Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dose Response ; 11(1): 49-59, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23550232

RESUMEN

Acrylonitrile(AN) is a neurotoxin both in animals and humans, but its effects on acetylcholinesterase (AChE) activity remain controversial. This study aimed to determine the dose-response effects of AN on AChE activity and the modulatory role of ethanol pre-treatment. A total of 144 Kunming mice were randomly divided into 18 groups: nine groups received 5% ethanol in their drinking water, and the remaining nine groups received regular tap water. One week later, both the ethanol and tap water only groups were given an intraperitoneal injection of AN at the following doses: 0 (control), 0.156, 0.3125, 0.625, 1.25, 2.5, 5, 10 or 20 mg AN/kg body weight. AChE activity was determined on whole blood and brain 24 h later. Blood AChE activity was higher in AN-injected mice than in controls at all doses. AChE activity in blood increased in a dose-dependent manner, peaking at 0.156 mg/kg, after which a gradual decrease ensued, displaying a ß-typed dose-response relationship. In contrast, brain AChE activity, following a single AN injection, was consistently lower than in control mice, and continued to fall up to a dose of 0.313 mg/kg, and thereafter increased gradually with higher doses. Mice receiving a 20 mg/kg dose of AN exhibited AChE brain activity indistinguishable from that of control mice, demonstrating a typical U-typed dose-response relationship. The activity of AChE in the blood and brain of the AN + ethanol-treated groups displayed a shift to the right, and the magnitude of the decrease in AChE activity induced by AN was attenuated relative to the AN-only group. These results suggest that AN affects AChE activity in both mouse blood and brain in a hormetic manner. Pretreatment with ethanol modifies the effect of AN on AChE, indicating that parent AN has a more prominent role than its metabolites in modulating enzyme activity.

2.
J Neurochem ; 121(1): 125-34, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22260250

RESUMEN

Ammonia is responsible for cerebral edema associated with acute liver failure, but the role of the vasogenic mechanism has been a matter of dispute. Here, we tested the hypothesis that ammonia induces changes in blood-brain barrier (BBB) permeability by a mechanism coupled to oxidative/nitrosative stress (ONS) evoked in the BBB-forming cerebral capillary endothelial cells. Treatment of a rat brain endothelial cell line with ammonia (5 mmol/L, 24 h) caused accumulation of ONS markers: reactive oxygen species, nitric oxide and peroxidation products of phospholipid-bound arachidonic acid, F2-isoprostanes. Concurrently, ammonia increased the activity of extracellular matrix metalloproteinases (MMP-2/MMP-9), increased cell permeability to fluorescein isothiocyanate-dextran (40 kDa), and increased the expression of y+LAT2, a transporter that mediates the uptake to the cells of the nitric oxide precursor, arginine. The increase of cell permeability was ameliorated upon co-treatment with a MMP inhibitor, SB-3CT and with an antioxidant, glutathione diethyl ester, which also reduced F2-isoprostanes. Ammonia-induced ONS was attenuated by cytoprotective agents l-ornithine, phenylbutyrate, and their conjugate l-ornithine phenylbutyrate, an ammonia-trapping drug used to treat hyperammonemia. The results support the concept that ONS and ONS-related activation of MMPs in cerebral capillary endothelial cells contribute to the alterations in BBB permeability and to the vasogenic component of cerebral edema associated with acute liver failure.


Asunto(s)
Amoníaco/farmacología , Encéfalo/irrigación sanguínea , Permeabilidad de la Membrana Celular/fisiología , Células Endoteliales/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Óxido Nítrico/fisiología , Estrés Oxidativo/fisiología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo
3.
Toxicol Appl Pharmacol ; 258(3): 394-402, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22178737

RESUMEN

Chronic, excessive exposure to manganese (Mn) may induce neurotoxicity and cause an irreversible brain disease, referred to as manganism. Efficacious therapies for the treatment of Mn are lacking, mandating the development of new interventions. The purpose of the present study was to investigate the efficacy of ebselen (Ebs) and para-aminosalicylic acid (PAS) in attenuating the neurotoxic effects of Mn in an in vivo rat model. Exposure biomarkers, inflammatory and oxidative stress biomarkers, as well as behavioral parameters were evaluated. Co-treatment with Mn plus Ebs or Mn plus PAS caused a significant decrease in blood and brain Mn concentrations (compared to rats treated with Mn alone), concomitant with reduced brain E2 prostaglandin (PGE2) and enhanced brain glutathione (GSH) levels, decreased serum prolactin (PRL) levels, and increased ambulation and rearing activities. Taken together, these results establish that both PAS and Ebs are efficacious in reducing Mn body burden, neuroinflammation, oxidative stress and locomotor activity impairments in a rat model of Mn-induced toxicity.


