Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L900-L911, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34585979

RESUMEN

Carbon black nanoparticles (CBNPs) are one of the most frequently used nanoparticles. Exposure to CBNPs during pregnancy (PrE to CBNPs) can directly induce inflammation, lung injury, and genotoxicity in dams and results in abnormalities in offspring. However, whether exposure to CBNPs during pregnancy enhances the susceptibility of offspring to environmental stimuli remains unknown. To address this issue, in this study, we intranasally treated pregnant mice with mock or CBNPs from gestational day (GD) 9 to GD18, and F1 and F2 offspring were normally obtained. By intratracheal instillation of mice with lipopolysaccharide (LPS) to trigger a classic animal model for acute lung injury, we intriguingly found that after LPS treatment, F1 and F2 offspring after exposure during pregnancy to CBNPs both exhibited more pronounced lung injury symptoms, including more degenerative histopathological changes, vascular leakage, elevated MPO activity, and activation of inflammation-related signaling transduction, compared with F1 and F2 offspring in the mock group, suggesting PrE to CBNPs would aggravate LPS-induced lung injury in offspring, and this effect was intergenerational. We also observed that PrE to CBNPs upregulated the mRNA expression of DNA methyltransferases (Dnmt) 1/3a/3b and DNA hypermethylation in both F1 and F2 offspring, which might partially account for the intergenerational effect. Together, our study demonstrates for the first time that PrE to CBNPs can enhance sensitivity to LPS in both F1 and F2 offspring, and this intergenerational effect may be related to DNA hypermethylation caused by CBNPs.


Asunto(s)
Daño del ADN/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Nanopartículas/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Femenino , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología , Masculino , Ratones , Embarazo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
2.
Food Chem Toxicol ; 151: 112114, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33722599

RESUMEN

Ferroptosis is a novel form of cell death that involves in the pathophysiological process of diverse brain diseases. However, how arsenite induces ferroptosis in the neuronal cells remains unsolved. In this study, by using in vitro and in vivo models, we demonstrated that arsenite was able to trigger ferroptosis in the neuronal cells. Exposure of arsenite for 6 months at 0.5, 5 and 50 mg/L arsenite via drinking water significantly reduced the number of neurons and caused the pathological changes in the mitochondria of hippocampus. Treatment of arsenite elevated the contents of lipid peroxidation products, disrupted the iron homeostasis, altered the expressions of ferroptosis-related proteins in the hippocampus and PC-12 cells. The results also showed that arsenite significantly decreased the expressions of ferritin and NCOA4, but sharply enhanced the level of autophagy marker LC3B, suggesting the activation of ferritinophagy by arsenite. Co-treatment of arsenite with ferroptosis inhibitor ferrostatin-1, or autophagy inhibitors 3-MA and BafA1, all remarkably attenuated the cytotoxic effects of arsenite. These findings not only present a novel mechanism that arsenite triggers ferroptosis in the neuronal cells via activation of ferritinophagy, but also indicate that regulating ferritinophagy to control iron level may provide a clue for prevention against arsenite neurotoxicity.


Asunto(s)
Arsenitos/farmacología , Ferritinas/metabolismo , Ferroptosis/efectos de los fármacos , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Arsenitos/toxicidad , Muerte Celular/efectos de los fármacos , Ciclohexilaminas/farmacología , Hipocampo/citología , Hipocampo/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Células PC12 , Fenilendiaminas/farmacología , Ratas , Sinapsis/efectos de los fármacos
3.
Int J Nanomedicine ; 15: 5299-5315, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32884256

