilase/farmacologia , Distribuição Tecidual
5.
Biochim Biophys Acta ; 1860(6): 1326-33, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27015763

RESUMO

BACKGROUND: We have previously reported that Whi2 enhances the toxicity of methylmercury in yeast. In the present study we examined the proteins known to interact with Whi2 to find those that influence the toxicity of methylmercury. METHODS: Gene disruption and site-directed mutagenesis were employed to examine the relationship of mercury toxicity and palmitoylation. Protein palmitoylation was examined using the acyl-biotinyl exchange method. Protein-protein interactions were detected by immunoprecipitation and immunoblotting. RESULTS: We found that deletion of Akr1, a palmitoyltransferase, rendered yeast cells highly sensitive to methylmercury, and Akr1 is necessary for the methylmercury resistance of Whi2-deleted yeast. Palmitoyltransferase activity of Akr1 has an important role in the alleviation of methylmercury toxicity. Whi2 deletion or methylmercury treatment enhanced the palmitoyltransferase activity of Akr1, and methylmercury treatment reduced the binding between Akr1 and Whi2. CONCLUSIONS: Whi2 bonds to Akr1 (a protein that is able to alleviate methylmercury toxicity) and thus inhibits Akr1's palmitoyltransferase activity, which leads to enhanced methylmercury toxicity. In contrast, methylmercury might break the bond between Whi2 and Akr1, which enhances the palmitoyltransferase activity of Akr1 to alleviate methylmercury toxicity. GENERAL SIGNIFICANCE: This study's findings propose that the Whi2/Akr1 system can be regarded as a defense mechanism that detects methylmercury incorporation of yeast cells and alleviates its toxicity.


Assuntos
Aciltransferases/antagonistas & inibidores , Compostos de Metilmercúrio/toxicidade , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Aciltransferases/fisiologia
6.
Biochim Biophys Acta Gen Subj ; 1861(7): 1729-1736, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28315413

RESUMO

BACKGROUND: We previously reported that palmitoyltransferase activity of Akr1 is required for alleviation of methylmercury toxicity in yeast. In this study, we identified a factor that alleviates methylmercury toxicity among the substrate proteins palmitoylated by Akr1, and investigated the role of this factor in methylmercury toxicity. METHODS: Gene disruption and site-directed mutagenesis were used to examine the relationship of methylmercury toxicity and vacuole function. Palmitoylation was investigated using the acyl-biotinyl exchange method. Vacuoles were stained with the fluorescent probe FM4-64. RESULTS: We found that Meh1 (alias Ego1), a substrate protein of Akr1, participates in the alleviation of methylmercury toxicity. Moreover, almost no palmitoylation of Meh1 when Akr1 was knocked out, and mutant Meh1, which is not palmitoylated, did not show alleviation of methylmercury toxicity. The palmitoylated Meh1 was involved in the alleviation of methylmercury toxicity as a constituent of EGO complex which suppresses autophagy. Methylmercury caused vacuole deformation, and this was greater in the yeasts knocking out the EGO complex subunits. 3-Methyladenine, an autophagy inhibitor, suppresses vacuole deformation and cytotoxicity caused by methylmercury. The elevated methylmercury sensitivity by Meh1 knockout almost completely disappeared in the presence of 3-methyladenine. CONCLUSIONS: Akr1 reduces methylmercury toxicity through palmitoylation of Meh1. Furthermore, the EGO complex including Meh1 reduces methylmercury toxicity by suppressing the induction of vacuole deformation caused by methylmercury. GENERAL SIGNIFICANCE: These findings propose that Meh1 palmitoylated by Akr1 may act as a constituent of the EGO complex when contributing to the decreased cytotoxicity by negatively controlling the induction of autophagy by methylmercury.