Asunto(s)
Ácido Aminosalicílico/farmacología , Azoles/farmacología , Manganeso/toxicidad , Síndromes de Neurotoxicidad/prevención & control , Compuestos de Organoselenio/farmacología , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/prevención & control , Isoindoles , Masculino , Manganeso/farmacocinética , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/etiología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Distribución Tisular
4.
Neurotoxicology ; 33(4): 763-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22186600

RESUMEN

Methylmercury (MeHg) is an environmental neurotoxicant associated with aberrant central nervous system (CNS) functions. In this study, we examined the protective effect of a novel anti-inflammatory and cytoprotective nonapeptide, termed IIIM1, against MeHg-induced toxicity in cultured rat neonatal primary astrocytes. Astrocytes were pretreated for 66 h with 5 µg/ml IIIM1 (4.95 µM) followed by 6 h exposure to MeHg (5 µM). MeHg significantly increased F(2)-isoprostane generation, a lipid peroxidation biomarker of oxidative injury and this effect was significantly reduced upon pre-treatment with IIIM1. The MeHg-induced increase in levels of prostaglandin E(2) (PGE(2)), biomarkers of inflammatory responses, was also decreased in the peptide-treated cells. Mass spectrometry analysis revealed no chemical or binding interaction between MeHg and IIIM1, indicating that intracellular cytoprotective mechanism of action accounts for the neuroprotection rather than direct intracellular neutralization of the neurotoxicant with the peptide. These findings point to therapeutic potential for IIIM1 in a plethora of conditions associated with reactive oxygen species (ROS) generation. The implication of these findings may prove beneficial in designing new treatment modalities that efficiently suppress neurotoxicity, triggered not only by MeHg, but also by other metals and environmental agents, as well as chronic disease conditions that inherently increase reactive radical production and inflammatory signaling.


Asunto(s)
Astrocitos/efectos de los fármacos , Histonas/farmacología , Compuestos de Metilmercurio/toxicidad , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Astrocitos/patología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , F2-Isoprostanos/metabolismo , Mediadores de Inflamación/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Espectrometría de Masas , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
5.
Toxicology ; 292(2-3): 90-8, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22154916

RESUMEN

BACKGROUND: Manganese (Mn) is a naturally occurring element and an essential nutrient for humans and animals. However, exposure to high levels of Mn may cause neurotoxic effects. The pathological mechanisms associated with Mn neurotoxicity are poorly understood, but several reports have established it is mediated, at least in part, by oxidative stress. OBJECTIVES: The present study was undertaken to test the hypothesis that a decrease in acetylcholinesterase (AChE) activity mediates Mn-induced neurotoxicity. METHODS: Groups of 6 rats received 4 or 8 intraperitoneal (i.p.) injections of 25mg MnCl(2)/kg/day, every 48 h. Twenty-four hours after the last injection, brain AChE activity and the levels of F(2)-isoprostanes (F(2)-IsoPs) and F(4)-neuroprostanes (F(4)-NPs) (biomarkers of oxidative stress), as well as prostaglandin E(2) (PGE(2)) (biomarker of neuroinflammation) were analyzed. RESULTS: The results showed that after either 4 or 8 Mn doses, brain AChE activity was significantly decreased (p<0.05), to 60 ± 16% and 55 ± 13% of control levels, respectively. Both treated groups exhibited clear signs of neurobehavioral toxicity, characterized by a significant (p<0.001) decrease in ambulation and rearings in open-field. Furthermore, Mn treatment caused a significant increase (p<0.05) in brain F(2)-IsoPs and PGE(2) levels, but only after 8 doses. In rats treated with 4 Mn doses, a significant increase (p<0.05) in brain F(4)-NPs levels was found. To evaluate cellular responses to oxidative stress, we assessed brain nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) and Mn-superoxide dismutase (Mn-SOD, SOD2) protein expression levels. A significant increase in Mn-SOD protein expression (p<0.05) and a trend towards increased Nrf2 protein expression was noted in rat brains after 4 Mn doses vs. the control group, but the expression of these proteins was decreased after 8 Mn doses. Taken together, these results suggest that the inhibitory effect of Mn on AChE activity promotes increased levels of neuronal oxidative stress and neuroinflammatory biomarkers.