RESUMEN

PURPOSE: Zinc oxide nanoparticles (ZnONPs) are one of the most important nanomaterials that are widely used in the food, cosmetic and medical industries. Humans are often exposed to ZnONPs via inhalation, and they may reach the brain where neurotoxic effects could occur via systemic distribution. However, the mechanisms underlying how ZnONPs produce neurotoxic effects in the brain remain unclear. In this study, we aimed to investigate the novel mechanism involved in ZnONPs-induced neurotoxicity. METHODS AND RESULTS: We demonstrated for the first time that pulmonary exposure to ZnONPs by intratracheal instillation could trigger ferroptosis, a new form of cell death, in the neuronal cells of mouse cerebral cortex. A similar phenomenon was also observed in cultured neuron-like PC-12 cell line. By using a specific inhibitor of ferroptosis ferrostatin-1 (Fer-1), our results showed that inhibition of ferroptosis by Fer-1 could significantly alleviate the ZnONPs-induced neuronal cell death both in vivo and in vitro. Mechanistic investigation revealed that ZnONPs selectively activated the JNK pathway and thus resulted in the ferroptotic phenotypes, JNK inhibitor SP600125 could reverse lipid peroxidation upregulation and ferroptotic cell death induced by ZnONPs in PC-12 cells. CONCLUSION: Taken together, this study not only demonstrates that pulmonary exposure of ZnONPs can induce JNK-involved ferroptotic cell death in mouse cortex and PC-12 cells, but also provides a clue that inhibition of ferroptosis by specific agents or drugs may serve as a feasible approach for reducing the untreatable neurotoxicity induced by ZnONPs.


Asunto(s)
Ferroptosis/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Óxido de Zinc/toxicidad , Administración por Inhalación , Animales , Antracenos/farmacología , Muerte Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Ciclohexilaminas/farmacología , Ferroptosis/fisiología , Peroxidación de Lípido/efectos de los fármacos , Pulmón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Nanopartículas del Metal/administración & dosificación , Ratones Endogámicos C57BL , Neuronas/patología , Síndromes de Neurotoxicidad/etiología , Células PC12 , Fenilendiaminas/farmacología , Ratas , Óxido de Zinc/administración & dosificación
4.
Neurotoxicology ; 81: 40-50, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32783905

RESUMEN

Increasing occupational and accidental exposure to carbon black nanoparticles (CBNPs) raise concerns over their possible effects on the nervous system. However, the influences of CBNPs on the neurodevelopment remain unclear. Thus, in this study, pregnant mice were exposed to different doses of CBNPs by intranasal instillation on gestation days 9-18. Our results demonstrated that maternal exposure to CBNPs caused significant changes on maternal behaviors. Pregnancy exposure to CBNPs also delayed the onset of incisor eruption, testes descent and vaginal opening in offspring, and caused the reduced body weight until adulthood. In the neurobehavioral tests, CBNPs-exposed offspring exhibited the elevated latency of negative geotaxis and surface right reflex, reduced grasping time and increased cliff avoidance. Histopathological changes were present in F1 generation but not in F2 generation. Intriguingly, our data revealed that the levels of total m6A modification were significantly decreased by CBNPs. Similar trends were observed on the mRNA expressions of m6A methyltransferases and demethylases. In summary, these findings provide the novel evidence that pregnancy exposure to CBNPs affects the maternal behaviors and partially induces the neurobehavioral, muscular and histopathological changes in offspring. Of note, these adverse effects may be associated with reduced levels of total m6A modification in brain.


Asunto(s)
Adenosina/análogos & derivados , Conducta Animal/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Nanopartículas/toxicidad , Síndromes de Neurotoxicidad/etiología , Efectos Tardíos de la Exposición Prenatal , Hollín/toxicidad , Adenosina/metabolismo , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Femenino , Edad Gestacional , Exposición Materna , Metiltransferasas/metabolismo , Ratones Endogámicos C57BL , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Embarazo
5.
Int J Nanomedicine ; 14: 9865-9875, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31849474