Assuntos
Aciltransferases/fisiologia , Proteínas de Membrana/fisiologia , Compostos de Metilmercúrio/toxicidade , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Lipoilação , Mutagênese Sítio-Dirigida , Ligação Proteica , Subunidades Proteicas , Fatores de Transcrição/fisiologia , Vacúolos/efeitos dos fármacos
7.
J Neural Transm (Vienna) ; 120(10): 1397-409, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23588349

RESUMO

cAMP/PKA/ERK/CREB signaling linked to CRE-mediated transcription is crucial for learning and memory. We originally found nobiletin as a natural compound that stimulates this intracellular signaling and exhibits anti-dementia action in animals. Citrus reticulata or C. unshiu peels are employed as "chinpi" and include a small amount of nobiletin. We here provide the first evidence for beneficial pharmacological actions on the cAMP/PKA/ERK/CREB cascade of extracts from nobiletin-rich C.reticulata peels designated as Nchinpi, the nobiletin content of which was 0.83 ± 0.13% of the dry weight or 16-fold higher than that of standard chinpi extracts. Nchinpi extracts potently facilitated CRE-mediated transcription in cultured hippocampal neurons, whereas the standard chinpi extracts showed no such activity. Also, the Nchinpi extract, but not the standard chinpi extract, stimulated PKA/ERK/CREB signaling. Interestingly, treatment with the Nchinpi extract at the concentration corresponding to approximately 5 µM nobiletin more potently facilitated CRE-mediated transcriptional activity than did 30 µM nobiletin alone. Consistently, sinensetin, tangeretin, 6-demethoxynobiletin, and 6-demethoxytangeretin were also identified as bioactive substances in Nchinpi that facilitated the CRE-mediated transcription. Purified sinensetin enhanced the transcription to a greater degree than nobiletin. Furthermore, samples reconstituted with the four purified compounds and nobiletin in the ratio of each constituent's content in the extract showed activity almost equal to that of the Nchinpi extract to stimulate CRE-mediated transcription. These findings suggest that above four compounds and nobiletin in the Nchinpi extract mainly cooperated to facilitate potently CRE-mediated transcription linked to the upstream cAMP/PKA/ERK/CREB pathway in hippocampal neurons.


Assuntos
Citrus/química , Medicamentos de Ervas Chinesas/farmacologia , Flavonas/farmacologia , Hipocampo/efeitos dos fármacos , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Células Cultivadas , Cromatografia Líquida de Alta Pressão , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Frutas/química , Hipocampo/fisiologia , Aprendizagem/fisiologia , Medicina Kampo , Memória/fisiologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Transfecção
8.
J Biol Chem ; 285(14): 10597-604, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20145245

RESUMO

Reactive oxygen species (ROS) generated during cellular metabolism are toxic to cells. As a result, cells must be able to identify ROS as a stress signal and induce stress response pathways that protect cells from ROS toxicity. Recently, peroxiredoxin (Prx)-induced relays of disulfide bond formation have been identified in budding yeast, namely the disulfide bond formation of Yap1, a crucial transcription factor for oxidative stress response, by a specific Prx Gpx3 and by a major Prx Tsa1. Here, we show that an atypical-type Prx Ahp1 can act as a receptor for alkylhydroperoxides, resulting in activation of the Cad1 transcription factor that is homologous to Yap1. We demonstrate that Ahp1 is required for the formation of intermolecular Cad1 disulfide bond(s) in both an in vitro redox system and in cells treated with alkylhydroperoxide. Furthermore, we found that Cad1-dependent transcriptional activation of the HSP82 gene is dependent on Ahp1. Our results suggest that, although the Gpx3-Yap1 pathway contributes more strongly to resistance than the Ahp1-Cad1 pathway, the Ahp1-induced activation of Cad1 can function as a defense system against stress induced by alkylhydroperoxides, possibly including lipid peroxides. Thus, the Prx family of proteins have an important role in determining peroxide response signals and in transmitting the signals to specific target proteins by inducing disulfide bond formation.


Assuntos
Dissulfetos/metabolismo , Regulação Fúngica da Expressão Gênica , Peroxirredoxinas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , terc-Butil Hidroperóxido/farmacologia , Imunoprecipitação da Cromatina , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/farmacologia , Immunoblotting , Imunoprecipitação , Oxidantes/farmacologia , Oxirredução , Estresse Oxidativo , Peroxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
9.
Anal Bioanal Chem ; 400(10): 3635-41, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21559759

RESUMO

We recently demonstrated that nobiletin, a citrus flavonoid, exhibits anti-dementia action in animals. However, no determination methods for the content of nobiletin with beneficial action in the brain of nobiletin-administered animals have been developed, nor has its pharmacokinetics been revealed completely. Here, we established the high-performance liquid chromatography/photodiode array detection method for nobiletin determination using Bond Elut C18 SPE cartridges for extraction, where the calibration curve was linear over 0.025-10 ng, with coefficient of variation of less than 6.76%. This method enabled us to determine pharmacokinetic parameters of nobiletin given intraperitoneally or per os in the brain of mice.