Asunto(s)
Acetilcolinesterasa/metabolismo , Encéfalo/efectos de los fármacos , Cloruros/toxicidad , Inhibidores de la Colinesterasa/toxicidad , Encefalitis/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Animales , Western Blotting , Encéfalo/enzimología , Encéfalo/metabolismo , Dinoprostona/metabolismo , Encefalitis/enzimología , Encefalitis/metabolismo , F2-Isoprostanos/metabolismo , Masculino , Compuestos de Manganeso , Actividad Motora/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba
7.
Methods Mol Biol ; 758: 195-204, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21815067

RESUMEN

Oxidative stress results from an imbalance between production of reactive oxygen and nitrogen species (ROS and RNS, respectively) and endogenous antioxidant defense mechanisms. Increased generation of ROS/RNS is implicated in the pathogenesis of a variety of human diseases, including neurodegenerative disease, atherosclerosis, cancer, and aging. However, measuring oxidative stress in biological systems is complex and requires accurate quantification of either free radicals or damaged biomolecules. One method to quantify oxidative injury is to measure lipid peroxidation. Lipids are readily attacked by free radicals, resulting in the formation of a number of peroxidation products. F2-isoprostanes (F2-IsoPs) are one group of these compounds and they are derived by the free radical peroxidation of arachidonic acid (AA). The F2-IsoPs, prostaglandine F2-like compounds, provide an accurate measure of oxidative stress both in vitro and in vivo. This protocol details current methodology used to quantify these molecules using gas chromatography-mass spectrometry (GC/MS).


Asunto(s)
Isoprostanos/metabolismo , Estrés Oxidativo , Animales , Biomarcadores/química , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Cromatografía de Gases , Humanos , Hidrólisis , Isoprostanos/química , Límite de Detección , Lípidos/química , Lípidos/aislamiento & purificación , Espectrometría de Masas/métodos
8.
Brain Res ; 1398: 1-12, 2011 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-21636077

RESUMEN

Selenoprotein P (Sepp1) is an important protein involved in selenium (Se) transport and homeostasis. Severe neurologic dysfunction develops in Sepp1 null mice (Sepp1(-/-)) fed a selenium-deficient diet. Sepp1(-/-) mice fed a selenium-deficient diet have extensive degeneration of the brainstem and thalamus, and even when supplemented with selenium exhibit subtle learning deficits and altered basal synaptic transmission and short-term plasticity in the CA1 region of the hippocampus. The goal of this study was to delineate the regional progression of neurodegeneration in the brain, determine the extent of neuronal cell death, and evaluate neurite structural changes within the hippocampus of Sepp1(-/-) mice. Whole brain serial sections of wild-type and Sepp1(-/-) mice maintained on selenium-deficient or supplemented diets over the course of 12 days from weaning were evaluated with amino cupric silver neurodegeneration stain. The neurodegeneration was present in all regions upon weaning and progressed over 12 days in Sepp1(-/-) mice fed selenium-deficient diet, except in the medial forebrain bundle and somatosensory cortex where the neurodegeneration developed post-weaning. The neurodegeneration was predominantly axonal, however the somatosensory cortex and lateral striatum showed silver-stained neurons. Morphologic analysis of the hippocampus revealed decreased dendritic length and spine density, suggesting that loss of Sepp1 also causes subtle changes in the brain that can contribute to functional deficits. These data illustrate that deletion of Sepp1, and presumably selenium deficiency in the brain, produce both neuronal and axonal degeneration as well as more moderate and potentially reversible neurite changes in the developing brain.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Enfermedades Neurodegenerativas/metabolismo , Selenio/deficiencia , Selenoproteína P/deficiencia , Selenoproteína P/genética , Animales , Axones/metabolismo , Axones/patología , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuritas/metabolismo , Neuritas/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología
9.
Toxicol Appl Pharmacol ; 256(3): 219-26, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21684300