RESUMEN

PURPOSE: Liver is regarded as one of the primary target organs for zinc oxide nanoparticles (ZnONPs) toxicity. Since liver represents the leading site for de novo cholesterol biosynthesis in mammals, the injuries of liver could result in the disruption of cholesterol biosynthesis. In this study, we aimed to investigate whether pulmonary ZnONPs exposure induces disturbance of cholesterol biosynthesis in mouse liver. METHODS AND RESULTS: Our data demonstrated intratracheally instilled with a single dose of 3, 6, and 12 µg/animal ZnONPs could induce histopathological deterioration in mouse liver in a dose-related manner at 3 days, but remission was observed at 7 days after treatment. Moreover, ZnONPs caused the disturbance of cholesterol biosynthesis by increasing both 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase and sterol regulatory element-binding protein 2 (SREBP2) protein expressions. To further reveal the underlying toxic mechanisms, we detected the biomarkers of autophagy and found that pulmonary ZnONPs exposure led to the elevation of LC3B-II and Beclin 1, suggesting ZnONPs might trigger autophagy in liver tissues. By using both beclin 1 +/+ and beclin 1 +/- mice, we demonstrated that inhibition of autophagy by heterozygous disruption of beclin 1 attenuated the disturbance of cholesterol biosynthesis induced by ZnONPs in liver. CONCLUSION: Pulmonary exposure of ZnONPs would induce the cholesterol biosynthesis disturbance in mouse liver through Beclin-1-dependent autophagy activation, suggesting that inhibition of autophagy may contribute to preventing the cholesterol biosynthesis disturbance and its associated pathologies induced by ZnONPs in liver.


Asunto(s)
Beclina-1/genética , Colesterol/biosíntesis , Hígado/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Óxido de Zinc/toxicidad , Animales , Autofagia/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Heterocigoto , Hígado/metabolismo , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Nanopartículas del Metal/administración & dosificación , Ratones Endogámicos C57BL , Ratones Mutantes , Óxido de Zinc/administración & dosificación
6.
Chemosphere ; 237: 124378, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31376700

RESUMEN

Benzo[a]pyrene (B[a]P) is a ubiquitous neurotoxic pollutant that widely distributes in the natural environment. However, the exact mechanism of B[a]P-induced neurotoxicity has not been well established. As one key synaptic protein, SNAP-25 plays an important role in the regulation of neurotransmitter release, including synaptic dopamine release. In this study, we demonstrated that, after intragastric administration of B[a]P in rats aged postnatal day 5 for 7 weeks, B[a]P significantly increased the level of dopamine and the expression of SNAP-25, dopamine receptor 1 (DRD1) and DRD 3. Moreover, treatment of B[a]P also caused the ultra-structural pathological changes in the cerebral cortex of rats. To further reveal the potential role of SNAP-25 in the regulation of DRDs, we treated the dopaminergic PC-12 cells with 20 µM B[a]P for 24 h. A significant cytotoxicity and apoptosis were observed, and more importantly, we found that SNAP-25, DRD 1 and DRD 3 co-localized in the cells, and down-regulation of SNAP-25 by CRISPR-Cas9 plasmid remarkably reduced the expression of DRD1 and DRD3. Together, our findings suggest that, synaptic dopamine release may be positively regulated by SNAP-25 via its receptors, and thus affecting the neurotoxicity induced by B[a]P.


Asunto(s)
Benzo(a)pireno/toxicidad , Dopamina/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Síndromes de Neurotoxicidad/etiología , Células PC12 , Ratas , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D3/metabolismo , Transmisión Sináptica
7.
Toxicology ; 425: 152244, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31302203

RESUMEN

Accumulating evidence shows that carbon black nanoparticles (CBNPs) (one of the most used nanoparticles) can induce toxicity via induction of inflammation, oxidative stress and genotoxicity in vitro and in vivo, and epidemiological studies have indicated that the possible correlation between maternal immune activation and risk of developing neuropsychiatric disorder in the offspring. However, whether pregnancy exposure of CBNPs (Pr-CBNPs) enhances the susceptibility to bleomycin (BLM)-induced lung fibrosis in offspring is unknown. Herein, we demonstrated that Pr-CBNPs during gestational day 9-18 via intranasal administration could confer enhanced susceptibility to BLM-induced fibrotic response in offspring, including deteriorative lung pathologic changes and more collagen deposition. Intriguingly, we found that Pr-CBNPs repressed the activation of autophagy (an anti-fibrotic mechanism), which was moderately activated in offspring from mock group. Moreover, Pr-CBNPs was likely to disrupt the LKB1-AMPK-ULK1 axis (a key regulatory pathway for autophagy induction). In summary, this study provides the first evidence that pregnancy exposure to CBNPs can exacerbate BLM-induced lung fibrotic response in offspring probably through disruption of LKB1-AMPK-ULK1 axis-mediated autophagy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia/efectos de los fármacos , Bleomicina/toxicidad , Nanopartículas/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Proteínas Serina-Treonina Quinasas/metabolismo , Fibrosis Pulmonar/inducido químicamente , Transducción de Señal/efectos de los fármacos , Hollín/toxicidad , Animales , Sinergismo Farmacológico , Femenino , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo
8.
Toxicology ; 422: 44-52, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31022427