Assuntos
Encéfalo/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Flavonas/farmacocinética , Animais , Antioxidantes , Calibragem , Cromatografia Líquida de Alta Pressão/instrumentação , Cromatografia Líquida de Alta Pressão/normas , Flavonas/administração & dosagem , Flavonas/sangue , Flavonoides , Camundongos , Reprodutibilidade dos Testes
10.
Toxicol Res ; 37(4): 451-458, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34631502

RESUMO

Methylmercury is an environmental pollutant that induces potent neurotoxicity. We previously identified transcription factor 3 (TCF3) as a transcription factor that is activated in the brains of mice treated with methylmercury, and reported that methylmercury sensitivity was increased in cells in which TCF3 expression was suppressed. However, the mechanisms involved in the activation of TCF3 by methylmercury and in the reduction of methylmercury toxicity by TCF3 remained unclear. We found that treatment of mouse neuronal C17.2 cells with methylmercury increased TCF3 protein levels and promoted the binding of TCF3 to DNA consensus sequences. In cells treated with actinomycin D, a transcription inhibitor, an increase in TCF3 protein levels was also observed under methylmercury exposure. However, in the presence of cycloheximide, a translation inhibitor, methylmercury delayed the degradation of TCF3 protein. In addition, treatment with MG132, a proteasome inhibitor, increased TCF3 protein levels, and there was not significant increase in TCF3 protein levels by methylmercury under these conditions. These results suggest that methylmercury may activate TCF3 by increasing its levels through inhibition of TCF3 degradation by the proteasome. It has been previously reported that the induction of apoptosis in neurons is involved in methylmercury-induced neuronal damage in the brain. Although apoptosis was induced in C17.2 cells treated with methylmercury, this induction was largely suppressed by overexpression of TCF3. These results indicate that TCF3, which is increased in the brain upon exposure to methylmercury, may be a novel defense factor against methylmercury-induced neurotoxicity.

11.
Sci Rep ; 11(1): 9832, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33972601

RESUMO

We recently found that tumor necrosis factor-α (TNF-α) may be involved in neuronal cell death induced by methylmercury in the mouse brain. Here, we examined the cells involved in the induction of TNF-α expression by methylmercury in the mouse brain by in situ hybridization. TNF-α-expressing cells were found throughout the brain and were identified as microglia by immunostaining for ionized calcium binding adaptor molecule 1 (Iba1). Methylmercury induced TNF-α expression in mouse primary microglia and mouse microglial cell line BV2. Knockdown of apoptosis signal-regulating kinase 1 (ASK1), an inflammatory cytokine up-regulator that is responsible for reactive oxygen species (ROS), decreased methylmercury-induced TNF-α expression through decreased phosphorylation of p38 MAP kinase in BV2 cells. Suppression of methylmercury-induced reactive oxygen species (ROS) by antioxidant treatment largely abolished the induction of TNF-α expression and phosphorylation of p38 by methylmercury in BV2 cells. Finally, in mouse brain slices, the TNF-α antagonist (WP9QY) inhibited neuronal cell death induced by methylmercury, as did the p38 inhibitor SB203580 and liposomal clodronate (a microglia-depleting agent). These results indicate that methylmercury induces mitochondrial ROS that are involved in activation of the ASK1/p38 pathway in microglia and that this is associated with induction of TNF-α expression and neuronal cell death.