RESUMEN

Exposure to excessive manganese (Mn) levels leads to neurotoxicity, referred to as manganism, which resembles Parkinson's disease (PD). Manganism is caused by neuronal injury in both cortical and subcortical regions, particularly in the basal ganglia. The basis for the selective neurotoxicity of Mn is not yet fully understood. However, several studies suggest that oxidative damage and inflammatory processes play prominent roles in the degeneration of dopamine-containing neurons. In the present study, we assessed the effects of Mn on reactive oxygen species (ROS) formation, changes in high-energy phosphates and associated neuronal dysfunctions both in vitro and in vivo. Results from our in vitro study showed a significant (p<0.01) increase in biomarkers of oxidative damage, F(2)-isoprostanes (F(2)-IsoPs), as well as the depletion of ATP in primary rat cortical neurons following exposure to Mn (500 µM) for 2h. These effects were protected when neurons were pretreated for 30 min with 100 of an antioxidant, the hydrophilic vitamin E analog, trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid), or an anti-inflammatory agent, indomethacin. Results from our in vivo study confirmed a significant increase in F(2)-IsoPs levels in conjunction with the progressive spine degeneration and dendritic damage of the striatal medium spiny neurons (MSNs) of mice exposed to Mn (100mg/kg, s.c.) 24h. Additionally, pretreatment with vitamin E (100mg/kg, i.p.) or ibuprofen (140 µg/ml in the drinking water for two weeks) attenuated the Mn-induced increase in cerebral F(2)-IsoPs? and protected the MSNs from dendritic atrophy and dendritic spine loss. Our findings suggest that the mediation of oxidative stress/mitochondrial dysfunction and the control of alterations in biomarkers of oxidative injury, neuroinflammation and synaptodendritic degeneration may provide an effective, multi-pronged therapeutic strategy for protecting dysfunctional dopaminergic transmission and slowing of the progression of Mn-induced neurodegenerative processes.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Intoxicación por Manganeso/prevención & control , Síndromes de Neurotoxicidad/etiología , Estrés Oxidativo/efectos de los fármacos , Adenosina Trifosfato/análisis , Animales , Células Cultivadas , Cerebro/química , Cerebro/efectos de los fármacos , Cromanos/farmacología , Dinoprostona/análisis , F2-Isoprostanos/análisis , Femenino , Ibuprofeno/farmacología , Indometacina/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/química , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/prevención & control , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/análisis
10.
Toxicol Appl Pharmacol ; 256(3): 300-13, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21704645

RESUMEN

Alzheimer's disease (AD) is characterized by a progressive cognitive decline and accumulation of neurotoxic oligomeric peptides amyloid-ß (Aß). Although the molecular events are not entirely known, it has become evident that inflammation, environmental and other risk factors may play a causal, disruptive and/or protective role in the development of AD. The present study investigated the ability of the chemokines, macrophage inflammatory protein-2 (MIP-2) and stromal cell-derived factor-1α (SDF-1α), the respective ligands for chemokine receptors CXCR2 and CXCR4, to suppress Aß-induced neurotoxicity in vitro and in vivo. Pretreatment with MIP-2 or SDF-1α significantly protected neurons from Aß-induced dendritic regression and apoptosis in vitro through activation of Akt, ERK1/2 and maintenance of metalloproteinase ADAM17 especially with SDF-1α. Intra-cerebroventricular (ICV) injection of Aß led to reduction in dendritic length and spine density of pyramidal neurons in the CA1 area of the hippocampus and increased oxidative damage 24h following the exposure. The Aß-induced morphometric changes of neurons and increase in biomarkers of oxidative damage, F(2)-isoprostanes, were significantly inhibited by pretreatment with the chemokines MIP-2 or SDF-1α. Additionally, MIP-2 or SDF-1α was able to suppress the aberrant mislocalization of p21-activated kinase (PAK), one of the proteins involved in the maintenance of dendritic spines. Furthermore, MIP-2 also protected neurons against Aß neurotoxicity in CXCR2-/- mice, potentially through observed up regulation of CXCR1 mRNA. Understanding the neuroprotective potential of chemokines is crucial in defining the role for their employment during the early stages of neurodegeneration.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Encéfalo/efectos de los fármacos , Quimiocina CXCL12/farmacología , Quimiocina CXCL2/farmacología , Quimiocinas/farmacología , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis/efectos de los fármacos , F2-Isoprostanos/análisis , Ratones , Ratones Endogámicos C57BL , Neuronas/química , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/prevención & control , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-8B/metabolismo
11.
Neurotoxicology ; 32(3): 312-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21376752