RESUMEN

Maternal exposure to carbon black nanoparticles (CBNPs) during pregnancy have been well documented to induce harmful outcomes of offspring on brain function. However, it remains largely unknown whether females exposed to CBNPs during sensitive period of pregnancy can cause the neurotoxic effects on their own body after parturition. In this study, our results showed that pregnancy CBNPs exposure induced the persistent pathological changes in the cerebral cortex tissues and impaired cerebrovascular function of mice manifested by significant alterations of endothelin-1, endothelial nitric oxide synthase, vascular endothelial growth factor-A and ATP-binding cassette transporter G1. Intriguingly, we observed that these deleterious effects on brain and cerebrovascular functions in mice could persist for 49 days after delivery of pups. By using in vitro human umbilical vein endothelial cells, we further verified the potential vascular dysfunction after CBNPs exposure. In summary, our results provide the first evidence that pregnancy CBNPs exposure-induced brain pathological changes and cerebrovascular dysfunction can persist for a relative long time. These finding suggest exposure to CBNPs during sensitive stages of pregnancy may not only show the harmful effects on offspring neurodevelopment, but also result in the irreversible brain damage on mother body.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Intercambio Materno-Fetal , Nanopartículas/toxicidad , Hollín/toxicidad , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Endotelina-1/genética , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/genética , Embarazo , Factor A de Crecimiento Endotelial Vascular/genética
9.
Nanotoxicology ; 12(9): 1068-1091, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30317896

RESUMEN

Pulmonary exposure to zinc oxide nanoparticles (ZnONPs) could cause acute lung injury (ALI), but the underlying molecular mechanism remains unclear. Herein, we established a ZnONPs-induced ALI mouse model, characterized by the histopathological changes (edema and infiltration of inflammatory cells in lung tissues), and the elevation of total protein and cytokine interleukin-6 in bronchoalveolar lavage fluid in time- and dose-dependent manners. This model also exhibited features like the disturbance of redox-state (reduced of glutathione to glutathione disulfide ratio, elevation of heme oxygenase-1 and superoxide dismutase 2), the decrease of adenosine triphosphate synthesis and the release of zinc ions in the lung tissues. Interestingly, we found that ZnONPs exposure caused the accumulation of autophagic vacuoles and the elevation of microtubule-associated proteins 1A/1B light chain (LC)3B-II and p62, indicating the impairment of autophagic flux. Our data indicated that the above process might be regulated by the activation of AMP-activated protein kinase but not the mammalian target of rapamycin pathway. The association between ZnONPs-induced ALI and autophagy was further verified by a classical autophagy inhibitor, 3-methyladenine (3-MA). 3-MA administration reduced the accumulation of autophagic vacuoles, the expression of LC3B-II and p62, followed by a significant attenuation of histopathological changes, inflammation, and oxidative stress. More importantly, 3-MA could directly decrease the release of zinc ions in lung tissues. Taken together, our study provides the evidence that ZnONPs-induced pulmonary toxicity is autophagy-dependent, suggests that limiting the release of zinc ions by inhibiting autophagy could be a feasible strategy for the prevention of ZnONPs-associated pulmonary toxicity.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Autofagia/efectos de los fármacos , Nanopartículas/toxicidad , Óxido de Zinc/toxicidad , Zinc/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Autofagosomas/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/análisis , Exposición por Inhalación , Iones , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Propiedades de Superficie , Óxido de Zinc/metabolismo
10.
Environ Toxicol Pharmacol ; 64: 1-10, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30265862