Assuntos
Encéfalo/patologia , Intoxicação do Sistema Nervoso por Mercúrio/patologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Linhagem Celular , Ácido Clodrônico/farmacologia , Modelos Animais de Doenças , Poluentes Ambientais/administração & dosagem , Poluentes Ambientais/toxicidade , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/farmacologia , MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Intoxicação do Sistema Nervoso por Mercúrio/etiologia , Compostos de Metilmercúrio/administração & dosagem , Compostos de Metilmercúrio/toxicidade , Camundongos , Microglia/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/patologia , Peptídeos Cíclicos/farmacologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Piridinas/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Toxics ; 8(4)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998228

RESUMO

Although homeobox protein B13 (HOXB13) is an oncogenic transcription factor, its role in stress response has rarely been examined. We previously reported that knockdown of HOXB13 reduces the cytotoxicity caused by various oxidative stress inducers. Here, we studied the role of HOXB13 in cytotoxicity caused by hydrogen peroxide in human lung epithelial A549 cells. The knockdown of HOXB13 reduced hydrogen peroxide-induced cytotoxicity; however, this phenomenon was largely absent in the presence of antioxidants (Trolox or N-acetyl cysteine (NAC)). This suggests that HOXB13 may be involved in the cytotoxicity caused by hydrogen peroxide via the production of reactive oxygen species (ROS). Hydrogen peroxide also increased both the mRNA and protein levels of HOXB13. However, these increases were rarely observed in the presence of a transcriptional inhibitor, which suggests that hydrogen peroxide increases protein levels via increased transcription of HOXB13. Furthermore, cell death occurred in A549 cells that highly expressed HOXB13. However, this cell death was mostly inhibited by treatment with antioxidants. Taken together, our findings indicate that HOXB13 may be a novel factor involved in the induction of oxidative stress, which causes cell death via intracellular ROS production.

13.
Life Sci ; 256: 118031, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32615186

RESUMO

AIMS: We had previously reported that addition of putrescine to the culture medium was reported to reduce methylmercury toxicity in C17.2 neural stem cells. Here, we have examined the inhibition of methylmercury-induced cytotoxicity by putrescine using ODC1-overexpressing C17.2 cells. MATERIALS AND METHODS: We established stable ODC1-overexpressing C17.2 cells and evaluated methylmercury-induced apoptosis by examining the TUNEL assay and cleaved caspase-3 levels. Mitochondria-mediated apoptosis was also evaluated by reduction of mitochondrial membrane potential and recruitment of Bax and Bak to the mitochondria. KEY FINDINGS: ODC is encoded by ODC1 gene, and putrescine levels in ODC1-overexpressing cells were significantly higher than in control cells. Overexpression of ODC1 and addition of putrescine to the culture medium suppressed DNA fragmentation and caspase-3 activation, which are observed when apoptosis is induced by methylmercury. Moreover, mitochondrial dysfunction and reactive oxygen species (ROS) generation, caused by methylmercury, were also inhibited by the overexpression of ODC1 and putrescine; pretreatment with ODC inhibitor, however, promoted both ROS generation and apoptosis by methylmercury. Finally, we found that Bax and Bak, the apoptosis-promoting factors, to be increased in mitochondria, following methylmercury treatment, and the same was inhibited by overexpression of ODC1. These results suggest that overexpression of ODC1 may prevent mitochondria-mediated apoptosis by methylmercury via increase of putrescine levels. SIGNIFICANCE: Our findings provide important clues to clarify mechanisms involved in the defense against methylmercury toxicity and suggest novel biological functions of putrescine.


Assuntos
Compostos de Metilmercúrio/toxicidade , Mitocôndrias/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Ornitina Descarboxilase/genética , Putrescina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Marcação In Situ das Extremidades Cortadas , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/patologia , Células-Tronco Neurais/patologia
14.
Cells ; 9(1)2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31878059

RESUMO

Homeobox protein B13 (HOXB13), a transcription factor, is related to methylmercury toxicity; however, the downstream factors involved in enhancing methylmercury toxicity remain unknown. We performed microarray analysis to search for downstream factors whose expression is induced by methylmercury via HOXB13 in human embryonic kidney cells (HEK293), which are useful model cells for analyzing molecular mechanisms. Methylmercury induced the expression of oncostatin M (OSM), a cytokine of the interleukin-6 family, and this was markedly suppressed by HOXB13 knockdown. OSM knockdown also conferred resistance to methylmercury in HEK293 cells, and no added methylmercury resistance was observed when both HOXB13 and OSM were knocked down. Binding of HOXB13 to the OSM gene promoter was increased by methylmercury, indicating the involvement of HOXB13 in the enhancement of its toxicity. Because addition of recombinant OSM to the medium enhanced methylmercury toxicity in OSM-knockdown cells, extracellularly released OSM was believed to enhance methylmercury toxicity via membrane receptors. We discovered tumor necrosis factor receptor (TNF) receptor 3 (TNFR3) to be a potential candidate involved in the enhancement of methylmercury toxicity by OSM. This toxicity mechanism was also confirmed in mouse neuronal stem cells. We report, for the first time, that HOXB13 is involved in enhancement of methylmercury toxicity via OSM-expression induction and that the synthesized OSM causes cell death by binding to TNFR3 extracellularly.