RESUMEN

The prostanoids, a naturally occurring subclass of eicosanoids, are lipid mediators generated through oxidative pathways from arachidonic acid. These cyclooxygenase metabolites, consisting of the prostaglandins (PG), prostacyclin and tromboxane, are released in response to a variety of physiological and pathological stimuli in almost all organs, including the brain. They are produced by various cell types and act upon targeted cells via specific G protein-coupled receptors. The existence of multiple receptors, cross-reactivity and coupling to different signal transduction pathways for each prostanoid, collectively establish their diverse effects. Notably, these effects can occur in functionally opposing directions within the same cell or organ. Prostaglandin E(2) (PGE(2)) is the most versatile prostanoid because of its receptors, E Prostanoid (EP) receptor subtypes 1 through 4, its biological heterogeneity and its differential expression on neuronal and glial cells throughout the central nervous system. Since PGE(2) plays an important role in processes associated with various neurological diseases, this review focuses on its dual neuroprotective and neurotoxic role in EP receptor subtype signaling pathways in different models of brain injury.


Asunto(s)
Encéfalo/metabolismo , Dinoprostona/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Animales , Humanos , Modelos Animales
12.
Neurotoxicology ; 32(3): 291-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21300091

RESUMEN

Methylmercury (MeHg) preferentially accumulates in glia of the central nervous system (CNS), but its toxic mechanisms have yet to be fully recognized. In the present study, we tested the hypothesis that MeHg induces neurotoxicity via oxidative stress mechanisms, and that these effects are attenuated by the antioxidant, ebselen. Rat neonatal primary cortical astrocytes were pretreated with or without 10 µM ebselen for 2h followed by MeHg (0, 1, 5, and 10 µM) treatments. MeHg-induced changes in astrocytic [(3)H]-glutamine uptake were assessed along with changes in mitochondrial membrane potential (ΔΨ(m)), using the potentiometric dye tetramethylrhodamine ethyl ester (TMRE). Western blot analysis was used to detect MeHg-induced ERK (extracellular-signal related kinase) phosphorylation and caspase-3 activation. MeHg treatment significantly decreased (p<0.05) astrocytic [(3)H]-glutamine uptake at all time points and concentrations. Ebselen fully reversed MeHg's (1 µM) effect on [(3)H]-glutamine uptake at 1 min. At higher MeHg concentrations, ebselen partially reversed the MeHg-induced astrocytic inhibition of [(3)H]-glutamine uptake [at 1 min (5 and 10 µM) (p<0.05); 5 min (1, 5 and 10 µM) (p<0.05)]. MeHg treatment (1h) significantly (p<0.05) dissipated the ΔΨ(m) in astrocytes as evidenced by a decrease in mitochondrial TMRE fluorescence. Ebselen fully reversed the effect of 1 µM MeHg treatment for 1h on astrocytic ΔΨ(m) and partially reversed the effect of 5 and 10 µM MeHg treatments for 1h on ΔΨ(m). In addition, ebselen inhibited MeHg-induced phosphorylation of ERK (p<0.05) and blocked MeHg-induced activation of caspase-3 (p<0.05-0.01). These results are consistent with the hypothesis that MeHg exerts its toxic effects via oxidative stress and that the phosphorylation of ERK and the dissipation of the astrocytic mitochondrial membrane potential are involved in MeHg toxicity. In addition, the protective effects elicited by ebselen reinforce the idea that organic selenocompounds represent promising strategies to counteract MeHg-induced neurotoxicity.