RESUMEN

Male spermatogenesis dysfunctions are associated with environmental pollutants, but the detailed mechanisms remain poorly understood. In this study, healthy C57BL/6 J mice were used to establish an animal model of maternal exposure to traffic pollutant during pregnancy, and the toxic effects on the reproductive system of F2 male mice were analysed using mRNA and miRNA microarray. Our results showed that 54 miRNAs and 1927 mRNAs were significantly altered in the exposed group. Gene Ontology (GO) analysis revealed that the most significant GO terms for biological process, molecular function and cellular component were myeloid cell differentiation, growth factor binding and main axon. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis demonstrated that the biosynthesis of amino acids was the most significant pathway and that the cytokine-cytokine receptor interaction was the most abundant pathway (37 genes). Protein-protein interaction (PPI) and the miRNA-mRNA network were constructed with Cytoscape. The hub genes, Tnf, Il10 and Gapdh, were closely related to immuno-regulation and their miRNA regulators were reversely changed. Together, our results indicate that maternal exposure to traffic pollutant can cause spermatogenesis damage in F2 male mice possibly through the destroyed immunoprivileged environment in testis mediated by the aberrant expression of miRNA and mRNA.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Espermatogénesis/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Animales , Femenino , Masculino , Intercambio Materno-Fetal , Ratones Endogámicos C57BL , Embarazo , Testículo/efectos de los fármacos , Testículo/metabolismo
11.
Toxicol Sci ; 165(2): 431-446, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982692

RESUMEN

Arsenite exposure is known to increase the risk of neurological disorders via alteration of dopamine content, but the detailed molecular mechanisms remain largely unknown. In this study, using both dopaminergic neurons of the PC-12 cell line and C57BL/6J mice as in vitro and in vivo models, our results demonstrated that 6 months of arsenite exposure via drinking water caused significant learning and memory impairment, anxiety-like behavior and alterations in conditioned avoidance and escape responses in male adult mice. We also were the first to reveal that the reduction in dopamine content induced by arsenite mainly resulted from deficits in dopaminergic neurotransmission in the synaptic cleft. The reversible N6- methyladenosine (m6A) modification is a novel epigenetic marker with broad roles in fundamental biological processes. We further evaluated the effect of arsenite on the m6A modification and tested if regulation of the m6A modification by demethylase fat mass and obesity-associated (FTO) could affect dopaminergic neurotransmission. Our data demonstrated for the first time that arsenite remarkably increased m6A modification, and FTO possessed the ability to alleviate the deficits in dopaminergic neurotransmission in response to arsenite exposure. Our findings not only provide valuable insight into the molecular neurotoxic pathogenesis of arsenite exposure, but are also the first evidence that regulation of FTO may be considered as a novel strategy for the prevention of arsenite-associated neurological disorders.


Asunto(s)
Adenosina/análogos & derivados , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Arsenitos/toxicidad , Conducta Animal/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Adenosina/genética , Adenosina/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Animales , Arsenitos/farmacocinética , Reacción de Prevención/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Relación Dosis-Respuesta a Droga , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Células PC12 , Modificación Traduccional de las Proteínas , Ratas
12.
Neurotoxicology ; 67: 27-36, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29678591