Assuntos
Proteínas de Homeodomínio/metabolismo , Compostos de Metilmercúrio/toxicidade , Oncostatina M/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Genes Homeobox , Células HEK293 , Humanos , Intoxicação por Mercúrio/metabolismo , Proteínas Nucleares/metabolismo , Oncostatina M/biossíntese , Transdução de Sinais/efeitos dos fármacos
15.
J Toxicol Sci ; 44(7): 471-479, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31270303

RESUMO

M1-microglia (neurotoxic microglia) regulate neuronal development and cell death and are involved in many pathologies in the brain. Although organotypic brain slice cultures are widely used to study the crosstalk between neurons and microglia, little is known about the properties of microglia in the mouse cerebral cortex slices. Here, we aimed to optimize the mouse cerebral slice cultures that reflect microglial functions and evaluate the effects of neurotoxic metals on M1-microglial activation. Most microglia in the cerebral slices prepared from postnatal day (P) 7 mice were similar to mature microglia in adult mice brains, but those in the slices prepared from P2 mice were immature, which is a conventional preparation condition. The degree of expression of M1-microglial markers (CD16 and CD32) and inflammatory cytokines (tumor necrosis factor-α and interleukin-1ß) by lipopolysaccharide, a representative microglia activator, in the cerebral slices of P7 mice were higher than that in the slices of P2 mice. These results indicate that M1-microglial activation can be evaluated more accurately in the cerebral slices of P7 mice than in those of P2 mice. Therefore, we next examined the effects of various neurotoxic metals on M1-microglial activation using the cerebral slices of P7 mice and found that methylmercury stimulated the activation to M1-microglia, but arsenite, lead, and tributyltin did not induce such activation. Altogether, the optimized mouse cerebral slice cultures used in this study can be a helpful tool to study the influence of various chemicals on the central nervous system in the presence of functionally mature microglia.


Assuntos
Córtex Cerebral/citologia , Metais/toxicidade , Microglia/efeitos dos fármacos , Microglia/fisiologia , Animais , Animais Recém-Nascidos , Arsenitos/toxicidade , Células Cultivadas , Córtex Cerebral/metabolismo , Citocinas/metabolismo , Expressão Gênica , Mediadores da Inflamação/metabolismo , Chumbo/toxicidade , Compostos de Metilmercúrio/toxicidade , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/fisiologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Compostos de Trialquitina/toxicidade
16.
Sci Rep ; 9(1): 4631, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874621

RESUMO

Methylmercury is an environmental pollutant that causes specific and serious damage to the central nervous system. We have previously shown that C-C motif chemokine ligand 4 (CCL4) protects cultured neural cells from methylmercury toxicity and expression of CCL4 is specifically induced in mouse brain by methylmercury. In this study, we examined the transcriptional regulatory mechanism that induces CCL4 expression by methylmercury using C17.2 mouse neural stem cells. The promoter region of the CCL4 gene was analyzed by a reporter assay, revealing that the region up to 50 bp upstream from the transcription start site was necessary for inducing expression of CCL4 by methylmercury. Nine transcription factors that might bind to this upstream region and be involved in the induction of CCL4 expression by methylmercury were selected, and the induction of CCL4 expression by methylmercury was suppressed by the knockdown of serum response factor (SRF). In addition, the nuclear level of SRF was elevated by methylmercury, and an increase in the amount bound to the CCL4 gene promoter was also observed. Furthermore, we examined the upstream signaling pathway involved in the induction of CCL4 expression by SRF, and confirmed that activation of p38 and ERK, which are part of the MAPK pathway, are involved. These results suggest that methylmercury induces the expression of CCL4 by activating SRF via the p38 and ERK signaling pathway. Our findings are important for elucidating the mechanism involved in the brain-specific induction of CCL4 expression by methylmercury.