Asunto(s)
Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Azoles/farmacología , Contaminantes Ambientales/toxicidad , Intoxicación del Sistema Nervioso por Mercurio/etiología , Compuestos de Metilmercurio/toxicidad , Fármacos Neuroprotectores/farmacología , Compuestos de Organoselenio/farmacología , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Astrocitos/patología , Western Blotting , Caspasa 3/metabolismo , Células Cultivadas , Citoprotección , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glutamina/metabolismo , Isoindoles , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Intoxicación del Sistema Nervioso por Mercurio/metabolismo , Intoxicación del Sistema Nervioso por Mercurio/patología , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
Neurotoxicology ; 32(5): 578-85, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21241737

RESUMEN

Among several available antimalarial drugs, mefloquine has proven to be effective against drug-resistant Plasmodium falciparum and remains the drug of choice for both therapy and chemoprophylaxis. However, mefloquine is known to cause adverse neurological and/or psychiatric symptoms, which offset its therapeutic advantage. The exact mechanisms leading to the adverse neurological effects of mefloquine are poorly defined. Alterations in neurotransmitter release and calcium homeostasis, the inhibition of cholinesterases and the interaction with adenosine A(2A) receptors have been hypothesized to play prominent roles in mediating the deleterious effects of this drug. Our recent data have established that mefloquine can also trigger oxidative damage and subsequent neurodegeneration in rat cortical primary neurons. Furthermore, we have utilized a system biology-centered approach and have constructed a pathway model of cellular responses to mefloquine, identifying non-receptor tyrosine kinase 2 (Pyk2) as a critical target in mediating mefloquine neurotoxicity. In this study, we sought to establish an experimental validation of Pyk2 using gene-silencing techniques (siRNA). We have examined whether the downregulation of Pyk2 in primary rat cortical neurons alters mefloquine neurotoxicity by evaluating cell viability, apoptosis and oxidative stress. Results from our study have confirmed that mefloquine neurotoxicity is associated with apoptotic response and oxidative injury, and we have demonstrated that mefloquine affects primary rat cortical neurons, at least in part, via Pyk2. The implication of these findings may prove beneficial in suppressing the neurological side effects of mefloquine and developing effective therapeutic modalities to offset its adverse effects.


Asunto(s)
Corteza Cerebral/enzimología , Mefloquina/toxicidad , TYK2 Quinasa/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley
14.
Neuromethods ; 56: 349-363, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-32132769
15.
Neurotoxicology ; 32(5): 518-25, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21159318

RESUMEN

γ-Glutamylcysteine (γ-GC) is an intermediate molecule of the glutathione (GSH) synthesis pathway. In the present study, we tested the hypothesis that γ-GC pretreatment in cultured astrocytes and neurons protects against hydrogen peroxide (H(2)O(2))-induced oxidative injury. We demonstrate that pretreatment with γ-GC increases the ratio of reduced:oxidized GSH levels in both neurons and astrocytes and increases total GSH levels in neurons. In addition, γ-GC pretreatment decreases isoprostane formation both in neurons and astrocytes, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation in astrocytes in response to H(2)O(2)-induced oxidative stress. Furthermore, GSH and isoprostane levels significantly correlate with increased neuron and astrocyte viability in cells pretreated with γ-GC. Finally, we demonstrate that administration of a single intravenous injection of γ-GC to mice significantly increases GSH levels in the brain, heart, lungs, liver, and in muscle tissues in vivo. These results support a potential therapeutic role for γ-GC in the reduction of oxidant stress-induced damage in tissues including the brain.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Dipéptidos/farmacología , Glutatión/metabolismo , Neuronas/metabolismo , Estrés Oxidativo/fisiología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
16.
PLoS Genet ; 6(8)2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20865164