RESUMEN

Ferroptosis is a novel iron-dependent form of cell death implicated in brain pathology. However, whether arsenite is an inducer of ferroptosis in the neuron remains completely unknown. In this study, the seven-week-old healthy C57BL/6 J male mice were treated with environmental related doses (0.5, 5 and 50 mg/L) of arsenite for 6 months via drinking water, and the ferroptosis-related indicators were further determined. Our results demonstrated for the first time that, arsenite exposure significantly reduced the number of neuron and caused the pathological changes of mitochondria in the cerebral cortex of mice. We further revealed that arsenite induced ferroptotic cell death in neuron by accumulation of reactive oxygen species and lipid peroxidation products, disruption of Fe2+ homeostasis, depletion of glutathione and adenosine triphosphate, inhibition of cysteine/glutamate antiporter, activation of mitogen-activated protein kinases and mitochondrial voltage-dependent anion channels pathways, up-regulation of endoplasmic reticulum stress, all of which were involved in the process of ferroptosis. These findings were also verified in the cultured PC-12 cells by using ferropotosis inhibitor, desferoxamine. Taken together, our results not only reveal a novel mechanism that chronic arsenite exposure may trigger the new form of cell death, ferroptosis, but also shed a new light on a potential clue for the intervention and prevention against arsenite-related neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenitos/toxicidad , Hierro/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Animales , Apoptosis/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Células PC12 , Distribución Aleatoria , Ratas , Especies Reactivas de Oxígeno/metabolismo , Teratógenos/toxicidad
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(2): 162-167, 2018 Feb 20.
Artículo en Chino | MEDLINE | ID: mdl-29502054

RESUMEN

OBJECTIVE: To analyze the changes in endogenous small molecule metabolites after benzo[a]pyrene (B[a]P) exposure in rat cerebral cortex and explore the mechanism of B[a]P neurotoxicity. METHODS: Five-day-old SD rats were subjected to gavage administration of 2 mg/kg B[a]P for 7 consecutive weeks. After the exposure, the rats were assessed for spatial learning ability using Morris water maze test, ultrastructural changes of the cortical neurons under electron microscope, and metabolite profiles of the cortex using GC/MS. The differential metabolites between the exposed and control rats were identified with partial least squares discriminant analysis (PLS-DA) and the metabolic pathways related with the differential metabolites were analyzed using Cytoscape software. RESULTS: Compared with the control group, the rats exposed to B[a]P showed significantly increased escape latency (P<0.05) and decreased time spent in the target area (P<0.05). The exposed rats exhibited widened synaptic cleft, thickened endplate membrane and swollen cytoplasm compared with the control rats. Eighteen differential metabolites (VIP>1, P<0.05) in the cortex were identified between the two groups, and 9 pathways associated with B[a]P neurotoxicity were identified involving amino acid metabolism, tricarboxylic acid cycle and Vitamin B3 (niacin and nicotinamide) metabolism. CONCLUSION: B[a]P can cause disturbance in normal metabolisms and its neurotoxicity is possibly related with disorders in amino acid metabolism, tricarboxylic acid cycle and vitamin metabolism.


Asunto(s)
Benzo(a)pireno/efectos adversos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Metaboloma , Animales , Aprendizaje por Laberinto , Ratas , Ratas Sprague-Dawley
14.
Toxicol Mech Methods ; 27(8): 598-608, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28583009

RESUMEN

Arsenite is a toxic metalloid that may increase the risk of Parkinson's disease by inducing dopaminergic neuronal apoptosis. Luteolin, a common dietary flavonoid, possesses variety of biological functions, but potential effects of luteolin on arsenite toxicity remain elusive. In this study, we demonstrated that luteolin prevented arsenite-induced apoptosis in the dopaminergic PC12 cells. Administration of luteolin to cells attenuated arsenite-induced ROS production, enhanced caspase-3 activity and γ-H2AX expression. Our results further showed the expression of α-Synuclein (α-Syn) was significantly increased in arsenite-treated cells, but co-treatment with luteolin reversed the expression of α-Syn back toward normal level. Inhibition of α-Syn by siRNA remarkably enhanced the beneficial effect of luteolin against arsenite-induced apoptotic cell death. Taken together, these results demonstrate that the ameliorative effects of luteolin against arsenite in the dopaminergic cell may be modulated by α-Syn, and indicating that luteolin may be developed as a chemopreventive supplementary agent to ameliorate dopaminergic cell apoptosis resulting from arsenite exposure.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenitos/toxicidad , Dieta , Dopamina/metabolismo , Luteolina/farmacología , alfa-Sinucleína/antagonistas & inhibidores , Animales , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...