Assuntos
Quimiocina CCL4/metabolismo , Compostos de Metilmercúrio/efeitos adversos , Fator de Resposta Sérica/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Quimiocina CCL4/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Compostos de Metilmercúrio/metabolismo , Compostos de Metilmercúrio/toxicidade , Camundongos , NF-kappa B/metabolismo , Células-Tronco Neurais/metabolismo , Regiões Promotoras Genéticas/genética , Fator de Resposta Sérica/fisiologia , Transdução de Sinais , Fatores de Transcrição/metabolismo
17.
Environ Toxicol Pharmacol ; 71: 103216, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31260942

RESUMO

Methylmercury is an environmental pollutant that shows selective toxicity to the central nervous system. We previously reported that brain-specific expression of chemokine CCL3 increases in mice administered methylmercury. However, the relationship between CCL3 and methylmercury toxicity has not been elucidated. Here, we confirmed that induction of CCL3 expression occurs before pathological change by methylmercury treatment was observed in the mouse brain. This induction was also observed in C17.2 mouse neural stem cells before methylmercury-induced cytotoxicity. In addition, cells in which CCL3 was knocked-down showed higher methylmercury sensitivity than did control cells. Moreover, activation of transcription factor NF-κB was observed following methylmercury treatment, and methylmercury-mediated induction of CCL3 expression was partially suppressed by knockdown of p65, an NF-κB subunit. Our results suggest that NF-κB plays a role in the induction of methylmercury-mediated CCL3 expression and that this action may be a cellular response to methylmercury toxicity.


Assuntos
Quimiocina CCL3/biossíntese , Poluentes Ambientais/toxicidade , Compostos de Metilmercúrio/toxicidade , NF-kappa B/biossíntese , Células-Tronco Neurais/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Cérebro/efeitos dos fármacos , Cérebro/metabolismo , Cérebro/patologia , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia
18.
Neurosci Lett ; 438(1): 54-8, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18479819

RESUMO

Metallothionein-3 (MT-3), also known as growth inhibitory factor (GIF), was originally identified in the brain. An essential step in elucidating the potential roles of MT-3 is to evaluate its expression levels in organs other than the brain. In this present study, we carried out RT-PCR, Western blot and immunohistochemical analyses to quantify MT-3 mRNA and its protein in the cerebrum, eye, heart, kidney, liver, prostate, testis, tongue, and muscle in male Wistar rats. MT-3 mRNA was detected in the cerebrum, the dorsolateral lobe of the prostate, testis, and tongue. Using a monoclonal anti-MT-3 antibody, we detected MT-3 in the cerebrum, the dorsolateral lobe of the prostate, testis, and tongue as a single band on an immunoblot. Immunohistochemical staining showed MT-3 in some astrocytes in the deep cortex, ependymal cells, and choroidal cells in the cerebrum. MT-3 was also detected in some cells of the glomerulus and the collective tubules in the kidney, some cells in the glandular epithelium of the dorsolateral lobe of the prostate, some Sertoli cells and Lydig cells in the testis, and taste bud cells in the tongue. Although MT-3 immunopositivity was obviously demonstrated in the kidney by the immnunohistochemical method, the expression of MT-3 was not fully detectable by RT-PCR and Western blot analyses. Interestingly, only a subset of cells showed positivity for MT-3, not all cells in all tissues. The localization of MT-3 in peripheral organs outside the brain suggests that MT-3 has roles in these tissues besides its role in growth inhibition of neurites.