RESUMEN

Parkinson's disease (PD)-mimicking drugs and pesticides, and more recently PD-associated gene mutations, have been studied in cell cultures and mammalian models to decipher the molecular basis of PD. Thus far, a dozen of genes have been identified that are responsible for inherited PD. However they only account for about 8% of PD cases, most of the cases likely involving environmental contributions. Environmental manganese (Mn) exposure represents an established risk factor for PD occurrence, and both PD and Mn-intoxicated patients display a characteristic extrapyramidal syndrome primarily involving dopaminergic (DAergic) neurodegeneration with shared common molecular mechanisms. To better understand the specificity of DAergic neurodegeneration, we studied Mn toxicity in vivo in Caenorhabditis elegans. Combining genetics and biochemical assays, we established that extracellular, and not intracellular, dopamine (DA) is responsible for Mn-induced DAergic neurodegeneration and that this process (1) requires functional DA-reuptake transporter (DAT-1) and (2) is associated with oxidative stress and lifespan reduction. Overexpression of the anti-oxidant transcription factor, SKN-1, affords protection against Mn toxicity, while the DA-dependency of Mn toxicity requires the NADPH dual-oxidase BLI-3. These results suggest that in vivo BLI-3 activity promotes the conversion of extracellular DA into toxic reactive species, which, in turn, can be taken up by DAT-1 in DAergic neurons, thus leading to oxidative stress and cell degeneration.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Dopamina/metabolismo , Espacio Extracelular/metabolismo , Manganeso/toxicidad , Degeneración Nerviosa , Estrés Oxidativo , Oxidorreductasas/metabolismo , Enfermedad de Parkinson/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Espacio Extracelular/genética , Femenino , Humanos , Esperanza de Vida , Masculino , Manganeso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Transporte de Proteínas
17.
Neurotoxicology ; 31(5): 608-20, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20620165

RESUMEN

The increasing exposure to environmental neurotoxicants in the last decades caused serious health problems in the world population. Some of the neurotoxic agents are being used in agriculture and household such as insecticides and rodenticides and others are of natural origin like snake and scorpion venoms. Additional group of harmful substances is the chemical warfare agents including nerve and blistering agents that are known for their disastrous effects on neuronal tissues. The present paper presents a combination of epidemiological/clinical and molecular approaches for investigating the effect of certain groups of neurotoxicants on a variety of pathologies. The work of Finkelstein and coworkers describes epidemiological and clinical studies on acute and chronic organophosphate (OP)-induced neurotoxicity in certain populations in Israel. They mainly investigated the neurotoxic effects of low-level long-term exposure to OP in agricultural areas but also dealt with acute exposures as well. A molecular approach to OP mechanism of neuronal injury was described by Milatovic and coworkers. They demonstrated OP-induced oxidative injury in pyramidal neurons in the CA1 hippocampal area and its suppression by antioxidants. Lecht and coworkers described the novel snake venom angioneurins as important mediators of the physiological cross-talk between the cardiovascular and nervous systems. They also showed that under certain conditions these angioneurins may induce pathologies such as tumor development or disruption of the vascular barrier function during envenomation. Additional mechanistic/therapeutic approach was presented by Brodsky, Rosengarten, Proscura, Shapira and Wormser. They developed a novel anti-inflammatory peptide that reduced skin irritation induced by heat and sulfur mustard (SM) stimuli. Since SM causes neuropsychiatric symptoms and alterations in neurological functions this peptide may serve as a potential treatment of neuronal injuries caused by environmental neurotoxicants. These reviews highlight different aspects of neurotoxicity, addressing epidemiology and mechanisms of toxicity; and identifying novel potential therapies.


Asunto(s)
Contaminantes Ambientales/toxicidad , Síndromes de Neurotoxicidad/etiología , Neurotoxinas/toxicidad , Humanos , Israel/epidemiología , Síndromes de Neurotoxicidad/epidemiología
18.
Neurotoxicology ; 31(5): 518-23, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20562019

RESUMEN

Mefloquine is an effective antimalarial that can cause adverse neurological events including headache, nausea, fatigue, insomnia, anxiety and depression. In this study, we examined the oxidative stress response in primary rat cortical neurons treated with mefloquine by quantifying oxidative stress markers glutathione (GSH) and F(2)-isoprostanes (F(2)-isoPs). Furthermore, we examined whether mefloquine induces synaptodendritic degeneration of primary rat cortical neurons. GSH was quantified in cortical neurons after 24-h treatment with mefloquine (0, 1, 5, 10 microM) using monochlorobimane. F(2)-isoPs were quantified in cortical neurons after 24-h treatment with mefloquine (0, 1, 5, 10 microM) using a stable isotope dilution method with detection by gas chromatography/mass spectrometry and selective ion monitoring. The concentration dependent decrease in GSH and the concomitant increase of F(2)-isoPs indicates the presence of oxidative stress in primary rat cortical neurons treated with mefloquine. Following a 24-h treatment with mefloquine, primary rat cortical neurons (0, 5, 10 microM) were fixed with 4% paraformaldehyde. Images from eight optical sections covering a distance of 2.88 microm on the z-axis were acquired using a confocal laser scanning unit. Traced images were analyzed with NeuroExplorer, a neurophysiological data analysis package. Mefloquine induces a concentration dependent decrease in the number of spines per neuron and the spine density, suggesting that mefloquine induced oxidative stress may be associated with the synaptodendritic degeneration. Together with previous work, there is strong evidence that a relationship exists between calcium homeostasis disruption, ER stress response, the oxidative stress response, and neurodegeneration. Understanding how oxidative stress alters the morphology of cortical neurons treated with mefloquine will provide further insight into the mechanism(s) related to clinically observed adverse neurological events.


Asunto(s)
Antimaláricos/farmacología , Corteza Cerebral/citología , Mefloquina/farmacología , Degeneración Nerviosa/inducido químicamente , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , F2-Isoprostanos/metabolismo , Glutatión/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Neuronas/citología , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
19.
Toxicol Sci ; 116(2): 590-603, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20421342

RESUMEN

The neurotoxicity of methylmercury (MeHg) is well documented in both humans and animals. MeHg causes acute and chronic damage to multiple organs, most profoundly the central nervous system (CNS). Microglial cells are derived from macrophage cell lineage, making up approximately 12% of cells in the CNS, yet their role in MeHg-induced neurotoxicity is not well defined. The purpose of the present study was to characterize microglial vulnerability to MeHg and their potential adaptive response to acute MeHg exposure. We examined the effects of MeHg on microglial viability, reactive oxygen species (ROS) generation, glutathione (GSH) level, redox homeostasis, and Nrf2 protein expression. Our data showed that MeHg (1-5 microM) treatment caused a rapid (within 1 min) concentration- and time-dependent increase in ROS generation, accompanied by a statistically significant decrease in the ratio of GSH and its oxidized form glutathione disulfide (GSSG) (GSH:GSSG ratio). MeHg increased the cytosolic Nrf2 protein level within 1 min of exposure, followed by its nuclear translocation after 10 min of treatment. Consistent with the nuclear translocation of Nrf2, quantitative real-time PCR revealed a concentration-dependent increase in the messenger RNA level of Ho-1, Nqo1, and xCT 30 min post MeHg exposure, whereas Nrf2 knockdown greatly reduced the upregulation of these genes. Furthermore, we observed increased microglial death upon Nrf2 knockdown by the small hairpin RNA approach. Taken together, our study has demonstrated that microglial cells are exquisitely sensitive to MeHg and respond rapidly to MeHg by upregulating the Nrf2-mediated antioxidant response.


Asunto(s)
Compuestos de Metilmercurio/toxicidad , Microglía/efectos de los fármacos , Factor 2 Relacionado con NF-E2/análisis , Estrés Oxidativo/efectos de los fármacos , Sistema de Transporte de Aminoácidos y+/genética , Sistemas de Transporte de Aminoácidos Acídicos , Animales , Células Cultivadas , Glutatión/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Interleucina-6/biosíntesis , Microglía/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
20.
Curr Protoc Toxicol ; Chapter 12: Unit 12.16, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20401325

RESUMEN

The study of dendritic length and spine density has become a standard in the analysis of neuronal abnormalities since a considerable number of neurological diseases have their foundation in alterations in these structures. One of the best ways to study possible alterations in neuronal morphometry is the use of Golgi impregnation. Introduced more than a century ago, it is still the standard and state-of-the-art technique for visualization of neuronal architecture. We successfully applied the Golgi method to mouse, rat, monkey and human brain tissues for studying both the normal and abnormal morphology of neurons. We were able to discover subtle morphological alterations in neuronal dendrites and dendritic spines in different brain areas. Although Golgi preparations can be examined by electronic microscopy, we used light microscopy and Neurolucida reconstruction to quantitatively explore the relationship between total dendritic length and spine density in different types of neurons. This review summarizes the methodology used to quantify neuronal abnormalities and discusses the utility of these techniques in different models of neurodegeneration.


Asunto(s)
Encéfalo/ultraestructura , Dendritas/ultraestructura , Técnicas de Preparación Histocitológica/métodos , Enfermedades Neurodegenerativas/patología , Animales , Espinas Dendríticas/ultraestructura , Humanos , Células de Purkinje/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...