Assuntos
Encéfalo/enzimologia , Células Epiteliais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Vísceras/enzimologia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Western Blotting , Encéfalo/anatomia & histologia , Plexo Corióideo/citologia , Plexo Corióideo/metabolismo , Epêndima/citologia , Epêndima/metabolismo , Células Epiteliais/citologia , Inibidores do Crescimento/metabolismo , Imuno-Histoquímica , Rim/citologia , Rim/metabolismo , Masculino , Metalotioneína 3 , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Próstata/citologia , Próstata/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Papilas Gustativas/citologia , Papilas Gustativas/metabolismo , Testículo/citologia , Testículo/metabolismo , Língua/citologia , Língua/metabolismo , Vísceras/citologia
19.
Life Sci ; 82(23-24): 1182-5, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18466927

RESUMO

The expression of the genes encoding the ubiquitin-conjugating enzymes, Ubc4, Ubc5, and Ubc7, has been reported to be induced by cadmium in budding yeast. In contrast, we have reported that the overexpression of Cdc34, another ubiquitin-conjugating enzyme, confers resistance to cadmium. In the present study, we examined the effects of overexpression of Ubc4, Ubc5, or Ubc7 on the sensitivity of budding yeast to cadmium. We found that yeast cells that overexpressed Ubc4, but not Ubc5 or Ubc7, showed similar cadmium resistance as yeast cells that overexpressed Cdc34. The ubiquitination levels of cellular proteins were significantly increased by overexpression of Ubc4 as well as by Cdc34. As previously reported, yeast cells overexpressing Cdc34 were resistant to cadmium even in the presence of the proteasome inhibitor MG132. However, the acquired resistance to cadmium by overexpression of Ubc4 was not observed in the presence of MG132. Cdc34 overexpression has been shown to inactivate the transcriptional activity of Met4 by accelerating its ubiquitination and to reduce expression of the MET25 gene, a target gene of Met4. Unlike Cdc34, overexpression of Ubc4 did not affect the expression of the MET25 gene. These findings suggest that the mechanism of acquired resistance to cadmium by overexpression of Ubc4 is different from that of Cdc34 and that Ubc4 confers resistance to cadmium by ubiquitination of proteins other than Met4 and accelerates the degradation of these proteins in the proteasomes.


Assuntos
Cloreto de Cádmio/toxicidade , Farmacorresistência Fúngica , Poluentes Ambientais/toxicidade , Proteínas de Saccharomyces cerevisiae/biossíntese , Saccharomyces cerevisiae/efeitos dos fármacos , Enzimas de Conjugação de Ubiquitina/biossíntese , Complexos Ubiquitina-Proteína Ligase/biossíntese , Ciclossomo-Complexo Promotor de Anáfase , Clonagem Molecular , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Farmacorresistência Fúngica/genética , Leupeptinas/farmacologia , Plasmídeos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , beta-Galactosidase/metabolismo
20.
Cancer Res ; 66(24): 11932-7, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17178891

RESUMO

To elucidate the mechanism of acquired resistance to Adriamycin, we searched for genes that, when overexpressed, render Saccharomyces cerevisiae resistant to Adriamycin. We identified AKL1, a gene of which the function is unknown but is considered, nonetheless, to be a member of the Ark/Prk kinase family, which is involved in the regulation of endocytosis, on the basis of its deduced amino acid sequence. Among tested members of the Ark/Prk kinase family (Ark1, Prk1, and Akl1), overexpressed Prk1 also conferred Adriamycin resistance on yeast cells. Prk1 is known to dissociate the Sla1/Pan1/End3 complex, which is involved in endocytosis, by phosphorylating Sla1 and Pan1 in the complex. We showed that Akl1 promotes phosphorylation of Pan1 in this complex and reduces the endocytic ability of the cell, as does Prk1. Sla1- and End3-defective yeast cells were also resistant to Adriamycin and overexpression of Akl1 in these defective cells did not increase the degree of Adriamycin resistance, suggesting that Akl1 might reduce Adriamycin toxicity by reducing the endocytic ability of cells via a mechanism that involves the Sla1/Pan1/End3 complex and the phosphorylation of Pan1. We also found that HEK293 cells that overexpressed AAK1, a member of the human Ark/Prk family, were Adriamycin resistant. Our findings suggest that endocytosis might be involved in the mechanism of Adriamycin toxicity in yeast and human cells.


Assuntos
Doxorrubicina/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Aurora Quinase A , Aurora Quinases , Linhagem Celular , Primers do DNA , Resistência a Medicamentos , Endocitose , Humanos , Isoquinolinas/farmacocinética , Rim/efeitos dos fármacos , Rim/embriologia , Mutagênese Sítio-Dirigida , Fosforilação , Mutação Puntual , Reação em Cadeia da Polimerase , Proteína Quinase C , Saccharomyces cerevisiae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA